PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 33688971-0 2021 Effect of irradiation on the expression of E-cadherin and beta-catenin in early and late radiation sequelae of the urinary bladder and its modulation by NF-kappaB inhibitor thalidomide. Thalidomide 173-184 cadherin 1 Mus musculus 43-53 6548496-0 1984 Thalidomide in lepra reaction (ENL) in lepromatous leprosy patients. Thalidomide 0-11 MLLT1 super elongation complex subunit Homo sapiens 31-34 6548496-6 1984 Control of ENL reaction by thalidomide in these patients is probably due to its immunosuppressive effect, more likely by its stablising action on lysosomes. Thalidomide 27-38 MLLT1 super elongation complex subunit Homo sapiens 11-14 5300279-0 1966 [Thalidomide: effect on serum ceruloplasmin]. Thalidomide 1-12 ceruloplasmin Homo sapiens 30-43 33878647-0 2021 Design and linkage optimization of ursane-thalidomide-based PROTACs and identification of their targeted-degradation properties to MDM2 protein. Thalidomide 42-53 transformed mouse 3T3 cell double minute 2 Mus musculus 131-135 3970671-7 1985 Glucagon administration (GLUPAN) transiently increased CI and QP but failed to improve O2CP and did not change QAVS. Thalidomide 25-31 glucagon Canis lupus familiaris 0-8 700507-3 1978 Contrary to N-ethyl-maleimide, thalidomide does not block the SH groups irreversible, and they can be reactivated with BAL. Thalidomide 31-42 solute carrier family 27 member 5 Rattus norvegicus 119-122 5593734-1 1967 On the necessary 2nd noxa in experimental thalidomide embryopathies]. Thalidomide 42-53 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 21-25 33780898-0 2021 Discovery of thalidomide-based PROTAC small molecules as the highly efficient SHP2 degraders. Thalidomide 13-24 protein tyrosine phosphatase non-receptor type 11 Homo sapiens 78-82 33780898-4 2021 Herein we described the design, synthesis and evaluation of a series of thalidomide-based heterobifunctional molecules and identified 11(ZB-S-29) as the highly efficient SHP2 degrader with a DC50 of 6.02 nM. Thalidomide 72-83 protein tyrosine phosphatase non-receptor type 11 Homo sapiens 170-174 33688971-0 2021 Effect of irradiation on the expression of E-cadherin and beta-catenin in early and late radiation sequelae of the urinary bladder and its modulation by NF-kappaB inhibitor thalidomide. Thalidomide 173-184 catenin (cadherin associated protein), beta 1 Mus musculus 58-70 33688971-0 2021 Effect of irradiation on the expression of E-cadherin and beta-catenin in early and late radiation sequelae of the urinary bladder and its modulation by NF-kappaB inhibitor thalidomide. Thalidomide 173-184 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 153-162 33688971-1 2021 PURPOSE: In a previous study we have shown in a mouse model that administration of nuclear factor-kappa B (NF-kappaB) inhibitor thalidomide has promising therapeutic effects on early radiation cystitis (ERC) and late radiation sequelae (LRS) of the urinary bladder. Thalidomide 128-139 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 83-105 33688971-1 2021 PURPOSE: In a previous study we have shown in a mouse model that administration of nuclear factor-kappa B (NF-kappaB) inhibitor thalidomide has promising therapeutic effects on early radiation cystitis (ERC) and late radiation sequelae (LRS) of the urinary bladder. Thalidomide 128-139 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 107-116 33688971-7 2021 Early administration of thalidomide (day 1-15) resulted in a steeper rise in the first days, an extended and increased expression at the end of the early phase and a higher expression of beta-catenin alone at the beginning of the late phase. Thalidomide 24-35 catenin (cadherin associated protein), beta 1 Mus musculus 187-199 33688971-9 2021 Early administration of thalidomide improves these compensatory mechanisms by inhibiting NF-kappaB signaling and its interfering effects. Thalidomide 24-35 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 89-98 33751979-3 2021 We firstly screened CRBN ligands as the E3 ligase moiety, then obtained a series of potent ALK degraders based on different CRBN ligands, exemplified by SIAIS091 and SIAIS001 with lenalidomide/thalidomide-based linkers. Thalidomide 193-204 ALK receptor tyrosine kinase Homo sapiens 91-94 34048672-6 2021 Fusing thalidomide-the ligand of the cereblon ubiquitin ligase-to O4I2 led to a new PROTAC-O4I2, which selectively degraded SF3B1 and induced cellular apoptosis in a CRBN-dependent manner. Thalidomide 7-18 splicing factor 3b subunit 1 Homo sapiens 124-129 34048672-6 2021 Fusing thalidomide-the ligand of the cereblon ubiquitin ligase-to O4I2 led to a new PROTAC-O4I2, which selectively degraded SF3B1 and induced cellular apoptosis in a CRBN-dependent manner. Thalidomide 7-18 cereblon Homo sapiens 166-170 33975771-0 2021 Thalidomide reduces glycerol-induced acute kidney injury by inhibition of NF-kappaB, NLRP3 inflammasome, COX-2 and inflammatory cytokines. Thalidomide 0-11 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 74-83 33975771-0 2021 Thalidomide reduces glycerol-induced acute kidney injury by inhibition of NF-kappaB, NLRP3 inflammasome, COX-2 and inflammatory cytokines. Thalidomide 0-11 NLR family, pyrin domain containing 3 Mus musculus 85-90 33975771-0 2021 Thalidomide reduces glycerol-induced acute kidney injury by inhibition of NF-kappaB, NLRP3 inflammasome, COX-2 and inflammatory cytokines. Thalidomide 0-11 cytochrome c oxidase II, mitochondrial Mus musculus 105-110 33687083-12 2021 Thalidomide (30-100 microM) inhibited noradrenaline-, phenylephrine-, and methoxamine-induced contractions, as well as nonadrenergic contractions induced by endothelin-1 and U46619, and neurogenic contractions induced by EFS. Thalidomide 0-11 endothelin 1 Homo sapiens 157-169 34033753-1 2021 Cereblon (CRBN), originally identified as a gene associated with intellectual disability, was identified as primary target of thalidomide. Thalidomide 126-137 cereblon Homo sapiens 0-8 34033753-1 2021 Cereblon (CRBN), originally identified as a gene associated with intellectual disability, was identified as primary target of thalidomide. Thalidomide 126-137 cereblon Homo sapiens 10-14 34033753-2 2021 Accumulating evidence has shown that CRBN is a substrate receptor of Cullin Ring E3 ubiquitin ligase 4 (CRL4) containing DDB1, CUL4, and RBX1, which recognizes specific neosubstrates in the presence of thalidomide or its analogs and induces their ubiquitination and proteasomal degradation. Thalidomide 202-213 cereblon Homo sapiens 37-41 34033753-2 2021 Accumulating evidence has shown that CRBN is a substrate receptor of Cullin Ring E3 ubiquitin ligase 4 (CRL4) containing DDB1, CUL4, and RBX1, which recognizes specific neosubstrates in the presence of thalidomide or its analogs and induces their ubiquitination and proteasomal degradation. Thalidomide 202-213 CDK2 associated cullin domain 1 Homo sapiens 69-75 34033753-2 2021 Accumulating evidence has shown that CRBN is a substrate receptor of Cullin Ring E3 ubiquitin ligase 4 (CRL4) containing DDB1, CUL4, and RBX1, which recognizes specific neosubstrates in the presence of thalidomide or its analogs and induces their ubiquitination and proteasomal degradation. Thalidomide 202-213 interleukin 17 receptor B Homo sapiens 104-108 34033753-2 2021 Accumulating evidence has shown that CRBN is a substrate receptor of Cullin Ring E3 ubiquitin ligase 4 (CRL4) containing DDB1, CUL4, and RBX1, which recognizes specific neosubstrates in the presence of thalidomide or its analogs and induces their ubiquitination and proteasomal degradation. Thalidomide 202-213 damage specific DNA binding protein 1 Homo sapiens 121-125 34033753-2 2021 Accumulating evidence has shown that CRBN is a substrate receptor of Cullin Ring E3 ubiquitin ligase 4 (CRL4) containing DDB1, CUL4, and RBX1, which recognizes specific neosubstrates in the presence of thalidomide or its analogs and induces their ubiquitination and proteasomal degradation. Thalidomide 202-213 ring-box 1 Homo sapiens 137-141 33938033-1 2021 Thalidomide and its derivatives lenalidomide and pomalidomide, known as immunomodulatory drugs, (IMiDs) bind directly to cereblon (CRBN), a substrate receptor of an E3 ubiquitin ligase, resulting in the rapid ubiquitination and degradation of the substrate protein. Thalidomide 0-11 cereblon Homo sapiens 131-135 33938033-3 2021 To date, almost all CRBN ligands are derived from thalidomide and there are few structural differences between them. Thalidomide 50-61 cereblon Homo sapiens 20-24 33206273-0 2021 Thalidomide Inhibits Angiogenesis via Downregulation of VEGF and Angiopoietin-2 in Crohn"s Disease. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 56-60 33926033-0 2021 Chemical Degradation of Androgen Receptor (AR) Using Bicalutamide Analog-Thalidomide PROTACs. Thalidomide 73-84 androgen receptor Homo sapiens 24-41 33926033-0 2021 Chemical Degradation of Androgen Receptor (AR) Using Bicalutamide Analog-Thalidomide PROTACs. Thalidomide 73-84 androgen receptor Homo sapiens 43-45 33926033-1 2021 A series of PROTACs (PROteolysis-TArgeting Chimeras) consisting of bicalutamide analogs and thalidomides were designed, synthesized, and biologically evaluated as novel androgen receptor (AR) degraders. Thalidomide 92-104 androgen receptor Homo sapiens 169-186 33926033-1 2021 A series of PROTACs (PROteolysis-TArgeting Chimeras) consisting of bicalutamide analogs and thalidomides were designed, synthesized, and biologically evaluated as novel androgen receptor (AR) degraders. Thalidomide 92-104 androgen receptor Homo sapiens 188-190 33206273-8 2021 Thalidomide produced a significant reduction in protein expression of Ang-2 and VEGF, along with a decrease in mRNA expression of Ang-2. Thalidomide 0-11 angiopoietin 2 Homo sapiens 70-75 33206273-8 2021 Thalidomide produced a significant reduction in protein expression of Ang-2 and VEGF, along with a decrease in mRNA expression of Ang-2. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 80-84 33206273-0 2021 Thalidomide Inhibits Angiogenesis via Downregulation of VEGF and Angiopoietin-2 in Crohn"s Disease. Thalidomide 0-11 angiopoietin 2 Homo sapiens 65-79 33206273-8 2021 Thalidomide produced a significant reduction in protein expression of Ang-2 and VEGF, along with a decrease in mRNA expression of Ang-2. Thalidomide 0-11 angiopoietin 2 Homo sapiens 130-135 33206273-13 2021 It also indicated that thalidomide can be able to deactivate endothelium by the downregulation effect on angiogenic factors by targeting VEGF and Ang-2. Thalidomide 23-34 vascular endothelial growth factor A Homo sapiens 137-141 33206273-13 2021 It also indicated that thalidomide can be able to deactivate endothelium by the downregulation effect on angiogenic factors by targeting VEGF and Ang-2. Thalidomide 23-34 angiopoietin 2 Homo sapiens 146-151 33465355-1 2021 In the present investigation, we tested the hypothesis that suppression of the phospho-extracellular signal regulated kinase (pERK1/2)-nuclear factor kappa (NFkappa)-B signaling, subsequent to tumor necrosis factor-alpha (TNF-alpha) inhibition, underlies thalidomide (TLM) mediated neuroprotection. Thalidomide 255-266 tumor necrosis factor Rattus norvegicus 193-220 33249990-2 2021 The pharmacological effects of thalidomide are attributed to its anti-angiogenic, anti-inflammatory and modulatory effect on cytokines principally tumor necrosis factor-alpha, while the teratogenic effects are linked to two molecular targets, namely cereblon and tubulin. Thalidomide 31-42 tumor necrosis factor Homo sapiens 147-174 33517358-8 2021 Metformin, thalidomide and cytokines (IFN, tumor necrosis factor (TNF), interleukin-2 (IL-2) and others) have pleiomorphic activities, some of which can enhance tumorigenesis. Thalidomide 11-22 tumor necrosis factor Homo sapiens 43-64 33517358-8 2021 Metformin, thalidomide and cytokines (IFN, tumor necrosis factor (TNF), interleukin-2 (IL-2) and others) have pleiomorphic activities, some of which can enhance tumorigenesis. Thalidomide 11-22 tumor necrosis factor Homo sapiens 66-69 33517358-8 2021 Metformin, thalidomide and cytokines (IFN, tumor necrosis factor (TNF), interleukin-2 (IL-2) and others) have pleiomorphic activities, some of which can enhance tumorigenesis. Thalidomide 11-22 interleukin 2 Homo sapiens 72-85 33517358-8 2021 Metformin, thalidomide and cytokines (IFN, tumor necrosis factor (TNF), interleukin-2 (IL-2) and others) have pleiomorphic activities, some of which can enhance tumorigenesis. Thalidomide 11-22 interleukin 2 Homo sapiens 87-91 33465355-1 2021 In the present investigation, we tested the hypothesis that suppression of the phospho-extracellular signal regulated kinase (pERK1/2)-nuclear factor kappa (NFkappa)-B signaling, subsequent to tumor necrosis factor-alpha (TNF-alpha) inhibition, underlies thalidomide (TLM) mediated neuroprotection. Thalidomide 255-266 tumor necrosis factor Rattus norvegicus 222-231 33465355-1 2021 In the present investigation, we tested the hypothesis that suppression of the phospho-extracellular signal regulated kinase (pERK1/2)-nuclear factor kappa (NFkappa)-B signaling, subsequent to tumor necrosis factor-alpha (TNF-alpha) inhibition, underlies thalidomide (TLM) mediated neuroprotection. Thalidomide 268-271 tumor necrosis factor Rattus norvegicus 193-220 33465355-1 2021 In the present investigation, we tested the hypothesis that suppression of the phospho-extracellular signal regulated kinase (pERK1/2)-nuclear factor kappa (NFkappa)-B signaling, subsequent to tumor necrosis factor-alpha (TNF-alpha) inhibition, underlies thalidomide (TLM) mediated neuroprotection. Thalidomide 268-271 tumor necrosis factor Rattus norvegicus 222-231 33465355-5 2021 Unilateral intra-hippocampal TLM ameliorated these behavioral deficits along with the following ex vivo hippocampal effects: (i) abrogation of the IR-evoked elevations in hippocampal TNF-alpha, pERK1/2, NFkappaB, BDNF, iNOS contents and (ii) partial restoration of the reduced anti-inflammatory cytokine IL-10 and p-nNOS S852. Thalidomide 29-32 tumor necrosis factor Rattus norvegicus 183-192 33465355-5 2021 Unilateral intra-hippocampal TLM ameliorated these behavioral deficits along with the following ex vivo hippocampal effects: (i) abrogation of the IR-evoked elevations in hippocampal TNF-alpha, pERK1/2, NFkappaB, BDNF, iNOS contents and (ii) partial restoration of the reduced anti-inflammatory cytokine IL-10 and p-nNOS S852. Thalidomide 29-32 brain-derived neurotrophic factor Rattus norvegicus 213-217 33465355-5 2021 Unilateral intra-hippocampal TLM ameliorated these behavioral deficits along with the following ex vivo hippocampal effects: (i) abrogation of the IR-evoked elevations in hippocampal TNF-alpha, pERK1/2, NFkappaB, BDNF, iNOS contents and (ii) partial restoration of the reduced anti-inflammatory cytokine IL-10 and p-nNOS S852. Thalidomide 29-32 nitric oxide synthase 2 Rattus norvegicus 219-223 33465355-5 2021 Unilateral intra-hippocampal TLM ameliorated these behavioral deficits along with the following ex vivo hippocampal effects: (i) abrogation of the IR-evoked elevations in hippocampal TNF-alpha, pERK1/2, NFkappaB, BDNF, iNOS contents and (ii) partial restoration of the reduced anti-inflammatory cytokine IL-10 and p-nNOS S852. Thalidomide 29-32 interleukin 10 Rattus norvegicus 304-309 33465355-5 2021 Unilateral intra-hippocampal TLM ameliorated these behavioral deficits along with the following ex vivo hippocampal effects: (i) abrogation of the IR-evoked elevations in hippocampal TNF-alpha, pERK1/2, NFkappaB, BDNF, iNOS contents and (ii) partial restoration of the reduced anti-inflammatory cytokine IL-10 and p-nNOS S852. Thalidomide 29-32 nitric oxide synthase 1 Rattus norvegicus 316-320 33015978-8 2021 Altered expression of miR-141 and HDAC6 was introduced to identify their functions in thalidomide-mediated events. Thalidomide 86-97 histone deacetylase 6 Rattus norvegicus 34-39 33470442-0 2021 Thalidomide and its metabolite 5-hydroxythalidomide induce teratogenicity via the cereblon neosubstrate PLZF. Thalidomide 0-11 zinc finger and BTB domain containing 16 Homo sapiens 104-108 33470442-1 2021 Thalidomide causes teratogenic effects by inducing protein degradation via cereblon (CRBN)-containing ubiquitin ligase and modification of its substrate specificity. Thalidomide 0-11 cereblon Gallus gallus 85-89 33470442-3 2021 Here, we report that promyelocytic leukaemia zinc finger (PLZF)/ZBTB16 is a CRBN target protein whose degradation is involved in thalidomide- and 5-hydroxythalidomide-induced teratogenicity. Thalidomide 129-140 zinc finger and BTB domain containing 16 Homo sapiens 58-62 33470442-3 2021 Here, we report that promyelocytic leukaemia zinc finger (PLZF)/ZBTB16 is a CRBN target protein whose degradation is involved in thalidomide- and 5-hydroxythalidomide-induced teratogenicity. Thalidomide 129-140 zinc finger and BTB domain containing 32 Gallus gallus 64-70 33470442-3 2021 Here, we report that promyelocytic leukaemia zinc finger (PLZF)/ZBTB16 is a CRBN target protein whose degradation is involved in thalidomide- and 5-hydroxythalidomide-induced teratogenicity. Thalidomide 129-140 cereblon Homo sapiens 76-80 33547076-3 2021 For example, thalidomide-like drugs (IMiDs) degrade the critical multiple myeloma transcription factors IKZF1 and IKZF3 by recruiting them to the cereblon E3 ubiquitin ligase. Thalidomide 13-24 IKAROS family zinc finger 1 Homo sapiens 104-109 33547076-3 2021 For example, thalidomide-like drugs (IMiDs) degrade the critical multiple myeloma transcription factors IKZF1 and IKZF3 by recruiting them to the cereblon E3 ubiquitin ligase. Thalidomide 13-24 IKAROS family zinc finger 3 Homo sapiens 114-119 33015978-8 2021 Altered expression of miR-141 and HDAC6 was introduced to identify their functions in thalidomide-mediated events. Thalidomide 86-97 microRNA 141 Rattus norvegicus 22-29 33015978-12 2021 miR-141 was responsible for thalidomide-mediated protective events by targeting HDAC6. Thalidomide 28-39 microRNA 141 Rattus norvegicus 0-7 33015978-12 2021 miR-141 was responsible for thalidomide-mediated protective events by targeting HDAC6. Thalidomide 28-39 histone deacetylase 6 Rattus norvegicus 80-85 33470442-4 2021 Using a human transcription factor protein array produced in a wheat cell-free protein synthesis system, PLZF was identified as a thalidomide-dependent CRBN substrate. Thalidomide 130-141 zinc finger and BTB domain containing 16 Homo sapiens 105-109 33470442-4 2021 Using a human transcription factor protein array produced in a wheat cell-free protein synthesis system, PLZF was identified as a thalidomide-dependent CRBN substrate. Thalidomide 130-141 cereblon Homo sapiens 152-156 33470442-5 2021 PLZF is degraded by the ubiquitin ligase CRL4CRBN in complex with thalidomide, its derivatives or 5-hydroxythalidomide in a manner dependent on the conserved first and third zinc finger domains of PLZF. Thalidomide 66-77 zinc finger and BTB domain containing 16 Homo sapiens 0-4 33470442-5 2021 PLZF is degraded by the ubiquitin ligase CRL4CRBN in complex with thalidomide, its derivatives or 5-hydroxythalidomide in a manner dependent on the conserved first and third zinc finger domains of PLZF. Thalidomide 66-77 cereblon Homo sapiens 45-49 33470442-5 2021 PLZF is degraded by the ubiquitin ligase CRL4CRBN in complex with thalidomide, its derivatives or 5-hydroxythalidomide in a manner dependent on the conserved first and third zinc finger domains of PLZF. Thalidomide 66-77 zinc finger and BTB domain containing 16 Homo sapiens 197-201 33470442-6 2021 Surprisingly, thalidomide and 5-hydroxythalidomide confer distinctly different substrate specificities to mouse and chicken CRBN, and both compounds cause teratogenic phenotypes in chicken embryos. Thalidomide 14-25 cereblon Gallus gallus 124-128 33470442-8 2021 Most importantly, degradation of PLZF protein, but not of the known thalidomide-dependent CRBN substrate SALL4, was induced by thalidomide or 5-hydroxythalidomide treatment in chicken embryos. Thalidomide 127-138 zinc finger and BTB domain containing 16 Homo sapiens 33-37 33470442-9 2021 Furthermore, PLZF overexpression partially rescued the thalidomide-induced phenotypes. Thalidomide 55-66 zinc finger and BTB domain containing 16 Homo sapiens 13-17 33470442-10 2021 Our findings implicate PLZF as an important thalidomide-induced CRBN neosubstrate involved in thalidomide teratogenicity. Thalidomide 44-55 zinc finger and BTB domain containing 16 Homo sapiens 23-27 33470442-10 2021 Our findings implicate PLZF as an important thalidomide-induced CRBN neosubstrate involved in thalidomide teratogenicity. Thalidomide 44-55 cereblon Gallus gallus 64-68 33470442-10 2021 Our findings implicate PLZF as an important thalidomide-induced CRBN neosubstrate involved in thalidomide teratogenicity. Thalidomide 94-105 zinc finger and BTB domain containing 16 Homo sapiens 23-27 33470442-10 2021 Our findings implicate PLZF as an important thalidomide-induced CRBN neosubstrate involved in thalidomide teratogenicity. Thalidomide 94-105 cereblon Gallus gallus 64-68 33015978-13 2021 Overexpression of HDAC6 blocked the protection of thalidomide against PQ-induced injury via activating the IkBalpha-NF-kappaB signalling pathway. Thalidomide 50-61 histone deacetylase 6 Rattus norvegicus 18-23 33015978-14 2021 Collectively, this study evidenced that thalidomide protects lung tissues from PQ-induced injury through a miR-141/HDAC6/IkBalpha-NF-kappaB axis. Thalidomide 40-51 microRNA 141 Rattus norvegicus 107-114 33015978-14 2021 Collectively, this study evidenced that thalidomide protects lung tissues from PQ-induced injury through a miR-141/HDAC6/IkBalpha-NF-kappaB axis. Thalidomide 40-51 histone deacetylase 6 Rattus norvegicus 115-120 33333254-7 2021 The mean reduction rates at 7-10 days after treatment for serum interleukin-6 and interferon-gamma concentrations were greater in the thalidomide group compared to the controls. Thalidomide 134-145 interleukin 6 Homo sapiens 64-77 33333254-7 2021 The mean reduction rates at 7-10 days after treatment for serum interleukin-6 and interferon-gamma concentrations were greater in the thalidomide group compared to the controls. Thalidomide 134-145 interferon gamma Homo sapiens 82-98 33416146-2 2021 The expression level of DEPTOR is closely related to the prognosis of patients with MM treated with the antiangiogenic agent thalidomide; however, its role in the regulation of angiogenesis has not yet been elucidated. Thalidomide 125-136 DEP domain containing MTOR interacting protein Homo sapiens 24-30 32008086-0 2020 Thalidomide targets EGFL6 to inhibit EGFL6/PAX6 axis-driven angiogenesis in small bowel vascular malformation. Thalidomide 0-11 EGF like domain multiple 6 Homo sapiens 20-25 33127336-8 2021 An examination of the contribution of Tnfa in allograft response revealed that it was prospectively antagonized by etanercept or thalidomide, which resolved cytokine, chemokine, and receptor cascades. Thalidomide 129-140 tumor necrosis factor Rattus norvegicus 38-42 33310190-1 2021 Cereblon (CRBN), the substrate receptor of an E3 ubiquitin ligase complex, is a target of thalidomide and thalidomide-derived immunomodulatory drugs (IMiDs). Thalidomide 90-101 cereblon Homo sapiens 0-8 33310190-1 2021 Cereblon (CRBN), the substrate receptor of an E3 ubiquitin ligase complex, is a target of thalidomide and thalidomide-derived immunomodulatory drugs (IMiDs). Thalidomide 90-101 cereblon Homo sapiens 10-14 33310190-1 2021 Cereblon (CRBN), the substrate receptor of an E3 ubiquitin ligase complex, is a target of thalidomide and thalidomide-derived immunomodulatory drugs (IMiDs). Thalidomide 106-117 cereblon Homo sapiens 0-8 33310190-1 2021 Cereblon (CRBN), the substrate receptor of an E3 ubiquitin ligase complex, is a target of thalidomide and thalidomide-derived immunomodulatory drugs (IMiDs). Thalidomide 106-117 cereblon Homo sapiens 10-14 33745380-7 2021 Pharmacological inhibition of PI3K or JNK, using thalidomide, quercetin, or SP600125, attenuated the development of morphine tolerance in mice with SNL as measured by thermal paw withdrawal. Thalidomide 49-60 mitogen-activated protein kinase 8 Mus musculus 38-41 33317522-0 2020 Oxidative stress mediates thalidomide-induced pain by targeting peripheral TRPA1 and central TRPV4. Thalidomide 26-37 transient receptor potential cation channel subfamily A member 1 Homo sapiens 75-80 33317522-0 2020 Oxidative stress mediates thalidomide-induced pain by targeting peripheral TRPA1 and central TRPV4. Thalidomide 26-37 transient receptor potential cation channel subfamily V member 4 Homo sapiens 93-98 33317522-11 2020 CONCLUSIONS: Targeting of peripheral TRPA1 and central TRPV4 may be required to attenuate pain associated with CIPN elicited by thalidomide and related drugs. Thalidomide 128-139 transient receptor potential cation channel subfamily A member 1 Homo sapiens 37-42 33317522-11 2020 CONCLUSIONS: Targeting of peripheral TRPA1 and central TRPV4 may be required to attenuate pain associated with CIPN elicited by thalidomide and related drugs. Thalidomide 128-139 transient receptor potential cation channel subfamily V member 4 Homo sapiens 55-60 33527793-8 2021 Corresponding to increased CTLA-4 expression in T cells, the expression of ligand molecules for CTLA-4 significantly increased in splenic dendritic cells in TM/DX-treated groups. Thalidomide 157-159 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 27-33 33527793-8 2021 Corresponding to increased CTLA-4 expression in T cells, the expression of ligand molecules for CTLA-4 significantly increased in splenic dendritic cells in TM/DX-treated groups. Thalidomide 157-159 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 96-102 32979866-3 2021 However, this may not be the sole mechanism involved since the blockade of TNF-alpha synthesis by thalidomide prevents hyperalgesia without interrupting the synthesis of IL-1beta, IL-6 and CINC-1. Thalidomide 98-109 tumor necrosis factor Rattus norvegicus 75-84 33275735-2 2020 Currently, there are 3 CD38 antibody-based regimens approved for the treatment of both transplant-eligible (daratumumab plus bortezomib-thalidomide-dexamethasone [D-VTd]) and transplant-ineligible (daratumumab plus lenalidomide-dexamethasone [D-Rd] or daratumumab plus bortezomib-melphalan-prednisone [D-VMP]) patients with newly diagnosed MM (NDMM). Thalidomide 136-147 CD38 molecule Homo sapiens 23-27 32008086-0 2020 Thalidomide targets EGFL6 to inhibit EGFL6/PAX6 axis-driven angiogenesis in small bowel vascular malformation. Thalidomide 0-11 EGF like domain multiple 6 Homo sapiens 37-42 32008086-0 2020 Thalidomide targets EGFL6 to inhibit EGFL6/PAX6 axis-driven angiogenesis in small bowel vascular malformation. Thalidomide 0-11 paired box 6 Homo sapiens 43-47 32008086-8 2020 Furthermore, through in vitro and in vivo assays, we elucidated that thalidomide can function as anti-angiogenesis medicine through the regulation of EGFL6 in a proteasome-dependent manner. Thalidomide 69-80 EGF like domain multiple 6 Homo sapiens 150-155 32008086-9 2020 Finally, we found that CRBN can mediate the effect of thalidomide on EGFL6 expression and that the CRBN protein interacts with EGFL6 via a Lon N-terminal peptide. Thalidomide 54-65 cereblon Homo sapiens 23-27 32008086-9 2020 Finally, we found that CRBN can mediate the effect of thalidomide on EGFL6 expression and that the CRBN protein interacts with EGFL6 via a Lon N-terminal peptide. Thalidomide 54-65 EGF like domain multiple 6 Homo sapiens 69-74 32008086-9 2020 Finally, we found that CRBN can mediate the effect of thalidomide on EGFL6 expression and that the CRBN protein interacts with EGFL6 via a Lon N-terminal peptide. Thalidomide 54-65 EGF like domain multiple 6 Homo sapiens 127-132 32008086-10 2020 CONCLUSION: Our findings revealed a key role for EGFL6 in SBVM pathogenesis and provided a mechanism explaining why thalidomide can cure small bowel bleeding resulting from SBVM. Thalidomide 116-127 EGF like domain multiple 6 Homo sapiens 49-54 32865967-1 2020 Cereblon (CRBN) is an E3 ligase adapter protein that can be reprogrammed by imide-class compounds such as thalidomide, lenalidomide, and pomalidomide to induce the degradation of neo-substrate proteins. Thalidomide 106-117 cereblon Homo sapiens 0-8 32778513-12 2020 Splicing mutations were common in both cohorts, though thalidomide-treated patients were more likely to have a high-risk SRSF2 or U2AF1 Q157 mutation (P = .01). Thalidomide 55-66 serine and arginine rich splicing factor 2 Homo sapiens 121-126 32778513-12 2020 Splicing mutations were common in both cohorts, though thalidomide-treated patients were more likely to have a high-risk SRSF2 or U2AF1 Q157 mutation (P = .01). Thalidomide 55-66 U2 small nuclear RNA auxiliary factor 1 Homo sapiens 130-135 33070611-7 2020 Our cereblon TR-FRET binding assay was very stable and detected changes in phthalimide activity due to thalidomide isomerization. Thalidomide 103-114 cereblon Homo sapiens 4-12 32865967-1 2020 Cereblon (CRBN) is an E3 ligase adapter protein that can be reprogrammed by imide-class compounds such as thalidomide, lenalidomide, and pomalidomide to induce the degradation of neo-substrate proteins. Thalidomide 106-117 cereblon Homo sapiens 10-14 32729462-1 2020 None: Recent developments in the genetics of restless legs syndrome (RLS) revealed associations of disease risk with genetic loci containing the genes coding cereblon, the protein bound by thalidomide, and its endogenous substrate MEIS2 whose degradation is inhibited by the thalidomide-cereblon interaction. Thalidomide 275-286 Meis homeobox 2 Homo sapiens 231-236 32848052-1 2020 Thalidomide exerts its teratogenic and immunomodulatory effects by binding to cereblon (CRBN) and thereby inhibiting/modifying the CRBN-mediated ubiquitination pathway consisting of the Cullin4-DDB1-ROC1 E3 ligase complex. Thalidomide 0-11 cereblon Mus musculus 88-92 33008427-5 2020 METHODS: MUK eight is a randomised, controlled, open, parallel group, multi-centre phase II trial that will recruit patients with RRMM who have relapsed after treatment with thalidomide, lenalidomide, and a proteasome inhibitor. Thalidomide 174-185 mitogen-activated protein kinase kinase kinase 12 Homo sapiens 9-12 32800337-5 2020 Immunofluorescence analysis showed decrease of VEGFA expression in both ARPE-19 cells and HUVECs after treatment only with thalidomide but not with senicapoc or sodium butyrate. Thalidomide 123-134 vascular endothelial growth factor A Homo sapiens 47-52 32800337-9 2020 Thalidomide also reduced cobalt chloride induced increase of VEGFA mRNA in ARPE-19 (-33%) and protein in culture medium (-20%). Thalidomide 0-11 vascular endothelial growth factor A Mus musculus 61-66 32848052-1 2020 Thalidomide exerts its teratogenic and immunomodulatory effects by binding to cereblon (CRBN) and thereby inhibiting/modifying the CRBN-mediated ubiquitination pathway consisting of the Cullin4-DDB1-ROC1 E3 ligase complex. Thalidomide 0-11 cereblon Mus musculus 131-135 32929090-2 2020 Here, we report the structural bases for the SALL4-specific proteasomal degradation induced by 5-hydroxythalidomide, a primary thalidomide metabolite generated by the enzymatic activity of cytochrome P450 isozymes, through the interaction with cereblon (CRBN). Thalidomide 104-115 spalt like transcription factor 4 Homo sapiens 45-50 32848052-1 2020 Thalidomide exerts its teratogenic and immunomodulatory effects by binding to cereblon (CRBN) and thereby inhibiting/modifying the CRBN-mediated ubiquitination pathway consisting of the Cullin4-DDB1-ROC1 E3 ligase complex. Thalidomide 0-11 damage specific DNA binding protein 1 Mus musculus 194-198 32848052-1 2020 Thalidomide exerts its teratogenic and immunomodulatory effects by binding to cereblon (CRBN) and thereby inhibiting/modifying the CRBN-mediated ubiquitination pathway consisting of the Cullin4-DDB1-ROC1 E3 ligase complex. Thalidomide 0-11 ring-box 1 Mus musculus 199-203 32848052-3 2020 Here we examined whether CRBN is involved in the hypnotic effect of thalidomide by generating mice harboring a thalidomide-resistant mutant allele of Crbn (Crbn YW/AA knock-in mice). Thalidomide 68-79 cereblon Mus musculus 25-29 32848052-4 2020 Thalidomide increased non-REM sleep time in Crbn YW/AA knock-in homozygotes and heterozygotes to a similar degree as seen in wild-type littermates. Thalidomide 0-11 cereblon Mus musculus 44-48 32848052-5 2020 Thalidomide similarly depressed excitatory synaptic transmission in the cortical slices obtained from wild-type and Crbn YW/AA homozygous knock-in mice without affecting GABAergic inhibition. Thalidomide 0-11 cereblon Mus musculus 116-120 32848052-6 2020 Thalidomide induced Fos expression in vasopressin-containing neurons of the supraoptic nucleus and reduced Fos expression in the tuberomammillary nuclei. Thalidomide 0-11 FBJ osteosarcoma oncogene Mus musculus 20-23 32848052-6 2020 Thalidomide induced Fos expression in vasopressin-containing neurons of the supraoptic nucleus and reduced Fos expression in the tuberomammillary nuclei. Thalidomide 0-11 FBJ osteosarcoma oncogene Mus musculus 107-110 32929090-1 2020 Thalidomide and its derivatives exert not only therapeutic effects as immunomodulatory drugs (IMiDs) but also adverse effects such as teratogenicity, which are due in part to different C2H2 zinc-finger (ZF) transcription factors, IKZF1 (or IKZF3) and SALL4, respectively. Thalidomide 0-11 IKAROS family zinc finger 1 Homo sapiens 230-235 32929090-1 2020 Thalidomide and its derivatives exert not only therapeutic effects as immunomodulatory drugs (IMiDs) but also adverse effects such as teratogenicity, which are due in part to different C2H2 zinc-finger (ZF) transcription factors, IKZF1 (or IKZF3) and SALL4, respectively. Thalidomide 0-11 IKAROS family zinc finger 3 Homo sapiens 240-245 32929090-1 2020 Thalidomide and its derivatives exert not only therapeutic effects as immunomodulatory drugs (IMiDs) but also adverse effects such as teratogenicity, which are due in part to different C2H2 zinc-finger (ZF) transcription factors, IKZF1 (or IKZF3) and SALL4, respectively. Thalidomide 0-11 spalt like transcription factor 4 Homo sapiens 251-256 32929090-2 2020 Here, we report the structural bases for the SALL4-specific proteasomal degradation induced by 5-hydroxythalidomide, a primary thalidomide metabolite generated by the enzymatic activity of cytochrome P450 isozymes, through the interaction with cereblon (CRBN). Thalidomide 104-115 cereblon Homo sapiens 254-258 32367505-7 2020 In contrast to this, Snap25 expression, studied in males, was increased in the hippocampus and cerebellum in both, VPA- and THAL-treated rats. Thalidomide 124-128 synaptosome associated protein 25 Rattus norvegicus 21-27 32387854-0 2020 The association of HBG2, BCL11A, and HBS1L-MYB polymorphisms to thalidomide response in Chinese beta-thalassemia patients. Thalidomide 64-75 hemoglobin subunit gamma 2 Homo sapiens 19-23 32387854-0 2020 The association of HBG2, BCL11A, and HBS1L-MYB polymorphisms to thalidomide response in Chinese beta-thalassemia patients. Thalidomide 64-75 HBS1 like translational GTPase Homo sapiens 37-42 32387854-0 2020 The association of HBG2, BCL11A, and HBS1L-MYB polymorphisms to thalidomide response in Chinese beta-thalassemia patients. Thalidomide 64-75 MYB proto-oncogene, transcription factor Homo sapiens 43-46 32387854-10 2020 It was demonstrated that SNPs in HBG2 and HBS1L-MYB contributed significantly to thalidomide response in Chinese patients with beta-thalassemia and that the cumulative number of minor SNP alleles may serve as good predictors of treatment response in this population. Thalidomide 81-92 hemoglobin subunit gamma 2 Homo sapiens 33-37 32387854-10 2020 It was demonstrated that SNPs in HBG2 and HBS1L-MYB contributed significantly to thalidomide response in Chinese patients with beta-thalassemia and that the cumulative number of minor SNP alleles may serve as good predictors of treatment response in this population. Thalidomide 81-92 HBS1 like translational GTPase Homo sapiens 42-47 32387854-10 2020 It was demonstrated that SNPs in HBG2 and HBS1L-MYB contributed significantly to thalidomide response in Chinese patients with beta-thalassemia and that the cumulative number of minor SNP alleles may serve as good predictors of treatment response in this population. Thalidomide 81-92 MYB proto-oncogene, transcription factor Homo sapiens 48-51 32634680-2 2020 These proteolysis targeted chimeras (PROTACs) are based on an ATP competitive inhibitor of p38alpha and p38beta, which is linked to thalidomide analogues to recruit the Cereblon E3 ubiquitin ligase complex. Thalidomide 132-143 mitogen-activated protein kinase 14 Homo sapiens 91-99 32634680-2 2020 These proteolysis targeted chimeras (PROTACs) are based on an ATP competitive inhibitor of p38alpha and p38beta, which is linked to thalidomide analogues to recruit the Cereblon E3 ubiquitin ligase complex. Thalidomide 132-143 mitogen-activated protein kinase 11 Homo sapiens 104-111 32935774-1 2020 INTRODUCTION: Thalidomide is an anti- tumor necrosis factor alpha (TNF-a) drug used mainly in the management of moderate to severe form of Erythema Nodosum Leprosum (ENL). Thalidomide 14-25 tumor necrosis factor Homo sapiens 38-65 32935774-1 2020 INTRODUCTION: Thalidomide is an anti- tumor necrosis factor alpha (TNF-a) drug used mainly in the management of moderate to severe form of Erythema Nodosum Leprosum (ENL). Thalidomide 14-25 tumor necrosis factor Homo sapiens 67-72 32819116-0 2020 Thalidomide inhibits the gene promoter of connective tissue growth factor in human embryonic lung fibroblasts. Thalidomide 0-11 cellular communication network factor 2 Homo sapiens 42-73 32819116-2 2020 It"s necessary to investigate the effect of thalidomide on the gene promoter activation of connective tissue growth factor (CTGF) induced by transforming growth factor-beta1 (TGF-beta1) in human embryonic lung fibroblast (HELF). Thalidomide 44-55 cellular communication network factor 2 Homo sapiens 91-122 32819116-2 2020 It"s necessary to investigate the effect of thalidomide on the gene promoter activation of connective tissue growth factor (CTGF) induced by transforming growth factor-beta1 (TGF-beta1) in human embryonic lung fibroblast (HELF). Thalidomide 44-55 cellular communication network factor 2 Homo sapiens 124-128 32819116-2 2020 It"s necessary to investigate the effect of thalidomide on the gene promoter activation of connective tissue growth factor (CTGF) induced by transforming growth factor-beta1 (TGF-beta1) in human embryonic lung fibroblast (HELF). Thalidomide 44-55 transforming growth factor beta 1 Homo sapiens 141-173 32819116-2 2020 It"s necessary to investigate the effect of thalidomide on the gene promoter activation of connective tissue growth factor (CTGF) induced by transforming growth factor-beta1 (TGF-beta1) in human embryonic lung fibroblast (HELF). Thalidomide 44-55 transforming growth factor beta 1 Homo sapiens 175-184 32819116-5 2020 The activity of luciferase was measured to observe the effect of TGF-beta1 and THALIDOMIDE on the activity of CTGF gene promoter. Thalidomide 79-90 cellular communication network factor 2 Homo sapiens 110-114 32819116-7 2020 The thalidomide inhibited the TGF-beta1-induced activation of CTGF gene promoter in HELF, and the effect peaked in the 25 microg/L group (all P<0.001). Thalidomide 4-15 transforming growth factor beta 1 Homo sapiens 30-39 32819116-7 2020 The thalidomide inhibited the TGF-beta1-induced activation of CTGF gene promoter in HELF, and the effect peaked in the 25 microg/L group (all P<0.001). Thalidomide 4-15 cellular communication network factor 2 Homo sapiens 62-66 32819116-8 2020 CONCLUSIONS: Thalidomide produces a significant inhibitory effect on the gene promoter activation of CTGF induced by TGF-beta1 in HELF, it may be a potentially effective drug for the treatment of pulmonary fibrosis. Thalidomide 13-24 cellular communication network factor 2 Homo sapiens 101-105 32819116-8 2020 CONCLUSIONS: Thalidomide produces a significant inhibitory effect on the gene promoter activation of CTGF induced by TGF-beta1 in HELF, it may be a potentially effective drug for the treatment of pulmonary fibrosis. Thalidomide 13-24 transforming growth factor beta 1 Homo sapiens 117-126 32268442-1 2020 The use of dexamethasone plus thalidomide in combination with a continuous infusion of cisplatin, doxorubicin, cyclophosphamide, and etoposide (DTPACE) was first described in relapsed myeloma (MM) and showed effectiveness as induction therapy before autologous stem cell transplant (ASCT)1 . Thalidomide 30-41 solute carrier family 1 member 4 Homo sapiens 283-289 32908940-11 2020 Tha 200 mug/ml suppressed iNOS and TNF-alpha, and promoted CD206 and Arg-1 expressions significantly compared to the HG group (p < 0.05). Thalidomide 0-3 nitric oxide synthase 2, inducible Mus musculus 26-30 32908940-11 2020 Tha 200 mug/ml suppressed iNOS and TNF-alpha, and promoted CD206 and Arg-1 expressions significantly compared to the HG group (p < 0.05). Thalidomide 0-3 tumor necrosis factor Mus musculus 35-44 32908940-11 2020 Tha 200 mug/ml suppressed iNOS and TNF-alpha, and promoted CD206 and Arg-1 expressions significantly compared to the HG group (p < 0.05). Thalidomide 0-3 mannose receptor, C type 1 Mus musculus 59-64 32908940-11 2020 Tha 200 mug/ml suppressed iNOS and TNF-alpha, and promoted CD206 and Arg-1 expressions significantly compared to the HG group (p < 0.05). Thalidomide 0-3 arginase, liver Mus musculus 69-74 32908940-13 2020 The (Tha 200 mug/ml) MS group exhibited a decrease in TNF-alpha and IL-1beta level, and an upregulation in nephrin and podocin expressions significantly compared to the (HG) MS group (p < 0.05). Thalidomide 5-8 tumor necrosis factor Mus musculus 54-63 32908940-13 2020 The (Tha 200 mug/ml) MS group exhibited a decrease in TNF-alpha and IL-1beta level, and an upregulation in nephrin and podocin expressions significantly compared to the (HG) MS group (p < 0.05). Thalidomide 5-8 interleukin 1 alpha Rattus norvegicus 68-76 32908940-13 2020 The (Tha 200 mug/ml) MS group exhibited a decrease in TNF-alpha and IL-1beta level, and an upregulation in nephrin and podocin expressions significantly compared to the (HG) MS group (p < 0.05). Thalidomide 5-8 NPHS1 adhesion molecule, nephrin Rattus norvegicus 107-114 32908940-13 2020 The (Tha 200 mug/ml) MS group exhibited a decrease in TNF-alpha and IL-1beta level, and an upregulation in nephrin and podocin expressions significantly compared to the (HG) MS group (p < 0.05). Thalidomide 5-8 NPHS2 stomatin family member, podocin Rattus norvegicus 119-126 32908940-14 2020 Our research confirmed that HG-activated macrophage differentiation aggravates HG-induced podocyte injury in vitro and the protective effects of Tha might be related to its actions on TNF-alpha and IL-1beta levels via its modulation on M1/M2 differentiation. Thalidomide 145-148 tumor necrosis factor Mus musculus 184-193 32908940-14 2020 Our research confirmed that HG-activated macrophage differentiation aggravates HG-induced podocyte injury in vitro and the protective effects of Tha might be related to its actions on TNF-alpha and IL-1beta levels via its modulation on M1/M2 differentiation. Thalidomide 145-148 interleukin 1 alpha Rattus norvegicus 198-206 32573072-5 2020 To exert the anti-metastatic effects of thalidomide, both IFN-g production and direct cytotoxicity of NK cells were essential, without notable contribution from T cells. Thalidomide 40-51 interferon gamma Mus musculus 58-63 32573072-6 2020 In thalidomide-treated mice, there was a significant increase in the terminally differentiated mature CD27lo NK cells in the peripheral tissues and NK cells in thalidomide-treated mice exhibited significantly higher cytotoxicity and IFN-g production. Thalidomide 3-14 CD27 antigen Mus musculus 102-106 32573072-6 2020 In thalidomide-treated mice, there was a significant increase in the terminally differentiated mature CD27lo NK cells in the peripheral tissues and NK cells in thalidomide-treated mice exhibited significantly higher cytotoxicity and IFN-g production. Thalidomide 3-14 interferon gamma Mus musculus 233-238 32573072-6 2020 In thalidomide-treated mice, there was a significant increase in the terminally differentiated mature CD27lo NK cells in the peripheral tissues and NK cells in thalidomide-treated mice exhibited significantly higher cytotoxicity and IFN-g production. Thalidomide 160-171 interferon gamma Mus musculus 233-238 32573072-7 2020 The NK cell expression of T-bet was up-regulated by thalidomide treatment and the down-regulation of glycogen synthase kinase 3b expression was observed in thalidomide-treated NK cells. Thalidomide 52-63 T-box 21 Mus musculus 26-31 32573072-7 2020 The NK cell expression of T-bet was up-regulated by thalidomide treatment and the down-regulation of glycogen synthase kinase 3b expression was observed in thalidomide-treated NK cells. Thalidomide 156-167 T-box 21 Mus musculus 26-31 32573072-7 2020 The NK cell expression of T-bet was up-regulated by thalidomide treatment and the down-regulation of glycogen synthase kinase 3b expression was observed in thalidomide-treated NK cells. Thalidomide 156-167 glycogen synthase kinase 3 beta Mus musculus 101-128 32573072-8 2020 Collectively, our study suggests that thalidomide induces the functional maturation of peripheral NK cells through alteration of T-bet expression to inhibit lung metastasis of cancer cells. Thalidomide 38-49 T-box 21 Mus musculus 129-134 32447240-16 2020 NO synthase (NOS) inhibitors significantly augmented the anticonvulsant effects of combined low doses of thalidomide and morphine, whereas the inducible NOS inhibitor aminoguanidine (100 mg/kg, i.p.) Thalidomide 105-116 nitric oxide synthase 1, neuronal Mus musculus 0-11 32766176-6 2020 Analytical testing (immunohistochemistry, western blot, and qRT-PCR) shows that IL-6, IL-17, IL-23, and RORgammat protein and mRNA expression levels were significantly reduced by thalidomide (p < 0.05 for all) and that these levels were significantly lower in the high-dose thalidomide group than in the low-dose thalidomide group (p < 0.05 for all). Thalidomide 179-190 interleukin 6 Rattus norvegicus 80-84 32407871-6 2020 In addition, thalidomide treatment also regulated cytokine production by inhibiting the production of angiogenesis-related cytokines, such as G-CSF, VEGF-B, VEGFR1, VEGFR3 and IL-10. Thalidomide 13-24 colony stimulating factor 3 (granulocyte) Mus musculus 142-147 32407871-6 2020 In addition, thalidomide treatment also regulated cytokine production by inhibiting the production of angiogenesis-related cytokines, such as G-CSF, VEGF-B, VEGFR1, VEGFR3 and IL-10. Thalidomide 13-24 vascular endothelial growth factor B Mus musculus 149-155 32407871-6 2020 In addition, thalidomide treatment also regulated cytokine production by inhibiting the production of angiogenesis-related cytokines, such as G-CSF, VEGF-B, VEGFR1, VEGFR3 and IL-10. Thalidomide 13-24 FMS-like tyrosine kinase 1 Mus musculus 157-163 32407871-6 2020 In addition, thalidomide treatment also regulated cytokine production by inhibiting the production of angiogenesis-related cytokines, such as G-CSF, VEGF-B, VEGFR1, VEGFR3 and IL-10. Thalidomide 13-24 FMS-like tyrosine kinase 4 Mus musculus 165-171 32407871-6 2020 In addition, thalidomide treatment also regulated cytokine production by inhibiting the production of angiogenesis-related cytokines, such as G-CSF, VEGF-B, VEGFR1, VEGFR3 and IL-10. Thalidomide 13-24 interleukin 10 Mus musculus 176-181 32622642-0 2020 Facile skin targeting of a thalidomide analog containing benzyl chloride moiety alleviates experimental psoriasis via the suppression of MAPK/NF-kappaB/AP-1 phosphorylation in keratinocytes. Thalidomide 27-38 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 142-151 32622642-0 2020 Facile skin targeting of a thalidomide analog containing benzyl chloride moiety alleviates experimental psoriasis via the suppression of MAPK/NF-kappaB/AP-1 phosphorylation in keratinocytes. Thalidomide 27-38 jun proto-oncogene Mus musculus 152-156 32622642-1 2020 BACKGROUND: Thalidomide can be a TNF-alpha inhibitor for treating skin inflammation. Thalidomide 12-23 tumor necrosis factor Mus musculus 33-42 32801749-9 2020 It follows that PD-1 antibody, chidamide, etoposide, and thalidomide (PCET) regimen may be a promising choice for patients with R/R ENKTL and warrants further investigation. Thalidomide 57-68 programmed cell death 1 Homo sapiens 16-20 32766176-6 2020 Analytical testing (immunohistochemistry, western blot, and qRT-PCR) shows that IL-6, IL-17, IL-23, and RORgammat protein and mRNA expression levels were significantly reduced by thalidomide (p < 0.05 for all) and that these levels were significantly lower in the high-dose thalidomide group than in the low-dose thalidomide group (p < 0.05 for all). Thalidomide 179-190 interleukin 17A Rattus norvegicus 86-91 32035188-9 2020 Both early thalidomide treatments reduced NF-kappaB activation and shifted the ED50 value for ERC and late radiation sequelae (LRS) to higher doses. Thalidomide 11-22 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 42-51 32304852-4 2020 Thalidomide analogues were recently found to hijack CRL4CRBN ubiquitin ligase to target a number of cellular proteins for ubiquitination and proteasomal degradation. Thalidomide 0-11 interleukin 17 receptor B Homo sapiens 52-56 32304852-5 2020 Thalidomide-mediated degradation of SALL4 and p63, transcription factors essential for embryonic development, very likely plays a critical role in thalidomide embryopathy. Thalidomide 0-11 spalt like transcription factor 4 Homo sapiens 36-41 32304852-5 2020 Thalidomide-mediated degradation of SALL4 and p63, transcription factors essential for embryonic development, very likely plays a critical role in thalidomide embryopathy. Thalidomide 0-11 tumor protein p63 Homo sapiens 46-49 32304852-5 2020 Thalidomide-mediated degradation of SALL4 and p63, transcription factors essential for embryonic development, very likely plays a critical role in thalidomide embryopathy. Thalidomide 147-158 spalt like transcription factor 4 Homo sapiens 36-41 32304852-5 2020 Thalidomide-mediated degradation of SALL4 and p63, transcription factors essential for embryonic development, very likely plays a critical role in thalidomide embryopathy. Thalidomide 147-158 tumor protein p63 Homo sapiens 46-49 32172013-6 2020 Response to thalidomide was better in patients with POEMS-CD than those with POEMS alone (90% vs 43% clinical response, [p = .012]; 80% vs 45% normalization of serum VEGF levels, [p = .079]). Thalidomide 12-23 vascular endothelial growth factor A Homo sapiens 166-170 31721534-13 2020 Conclusion: Combined treatment with ATO and THAL can inhibit proliferation and invasion of AML cells by down-regulating ULK1 and BECLIN1 and up-regulating PTEN and IL6, and this effect was more marked than the effects of ATO and THAL alone. Thalidomide 44-48 unc-51 like autophagy activating kinase 1 Homo sapiens 120-124 31721534-13 2020 Conclusion: Combined treatment with ATO and THAL can inhibit proliferation and invasion of AML cells by down-regulating ULK1 and BECLIN1 and up-regulating PTEN and IL6, and this effect was more marked than the effects of ATO and THAL alone. Thalidomide 44-48 beclin 1 Homo sapiens 129-136 31721534-13 2020 Conclusion: Combined treatment with ATO and THAL can inhibit proliferation and invasion of AML cells by down-regulating ULK1 and BECLIN1 and up-regulating PTEN and IL6, and this effect was more marked than the effects of ATO and THAL alone. Thalidomide 44-48 phosphatase and tensin homolog Homo sapiens 155-159 31721534-13 2020 Conclusion: Combined treatment with ATO and THAL can inhibit proliferation and invasion of AML cells by down-regulating ULK1 and BECLIN1 and up-regulating PTEN and IL6, and this effect was more marked than the effects of ATO and THAL alone. Thalidomide 44-48 interleukin 6 Homo sapiens 164-167 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 26-30 chemokine (C-C motif) ligand 3 Mus musculus 147-151 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 26-30 interferon gamma Mus musculus 153-162 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 26-30 vascular endothelial growth factor A Mus musculus 167-171 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 26-30 interleukin 6 Mus musculus 205-209 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 26-30 interleukin 1 beta Mus musculus 211-219 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 26-30 interleukin 17A Mus musculus 221-226 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 26-30 transforming growth factor, beta 1 Mus musculus 232-241 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 39-43 chemokine (C-C motif) ligand 3 Mus musculus 147-151 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 39-43 interferon gamma Mus musculus 153-162 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 39-43 vascular endothelial growth factor A Mus musculus 167-171 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 39-43 interleukin 6 Mus musculus 205-209 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 39-43 interleukin 1 beta Mus musculus 211-219 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 39-43 interleukin 17A Mus musculus 221-226 31578565-8 2020 The mice treated with ITC/Thal and CMX/Thal showed intense weight loss, increased deposition of reticulin fibers, high pulmonary concentrations of CCL3, IFN-gamma and VEGF, and decreased concentrations of IL-6, IL-1beta, IL-17, and TGF-beta1. Thalidomide 39-43 transforming growth factor, beta 1 Mus musculus 232-241 32621419-8 2020 On the other hand, several CRL activators have been reported, such as plant auxin and immunomodulatory imide drugs, thalidomide. Thalidomide 116-127 interleukin 31 receptor A Homo sapiens 27-30 32223235-5 2020 In this study, we developed thalidomide-based heterobifunctional compounds that induced selective degradation of BRAF-V600E, but not the wild-type BRAF. Thalidomide 28-39 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 113-117 32443710-0 2020 Thalidomide Reduces Vascular Endothelial Growth Factor Immunostaining in Canine Splenic Hemangiosarcoma. Thalidomide 0-11 vascular endothelial growth factor A Canis lupus familiaris 20-54 32443710-2 2020 Recently it was demonstrated that thalidomide extends the survival time of dogs with HSA, potentially due to thalidomide-induced inhibition of vascular endothelial growth factor (VEGF) production by the neoplastic cells. Thalidomide 34-45 vascular endothelial growth factor A Canis lupus familiaris 143-177 32443710-2 2020 Recently it was demonstrated that thalidomide extends the survival time of dogs with HSA, potentially due to thalidomide-induced inhibition of vascular endothelial growth factor (VEGF) production by the neoplastic cells. Thalidomide 34-45 vascular endothelial growth factor A Canis lupus familiaris 179-183 32443710-2 2020 Recently it was demonstrated that thalidomide extends the survival time of dogs with HSA, potentially due to thalidomide-induced inhibition of vascular endothelial growth factor (VEGF) production by the neoplastic cells. Thalidomide 109-120 vascular endothelial growth factor A Canis lupus familiaris 143-177 32443710-2 2020 Recently it was demonstrated that thalidomide extends the survival time of dogs with HSA, potentially due to thalidomide-induced inhibition of vascular endothelial growth factor (VEGF) production by the neoplastic cells. Thalidomide 109-120 vascular endothelial growth factor A Canis lupus familiaris 179-183 32443710-3 2020 To investigate this, immunostaining was used to evaluate VEGF within HSA metastases that developed after thalidomide treatment. Thalidomide 105-116 vascular endothelial growth factor A Canis lupus familiaris 57-61 32443710-7 2020 This supports the hypothesis that thalidomide prolongs survival time in dogs with HSA due to inhibition of VEGF production by the neoplastic cells. Thalidomide 34-45 vascular endothelial growth factor A Canis lupus familiaris 107-111 32443710-8 2020 As VEGF remained visible within HSAs exposed to thalidomide, additional treatments to inhibit VEGF production may further prolong survival times of dogs with these common canine neoplasms. Thalidomide 48-59 vascular endothelial growth factor A Canis lupus familiaris 3-7 32443710-8 2020 As VEGF remained visible within HSAs exposed to thalidomide, additional treatments to inhibit VEGF production may further prolong survival times of dogs with these common canine neoplasms. Thalidomide 48-59 vascular endothelial growth factor A Canis lupus familiaris 94-98 32009091-1 2020 Objective A randomized controlled trial has shown the efficacy of thalidomide against Polyneuropathy, organomegaly, endocrinopathy, M-protein, and skin changes (POEMS) syndrome; however, there are still refractory patients. Thalidomide 66-77 myomesin 2 Homo sapiens 132-141 32414180-3 2020 The discovery of cereblon (CRBN), the direct target of thalidomide, a decade ago greatly improved our understanding of its mechanism of action. Thalidomide 55-66 cereblon Homo sapiens 27-31 32414180-4 2020 Accumulating evidence has shown that CRBN functions as a substrate of Cullin RING E3 ligase (CRL4CRBN), whose specificity is controlled by ligands such as thalidomide. Thalidomide 155-166 cereblon Homo sapiens 37-41 32414180-4 2020 Accumulating evidence has shown that CRBN functions as a substrate of Cullin RING E3 ligase (CRL4CRBN), whose specificity is controlled by ligands such as thalidomide. Thalidomide 155-166 CDK2 associated cullin domain 1 Homo sapiens 70-76 32414180-5 2020 For example, lenalidomide and pomalidomide, well-known thalidomide derivatives, degrade the neosubstrates Ikaros and Aiolos, resulting in anti-proliferative effects in multiple myeloma. Thalidomide 55-66 IKAROS family zinc finger 1 Homo sapiens 106-112 32414180-5 2020 For example, lenalidomide and pomalidomide, well-known thalidomide derivatives, degrade the neosubstrates Ikaros and Aiolos, resulting in anti-proliferative effects in multiple myeloma. Thalidomide 55-66 IKAROS family zinc finger 3 Homo sapiens 117-123 32414180-9 2020 Recently, several CRBN neosubstrates (e.g., SALL4 (Spalt Like Transcription Factor 4) and p63 (Tumor Protein P63)) have been identified as candidate mediators of thalidomide teratogenicity. Thalidomide 162-173 cereblon Homo sapiens 18-22 32414180-9 2020 Recently, several CRBN neosubstrates (e.g., SALL4 (Spalt Like Transcription Factor 4) and p63 (Tumor Protein P63)) have been identified as candidate mediators of thalidomide teratogenicity. Thalidomide 162-173 spalt like transcription factor 4 Homo sapiens 44-49 32414180-9 2020 Recently, several CRBN neosubstrates (e.g., SALL4 (Spalt Like Transcription Factor 4) and p63 (Tumor Protein P63)) have been identified as candidate mediators of thalidomide teratogenicity. Thalidomide 162-173 spalt like transcription factor 4 Homo sapiens 51-84 32414180-9 2020 Recently, several CRBN neosubstrates (e.g., SALL4 (Spalt Like Transcription Factor 4) and p63 (Tumor Protein P63)) have been identified as candidate mediators of thalidomide teratogenicity. Thalidomide 162-173 tumor protein p63 Homo sapiens 90-93 32414180-9 2020 Recently, several CRBN neosubstrates (e.g., SALL4 (Spalt Like Transcription Factor 4) and p63 (Tumor Protein P63)) have been identified as candidate mediators of thalidomide teratogenicity. Thalidomide 162-173 tumor protein p63 Homo sapiens 95-113 32223235-5 2020 In this study, we developed thalidomide-based heterobifunctional compounds that induced selective degradation of BRAF-V600E, but not the wild-type BRAF. Thalidomide 28-39 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 147-151 32018067-9 2020 Protein content, N-acetyl-b-glucosaminidase (NAG) and myeloperoxidase (MPO) activities were elevated in ocular tissues after disease induction, further decreasing post-treatment with intravitreal thalidomide. Thalidomide 196-207 N-acetyl-alpha-glucosaminidase Homo sapiens 17-43 32317922-7 2020 We then identified the autophosphorylation site T2524 as a so far not described 14-3-3 binding site at the very C-terminus of LRRK2. Thalidomide 48-53 leucine rich repeat kinase 2 Homo sapiens 126-131 32018067-9 2020 Protein content, N-acetyl-b-glucosaminidase (NAG) and myeloperoxidase (MPO) activities were elevated in ocular tissues after disease induction, further decreasing post-treatment with intravitreal thalidomide. Thalidomide 196-207 myeloperoxidase Homo sapiens 71-74 32018067-9 2020 Protein content, N-acetyl-b-glucosaminidase (NAG) and myeloperoxidase (MPO) activities were elevated in ocular tissues after disease induction, further decreasing post-treatment with intravitreal thalidomide. Thalidomide 196-207 N-acetyl-alpha-glucosaminidase Homo sapiens 45-48 32018067-9 2020 Protein content, N-acetyl-b-glucosaminidase (NAG) and myeloperoxidase (MPO) activities were elevated in ocular tissues after disease induction, further decreasing post-treatment with intravitreal thalidomide. Thalidomide 196-207 myeloperoxidase Homo sapiens 54-69 33133483-6 2020 Proteolysis targeting chimeras (PROTACs) structurally based on the cereblon (CRBN) ligand thalidomide have recently been described to degrade the targets CDK4/6. Thalidomide 90-101 cereblon Homo sapiens 77-81 32251415-1 2020 Thalidomide-dependent degradation of the embryonic transcription factor SALL4 by the CRL4CRBN E3 ubiquitin ligase is a plausible major driver of thalidomide teratogenicity. Thalidomide 0-11 spalt like transcription factor 4 Homo sapiens 72-77 32251415-1 2020 Thalidomide-dependent degradation of the embryonic transcription factor SALL4 by the CRL4CRBN E3 ubiquitin ligase is a plausible major driver of thalidomide teratogenicity. Thalidomide 145-156 spalt like transcription factor 4 Homo sapiens 72-77 33133483-6 2020 Proteolysis targeting chimeras (PROTACs) structurally based on the cereblon (CRBN) ligand thalidomide have recently been described to degrade the targets CDK4/6. Thalidomide 90-101 cyclin dependent kinase 4 Homo sapiens 154-160 31840939-7 2020 CONCLUSION: Thal can effectively prevent skin and pulmonary tissue fibrosis in a mouse model of SSc through the TGF-beta1/Smad3 signaling pathway and can rectify the distortion of the Th17/Treg balance in SSc by potentially regulating Th17 and Treg cell production, as well as their related factors expression. Thalidomide 12-16 transforming growth factor, beta 1 Mus musculus 112-121 32132601-7 2020 By contrast, the cytoplasmic translation factor GSPT1 was degraded following treatment with the thalidomide derivative CC-885 only when CRBN was present in the cytoplasm, indicating that subcellular distribution of CRBN is critical for the efficacy of thalidomide-based medications. Thalidomide 96-107 G1 to S phase transition 1 Homo sapiens 48-53 32132601-7 2020 By contrast, the cytoplasmic translation factor GSPT1 was degraded following treatment with the thalidomide derivative CC-885 only when CRBN was present in the cytoplasm, indicating that subcellular distribution of CRBN is critical for the efficacy of thalidomide-based medications. Thalidomide 96-107 cereblon Homo sapiens 136-140 32132601-7 2020 By contrast, the cytoplasmic translation factor GSPT1 was degraded following treatment with the thalidomide derivative CC-885 only when CRBN was present in the cytoplasm, indicating that subcellular distribution of CRBN is critical for the efficacy of thalidomide-based medications. Thalidomide 96-107 cereblon Homo sapiens 215-219 32132601-7 2020 By contrast, the cytoplasmic translation factor GSPT1 was degraded following treatment with the thalidomide derivative CC-885 only when CRBN was present in the cytoplasm, indicating that subcellular distribution of CRBN is critical for the efficacy of thalidomide-based medications. Thalidomide 252-263 G1 to S phase transition 1 Homo sapiens 48-53 32132601-7 2020 By contrast, the cytoplasmic translation factor GSPT1 was degraded following treatment with the thalidomide derivative CC-885 only when CRBN was present in the cytoplasm, indicating that subcellular distribution of CRBN is critical for the efficacy of thalidomide-based medications. Thalidomide 252-263 cereblon Homo sapiens 215-219 31840939-6 2020 RESULTS: Thal treatment: (a) reduced skin, and pulmonary tissue fibrosis, inflammation score, and hydroxyproline content (P < .001) in BLM-induced SSc mice; (b) reduced the percentages of Th17 cells and associated interleukin (IL)-17A expression (both P < .05) but increased the percentages of Treg cells and its transcription factor Foxp3 expression (both P < .05); (c) correlation analysis found positive correlations between Th17/Treg ratio, the inflammatory score of the skin and pulmonary tissues, hydroxyproline content, and type I collagen messenger RNA expression (r = .8546, .8656, .6902, .6807, .8118, and .8424, respectively, P < .01); (d) Thal inhibited TGF-beta1 expression and Smad3 phosphorylation (both P < .05); (e) TGF-beta1 was positively correlated with the IL-17A and Th17/Treg ratio (r = .5856, P = .005; r = .6684, P = .0107, respectively). Thalidomide 9-13 forkhead box P3 Mus musculus 340-345 31840939-7 2020 CONCLUSION: Thal can effectively prevent skin and pulmonary tissue fibrosis in a mouse model of SSc through the TGF-beta1/Smad3 signaling pathway and can rectify the distortion of the Th17/Treg balance in SSc by potentially regulating Th17 and Treg cell production, as well as their related factors expression. Thalidomide 12-16 SMAD family member 3 Mus musculus 122-127 31840939-6 2020 RESULTS: Thal treatment: (a) reduced skin, and pulmonary tissue fibrosis, inflammation score, and hydroxyproline content (P < .001) in BLM-induced SSc mice; (b) reduced the percentages of Th17 cells and associated interleukin (IL)-17A expression (both P < .05) but increased the percentages of Treg cells and its transcription factor Foxp3 expression (both P < .05); (c) correlation analysis found positive correlations between Th17/Treg ratio, the inflammatory score of the skin and pulmonary tissues, hydroxyproline content, and type I collagen messenger RNA expression (r = .8546, .8656, .6902, .6807, .8118, and .8424, respectively, P < .01); (d) Thal inhibited TGF-beta1 expression and Smad3 phosphorylation (both P < .05); (e) TGF-beta1 was positively correlated with the IL-17A and Th17/Treg ratio (r = .5856, P = .005; r = .6684, P = .0107, respectively). Thalidomide 9-13 transforming growth factor, beta 1 Mus musculus 678-687 31422568-10 2020 Hesperidin found stronger binding strength and IC50 was relative to thalidomide on TNF alpha, IL 6, and caspase 3. Thalidomide 68-79 interleukin 6 Rattus norvegicus 94-98 31840939-6 2020 RESULTS: Thal treatment: (a) reduced skin, and pulmonary tissue fibrosis, inflammation score, and hydroxyproline content (P < .001) in BLM-induced SSc mice; (b) reduced the percentages of Th17 cells and associated interleukin (IL)-17A expression (both P < .05) but increased the percentages of Treg cells and its transcription factor Foxp3 expression (both P < .05); (c) correlation analysis found positive correlations between Th17/Treg ratio, the inflammatory score of the skin and pulmonary tissues, hydroxyproline content, and type I collagen messenger RNA expression (r = .8546, .8656, .6902, .6807, .8118, and .8424, respectively, P < .01); (d) Thal inhibited TGF-beta1 expression and Smad3 phosphorylation (both P < .05); (e) TGF-beta1 was positively correlated with the IL-17A and Th17/Treg ratio (r = .5856, P = .005; r = .6684, P = .0107, respectively). Thalidomide 9-13 SMAD family member 3 Mus musculus 703-708 31840939-6 2020 RESULTS: Thal treatment: (a) reduced skin, and pulmonary tissue fibrosis, inflammation score, and hydroxyproline content (P < .001) in BLM-induced SSc mice; (b) reduced the percentages of Th17 cells and associated interleukin (IL)-17A expression (both P < .05) but increased the percentages of Treg cells and its transcription factor Foxp3 expression (both P < .05); (c) correlation analysis found positive correlations between Th17/Treg ratio, the inflammatory score of the skin and pulmonary tissues, hydroxyproline content, and type I collagen messenger RNA expression (r = .8546, .8656, .6902, .6807, .8118, and .8424, respectively, P < .01); (d) Thal inhibited TGF-beta1 expression and Smad3 phosphorylation (both P < .05); (e) TGF-beta1 was positively correlated with the IL-17A and Th17/Treg ratio (r = .5856, P = .005; r = .6684, P = .0107, respectively). Thalidomide 9-13 transforming growth factor, beta 1 Mus musculus 748-757 31840939-6 2020 RESULTS: Thal treatment: (a) reduced skin, and pulmonary tissue fibrosis, inflammation score, and hydroxyproline content (P < .001) in BLM-induced SSc mice; (b) reduced the percentages of Th17 cells and associated interleukin (IL)-17A expression (both P < .05) but increased the percentages of Treg cells and its transcription factor Foxp3 expression (both P < .05); (c) correlation analysis found positive correlations between Th17/Treg ratio, the inflammatory score of the skin and pulmonary tissues, hydroxyproline content, and type I collagen messenger RNA expression (r = .8546, .8656, .6902, .6807, .8118, and .8424, respectively, P < .01); (d) Thal inhibited TGF-beta1 expression and Smad3 phosphorylation (both P < .05); (e) TGF-beta1 was positively correlated with the IL-17A and Th17/Treg ratio (r = .5856, P = .005; r = .6684, P = .0107, respectively). Thalidomide 9-13 interleukin 17A Mus musculus 793-799 32071327-0 2020 Thalidomide Inhibits Human iPSC Mesendoderm Differentiation by Modulating CRBN-dependent Degradation of SALL4. Thalidomide 0-11 cereblon Homo sapiens 74-78 32071327-0 2020 Thalidomide Inhibits Human iPSC Mesendoderm Differentiation by Modulating CRBN-dependent Degradation of SALL4. Thalidomide 0-11 spalt like transcription factor 4 Homo sapiens 104-109 32071327-2 2020 Thalidomide-induced teratogenicity is dependent on its binding to cereblon (CRBN), the substrate receptor of the Cul4A-DDB1-CRBN-RBX1 E3 ubiquitin ligase complex. Thalidomide 0-11 cereblon Rattus norvegicus 76-80 32071327-2 2020 Thalidomide-induced teratogenicity is dependent on its binding to cereblon (CRBN), the substrate receptor of the Cul4A-DDB1-CRBN-RBX1 E3 ubiquitin ligase complex. Thalidomide 0-11 cullin 4A Homo sapiens 113-118 32071327-2 2020 Thalidomide-induced teratogenicity is dependent on its binding to cereblon (CRBN), the substrate receptor of the Cul4A-DDB1-CRBN-RBX1 E3 ubiquitin ligase complex. Thalidomide 0-11 damage specific DNA binding protein 1 Homo sapiens 119-123 32071327-2 2020 Thalidomide-induced teratogenicity is dependent on its binding to cereblon (CRBN), the substrate receptor of the Cul4A-DDB1-CRBN-RBX1 E3 ubiquitin ligase complex. Thalidomide 0-11 cereblon Rattus norvegicus 124-128 32071327-2 2020 Thalidomide-induced teratogenicity is dependent on its binding to cereblon (CRBN), the substrate receptor of the Cul4A-DDB1-CRBN-RBX1 E3 ubiquitin ligase complex. Thalidomide 0-11 ring-box 1 Homo sapiens 129-133 32071327-3 2020 Thalidomide binding to CRBN elicits subsequent ubiquitination and proteasomal degradation of CRBN neosubstrates including SALL4, a transcription factor of which polymorphisms phenocopy thalidomide-induced limb defects in humans. Thalidomide 0-11 cereblon Homo sapiens 23-27 32071327-3 2020 Thalidomide binding to CRBN elicits subsequent ubiquitination and proteasomal degradation of CRBN neosubstrates including SALL4, a transcription factor of which polymorphisms phenocopy thalidomide-induced limb defects in humans. Thalidomide 0-11 cereblon Homo sapiens 93-97 32071327-3 2020 Thalidomide binding to CRBN elicits subsequent ubiquitination and proteasomal degradation of CRBN neosubstrates including SALL4, a transcription factor of which polymorphisms phenocopy thalidomide-induced limb defects in humans. Thalidomide 0-11 spalt like transcription factor 4 Homo sapiens 122-127 32071327-3 2020 Thalidomide binding to CRBN elicits subsequent ubiquitination and proteasomal degradation of CRBN neosubstrates including SALL4, a transcription factor of which polymorphisms phenocopy thalidomide-induced limb defects in humans. Thalidomide 185-196 cereblon Homo sapiens 23-27 32071327-3 2020 Thalidomide binding to CRBN elicits subsequent ubiquitination and proteasomal degradation of CRBN neosubstrates including SALL4, a transcription factor of which polymorphisms phenocopy thalidomide-induced limb defects in humans. Thalidomide 185-196 cereblon Homo sapiens 93-97 32071327-3 2020 Thalidomide binding to CRBN elicits subsequent ubiquitination and proteasomal degradation of CRBN neosubstrates including SALL4, a transcription factor of which polymorphisms phenocopy thalidomide-induced limb defects in humans. Thalidomide 185-196 spalt like transcription factor 4 Homo sapiens 122-127 32071327-4 2020 Herein, thalidomide-induced degradation of SALL4 was examined in human induced pluripotent stem cells (hiPSCs) that were differentiated either to lateral plate mesoderm (LPM)-like cells, the developmental ontology of the limb bud, or definitive endoderm. Thalidomide 8-19 spalt like transcription factor 4 Homo sapiens 43-48 32071327-6 2020 Thalidomide- and IMiD-induced SALL4 degradation can be abrogated by CRBN V388I mutation or SALL4 G416A mutation in hiPSCs. Thalidomide 0-11 spalt like transcription factor 4 Homo sapiens 30-35 32071327-6 2020 Thalidomide- and IMiD-induced SALL4 degradation can be abrogated by CRBN V388I mutation or SALL4 G416A mutation in hiPSCs. Thalidomide 0-11 cereblon Homo sapiens 68-72 32071327-6 2020 Thalidomide- and IMiD-induced SALL4 degradation can be abrogated by CRBN V388I mutation or SALL4 G416A mutation in hiPSCs. Thalidomide 0-11 spalt like transcription factor 4 Homo sapiens 91-96 32071327-9 2020 The data here show that thalidomide, lenalidomide, and pomalidomide affect stem cell mesendoderm differentiation through CRBN-mediated degradation of SALL4 and highlight the utility of the LPM differentiation model for studying the teratogenicity of new CRBN modulating agents. Thalidomide 24-35 cereblon Homo sapiens 121-125 32071327-9 2020 The data here show that thalidomide, lenalidomide, and pomalidomide affect stem cell mesendoderm differentiation through CRBN-mediated degradation of SALL4 and highlight the utility of the LPM differentiation model for studying the teratogenicity of new CRBN modulating agents. Thalidomide 24-35 spalt like transcription factor 4 Homo sapiens 150-155 32071327-9 2020 The data here show that thalidomide, lenalidomide, and pomalidomide affect stem cell mesendoderm differentiation through CRBN-mediated degradation of SALL4 and highlight the utility of the LPM differentiation model for studying the teratogenicity of new CRBN modulating agents. Thalidomide 24-35 cereblon Homo sapiens 254-258 31392724-5 2020 In the LOT1 setting, from 2008 to 2015, there was a decrease in the use of thalidomide-based therapy, from 66 7% to 42 6%, and chemotherapy from, 20 2% to 5 9%, whereas use of bortezomib-based therapy or bortezomib + thalidomide increased from 10 7% to 45 5%. Thalidomide 75-86 PLAG1 like zinc finger 1 Homo sapiens 7-11 31392724-5 2020 In the LOT1 setting, from 2008 to 2015, there was a decrease in the use of thalidomide-based therapy, from 66 7% to 42 6%, and chemotherapy from, 20 2% to 5 9%, whereas use of bortezomib-based therapy or bortezomib + thalidomide increased from 10 7% to 45 5%. Thalidomide 217-228 PLAG1 like zinc finger 1 Homo sapiens 7-11 31620907-7 2020 In addition, the PDCD1LG1 rs4143815 CC and CG types (high-expression types) were associated significantly with higher frequency of patients who were treated with thalidomide and/or bortezomib. Thalidomide 162-173 CD274 molecule Homo sapiens 17-25 31916702-1 2020 OBJECTIVE: To explore the relationship between hepatic cytochrome P450 2C19 (CYP2C19) gene polymorphisms and the effectiveness and safety of thalidomide in the treatment of patients with immune-related bowel disease (IRBD). Thalidomide 141-152 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 55-75 31916702-1 2020 OBJECTIVE: To explore the relationship between hepatic cytochrome P450 2C19 (CYP2C19) gene polymorphisms and the effectiveness and safety of thalidomide in the treatment of patients with immune-related bowel disease (IRBD). Thalidomide 141-152 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 77-84 31916702-2 2020 METHODS: CYP2C19 variants in 79 patients treated with thalidomide were analyzed using a polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Thalidomide 54-65 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 9-16 31422568-10 2020 Hesperidin found stronger binding strength and IC50 was relative to thalidomide on TNF alpha, IL 6, and caspase 3. Thalidomide 68-79 tumor necrosis factor Rattus norvegicus 83-92 31422568-10 2020 Hesperidin found stronger binding strength and IC50 was relative to thalidomide on TNF alpha, IL 6, and caspase 3. Thalidomide 68-79 caspase 3 Rattus norvegicus 104-113 31767403-3 2020 We describe herein the design, synthesis, and evaluation of PROTAC BET degraders, based on the BET inhibitor with selectivity for the first Bromodomain benzo[cd]indole-2-one, alkylamide linker and cereblon ligand thalidomide. Thalidomide 213-224 delta/notch like EGF repeat containing Homo sapiens 67-70 31964914-0 2020 CRL4-Cereblon complex in Thalidomide Embryopathy: a translational investigation. Thalidomide 25-36 interleukin 17 receptor B Homo sapiens 0-4 31964914-0 2020 CRL4-Cereblon complex in Thalidomide Embryopathy: a translational investigation. Thalidomide 25-36 cereblon Homo sapiens 5-13 31964914-1 2020 The Cereblon-CRL4 complex has been studied predominantly with regards to thalidomide treatment of multiple myeloma. Thalidomide 73-84 cereblon Homo sapiens 4-12 31964914-1 2020 The Cereblon-CRL4 complex has been studied predominantly with regards to thalidomide treatment of multiple myeloma. Thalidomide 73-84 interleukin 17 receptor B Homo sapiens 13-17 31964914-2 2020 Nevertheless, the role of Cereblon-CRL4 in Thalidomide Embryopathy (TE) is still not understood. Thalidomide 43-54 cereblon Homo sapiens 26-34 31964914-2 2020 Nevertheless, the role of Cereblon-CRL4 in Thalidomide Embryopathy (TE) is still not understood. Thalidomide 43-54 interleukin 17 receptor B Homo sapiens 35-39 31964914-9 2020 In conclusion, we suggest that CRL4-Cereblon variants act through several regulatory mechanisms, which may influence CRL4-Cereblon complex assembly and its ability to bind thalidomide. Thalidomide 172-183 interleukin 17 receptor B Homo sapiens 31-35 31964914-9 2020 In conclusion, we suggest that CRL4-Cereblon variants act through several regulatory mechanisms, which may influence CRL4-Cereblon complex assembly and its ability to bind thalidomide. Thalidomide 172-183 cereblon Homo sapiens 36-44 31964914-9 2020 In conclusion, we suggest that CRL4-Cereblon variants act through several regulatory mechanisms, which may influence CRL4-Cereblon complex assembly and its ability to bind thalidomide. Thalidomide 172-183 interleukin 17 receptor B Homo sapiens 117-121 31964914-9 2020 In conclusion, we suggest that CRL4-Cereblon variants act through several regulatory mechanisms, which may influence CRL4-Cereblon complex assembly and its ability to bind thalidomide. Thalidomide 172-183 cereblon Homo sapiens 122-130 31963193-13 2020 IMiDs decreased TLR4-induced mediator release; this effect was stronger for pomalidomide than for lenalidomide and especially thalidomide. Thalidomide 126-137 toll like receptor 4 Homo sapiens 16-20 32021103-0 2020 Curcumin Combined with Thalidomide Reduces Expression of STAT3 and Bcl-xL, Leading to Apoptosis in Acute Myeloid Leukemia Cell Lines. Thalidomide 23-34 signal transducer and activator of transcription 3 Homo sapiens 57-62 32021103-0 2020 Curcumin Combined with Thalidomide Reduces Expression of STAT3 and Bcl-xL, Leading to Apoptosis in Acute Myeloid Leukemia Cell Lines. Thalidomide 23-34 BCL2 like 1 Homo sapiens 67-73 32021103-9 2020 The expression level of STAT3 and BCL-XL was significantly down-regulated in KG-1 cells after treatment by CUR and THAL and their combination. Thalidomide 115-119 signal transducer and activator of transcription 3 Homo sapiens 24-29 32021103-9 2020 The expression level of STAT3 and BCL-XL was significantly down-regulated in KG-1 cells after treatment by CUR and THAL and their combination. Thalidomide 115-119 BCL2 like 1 Homo sapiens 34-40 32021103-10 2020 Conclusion: Overall, our findings suggested that down-regulation of STAT3 and BCL-XL mRNA expression in response to CUR and THAL treatment lead to inhibition of cell growth and induction of apoptosis. Thalidomide 124-128 signal transducer and activator of transcription 3 Homo sapiens 68-73 32021103-10 2020 Conclusion: Overall, our findings suggested that down-regulation of STAT3 and BCL-XL mRNA expression in response to CUR and THAL treatment lead to inhibition of cell growth and induction of apoptosis. Thalidomide 124-128 BCL2 like 1 Homo sapiens 78-84 32010638-7 2019 Thalidomide, an inhibitor of monocyte-derived TNF-alpha, was evaluated in the rabbit model of TBM and shown to improve survival and reduce inflammation of the brain and the meninges. Thalidomide 0-11 tumor necrosis factor Oryctolagus cuniculus 46-55 31767403-3 2020 We describe herein the design, synthesis, and evaluation of PROTAC BET degraders, based on the BET inhibitor with selectivity for the first Bromodomain benzo[cd]indole-2-one, alkylamide linker and cereblon ligand thalidomide. Thalidomide 213-224 delta/notch like EGF repeat containing Homo sapiens 95-98 31044460-0 2019 Blockade of angiogenin by thalidomide inhibits the tumorigenesis of murine hemangioendothelioma. Thalidomide 26-37 angiogenin, ribonuclease, RNase A family, 5 Mus musculus 12-22 31920668-4 2019 Herein, our study demonstrated that thalidomide protected colon mucosa against trinitro-benzene-sulfonic acid (TNBS)-induced injury, diminished inflammatory infiltration and levels of IFN-gamma, IGF-1, IL-6, IL-17, TNF-alpha, while increased the levels of IL-10 and TGF-gamma. Thalidomide 36-47 interferon gamma Mus musculus 184-193 31920668-4 2019 Herein, our study demonstrated that thalidomide protected colon mucosa against trinitro-benzene-sulfonic acid (TNBS)-induced injury, diminished inflammatory infiltration and levels of IFN-gamma, IGF-1, IL-6, IL-17, TNF-alpha, while increased the levels of IL-10 and TGF-gamma. Thalidomide 36-47 insulin-like growth factor 1 Mus musculus 195-200 31920668-4 2019 Herein, our study demonstrated that thalidomide protected colon mucosa against trinitro-benzene-sulfonic acid (TNBS)-induced injury, diminished inflammatory infiltration and levels of IFN-gamma, IGF-1, IL-6, IL-17, TNF-alpha, while increased the levels of IL-10 and TGF-gamma. Thalidomide 36-47 interleukin 6 Mus musculus 202-206 31920668-4 2019 Herein, our study demonstrated that thalidomide protected colon mucosa against trinitro-benzene-sulfonic acid (TNBS)-induced injury, diminished inflammatory infiltration and levels of IFN-gamma, IGF-1, IL-6, IL-17, TNF-alpha, while increased the levels of IL-10 and TGF-gamma. Thalidomide 36-47 interleukin 17A Mus musculus 208-213 31920668-4 2019 Herein, our study demonstrated that thalidomide protected colon mucosa against trinitro-benzene-sulfonic acid (TNBS)-induced injury, diminished inflammatory infiltration and levels of IFN-gamma, IGF-1, IL-6, IL-17, TNF-alpha, while increased the levels of IL-10 and TGF-gamma. Thalidomide 36-47 tumor necrosis factor Mus musculus 215-224 31920668-4 2019 Herein, our study demonstrated that thalidomide protected colon mucosa against trinitro-benzene-sulfonic acid (TNBS)-induced injury, diminished inflammatory infiltration and levels of IFN-gamma, IGF-1, IL-6, IL-17, TNF-alpha, while increased the levels of IL-10 and TGF-gamma. Thalidomide 36-47 interleukin 10 Mus musculus 256-261 30914422-1 2020 AIM: To determine whether changes in the serum levels of vascular endothelial growth factor (VEGF) after thalidomide therapy will affect the peripapillary retinal thickness (pRT) associated with optic disc oedema (ODE) in patients with polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy and skin changes (POEMS) syndrome. Thalidomide 105-116 vascular endothelial growth factor A Homo sapiens 57-91 30914422-4 2020 We determined whether a change in the pRT from baseline was significantly correlated with the serum level of VEGF from that at 6 months after the thalidomide treatment. Thalidomide 146-157 vascular endothelial growth factor A Homo sapiens 109-113 32522938-2 2020 Although the molecular mechanisms of thalidomide and its derivatives remained poorly understood until recently, we identified cereblon (CRBN), a primary direct target of thalidomide, using ferrite glycidyl methacrylate (FG) beads. Thalidomide 37-48 cereblon Homo sapiens 136-140 32522938-2 2020 Although the molecular mechanisms of thalidomide and its derivatives remained poorly understood until recently, we identified cereblon (CRBN), a primary direct target of thalidomide, using ferrite glycidyl methacrylate (FG) beads. Thalidomide 170-181 cereblon Homo sapiens 136-140 32522938-4 2020 When a ligand such as thalidomide binds to CRBN, it recognizes various "neosubstrates" depending on the shape of the ligand. Thalidomide 22-33 cereblon Homo sapiens 43-47 31867326-4 2019 Thalidomide and its derivatives, termed immunomodulatory imide drugs (IMiDs), are a class of drugs that target the 3"-untranslated region (3"-UTR) of TNF-alpha mRNA, inhibiting TNF-alpha production. Thalidomide 0-11 tumor necrosis factor Homo sapiens 150-159 31867326-4 2019 Thalidomide and its derivatives, termed immunomodulatory imide drugs (IMiDs), are a class of drugs that target the 3"-untranslated region (3"-UTR) of TNF-alpha mRNA, inhibiting TNF-alpha production. Thalidomide 0-11 tumor necrosis factor Homo sapiens 177-186 31044460-5 2019 By qRT-PCR screening, we observed that the expression of angiogenin was downregulated by thalidomide treatment. Thalidomide 89-100 angiogenin, ribonuclease, RNase A family, 5 Mus musculus 57-67 31044460-7 2019 Moreover, we confirmed that the antitumoral role of thalidomide is dependent on angiogenin expression both in vivo and in vitro. Thalidomide 52-63 angiogenin Homo sapiens 80-90 32186620-2 2019 Since 2013, the treatment of these patients in the public system is based on CTD (cyclophosphamide, thalidomide, and dexamethasone). Thalidomide 100-111 CTD Homo sapiens 77-80 30890040-3 2019 This study is to assess if serum PF4 could be a prognostic factor in predicting treatment response and survival of MM treated with thalidomide and VAD regimens. Thalidomide 131-142 platelet factor 4 Homo sapiens 33-36 30890040-12 2019 CONCLUSIONS: We speculate serum PF4 is a promising response and prognostic factor in newly diagnosed MM treated with thalidomide and VAD regimens. Thalidomide 117-128 platelet factor 4 Homo sapiens 32-35 31335998-1 2019 BACKGROUND: Thalidomide and closely related analogues are used clinically for their immunomodulatory and antiangiogenic properties mediated by the inhibition of the proinflammatory cytokine tumor necrosis factor alpha. Thalidomide 12-23 tumor necrosis factor Rattus norvegicus 190-217 31591562-0 2019 p63 is a cereblon substrate involved in thalidomide teratogenicity. Thalidomide 40-51 tumor protein p63 Danio rerio 0-3 31819606-11 2019 Furthermore, the GFAP and Iba1 expressions and TNF-alpha levels of the ipsilateral spinal dorsal horn in SNL+Tha group were significantly weaker from day 3 to day 14 after SNL than those in SNL+Veh group (P < 0.05). Thalidomide 109-112 glial fibrillary acidic protein Homo sapiens 17-21 31819606-11 2019 Furthermore, the GFAP and Iba1 expressions and TNF-alpha levels of the ipsilateral spinal dorsal horn in SNL+Tha group were significantly weaker from day 3 to day 14 after SNL than those in SNL+Veh group (P < 0.05). Thalidomide 109-112 allograft inflammatory factor 1 Homo sapiens 26-30 31819606-11 2019 Furthermore, the GFAP and Iba1 expressions and TNF-alpha levels of the ipsilateral spinal dorsal horn in SNL+Tha group were significantly weaker from day 3 to day 14 after SNL than those in SNL+Veh group (P < 0.05). Thalidomide 109-112 tumor necrosis factor Homo sapiens 47-56 32128089-0 2019 Thalidomide attenuates the hyporesponsiveness of isolated atria to chronotropic stimulation in BDL rats: The involvement of TNF-alpha, IL-6 inhibition, and SOCS1 activation. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 124-133 32128089-0 2019 Thalidomide attenuates the hyporesponsiveness of isolated atria to chronotropic stimulation in BDL rats: The involvement of TNF-alpha, IL-6 inhibition, and SOCS1 activation. Thalidomide 0-11 interleukin 6 Rattus norvegicus 135-139 32128089-0 2019 Thalidomide attenuates the hyporesponsiveness of isolated atria to chronotropic stimulation in BDL rats: The involvement of TNF-alpha, IL-6 inhibition, and SOCS1 activation. Thalidomide 0-11 suppressor of cytokine signaling 1 Rattus norvegicus 156-161 32128089-14 2019 On the other hand, hepatic SOCS1 expression was up-regulated by thalidomide treatment in this group. Thalidomide 64-75 suppressor of cytokine signaling 1 Rattus norvegicus 27-32 30962579-4 2019 WL40 was synthesized by linking the Rpn13 covalent inhibitor RA190 with the cereblon (CRBN) binding ligand thalidomide. Thalidomide 107-118 cereblon Homo sapiens 86-90 31591562-1 2019 Cereblon (CRBN) is a primary target of thalidomide and mediates its multiple pharmacological activities, including teratogenic and antimyeloma activities. Thalidomide 39-50 cereblon Danio rerio 10-14 31591562-2 2019 CRBN functions as a substrate receptor of the E3 ubiquitin ligase CRL4, whose substrate specificity is modulated by thalidomide and its analogs. Thalidomide 116-127 cereblon Danio rerio 0-4 31591562-4 2019 Here we show that p63 isoforms are thalidomide-dependent CRL4CRBN neosubstrates that are responsible, at least in part, for its teratogenic effects. Thalidomide 35-46 tumor protein p63 Danio rerio 18-21 30934098-0 2019 A Cautious Note on Thalidomide Usage in Cancer Treatment: Genetic Profiling of the TBX2 Sub-Family Gene Expression is Required. Thalidomide 19-30 T-box transcription factor 2 Homo sapiens 83-87 31670229-11 2019 Compared with vehicle (phosphate-buffered saline), thalidomide significantly inhibited the secretion of IFN-gamma and IL-17 in ITP mouse and reduced the expression of CD68 in spleen. Thalidomide 51-62 interferon gamma Mus musculus 104-113 31670229-11 2019 Compared with vehicle (phosphate-buffered saline), thalidomide significantly inhibited the secretion of IFN-gamma and IL-17 in ITP mouse and reduced the expression of CD68 in spleen. Thalidomide 51-62 interleukin 17A Mus musculus 118-123 31670229-11 2019 Compared with vehicle (phosphate-buffered saline), thalidomide significantly inhibited the secretion of IFN-gamma and IL-17 in ITP mouse and reduced the expression of CD68 in spleen. Thalidomide 51-62 CD68 antigen Mus musculus 167-171 31487824-8 2019 Based on the structure of dicarboximides and thalidomide, novel proteolysis-targeting chimeras (PROTACs) were synthesized and used as tools to downregulate ABC50 in leukemia cells. Thalidomide 45-56 ATP binding cassette subfamily F member 1 Homo sapiens 156-161 31487824-0 2019 New Thalidomide-Resembling Dicarboximides Target ABC50 Protein and Show Antileukemic and Immunomodulatory Activities. Thalidomide 4-15 ATP binding cassette subfamily F member 1 Homo sapiens 49-54 31115923-0 2019 Polymorphisms in the promotor region of the CRBN gene as a predictive factor for peripheral neuropathy in the course of thalidomide-based chemotherapy in multiple myeloma patients. Thalidomide 120-131 cereblon Homo sapiens 44-48 31115923-2 2019 This study aimed to analyse the influence of clinical and molecular factors - single nucleotide polymorphisms (SNPs) of the CRBN gene: rs6768972 and rs1672753, on the risk of adverse effects (AEs) of thalidomide-based chemotherapy in patients with MM. Thalidomide 200-211 cereblon Homo sapiens 124-128 31115923-8 2019 Selected CRBN SNPs may be useful in assessing the probability of AEs in the form of peripheral polyneuropathy and gastrointestinal motility disorders associated with the use of thalidomide in patients with MM. Thalidomide 177-188 cereblon Homo sapiens 9-13 30934098-5 2019 The identification of CRBN and TBX5 as potential direct ligands for this drug have opened the way to better understand its efficiency and its failure.We hereby review these mechanisms and provide evidence that could explain why thalidomide failed to make it as a drug of choice in lung cancer treatment. Thalidomide 228-239 cereblon Homo sapiens 22-26 30934098-5 2019 The identification of CRBN and TBX5 as potential direct ligands for this drug have opened the way to better understand its efficiency and its failure.We hereby review these mechanisms and provide evidence that could explain why thalidomide failed to make it as a drug of choice in lung cancer treatment. Thalidomide 228-239 T-box transcription factor 5 Homo sapiens 31-35 30934098-6 2019 Linking the genetic signature of TBX2 subfamily in these tumors to their inability to respond properly to thalidomide raises concerns of worsening lung cancer patients" health if this drug is utilized. Thalidomide 106-117 T-box transcription factor 2 Homo sapiens 33-37 31175889-3 2019 In this study, we used two mouse models of depression and investigated the possible role of nitric oxide (NO), NO synthase (NOS) and inducible NOS (iNOS) in the ant-depressant-like effects of thalidomide. Thalidomide 192-203 nitric oxide synthase 2, inducible Mus musculus 133-146 30806135-0 2019 Thalidomide alleviates bone cancer pain by down-regulating expressions of NF-kappaB and GFAP in spinal astrocytes in a mouse model. Thalidomide 0-11 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 74-83 30806135-0 2019 Thalidomide alleviates bone cancer pain by down-regulating expressions of NF-kappaB and GFAP in spinal astrocytes in a mouse model. Thalidomide 0-11 glial fibrillary acidic protein Mus musculus 88-92 30806135-2 2019 Previous study has shown that thalidomide decreases the expression of tumor necrosis factor alpha in the mouse spinal cord. Thalidomide 30-41 tumor necrosis factor Mus musculus 70-97 30806135-4 2019 Here, we investigated the effect of thalidomide on the expression level of NF-kappaB as well as glial fibrillary acidic protein (GFAP) in the spinal cord astrocyte in a mice model. Thalidomide 36-47 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 75-84 30806135-11 2019 Treating the tumor-bearing mice with thalidomide results in a dramatic reduction in pain behaviors and a significant decrease of NF-kappaB and GFAP expressions. Thalidomide 37-48 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 129-138 30806135-11 2019 Treating the tumor-bearing mice with thalidomide results in a dramatic reduction in pain behaviors and a significant decrease of NF-kappaB and GFAP expressions. Thalidomide 37-48 glial fibrillary acidic protein Mus musculus 143-147 30806135-12 2019 Conclusions: Thalidomide alleviates the pain behaviors probably by down-regulating the expression of NF-kappaB and GFAP. Thalidomide 13-24 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 101-110 30806135-12 2019 Conclusions: Thalidomide alleviates the pain behaviors probably by down-regulating the expression of NF-kappaB and GFAP. Thalidomide 13-24 glial fibrillary acidic protein Mus musculus 115-119 31168767-0 2019 Post-treatment improvement of NK cell numbers predicts better survival in myeloma patients treated with thalidomide-based regimens. Thalidomide 104-115 solute carrier family 35 member G1 Homo sapiens 0-4 31388035-0 2019 The role of ESCO2, SALL4 and TBX5 genes in the susceptibility to thalidomide teratogenesis. Thalidomide 65-76 establishment of sister chromatid cohesion N-acetyltransferase 2 Homo sapiens 12-17 31132355-0 2019 Synergistic effects of gefitinib and thalidomide treatment on EGFR-TKI-sensitive and -resistant NSCLC. Thalidomide 37-48 epidermal growth factor receptor Homo sapiens 62-66 31132355-8 2019 The inhibition of EGFR phosphorylation and downstream signaling was more pronounced in the thalidomide and gefitinib co-treatment group as compared with the single agent treatment groups. Thalidomide 91-102 epidermal growth factor receptor Homo sapiens 18-22 31132355-9 2019 Further study revealed that the inhibitors of AKT, MEK/ERK, and p38 increased the antiproliferative and proapoptotic effects of the combined treatment of thalidomide and gefitinib. Thalidomide 154-165 AKT serine/threonine kinase 1 Homo sapiens 46-49 31132355-9 2019 Further study revealed that the inhibitors of AKT, MEK/ERK, and p38 increased the antiproliferative and proapoptotic effects of the combined treatment of thalidomide and gefitinib. Thalidomide 154-165 mitogen-activated protein kinase kinase 7 Homo sapiens 51-54 31132355-9 2019 Further study revealed that the inhibitors of AKT, MEK/ERK, and p38 increased the antiproliferative and proapoptotic effects of the combined treatment of thalidomide and gefitinib. Thalidomide 154-165 EPH receptor B2 Homo sapiens 55-58 31132355-9 2019 Further study revealed that the inhibitors of AKT, MEK/ERK, and p38 increased the antiproliferative and proapoptotic effects of the combined treatment of thalidomide and gefitinib. Thalidomide 154-165 mitogen-activated protein kinase 14 Homo sapiens 64-67 31132355-11 2019 In conclusion, thalidomide and gefitinib exhibit synergistic effects on both TKI-sensitive and -resistant NSCLC cells by targeting the EGFR signaling pathways, suggesting that the combination strategy is promising for the treatment of NSCLC. Thalidomide 15-26 epidermal growth factor receptor Homo sapiens 135-139 31388035-0 2019 The role of ESCO2, SALL4 and TBX5 genes in the susceptibility to thalidomide teratogenesis. Thalidomide 65-76 spalt like transcription factor 4 Homo sapiens 19-24 31388035-0 2019 The role of ESCO2, SALL4 and TBX5 genes in the susceptibility to thalidomide teratogenesis. Thalidomide 65-76 T-box transcription factor 5 Homo sapiens 29-33 31388035-4 2019 Recently, SALL4 and TBX5 were demonstrated to be thalidomide targets. Thalidomide 49-60 spalt like transcription factor 4 Homo sapiens 10-15 31388035-4 2019 Recently, SALL4 and TBX5 were demonstrated to be thalidomide targets. Thalidomide 49-60 T-box transcription factor 5 Homo sapiens 20-24 31132668-13 2019 Particularly, we showed thalidomide reduced CD4+ T helper cell infiltration and downregulated Th1- and Th17-polarizing genes. Thalidomide 24-35 CD4 antigen Mus musculus 44-47 31388035-10 2019 DGE analysis showed a significant reduction of ESCO2 in hPSCs after thalidomide exposure. Thalidomide 68-79 establishment of sister chromatid cohesion N-acetyltransferase 2 Homo sapiens 47-52 31132668-13 2019 Particularly, we showed thalidomide reduced CD4+ T helper cell infiltration and downregulated Th1- and Th17-polarizing genes. Thalidomide 24-35 negative elongation factor complex member C/D, Th1l Mus musculus 94-97 31043423-4 2019 Thalidomide analogs bind the CRL4CRBN ubiquitin ligase and induce degradation of particular proteins, but each of the molecules studied has distinct patterns of substrate specificity that likely underlie the clinical activity and toxicities of each drug. Thalidomide 0-11 interleukin 17 receptor B Homo sapiens 29-33 31132668-14 2019 In addition, thalidomide treatment lowered the microvessel density and vascular endothelial growth factor (VEGF) expression. Thalidomide 13-24 vascular endothelial growth factor A Mus musculus 71-105 31132668-14 2019 In addition, thalidomide treatment lowered the microvessel density and vascular endothelial growth factor (VEGF) expression. Thalidomide 13-24 vascular endothelial growth factor A Mus musculus 107-111 31132668-15 2019 We further demonstrated that thalidomide suppressed NF-kappaB activation in LL37-treated skin and in TNF-alpha-stimulated HaCaT keratinocytes in vitro. Thalidomide 29-40 tumor necrosis factor Mus musculus 101-110 31251063-0 2019 De-Novo Design of Cereblon (CRBN) Effectors Guided by Natural Hydrolysis Products of Thalidomide Derivatives. Thalidomide 85-96 cereblon Homo sapiens 18-26 31251063-0 2019 De-Novo Design of Cereblon (CRBN) Effectors Guided by Natural Hydrolysis Products of Thalidomide Derivatives. Thalidomide 85-96 cereblon Homo sapiens 28-32 31030100-0 2019 Thalidomide induce response in patients with corticosteroid-resistant or relapsed ITP by upregulating Neuropilin-1 expression. Thalidomide 0-11 neuropilin 1 Homo sapiens 102-114 31030100-10 2019 In vitro, THD treatment upregulated expression of NRP-1and Tregs only in ITP patients. Thalidomide 10-13 neuropilin 1 Homo sapiens 50-55 31030100-12 2019 Additionally, rhTPO plus THD or THD therapy significantly increased the levels of NRP-1, Tregs and Bregs in responders. Thalidomide 25-28 neuropilin 1 Homo sapiens 82-87 31030100-12 2019 Additionally, rhTPO plus THD or THD therapy significantly increased the levels of NRP-1, Tregs and Bregs in responders. Thalidomide 32-35 neuropilin 1 Homo sapiens 82-87 31030100-13 2019 CONCLUSIONS: Our study shows for the first time that NRP-1 is involved in the pathogenesis of ITP, THD could induce response in ITP patients by upregulating NRP-1 expression and restoring the proportion of Tregs and Bregs. Thalidomide 99-102 neuropilin 1 Homo sapiens 53-58 31030100-13 2019 CONCLUSIONS: Our study shows for the first time that NRP-1 is involved in the pathogenesis of ITP, THD could induce response in ITP patients by upregulating NRP-1 expression and restoring the proportion of Tregs and Bregs. Thalidomide 99-102 neuropilin 1 Homo sapiens 157-162 31055217-1 2019 Thalidomide is a teratogen that causes multiple malformations in the developing baby through its interaction with cereblon (CRBN), a substrate receptor subunit of the CRL4 E3 ubiquitin ligase complex. Thalidomide 0-11 cereblon Danio rerio 124-128 31289557-7 2019 The present data indicate that thalidomide contributes to an improvement of prognosis for patients with locally advanced EC with elevated serum VEGF levels during radiotherapy. Thalidomide 31-42 vascular endothelial growth factor A Homo sapiens 144-148 31464258-1 2019 Thalidomide, which is an angiogenesis inhibitor and immunomodulator that reduces tumor necrosis factor-alpha, has regained value in the treatment of multiple myeloma. Thalidomide 0-11 tumor necrosis factor Homo sapiens 81-108 31055217-5 2019 Knockdown of CRBN in zebrafish embryos impaired brain development and led to small brains, as did treatment with thalidomide. Thalidomide 113-124 cereblon Danio rerio 13-17 30612232-7 2019 Long-term follow-up found that all 12 patients received autologous stem cell transplant, melphalan-based therapy or lenalidomide/thalidomide-based therapy obtained clinical improvement, of which eight experienced decreased levels of VEGF by 50% or back to normal. Thalidomide 129-140 vascular endothelial growth factor A Homo sapiens 233-237 30834740-0 2019 Thalidomide and Its Analogs Differentially Target Fibroblast Growth Factor Receptors: Thalidomide Suppresses FGFR Gene Expression while Pomalidomide Dampens FGFR2 Activity. Thalidomide 0-11 fibroblast growth factor receptor 2 Gallus gallus 157-162 30834740-8 2019 Corroboration with previous studies suggests that thalidomide might affect FGFR expression through the transcription factor, E2F1. Thalidomide 50-61 E2F transcription factor 1 Gallus gallus 125-129 30834740-11 2019 Kinome analysis revealed that kinase activities of FGFR2 and FGFR3 (G697C) reduced by 31% and 65%, respectively, in the presence of 10 muM thalidomide. Thalidomide 139-150 fibroblast growth factor receptor 2 Gallus gallus 51-56 30834740-11 2019 Kinome analysis revealed that kinase activities of FGFR2 and FGFR3 (G697C) reduced by 31% and 65%, respectively, in the presence of 10 muM thalidomide. Thalidomide 139-150 fibroblast growth factor receptor 3 Gallus gallus 61-66 31157769-1 2019 The immunomodulatory drugs (IMiDs) thalidomide and its analogs, lenalidomide and pomalidomide, all FDA approved drugs for the treatment of multiple myeloma, induce ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase for proteasomal degradation. Thalidomide 35-46 IKAROS family zinc finger 1 Homo sapiens 241-246 31157769-1 2019 The immunomodulatory drugs (IMiDs) thalidomide and its analogs, lenalidomide and pomalidomide, all FDA approved drugs for the treatment of multiple myeloma, induce ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase for proteasomal degradation. Thalidomide 35-46 IKAROS family zinc finger 3 Homo sapiens 252-258 31157769-1 2019 The immunomodulatory drugs (IMiDs) thalidomide and its analogs, lenalidomide and pomalidomide, all FDA approved drugs for the treatment of multiple myeloma, induce ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase for proteasomal degradation. Thalidomide 35-46 IKAROS family zinc finger 3 Homo sapiens 260-265 31157769-1 2019 The immunomodulatory drugs (IMiDs) thalidomide and its analogs, lenalidomide and pomalidomide, all FDA approved drugs for the treatment of multiple myeloma, induce ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase for proteasomal degradation. Thalidomide 35-46 cereblon Homo sapiens 285-289 31157769-1 2019 The immunomodulatory drugs (IMiDs) thalidomide and its analogs, lenalidomide and pomalidomide, all FDA approved drugs for the treatment of multiple myeloma, induce ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase for proteasomal degradation. Thalidomide 35-46 mitochondrial E3 ubiquitin protein ligase 1 Homo sapiens 291-310 30687387-8 2018 The observed limb phenotype is similar to the severe proximal truncations observed in human babies exposed to thalidomide, which has been proposed to block the Fgf10-AER-Fgf8 feedback loop. Thalidomide 110-121 fibroblast growth factor 10 Homo sapiens 160-165 30683719-3 2019 The thalidomide-like molecules (collectively called the IMiDs) bind to the ubiquitously expressed cereblon ubiquitin ligase complex and alter its substrate specificity such that it targets the IKZF1 and IKZF3 lymphocyte transcription factors for destruction. Thalidomide 4-15 IKAROS family zinc finger 1 Homo sapiens 193-198 30683719-3 2019 The thalidomide-like molecules (collectively called the IMiDs) bind to the ubiquitously expressed cereblon ubiquitin ligase complex and alter its substrate specificity such that it targets the IKZF1 and IKZF3 lymphocyte transcription factors for destruction. Thalidomide 4-15 IKAROS family zinc finger 3 Homo sapiens 203-208 30736806-7 2019 Importantly, intraperitoneal injection of the TNF-alpha inhibitor thalidomide reduced the phosphorylation of STAT3 and decreased the recruitment of STAT3 and histone H4 hyperacetylation in the Scn8a promoter, thus subsequently attenuating Nav1.6 upregulation in DRG neurons and mechanical allodynia induced by L5-VRT. Thalidomide 66-77 tumor necrosis factor Homo sapiens 46-55 30736806-7 2019 Importantly, intraperitoneal injection of the TNF-alpha inhibitor thalidomide reduced the phosphorylation of STAT3 and decreased the recruitment of STAT3 and histone H4 hyperacetylation in the Scn8a promoter, thus subsequently attenuating Nav1.6 upregulation in DRG neurons and mechanical allodynia induced by L5-VRT. Thalidomide 66-77 signal transducer and activator of transcription 3 Homo sapiens 109-114 30736806-7 2019 Importantly, intraperitoneal injection of the TNF-alpha inhibitor thalidomide reduced the phosphorylation of STAT3 and decreased the recruitment of STAT3 and histone H4 hyperacetylation in the Scn8a promoter, thus subsequently attenuating Nav1.6 upregulation in DRG neurons and mechanical allodynia induced by L5-VRT. Thalidomide 66-77 signal transducer and activator of transcription 3 Homo sapiens 148-153 30736806-7 2019 Importantly, intraperitoneal injection of the TNF-alpha inhibitor thalidomide reduced the phosphorylation of STAT3 and decreased the recruitment of STAT3 and histone H4 hyperacetylation in the Scn8a promoter, thus subsequently attenuating Nav1.6 upregulation in DRG neurons and mechanical allodynia induced by L5-VRT. Thalidomide 66-77 sodium voltage-gated channel alpha subunit 8 Homo sapiens 193-198 30736806-7 2019 Importantly, intraperitoneal injection of the TNF-alpha inhibitor thalidomide reduced the phosphorylation of STAT3 and decreased the recruitment of STAT3 and histone H4 hyperacetylation in the Scn8a promoter, thus subsequently attenuating Nav1.6 upregulation in DRG neurons and mechanical allodynia induced by L5-VRT. Thalidomide 66-77 neuron navigator 1 Homo sapiens 239-243 30717761-10 2019 Thalidomide AEs occurred at more similar rates in males (25 AEs in 41 men, 60.9%) and females (12 in 26 (46.2%), but were more common in ENG patients (17 in 17) than in ACVRL1 (14 in 34, p < 0.0001). Thalidomide 0-11 activin A receptor like type 1 Homo sapiens 169-175 30682785-4 2019 We have previously shown that 3,6"-dithiothalidomide (3,6"-DT), a thalidomide analog that is more potent than thalidomide with similar brain penetration, selectively inhibits the synthesis of TNF-alpha in cultured cells and reverses behavioral impairments induced by mild TBI in mice. Thalidomide 41-52 tumor necrosis factor Mus musculus 192-201 30682785-4 2019 We have previously shown that 3,6"-dithiothalidomide (3,6"-DT), a thalidomide analog that is more potent than thalidomide with similar brain penetration, selectively inhibits the synthesis of TNF-alpha in cultured cells and reverses behavioral impairments induced by mild TBI in mice. Thalidomide 66-77 tumor necrosis factor Mus musculus 192-201 30687387-8 2018 The observed limb phenotype is similar to the severe proximal truncations observed in human babies exposed to thalidomide, which has been proposed to block the Fgf10-AER-Fgf8 feedback loop. Thalidomide 110-121 fibroblast growth factor 8 Homo sapiens 170-174 30362282-8 2019 We emphasize targeting a more specific Th1 mechanism via the tumour necrosis factor (TNF)-alpha cytokine antagonist thalidomide, as it is the only TNF-alpha antagonist currently approved for use in infectious disease settings and has been shown to decrease Th1 overreaction, restoring immune balance in HIV-CM IRIS. Thalidomide 116-127 negative elongation factor complex member C/D Homo sapiens 39-42 30602127-3 2019 Notably, targeting CRL4 has recently emerged as a noval anti-cancer strategy, including thalidomide and its derivatives that bind to the substrate recognition receptor cereblon (CRBN), and anticancer sulfonamides that target DCAF15 to suppress the neoplastic proliferation of multiple myeloma and colorectal cancers, respectively. Thalidomide 88-99 cereblon Homo sapiens 178-182 30651882-12 2019 Taken together, these results suggest that Thd may have therapeutic potential in diabetic renal injury via the AMPK signaling pathway. Thalidomide 43-46 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 111-115 30651882-0 2019 Thalidomide decreases high glucose-induced extracellular matrix protein synthesis in mesangial cells via the AMPK pathway. Thalidomide 0-11 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 109-113 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 61-72 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 138-186 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 61-72 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 188-192 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 61-72 nuclear factor kappa B subunit 1 Homo sapiens 239-248 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 61-72 C-C motif chemokine ligand 2 Rattus norvegicus 250-284 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 61-72 C-C motif chemokine ligand 2 Rattus norvegicus 286-291 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 61-72 transforming growth factor, beta 1 Rattus norvegicus 297-335 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 74-77 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 138-186 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 74-77 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 188-192 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 74-77 nuclear factor kappa B subunit 1 Homo sapiens 239-248 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 74-77 C-C motif chemokine ligand 2 Rattus norvegicus 250-284 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 74-77 C-C motif chemokine ligand 2 Rattus norvegicus 286-291 30651882-1 2019 A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor kappaB (NF-kappaB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1/mothers against decapentaplegic homolog signaling pathways. Thalidomide 74-77 transforming growth factor, beta 1 Rattus norvegicus 297-335 30362282-8 2019 We emphasize targeting a more specific Th1 mechanism via the tumour necrosis factor (TNF)-alpha cytokine antagonist thalidomide, as it is the only TNF-alpha antagonist currently approved for use in infectious disease settings and has been shown to decrease Th1 overreaction, restoring immune balance in HIV-CM IRIS. Thalidomide 116-127 tumor necrosis factor Homo sapiens 61-95 30362282-8 2019 We emphasize targeting a more specific Th1 mechanism via the tumour necrosis factor (TNF)-alpha cytokine antagonist thalidomide, as it is the only TNF-alpha antagonist currently approved for use in infectious disease settings and has been shown to decrease Th1 overreaction, restoring immune balance in HIV-CM IRIS. Thalidomide 116-127 tumor necrosis factor Homo sapiens 147-156 30362282-8 2019 We emphasize targeting a more specific Th1 mechanism via the tumour necrosis factor (TNF)-alpha cytokine antagonist thalidomide, as it is the only TNF-alpha antagonist currently approved for use in infectious disease settings and has been shown to decrease Th1 overreaction, restoring immune balance in HIV-CM IRIS. Thalidomide 116-127 negative elongation factor complex member C/D Homo sapiens 257-260 30155589-1 2018 Cancer-related fatigue (CRF) is one of the adverse events in multiple myeloma (MM) patients treated with cytotoxic agents, proteasome inhibitors (PIs), and immunomodulatory drugs (IMiDs) such as bortezomib, lenalidomide, and thalidomide. Thalidomide 225-236 complement C1q like 1 Homo sapiens 0-22 31031470-2 2019 Erythema nodosum leprosum(ENL) in special cases need to be managed with capsule thalidomide in varying doses. Thalidomide 80-91 MLLT1 super elongation complex subunit Homo sapiens 26-29 31031470-3 2019 We report such a case of bradycardia in thalidomide dose dependent manner in a young ENL male. Thalidomide 40-51 MLLT1 super elongation complex subunit Homo sapiens 85-88 30597765-0 2018 Thalidomide Teratogenic Effects Linked to Degradation of SALL4: After 60 years, researchers have now shed light on the mechanism underlying thalidomide"s devastating teratogenic effects. Thalidomide 0-11 spalt like transcription factor 4 Homo sapiens 57-62 30597765-0 2018 Thalidomide Teratogenic Effects Linked to Degradation of SALL4: After 60 years, researchers have now shed light on the mechanism underlying thalidomide"s devastating teratogenic effects. Thalidomide 140-151 spalt like transcription factor 4 Homo sapiens 57-62 30339882-0 2018 Thalidomide (THD) alleviates radiation induced lung fibrosis (RILF) via down-regulation of TGF-beta/Smad3 signaling pathway in an Nrf2-dependent manner. Thalidomide 0-11 SMAD family member 3 Mus musculus 100-105 30339882-0 2018 Thalidomide (THD) alleviates radiation induced lung fibrosis (RILF) via down-regulation of TGF-beta/Smad3 signaling pathway in an Nrf2-dependent manner. Thalidomide 0-11 nuclear factor, erythroid derived 2, like 2 Mus musculus 130-134 31068561-5 2019 A randomized controlled trial on thalidomide plus dexamethasone for POEMS syndrome showed reduced serum VEGF levels after therapy; however, the incidence of peripheral neuropathy, a well-known side effect of both thalidomide and bortezomib, increased. Thalidomide 33-44 vascular endothelial growth factor A Homo sapiens 104-108 30344721-0 2018 Association of CD117 and HLA-DR expression with shorter overall survival and/or progression-free survival in patients with multiple myeloma treated with bortezomib and thalidomide combination treatment without transplantation. Thalidomide 168-179 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 15-20 30534560-2 2018 Based on reports that vascular endothelial growth factor- (VEGF-) mediated angiogenesis is critical for MM pathogenesis, the antiangiogenic compound thalidomide has been added to VAD (T-VAD). Thalidomide 149-160 vascular endothelial growth factor A Homo sapiens 22-64 30373817-7 2018 We have also tested a thalidomide derivative, FPFT-2216, which showed an inhibitory effect toward IKZF1 protein level. Thalidomide 22-33 IKAROS family zinc finger 1 Mus musculus 98-103 30385546-0 2018 Defining the human C2H2 zinc finger degrome targeted by thalidomide analogs through CRBN. Thalidomide 56-67 cereblon Homo sapiens 84-88 30385546-1 2018 The small molecules thalidomide, lenalidomide, and pomalidomide induce the ubiquitination and proteasomal degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) by recruiting a Cys2-His2 (C2H2) zinc finger domain to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase. Thalidomide 20-31 IKAROS family zinc finger 1 Homo sapiens 155-160 30385546-1 2018 The small molecules thalidomide, lenalidomide, and pomalidomide induce the ubiquitination and proteasomal degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) by recruiting a Cys2-His2 (C2H2) zinc finger domain to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase. Thalidomide 20-31 IKAROS family zinc finger 3 Homo sapiens 174-179 30385546-1 2018 The small molecules thalidomide, lenalidomide, and pomalidomide induce the ubiquitination and proteasomal degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) by recruiting a Cys2-His2 (C2H2) zinc finger domain to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase. Thalidomide 20-31 cereblon Homo sapiens 236-244 30385546-1 2018 The small molecules thalidomide, lenalidomide, and pomalidomide induce the ubiquitination and proteasomal degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) by recruiting a Cys2-His2 (C2H2) zinc finger domain to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase. Thalidomide 20-31 cereblon Homo sapiens 246-250 30252647-1 2018 A transcription factor called SALL4 could be the missing link between thalidomide and the limb defects caused by the drug. Thalidomide 70-81 spalt like transcription factor 4 Homo sapiens 30-35 30190590-0 2018 SALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrate. Thalidomide 35-46 spalt like transcription factor 4 Homo sapiens 0-5 30190590-4 2018 Instead, we identify SALL4 as a thalidomide-dependent cereblon neosubstrate. Thalidomide 32-43 spalt like transcription factor 4 Homo sapiens 21-26 30190590-5 2018 Human mutations in SALL4 cause Duane-radial ray, IVIC, and acro-renal-ocular syndromes with overlapping clinical presentations to thalidomide embryopathy, including phocomelia. Thalidomide 130-141 spalt like transcription factor 4 Homo sapiens 19-24 30301277-5 2018 Among them, 2-[1-(3-chlorobenzyl)-2,6-dioxopiperidin-3-yl]isoindoline-1,3-dione (5c) was found to inhibit TNF-alpha and IL-6 expression in HaCaT cells with a higher potency than thalidomide and no significant cell cytotoxicity was detected at 10 muM. Thalidomide 178-189 tumor necrosis factor Homo sapiens 106-115 30064974-0 2018 Crbn I391V is sufficient to confer in vivo sensitivity to thalidomide and its derivatives in mice. Thalidomide 58-69 cereblon Mus musculus 0-4 30064974-3 2018 Here, we report the development of a mouse model that is sensitive to thalidomide derivatives because of a single amino acid change in the direct target of thalidomide derivatives, cereblon (Crbn). Thalidomide 70-81 cereblon Mus musculus 191-195 30064974-3 2018 Here, we report the development of a mouse model that is sensitive to thalidomide derivatives because of a single amino acid change in the direct target of thalidomide derivatives, cereblon (Crbn). Thalidomide 156-167 cereblon Mus musculus 191-195 30064974-4 2018 In human cells, thalidomide and its analogs bind CRBN and recruit protein targets to the CRL4CRBN E3 ubiquitin ligase, resulting in their ubiquitination and subsequent degradation by the proteasome. Thalidomide 16-27 cereblon Homo sapiens 49-53 30064974-5 2018 We show that mice with a single I391V amino acid change in Crbn exhibit thalidomide-induced degradation of drug targets previously identified in human cells, including Ikaros (Ikzf1), Aiolos (Ikzf3), Zfp91, and casein kinase 1a1 (Ck1alpha), both in vitro and in vivo. Thalidomide 72-83 cereblon Mus musculus 59-63 30064974-5 2018 We show that mice with a single I391V amino acid change in Crbn exhibit thalidomide-induced degradation of drug targets previously identified in human cells, including Ikaros (Ikzf1), Aiolos (Ikzf3), Zfp91, and casein kinase 1a1 (Ck1alpha), both in vitro and in vivo. Thalidomide 72-83 IKAROS family zinc finger 1 Homo sapiens 168-174 30064974-5 2018 We show that mice with a single I391V amino acid change in Crbn exhibit thalidomide-induced degradation of drug targets previously identified in human cells, including Ikaros (Ikzf1), Aiolos (Ikzf3), Zfp91, and casein kinase 1a1 (Ck1alpha), both in vitro and in vivo. Thalidomide 72-83 IKAROS family zinc finger 1 Homo sapiens 176-181 30064974-5 2018 We show that mice with a single I391V amino acid change in Crbn exhibit thalidomide-induced degradation of drug targets previously identified in human cells, including Ikaros (Ikzf1), Aiolos (Ikzf3), Zfp91, and casein kinase 1a1 (Ck1alpha), both in vitro and in vivo. Thalidomide 72-83 IKAROS family zinc finger 3 Homo sapiens 192-197 30064974-5 2018 We show that mice with a single I391V amino acid change in Crbn exhibit thalidomide-induced degradation of drug targets previously identified in human cells, including Ikaros (Ikzf1), Aiolos (Ikzf3), Zfp91, and casein kinase 1a1 (Ck1alpha), both in vitro and in vivo. Thalidomide 72-83 ZFP91 zinc finger protein, atypical E3 ubiquitin ligase Homo sapiens 200-205 30064974-8 2018 We further demonstrate that Crbn I391V is sufficient to confer thalidomide-induced fetal loss in mice, capturing a major toxicity of this class of drugs. Thalidomide 63-74 cereblon Mus musculus 28-32 30118587-7 2018 Homobifunctional CRBN degraders will be useful tools for future biomedical investigations of CRBN-related signaling and may help to further elucidate the molecular mechanism of thalidomide analogues. Thalidomide 177-188 cereblon Homo sapiens 17-21 30118587-1 2018 The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. Thalidomide 35-46 IKAROS family zinc finger 1 Homo sapiens 182-188 30118587-1 2018 The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. Thalidomide 35-46 IKAROS family zinc finger 1 Homo sapiens 190-195 30118587-7 2018 Homobifunctional CRBN degraders will be useful tools for future biomedical investigations of CRBN-related signaling and may help to further elucidate the molecular mechanism of thalidomide analogues. Thalidomide 177-188 cereblon Homo sapiens 93-97 30118587-1 2018 The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. Thalidomide 35-46 IKAROS family zinc finger 3 Homo sapiens 201-207 30118587-1 2018 The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. Thalidomide 35-46 IKAROS family zinc finger 3 Homo sapiens 209-214 29938270-9 2018 Thalidomide, as an inhibitor of ubiquitin ligase, was able to prevent the decrease in claudin-1 protein expression and the TEER reduction in Caco-2 cells. Thalidomide 0-11 claudin 1 Homo sapiens 86-95 30118587-1 2018 The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. Thalidomide 35-46 cereblon Homo sapiens 234-238 30288348-5 2018 Results: We found that high IKZF3, but not IKZF1, expression in T-cells correlates with superior overall survival in MMIII patients treated with immunomodulatory drugs (thalidomide, lenalidomide and pomalidomide). Thalidomide 169-180 IKAROS family zinc finger 3 Homo sapiens 28-33 30029933-15 2018 CONCLUSIONS: Thalidomide, which is a specific VEGF inhibitor, significantly inhibited neointimal hyperplasia and vascular restenosis after balloon injury to the carotid artery in rats, thus potentially providing a novel method for the prevention and treatment of restenosis, especially in-stent restenosis. Thalidomide 13-24 vascular endothelial growth factor A Rattus norvegicus 46-50 30067223-0 2018 Thalidomide promotes degradation of SALL4, a transcription factor implicated in Duane Radial Ray syndrome. Thalidomide 0-11 spalt like transcription factor 4 Homo sapiens 36-41 30067223-4 2018 Strikingly, heterozygous loss of function mutations in SALL4 result in a human developmental condition that phenocopies thalidomide-induced birth defects such as absence of thumbs, phocomelia, defects in ear and eye development, and congenital heart disease. Thalidomide 120-131 spalt like transcription factor 4 Homo sapiens 55-60 30067223-5 2018 We find that thalidomide induces degradation of SALL4 exclusively in humans, primates, and rabbits, but not in rodents or fish, providing a mechanistic link for the species-specific pathogenesis of thalidomide syndrome. Thalidomide 13-24 spalt like transcription factor 4 Homo sapiens 48-53 30067223-5 2018 We find that thalidomide induces degradation of SALL4 exclusively in humans, primates, and rabbits, but not in rodents or fish, providing a mechanistic link for the species-specific pathogenesis of thalidomide syndrome. Thalidomide 198-209 spalt like transcription factor 4 Homo sapiens 48-53 30069488-0 2018 PYR-41 and Thalidomide Impair Dendritic Cell Cross-Presentation by Inhibiting Myddosome Formation and Attenuating the Endosomal Recruitments of p97 and Sec61 via NF-kappaB Inactivation. Thalidomide 11-22 melanotransferrin Homo sapiens 144-147 30069488-0 2018 PYR-41 and Thalidomide Impair Dendritic Cell Cross-Presentation by Inhibiting Myddosome Formation and Attenuating the Endosomal Recruitments of p97 and Sec61 via NF-kappaB Inactivation. Thalidomide 11-22 SEC61 translocon subunit alpha 1 Homo sapiens 152-157 30069488-0 2018 PYR-41 and Thalidomide Impair Dendritic Cell Cross-Presentation by Inhibiting Myddosome Formation and Attenuating the Endosomal Recruitments of p97 and Sec61 via NF-kappaB Inactivation. Thalidomide 11-22 nuclear factor kappa B subunit 1 Homo sapiens 162-171 30069488-5 2018 We demonstrated that the inhibition of endosomal recruitment of p97 and Sec61, together with attenuated NF-kappaB activation and Myddosome formation, contributes to PYR-41- and thalidomide-impaired cross-presentation and thereby reverses cross-activation of T cells. Thalidomide 177-188 melanotransferrin Homo sapiens 64-67 30069488-5 2018 We demonstrated that the inhibition of endosomal recruitment of p97 and Sec61, together with attenuated NF-kappaB activation and Myddosome formation, contributes to PYR-41- and thalidomide-impaired cross-presentation and thereby reverses cross-activation of T cells. Thalidomide 177-188 SEC61 translocon subunit alpha 1 Homo sapiens 72-77 30069488-5 2018 We demonstrated that the inhibition of endosomal recruitment of p97 and Sec61, together with attenuated NF-kappaB activation and Myddosome formation, contributes to PYR-41- and thalidomide-impaired cross-presentation and thereby reverses cross-activation of T cells. Thalidomide 177-188 nuclear factor kappa B subunit 1 Homo sapiens 104-113 30069488-6 2018 These observations suggest that NF-kappaB signaling and p97 and Sec61 molecules are candidates for dealing with the side effects of PYR-41 and thalidomide. Thalidomide 143-154 nuclear factor kappa B subunit 1 Homo sapiens 32-41 30069488-6 2018 These observations suggest that NF-kappaB signaling and p97 and Sec61 molecules are candidates for dealing with the side effects of PYR-41 and thalidomide. Thalidomide 143-154 melanotransferrin Homo sapiens 56-59 30069488-6 2018 These observations suggest that NF-kappaB signaling and p97 and Sec61 molecules are candidates for dealing with the side effects of PYR-41 and thalidomide. Thalidomide 143-154 SEC61 translocon subunit alpha 1 Homo sapiens 64-69 29763936-0 2018 Synergistic Inhibition of Thalidomide and Icotinib on Human Non-Small Cell Lung Carcinomas Through ERK and AKT Signaling. Thalidomide 26-37 AKT serine/threonine kinase 1 Homo sapiens 107-110 30029250-7 2018 Results: Thalidomide and three novel analogs all showed inhibitory effects on endothelial cell proliferation and VEGF expression in vitro. Thalidomide 9-20 vascular endothelial growth factor A Rattus norvegicus 113-117 29750304-2 2018 Thalidomide is a drug that has been reported to inhibit angiogenesis and reduce VEGF production by downregulating VEGF expression. Thalidomide 0-11 vascular endothelial growth factor A Mus musculus 80-84 29750304-2 2018 Thalidomide is a drug that has been reported to inhibit angiogenesis and reduce VEGF production by downregulating VEGF expression. Thalidomide 0-11 vascular endothelial growth factor A Mus musculus 114-118 29750304-13 2018 Thus, the results of the present study demonstrated that intraperitoneal administration of thalidomide may alleviate the development of early OA by suppressing VEGF expression in mice and may have potential as a novel therapy for the treatment of OA. Thalidomide 91-102 vascular endothelial growth factor A Mus musculus 160-164 29763936-0 2018 Synergistic Inhibition of Thalidomide and Icotinib on Human Non-Small Cell Lung Carcinomas Through ERK and AKT Signaling. Thalidomide 26-37 EPH receptor B2 Homo sapiens 99-102 29750304-0 2018 Intraperitoneal injection of thalidomide alleviates early osteoarthritis development by suppressing vascular endothelial growth factor expression in mice. Thalidomide 29-40 vascular endothelial growth factor A Mus musculus 100-134 29854627-4 2018 Thalidomide and carboplatin induced up-regulation of the expression of p53, tumor necrosis factor-alpha and interleukin-6 in brain and kidney. Thalidomide 0-11 tumor protein p53 Homo sapiens 71-74 29593101-13 2018 Thalidomide reduced the burden of CD68+ cells and the expression of inflammatory cytokines in the bAVM lesions. Thalidomide 0-11 CD68 antigen Mus musculus 34-38 29593101-15 2018 Thalidomide increased Pdgfb expression in bAVM lesion. Thalidomide 0-11 platelet derived growth factor, B polypeptide Mus musculus 22-27 29593101-16 2018 Overexpression of PDGFB mimicked the effect of thalidomide. Thalidomide 47-58 platelet derived growth factor, B polypeptide Mus musculus 18-23 29593101-17 2018 CONCLUSIONS: Thalidomide and lenalidomide improve mural cell coverage of bAVM vessels and reduce bAVM hemorrhage, which is likely through upregulation of Pdgfb expression. Thalidomide 13-24 platelet derived growth factor, B polypeptide Mus musculus 154-159 29854627-8 2018 GSPE co-treatment with thalidomide and carboplatin reduced their brain and renal damage, oxidative stress, diminished cytokines, p53, neurotransmitters and biochemical parameters, and inhibited brain and renal cell apoptosis. Thalidomide 23-34 tumor protein p53 Homo sapiens 129-132 29746508-0 2018 CUL5 is required for thalidomide-dependent inhibition of cellular proliferation. Thalidomide 21-32 cullin 5 Homo sapiens 0-4 29746508-3 2018 We discovered that CUL5 (previously identified as VACM-1), a scaffold protein in E3 ligase complexes, is involved in thalidomide-dependent inhibition of endothelial cell growth. Thalidomide 117-128 cullin 5 Homo sapiens 19-23 29746508-3 2018 We discovered that CUL5 (previously identified as VACM-1), a scaffold protein in E3 ligase complexes, is involved in thalidomide-dependent inhibition of endothelial cell growth. Thalidomide 117-128 cullin 5 Homo sapiens 50-56 29746508-4 2018 Our results show that in human endothelial cells (HUVEC), thalidomide-dependent decrease in cell growth was associated with decreased nuclear localization of CUL5. Thalidomide 58-69 cullin 5 Homo sapiens 158-162 29746508-7 2018 In this study, the antiproliferative response to thalidomide was compromised in RAMEC expressing mutated CUL5. Thalidomide 49-60 cullin 5 Homo sapiens 105-109 29746508-8 2018 These results suggest that CUL5 protein is involved in the thalidomide-dependent regulation of cellular proliferation in vitro. Thalidomide 59-70 cullin 5 Homo sapiens 27-31 29746508-9 2018 Consequently, CUL5 may be an important part of the mechanism for thalidomide-dependent inhibition of cellular proliferation, as well as a novel biomarker for predicting a response to thalidomide for the treatment of disorders such as multiple myeloma and HIV infection. Thalidomide 65-76 cullin 5 Homo sapiens 14-18 29746508-9 2018 Consequently, CUL5 may be an important part of the mechanism for thalidomide-dependent inhibition of cellular proliferation, as well as a novel biomarker for predicting a response to thalidomide for the treatment of disorders such as multiple myeloma and HIV infection. Thalidomide 183-194 cullin 5 Homo sapiens 14-18 29844872-2 2018 The aim of this study was to evaluate the influence of selected clinical and molecular factors including single nucleotide polymorphisms (SNPs) in CRBN gene on the efficacy of first line CTD (cyclophosphamide, thalidomide, dexamethasone) chemotherapy in patients with multiple myeloma. Thalidomide 210-221 cereblon Homo sapiens 147-151 29844872-2 2018 The aim of this study was to evaluate the influence of selected clinical and molecular factors including single nucleotide polymorphisms (SNPs) in CRBN gene on the efficacy of first line CTD (cyclophosphamide, thalidomide, dexamethasone) chemotherapy in patients with multiple myeloma. Thalidomide 210-221 CTD Homo sapiens 187-190 29844872-11 2018 In conclusion, SNPs of the CRBN gene may be useful in qualifying patients for treatment with regimens containing thalidomide. Thalidomide 113-124 cereblon Homo sapiens 27-31 29653983-5 2018 Adoptive transfer of wild-type Gr-1+CD11b+ MDSCs from tumor-bearing mice reversed this antitumor response, whereas caspase-1 inhibiting thalidomide-treated MDSCs phenocopied the antitumor response found in caspase-1 null mice. Thalidomide 136-147 caspase 1 Mus musculus 115-124 29653983-5 2018 Adoptive transfer of wild-type Gr-1+CD11b+ MDSCs from tumor-bearing mice reversed this antitumor response, whereas caspase-1 inhibiting thalidomide-treated MDSCs phenocopied the antitumor response found in caspase-1 null mice. Thalidomide 136-147 caspase 1 Mus musculus 206-215 29854627-4 2018 Thalidomide and carboplatin induced up-regulation of the expression of p53, tumor necrosis factor-alpha and interleukin-6 in brain and kidney. Thalidomide 0-11 tumor necrosis factor Homo sapiens 76-103 29854627-4 2018 Thalidomide and carboplatin induced up-regulation of the expression of p53, tumor necrosis factor-alpha and interleukin-6 in brain and kidney. Thalidomide 0-11 interleukin 6 Homo sapiens 108-121 29449372-1 2018 Upon binding to thalidomide and other immunomodulatory drugs, the E3 ligase substrate receptor cereblon (CRBN) promotes proteosomal destruction by engaging the DDB1-CUL4A-Roc1-RBX1 E3 ubiquitin ligase in human cells but not in mouse cells, suggesting that sequence variations in CRBN may cause its inactivation. Thalidomide 16-27 cereblon Mus musculus 105-109 29719394-1 2018 Introduction: Thalidomide (THA) is an angiogenesis inhibitor and an efficient inhibitor of the tumor necrosis factor-alpha (TNF-alpha). Thalidomide 14-25 tumor necrosis factor Homo sapiens 95-122 29719394-1 2018 Introduction: Thalidomide (THA) is an angiogenesis inhibitor and an efficient inhibitor of the tumor necrosis factor-alpha (TNF-alpha). Thalidomide 14-25 tumor necrosis factor Homo sapiens 124-133 29719394-1 2018 Introduction: Thalidomide (THA) is an angiogenesis inhibitor and an efficient inhibitor of the tumor necrosis factor-alpha (TNF-alpha). Thalidomide 27-30 tumor necrosis factor Homo sapiens 95-122 29719394-1 2018 Introduction: Thalidomide (THA) is an angiogenesis inhibitor and an efficient inhibitor of the tumor necrosis factor-alpha (TNF-alpha). Thalidomide 27-30 tumor necrosis factor Homo sapiens 124-133 29695106-3 2018 Additionally, BSG is implicated in response to treatment with immunomodulatory drugs (thalidomide and its derivatives). Thalidomide 86-97 basigin (Ok blood group) Homo sapiens 14-17 29449372-1 2018 Upon binding to thalidomide and other immunomodulatory drugs, the E3 ligase substrate receptor cereblon (CRBN) promotes proteosomal destruction by engaging the DDB1-CUL4A-Roc1-RBX1 E3 ubiquitin ligase in human cells but not in mouse cells, suggesting that sequence variations in CRBN may cause its inactivation. Thalidomide 16-27 damage specific DNA binding protein 1 Mus musculus 160-164 29449372-1 2018 Upon binding to thalidomide and other immunomodulatory drugs, the E3 ligase substrate receptor cereblon (CRBN) promotes proteosomal destruction by engaging the DDB1-CUL4A-Roc1-RBX1 E3 ubiquitin ligase in human cells but not in mouse cells, suggesting that sequence variations in CRBN may cause its inactivation. Thalidomide 16-27 cullin 4A Mus musculus 165-182 29449372-1 2018 Upon binding to thalidomide and other immunomodulatory drugs, the E3 ligase substrate receptor cereblon (CRBN) promotes proteosomal destruction by engaging the DDB1-CUL4A-Roc1-RBX1 E3 ubiquitin ligase in human cells but not in mouse cells, suggesting that sequence variations in CRBN may cause its inactivation. Thalidomide 16-27 cereblon Mus musculus 279-283 29449372-4 2018 With a series of recombinant thalidomide-binding domain (TBD) proteins, we show that CRBN sequence variants retain their drug-binding properties to both classical immunomodulatory drugs and dBET1, a chemical compound and targeting ligand designed to degrade bromodomain-containing 4 (BRD4) via a CRBN-dependent mechanism. Thalidomide 29-40 cereblon Mus musculus 85-89 29449372-4 2018 With a series of recombinant thalidomide-binding domain (TBD) proteins, we show that CRBN sequence variants retain their drug-binding properties to both classical immunomodulatory drugs and dBET1, a chemical compound and targeting ligand designed to degrade bromodomain-containing 4 (BRD4) via a CRBN-dependent mechanism. Thalidomide 29-40 Blocked early in transport 1 Drosophila melanogaster 190-195 29552137-7 2018 Furthermore, vincristine administration induced the upregulation of tumor necrosis factor (TNF)-alpha production in DRGs, and inhibition of TNF-alpha synthesis with thalidomide in vivo reversed TRPV1 protein expression, as well as pain hypersensitivity induced by vincristine in rats. Thalidomide 165-176 tumor necrosis factor Rattus norvegicus 140-149 29530986-8 2018 These results strongly suggest that increased BK channel activity is the pathological mechanism of intellectual disability in CRBN mutations.SIGNIFICANCE STATEMENTCereblon (CRBN), a well known target of the immunomodulatory drug thalidomide, was originally identified as a gene that causes human intellectual disability when mutated. Thalidomide 229-240 cereblon Homo sapiens 126-130 29530986-8 2018 These results strongly suggest that increased BK channel activity is the pathological mechanism of intellectual disability in CRBN mutations.SIGNIFICANCE STATEMENTCereblon (CRBN), a well known target of the immunomodulatory drug thalidomide, was originally identified as a gene that causes human intellectual disability when mutated. Thalidomide 229-240 cereblon Homo sapiens 173-177 29552137-7 2018 Furthermore, vincristine administration induced the upregulation of tumor necrosis factor (TNF)-alpha production in DRGs, and inhibition of TNF-alpha synthesis with thalidomide in vivo reversed TRPV1 protein expression, as well as pain hypersensitivity induced by vincristine in rats. Thalidomide 165-176 transient receptor potential cation channel, subfamily V, member 1 Rattus norvegicus 194-199 29632537-3 2018 Recent studies also show that thalidomide analogs, cyclophosphamide, and other small molecules are able to act on TNFR2, resulting in the elimination of TNFR2-expressing Tregs. Thalidomide 30-41 TNF receptor superfamily member 1B Homo sapiens 114-119 29632537-3 2018 Recent studies also show that thalidomide analogs, cyclophosphamide, and other small molecules are able to act on TNFR2, resulting in the elimination of TNFR2-expressing Tregs. Thalidomide 30-41 TNF receptor superfamily member 1B Homo sapiens 153-158 29496670-5 2018 Using a thalidomide analog bead pull-down assay, we showed that IMiD compounds directly bind CRBN in CD34+ cells. Thalidomide 8-19 cereblon Mus musculus 93-97 29193903-3 2018 In this study, seven polymorphisms in genes related to development (FGF8, FGF10, BMP4, SHH, TP53, TP63, and TP73) were analyzed in people with thalidomide embryopathy (TE) and compared to people without malformations. Thalidomide 143-154 fibroblast growth factor 8 Homo sapiens 68-72 29193903-3 2018 In this study, seven polymorphisms in genes related to development (FGF8, FGF10, BMP4, SHH, TP53, TP63, and TP73) were analyzed in people with thalidomide embryopathy (TE) and compared to people without malformations. Thalidomide 143-154 fibroblast growth factor 10 Homo sapiens 74-79 29193903-3 2018 In this study, seven polymorphisms in genes related to development (FGF8, FGF10, BMP4, SHH, TP53, TP63, and TP73) were analyzed in people with thalidomide embryopathy (TE) and compared to people without malformations. Thalidomide 143-154 bone morphogenetic protein 4 Homo sapiens 81-85 29193903-3 2018 In this study, seven polymorphisms in genes related to development (FGF8, FGF10, BMP4, SHH, TP53, TP63, and TP73) were analyzed in people with thalidomide embryopathy (TE) and compared to people without malformations. Thalidomide 143-154 sonic hedgehog signaling molecule Homo sapiens 87-90 29193903-3 2018 In this study, seven polymorphisms in genes related to development (FGF8, FGF10, BMP4, SHH, TP53, TP63, and TP73) were analyzed in people with thalidomide embryopathy (TE) and compared to people without malformations. Thalidomide 143-154 tumor protein p53 Homo sapiens 92-96 29193903-3 2018 In this study, seven polymorphisms in genes related to development (FGF8, FGF10, BMP4, SHH, TP53, TP63, and TP73) were analyzed in people with thalidomide embryopathy (TE) and compared to people without malformations. Thalidomide 143-154 tumor protein p63 Homo sapiens 98-102 29193903-3 2018 In this study, seven polymorphisms in genes related to development (FGF8, FGF10, BMP4, SHH, TP53, TP63, and TP73) were analyzed in people with thalidomide embryopathy (TE) and compared to people without malformations. Thalidomide 143-154 tumor protein p73 Homo sapiens 108-112 29496670-5 2018 Using a thalidomide analog bead pull-down assay, we showed that IMiD compounds directly bind CRBN in CD34+ cells. Thalidomide 8-19 CD34 antigen Mus musculus 101-105 29681766-12 2018 In addition, the TNF-alpha synthesis inhibitor thalidomide significantly attenuated the number of paclitaxel-induced TUNEL-positive neurons in the hippocampus and restored the impaired spatial learning and memory function in paclitaxel-treated rats. Thalidomide 47-58 tumor necrosis factor Rattus norvegicus 17-26 29223784-0 2018 Thalidomide attenuates development of morphine dependence in mice by inhibiting PI3K/Akt and nitric oxide signaling pathways. Thalidomide 0-11 thymoma viral proto-oncogene 1 Mus musculus 85-88 29223784-6 2018 In the present study, we aimed to explore the effect of thalidomide on the development of morphine dependence targeting PI3K/Akt (PKB) and nitric oxide (NO) pathways. Thalidomide 56-67 thymoma viral proto-oncogene 1 Mus musculus 125-128 29223784-6 2018 In the present study, we aimed to explore the effect of thalidomide on the development of morphine dependence targeting PI3K/Akt (PKB) and nitric oxide (NO) pathways. Thalidomide 56-67 AKT serine/threonine kinase 1 Homo sapiens 130-133 29223784-8 2018 In both models the consequent effect of thalidomide on PI3K/Akt and/or NO signaling in morphine dependence was determined. Thalidomide 40-51 thymoma viral proto-oncogene 1 Mus musculus 60-63 29223784-9 2018 Thalidomide alone or in combination with PI3K inhibitor, Akt inhibitor or nitric oxide synthase (NOS) inhibitors significantly reduced naloxone induced withdrawal signs in morphine dependent mice. Thalidomide 0-11 nitric oxide synthase 1, neuronal Mus musculus 74-95 29223784-11 2018 In T98G human glioblastoma cells, thalidomide alone or in combination with PI3K and Akt inhibitors significantly reduced iNOS expression in comparison to the morphine treated cells. Thalidomide 34-45 nitric oxide synthase 2 Homo sapiens 121-125 29223784-12 2018 Also, morphine-induced p-Akt was suppressed when T98G cells were pretreated with thalidomide. Thalidomide 81-92 thymoma viral proto-oncogene 1 Mus musculus 25-28 29223784-14 2018 Moreover, these data indicate that thalidomide attenuates the development of morphine dependence in vivo and in vitro by inhibition of PI3K/Akt and nitric oxide signaling pathways. Thalidomide 35-46 thymoma viral proto-oncogene 1 Mus musculus 140-143 29383859-6 2018 The pro-angiogenic effect of GDF15 was through the activation of Src and its downstream AKT, MAPK, and NF-kappaB signaling, which was blocked by thalidomide. Thalidomide 145-156 growth differentiation factor 15 Mus musculus 29-34 29383859-6 2018 The pro-angiogenic effect of GDF15 was through the activation of Src and its downstream AKT, MAPK, and NF-kappaB signaling, which was blocked by thalidomide. Thalidomide 145-156 Rous sarcoma oncogene Mus musculus 65-68 29383859-6 2018 The pro-angiogenic effect of GDF15 was through the activation of Src and its downstream AKT, MAPK, and NF-kappaB signaling, which was blocked by thalidomide. Thalidomide 145-156 thymoma viral proto-oncogene 1 Mus musculus 88-91 29410497-0 2018 The Neuroprotective Effect of Thalidomide against Ischemia through the Cereblon-mediated Repression of AMPK Activity. Thalidomide 30-41 cereblon Homo sapiens 71-79 29410497-4 2018 Here we examined the molecular mechanism of thalidomide"s neuroprotective effects, focusing on its target protein, cereblon (CRBN), and its binding protein, AMP-activated protein kinase (AMPK), which plays an important role in maintaining intracellular energy homeostasis in the brain. Thalidomide 44-55 cereblon Homo sapiens 115-123 29410497-4 2018 Here we examined the molecular mechanism of thalidomide"s neuroprotective effects, focusing on its target protein, cereblon (CRBN), and its binding protein, AMP-activated protein kinase (AMPK), which plays an important role in maintaining intracellular energy homeostasis in the brain. Thalidomide 44-55 cereblon Homo sapiens 125-129 29410497-8 2018 Furthermore, we considered that the AMPK-CRBN interaction was altered when neuroprotective action by thalidomide occurred in cells under ischemic conditions. Thalidomide 101-112 cereblon Homo sapiens 41-45 29410497-10 2018 However, when thalidomide was administered at the same time as H2O2, the binding of AMPK and CRBN was partly restored. Thalidomide 14-25 cereblon Homo sapiens 93-97 29410497-11 2018 These results suggest that thalidomide inhibits the activity of AMPK via CRBN under oxidative stress and suppresses nerve cell death. Thalidomide 27-38 cereblon Homo sapiens 73-77 29108926-3 2018 Thalidomide analogs have the ability to induce HbF production while inhibiting the release of tumor necrosis factor-alpha. Thalidomide 0-11 tumor necrosis factor Mus musculus 94-121 28786198-12 2018 Moreover, IL23R polymorphisms may contribute to poor prognosis in patients with MM treated with thalidomide and/or bortezomib. Thalidomide 96-107 interleukin 23 receptor Homo sapiens 10-15 29130642-1 2018 Thalidomide and its derivatives, lenalidomide and pomalidomide (also known as IMiDs), have significantly changed the treatment landscape of multiple myeloma, and the recent discovery of cereblon (CRBN) as their direct biological target has led to a deeper understanding of their complex mechanism of action. Thalidomide 0-11 cereblon Homo sapiens 196-200 28758232-8 2018 Finally, we found that repeated thalidomide treatment robustly enhanced CB2 receptor expression, but slightly reduced CB1 receptor expression, in the RVM. Thalidomide 32-43 cannabinoid receptor 2 Rattus norvegicus 72-75 28758232-8 2018 Finally, we found that repeated thalidomide treatment robustly enhanced CB2 receptor expression, but slightly reduced CB1 receptor expression, in the RVM. Thalidomide 32-43 cannabinoid receptor 1 Rattus norvegicus 118-121 29291352-7 2018 The immunosuppressive drug thalidomide prevented NF-kappaB from binding to the promoters of its target genes (including TNF and IFNG), and combination treatment with thalidomide and dexamethasone extended survival of mice engrafted with ECSIT-T419C-transfected ENKTL cells. Thalidomide 166-177 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 49-58 29251720-2 2018 We characterized NVP-2, a selective ATP-competitive CDK9 inhibitor, and THAL-SNS-032, a selective CDK9 degrader consisting of a CDK-binding SNS-032 ligand linked to a thalidomide derivative that binds the E3 ubiquitin ligase Cereblon (CRBN). Thalidomide 167-178 cyclin dependent kinase 9 Homo sapiens 98-102 29291352-7 2018 The immunosuppressive drug thalidomide prevented NF-kappaB from binding to the promoters of its target genes (including TNF and IFNG), and combination treatment with thalidomide and dexamethasone extended survival of mice engrafted with ECSIT-T419C-transfected ENKTL cells. Thalidomide 27-38 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 49-58 29291352-7 2018 The immunosuppressive drug thalidomide prevented NF-kappaB from binding to the promoters of its target genes (including TNF and IFNG), and combination treatment with thalidomide and dexamethasone extended survival of mice engrafted with ECSIT-T419C-transfected ENKTL cells. Thalidomide 166-177 tumor necrosis factor Mus musculus 120-123 29291352-7 2018 The immunosuppressive drug thalidomide prevented NF-kappaB from binding to the promoters of its target genes (including TNF and IFNG), and combination treatment with thalidomide and dexamethasone extended survival of mice engrafted with ECSIT-T419C-transfected ENKTL cells. Thalidomide 27-38 tumor necrosis factor Mus musculus 120-123 29291352-7 2018 The immunosuppressive drug thalidomide prevented NF-kappaB from binding to the promoters of its target genes (including TNF and IFNG), and combination treatment with thalidomide and dexamethasone extended survival of mice engrafted with ECSIT-T419C-transfected ENKTL cells. Thalidomide 27-38 interferon gamma Mus musculus 128-132 29291352-7 2018 The immunosuppressive drug thalidomide prevented NF-kappaB from binding to the promoters of its target genes (including TNF and IFNG), and combination treatment with thalidomide and dexamethasone extended survival of mice engrafted with ECSIT-T419C-transfected ENKTL cells. Thalidomide 166-177 interferon gamma Mus musculus 128-132 29291352-7 2018 The immunosuppressive drug thalidomide prevented NF-kappaB from binding to the promoters of its target genes (including TNF and IFNG), and combination treatment with thalidomide and dexamethasone extended survival of mice engrafted with ECSIT-T419C-transfected ENKTL cells. Thalidomide 27-38 ECSIT signalling integrator Mus musculus 237-242 29291352-7 2018 The immunosuppressive drug thalidomide prevented NF-kappaB from binding to the promoters of its target genes (including TNF and IFNG), and combination treatment with thalidomide and dexamethasone extended survival of mice engrafted with ECSIT-T419C-transfected ENKTL cells. Thalidomide 166-177 ECSIT signalling integrator Mus musculus 237-242 29291352-8 2018 We added thalidomide to the conventional dexamethasone-containing therapy regimen for two patients with HPS who expressed ECSIT-V140A, and we observed reversal of their HPS and disease-free survival for longer than 3 years. Thalidomide 9-20 ECSIT signaling integrator Homo sapiens 122-127 28365861-4 2018 Application of the TNF-synthesis inhibitor thalidomide and the TNF-alpha antagonist etanercept dose-dependently suppressed acute itch. Thalidomide 43-54 itchy, E3 ubiquitin protein ligase Mus musculus 129-133 28365861-7 2018 Furthermore, chronic itch induced by dry skin was reduced by administration of thalidomide and etanercept and in TNFR1/R2 DKO mice. Thalidomide 79-90 itchy, E3 ubiquitin protein ligase Mus musculus 21-25 29397841-6 2018 The ratio of Th17 cells in peripheral blood and serum levels of IL-17 in patients with multiple myeloma after treatment with thalidomide were significantly lower than those before treatment, and the ratio of Treg cells and levels of IL-35 were significantly higher than those before treatment, and the ratio of Th17 / Treg cells was higher than that before treatment (P<0.05). Thalidomide 125-136 interleukin 17A Homo sapiens 64-69 29108922-2 2018 In our previously clinical practice, we observed that thalidomide, a potent VEGF inhibitor, can significantly decrease the tumor size of one EGFR-TKI resistance patient with lung cancer cachexia. Thalidomide 54-65 vascular endothelial growth factor A Homo sapiens 76-80 29108922-2 2018 In our previously clinical practice, we observed that thalidomide, a potent VEGF inhibitor, can significantly decrease the tumor size of one EGFR-TKI resistance patient with lung cancer cachexia. Thalidomide 54-65 epidermal growth factor receptor Homo sapiens 141-145 29397841-9 2018 The anti-MM mechanism of thalidomide may relate with the regulation of Th17 / Treg cell ratio and expression levels of IL-17 and IL-35. Thalidomide 25-36 interleukin 17A Homo sapiens 119-124 29513882-1 2018 In the present study, we successfully developed a docetaxel (DTX) and thalidomide (TDD) co-delivery system based on low density lipoprotein (LDL) modified silica nanoparticles (LDL/SLN/DTX/TDD). Thalidomide 70-81 aldo-keto reductase family 1 member C2 Homo sapiens 83-86 28379698-1 2018 Here we report the development of a proteolysis targeting chimera (PROTAC) based on the combination of the unique features of the sirtuin rearranging ligands (SirReals) as highly potent and isotype-selective Sirt2 inhibitors with thalidomide, a bona fide cereblon ligand. Thalidomide 230-241 sirtuin 2 Homo sapiens 208-213 29732020-6 2018 The rele-vance of TNF-alpha to the pathogenesis of RAS has stemmed from the observations that anti- TNF-alpha drugs such as thalidomide and pentoxifylline have been found to be effective in the treatment of RAS. Thalidomide 124-135 tumor necrosis factor Homo sapiens 18-27 29386886-9 2018 Compared with diabetic control rats, those treated with thalidomide showed significantly improved histological alterations and biomarkers of renal function, as well as reduced expression of renal inflammatory cytokines, including NF-kappaB and MCP-1. Thalidomide 56-67 mast cell protease 1-like 1 Rattus norvegicus 244-249 29358579-0 2018 Structural basis of thalidomide enantiomer binding to cereblon. Thalidomide 20-31 cereblon Danio rerio 54-62 29358579-3 2018 Here, we present structural and biochemical studies of (S)- and (R)-enantiomers bound to the primary target of thalidomide, cereblon (CRBN). Thalidomide 111-122 cereblon Danio rerio 124-132 29358579-3 2018 Here, we present structural and biochemical studies of (S)- and (R)-enantiomers bound to the primary target of thalidomide, cereblon (CRBN). Thalidomide 111-122 cereblon Danio rerio 134-138 29358579-5 2018 The crystal structures of the thalidomide-binding domain of CRBN bound to each enantiomer show that both enantiomers bind the tri-Trp pocket, although the bound form of the (S)-enantiomer exhibited a more relaxed glutarimide ring conformation. Thalidomide 30-41 cereblon Danio rerio 60-64 28982338-0 2018 Thalidomide Improves Psoriasis-like Lesions and Inhibits Cutaneous VEGF Expression without Alteration of Microvessel Density in Imiquimod- induced Psoriatic Mouse Model. Thalidomide 0-11 vascular endothelial growth factor A Mus musculus 67-71 28982338-3 2018 Thalidomide has been reported being able to inhibit the effects of fibroblast growth factor 2 and vascular endothelial growth factor (VEGF), and inhibit tumour necrosis factor-alpha synthesis, and suppress tumour necrosis factor-induced nuclear factor-kappa B activation in Jurkat cells, resulting in suppression of proliferation inflammation, angiogenesis, and the immune system, which are related to the pathogenesis of psoriasis. Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 67-93 28982338-3 2018 Thalidomide has been reported being able to inhibit the effects of fibroblast growth factor 2 and vascular endothelial growth factor (VEGF), and inhibit tumour necrosis factor-alpha synthesis, and suppress tumour necrosis factor-induced nuclear factor-kappa B activation in Jurkat cells, resulting in suppression of proliferation inflammation, angiogenesis, and the immune system, which are related to the pathogenesis of psoriasis. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 98-132 28982338-3 2018 Thalidomide has been reported being able to inhibit the effects of fibroblast growth factor 2 and vascular endothelial growth factor (VEGF), and inhibit tumour necrosis factor-alpha synthesis, and suppress tumour necrosis factor-induced nuclear factor-kappa B activation in Jurkat cells, resulting in suppression of proliferation inflammation, angiogenesis, and the immune system, which are related to the pathogenesis of psoriasis. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 134-138 28982338-10 2018 The cutaneous VEGF protein levels down-expressed significantly in the moderate- and high-dose thalidomide groups (p<0.05 for both), while those in the low-dose thalidomide and acitretin did not (p>0.05 for both). Thalidomide 94-105 vascular endothelial growth factor A Mus musculus 14-18 28982338-12 2018 CONCLUSION: Thalidomide can improve the psoriasis-like lesions and inhibit the expression of cutaneous VEGF in imiquimod-induced psoriatic model with dose-dependence, however, it does not alter circulating VEGF-A levels and microvessel density in dermis. Thalidomide 12-23 vascular endothelial growth factor A Mus musculus 103-107 29732020-6 2018 The rele-vance of TNF-alpha to the pathogenesis of RAS has stemmed from the observations that anti- TNF-alpha drugs such as thalidomide and pentoxifylline have been found to be effective in the treatment of RAS. Thalidomide 124-135 tumor necrosis factor Homo sapiens 100-109 29272525-6 2017 Thalidomide treatment reduced PTX3 in the serum 7 days after starting treatment. Thalidomide 0-11 pentraxin 3 Homo sapiens 30-34 30175757-5 2018 To recruit the von Hippel-Lindau (VHL) E3 ligase complex and the cereblon (CRBN) E3 ligase complex, a VHL inhibitor and a thalidomide derivative have been integrated into PROTAC and Degronimid constructs, respectively. Thalidomide 122-133 cereblon Homo sapiens 75-79 29272525-7 2017 In situ analyses have also demonstrated enhancement of PTX3 in ENL lesions and showed that treatment with thalidomide reduced its expression and the prominent neutrophilic infiltrate, a hallmark of the disease. Thalidomide 106-117 pentraxin 3 Homo sapiens 55-59 29172279-0 2017 Anti-Vascular Endothelial Growth Factor Targeting by Curcumin and Thalidomide in Acute Myeloid Leukemia Cells Acute myeloid leukemias (AMLs) are blood disorders that exhibit uncontrolled growth and reduction of apoptosisrates. Thalidomide 66-77 vascular endothelial growth factor A Homo sapiens 5-39 27405747-0 2017 Polymorphism of IL-10 receptor beta affects the prognosis of multiple myeloma patients treated with thalidomide and/or bortezomib. Thalidomide 100-111 interleukin 10 Homo sapiens 16-21 27405747-12 2017 In particular, IL-10RB K47E polymorphism may contribute to the poor prognosis of MM patients treated with thalidomide and/or bortezomib. Thalidomide 106-117 interleukin 10 receptor subunit beta Homo sapiens 15-22 29344159-7 2017 P38 cDNA was transfected into esophageal cancer cells to assess tumor cell viability, sensitivity to thalidomide treatment and IDO gene expression. Thalidomide 101-112 mitogen-activated protein kinase 14 Homo sapiens 0-3 29344159-10 2017 In vitro experiments revealed that the expression of p-p38 induced esophageal cancer Eca-109 and TE-1 cell viability, and resistance to thalidomide treatment, as well as in the expression of IDO without the application of lipopolysaccharides. Thalidomide 136-147 mitogen-activated protein kinase 14 Homo sapiens 55-58 29238068-7 2017 Thalidomide, a TNF-alpha and NF-kappaB inhibitor, significantly weakened the metastasis and homing capacity of miR-105-overexpressed CRC cells in nude mice. Thalidomide 0-11 tumor necrosis factor Mus musculus 15-24 29238068-7 2017 Thalidomide, a TNF-alpha and NF-kappaB inhibitor, significantly weakened the metastasis and homing capacity of miR-105-overexpressed CRC cells in nude mice. Thalidomide 0-11 microRNA 105 Mus musculus 111-118 29172279-11 2017 Thalidomide as a VEGF inhibitor in combination with curcumin appears to have a synergistic impacton inhibition of cell proliferation and promotion of apoptosis. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 17-21 28869897-9 2017 Single administration of thalidomide significantly reduced production of cytokines (tumor necrosis factor (TNF)-alpha and interleukin (IL)-1beta) in serum but not in the brain, and attenuated upregulation of NMDA receptor (NR) 2A/B subunits in the hippocampus at POD 14. Thalidomide 25-36 tumor necrosis factor Rattus norvegicus 84-117 29116196-0 2017 Thalidomide Inhibits TGF-beta1-induced Epithelial to Mesenchymal Transition in Alveolar Epithelial Cells via Smad-Dependent and Smad-Independent Signaling Pathways. Thalidomide 0-11 transforming growth factor beta 1 Homo sapiens 21-30 29116196-4 2017 CCL-149 cells were treated with TGF-beta1 in the presence of THL at the indicated concentrations. Thalidomide 61-64 transforming growth factor beta 1 Homo sapiens 32-41 29116196-7 2017 THL inhibited the TGF-beta1 induction of alpha-SMA, vimentin, MMP-2/-9 and collagen type IV expression and restored the morphological changes in primary alveolar epithelial cells caused by TGF-beta1. Thalidomide 0-3 transforming growth factor beta 1 Homo sapiens 18-27 29116196-7 2017 THL inhibited the TGF-beta1 induction of alpha-SMA, vimentin, MMP-2/-9 and collagen type IV expression and restored the morphological changes in primary alveolar epithelial cells caused by TGF-beta1. Thalidomide 0-3 vimentin Homo sapiens 52-60 29116196-7 2017 THL inhibited the TGF-beta1 induction of alpha-SMA, vimentin, MMP-2/-9 and collagen type IV expression and restored the morphological changes in primary alveolar epithelial cells caused by TGF-beta1. Thalidomide 0-3 matrix metallopeptidase 2 Homo sapiens 62-70 29116196-7 2017 THL inhibited the TGF-beta1 induction of alpha-SMA, vimentin, MMP-2/-9 and collagen type IV expression and restored the morphological changes in primary alveolar epithelial cells caused by TGF-beta1. Thalidomide 0-3 transforming growth factor beta 1 Homo sapiens 189-198 29116196-9 2017 Moreover, THL inhibited TGF-beta1-induced phosphorylation of p38, JNK, ERK, Akt, GSK3beta, Smad 2 and Smad3 without altering the total expression levels of those proteins. Thalidomide 10-13 transforming growth factor beta 1 Homo sapiens 24-33 29116196-9 2017 Moreover, THL inhibited TGF-beta1-induced phosphorylation of p38, JNK, ERK, Akt, GSK3beta, Smad 2 and Smad3 without altering the total expression levels of those proteins. Thalidomide 10-13 mitogen-activated protein kinase 14 Homo sapiens 61-64 29116196-9 2017 Moreover, THL inhibited TGF-beta1-induced phosphorylation of p38, JNK, ERK, Akt, GSK3beta, Smad 2 and Smad3 without altering the total expression levels of those proteins. Thalidomide 10-13 mitogen-activated protein kinase 8 Homo sapiens 66-69 29116196-9 2017 Moreover, THL inhibited TGF-beta1-induced phosphorylation of p38, JNK, ERK, Akt, GSK3beta, Smad 2 and Smad3 without altering the total expression levels of those proteins. Thalidomide 10-13 mitogen-activated protein kinase 1 Homo sapiens 71-74 29116196-9 2017 Moreover, THL inhibited TGF-beta1-induced phosphorylation of p38, JNK, ERK, Akt, GSK3beta, Smad 2 and Smad3 without altering the total expression levels of those proteins. Thalidomide 10-13 AKT serine/threonine kinase 1 Homo sapiens 76-79 29116196-9 2017 Moreover, THL inhibited TGF-beta1-induced phosphorylation of p38, JNK, ERK, Akt, GSK3beta, Smad 2 and Smad3 without altering the total expression levels of those proteins. Thalidomide 10-13 glycogen synthase kinase 3 beta Homo sapiens 81-89 29116196-9 2017 Moreover, THL inhibited TGF-beta1-induced phosphorylation of p38, JNK, ERK, Akt, GSK3beta, Smad 2 and Smad3 without altering the total expression levels of those proteins. Thalidomide 10-13 SMAD family member 2 Homo sapiens 91-97 29116196-9 2017 Moreover, THL inhibited TGF-beta1-induced phosphorylation of p38, JNK, ERK, Akt, GSK3beta, Smad 2 and Smad3 without altering the total expression levels of those proteins. Thalidomide 10-13 SMAD family member 3 Homo sapiens 102-107 29116196-10 2017 These findings indicate that TGF-beta1-induced EMT in alveolar epithelial cells is inhibited by THL via both Smad-dependent and non-Smad-dependent signaling pathways and suggests therapeutic approaches for targeting this process in pulmonary fibrosis. Thalidomide 96-99 transforming growth factor beta 1 Homo sapiens 29-38 28869897-9 2017 Single administration of thalidomide significantly reduced production of cytokines (tumor necrosis factor (TNF)-alpha and interleukin (IL)-1beta) in serum but not in the brain, and attenuated upregulation of NMDA receptor (NR) 2A/B subunits in the hippocampus at POD 14. Thalidomide 25-36 interleukin 1 beta Rattus norvegicus 122-144 28993730-8 2017 However, targeting the altered biological pathways (mTOR, PDGFRB, FGF2, HDAC) guided identification of possibly beneficial treatment with a combination of sirolimus, thalidomide, sunitinib, and vorinostat. Thalidomide 166-177 mechanistic target of rapamycin kinase Homo sapiens 52-56 28672061-8 2017 Measurement of intragraft cytokine levels after thalidomide treatment revealed downregulation of matrix metalloproteinase 8 and monocyte chemotactic protein 1, cytokines involved in tissue remodelling and inflammation, respectively. Thalidomide 48-59 matrix metallopeptidase 8 Rattus norvegicus 97-123 28672061-8 2017 Measurement of intragraft cytokine levels after thalidomide treatment revealed downregulation of matrix metalloproteinase 8 and monocyte chemotactic protein 1, cytokines involved in tissue remodelling and inflammation, respectively. Thalidomide 48-59 C-C motif chemokine ligand 2 Rattus norvegicus 128-158 28538510-3 2017 Thalidomide, a drug known for its immunomodulating and antiangiogenic properties, has recently been demonstrated to induce gamma-globin gene expression and to increase the proliferation of erythroid cells. Thalidomide 0-11 hemoglobin subunit gamma 1 Homo sapiens 123-135 29151411-13 2017 CONCLUSIONS: Thalidomide has a protective effect on ALI induced by PQ poisoning in rats in a dose-dependent manner, the mechanism may be achieved by reducing the level of oxygen free radicals, reducing the inflammatory factor and inhibiting the IkappaB-alpha/NF-kappaB signal pathway activation. Thalidomide 13-24 NFKB inhibitor alpha Rattus norvegicus 245-258 28902348-4 2017 Silencing CUL4A expression or CUL4A inhibition by thalidomide suppressed the EMT process induced by TGF-beta1. Thalidomide 50-61 cullin 4A Homo sapiens 30-35 28902348-4 2017 Silencing CUL4A expression or CUL4A inhibition by thalidomide suppressed the EMT process induced by TGF-beta1. Thalidomide 50-61 transforming growth factor beta 1 Homo sapiens 100-109 28993730-8 2017 However, targeting the altered biological pathways (mTOR, PDGFRB, FGF2, HDAC) guided identification of possibly beneficial treatment with a combination of sirolimus, thalidomide, sunitinib, and vorinostat. Thalidomide 166-177 histone deacetylase 9 Homo sapiens 72-76 28636891-2 2017 Thalidomide or bortezomib may be combined with cyclophosphamide and dexamethasone, in what are known as the CTD and VCD protocols, respectively. Thalidomide 0-11 CTD Homo sapiens 108-111 28822719-11 2017 During thalidomide treatment, a decrease in CD4/CD8 ratio (p=0.04), a decrease in CD4 count (p=0.04), an increase in cell activation calculated by the percentage of CD38 +/HLA-DR+ CD8 cells (p<0.05) and an increase in US-CRP (p<0.01) were observed in the Thalidomide group, with all parameters returning to baseline levels after thalidomide interruption. Thalidomide 7-18 CD4 molecule Homo sapiens 44-47 28822719-11 2017 During thalidomide treatment, a decrease in CD4/CD8 ratio (p=0.04), a decrease in CD4 count (p=0.04), an increase in cell activation calculated by the percentage of CD38 +/HLA-DR+ CD8 cells (p<0.05) and an increase in US-CRP (p<0.01) were observed in the Thalidomide group, with all parameters returning to baseline levels after thalidomide interruption. Thalidomide 7-18 CD8a molecule Homo sapiens 48-51 28822719-11 2017 During thalidomide treatment, a decrease in CD4/CD8 ratio (p=0.04), a decrease in CD4 count (p=0.04), an increase in cell activation calculated by the percentage of CD38 +/HLA-DR+ CD8 cells (p<0.05) and an increase in US-CRP (p<0.01) were observed in the Thalidomide group, with all parameters returning to baseline levels after thalidomide interruption. Thalidomide 7-18 CD4 molecule Homo sapiens 82-85 28822719-11 2017 During thalidomide treatment, a decrease in CD4/CD8 ratio (p=0.04), a decrease in CD4 count (p=0.04), an increase in cell activation calculated by the percentage of CD38 +/HLA-DR+ CD8 cells (p<0.05) and an increase in US-CRP (p<0.01) were observed in the Thalidomide group, with all parameters returning to baseline levels after thalidomide interruption. Thalidomide 7-18 CD38 molecule Homo sapiens 165-169 28822719-11 2017 During thalidomide treatment, a decrease in CD4/CD8 ratio (p=0.04), a decrease in CD4 count (p=0.04), an increase in cell activation calculated by the percentage of CD38 +/HLA-DR+ CD8 cells (p<0.05) and an increase in US-CRP (p<0.01) were observed in the Thalidomide group, with all parameters returning to baseline levels after thalidomide interruption. Thalidomide 7-18 CD8a molecule Homo sapiens 180-183 28822719-14 2017 CONCLUSIONS: Short-term use of thalidomide led to an intense transient increase in T cell activation and inflammation, with a decrease in the CD4+ cell count without changes to the CD8+ cell count. Thalidomide 31-42 CD4 molecule Homo sapiens 142-145 28822719-14 2017 CONCLUSIONS: Short-term use of thalidomide led to an intense transient increase in T cell activation and inflammation, with a decrease in the CD4+ cell count without changes to the CD8+ cell count. Thalidomide 31-42 CD8a molecule Homo sapiens 181-184 28500554-1 2017 PURPOSE: Circularly permuted TRAIL (CPT) has exhibited promising efficacy as a mono-therapy or in combination with thalidomide for patients with multiple myeloma (MM). Thalidomide 115-126 TNF superfamily member 10 Homo sapiens 29-34 28737650-0 2017 Thalidomide in Transfusion Dependent Thalassemia: Hope or Hype. Thalidomide 0-11 FIC domain protein adenylyltransferase Homo sapiens 58-62 28665417-0 2017 VEGF, VEGFR2 and GSTM1 polymorphisms in outcome of multiple myeloma patients treated with thalidomide-based regimens. Thalidomide 90-101 vascular endothelial growth factor A Homo sapiens 0-4 28665417-0 2017 VEGF, VEGFR2 and GSTM1 polymorphisms in outcome of multiple myeloma patients treated with thalidomide-based regimens. Thalidomide 90-101 glutathione S-transferase mu 1 Homo sapiens 17-22 27707692-1 2017 AIMS: To determine the changes in the subfoveal choroidal thickness (CT), the foveal thickness (FT) and the serum level of vascular endothelial growth factor (VEGF) after thalidomide treatment in patients with polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy and skin changes (POEMS) syndrome. Thalidomide 171-182 vascular endothelial growth factor A Homo sapiens 123-157 27707692-1 2017 AIMS: To determine the changes in the subfoveal choroidal thickness (CT), the foveal thickness (FT) and the serum level of vascular endothelial growth factor (VEGF) after thalidomide treatment in patients with polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy and skin changes (POEMS) syndrome. Thalidomide 171-182 vascular endothelial growth factor A Homo sapiens 159-163 28811745-4 2017 The results showed a role for direct and indirect TNF-alpha inhibition through agents such as thalidomide, 3,6-dithiothalidomide, etanercept, infliximab, exendin-4, sodium hydrosulfide, minocycline, imipramine, and atorvastatin. Thalidomide 94-105 tumor necrosis factor Rattus norvegicus 50-59 28161597-6 2017 The STRING-tool suggested nitric oxide and beta-catenin as the central angiogenesis-related molecules affected by thalidomide"s antiangiogenic property. Thalidomide 114-125 catenin beta 1 Homo sapiens 43-55 28409562-5 2017 Interestingly, abrogation of AIM2 signalling, either in AIM2-deficient mice or by a pharmacological inhibitor such as thalidomide, significantly reduces the incidence of drug-induced diarrhoea without affecting the anticancer efficacy of CPT-11. Thalidomide 118-129 absent in melanoma 2 Mus musculus 29-33 28422447-5 2017 We also revealed that monohydrolyzed and monohydroxylated metabolites of thalidomide were likely to generate mainly by a C-5 oxidation of thalidomide and subsequent ring opening of the hydroxylated metabolite. Thalidomide 73-84 complement C5 Homo sapiens 121-124 28422447-5 2017 We also revealed that monohydrolyzed and monohydroxylated metabolites of thalidomide were likely to generate mainly by a C-5 oxidation of thalidomide and subsequent ring opening of the hydroxylated metabolite. Thalidomide 138-149 complement C5 Homo sapiens 121-124 28521705-5 2017 Here we report a case of DVT induced after 8 months of use of thalidomide in a young patient of 22 years age suffering from ENL. Thalidomide 62-73 MLLT1 super elongation complex subunit Homo sapiens 124-127 28415757-8 2017 However, while mice injected with thalidomide displayed no increased surface expression of platelet P-selectin or alphaIIbbeta3, there was a significantly shortened tail bleeding time, thrombin time, prothrombin time together with higher levels of Factor IX and fibrinogen. Thalidomide 34-45 coagulation factor II Mus musculus 185-193 28469241-0 2017 A HAND to TBX5 Explains the Link Between Thalidomide and Cardiac Diseases. Thalidomide 41-52 T-box transcription factor 5 Homo sapiens 10-14 27753513-5 2017 Furthermore, ubiquitin ligase inhibitor, thalidomide, was added in culture process, and resulted in efficient and stable secretion of HSA-HGF (R494E) in CHO cells. Thalidomide 41-52 hepatocyte growth factor Cricetulus griseus 138-141 28469241-4 2017 We used both in silico docking software, and in vitro molecular and biochemical methods to document a novel interaction involving Thalidomide, TBX5, and HAND2. Thalidomide 130-141 T-box transcription factor 5 Homo sapiens 143-147 28469241-4 2017 We used both in silico docking software, and in vitro molecular and biochemical methods to document a novel interaction involving Thalidomide, TBX5, and HAND2. Thalidomide 130-141 heart and neural crest derivatives expressed 2 Homo sapiens 153-158 28469241-5 2017 Thalidomide binds readily to TBX5 through amino acids R81, R82, and K226 all implicated in DNA binding. Thalidomide 0-11 T-box transcription factor 5 Homo sapiens 29-33 28469241-8 2017 Similarly, thalidomide inhibited the TBX5/HAND2 physical interaction, and the in silico docking revealed that the same amino acids involved in the interaction of TBX5 with DNA are also involved in its binding to HAND2. Thalidomide 11-22 T-box transcription factor 5 Homo sapiens 37-41 28469241-8 2017 Similarly, thalidomide inhibited the TBX5/HAND2 physical interaction, and the in silico docking revealed that the same amino acids involved in the interaction of TBX5 with DNA are also involved in its binding to HAND2. Thalidomide 11-22 heart and neural crest derivatives expressed 2 Homo sapiens 42-47 28469241-8 2017 Similarly, thalidomide inhibited the TBX5/HAND2 physical interaction, and the in silico docking revealed that the same amino acids involved in the interaction of TBX5 with DNA are also involved in its binding to HAND2. Thalidomide 11-22 T-box transcription factor 5 Homo sapiens 162-166 28469241-8 2017 Similarly, thalidomide inhibited the TBX5/HAND2 physical interaction, and the in silico docking revealed that the same amino acids involved in the interaction of TBX5 with DNA are also involved in its binding to HAND2. Thalidomide 11-22 heart and neural crest derivatives expressed 2 Homo sapiens 212-217 28124585-3 2017 Pre-clinical data obtained in APP23 mice suggested that the anti-cancer drug thalidomide decreases brainBACE1 and Abeta levels. Thalidomide 77-88 amyloid beta (A4) precursor protein Mus musculus 114-119 28300056-7 2017 The intracerebroventricular (ICV) injection of PGRN or intraperitoneal injection of TNFalpha synthesis blocker thalidomide (25 mg/kg), prevented the memory impairment and anxiety behaviors induced by S-DEP. Thalidomide 111-122 tumor necrosis factor Mus musculus 84-92 28099781-4 2017 Herein we report some effects on the growth and progression of MCF-7 and MDA-MB-231 breast cancer cells by a small series of thalidomide-correlated compounds, which are very effective at inducing cancer cell death by triggering TNFalpha-mediated apoptosis. Thalidomide 125-136 tumor necrosis factor Homo sapiens 228-236 27889230-0 2017 Anticancer effects of novel thalidomide analogs in A549 cells through inhibition of vascular endothelial growth factor and matrix metalloproteinase-2. Thalidomide 28-39 matrix metallopeptidase 2 Homo sapiens 123-149 28516791-8 2017 TNF-alpha levels were found statistically significantly decreased in LPS+etanercept and LPS+thalidomide+etanercept treated groups when compared with LPS group (p < 0.05). Thalidomide 92-103 tumor necrosis factor Rattus norvegicus 0-9 28516791-9 2017 For IL-1beta and IL-6 levels a statistically significant decline was observed in the LPS+thalidomide and LPS+etanercept treated groups compared to the LPS group (p < 0.05). Thalidomide 89-100 interleukin 1 beta Rattus norvegicus 4-12 28516791-9 2017 For IL-1beta and IL-6 levels a statistically significant decline was observed in the LPS+thalidomide and LPS+etanercept treated groups compared to the LPS group (p < 0.05). Thalidomide 89-100 interleukin 6 Rattus norvegicus 17-21 28298557-2 2017 An increased understanding of the molecular mechanism of thalidomide and its analogues following their clinical use has unlocked small-molecule modulation of the substrate specificity of the E3 ligase cereblon (CRBN), which in turn has resulted in the advancement of new immunomodulatory drugs (IMiDs) into the clinic. Thalidomide 57-68 cereblon Homo sapiens 211-215 27750412-2 2017 Thalidomide exerts its various pharmacological and toxic actions in primates through multiple mechanisms, including nonspecific modification of many protein networks after bioactivation by autoinduced human P450 enzymes. Thalidomide 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 207-211 27899254-0 2017 Thalidomide attenuates the development and expression of antinociceptive tolerance to mu-opioid agonist morphine through l-arginine-iNOS and nitric oxide pathway. Thalidomide 0-11 nitric oxide synthase 2 Homo sapiens 132-136 27899254-11 2017 In addition, thalidomide (20muM) also significantly inhibited the overexpression of iNOS gene induced by morphine (2.5muM) in T98G cell line. Thalidomide 13-24 nitric oxide synthase 2 Homo sapiens 84-88 27899254-13 2017 It is also evident that this effect of thalidomide is induced by the inhibition of NOS enzyme predominantly iNOS. Thalidomide 39-50 nitric oxide synthase 2 Homo sapiens 108-112 28009008-0 2016 Down-regulation of common NFkappaB-iNOS pathway by chronic Thalidomide treatment improves Hepatopulmonary Syndrome and Muscle Wasting in rats with Biliary Cirrhosis. Thalidomide 59-70 nitric oxide synthase 2 Rattus norvegicus 35-39 28496040-6 2017 Thalidomide significantly induced P450 3A4/5, 2B6, and pregnane X receptor (PXR) mRNA levels 2 to 3-fold, but rifampicin only enhanced P450 3A5 and PXR mRNA under the modified media conditions. Thalidomide 0-11 nuclear receptor subfamily 1 group I member 2 Homo sapiens 55-74 28496040-6 2017 Thalidomide significantly induced P450 3A4/5, 2B6, and pregnane X receptor (PXR) mRNA levels 2 to 3-fold, but rifampicin only enhanced P450 3A5 and PXR mRNA under the modified media conditions. Thalidomide 0-11 nuclear receptor subfamily 1 group I member 2 Homo sapiens 76-79 28571559-3 2017 Through an increase in the degradation of TNFalpha-mRNA, thalidomide reduces the production of TNFalpha by monocytes and macrophages stimulated by lipopolysaccharide or by T lymphocytes induced by mitogenic stimuli. Thalidomide 57-68 tumor necrosis factor Homo sapiens 42-50 28571559-3 2017 Through an increase in the degradation of TNFalpha-mRNA, thalidomide reduces the production of TNFalpha by monocytes and macrophages stimulated by lipopolysaccharide or by T lymphocytes induced by mitogenic stimuli. Thalidomide 57-68 tumor necrosis factor Homo sapiens 95-103 28571559-5 2017 Furthermore, thalidomide affects VEGF levels by down-regulating its expression. Thalidomide 13-24 vascular endothelial growth factor A Homo sapiens 33-37 28009008-1 2016 Thalidomide can modulate the TNFalpha-NFkappaB and iNOS pathway, which involve in the pathogenesis of hepatopulmonary syndrome (HPS) and muscle wasting in cirrhosis. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 29-37 28009008-1 2016 Thalidomide can modulate the TNFalpha-NFkappaB and iNOS pathway, which involve in the pathogenesis of hepatopulmonary syndrome (HPS) and muscle wasting in cirrhosis. Thalidomide 0-11 nitric oxide synthase 2 Rattus norvegicus 51-55 28009008-4 2016 The in vitro data demonstrated an iNOS-dependent inhibition of thalidomide on the TNFalpha-stimulated angiogenesis and myogenesis in human pulmonary artery endothelial cells (HPAECs) and C2C12 myoblasts. Thalidomide 63-74 inositol-3-phosphate synthase 1 Homo sapiens 34-38 28009008-4 2016 The in vitro data demonstrated an iNOS-dependent inhibition of thalidomide on the TNFalpha-stimulated angiogenesis and myogenesis in human pulmonary artery endothelial cells (HPAECs) and C2C12 myoblasts. Thalidomide 63-74 tumor necrosis factor Rattus norvegicus 82-90 28009008-6 2016 Our findings demonstrate that TNFalpha inhibitor thalidomide markedly diminishes the severity of experimental HPS and muscle wasting by down-regulation of common peripheral and local NFkappaB-iNOS pathway. Thalidomide 49-60 tumor necrosis factor Rattus norvegicus 30-38 28009008-6 2016 Our findings demonstrate that TNFalpha inhibitor thalidomide markedly diminishes the severity of experimental HPS and muscle wasting by down-regulation of common peripheral and local NFkappaB-iNOS pathway. Thalidomide 49-60 nitric oxide synthase 2 Rattus norvegicus 192-196 27991432-5 2016 The concommitant use of omeprazole/esomeprazole, therefore, could have critical clinical relevance in individualizing medications metabolized primarily by CYP2C19 such as PPI, clopidogrel, phenytoin, cyclophosphamide, thalidomide, citalopram, clonazepam, diazepam, proguanil, tivantinib etc. Thalidomide 218-229 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 155-162 27618360-6 2016 RESULTS: A treatment response (complete remission, very good partial remission, partial remission) was achieved in 83.7% of the patients and correlated with high CRBN expression (P = .006), mainly in the patients treated with thalidomide (P = .028). Thalidomide 226-237 cereblon Homo sapiens 162-166 27492707-5 2016 In addition, thalidomide and its analogs inhibit release of tumor necrosis factor-alpha and increase interleukin-2 (IL-2) and interferon-gamma release from T cells. Thalidomide 13-24 tumor necrosis factor Homo sapiens 60-87 27492707-5 2016 In addition, thalidomide and its analogs inhibit release of tumor necrosis factor-alpha and increase interleukin-2 (IL-2) and interferon-gamma release from T cells. Thalidomide 13-24 interleukin 2 Homo sapiens 101-114 27492707-5 2016 In addition, thalidomide and its analogs inhibit release of tumor necrosis factor-alpha and increase interleukin-2 (IL-2) and interferon-gamma release from T cells. Thalidomide 13-24 interleukin 2 Homo sapiens 116-120 27492707-5 2016 In addition, thalidomide and its analogs inhibit release of tumor necrosis factor-alpha and increase interleukin-2 (IL-2) and interferon-gamma release from T cells. Thalidomide 13-24 interferon gamma Homo sapiens 126-142 27492707-6 2016 The underlying molecular mechanisms for these pleiotropic effects remained obscure until the identification of the cereblon (CRBN) E3 ubiquitin ligase as the primary target of thalidomide and its analogs in 2010. Thalidomide 176-187 cereblon Homo sapiens 125-129 27492707-7 2016 Binding of thalidomide or lenalidomide increases the affinity of CRBN to the transcription factors IKZF1 and IKZF3 and casein kinase 1alpha (CK1alpha). Thalidomide 11-22 cereblon Homo sapiens 65-69 27573481-6 2016 Finally, STZ-induced diabetes promoted PKC activation and enhanced TNFalpha level in the spinal cord, which were attenuated by intrathecal administrations of thalidomide. Thalidomide 158-169 tumor necrosis factor Rattus norvegicus 67-75 27573481-7 2016 Taken together, these results suggested that thalidomide may potentiate morphine efficacy on diabetic neuropathy and prevent the development of morphine tolerance by suppressing PKC activation and TNFalpha level in the spinal cord. Thalidomide 45-56 tumor necrosis factor Rattus norvegicus 197-205 27702999-3 2016 Furthermore, human CRBN was identified as the primary target of thalidomide teratogenicity. Thalidomide 64-75 cereblon Homo sapiens 19-23 28083618-8 2016 The CRBN (rs711613) A allele-carriers were better responders to the treatment (P = 0.012), in particular to thalidomide including therapy (P = 0.023). Thalidomide 108-119 cereblon Homo sapiens 4-8 27748909-3 2016 Thalidomide was added to osteosarcoma cells and studied by cytotoxicity assay, evaluating apoptosis, cell cycle arrest, mitochondrial membrane potential (DeltaPsim), and reactive oxygen species (ROS) levels and the expression of Bcl-2, Bax, caspase-3 and NF-kappaB. Thalidomide 0-11 BCL2 apoptosis regulator Homo sapiens 229-234 27748909-3 2016 Thalidomide was added to osteosarcoma cells and studied by cytotoxicity assay, evaluating apoptosis, cell cycle arrest, mitochondrial membrane potential (DeltaPsim), and reactive oxygen species (ROS) levels and the expression of Bcl-2, Bax, caspase-3 and NF-kappaB. Thalidomide 0-11 BCL2 associated X, apoptosis regulator Homo sapiens 236-239 27748909-3 2016 Thalidomide was added to osteosarcoma cells and studied by cytotoxicity assay, evaluating apoptosis, cell cycle arrest, mitochondrial membrane potential (DeltaPsim), and reactive oxygen species (ROS) levels and the expression of Bcl-2, Bax, caspase-3 and NF-kappaB. Thalidomide 0-11 caspase 3 Homo sapiens 241-250 27748909-3 2016 Thalidomide was added to osteosarcoma cells and studied by cytotoxicity assay, evaluating apoptosis, cell cycle arrest, mitochondrial membrane potential (DeltaPsim), and reactive oxygen species (ROS) levels and the expression of Bcl-2, Bax, caspase-3 and NF-kappaB. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 255-264 27748909-8 2016 By western blot analysis, thalidomide resulted in the decreasing expression of Bcl-2 and NF-kappaB, and the increasing expression of Bcl-2/Bax and caspase-3. Thalidomide 26-37 BCL2 apoptosis regulator Homo sapiens 79-84 27748909-8 2016 By western blot analysis, thalidomide resulted in the decreasing expression of Bcl-2 and NF-kappaB, and the increasing expression of Bcl-2/Bax and caspase-3. Thalidomide 26-37 nuclear factor kappa B subunit 1 Homo sapiens 89-98 27748909-8 2016 By western blot analysis, thalidomide resulted in the decreasing expression of Bcl-2 and NF-kappaB, and the increasing expression of Bcl-2/Bax and caspase-3. Thalidomide 26-37 BCL2 apoptosis regulator Homo sapiens 133-138 27748909-8 2016 By western blot analysis, thalidomide resulted in the decreasing expression of Bcl-2 and NF-kappaB, and the increasing expression of Bcl-2/Bax and caspase-3. Thalidomide 26-37 BCL2 associated X, apoptosis regulator Homo sapiens 139-142 27748909-8 2016 By western blot analysis, thalidomide resulted in the decreasing expression of Bcl-2 and NF-kappaB, and the increasing expression of Bcl-2/Bax and caspase-3. Thalidomide 26-37 caspase 3 Homo sapiens 147-156 27751757-2 2016 Investigations with Cereblon, a thalidomide target protein encoded by CRBN gene, have helped to understand thalidomide therapeutic and teratogenic properties. Thalidomide 32-43 cereblon Homo sapiens 70-74 27751757-2 2016 Investigations with Cereblon, a thalidomide target protein encoded by CRBN gene, have helped to understand thalidomide therapeutic and teratogenic properties. Thalidomide 107-118 cereblon Homo sapiens 70-74 27751757-3 2016 We sequenced CRBN-thalidomide binding region in 38 TE individuals and 136 Brazilians without congenital anomalies, and performed in silico analyses. Thalidomide 18-29 cereblon Homo sapiens 13-17 27751757-8 2016 Comparison of the Cereblon-thalidomide binding domains in mammals demonstrated that CRBN is highly conserved across species. Thalidomide 27-38 cereblon Homo sapiens 84-88 27553604-8 2016 Albeit remaining within the reference ranges, erythrocytes, haematocrit, total leucocyte count, neutrophils, lymphocytes, monocytes and gamma-glutamyltranspeptidase showed significant alteration associated to thalidomide treatment. Thalidomide 209-220 inactive glutathione hydrolase 2 Homo sapiens 136-164 27618360-8 2016 In the Cox regression model, low CRBN expression was the most important prognostic parameter that influenced PFS in the thalidomide-treated patients (P = .012). Thalidomide 120-131 cytochrome c oxidase subunit 8A Homo sapiens 7-10 27618360-8 2016 In the Cox regression model, low CRBN expression was the most important prognostic parameter that influenced PFS in the thalidomide-treated patients (P = .012). Thalidomide 120-131 cereblon Homo sapiens 33-37 27618360-9 2016 CONCLUSION: CRBN expression is of prognostic value in MM patients ineligible for ASCT treated with thalidomide as an immunomodulatory drug. Thalidomide 99-110 cereblon Homo sapiens 12-16 27739411-0 2016 Inhibition of Uterine Contractility by Thalidomide Analogs via Phosphodiesterase-4 Inhibition and Calcium Entry Blockade. Thalidomide 39-50 phosphodiesterase 4A Homo sapiens 63-82 27748917-7 2016 It was identified that treatment of cells with lenalidomide and thalidomide led to a dose-dependent inhibition of cell proliferation, and the two drugs were able to induce cells apoptosis and inhibit VEGF expression in HCC cells. Thalidomide 64-75 vascular endothelial growth factor A Homo sapiens 200-204 27739411-3 2016 Some thalidomide analogs are PDE-4 inhibitors. Thalidomide 5-16 phosphodiesterase 4A Homo sapiens 29-34 27739411-11 2016 The inhibitory effects of thalidomide analogs on the contractions of pregnant human myometrium tissue may be due to their PDE-4 inhibitory effect and novel mechanism as calcium-channel blockers. Thalidomide 26-37 phosphodiesterase 4A Homo sapiens 122-127 27695085-7 2016 We further analysed monocyte TLR and CD14 expression upon exposure to the IMID immunomodulatory drug Pomalidomide (CC-4047), a Thalidomide analogue known to downregulate PU.1. Thalidomide 128-139 Spi-1 proto-oncogene Homo sapiens 171-175 27365142-1 2016 Cereblon (CRBN) has been identified as a primary target of immunomodulatory drugs and is considered a biomarker for the prediction of outcomes after thalidomide- or lenalidomide-based treatments. Thalidomide 149-160 cereblon Homo sapiens 10-14 27365142-6 2016 Among patients treated with thalidomide-based regimens, CRBN(+) patients showed a better treatment response than did CRBN-negative patients (35.0 vs. 11.8 % complete response rate, respectively; HR = 4.038, P = 0.137). Thalidomide 28-39 cereblon Homo sapiens 56-60 27365142-6 2016 Among patients treated with thalidomide-based regimens, CRBN(+) patients showed a better treatment response than did CRBN-negative patients (35.0 vs. 11.8 % complete response rate, respectively; HR = 4.038, P = 0.137). Thalidomide 28-39 cereblon Homo sapiens 117-121 27365142-9 2016 These data suggest that thalidomide-based regimens are as effective as bortezomib-based regimens in elderly patients with MM who are CRBN(+). Thalidomide 24-35 cereblon Homo sapiens 133-137 27422610-5 2016 Here, we modulate the permeability of tumor vessels by thalidomide (THD) in a sarcoma-bearing EPR mouse model via monitoring endogenous deoxygenated hemoglobin in circulation, and then we confirm the effect of THD on tumor vessel permeability by vessel density, vessel maturity, VEGF expression and so on. Thalidomide 68-71 vascular endothelial growth factor A Mus musculus 279-283 27496680-3 2016 Thalidomide suppresses VEGF and plasma cell proliferation. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 23-27 27496680-12 2016 The adjusted mean VEGF concentration reduction rate at 24 weeks was 0 39 (SD 0 34) in the thalidomide group compared with -0 02 (0 54) in the placebo group (adjusted mean difference 0 41, 95% CI 0 02-0 80; p=0 04). Thalidomide 90-101 vascular endothelial growth factor A Homo sapiens 18-22 27496680-18 2016 INTERPRETATION: Thalidomide reduces serum VEGF concentrations and represents a new treatment for patients with POEMS syndrome who are not eligible for stem-cell transplantation. Thalidomide 16-27 vascular endothelial growth factor A Homo sapiens 42-46 27343012-2 2016 Several key findings suggest diverse roles of CRBN, including its regulation of the large-conductance calcium- and voltage-activated potassium (BKCa) channels, regulation of thalidomide-binding proteins, and mediation of lenalidomide treatment in multiple myeloma. Thalidomide 174-185 cereblon Homo sapiens 46-50 27556927-6 2016 Several mechanisms have been proposed to explain the efficacy of thalidomide in ENL, including the inhibition of TNF production. Thalidomide 65-76 tumor necrosis factor Homo sapiens 113-116 27556927-12 2016 The present study demonstrated that CD64 mRNA and protein were expressed in ENL lesions and that thalidomide treatment reduced CD64 expression and neutrophil infiltrates-a hallmark of ENL. Thalidomide 97-108 Fc gamma receptor Ia Homo sapiens 127-131 27460676-4 2016 Although the molecular mechanism of thalidomide and IMiDs remained unclear for a long time, cereblon, a substrate receptor of the CRL4 E3 ubiquitin ligase was identified as a primary direct target by a new affinity technique. Thalidomide 36-47 interleukin 17 receptor B Homo sapiens 130-134 27693314-6 2016 The results demonstrated that SNAP-25 mRNA levels were increased by 49 and 66% by TBBPA and THAL, respectively, whereas VPA and NAg reduced these levels to 48 and 64% of the control, respectively. Thalidomide 92-96 synaptosome associated protein 25 Rattus norvegicus 30-37 27693314-7 2016 The SNAP-25 protein content in CGCs was increased by 79% by TBBPA, 25% by THAL and 21% by NAg; VPA and TH reduced these levels to 73 and 69% of the control, respectively. Thalidomide 74-78 synaptosome associated protein 25 Rattus norvegicus 4-11 27329811-7 2016 CRBN was also identified as a thalidomide-binding protein and component of the cullin-4-containing E3 ubiquitin ligase complex. Thalidomide 30-41 cereblon Homo sapiens 0-4 27329811-8 2016 Thalidomide has been reported to be involved in the regulation of the transcription factor Ikaros by CRBN-mediated degradation. Thalidomide 0-11 IKAROS family zinc finger 1 Homo sapiens 91-97 27329811-8 2016 Thalidomide has been reported to be involved in the regulation of the transcription factor Ikaros by CRBN-mediated degradation. Thalidomide 0-11 cereblon Homo sapiens 101-105 27282278-3 2016 The present study was initiated to determine the effect of the NF-kappaB inhibitor thalidomide on the clinical manifestation of oral mucositis in the established mouse tongue model. Thalidomide 83-94 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 63-72 27282278-10 2016 CONCLUSION: Thalidomide treatment demonstrated a significant mucositis-ameliorating effect during fractionated irradiation, which is likely to result from NF-kappaB inhibition. Thalidomide 12-23 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 155-164 27531780-1 2016 OBJECTIVE: To investigate the expression of PIGF and its receptor Flt-1 in patients with multiple myeloma, and to analyze their correlation with the efficacy of thalidomide-based chemotherapy so as to provide further theoretical basis for individualized treatment. Thalidomide 161-172 phosphatidylinositol glycan anchor biosynthesis class F Homo sapiens 44-48 27531780-11 2016 CONCLUSION: PIGF and FLT-1 are highly expressed in patients with multiple myeloma, and their expression levels positively correlates with curative effecacy of thalidomide-based chemotherapy. Thalidomide 159-170 phosphatidylinositol glycan anchor biosynthesis class F Homo sapiens 12-16 27531780-11 2016 CONCLUSION: PIGF and FLT-1 are highly expressed in patients with multiple myeloma, and their expression levels positively correlates with curative effecacy of thalidomide-based chemotherapy. Thalidomide 159-170 fms related receptor tyrosine kinase 1 Homo sapiens 21-26 27417535-3 2016 The intellectual disability-associated and thalidomide-binding protein cereblon (CRBN) contains a large, highly conserved Lon domain. Thalidomide 43-54 cereblon Homo sapiens 81-85 27294876-8 2016 Finally, we show that BSG (CD147) knockdown phenocopies the teratogenic effects of thalidomide exposure in zebrafish. Thalidomide 83-94 basigin Danio rerio 22-25 27346278-6 2016 Intra-RVM administrations of thalidomide dose-dependently attenuated mechanical allodynia and thermal hyperalgesia, and this phenomenon was associated with reduced levels of TNFalpha, IL-1beta, and NFkappaB in the RVM, without altering serum levels of TNFalpha or IL-1beta. Thalidomide 29-40 tumor necrosis factor Rattus norvegicus 174-182 27346278-6 2016 Intra-RVM administrations of thalidomide dose-dependently attenuated mechanical allodynia and thermal hyperalgesia, and this phenomenon was associated with reduced levels of TNFalpha, IL-1beta, and NFkappaB in the RVM, without altering serum levels of TNFalpha or IL-1beta. Thalidomide 29-40 interleukin 1 beta Rattus norvegicus 184-192 27346278-6 2016 Intra-RVM administrations of thalidomide dose-dependently attenuated mechanical allodynia and thermal hyperalgesia, and this phenomenon was associated with reduced levels of TNFalpha, IL-1beta, and NFkappaB in the RVM, without altering serum levels of TNFalpha or IL-1beta. Thalidomide 29-40 tumor necrosis factor Rattus norvegicus 252-260 27346278-6 2016 Intra-RVM administrations of thalidomide dose-dependently attenuated mechanical allodynia and thermal hyperalgesia, and this phenomenon was associated with reduced levels of TNFalpha, IL-1beta, and NFkappaB in the RVM, without altering serum levels of TNFalpha or IL-1beta. Thalidomide 29-40 interleukin 1 beta Rattus norvegicus 264-272 27346278-7 2016 These results suggested that supraspinal mechanisms of thalidomide play a critical role in modulations of type 2 diabetes induced neuropathic pain, which is likely mediated by TNFalpha and IL-1beta in the RVM. Thalidomide 55-66 tumor necrosis factor Rattus norvegicus 176-184 27346278-7 2016 These results suggested that supraspinal mechanisms of thalidomide play a critical role in modulations of type 2 diabetes induced neuropathic pain, which is likely mediated by TNFalpha and IL-1beta in the RVM. Thalidomide 55-66 interleukin 1 beta Rattus norvegicus 189-197 27294876-8 2016 Finally, we show that BSG (CD147) knockdown phenocopies the teratogenic effects of thalidomide exposure in zebrafish. Thalidomide 83-94 basigin Danio rerio 27-32 27249651-7 2016 In vivo zebrafish experiments suggested that HIF-2alpha overexpression was associated with abnormal subintestinal vascular (SIV) sprouting, which was reversed by thalidomide. Thalidomide 162-173 hypoxia inducible factor 1 subunit alpha, like 2 Danio rerio 45-55 26865412-1 2016 Thalidomide and its derivatives, collectively referred to as immunomodulatory drugs (IMiDs), are effective inhibitors of inflammation and are known to inhibit TLR-induced TNFalpha production. Thalidomide 0-11 tumor necrosis factor Mus musculus 171-179 26689580-9 2016 miR-202 over-expression sensitized MM cells to bortezomib (Bort) but less to Thalidomide (Thal) and dexamethasone (Dex). Thalidomide 77-88 microRNA 202 Homo sapiens 0-7 26689580-9 2016 miR-202 over-expression sensitized MM cells to bortezomib (Bort) but less to Thalidomide (Thal) and dexamethasone (Dex). Thalidomide 77-81 microRNA 202 Homo sapiens 0-7 27249651-8 2016 This result indicated that thalidomide regulated angiogenesis via the inhibition of HIF-1alpha and HIF-2alpha expression, which further regulated downstream factors, including VEGF, NOTCH1, DLL4, and Ang2. Thalidomide 27-38 vascular endothelial growth factor A Homo sapiens 176-180 27249651-8 2016 This result indicated that thalidomide regulated angiogenesis via the inhibition of HIF-1alpha and HIF-2alpha expression, which further regulated downstream factors, including VEGF, NOTCH1, DLL4, and Ang2. Thalidomide 27-38 notch receptor 1 Homo sapiens 182-188 27249651-8 2016 This result indicated that thalidomide regulated angiogenesis via the inhibition of HIF-1alpha and HIF-2alpha expression, which further regulated downstream factors, including VEGF, NOTCH1, DLL4, and Ang2. Thalidomide 27-38 delta like canonical Notch ligand 4 Homo sapiens 190-194 27249651-8 2016 This result indicated that thalidomide regulated angiogenesis via the inhibition of HIF-1alpha and HIF-2alpha expression, which further regulated downstream factors, including VEGF, NOTCH1, DLL4, and Ang2. Thalidomide 27-38 angiopoietin 2 Homo sapiens 200-204 27629726-8 2016 The ELISA assay revealed that both 350 mug/mL of thalidomide and 1400 U/mL of IFN could reduce the VEGF levels in cell culture supernatants; the two-drug combination group had a further decreased VEGF concentration. Thalidomide 49-60 vascular endothelial growth factor A Homo sapiens 99-103 27129176-0 2016 Thalidomide enhanced the efficacy of CHOP chemotherapy in the treatment of diffuse large B cell lymphoma: A phase II study. Thalidomide 0-11 DNA damage inducible transcript 3 Homo sapiens 37-41 27129176-11 2016 We concluded that the addition of thalidomide to the CHOP regimen significantly improved the CRR and showed a trend of improving clinical outcome in patients with DLBCL, especially for patients with Bcl-2 positive and Bcl-6 negative B-cell phenotype, without increased toxicity. Thalidomide 34-45 DNA damage inducible transcript 3 Homo sapiens 53-57 27129176-11 2016 We concluded that the addition of thalidomide to the CHOP regimen significantly improved the CRR and showed a trend of improving clinical outcome in patients with DLBCL, especially for patients with Bcl-2 positive and Bcl-6 negative B-cell phenotype, without increased toxicity. Thalidomide 34-45 BCL2 apoptosis regulator Homo sapiens 199-204 27129176-11 2016 We concluded that the addition of thalidomide to the CHOP regimen significantly improved the CRR and showed a trend of improving clinical outcome in patients with DLBCL, especially for patients with Bcl-2 positive and Bcl-6 negative B-cell phenotype, without increased toxicity. Thalidomide 34-45 BCL6 transcription repressor Homo sapiens 218-223 27386947-13 2016 Meanwhile, we found that isoform D of vascular endothelial growth factor was elevated, which was normalized after the introduction of thalidomide. Thalidomide 134-145 vascular endothelial growth factor A Homo sapiens 38-72 27629726-9 2016 Forty-eighthour treatment of thalidomide 350 mug/mL and IFN 1400 U/mL could significantly decrease Bcl-2 expression and increase the expression levels of phosphor-P38, BAX, cytochrome c, and cleaved caspase-3, -8, and -9 as compared to the control group. Thalidomide 29-40 BCL2 apoptosis regulator Homo sapiens 99-104 27629726-9 2016 Forty-eighthour treatment of thalidomide 350 mug/mL and IFN 1400 U/mL could significantly decrease Bcl-2 expression and increase the expression levels of phosphor-P38, BAX, cytochrome c, and cleaved caspase-3, -8, and -9 as compared to the control group. Thalidomide 29-40 BCL2 associated X, apoptosis regulator Homo sapiens 168-171 27629726-9 2016 Forty-eighthour treatment of thalidomide 350 mug/mL and IFN 1400 U/mL could significantly decrease Bcl-2 expression and increase the expression levels of phosphor-P38, BAX, cytochrome c, and cleaved caspase-3, -8, and -9 as compared to the control group. Thalidomide 29-40 cytochrome c, somatic Homo sapiens 173-185 27629726-8 2016 The ELISA assay revealed that both 350 mug/mL of thalidomide and 1400 U/mL of IFN could reduce the VEGF levels in cell culture supernatants; the two-drug combination group had a further decreased VEGF concentration. Thalidomide 49-60 vascular endothelial growth factor A Homo sapiens 196-200 27629726-11 2016 CONCLUSIONS: Thalidomide and IFN can synergistically inhibit Kasumi-1 cell proliferation, which is possibly achieved through the mitochondrial and death receptor pathways and through the activation of the P38 signaling pathway to induce apoptosis and by inhibiting Kasumi-1 cell autocrine VEGF secretion. Thalidomide 13-24 vascular endothelial growth factor A Homo sapiens 289-293 27320601-0 2016 Role of Thalidomide on the Expression of OX40, 4-1BB, and GITR in T Cell Subsets. Thalidomide 8-19 tumor necrosis factor receptor superfamily, member 4 Mus musculus 41-45 27320601-0 2016 Role of Thalidomide on the Expression of OX40, 4-1BB, and GITR in T Cell Subsets. Thalidomide 8-19 tumor necrosis factor receptor superfamily, member 9 Mus musculus 47-52 27320601-0 2016 Role of Thalidomide on the Expression of OX40, 4-1BB, and GITR in T Cell Subsets. Thalidomide 8-19 tumor necrosis factor receptor superfamily, member 18 Mus musculus 58-62 26990986-4 2016 Binding of acetylated degron peptides to CRBN depends on an intact thalidomide-binding pocket but is not competitive with IMiDs. Thalidomide 67-78 cereblon Homo sapiens 41-45 26692341-0 2016 Thalidomide analogues: Tumor necrosis factor-alpha inhibitors and their evaluation as anti-inflammatory agents. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 23-50 26990986-1 2016 Cereblon (CRBN), a substrate receptor for the cullin-RING ubiquitin ligase 4 (CRL4) complex, is a direct protein target for thalidomide teratogenicity and antitumor activity of immunomodulatory drugs (IMiDs). Thalidomide 124-135 cereblon Homo sapiens 0-8 26990986-1 2016 Cereblon (CRBN), a substrate receptor for the cullin-RING ubiquitin ligase 4 (CRL4) complex, is a direct protein target for thalidomide teratogenicity and antitumor activity of immunomodulatory drugs (IMiDs). Thalidomide 124-135 cereblon Homo sapiens 10-14 26903378-0 2016 Thalidomide-induced limb abnormalities in a humanized CYP3A mouse model. Thalidomide 0-11 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 54-59 26903378-8 2016 Thalidomide-treated embryos with the human CYP3A gene cluster showed limb abnormalities, and human CYP3A was expressed in the placenta, suggesting that human CYP3A in the placenta may contribute to the teratogenicity of thalidomide. Thalidomide 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-48 26903378-8 2016 Thalidomide-treated embryos with the human CYP3A gene cluster showed limb abnormalities, and human CYP3A was expressed in the placenta, suggesting that human CYP3A in the placenta may contribute to the teratogenicity of thalidomide. Thalidomide 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-104 26903378-8 2016 Thalidomide-treated embryos with the human CYP3A gene cluster showed limb abnormalities, and human CYP3A was expressed in the placenta, suggesting that human CYP3A in the placenta may contribute to the teratogenicity of thalidomide. Thalidomide 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-104 26903378-8 2016 Thalidomide-treated embryos with the human CYP3A gene cluster showed limb abnormalities, and human CYP3A was expressed in the placenta, suggesting that human CYP3A in the placenta may contribute to the teratogenicity of thalidomide. Thalidomide 220-231 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-48 26903378-8 2016 Thalidomide-treated embryos with the human CYP3A gene cluster showed limb abnormalities, and human CYP3A was expressed in the placenta, suggesting that human CYP3A in the placenta may contribute to the teratogenicity of thalidomide. Thalidomide 220-231 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-104 26903378-8 2016 Thalidomide-treated embryos with the human CYP3A gene cluster showed limb abnormalities, and human CYP3A was expressed in the placenta, suggesting that human CYP3A in the placenta may contribute to the teratogenicity of thalidomide. Thalidomide 220-231 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-104 27003992-4 2016 Thalidomide, which caused severe deformities in newborn children in the 1960s, is now increasingly used after it was shown to suppress tumor necrosis factor alpha, inhibit angiogenesis and to be also effective for treatment of multiple myeloma. Thalidomide 0-11 tumor necrosis factor Homo sapiens 135-162 26998101-4 2016 The exact mechanism of action is not yet clear, however, we hypothesize that thalidomide may function through decreasing the secretion of interleukin-6 and affecting the growth of plasma cells. Thalidomide 77-88 interleukin 6 Homo sapiens 138-151 26820153-1 2016 BACKGROUND AND AIMS: By blocking TNFalpha-related effects, thalidomide not only inhibits hepatic fibrogenesis but improves peripheral vasodilatation and portal hypertension in cirrhotic rats. Thalidomide 59-70 tumor necrosis factor Rattus norvegicus 33-41 26373954-12 2016 Co-administration of thalidomide (TNF-alpha synthesis inhibitor) and IL-1ra prevented BTZ-induced mechanical allodynia. Thalidomide 21-32 tumor necrosis factor Rattus norvegicus 34-43 26781879-7 2016 of the TNF-alpha synthesis inhibitor thalidomide reduced the SNI-induced mechanical hypersensitivity and inhibited the expression of CXCL12 in the DRG and spinal cord. Thalidomide 37-48 tumor necrosis factor Rattus norvegicus 7-16 26781879-7 2016 of the TNF-alpha synthesis inhibitor thalidomide reduced the SNI-induced mechanical hypersensitivity and inhibited the expression of CXCL12 in the DRG and spinal cord. Thalidomide 37-48 C-X-C motif chemokine ligand 12 Rattus norvegicus 133-139 26820153-10 2016 Additionally, acute incubation with thalidomide abolished TNFalpha-augmented VEGF-mediated migration of and tube formation of human umbilical vein endothelial cells, which was accompanied by corresponding changes in inflammatory and angiogenic substances release. Thalidomide 36-47 tumor necrosis factor Homo sapiens 58-66 26820153-10 2016 Additionally, acute incubation with thalidomide abolished TNFalpha-augmented VEGF-mediated migration of and tube formation of human umbilical vein endothelial cells, which was accompanied by corresponding changes in inflammatory and angiogenic substances release. Thalidomide 36-47 vascular endothelial growth factor A Homo sapiens 77-81 26554332-5 2016 Chorioallantoic membrane assay (CAM) shows that antiangiogenic activity of latent and oxidized AT are better than thalidomide, a potent antiangiogenic drug. Thalidomide 114-125 serpin family C member 1 Homo sapiens 95-97 26711561-0 2016 Thalidomide represses inflammatory response and reduces radiculopathic pain by inhibiting IRAK-1 and NF-kappaB/p38/JNK signaling. Thalidomide 0-11 interleukin-1 receptor-associated kinase 1 Rattus norvegicus 90-96 26711561-0 2016 Thalidomide represses inflammatory response and reduces radiculopathic pain by inhibiting IRAK-1 and NF-kappaB/p38/JNK signaling. Thalidomide 0-11 mitogen activated protein kinase 14 Rattus norvegicus 111-114 26711561-0 2016 Thalidomide represses inflammatory response and reduces radiculopathic pain by inhibiting IRAK-1 and NF-kappaB/p38/JNK signaling. Thalidomide 0-11 mitogen-activated protein kinase 8 Rattus norvegicus 115-118 26711561-6 2016 Our results indicated that thalidomide significantly inhibited the expression of pro-inflammatory mediators and matrix metalloproteinases in vitro, as well as radiculopathic pain in vivo, most probably by modulation of the activity of IRAK-1 and its downstream effectors p38, JNK and NF-kappaB. Thalidomide 27-38 interleukin-1 receptor-associated kinase 1 Rattus norvegicus 235-241 26711561-6 2016 Our results indicated that thalidomide significantly inhibited the expression of pro-inflammatory mediators and matrix metalloproteinases in vitro, as well as radiculopathic pain in vivo, most probably by modulation of the activity of IRAK-1 and its downstream effectors p38, JNK and NF-kappaB. Thalidomide 27-38 mitogen activated protein kinase 14 Rattus norvegicus 271-274 26711561-6 2016 Our results indicated that thalidomide significantly inhibited the expression of pro-inflammatory mediators and matrix metalloproteinases in vitro, as well as radiculopathic pain in vivo, most probably by modulation of the activity of IRAK-1 and its downstream effectors p38, JNK and NF-kappaB. Thalidomide 27-38 mitogen-activated protein kinase 8 Rattus norvegicus 276-279 29741823-8 2016 To conclude, for the management of recurrent/chronic ENL, both the combinations-prednisolone with thalidomide or clofazimine appear to be efficacious. Thalidomide 98-109 MLLT1 super elongation complex subunit Homo sapiens 53-56 26951036-5 2016 But because of their high cost we chose thalidomide, which also has TNF-alpha inhibiting properties to successfully treat a long-standing case of ulcerative NL, which was refractory to various treatment modalities. Thalidomide 40-51 tumor necrosis factor Homo sapiens 68-77 27047649-0 2016 Thalidomide is more efficient than sodium butyrate in enhancing GATA-1 and EKLF gene expression in erythroid progenitors derived from HSCs with beta-globin gene mutation. Thalidomide 0-11 GATA binding protein 1 Homo sapiens 64-70 26440459-0 2016 LASSBio-1425, an analog of thalidomide, decreases triglyceride and increases HDL cholesterol levels by inhibition of TNF-alpha production. Thalidomide 27-38 tumor necrosis factor Homo sapiens 117-126 26586093-12 2016 CONCLUSION: Metronomic capecitabine and thalidomide after RFA significantly reduced recurrence of HCC and extended PFS, especially for HCC outside the Milan criteria, perhaps via reduction of serum CECs and VEGF levels and inhibition of tumour angiogenesis. Thalidomide 40-51 vascular endothelial growth factor A Homo sapiens 207-211 27047649-0 2016 Thalidomide is more efficient than sodium butyrate in enhancing GATA-1 and EKLF gene expression in erythroid progenitors derived from HSCs with beta-globin gene mutation. Thalidomide 0-11 Kruppel like factor 1 Homo sapiens 75-79 27047649-7 2016 RESULTS: Thalidomide and sodium butyrate increased GATA-1 and EKLF gene expression, compared to the non-treated control (P<0.05). Thalidomide 9-20 GATA binding protein 1 Homo sapiens 51-57 27047649-7 2016 RESULTS: Thalidomide and sodium butyrate increased GATA-1 and EKLF gene expression, compared to the non-treated control (P<0.05). Thalidomide 9-20 Kruppel like factor 1 Homo sapiens 62-66 27047649-8 2016 CONCLUSION: Thalidomide was more efficient than sodium butyrate in augmenting expression of GATA-1 and EKLF genes. Thalidomide 12-23 GATA binding protein 1 Homo sapiens 92-98 27047649-8 2016 CONCLUSION: Thalidomide was more efficient than sodium butyrate in augmenting expression of GATA-1 and EKLF genes. Thalidomide 12-23 Kruppel like factor 1 Homo sapiens 103-107 27047649-9 2016 It seems that GATA-1 and EKLF have crucial roles in the efficient induction of HbF by thalidomide. Thalidomide 86-97 GATA binding protein 1 Homo sapiens 14-20 27047649-9 2016 It seems that GATA-1 and EKLF have crucial roles in the efficient induction of HbF by thalidomide. Thalidomide 86-97 Kruppel like factor 1 Homo sapiens 25-29 27863757-4 2016 The objective of this study was to assess the number of CD34+ cells mobilized using granulocyte-colony stimulating factor with and without cyclophosphamide after induction with cyclophosphamide, thalidomide and dexamethasone. Thalidomide 195-206 CD34 molecule Homo sapiens 56-60 26546878-9 2015 Spot-forming cell (SFC) counts in the IFN-gamma ELISPOT assay decreased from baseline after bortezomib (p=0.011) or thalidomide (p=0.096) treatment. Thalidomide 116-127 interferon gamma Homo sapiens 38-47 26231201-2 2015 Thalidomide binds to cereblon (CRBN), a substrate receptor of the cullin-4 really interesting new gene (RING) E3 ligase complex. Thalidomide 0-11 cereblon Homo sapiens 31-35 26231201-3 2015 Here, we examine the effect of thalidomide and its analogs on CRBN ubiquitination and its functions in human cell lines. Thalidomide 31-42 cereblon Homo sapiens 62-66 26231201-4 2015 We find that the ubiquitin modification of CRBN includes K48-linked polyubiquitin chains and that thalidomide blocks the formation of CRBN-ubiquitin conjugates. Thalidomide 98-109 cereblon Homo sapiens 134-138 26231201-6 2015 Treatment of human myeloma cell lines such as MM1.S, OPM2, and U266 with thalidomide (100 muM) and its structural analog lenalidomide (10 muM) results in stabilization of CRBN and elevation of CRBN protein levels. Thalidomide 73-84 latexin Homo sapiens 90-93 26231201-6 2015 Treatment of human myeloma cell lines such as MM1.S, OPM2, and U266 with thalidomide (100 muM) and its structural analog lenalidomide (10 muM) results in stabilization of CRBN and elevation of CRBN protein levels. Thalidomide 73-84 latexin Homo sapiens 138-141 26231201-6 2015 Treatment of human myeloma cell lines such as MM1.S, OPM2, and U266 with thalidomide (100 muM) and its structural analog lenalidomide (10 muM) results in stabilization of CRBN and elevation of CRBN protein levels. Thalidomide 73-84 cereblon Homo sapiens 171-175 26231201-6 2015 Treatment of human myeloma cell lines such as MM1.S, OPM2, and U266 with thalidomide (100 muM) and its structural analog lenalidomide (10 muM) results in stabilization of CRBN and elevation of CRBN protein levels. Thalidomide 73-84 cereblon Homo sapiens 193-197 26231201-8 2015 Our results reveal a novel mechanism by which thalidomide and its analogs modulate the CRBN function in cells. Thalidomide 46-57 cereblon Homo sapiens 87-91 26231201-9 2015 Through inhibition of CRBN ubiquitination, thalidomide and its analogs allow CRBN to accumulate, leading to the increased cullin-4 RING E3 ligase-mediated degradation of target proteins. Thalidomide 43-54 cereblon Homo sapiens 22-26 26231201-9 2015 Through inhibition of CRBN ubiquitination, thalidomide and its analogs allow CRBN to accumulate, leading to the increased cullin-4 RING E3 ligase-mediated degradation of target proteins. Thalidomide 43-54 cereblon Homo sapiens 77-81 26503997-5 2015 Herein the signalling pathways modulated by thalidomides via the mediation of vascular endothelial growth factor, phosphoinositide-kinase/protein kinase B and nuclear factor kappa B, and mammalian target of rapamycin, which integrates these signalling systems, are discussed. Thalidomide 44-56 vascular endothelial growth factor A Homo sapiens 78-112 26521987-1 2015 Recent studies have suggested that cereblon (CRBN) is essential for the anti-myeloma (MM) activity of immunomodulatory drugs (IMiDs), such as thalidomide and lenalidomide, and that dysregulation of Wnt/beta-catenin pathway may be one of possible reasons of lenalidomide resistance. Thalidomide 142-153 cereblon Homo sapiens 45-49 26521987-6 2015 Patients carrying the CTNNB1 (rs4533622) AA genotype were better responders to the first line therapy with thalidomide containing regimens (p<0.05). Thalidomide 107-118 catenin beta 1 Homo sapiens 22-28 26481541-0 2015 Thalidomide can promote erythropoiesis by induction of STAT5 and repression of external pathway of apoptosis resulting in increased expression of GATA-1 transcription factor. Thalidomide 0-11 signal transducer and activator of transcription 5A Homo sapiens 55-60 26481541-0 2015 Thalidomide can promote erythropoiesis by induction of STAT5 and repression of external pathway of apoptosis resulting in increased expression of GATA-1 transcription factor. Thalidomide 0-11 GATA binding protein 1 Homo sapiens 146-152 26481541-8 2015 CONCLUSION: Our results suggest that thalidomide enhances expression of STAT5 in response of erythroid cells to erythropoietin and as a result of caspase 3 suppression. Thalidomide 37-48 signal transducer and activator of transcription 5A Homo sapiens 72-77 26481541-8 2015 CONCLUSION: Our results suggest that thalidomide enhances expression of STAT5 in response of erythroid cells to erythropoietin and as a result of caspase 3 suppression. Thalidomide 37-48 erythropoietin Homo sapiens 112-126 26481541-8 2015 CONCLUSION: Our results suggest that thalidomide enhances expression of STAT5 in response of erythroid cells to erythropoietin and as a result of caspase 3 suppression. Thalidomide 37-48 caspase 3 Homo sapiens 146-155 26608985-8 2015 Dynamic real-time polymerase chain reaction assay, ELISA and Western blotting from patient"s peripheral blood mononuclear cells and plasma were used to detect the expression of transforming growth factor beta 3 (TGF-beta3) messenger RNA (mRNA) and vascular endothelial growth factor (VEGF) protein before and after 6 months of thalidomide treatment. Thalidomide 327-338 transforming growth factor beta 3 Homo sapiens 177-210 26608985-11 2015 The Fli-EGFP zebrafish model manifested discontinuous vessel development and vascular occlusion (7 of 10 fishes), and the TGF-beta3 mRNA expression of five patients was lower after thalidomide therapy. Thalidomide 181-192 Fli-1 proto-oncogene, ETS transcription factor Danio rerio 4-7 26608985-11 2015 The Fli-EGFP zebrafish model manifested discontinuous vessel development and vascular occlusion (7 of 10 fishes), and the TGF-beta3 mRNA expression of five patients was lower after thalidomide therapy. Thalidomide 181-192 transforming growth factor beta 3 Homo sapiens 122-131 26608985-13 2015 CONCLUSIONS: Thalidomide reverses telangiectasia and controls nosebleeds by down-regulating the expression of TGF-beta3 and VEGF in HHT patients. Thalidomide 13-24 transforming growth factor beta 3 Homo sapiens 110-119 26608985-13 2015 CONCLUSIONS: Thalidomide reverses telangiectasia and controls nosebleeds by down-regulating the expression of TGF-beta3 and VEGF in HHT patients. Thalidomide 13-24 vascular endothelial growth factor A Homo sapiens 124-128 26503997-5 2015 Herein the signalling pathways modulated by thalidomides via the mediation of vascular endothelial growth factor, phosphoinositide-kinase/protein kinase B and nuclear factor kappa B, and mammalian target of rapamycin, which integrates these signalling systems, are discussed. Thalidomide 44-56 protein tyrosine kinase 2 beta Homo sapiens 138-181 26503997-5 2015 Herein the signalling pathways modulated by thalidomides via the mediation of vascular endothelial growth factor, phosphoinositide-kinase/protein kinase B and nuclear factor kappa B, and mammalian target of rapamycin, which integrates these signalling systems, are discussed. Thalidomide 44-56 mechanistic target of rapamycin kinase Homo sapiens 187-216 26220057-5 2015 The sequential drug release of LDC and THD from the developed LDC-THD-GO nanosheets exhibited a synergistic effect on neuropathic pain in vitro and in vivo, as evidenced by the increased pain threshold in mechanical allodynia and hyperalgesic response tests, and the improved inhibition of proinflammatory cytokines TNF-alpha, IL-1beta, IL-6, and nitric oxide. Thalidomide 39-42 tumor necrosis factor Homo sapiens 316-325 26486925-7 2015 RESULTS: Among our 64 patients with gastrointestinal involvement of Behcet"s syndrome, we identified 13 (20%) (7 women, 6 men, mean age 27.4+-9.4) who had been treated with TNF-alpha antagonists and/or thalidomide. Thalidomide 202-213 tumor necrosis factor Homo sapiens 173-182 26398114-5 2015 Thalidomide-gamma-cyclodextrin complex exerted a concentration-dependent cytotoxicity in TCC cells, but was relatively less cytotoxic (with IC50 of 200 microM) in BFTC905 cells than the other 3 TCC cell lines, possibly due to upregulation of Bcl-xL and HIF-1alpha mediated carbonic anhydrase IX, and promotion of quiescence. Thalidomide 0-11 BCL2 like 1 Homo sapiens 242-248 26398114-5 2015 Thalidomide-gamma-cyclodextrin complex exerted a concentration-dependent cytotoxicity in TCC cells, but was relatively less cytotoxic (with IC50 of 200 microM) in BFTC905 cells than the other 3 TCC cell lines, possibly due to upregulation of Bcl-xL and HIF-1alpha mediated carbonic anhydrase IX, and promotion of quiescence. Thalidomide 0-11 hypoxia inducible factor 1 subunit alpha Homo sapiens 253-263 26398114-5 2015 Thalidomide-gamma-cyclodextrin complex exerted a concentration-dependent cytotoxicity in TCC cells, but was relatively less cytotoxic (with IC50 of 200 microM) in BFTC905 cells than the other 3 TCC cell lines, possibly due to upregulation of Bcl-xL and HIF-1alpha mediated carbonic anhydrase IX, and promotion of quiescence. Thalidomide 0-11 carbonic anhydrase 9 Homo sapiens 273-294 26398114-7 2015 Thalidomide induced apoptosis through downregulation of survivin and securin. Thalidomide 0-11 PTTG1 regulator of sister chromatid separation, securin Homo sapiens 69-76 26398114-8 2015 The secretion of VEGF and TNF-alpha was ameliorated by thalidomide, but they did not affect cell proliferation. Thalidomide 55-66 vascular endothelial growth factor A Homo sapiens 17-21 26398114-8 2015 The secretion of VEGF and TNF-alpha was ameliorated by thalidomide, but they did not affect cell proliferation. Thalidomide 55-66 tumor necrosis factor Homo sapiens 26-35 26398114-12 2015 Thalidomide also inhibited TNF-alpha induced invasion through inhibition of NF-kappaB, and downregulation of effectors, ICAM-1 and MMP-9. Thalidomide 0-11 tumor necrosis factor Homo sapiens 27-36 26398114-12 2015 Thalidomide also inhibited TNF-alpha induced invasion through inhibition of NF-kappaB, and downregulation of effectors, ICAM-1 and MMP-9. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 76-85 26398114-12 2015 Thalidomide also inhibited TNF-alpha induced invasion through inhibition of NF-kappaB, and downregulation of effectors, ICAM-1 and MMP-9. Thalidomide 0-11 intercellular adhesion molecule 1 Homo sapiens 120-126 26398114-12 2015 Thalidomide also inhibited TNF-alpha induced invasion through inhibition of NF-kappaB, and downregulation of effectors, ICAM-1 and MMP-9. Thalidomide 0-11 matrix metallopeptidase 9 Homo sapiens 131-136 26220057-5 2015 The sequential drug release of LDC and THD from the developed LDC-THD-GO nanosheets exhibited a synergistic effect on neuropathic pain in vitro and in vivo, as evidenced by the increased pain threshold in mechanical allodynia and hyperalgesic response tests, and the improved inhibition of proinflammatory cytokines TNF-alpha, IL-1beta, IL-6, and nitric oxide. Thalidomide 39-42 interleukin 1 beta Homo sapiens 327-335 26220057-5 2015 The sequential drug release of LDC and THD from the developed LDC-THD-GO nanosheets exhibited a synergistic effect on neuropathic pain in vitro and in vivo, as evidenced by the increased pain threshold in mechanical allodynia and hyperalgesic response tests, and the improved inhibition of proinflammatory cytokines TNF-alpha, IL-1beta, IL-6, and nitric oxide. Thalidomide 39-42 interleukin 6 Homo sapiens 337-341 26446639-0 2015 Thalidomide Accelerates the Degradation of Extracellular Matrix in Rat Hepatic Cirrhosis via Down-Regulation of Transforming Growth Factor-beta1. Thalidomide 0-11 transforming growth factor, beta 1 Rattus norvegicus 112-144 26446639-8 2015 The levels of TIMP-1 and TGF-beta1 mRNA and protein expressions were decreased significantly and MMP-13 mRNA and protein in the liver were significantly elevated in the thalidomide-treated group. Thalidomide 169-180 TIMP metallopeptidase inhibitor 1 Rattus norvegicus 14-20 26446639-8 2015 The levels of TIMP-1 and TGF-beta1 mRNA and protein expressions were decreased significantly and MMP-13 mRNA and protein in the liver were significantly elevated in the thalidomide-treated group. Thalidomide 169-180 transforming growth factor, beta 1 Rattus norvegicus 25-34 26446639-8 2015 The levels of TIMP-1 and TGF-beta1 mRNA and protein expressions were decreased significantly and MMP-13 mRNA and protein in the liver were significantly elevated in the thalidomide-treated group. Thalidomide 169-180 matrix metallopeptidase 13 Rattus norvegicus 97-103 26446639-9 2015 CONCLUSION: Thalidomide may exert its effects on the regulation of MMP-13 and TIMP-1 via inhibition of the TGF-beta1 signaling pathway, which enhances the degradation of extracellular matrix and accelerates the regression of hepatic cirrhosis in rats. Thalidomide 12-23 matrix metallopeptidase 13 Rattus norvegicus 67-73 26446639-9 2015 CONCLUSION: Thalidomide may exert its effects on the regulation of MMP-13 and TIMP-1 via inhibition of the TGF-beta1 signaling pathway, which enhances the degradation of extracellular matrix and accelerates the regression of hepatic cirrhosis in rats. Thalidomide 12-23 TIMP metallopeptidase inhibitor 1 Rattus norvegicus 78-84 26446639-9 2015 CONCLUSION: Thalidomide may exert its effects on the regulation of MMP-13 and TIMP-1 via inhibition of the TGF-beta1 signaling pathway, which enhances the degradation of extracellular matrix and accelerates the regression of hepatic cirrhosis in rats. Thalidomide 12-23 transforming growth factor, beta 1 Rattus norvegicus 107-116 26126925-12 2015 Treatment with fluoxetine or thalidomide reversed crush-induced mechanical hyperalgesia, depressive-like behaviors and the increased TNF-alpha levels in the sciatic nerve, prefrontal cortex and hippocampus. Thalidomide 29-40 tumor necrosis factor Mus musculus 133-142 26632468-12 2015 CONCLUSION: CsA combined with thalidomide regime could improve the anemia symptom in low/int-1 risk MDS patients without del(5q). Thalidomide 30-41 Wnt family member 1 Homo sapiens 89-94 26362268-2 2015 However, RA may also induce the expression of resistance genes such as HOXB7 which can be suppressed by Thalidomide (THAL). Thalidomide 104-115 homeobox B7 Homo sapiens 71-76 26362268-2 2015 However, RA may also induce the expression of resistance genes such as HOXB7 which can be suppressed by Thalidomide (THAL). Thalidomide 117-121 homeobox B7 Homo sapiens 71-76 26450624-7 2015 An important and novel finding of our study was that a thalidomide derivative lenalidomide induced 3C10-CAR PBMC proliferation and enhanced the persistent antitumor effect of the cells in vivo. Thalidomide 55-66 nuclear receptor subfamily 1, group I, member 3 Mus musculus 104-107 25895164-9 2015 Intraperitoneal injection of an inhibitor of TNF-alpha synthesis thalidomide at 50 or 100 mg/kg dose (but not 10 mg/kg dose) significantly ameliorated the reduced paw withdrawal threshold induced by SMIR surgery. Thalidomide 65-76 tumor necrosis factor Rattus norvegicus 45-54 26524034-7 2015 The percentage of Th17 cells, the mRNA expression of RORgammat and the plasm IL-17 levels in patients with response to thalidomide were statistically lower than those in patients before treatment (P < 0.05). Thalidomide 119-130 interleukin 17A Homo sapiens 77-82 26126925-14 2015 Furthermore, inhibitors of TNF-alpha synthesis, like thalidomide, have a potential to treat depressive disorders associated with neuropathic pain. Thalidomide 53-64 tumor necrosis factor Mus musculus 27-36 28913056-10 2015 CONCLUSION: Thalidomide provides volumetric and histopathologic recovery in implants particularly because the VEGF inhibition and anti-angiogenic effect, which suggests that it could be effective in the treatment of endometriosis. Thalidomide 12-23 vascular endothelial growth factor A Rattus norvegicus 110-114 26134001-6 2015 Immunohistochemistry labeling assays with CD-31 (PECAM-1) antibody showed microvascular density (MVD) was significantly reduced in thalidomide, 2a and 2b groups (30+-4.9, 64.6+-1.8 and 46.5+-19.5%, respectively) (p<0.05). Thalidomide 131-142 platelet/endothelial cell adhesion molecule 1 Mus musculus 42-47 26134001-6 2015 Immunohistochemistry labeling assays with CD-31 (PECAM-1) antibody showed microvascular density (MVD) was significantly reduced in thalidomide, 2a and 2b groups (30+-4.9, 64.6+-1.8 and 46.5+-19.5%, respectively) (p<0.05). Thalidomide 131-142 platelet/endothelial cell adhesion molecule 1 Mus musculus 49-56 26131937-5 2015 We further demonstrate that minor side chain modifications in thalidomide and a novel analogue, CC-122, can modulate the spectrum of substrates targeted by CRL4(CRBN). Thalidomide 62-73 cereblon Mus musculus 161-165 26462773-2 2015 METHODS: The inhibitiory effect of Kasumi- 1 cells by thalidomide, interferon or combination was detected by CCK- 8 method, the apoptosis by flow cytometry, the expression of apoptosis related proteins by Western blot, vascular endothelial growth factor (VEGF) concentration in culture supernatant by ELISA. Thalidomide 54-65 vascular endothelial growth factor A Homo sapiens 219-253 26462773-2 2015 METHODS: The inhibitiory effect of Kasumi- 1 cells by thalidomide, interferon or combination was detected by CCK- 8 method, the apoptosis by flow cytometry, the expression of apoptosis related proteins by Western blot, vascular endothelial growth factor (VEGF) concentration in culture supernatant by ELISA. Thalidomide 54-65 vascular endothelial growth factor A Homo sapiens 255-259 26462773-5 2015 The VEGF concentrations of combination group [(94.61 +- 5.46) ng/L decreased significantly, as compared to thalidomide group [(141.11 +- 3.70) ng/L and IFN group [(119.90 +- 2.00) ng/L (P < 0.05). Thalidomide 107-118 vascular endothelial growth factor A Homo sapiens 4-8 26462773-7 2015 CONCLUSION: Thalidomide and IFN could synergistically inhibit the proliferation of Kasumi-1 cells probably through inducing apoptosis via the mitochondrial pathway, death receptor pathway and P38 MAPK pathway, as well as inhibiting VEGF secretion. Thalidomide 12-23 vascular endothelial growth factor A Homo sapiens 232-236 26221080-6 2015 Thalidomide treatment after PQ exposure resulted in significantly reduced serum levels of IL-6, TNF-alpha, TGF-beta1 and COL1A1, as compared to PQ group. Thalidomide 0-11 interleukin 6 Rattus norvegicus 90-94 26221080-6 2015 Thalidomide treatment after PQ exposure resulted in significantly reduced serum levels of IL-6, TNF-alpha, TGF-beta1 and COL1A1, as compared to PQ group. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 96-105 26221080-6 2015 Thalidomide treatment after PQ exposure resulted in significantly reduced serum levels of IL-6, TNF-alpha, TGF-beta1 and COL1A1, as compared to PQ group. Thalidomide 0-11 transforming growth factor, beta 1 Rattus norvegicus 107-116 26221080-6 2015 Thalidomide treatment after PQ exposure resulted in significantly reduced serum levels of IL-6, TNF-alpha, TGF-beta1 and COL1A1, as compared to PQ group. Thalidomide 0-11 collagen type I alpha 1 chain Rattus norvegicus 121-127 26221080-7 2015 PCR analysis demonstrated that expression levels of IL-6, TNF-alpha, TGF-beta1 and COL1A1 in lung tissue were significantly increased after PQ exposure but reduced by thalidomide, which were confirmed by immunohistochemistry staining. Thalidomide 167-178 interleukin 6 Rattus norvegicus 52-56 26221080-7 2015 PCR analysis demonstrated that expression levels of IL-6, TNF-alpha, TGF-beta1 and COL1A1 in lung tissue were significantly increased after PQ exposure but reduced by thalidomide, which were confirmed by immunohistochemistry staining. Thalidomide 167-178 tumor necrosis factor Rattus norvegicus 58-67 26221080-7 2015 PCR analysis demonstrated that expression levels of IL-6, TNF-alpha, TGF-beta1 and COL1A1 in lung tissue were significantly increased after PQ exposure but reduced by thalidomide, which were confirmed by immunohistochemistry staining. Thalidomide 167-178 transforming growth factor, beta 1 Rattus norvegicus 69-78 26221080-7 2015 PCR analysis demonstrated that expression levels of IL-6, TNF-alpha, TGF-beta1 and COL1A1 in lung tissue were significantly increased after PQ exposure but reduced by thalidomide, which were confirmed by immunohistochemistry staining. Thalidomide 167-178 collagen type I alpha 1 chain Rattus norvegicus 83-89 25978432-1 2015 Immunomodulatory drugs (IMiDs) thalidomide, lenalidomide (Len) and pomalidomide trigger anti-tumor activities in multiple myeloma (MM) by targetting cereblon and thereby impacting IZF1/3, c-Myc and IRF4. Thalidomide 31-42 MYC proto-oncogene, bHLH transcription factor Homo sapiens 188-193 25929448-0 2015 Involvement of peripheral TRPV1 channels in the analgesic effects of thalidomide. Thalidomide 69-80 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 26-31 25929448-4 2015 Many studies suggest that the analgesic effect of thalidomide may be due to its immunomodulatory and anti-inflammatory properties as it suppresses the production of tumor necrosis factor alpha (TNF-alpha) selectively. Thalidomide 50-61 tumor necrosis factor Mus musculus 165-192 25929448-4 2015 Many studies suggest that the analgesic effect of thalidomide may be due to its immunomodulatory and anti-inflammatory properties as it suppresses the production of tumor necrosis factor alpha (TNF-alpha) selectively. Thalidomide 50-61 tumor necrosis factor Mus musculus 194-203 25929448-6 2015 In this study, we demonstrated that the peripheral vanilloid receptor 1 (TRPV1) channel was also involved in the analgesic effect of thalidomide in different cell and animal models. Thalidomide 133-144 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 51-71 25929448-6 2015 In this study, we demonstrated that the peripheral vanilloid receptor 1 (TRPV1) channel was also involved in the analgesic effect of thalidomide in different cell and animal models. Thalidomide 133-144 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 73-78 25929448-8 2015 And such attenuation in the TRPV1 activity was in a dose-dependent manner of thalidomide. Thalidomide 77-88 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 28-33 25929448-9 2015 In an acetic acid writhing test, pre-treatment of thalidomide decreased the writhing number in the wild type mice, while it did not happen in TRPV1 knockout mice, suggesting that the TRPV1 channel was involved in the pain relief by thalidomide. Thalidomide 50-61 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 183-188 25929448-9 2015 In an acetic acid writhing test, pre-treatment of thalidomide decreased the writhing number in the wild type mice, while it did not happen in TRPV1 knockout mice, suggesting that the TRPV1 channel was involved in the pain relief by thalidomide. Thalidomide 232-243 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 183-188 26171197-1 2015 The objective of this study was to assess the efficacy of granulocyte-macrophage colony-stimulating factor (GM-CSF) in combination with thalidomide for prostate-specific antigen (PSA) reduction in older patients (aged >=70 years, life expectancy of >1 year) with castration-resistant prostate cancer (CRPC). Thalidomide 136-147 kallikrein related peptidase 3 Homo sapiens 152-183 25929448-10 2015 Taken together, the study showed that TRPV1 channels were involved in the analgesic effects of thalidomide. Thalidomide 95-106 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 38-43 25929448-11 2015 Such alteration in the action of TRPV1 channels by thalidomide may help understand how thalidomide takes analgesic effect in the body in addition to its selective inhibition of TNF-alpha production. Thalidomide 51-62 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 33-38 25929448-11 2015 Such alteration in the action of TRPV1 channels by thalidomide may help understand how thalidomide takes analgesic effect in the body in addition to its selective inhibition of TNF-alpha production. Thalidomide 87-98 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 33-38 25840743-10 2015 In contrast, Thal and Dex had a direct inhibitory effect on the expression of EPCR and TM, and this inhibitory effect was especially significant when MM serum was added. Thalidomide 13-17 protein C receptor Homo sapiens 78-82 25978432-1 2015 Immunomodulatory drugs (IMiDs) thalidomide, lenalidomide (Len) and pomalidomide trigger anti-tumor activities in multiple myeloma (MM) by targetting cereblon and thereby impacting IZF1/3, c-Myc and IRF4. Thalidomide 31-42 interferon regulatory factor 4 Homo sapiens 198-202 25905462-8 2015 RESULTS: The mice with thalidomide treatment showed significantly reduced levels of allergen-induced BALF and lung inflammation, AHR, and the expression of a number of pro-inflammatory cytokines and mediators including Th2 related, IL-17 cytokines, and altered levels of allergen-specific IgG1/IgG2a. Thalidomide 23-34 heart and neural crest derivatives expressed 2 Mus musculus 219-222 25905462-8 2015 RESULTS: The mice with thalidomide treatment showed significantly reduced levels of allergen-induced BALF and lung inflammation, AHR, and the expression of a number of pro-inflammatory cytokines and mediators including Th2 related, IL-17 cytokines, and altered levels of allergen-specific IgG1/IgG2a. Thalidomide 23-34 interleukin 17A Mus musculus 232-237 25905462-8 2015 RESULTS: The mice with thalidomide treatment showed significantly reduced levels of allergen-induced BALF and lung inflammation, AHR, and the expression of a number of pro-inflammatory cytokines and mediators including Th2 related, IL-17 cytokines, and altered levels of allergen-specific IgG1/IgG2a. Thalidomide 23-34 LOC105243590 Mus musculus 289-293 25905462-8 2015 RESULTS: The mice with thalidomide treatment showed significantly reduced levels of allergen-induced BALF and lung inflammation, AHR, and the expression of a number of pro-inflammatory cytokines and mediators including Th2 related, IL-17 cytokines, and altered levels of allergen-specific IgG1/IgG2a. Thalidomide 23-34 immunoglobulin heavy variable V1-9 Mus musculus 294-299 25905462-9 2015 Of interesting note, thalidomide treatment significantly reduced expression levels of allergen- or Th2 cytokine-stimulated alternative activation of macrophages in vivo and in vitro. Thalidomide 21-32 heart and neural crest derivatives expressed 2 Mus musculus 99-102 25749497-5 2015 Thalidomide (Thal) has been used to treat multiple myeloma due to its inhibitory effects on IL-6-induced cell growth. Thalidomide 0-11 interleukin 6 Mus musculus 92-96 25818676-9 2015 Thalidomide also lowered significantly the number of TUNEL-positive cells, levels of Bax, caspase-3, lipoperoxidation, and pro-inflammatory cytokines, and increased the levels of SOD1, Bcl-2 and pAkt. Thalidomide 0-11 BCL2 associated X, apoptosis regulator Rattus norvegicus 85-88 25818676-9 2015 Thalidomide also lowered significantly the number of TUNEL-positive cells, levels of Bax, caspase-3, lipoperoxidation, and pro-inflammatory cytokines, and increased the levels of SOD1, Bcl-2 and pAkt. Thalidomide 0-11 caspase 3 Rattus norvegicus 90-99 25818676-9 2015 Thalidomide also lowered significantly the number of TUNEL-positive cells, levels of Bax, caspase-3, lipoperoxidation, and pro-inflammatory cytokines, and increased the levels of SOD1, Bcl-2 and pAkt. Thalidomide 0-11 superoxide dismutase 1 Rattus norvegicus 179-183 25818676-9 2015 Thalidomide also lowered significantly the number of TUNEL-positive cells, levels of Bax, caspase-3, lipoperoxidation, and pro-inflammatory cytokines, and increased the levels of SOD1, Bcl-2 and pAkt. Thalidomide 0-11 BCL2, apoptosis regulator Rattus norvegicus 185-190 25892862-0 2015 Thalidomide ameliorates portal hypertension via nitric oxide synthase independent reduced systolic blood pressure. Thalidomide 0-11 nitric oxide synthase 1, neuronal Mus musculus 48-69 25892862-4 2015 One hypothesis is that thalidomide destabilizes tumor necrosis factor alpha (TNFalpha) mRNA and therefore diminishes TNFalpha induction of nitric oxide synthase (NOS) and the production of nitric oxide (NO). Thalidomide 23-34 tumor necrosis factor Mus musculus 48-75 25892862-4 2015 One hypothesis is that thalidomide destabilizes tumor necrosis factor alpha (TNFalpha) mRNA and therefore diminishes TNFalpha induction of nitric oxide synthase (NOS) and the production of nitric oxide (NO). Thalidomide 23-34 tumor necrosis factor Mus musculus 77-85 25892862-4 2015 One hypothesis is that thalidomide destabilizes tumor necrosis factor alpha (TNFalpha) mRNA and therefore diminishes TNFalpha induction of nitric oxide synthase (NOS) and the production of nitric oxide (NO). Thalidomide 23-34 tumor necrosis factor Mus musculus 117-125 25892862-4 2015 One hypothesis is that thalidomide destabilizes tumor necrosis factor alpha (TNFalpha) mRNA and therefore diminishes TNFalpha induction of nitric oxide synthase (NOS) and the production of nitric oxide (NO). Thalidomide 23-34 nitric oxide synthase 1, neuronal Mus musculus 139-160 24950782-12 2015 Thalidomide treatment also significantly reduced tissue levels of the proinflammatory cytokines, MDA, MPO, and NO and increased anti-inflammatory cytokine IL-10. Thalidomide 0-11 myeloperoxidase Mus musculus 102-105 24950782-12 2015 Thalidomide treatment also significantly reduced tissue levels of the proinflammatory cytokines, MDA, MPO, and NO and increased anti-inflammatory cytokine IL-10. Thalidomide 0-11 interleukin 10 Mus musculus 155-160 25749497-5 2015 Thalidomide (Thal) has been used to treat multiple myeloma due to its inhibitory effects on IL-6-induced cell growth. Thalidomide 0-4 interleukin 6 Mus musculus 92-96 25749497-12 2015 In vitro studies revealed that Thal decreased 1) the expression of IL-1beta and IL-6 in human lung epithelial cells, and 2) CSE-induced apoptosis and the inhibition of cell growth, which may be the underlying mechanisms for the preventative effects of Thal on emphysema. Thalidomide 31-35 interleukin 1 beta Homo sapiens 67-75 25596877-15 2015 Our findings are in line with the findings reported in the literature and encourage further studies with TNF-alpha antagonists, in particular Thalidomide. Thalidomide 142-153 tumor necrosis factor Rattus norvegicus 105-114 25749497-12 2015 In vitro studies revealed that Thal decreased 1) the expression of IL-1beta and IL-6 in human lung epithelial cells, and 2) CSE-induced apoptosis and the inhibition of cell growth, which may be the underlying mechanisms for the preventative effects of Thal on emphysema. Thalidomide 31-35 interleukin 6 Homo sapiens 80-84 25617013-3 2015 Thalidomide has immunomodulatory and anti-tumor necrosis factor-alpha effects as well as antiangiogenic properties, making it useful for a broad spectrum of inflammatory disorders. Thalidomide 0-11 tumor necrosis factor Homo sapiens 42-69 25617013-8 2015 We review the mechanism of action and propose that thalidomide is an alternative treatment for patients who fail or have contraindications to anti-tumor necrosis factor-alpha biologics. Thalidomide 51-62 tumor necrosis factor Homo sapiens 147-174 25596877-3 2015 Recent reports indicate a protective role of Thalidomide, Etanercept, and Infliximab, all of which have anti-TNF-alpha activity, against cognitive and neuropathological changes in experimental and clinical studies. Thalidomide 45-56 tumor necrosis factor Rattus norvegicus 109-118 25891732-10 2015 RESULTS: Thalidomide significantly decreased the proliferation of CD4(+) Teffs in a dose-dependent manner (P < .01). Thalidomide 9-20 CD4 antigen Mus musculus 66-69 25891732-11 2015 In contrast, conversion to CD4(+)FoxP3(+) Tregs tended to increase by thalidomide treatment, although the increase was not statistically significant. Thalidomide 70-81 CD4 antigen Mus musculus 27-30 25891732-11 2015 In contrast, conversion to CD4(+)FoxP3(+) Tregs tended to increase by thalidomide treatment, although the increase was not statistically significant. Thalidomide 70-81 forkhead box P3 Mus musculus 33-38 25891732-12 2015 CONCLUSION: These findings suggest that thalidomide may have an immune modulatory effect by selectively suppressing CD4(+) Teff proliferation. Thalidomide 40-51 CD4 antigen Mus musculus 116-119 25886177-3 2015 In a 2012 case report, we described our breakthrough finding in two advanced HCC patients, of whom one achieved complete remission of liver tumors and the other a normalized alpha-fetoprotein level, along with complete remission of their lung metastases, after the concomitant use of thalidomide and cyproheptadine. Thalidomide 284-295 alpha fetoprotein Homo sapiens 174-191 25584775-9 2015 The combined treatment with pentoxifylline or thalidomide with morphine, at doses that are ineffective as single treatment, diminished G-CSF-induced hyperalgesia through inhibiting cytokine production. Thalidomide 46-57 colony stimulating factor 3 (granulocyte) Mus musculus 135-140 25584775-8 2015 G-CSF-induced hyperalgesia was dose-dependently inhibited by thalidomide (5-45mg/kg) and pentoxifylline (0.5-13.5mg/kg), and treatment with these drugs inhibited G-CSF-induced TNFalpha, IL-1beta and IL-10 production. Thalidomide 61-72 colony stimulating factor 3 (granulocyte) Mus musculus 0-5 25724264-4 2015 In both circumstances thalidomide improved the neurotoxicity induced by MPTP as seen by a significant raise of the striatal contents of dopamine and simultaneous decrease of monoamine-oxidase-B (MAO-B). Thalidomide 22-33 monoamine oxidase B Mus musculus 174-193 25724264-4 2015 In both circumstances thalidomide improved the neurotoxicity induced by MPTP as seen by a significant raise of the striatal contents of dopamine and simultaneous decrease of monoamine-oxidase-B (MAO-B). Thalidomide 22-33 monoamine oxidase B Mus musculus 195-200 25741973-1 2015 Neurologic tuberculous pseudoabscesses that clinically progress despite conventional antituberculosis therapy may be responsive to adjuvant thalidomide, a potent tumor necrosis factor-alpha inhibitor. Thalidomide 140-151 tumor necrosis factor Homo sapiens 162-189 25302852-0 2015 Thalidomide-prednisone maintenance following autologous stem cell transplant for multiple myeloma: effect on thrombin generation and procoagulant markers in NCIC CTG MY.10. Thalidomide 0-11 coagulation factor II, thrombin Homo sapiens 109-117 25370353-3 2015 Thalidomide (40 microg/ml) exerted differing effects on the activities of ADAM10 and NEP enzymes. Thalidomide 0-11 a disintegrin and metallopeptidase domain 10 Mus musculus 74-80 25370353-3 2015 Thalidomide (40 microg/ml) exerted differing effects on the activities of ADAM10 and NEP enzymes. Thalidomide 0-11 membrane metallo endopeptidase Mus musculus 85-88 25370353-6 2015 By contrast, in the 4THMpc cell line, 40 microg/ml thalidomide alone induced a 66.6% increase in ADAM10 enzyme activity. Thalidomide 51-62 a disintegrin and metallopeptidase domain 10 Mus musculus 97-103 25370353-7 2015 Radiotherapy alone and thalidomide with 60Co combined therapy caused a 33.3 and 40% inhibition of ADAM10 activity, respectively. Thalidomide 23-34 a disintegrin and metallopeptidase domain 10 Mus musculus 98-104 25370353-8 2015 In 4T1 cells, thalidomide alone caused a 40.9% increase in NEP activity. Thalidomide 14-25 membrane metallo endopeptidase Mus musculus 59-62 25370353-10 2015 In more aggressive 4THMpc cells, thalidomide alone caused a 26.6% increase in NEP activity. Thalidomide 33-44 membrane metallo endopeptidase Mus musculus 78-81 25370353-12 2015 To the best of our knowledge, the present study is the first to demonstrate that thalidomide alone or in combination with radiotherapy exhibits significant cytotoxic effects on 4T1 and 4THMpc mouse breast cancer cell lines indicating that this drug affects the enzymatic activity of ADAM10 and NEP in vitro. Thalidomide 81-92 a disintegrin and metallopeptidase domain 10 Mus musculus 283-289 25370353-12 2015 To the best of our knowledge, the present study is the first to demonstrate that thalidomide alone or in combination with radiotherapy exhibits significant cytotoxic effects on 4T1 and 4THMpc mouse breast cancer cell lines indicating that this drug affects the enzymatic activity of ADAM10 and NEP in vitro. Thalidomide 81-92 membrane metallo endopeptidase Mus musculus 294-297 25222109-8 2015 In addition, significant decreases of NAG release, TNF-alpha concentrations and p22(phox) protein levels in HK2 cells were observed in high glucose-treated group with thalidomide. Thalidomide 167-178 tumor necrosis factor Homo sapiens 51-60 25222109-8 2015 In addition, significant decreases of NAG release, TNF-alpha concentrations and p22(phox) protein levels in HK2 cells were observed in high glucose-treated group with thalidomide. Thalidomide 167-178 O-GlcNAcase Homo sapiens 38-41 25222109-8 2015 In addition, significant decreases of NAG release, TNF-alpha concentrations and p22(phox) protein levels in HK2 cells were observed in high glucose-treated group with thalidomide. Thalidomide 167-178 calcineurin like EF-hand protein 1 Homo sapiens 80-83 25687062-6 2015 Meanwhile, the level of plasma TM in the group of thalidomide combined with chemotherapy was higer than that in newly diagnosed patients (P < 0.05). Thalidomide 50-61 thrombomodulin Homo sapiens 31-33 25687062-12 2015 The level of plasma TM and D-Dimer can be elevated when thalidomide used, which indirectly suggested the tendency for thrombosis in MM patients. Thalidomide 56-67 thrombomodulin Homo sapiens 20-22 25569776-2 2015 The protein cereblon, which forms an E3 ubiquitin ligase compex together with damaged DNA-binding protein 1 (DDB1) and cullin 4A, has been recently indentified as a primary target of thalidomide and its C-terminal part as responsible for binding thalidomide within a domain carrying several invariant cysteine and tryptophan residues. Thalidomide 183-194 damage specific DNA binding protein 1 Homo sapiens 78-107 25569776-2 2015 The protein cereblon, which forms an E3 ubiquitin ligase compex together with damaged DNA-binding protein 1 (DDB1) and cullin 4A, has been recently indentified as a primary target of thalidomide and its C-terminal part as responsible for binding thalidomide within a domain carrying several invariant cysteine and tryptophan residues. Thalidomide 183-194 damage specific DNA binding protein 1 Homo sapiens 109-113 25569776-2 2015 The protein cereblon, which forms an E3 ubiquitin ligase compex together with damaged DNA-binding protein 1 (DDB1) and cullin 4A, has been recently indentified as a primary target of thalidomide and its C-terminal part as responsible for binding thalidomide within a domain carrying several invariant cysteine and tryptophan residues. Thalidomide 183-194 cullin 4A Homo sapiens 119-128 25441253-9 2015 RESULTS: In obese mice, thalidomide administration induced a reduction in adiposity accompanied by a reduction of tumor necrosis factor-alpha (TNF-alpha), leptin and MCP-1 adipose tissue production, macrophage infiltration and JNK activation. Thalidomide 24-35 tumor necrosis factor Mus musculus 114-141 25478868-7 2015 Thalidomide pretreatment significantly reduced the levels of pro-inflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6], malondialdehyde (MDA) and myeloperoxidase (MPO) activity. Thalidomide 0-11 tumor necrosis factor Mus musculus 89-122 25478868-7 2015 Thalidomide pretreatment significantly reduced the levels of pro-inflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6], malondialdehyde (MDA) and myeloperoxidase (MPO) activity. Thalidomide 0-11 interleukin 1 beta Mus musculus 124-146 25478868-7 2015 Thalidomide pretreatment significantly reduced the levels of pro-inflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6], malondialdehyde (MDA) and myeloperoxidase (MPO) activity. Thalidomide 0-11 interleukin 6 Mus musculus 148-152 25478868-7 2015 Thalidomide pretreatment significantly reduced the levels of pro-inflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6], malondialdehyde (MDA) and myeloperoxidase (MPO) activity. Thalidomide 0-11 myeloperoxidase Mus musculus 181-196 25478868-7 2015 Thalidomide pretreatment significantly reduced the levels of pro-inflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6], malondialdehyde (MDA) and myeloperoxidase (MPO) activity. Thalidomide 0-11 myeloperoxidase Mus musculus 198-201 25441253-9 2015 RESULTS: In obese mice, thalidomide administration induced a reduction in adiposity accompanied by a reduction of tumor necrosis factor-alpha (TNF-alpha), leptin and MCP-1 adipose tissue production, macrophage infiltration and JNK activation. Thalidomide 24-35 tumor necrosis factor Mus musculus 143-152 25441253-9 2015 RESULTS: In obese mice, thalidomide administration induced a reduction in adiposity accompanied by a reduction of tumor necrosis factor-alpha (TNF-alpha), leptin and MCP-1 adipose tissue production, macrophage infiltration and JNK activation. Thalidomide 24-35 chemokine (C-C motif) ligand 2 Mus musculus 166-171 25441253-9 2015 RESULTS: In obese mice, thalidomide administration induced a reduction in adiposity accompanied by a reduction of tumor necrosis factor-alpha (TNF-alpha), leptin and MCP-1 adipose tissue production, macrophage infiltration and JNK activation. Thalidomide 24-35 mitogen-activated protein kinase 8 Mus musculus 227-230 25441253-10 2015 TNF-alpha and leptin serum levels were also reduced by thalidomide treatment in obese mice. Thalidomide 55-66 tumor necrosis factor Mus musculus 0-9 26763359-0 2015 CD200 Expression on Plasma Cell Myeloma Cells is Associated with the Efficacies of Bortezomib, Lenalidomide and Thalidomide. Thalidomide 112-123 CD200 molecule Homo sapiens 0-5 23634876-1 2015 A 61-year-old man with newly diagnosed INT-1 risk myelodysplastic syndrome--refractory cytopenia with multilineage dysplasia (MDS-RCMD) was not responsive to treatment, such as androgen, thalidomide, granulocyte--colony stimulating factor (G-CSF) combined with erythropoietin (EPO), interleukin-11 (IL-11) and thrombopoietin (TPO), and became transfusion dependent. Thalidomide 187-198 Wnt family member 1 Homo sapiens 39-44 25186973-9 2014 Interestingly, an anti-inflammatory drug, such as a corticosteroid or thalidomide, but not anti-bacterial or anti-fungal drugs, improved CT image findings and reduced their KL-6 levels. Thalidomide 70-81 mucin 1, cell surface associated Homo sapiens 173-177 25326112-0 2014 Inhibition of endothelial Slit2/Robo1 signaling by thalidomide restrains angiogenesis by blocking the PI3K/Akt pathway. Thalidomide 51-62 slit guidance ligand 2 Homo sapiens 26-31 25326112-0 2014 Inhibition of endothelial Slit2/Robo1 signaling by thalidomide restrains angiogenesis by blocking the PI3K/Akt pathway. Thalidomide 51-62 roundabout guidance receptor 1 Homo sapiens 32-37 25326112-0 2014 Inhibition of endothelial Slit2/Robo1 signaling by thalidomide restrains angiogenesis by blocking the PI3K/Akt pathway. Thalidomide 51-62 AKT serine/threonine kinase 1 Homo sapiens 107-110 25326112-4 2014 PURPOSE: The aim of this study was to explore the expression and effects of Robo1 and Slit2 in angiodysplasia and to identify the possible therapeutic mechanisms of thalidomide. Thalidomide 165-176 roundabout guidance receptor 1 Homo sapiens 76-81 25326112-10 2014 High concentrations of Slit2 increased the levels of VEGF in HUVECs via signaling through the PI3K/Akt pathway-an effect that could be inhibited by thalidomide. Thalidomide 148-159 slit guidance ligand 2 Homo sapiens 23-28 25326112-10 2014 High concentrations of Slit2 increased the levels of VEGF in HUVECs via signaling through the PI3K/Akt pathway-an effect that could be inhibited by thalidomide. Thalidomide 148-159 vascular endothelial growth factor A Homo sapiens 53-57 25326112-10 2014 High concentrations of Slit2 increased the levels of VEGF in HUVECs via signaling through the PI3K/Akt pathway-an effect that could be inhibited by thalidomide. Thalidomide 148-159 AKT serine/threonine kinase 1 Homo sapiens 99-102 24912813-4 2014 The results showed that Thd significantly improved the survival rate, reduced the infiltration of inflammatory cells and cytokine (e.g., IL-6, TNF-alpha) and chemokine (e.g., RANTES, IP-10) levels, and inhibited activated p-NFkappaB p65 in infected mice. Thalidomide 24-27 interleukin 6 Mus musculus 137-141 24912813-4 2014 The results showed that Thd significantly improved the survival rate, reduced the infiltration of inflammatory cells and cytokine (e.g., IL-6, TNF-alpha) and chemokine (e.g., RANTES, IP-10) levels, and inhibited activated p-NFkappaB p65 in infected mice. Thalidomide 24-27 tumor necrosis factor Mus musculus 143-152 24912813-4 2014 The results showed that Thd significantly improved the survival rate, reduced the infiltration of inflammatory cells and cytokine (e.g., IL-6, TNF-alpha) and chemokine (e.g., RANTES, IP-10) levels, and inhibited activated p-NFkappaB p65 in infected mice. Thalidomide 24-27 chemokine (C-C motif) ligand 5 Mus musculus 175-181 24912813-4 2014 The results showed that Thd significantly improved the survival rate, reduced the infiltration of inflammatory cells and cytokine (e.g., IL-6, TNF-alpha) and chemokine (e.g., RANTES, IP-10) levels, and inhibited activated p-NFkappaB p65 in infected mice. Thalidomide 24-27 chemokine (C-X-C motif) ligand 10 Mus musculus 183-188 24912813-4 2014 The results showed that Thd significantly improved the survival rate, reduced the infiltration of inflammatory cells and cytokine (e.g., IL-6, TNF-alpha) and chemokine (e.g., RANTES, IP-10) levels, and inhibited activated p-NFkappaB p65 in infected mice. Thalidomide 24-27 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 233-236 25579023-10 2014 CONCLUSIONS: Lipid peroxide damage was one of the mechanisms of paraquat poisioning, thalidomide could attenuate paraquat-induced acute lung injury and its mechanism may be activating the Nrf2-ARE signaling pathway to protect mouse from Lipid peroxide damage. Thalidomide 85-96 nuclear factor, erythroid derived 2, like 2 Mus musculus 188-192 24392952-11 2014 Compared with BOR and DEX (minimum 72 and 56%, respectively), the PBF of THA (37%) was significantly lower (P < 0.001). Thalidomide 73-76 PTTG1 interacting protein Homo sapiens 66-69 24859059-9 2014 Thalidomide analogs 1 and 2 demonstrated more potent activity to suppress expression levels of IL-6, IL-8, TNF-alpha, VEGF165, and MMP-2 than thalidomide. Thalidomide 0-11 interleukin 6 Homo sapiens 95-99 25015938-6 2014 Moreover, a detrimental effect of thalidomide, even when administered in association with bortezomib, was observed in patients with abnormal chromosome 1 as well as in those with 17p deletion, while the benefit of adding thalidomide to the bortezomib-melphalan-prednisone regimen was noted in patients carrying an aggressive CD19(+)/CD117(-) bone marrow plasma cell immunophenotype. Thalidomide 34-45 CD19 molecule Homo sapiens 325-329 24718779-8 2014 Further, levels of NK-1R in the cisplatin, thalidomide, and granisetron group were significantly increased at both 5 and 33 h (p < 0.05 vs. control group), with no obvious difference among these three groups. Thalidomide 43-54 tachykinin receptor 1 Rattus norvegicus 19-24 25108355-0 2014 Structure of the human Cereblon-DDB1-lenalidomide complex reveals basis for responsiveness to thalidomide analogs. Thalidomide 94-105 damage specific DNA binding protein 1 Homo sapiens 32-36 25108355-1 2014 The Cul4-Rbx1-DDB1-Cereblon E3 ubiquitin ligase complex is the target of thalidomide, lenalidomide and pomalidomide, therapeutically important drugs for multiple myeloma and other B-cell malignancies. Thalidomide 73-84 ring-box 1 Homo sapiens 9-13 25108355-1 2014 The Cul4-Rbx1-DDB1-Cereblon E3 ubiquitin ligase complex is the target of thalidomide, lenalidomide and pomalidomide, therapeutically important drugs for multiple myeloma and other B-cell malignancies. Thalidomide 73-84 damage specific DNA binding protein 1 Homo sapiens 14-18 25108355-4 2014 A hydrophobic pocket in the thalidomide-binding domain (TBD) of CRBN accommodates the glutarimide moiety of lenalidomide, whereas the isoindolinone ring is exposed to solvent. Thalidomide 28-39 cereblon Homo sapiens 64-68 24939146-0 2014 Thalidomide alleviates acute pancreatitis-associated lung injury via down-regulation of NFkappaB induced TNF-alpha. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 105-114 24939146-1 2014 AIMS: We studied the effect of thalidomide on NFkappaB-induced TNF-alpha in acute pancreatitis-associated lung injury in the rat. Thalidomide 31-42 tumor necrosis factor Rattus norvegicus 63-72 24939146-6 2014 CONCLUSION: Thalidomide may inhibit TNF-alpha expression via down-regulation of the NFkappaB signaling pathway to alleviate acute pancreatitis-associated lung injury in rats. Thalidomide 12-23 tumor necrosis factor Rattus norvegicus 36-45 24687382-1 2014 Cereblon (CRBN) is essential for the anti-myeloma (MM) activity of immunomodulatory drugs (IMiDs), such as thalidomide and lenalidomide. Thalidomide 107-118 cereblon Homo sapiens 10-14 24687382-3 2014 Using immunohistochemical (IHC) staining on paraffin-embedded bone marrow sections, the expression of CRBN protein in myeloma cells (MCs) was assessed in 40 relapsed/refractory MM (RRMM) patients who received lenalidomide/dexamethasone (LD) and 45 and 22 newly diagnosed MM (NDMM) patients who received thalidomide/dexamethasone (TD) and melphalan/bortezomib/prednisolone (MVP), respectively. Thalidomide 303-314 cereblon Homo sapiens 102-106 25141548-4 2014 Extracts (25, 50 and 100 microg) were screened in the vascular endothelial growth factor (VEGF) induced angiogenesis in inovo chick chorioallontoic membrane assay (CAM) at various concentrations using thalidomide and normal saline as positive and untreated control groups respectively. Thalidomide 201-212 vascular endothelial growth factor A Gallus gallus 54-88 25141548-4 2014 Extracts (25, 50 and 100 microg) were screened in the vascular endothelial growth factor (VEGF) induced angiogenesis in inovo chick chorioallontoic membrane assay (CAM) at various concentrations using thalidomide and normal saline as positive and untreated control groups respectively. Thalidomide 201-212 vascular endothelial growth factor A Gallus gallus 90-94 24893330-5 2014 Inhibiting the synthesis of tumor necrosis factor-alpha (TNF-alpha) by thalidomide prevented DN, accompanied by strongly blocking the up-regulation of Nav1.7, TNF-alpha and p-nucleus factor-kappa B (p-NF-kappaB) in DRG. Thalidomide 71-82 tumor necrosis factor Rattus norvegicus 28-55 24893330-5 2014 Inhibiting the synthesis of tumor necrosis factor-alpha (TNF-alpha) by thalidomide prevented DN, accompanied by strongly blocking the up-regulation of Nav1.7, TNF-alpha and p-nucleus factor-kappa B (p-NF-kappaB) in DRG. Thalidomide 71-82 tumor necrosis factor Rattus norvegicus 57-66 24893330-5 2014 Inhibiting the synthesis of tumor necrosis factor-alpha (TNF-alpha) by thalidomide prevented DN, accompanied by strongly blocking the up-regulation of Nav1.7, TNF-alpha and p-nucleus factor-kappa B (p-NF-kappaB) in DRG. Thalidomide 71-82 tumor necrosis factor Rattus norvegicus 159-168 25043012-0 2014 Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide. Thalidomide 63-74 damage specific DNA binding protein 1 Homo sapiens 17-21 25043012-0 2014 Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide. Thalidomide 63-74 cereblon Homo sapiens 22-26 25043012-0 2014 Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide. Thalidomide 63-74 Cbl proto-oncogene like 2 Homo sapiens 27-46 25043012-4 2014 Here we present crystal structures of the DDB1-CRBN complex bound to thalidomide, lenalidomide and pomalidomide. Thalidomide 69-80 damage specific DNA binding protein 1 Homo sapiens 42-46 25043012-4 2014 Here we present crystal structures of the DDB1-CRBN complex bound to thalidomide, lenalidomide and pomalidomide. Thalidomide 69-80 cereblon Homo sapiens 47-51 24840055-3 2014 However, the exact mechanism of action has been difficult to quantify until recently when it was shown that another IMiD, thalidomide, binds to an E3 ubiquitin ligase complex constituent, CRBN. Thalidomide 122-133 cereblon Homo sapiens 188-192 24859059-9 2014 Thalidomide analogs 1 and 2 demonstrated more potent activity to suppress expression levels of IL-6, IL-8, TNF-alpha, VEGF165, and MMP-2 than thalidomide. Thalidomide 0-11 matrix metallopeptidase 2 Homo sapiens 131-136 24859059-9 2014 Thalidomide analogs 1 and 2 demonstrated more potent activity to suppress expression levels of IL-6, IL-8, TNF-alpha, VEGF165, and MMP-2 than thalidomide. Thalidomide 0-11 C-X-C motif chemokine ligand 8 Homo sapiens 101-105 24859059-9 2014 Thalidomide analogs 1 and 2 demonstrated more potent activity to suppress expression levels of IL-6, IL-8, TNF-alpha, VEGF165, and MMP-2 than thalidomide. Thalidomide 0-11 tumor necrosis factor Homo sapiens 107-116 24754268-0 2014 Behavioural, metabolic, and endothelial effects of the TNF-alpha suppressor thalidomide on rats subjected to chronic mild stress and fed an atherogenic diet. Thalidomide 76-87 tumor necrosis factor Rattus norvegicus 55-64 25610725-1 2014 Lenalidomide and its analogs, thalidomide and pomalidomide, specifically inhibit growth of mature B-cell lymphomas, including multiple myeloma, and induce interleukin-2 (IL-2) release from T cells. Thalidomide 30-41 interleukin 2 Homo sapiens 155-168 25610725-1 2014 Lenalidomide and its analogs, thalidomide and pomalidomide, specifically inhibit growth of mature B-cell lymphomas, including multiple myeloma, and induce interleukin-2 (IL-2) release from T cells. Thalidomide 30-41 interleukin 2 Homo sapiens 170-174 24739012-1 2014 Lenalidomide (Revlimid; Selleck Chemicals, Houston, TX, USA), an analogue of thalidomide, possesses potent cytokine modulatory capacity through inhibition of cytokines such as tumour necrosis factor (TNF)-alpha, a cytokine pivotal for the onset and development of complications in obesity and diabetes mellitus. Thalidomide 77-88 tumor necrosis factor Mus musculus 176-210 24667589-1 2014 Thalidomide is an oral immunomodulatory and anti-inflammatory drug with antitumor necrosis factor-alpha (TNF-alpha) activity. Thalidomide 0-11 tumor necrosis factor Homo sapiens 105-114 24667589-3 2014 We report the effect and tolerability of thalidomide in 3 patients with moderate-to-severe CD who were not responsive to anti-TNF-alpha therapies, and review the relevant literature. Thalidomide 41-52 tumor necrosis factor Homo sapiens 126-135 24667589-8 2014 In our CD patients who experienced loss of response or were unable to tolerate anti-TNF-alpha drugs, thalidomide was an effective and well-tolerated therapy for inducing and maintaining long-term remission. Thalidomide 101-112 tumor necrosis factor Homo sapiens 84-93 24803825-10 2014 CONCLUSION: Thalidomide can down-regulate serum VEGF level in EC patients, and combined with radiotherapy may improve treatment outcome. Thalidomide 12-23 vascular endothelial growth factor A Homo sapiens 48-52 24530998-4 2014 Furthermore, the TNF-alpha synthesis inhibitor thalidomide significantly blocked the activation of both isoforms JNK1 and JNK2 in the DRG and attenuated mechanical allodynia following BTZ treatment. Thalidomide 47-58 tumor necrosis factor Mus musculus 17-26 24530998-4 2014 Furthermore, the TNF-alpha synthesis inhibitor thalidomide significantly blocked the activation of both isoforms JNK1 and JNK2 in the DRG and attenuated mechanical allodynia following BTZ treatment. Thalidomide 47-58 mitogen-activated protein kinase 8 Mus musculus 113-117 24530998-4 2014 Furthermore, the TNF-alpha synthesis inhibitor thalidomide significantly blocked the activation of both isoforms JNK1 and JNK2 in the DRG and attenuated mechanical allodynia following BTZ treatment. Thalidomide 47-58 mitogen-activated protein kinase 9 Mus musculus 122-126 24735834-5 2014 Some investigations revealed that thalidomide could elicit immunomodulatory or neuromodulatory properties by affecting different targets, including cytokines (such as TNF alpha), neurotransmitters, and nitric oxide (NO). Thalidomide 34-45 tumor necrosis factor Mus musculus 167-176 24361040-2 2014 We hypothesized that because thalidomide inhibits secretion of tumor necrosis factor alpha (TNF-alpha), survival of animals that received thalidomide would be significantly prolonged. Thalidomide 29-40 tumor necrosis factor Oryctolagus cuniculus 63-90 24361040-2 2014 We hypothesized that because thalidomide inhibits secretion of tumor necrosis factor alpha (TNF-alpha), survival of animals that received thalidomide would be significantly prolonged. Thalidomide 29-40 tumor necrosis factor Oryctolagus cuniculus 92-101 24361040-2 2014 We hypothesized that because thalidomide inhibits secretion of tumor necrosis factor alpha (TNF-alpha), survival of animals that received thalidomide would be significantly prolonged. Thalidomide 138-149 tumor necrosis factor Oryctolagus cuniculus 63-90 24361040-2 2014 We hypothesized that because thalidomide inhibits secretion of tumor necrosis factor alpha (TNF-alpha), survival of animals that received thalidomide would be significantly prolonged. Thalidomide 138-149 tumor necrosis factor Oryctolagus cuniculus 92-101 24769540-3 2014 Total three studies were finally included, indicating that docetaxel plus thalidomide exhibited better survival prognosis and greater prostate-specific antigen (PSA) decline than docetaxel alone. Thalidomide 74-85 kallikrein related peptidase 3 Homo sapiens 134-165 24134145-11 2014 Thalidomide is the drug of choice for severe atypical lepra reactions because of its anti-tumor necrosis factor-alpha action. Thalidomide 0-11 tumor necrosis factor Homo sapiens 90-117 24692723-5 2014 RESULTS: RA up-regulated FGF2 gene expression, which was abrogated by thalidomide. Thalidomide 70-81 fibroblast growth factor 2 Homo sapiens 25-29 24520265-16 2014 Thus, thalidomide eased the degree of BLM-induced pulmonary fibrosis in rats by downregulating p-JNK and alpha-SMA expression. Thalidomide 6-17 actin gamma 2, smooth muscle Rattus norvegicus 105-114 24487918-0 2014 Thalidomide influences atherogenesis in aortas of ApoE(-/-)/LDLR (-/-) double knockout mice: a nano-CT study. Thalidomide 0-11 apolipoprotein E Mus musculus 50-54 24487918-0 2014 Thalidomide influences atherogenesis in aortas of ApoE(-/-)/LDLR (-/-) double knockout mice: a nano-CT study. Thalidomide 0-11 low density lipoprotein receptor Mus musculus 60-64 25053990-0 2014 Internal ribosome entry site of bFGF is the target of thalidomide for IMiDs development in multiple myeloma. Thalidomide 54-65 fibroblast growth factor 2 Homo sapiens 32-36 24460184-0 2014 Thalidomide increases human hepatic cytochrome P450 3A enzymes by direct activation of the pregnane X receptor. Thalidomide 0-11 nuclear receptor subfamily 1 group I member 2 Homo sapiens 91-110 25053990-4 2014 Interestingly, we further demonstrated that although thalidomide down-regulated bFGF translation through the inhibition of IRES even at 0.1 mug/ml, pomalidomide did not have a similar affect bFGF levels. Thalidomide 53-64 fibroblast growth factor 2 Homo sapiens 80-84 25053990-5 2014 A colony formation assay demonstrated that thalidomide and the bFGF knock-down clones caused a significant reduction in the clonogenic ability of MM cells, and treatment with exogenous bFGF can recover the clonogenic ability of thalidomide-treated cells and knock-down clones, but not that of pomalidomide-treated cells. Thalidomide 228-239 fibroblast growth factor 2 Homo sapiens 63-67 25053990-5 2014 A colony formation assay demonstrated that thalidomide and the bFGF knock-down clones caused a significant reduction in the clonogenic ability of MM cells, and treatment with exogenous bFGF can recover the clonogenic ability of thalidomide-treated cells and knock-down clones, but not that of pomalidomide-treated cells. Thalidomide 228-239 fibroblast growth factor 2 Homo sapiens 185-189 25053990-6 2014 This implies that thalidomide, but not pomalidomide, targets the IRES of FGF-2. Thalidomide 18-29 fibroblast growth factor 2 Homo sapiens 73-78 24460184-8 2014 Thalidomide enhanced levels of P450 3A4 and 2B6 mRNA, protein expression, and/or oxidation activity in human hepatocytes, indirectly suggesting the activation of upstream transcription factors involved in detoxication, e.g., the nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR). Thalidomide 0-11 nuclear receptor subfamily 1 group I member 2 Homo sapiens 247-266 24460184-8 2014 Thalidomide enhanced levels of P450 3A4 and 2B6 mRNA, protein expression, and/or oxidation activity in human hepatocytes, indirectly suggesting the activation of upstream transcription factors involved in detoxication, e.g., the nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR). Thalidomide 0-11 nuclear receptor subfamily 1 group I member 2 Homo sapiens 268-271 24460184-8 2014 Thalidomide enhanced levels of P450 3A4 and 2B6 mRNA, protein expression, and/or oxidation activity in human hepatocytes, indirectly suggesting the activation of upstream transcription factors involved in detoxication, e.g., the nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR). Thalidomide 0-11 nuclear receptor subfamily 1 group I member 3 Homo sapiens 277-309 24460184-8 2014 Thalidomide enhanced levels of P450 3A4 and 2B6 mRNA, protein expression, and/or oxidation activity in human hepatocytes, indirectly suggesting the activation of upstream transcription factors involved in detoxication, e.g., the nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR). Thalidomide 0-11 nuclear receptor subfamily 1 group I member 3 Homo sapiens 311-314 24460184-10 2014 To investigate direct engagement and functional alteration of PXR and CAR by thalidomide, we utilized a peptide microarray with 154 coregulator-derived nuclear receptor-interaction motifs and coregulator and nuclear receptor boxes, which serves as a sensor for nuclear receptor conformation and activity status as a function of ligand. Thalidomide 77-88 nuclear receptor subfamily 1 group I member 2 Homo sapiens 62-65 24460184-10 2014 To investigate direct engagement and functional alteration of PXR and CAR by thalidomide, we utilized a peptide microarray with 154 coregulator-derived nuclear receptor-interaction motifs and coregulator and nuclear receptor boxes, which serves as a sensor for nuclear receptor conformation and activity status as a function of ligand. Thalidomide 77-88 nuclear receptor subfamily 1 group I member 3 Homo sapiens 70-73 24460184-11 2014 Thalidomide and its human proximate metabolite 5-hydroxythalidomide displayed significant modulation of coregulator interaction with PXR and CAR ligand-binding domains, similar to established agonists for these receptors. Thalidomide 0-11 nuclear receptor subfamily 1 group I member 2 Homo sapiens 133-136 24460184-11 2014 Thalidomide and its human proximate metabolite 5-hydroxythalidomide displayed significant modulation of coregulator interaction with PXR and CAR ligand-binding domains, similar to established agonists for these receptors. Thalidomide 0-11 nuclear receptor subfamily 1 group I member 3 Homo sapiens 141-144 24460184-12 2014 These results collectively suggest that thalidomide acts as a ligand for PXR and CAR and causes enzyme induction leading to increased P450 enzyme activity. Thalidomide 40-51 nuclear receptor subfamily 1 group I member 2 Homo sapiens 73-76 24460184-12 2014 These results collectively suggest that thalidomide acts as a ligand for PXR and CAR and causes enzyme induction leading to increased P450 enzyme activity. Thalidomide 40-51 nuclear receptor subfamily 1 group I member 3 Homo sapiens 81-84 24219762-4 2014 In addition, in vitro effect of thalidomide on Ang2, Notch1 and Dll4 in human umbilical vein endothelial cells (HUVEC) and on HUVEC proliferation was also investigated during normoxic and hypoxic conditions. Thalidomide 32-43 angiopoietin 2 Homo sapiens 47-51 24219762-4 2014 In addition, in vitro effect of thalidomide on Ang2, Notch1 and Dll4 in human umbilical vein endothelial cells (HUVEC) and on HUVEC proliferation was also investigated during normoxic and hypoxic conditions. Thalidomide 32-43 notch receptor 1 Homo sapiens 53-59 24219762-0 2014 Thalidomide-induced angiopoietin 2, Notch1 and Dll4 downregulation under hypoxic condition in tissues with gastrointestinal vascular malformation and human umbilical vein endothelial cells. Thalidomide 0-11 angiopoietin 2 Homo sapiens 20-34 24219762-4 2014 In addition, in vitro effect of thalidomide on Ang2, Notch1 and Dll4 in human umbilical vein endothelial cells (HUVEC) and on HUVEC proliferation was also investigated during normoxic and hypoxic conditions. Thalidomide 32-43 delta like canonical Notch ligand 4 Homo sapiens 64-68 24219762-0 2014 Thalidomide-induced angiopoietin 2, Notch1 and Dll4 downregulation under hypoxic condition in tissues with gastrointestinal vascular malformation and human umbilical vein endothelial cells. Thalidomide 0-11 notch receptor 1 Homo sapiens 36-42 24219762-0 2014 Thalidomide-induced angiopoietin 2, Notch1 and Dll4 downregulation under hypoxic condition in tissues with gastrointestinal vascular malformation and human umbilical vein endothelial cells. Thalidomide 0-11 delta like canonical Notch ligand 4 Homo sapiens 47-51 24219762-9 2014 CONCLUSION: Ang2, Notch1, Dll4 and Hif-1alpha may play an important role in the pathogenesis of GIVM and may be potential targets of thalidomide in the treatment of the disease. Thalidomide 133-144 angiopoietin 2 Homo sapiens 12-16 24219762-9 2014 CONCLUSION: Ang2, Notch1, Dll4 and Hif-1alpha may play an important role in the pathogenesis of GIVM and may be potential targets of thalidomide in the treatment of the disease. Thalidomide 133-144 notch receptor 1 Homo sapiens 18-24 24219762-9 2014 CONCLUSION: Ang2, Notch1, Dll4 and Hif-1alpha may play an important role in the pathogenesis of GIVM and may be potential targets of thalidomide in the treatment of the disease. Thalidomide 133-144 delta like canonical Notch ligand 4 Homo sapiens 26-30 24219762-9 2014 CONCLUSION: Ang2, Notch1, Dll4 and Hif-1alpha may play an important role in the pathogenesis of GIVM and may be potential targets of thalidomide in the treatment of the disease. Thalidomide 133-144 hypoxia inducible factor 1 subunit alpha Homo sapiens 35-45 24596000-0 2014 Thalidomide regulation of NF-kappaB proteins limits Tregs activity in chronic lymphocytic leukemia. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 26-35 24292623-6 2014 Analysis of myeloma cell lines revealed that loss of IKZF1 and IKZF3 is both necessary and sufficient for lenalidomide"s therapeutic effect, suggesting that the antitumor and teratogenic activities of thalidomide-like drugs are dissociable. Thalidomide 201-212 IKAROS family zinc finger 1 Homo sapiens 53-58 24292623-6 2014 Analysis of myeloma cell lines revealed that loss of IKZF1 and IKZF3 is both necessary and sufficient for lenalidomide"s therapeutic effect, suggesting that the antitumor and teratogenic activities of thalidomide-like drugs are dissociable. Thalidomide 201-212 IKAROS family zinc finger 3 Homo sapiens 63-68 24596000-4 2014 Thalidomide"s influence on NF-kappaB proteins and on regulatory T cells (Treg) in CLL was investigated. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 27-36 24596000-10 2014 RESULTS: It was found that thalidomide regulates NF-kappaB activity differentially, and the activity of certain NF-kappaB components correlated with TNF levels and T regulatory cell (CD4 + CD25 high GITR + ). Thalidomide 27-38 nuclear factor kappa B subunit 1 Homo sapiens 49-58 24596000-10 2014 RESULTS: It was found that thalidomide regulates NF-kappaB activity differentially, and the activity of certain NF-kappaB components correlated with TNF levels and T regulatory cell (CD4 + CD25 high GITR + ). Thalidomide 27-38 nuclear factor kappa B subunit 1 Homo sapiens 112-121 24596000-10 2014 RESULTS: It was found that thalidomide regulates NF-kappaB activity differentially, and the activity of certain NF-kappaB components correlated with TNF levels and T regulatory cell (CD4 + CD25 high GITR + ). Thalidomide 27-38 tumor necrosis factor Homo sapiens 149-152 24596000-10 2014 RESULTS: It was found that thalidomide regulates NF-kappaB activity differentially, and the activity of certain NF-kappaB components correlated with TNF levels and T regulatory cell (CD4 + CD25 high GITR + ). Thalidomide 27-38 CD4 molecule Homo sapiens 183-186 24596000-10 2014 RESULTS: It was found that thalidomide regulates NF-kappaB activity differentially, and the activity of certain NF-kappaB components correlated with TNF levels and T regulatory cell (CD4 + CD25 high GITR + ). Thalidomide 27-38 interleukin 2 receptor subunit alpha Homo sapiens 189-193 24596000-10 2014 RESULTS: It was found that thalidomide regulates NF-kappaB activity differentially, and the activity of certain NF-kappaB components correlated with TNF levels and T regulatory cell (CD4 + CD25 high GITR + ). Thalidomide 27-38 TNF receptor superfamily member 18 Homo sapiens 199-203 24596000-11 2014 CONCLUSIONS: These results might indicate that thalidomide not only regulates TNF but also directly interferes with NF-kappaB components. Thalidomide 47-58 tumor necrosis factor Homo sapiens 78-81 24596000-11 2014 CONCLUSIONS: These results might indicate that thalidomide not only regulates TNF but also directly interferes with NF-kappaB components. Thalidomide 47-58 nuclear factor kappa B subunit 1 Homo sapiens 116-125 24451995-6 2014 Similar to valproate, thalidomide was classified as a Class 2 agent, with weak embryotoxicity, by the mEST criteria, and was classified as Category 3 embryotoxic based on our criteria. Thalidomide 22-33 sulfotransferase family 1E, member 1 Mus musculus 102-106 25531359-9 2014 In patients not responding to anti-TNF therapy, thalidomide or tacrolimus may be considered. Thalidomide 48-59 tumor necrosis factor Homo sapiens 35-38 24118365-0 2014 Do baseline Cereblon gene expression and IL-6 receptor expression determine the response to thalidomide-dexamethasone treatment in multiple myeloma patients? Thalidomide 92-103 cereblon Homo sapiens 12-20 24118365-2 2014 Cereblon (CRBN) expression was described to be essential for the activity of thalidomide. Thalidomide 77-88 cereblon Homo sapiens 0-8 24118365-2 2014 Cereblon (CRBN) expression was described to be essential for the activity of thalidomide. Thalidomide 77-88 cereblon Homo sapiens 10-14 24118365-6 2014 The aim of the study was to evaluate CRBN and IL-6R expressions and their impact on clinical efficacy of dexamethasone-thalidomide therapy in multiple myeloma (MM) patients, in addition to their association with other clinical and prognostic parameters. Thalidomide 119-130 cereblon Homo sapiens 37-41 24118365-6 2014 The aim of the study was to evaluate CRBN and IL-6R expressions and their impact on clinical efficacy of dexamethasone-thalidomide therapy in multiple myeloma (MM) patients, in addition to their association with other clinical and prognostic parameters. Thalidomide 119-130 interleukin 6 receptor Homo sapiens 46-51 24931258-21 2014 On day 7 the expressions of TNF-alpha and IL-1beta in the thalidomide treatment group were lower than in the TNBS model group. Thalidomide 58-69 tumor necrosis factor Rattus norvegicus 28-37 24931258-21 2014 On day 7 the expressions of TNF-alpha and IL-1beta in the thalidomide treatment group were lower than in the TNBS model group. Thalidomide 58-69 interleukin 1 beta Rattus norvegicus 42-50 24931258-22 2014 On day 14, the expressions of TNF-alpha and IL-1beta in the thalidomide treatment group were significantly lower than in the TNBS model group (P < 0.05). Thalidomide 60-71 tumor necrosis factor Rattus norvegicus 30-39 24931258-22 2014 On day 14, the expressions of TNF-alpha and IL-1beta in the thalidomide treatment group were significantly lower than in the TNBS model group (P < 0.05). Thalidomide 60-71 interleukin 1 beta Rattus norvegicus 44-52 24931258-23 2014 On day 4, the IL-10 levels of the thalidomide treatment group became significantly elevated. Thalidomide 34-45 interleukin 10 Rattus norvegicus 14-19 23510862-3 2014 GM-CSF in combination with thalidomide induces prostate-specific antigen (PSA) responses in 20% to 25% of patients with castration-resistant prostate cancer (CRPC). Thalidomide 27-38 kallikrein related peptidase 3 Homo sapiens 47-78 24275522-7 2014 In cancer cell lines, thalidomide is antiangiogenic and antiproliferative via suppression of tumor necrosis factor-alpha (TNF-alpha) and downstream effects on the nuclear factor (NFkappaB) cascade. Thalidomide 22-33 tumor necrosis factor Homo sapiens 93-120 24275522-7 2014 In cancer cell lines, thalidomide is antiangiogenic and antiproliferative via suppression of tumor necrosis factor-alpha (TNF-alpha) and downstream effects on the nuclear factor (NFkappaB) cascade. Thalidomide 22-33 tumor necrosis factor Homo sapiens 122-131 24275522-7 2014 In cancer cell lines, thalidomide is antiangiogenic and antiproliferative via suppression of tumor necrosis factor-alpha (TNF-alpha) and downstream effects on the nuclear factor (NFkappaB) cascade. Thalidomide 22-33 nuclear factor kappa B subunit 1 Homo sapiens 179-187 24154490-7 2014 Specifically, 71 drug-like compounds with known in vivo effects, including thalidomide, were examined in the hPST. Thalidomide 75-86 sulfotransferase family 1A member 1 Homo sapiens 109-113 23810410-10 2013 Thalidomide decreased production of inflammatory and fibrogenic cytokine tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, and transforming growth factor (TGF)-beta1. Thalidomide 0-11 interleukin 1 beta Mus musculus 108-130 24030118-0 2013 Increased PAC-1 expression among patients with multiple myeloma on concurrent thalidomide and warfarin. Thalidomide 78-89 dual specificity phosphatase 2 Homo sapiens 10-15 24030118-7 2013 Increased expression marker for PAC-1 was observed after 4 weeks of thalidomide treatment (P < 0.05) indicating one aspect of platelet activation activity seen in these patients. Thalidomide 68-79 dual specificity phosphatase 2 Homo sapiens 32-37 24210217-6 2013 Bortezomib, thalidomide or lenalidomide extended OS of FGFR3 and/or c-MAF-positive patients. Thalidomide 12-23 fibroblast growth factor receptor 3 Homo sapiens 55-60 24210217-6 2013 Bortezomib, thalidomide or lenalidomide extended OS of FGFR3 and/or c-MAF-positive patients. Thalidomide 12-23 MAF bZIP transcription factor Homo sapiens 68-73 24055736-0 2013 Polymorphisms in the endothelial nitric oxide synthase gene in thalidomide embryopathy. Thalidomide 63-74 nitric oxide synthase 3 Homo sapiens 21-54 24187103-7 2013 Treatment with meloxicam, thalidomide, and etanercept for 1 month significantly reduced percentages of IL-21+CXCR5+CD4+ T cells and concentrations of serum IL-21 (p < 0.001, p < 0.001, respectively), accompanied by significantly minimized disease activity in drug responders, but not in the drug nonresponders. Thalidomide 26-37 interleukin 21 Homo sapiens 103-108 24187103-7 2013 Treatment with meloxicam, thalidomide, and etanercept for 1 month significantly reduced percentages of IL-21+CXCR5+CD4+ T cells and concentrations of serum IL-21 (p < 0.001, p < 0.001, respectively), accompanied by significantly minimized disease activity in drug responders, but not in the drug nonresponders. Thalidomide 26-37 C-X-C motif chemokine receptor 5 Homo sapiens 109-114 24187103-7 2013 Treatment with meloxicam, thalidomide, and etanercept for 1 month significantly reduced percentages of IL-21+CXCR5+CD4+ T cells and concentrations of serum IL-21 (p < 0.001, p < 0.001, respectively), accompanied by significantly minimized disease activity in drug responders, but not in the drug nonresponders. Thalidomide 26-37 interleukin 21 Homo sapiens 156-161 23952807-9 2013 There was significant regression of vessels in CAM incubated with thalidomide dispersion, thalidomide-loaded implants and vitreous samples from group I when compared to control. Thalidomide 66-77 calmodulin Oryctolagus cuniculus 47-50 23952807-9 2013 There was significant regression of vessels in CAM incubated with thalidomide dispersion, thalidomide-loaded implants and vitreous samples from group I when compared to control. Thalidomide 90-101 calmodulin Oryctolagus cuniculus 47-50 23810410-10 2013 Thalidomide decreased production of inflammatory and fibrogenic cytokine tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, and transforming growth factor (TGF)-beta1. Thalidomide 0-11 interleukin 6 Mus musculus 132-136 23810410-10 2013 Thalidomide decreased production of inflammatory and fibrogenic cytokine tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, and transforming growth factor (TGF)-beta1. Thalidomide 0-11 transforming growth factor, beta 1 Mus musculus 142-180 23810410-11 2013 In addition thalidomide reduced myeloperoxidase (MPO), nitric oxide (NO), and hydroxyproline content in lung tissue. Thalidomide 12-23 myeloperoxidase Mus musculus 32-47 23810410-11 2013 In addition thalidomide reduced myeloperoxidase (MPO), nitric oxide (NO), and hydroxyproline content in lung tissue. Thalidomide 12-23 myeloperoxidase Mus musculus 49-52 23926306-4 2013 According to the gene profile, the downregulation of caspase-8 and caspase-10, and upregulation of oct3/4 and tgf-beta1, may be associated with THD modulation. Thalidomide 144-147 caspase 8 Homo sapiens 53-62 23926306-4 2013 According to the gene profile, the downregulation of caspase-8 and caspase-10, and upregulation of oct3/4 and tgf-beta1, may be associated with THD modulation. Thalidomide 144-147 caspase 10 Homo sapiens 67-77 23926306-4 2013 According to the gene profile, the downregulation of caspase-8 and caspase-10, and upregulation of oct3/4 and tgf-beta1, may be associated with THD modulation. Thalidomide 144-147 POU class 5 homeobox 1 Homo sapiens 99-105 23926306-4 2013 According to the gene profile, the downregulation of caspase-8 and caspase-10, and upregulation of oct3/4 and tgf-beta1, may be associated with THD modulation. Thalidomide 144-147 transforming growth factor beta 1 Homo sapiens 110-119 23767831-2 2013 Prior attempts to block vascular endothelial growth factor (VEGF) with sunitinib, sorafenib and thalidomide have obtained disappointing results. Thalidomide 96-107 vascular endothelial growth factor A Homo sapiens 24-58 23447383-8 2013 Now we observe the reduction of the NRP1 expression on Tregs in vitro, thereby suggesting a possible target of thalidomide action. Thalidomide 111-122 neuropilin 1 Homo sapiens 36-40 23767831-2 2013 Prior attempts to block vascular endothelial growth factor (VEGF) with sunitinib, sorafenib and thalidomide have obtained disappointing results. Thalidomide 96-107 vascular endothelial growth factor A Homo sapiens 60-64 23500539-7 2013 Furthermore, pretreatment with thalidomide inhibited the TGF-beta1-induced phosphorylation of p38 and Smad3, but not that of ERK1/2, JNK, and Smad2. Thalidomide 31-42 transforming growth factor beta 1 Homo sapiens 57-66 23722461-5 2013 RESULTS: Thalidomide (20 muM) not only inhibited cell proliferation after 24 h [fold increase of cell number, 0.85 +- 0.09 vs. 1.47 +- 0.14 (treatment vs. control group); P < 0.01] and 48 h of incubation (0.85 +- 0.10 vs. 1.97 +- 0.12; P < 0.001), it also inhibited cell migration and slowed wound closure at 24 h (P < 0.001). Thalidomide 9-20 latexin Homo sapiens 25-28 23722461-6 2013 Thalidomide significantly attenuated TGF-beta1-induced VEGF expression at both the mRNA and protein levels. Thalidomide 0-11 transforming growth factor beta 1 Homo sapiens 37-46 23722461-6 2013 Thalidomide significantly attenuated TGF-beta1-induced VEGF expression at both the mRNA and protein levels. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 55-59 23722461-7 2013 Incubation of thalidomide with cells stimulated with TGF-beta1 significantly inhibited their production of collagen. Thalidomide 14-25 transforming growth factor beta 1 Homo sapiens 53-62 23722461-8 2013 Thalidomide inhibited Smad3, STAT3, and subsequent p44/42 kinase phosphorylation. Thalidomide 0-11 SMAD family member 3 Homo sapiens 22-27 23722461-8 2013 Thalidomide inhibited Smad3, STAT3, and subsequent p44/42 kinase phosphorylation. Thalidomide 0-11 signal transducer and activator of transcription 3 Homo sapiens 29-34 23722461-8 2013 Thalidomide inhibited Smad3, STAT3, and subsequent p44/42 kinase phosphorylation. Thalidomide 0-11 interferon induced protein 44 Homo sapiens 51-54 23500539-0 2013 Thalidomide inhibits fibronectin production in TGF-beta1-treated normal and keloid fibroblasts via inhibition of the p38/Smad3 pathway. Thalidomide 0-11 fibronectin 1 Homo sapiens 21-32 23500539-0 2013 Thalidomide inhibits fibronectin production in TGF-beta1-treated normal and keloid fibroblasts via inhibition of the p38/Smad3 pathway. Thalidomide 0-11 transforming growth factor beta 1 Homo sapiens 47-56 24284039-0 2013 Thalidomide distinctly affected TNF-alpha, IL-6 and MMP secretion by an ovarian cancer cell line (SKOV-3) and primary ovarian cancer cells. Thalidomide 0-11 tumor necrosis factor Homo sapiens 32-41 24284039-0 2013 Thalidomide distinctly affected TNF-alpha, IL-6 and MMP secretion by an ovarian cancer cell line (SKOV-3) and primary ovarian cancer cells. Thalidomide 0-11 interleukin 6 Homo sapiens 43-47 24284039-1 2013 BACKGROUND: Thalidomide inhibits TNF-alpha production in lipopolysaccharide-stimulated monocytes. Thalidomide 12-23 tumor necrosis factor Homo sapiens 33-42 24284039-2 2013 The aim of this study was to evaluate the effect of thalidomide on TNF-alpha, IL-6 and MMP secretion in epithelial ovarian carcinoma cells. Thalidomide 52-63 tumor necrosis factor Homo sapiens 67-76 24284039-2 2013 The aim of this study was to evaluate the effect of thalidomide on TNF-alpha, IL-6 and MMP secretion in epithelial ovarian carcinoma cells. Thalidomide 52-63 interleukin 6 Homo sapiens 78-82 24284039-8 2013 Thalidomide also significantly decreased the capacity of SKOV-3 cells, but not primary epithelial ovarian carcinoma cells, to secrete MMP-9 and MMP-2. Thalidomide 0-11 matrix metallopeptidase 9 Homo sapiens 134-139 24284039-8 2013 Thalidomide also significantly decreased the capacity of SKOV-3 cells, but not primary epithelial ovarian carcinoma cells, to secrete MMP-9 and MMP-2. Thalidomide 0-11 matrix metallopeptidase 2 Homo sapiens 144-149 24284039-10 2013 CONCLUSION: Our study suggests that thalidomide distinctly affected TNF-alpha, IL-6 and MMPs secretion by an ovarian carcinoma cell line (SKOV-3) and primary ovarian cancer cells. Thalidomide 36-47 tumor necrosis factor Homo sapiens 68-77 24284039-10 2013 CONCLUSION: Our study suggests that thalidomide distinctly affected TNF-alpha, IL-6 and MMPs secretion by an ovarian carcinoma cell line (SKOV-3) and primary ovarian cancer cells. Thalidomide 36-47 interleukin 6 Homo sapiens 79-83 24284039-10 2013 CONCLUSION: Our study suggests that thalidomide distinctly affected TNF-alpha, IL-6 and MMPs secretion by an ovarian carcinoma cell line (SKOV-3) and primary ovarian cancer cells. Thalidomide 36-47 matrix metallopeptidase 2 Homo sapiens 88-92 23500539-0 2013 Thalidomide inhibits fibronectin production in TGF-beta1-treated normal and keloid fibroblasts via inhibition of the p38/Smad3 pathway. Thalidomide 0-11 mitogen-activated protein kinase 1 Homo sapiens 117-120 23500539-0 2013 Thalidomide inhibits fibronectin production in TGF-beta1-treated normal and keloid fibroblasts via inhibition of the p38/Smad3 pathway. Thalidomide 0-11 SMAD family member 3 Homo sapiens 121-126 23500539-3 2013 The aims were to examine possible therapeutic effects of thalidomide on fibronectin expression in transforming growth factor-beta1 (TGF-beta1)-treated normal fibroblasts (NFs) and keloid-derived fibroblasts (KFs) and the underlying mechanism of action, especially the involvement of mitogen-activated protein kinase (MAPKs) and Sma- and Mad-related family (Smads) pathways. Thalidomide 57-68 fibronectin 1 Homo sapiens 72-83 23500539-3 2013 The aims were to examine possible therapeutic effects of thalidomide on fibronectin expression in transforming growth factor-beta1 (TGF-beta1)-treated normal fibroblasts (NFs) and keloid-derived fibroblasts (KFs) and the underlying mechanism of action, especially the involvement of mitogen-activated protein kinase (MAPKs) and Sma- and Mad-related family (Smads) pathways. Thalidomide 57-68 transforming growth factor beta 1 Homo sapiens 98-130 23500539-7 2013 Furthermore, pretreatment with thalidomide inhibited the TGF-beta1-induced phosphorylation of p38 and Smad3, but not that of ERK1/2, JNK, and Smad2. Thalidomide 31-42 mitogen-activated protein kinase 1 Homo sapiens 94-97 23500539-5 2013 TGF-beta1 induced significant fibronectin expression in NFs and KFs and the effect was inhibited by pretreatment with thalidomide. Thalidomide 118-129 transforming growth factor beta 1 Homo sapiens 0-9 23500539-5 2013 TGF-beta1 induced significant fibronectin expression in NFs and KFs and the effect was inhibited by pretreatment with thalidomide. Thalidomide 118-129 fibronectin 1 Homo sapiens 30-41 23500539-7 2013 Furthermore, pretreatment with thalidomide inhibited the TGF-beta1-induced phosphorylation of p38 and Smad3, but not that of ERK1/2, JNK, and Smad2. Thalidomide 31-42 SMAD family member 3 Homo sapiens 102-107 23500539-8 2013 In addition, thalidomide pretreatment inhibited the TGF-beta-induced DNA binding activity of AP-1 and Smad3/4, caused fibronectin degradation by increasing the activity of matrix metalloproteinase 9, and decreased production of TGF-beta1 and fibronectin and the number of fibroblasts in an in vivo keloid model. Thalidomide 13-24 transforming growth factor beta 1 Homo sapiens 52-60 23500539-8 2013 In addition, thalidomide pretreatment inhibited the TGF-beta-induced DNA binding activity of AP-1 and Smad3/4, caused fibronectin degradation by increasing the activity of matrix metalloproteinase 9, and decreased production of TGF-beta1 and fibronectin and the number of fibroblasts in an in vivo keloid model. Thalidomide 13-24 SMAD family member 3 Homo sapiens 102-107 23500539-8 2013 In addition, thalidomide pretreatment inhibited the TGF-beta-induced DNA binding activity of AP-1 and Smad3/4, caused fibronectin degradation by increasing the activity of matrix metalloproteinase 9, and decreased production of TGF-beta1 and fibronectin and the number of fibroblasts in an in vivo keloid model. Thalidomide 13-24 fibronectin 1 Homo sapiens 118-129 23500539-8 2013 In addition, thalidomide pretreatment inhibited the TGF-beta-induced DNA binding activity of AP-1 and Smad3/4, caused fibronectin degradation by increasing the activity of matrix metalloproteinase 9, and decreased production of TGF-beta1 and fibronectin and the number of fibroblasts in an in vivo keloid model. Thalidomide 13-24 matrix metallopeptidase 9 Homo sapiens 172-198 23500539-8 2013 In addition, thalidomide pretreatment inhibited the TGF-beta-induced DNA binding activity of AP-1 and Smad3/4, caused fibronectin degradation by increasing the activity of matrix metalloproteinase 9, and decreased production of TGF-beta1 and fibronectin and the number of fibroblasts in an in vivo keloid model. Thalidomide 13-24 transforming growth factor beta 1 Homo sapiens 228-237 23500539-8 2013 In addition, thalidomide pretreatment inhibited the TGF-beta-induced DNA binding activity of AP-1 and Smad3/4, caused fibronectin degradation by increasing the activity of matrix metalloproteinase 9, and decreased production of TGF-beta1 and fibronectin and the number of fibroblasts in an in vivo keloid model. Thalidomide 13-24 fibronectin 1 Homo sapiens 242-253 23500539-9 2013 These results show that thalidomide has an antifibrotic effect on keloid fibroblasts that is caused by suppression of TGF-beta1-induced p38 and Smad3 signaling. Thalidomide 24-35 transforming growth factor beta 1 Homo sapiens 118-127 23500539-9 2013 These results show that thalidomide has an antifibrotic effect on keloid fibroblasts that is caused by suppression of TGF-beta1-induced p38 and Smad3 signaling. Thalidomide 24-35 mitogen-activated protein kinase 1 Homo sapiens 136-139 23500539-9 2013 These results show that thalidomide has an antifibrotic effect on keloid fibroblasts that is caused by suppression of TGF-beta1-induced p38 and Smad3 signaling. Thalidomide 24-35 SMAD family member 3 Homo sapiens 144-149 23531412-10 2013 We show that thalidomide activity on organizer formation in hydra depends on the activity of casein kinase1 and the abundance of beta-catenin. Thalidomide 13-24 catenin beta 1 Homo sapiens 129-141 23686003-0 2013 A myeloma cell line established from a patient refractory to thalidomide therapy revealed high-risk cytogenetic abnormalities and produced vascular endothelial growth factor. Thalidomide 61-72 vascular endothelial growth factor A Homo sapiens 139-173 23737889-0 2013 Effect of thalidomide on the proliferation of hepatoma cells assessed by osteopontin levels in nude mice. Thalidomide 10-21 secreted phosphoprotein 1 Mus musculus 73-84 23717811-15 2013 An analog of thalidomide, CC-5103 increases the secretion of critical cytokines of the tumor microenvironment, including IL-2, IFN-gamma, TNF-alpha, and IL-10, and is currently being evaluated in clinical trials for the treatment of recurrent or refractory pediatric central nervous system tumors. Thalidomide 13-24 interleukin 2 Homo sapiens 121-125 23717811-15 2013 An analog of thalidomide, CC-5103 increases the secretion of critical cytokines of the tumor microenvironment, including IL-2, IFN-gamma, TNF-alpha, and IL-10, and is currently being evaluated in clinical trials for the treatment of recurrent or refractory pediatric central nervous system tumors. Thalidomide 13-24 interferon gamma Homo sapiens 127-136 23717811-15 2013 An analog of thalidomide, CC-5103 increases the secretion of critical cytokines of the tumor microenvironment, including IL-2, IFN-gamma, TNF-alpha, and IL-10, and is currently being evaluated in clinical trials for the treatment of recurrent or refractory pediatric central nervous system tumors. Thalidomide 13-24 tumor necrosis factor Homo sapiens 138-147 23717811-15 2013 An analog of thalidomide, CC-5103 increases the secretion of critical cytokines of the tumor microenvironment, including IL-2, IFN-gamma, TNF-alpha, and IL-10, and is currently being evaluated in clinical trials for the treatment of recurrent or refractory pediatric central nervous system tumors. Thalidomide 13-24 interleukin 10 Homo sapiens 153-158 23244235-2 2013 Thalidomide has a significant anti-inflammatory effect by inhibiting TNF-alpha, which plays role in Abeta neurotoxicity. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 69-78 23583898-4 2013 Here, we show that thalidomide suppressed TNF-alpha-induced NF-kappaB activation and ATP-induced IL-1beta secretion, but did not interrupt the production of IL-1beta, IL-6, IL-8, and TNF-alpha in response to lipopolysaccharide in CGD monocytes. Thalidomide 19-30 tumor necrosis factor Homo sapiens 42-51 23583898-4 2013 Here, we show that thalidomide suppressed TNF-alpha-induced NF-kappaB activation and ATP-induced IL-1beta secretion, but did not interrupt the production of IL-1beta, IL-6, IL-8, and TNF-alpha in response to lipopolysaccharide in CGD monocytes. Thalidomide 19-30 interleukin 1 beta Homo sapiens 97-105 23737889-11 2013 Thalidomide may inhibit the generation of OPN and thereby inhibit the infiltration and metastasis of tumors; the immediate use of thalidomide following hepatectomy in the present study may block the invasion and metasis for liver cancer more effectively. Thalidomide 0-11 secreted phosphoprotein 1 Mus musculus 42-45 23345625-15 2013 Thalidomide treatment significantly reduced gene expression of these cytokines and the activation of the NF-kappaB in the renal tissue and the circulating levels of cytokines. Thalidomide 0-11 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 105-114 23434428-0 2013 Induction of apoptosis in multiple myeloma cells by a statin-thalidomide combination can be enhanced by p38 MAPK inhibition. Thalidomide 61-72 mitogen-activated protein kinase 14 Homo sapiens 104-107 23434428-4 2013 What is more, the combination of thalidomide with statins inhibited cell migration and decreased the production of VEGF and MMP-9 in MM cells more effectively than each of these drugs used separately. Thalidomide 33-44 vascular endothelial growth factor A Homo sapiens 115-119 23434428-4 2013 What is more, the combination of thalidomide with statins inhibited cell migration and decreased the production of VEGF and MMP-9 in MM cells more effectively than each of these drugs used separately. Thalidomide 33-44 matrix metallopeptidase 9 Homo sapiens 124-129 23434428-5 2013 The combination of simvastatin and thalidomide augmented caspase 8 and 3 activation, and the additional application of p38 MAPK inhibitor resulted in enhanced apoptosis of MM cells concomitant with increased caspase 9 and 3 activation, as well as JNK phosphorylation. Thalidomide 35-46 caspase 8 Homo sapiens 57-66 23404341-2 2013 It was recently reported that the thalidomide analog 3,6"-dithiothalidomide (3,6"-DT) can selectively inhibit the synthesis of TNF in cultured cells. Thalidomide 34-45 tumor necrosis factor Mus musculus 127-130 23279849-7 2013 Suppression of STAT3 correlated with the down-regulation of various gene products linked to tumor survival, proliferation, and angiogenesis and led to sensitization of tumor cells to thalidomide and bortezomib. Thalidomide 183-194 signal transducer and activator of transcription 3 Homo sapiens 15-20 23538327-8 2013 FINDINGS: Compared to other treatment groups, CD133+ cells treated with thalidomide alone produced more hematopoietic colonies. Thalidomide 72-83 prominin 1 Homo sapiens 46-51 23442845-12 2013 We concluded that fluoxetine, anti-TNF-alpha antibody and thalidomide were effective in preventing depressive-like behaviour and the increase in TNF-alpha levels in the hippocampus of mice induced by an i.c.v. Thalidomide 58-69 tumor necrosis factor Mus musculus 145-154 22966948-2 2013 A landmark study recently identified cereblon (CRBN) as a primary target of thalidomide teratogenicity. Thalidomide 76-87 cereblon Homo sapiens 47-51 22966948-3 2013 Subsequently it was demonstrated that CRBN is also required for the anti-myeloma activity of thalidomide and related drugs, the so-called immune-modulatory drugs (IMiDs). Thalidomide 93-104 cereblon Homo sapiens 38-42 23614289-16 2013 In animals administered thalidomide in the dose of 60 mg/kg p.o., reduction mass of tibia, femur, and L-4 vertebra has been observed. Thalidomide 24-35 ribosomal protein L3 Rattus norvegicus 102-105 23142220-8 2013 Importantly, after cessation of Thalidomide therapy, this effect was reversible in the CD8 compartment. Thalidomide 32-43 CD8a molecule Homo sapiens 87-90 23261764-6 2013 Furthermore, inhibition of TNF-alpha synthesis by intraperitoneal thalidomide prevented both mechanical allodynia and the up-regulation of IL-6 in DRGs following L5-VRT. Thalidomide 66-77 tumor necrosis factor Homo sapiens 27-36 23261764-6 2013 Furthermore, inhibition of TNF-alpha synthesis by intraperitoneal thalidomide prevented both mechanical allodynia and the up-regulation of IL-6 in DRGs following L5-VRT. Thalidomide 66-77 interleukin 6 Homo sapiens 139-143 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 tumor necrosis factor Mus musculus 115-124 23426142-0 2013 Effect of thalidomide on the expression of vascular endothelial growth factor in a rat model of liver regeneration. Thalidomide 10-21 vascular endothelial growth factor A Rattus norvegicus 43-77 23426142-2 2013 The present study investigated the influence of thalidomide, an antiangiogenic agent, on vascular endothelial growth factor (VEGF) expression and liver regeneration after 70% partial hepatectomy (PH) in rats. Thalidomide 48-59 vascular endothelial growth factor A Rattus norvegicus 89-123 23426142-2 2013 The present study investigated the influence of thalidomide, an antiangiogenic agent, on vascular endothelial growth factor (VEGF) expression and liver regeneration after 70% partial hepatectomy (PH) in rats. Thalidomide 48-59 vascular endothelial growth factor A Rattus norvegicus 125-129 23426142-10 2013 The immunohistochemical staining revealed a waved pattern of VEGF expression which advanced from the periportal to pericentral area in both groups, but a slower advancement was detected in thalidomide-treated rats. Thalidomide 189-200 vascular endothelial growth factor A Rattus norvegicus 61-65 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 interleukin 1 beta Mus musculus 126-134 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 interleukin 6 Mus musculus 136-140 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 142-151 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 170-179 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 180-183 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 tumor necrosis factor Mus musculus 330-339 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 interleukin 1 beta Mus musculus 341-349 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 interleukin 6 Mus musculus 351-355 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 170-179 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 170-179 23510834-8 2013 Compared with the paraquat poisoning group, the thalidomide treatment groups had significantly decreased levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65 and wet/dry ratios of the lung (P < 0.05), and the 150 mg/kg thalidomide treatment group showed the most significant decrease in the levels of TNF-alpha, IL-1beta, IL-6, NF-kappaB mRNA, and nuclear NF-kappaB p65. Thalidomide 48-59 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 395-398 22710762-7 2013 Exposure to thalidomide (100 microg/ml) reduced the intracytoplasmic pro-inflammatory cytokine production of neonatal monocytes and the IFN-gamma production of neonatal lymphocytes. Thalidomide 12-23 interferon gamma Homo sapiens 136-145 22710762-8 2013 In supernatants, the addition of thalidomide resulted in reduction of TNF-alpha, IL-6, IL-10 and, by trend, IFN-gamma. Thalidomide 33-44 tumor necrosis factor Homo sapiens 70-79 22710762-8 2013 In supernatants, the addition of thalidomide resulted in reduction of TNF-alpha, IL-6, IL-10 and, by trend, IFN-gamma. Thalidomide 33-44 interleukin 6 Homo sapiens 81-85 22710762-8 2013 In supernatants, the addition of thalidomide resulted in reduction of TNF-alpha, IL-6, IL-10 and, by trend, IFN-gamma. Thalidomide 33-44 interleukin 10 Homo sapiens 87-92 22710762-8 2013 In supernatants, the addition of thalidomide resulted in reduction of TNF-alpha, IL-6, IL-10 and, by trend, IFN-gamma. Thalidomide 33-44 interferon gamma Homo sapiens 108-117 22710762-10 2013 Bcl-2 expression tended to be increased after addition of thalidomide. Thalidomide 58-69 BCL2 apoptosis regulator Homo sapiens 0-5 23233657-1 2013 Recently, cereblon (CRBN) expression was found to be essential for the activity of thalidomide and lenalidomide. Thalidomide 83-94 cereblon Homo sapiens 20-24 23233657-2 2013 In the present study, we investigated whether the clinical efficacy of thalidomide in multiple myeloma is associated with CRBN expression in myeloma cells. Thalidomide 71-82 cereblon Homo sapiens 122-126 23233657-7 2013 We conclude that CRBN expression may be associated with the clinical efficacy of thalidomide. Thalidomide 81-92 cereblon Homo sapiens 17-21 23405115-1 2013 Thalidomide is a tumor necrosis factor alpha (TNFalpha) inhibitor which has been found to have abilities against tumor growth, angiogenesis and inflammation. Thalidomide 0-11 tumor necrosis factor Mus musculus 17-44 23165864-8 2013 Genetic factors including single nucleotide polymorphisms (SNPs) that change cytochrome P450 (CYP) activity and epigenetic regulation that modifies CYP expression levels may contribute to the changes in pharmacokinetics and adverse drug reactions (ADRs) of thalidomide. Thalidomide 257-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 23165864-8 2013 Genetic factors including single nucleotide polymorphisms (SNPs) that change cytochrome P450 (CYP) activity and epigenetic regulation that modifies CYP expression levels may contribute to the changes in pharmacokinetics and adverse drug reactions (ADRs) of thalidomide. Thalidomide 257-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 23233155-1 2013 The CTD (cyclophosphamide, thalidomide, and dexamethasone) regimen is known to be an effective primary therapy in patients with newly diagnosed multiple myeloma (MM). Thalidomide 27-38 CTD Homo sapiens 4-7 23405115-1 2013 Thalidomide is a tumor necrosis factor alpha (TNFalpha) inhibitor which has been found to have abilities against tumor growth, angiogenesis and inflammation. Thalidomide 0-11 tumor necrosis factor Mus musculus 46-54 23405115-9 2013 We found that, in addition of dramatic decrease in the activation of both astrocytes and microglia, thalidomide significantly reduces Abeta load and plaque formation. Thalidomide 100-111 amyloid beta (A4) precursor protein Mus musculus 134-139 23405115-10 2013 Furthermore, we found a significant decrease in BACE1 level and activity with long-term thalidomide application. Thalidomide 88-99 beta-site APP cleaving enzyme 1 Mus musculus 48-53 22901832-9 2012 Thalidomide treatment also significantly decreased plasma levels of nitric oxide and pancreatic proinflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, IL-12, IL-17 and interferon (IFN)-gamma)] while increased anti-inflammatory cytokine IL-10. Thalidomide 0-11 tumor necrosis factor Mus musculus 123-156 22923758-2 2012 Moreover, thalidomide modulates the activity of NF-kappaB, which can up-regulate the expression of downstream genes involved in the pathophysiology of LN. Thalidomide 10-21 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 48-57 22923758-9 2012 Also, treatment with 10 mg/kg of thalidomide significantly decreased immune complex accumulation, reduced the serum concentration of anti-dsDNA, IgG2a and IgG2b and inhibited nuclear translocation of NF-kappaB in kidney tissues, comparable to standard therapy for LN. Thalidomide 33-44 immunoglobulin heavy variable V1-9 Mus musculus 145-150 22923758-9 2012 Also, treatment with 10 mg/kg of thalidomide significantly decreased immune complex accumulation, reduced the serum concentration of anti-dsDNA, IgG2a and IgG2b and inhibited nuclear translocation of NF-kappaB in kidney tissues, comparable to standard therapy for LN. Thalidomide 33-44 immunoglobulin heavy constant gamma 2B Mus musculus 155-160 22923758-9 2012 Also, treatment with 10 mg/kg of thalidomide significantly decreased immune complex accumulation, reduced the serum concentration of anti-dsDNA, IgG2a and IgG2b and inhibited nuclear translocation of NF-kappaB in kidney tissues, comparable to standard therapy for LN. Thalidomide 33-44 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 200-209 22901832-9 2012 Thalidomide treatment also significantly decreased plasma levels of nitric oxide and pancreatic proinflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, IL-12, IL-17 and interferon (IFN)-gamma)] while increased anti-inflammatory cytokine IL-10. Thalidomide 0-11 interleukin 1 beta Mus musculus 158-180 22901832-9 2012 Thalidomide treatment also significantly decreased plasma levels of nitric oxide and pancreatic proinflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, IL-12, IL-17 and interferon (IFN)-gamma)] while increased anti-inflammatory cytokine IL-10. Thalidomide 0-11 interleukin 6 Mus musculus 182-186 22901832-9 2012 Thalidomide treatment also significantly decreased plasma levels of nitric oxide and pancreatic proinflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, IL-12, IL-17 and interferon (IFN)-gamma)] while increased anti-inflammatory cytokine IL-10. Thalidomide 0-11 interleukin 17A Mus musculus 195-200 22901832-9 2012 Thalidomide treatment also significantly decreased plasma levels of nitric oxide and pancreatic proinflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, IL-12, IL-17 and interferon (IFN)-gamma)] while increased anti-inflammatory cytokine IL-10. Thalidomide 0-11 interferon gamma Mus musculus 205-227 22901832-9 2012 Thalidomide treatment also significantly decreased plasma levels of nitric oxide and pancreatic proinflammatory cytokines [tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, IL-12, IL-17 and interferon (IFN)-gamma)] while increased anti-inflammatory cytokine IL-10. Thalidomide 0-11 interleukin 10 Mus musculus 273-278 22855579-0 2012 Thalidomide in total therapy 2 overcomes inferior prognosis of myeloma with low expression of the glucocorticoid receptor gene NR3C1. Thalidomide 0-11 nuclear receptor subfamily 3 group C member 1 Homo sapiens 98-121 22552008-2 2012 The ubiquitously expressed E3 ligase protein cereblon (CRBN) has been identified as the primary teratogenic target of thalidomide. Thalidomide 118-129 cereblon Homo sapiens 45-53 22552008-2 2012 The ubiquitously expressed E3 ligase protein cereblon (CRBN) has been identified as the primary teratogenic target of thalidomide. Thalidomide 118-129 cereblon Homo sapiens 55-59 22552008-3 2012 Our studies demonstrate that thalidomide, lenalidomide and another immunomodulatory drug, pomalidomide, bound endogenous CRBN and recombinant CRBN-DNA damage binding protein-1 (DDB1) complexes. Thalidomide 29-40 cereblon Homo sapiens 121-125 22552008-3 2012 Our studies demonstrate that thalidomide, lenalidomide and another immunomodulatory drug, pomalidomide, bound endogenous CRBN and recombinant CRBN-DNA damage binding protein-1 (DDB1) complexes. Thalidomide 29-40 damage specific DNA binding protein 1 Homo sapiens 142-175 22552008-3 2012 Our studies demonstrate that thalidomide, lenalidomide and another immunomodulatory drug, pomalidomide, bound endogenous CRBN and recombinant CRBN-DNA damage binding protein-1 (DDB1) complexes. Thalidomide 29-40 damage specific DNA binding protein 1 Homo sapiens 177-181 22552008-5 2012 Lenalidomide and pomalidomide inhibited autoubiquitination of CRBN in HEK293T cells expressing thalidomide-binding competent wild-type CRBN, but not thalidomide-binding defective CRBN(YW/AA). Thalidomide 95-106 cereblon Homo sapiens 62-66 23076705-0 2012 Unexpected remission of hepatocellular carcinoma (HCC) with lung metastasis to the combination therapy of thalidomide and cyproheptadine: report of two cases and a preliminary HCC cell line study. Thalidomide 106-117 HCC Homo sapiens 50-53 23076705-0 2012 Unexpected remission of hepatocellular carcinoma (HCC) with lung metastasis to the combination therapy of thalidomide and cyproheptadine: report of two cases and a preliminary HCC cell line study. Thalidomide 106-117 HCC Homo sapiens 176-179 23076705-1 2012 We reported two cases of hepatocellular carcinoma (HCC) with lung metastases who were treated with a combination of thalidomide and cyproheptadine. Thalidomide 116-127 HCC Homo sapiens 51-54 22705333-10 2012 Likewise, the expression of FGF-1 showed weaker cytoplasmic positivity in the group treated with thalidomide compared with the control group. Thalidomide 97-108 fibroblast growth factor 1 Mus musculus 28-33 22705333-11 2012 The levels of two cytokines, VEGF (pro-angiogenic) and TNF-alpha (pro-inflammatory) were decreased in tumor samples of thalidomide-treated group compared with the control group. Thalidomide 119-130 vascular endothelial growth factor A Mus musculus 29-33 22705333-11 2012 The levels of two cytokines, VEGF (pro-angiogenic) and TNF-alpha (pro-inflammatory) were decreased in tumor samples of thalidomide-treated group compared with the control group. Thalidomide 119-130 tumor necrosis factor Mus musculus 55-64 22855579-0 2012 Thalidomide in total therapy 2 overcomes inferior prognosis of myeloma with low expression of the glucocorticoid receptor gene NR3C1. Thalidomide 0-11 nuclear receptor subfamily 3 group C member 1 Homo sapiens 127-132 22855579-3 2012 RESULTS: Low NR3C1 expression levels had a negative impact on progression-free survival (PFS; HR, 1.47; P = 0.030) and overall survival (OS; HR, 1.90; P = 0.002) in the no-thalidomide arm. Thalidomide 172-183 nuclear receptor subfamily 3 group C member 1 Homo sapiens 13-18 22855579-4 2012 Conversely, there was a significant clinical benefit of thalidomide for patients with low receptor levels (OS: HR, 0.54; P = 0.015; PFS: HR, 0.54; P = 0.004), mediated most likely by thalidomide"s upregulation of NR3C1. Thalidomide 56-67 nuclear receptor subfamily 3 group C member 1 Homo sapiens 213-218 22855579-7 2012 The identification of an interaction term between thalidomide and NR3C1 underscores the importance of pharmacogenomic studies in the systematic study of new drugs. Thalidomide 50-61 nuclear receptor subfamily 3 group C member 1 Homo sapiens 66-71 22843924-9 2012 Net reduced levels of vascular endothelial growth factor and basic fibroblast growth factor cytokines were observed in the peripheral blood and the bone marrow of thalidomide-treated patients. Thalidomide 163-174 vascular endothelial growth factor A Homo sapiens 22-56 22880820-9 2012 Tumor necrosis factor (TNF)-alpha (p=0.04) and its receptors TNFR1 (p=0.04), TNFR2 (p=0.04), and interleukin (IL)-8 (p=0.04) were statistically significant in the thalidomide group. Thalidomide 163-174 tumor necrosis factor Homo sapiens 0-33 22880820-9 2012 Tumor necrosis factor (TNF)-alpha (p=0.04) and its receptors TNFR1 (p=0.04), TNFR2 (p=0.04), and interleukin (IL)-8 (p=0.04) were statistically significant in the thalidomide group. Thalidomide 163-174 TNF receptor superfamily member 1A Homo sapiens 61-66 22139971-0 2012 Vascular endothelial growth factor (VEGF) gene polymorphisms may influence the efficacy of thalidomide in multiple myeloma. Thalidomide 91-102 vascular endothelial growth factor A Homo sapiens 0-34 22139971-0 2012 Vascular endothelial growth factor (VEGF) gene polymorphisms may influence the efficacy of thalidomide in multiple myeloma. Thalidomide 91-102 vascular endothelial growth factor A Homo sapiens 36-40 22139971-4 2012 In this study, we evaluated the association between genetic variations in the VEGF gene in patients with multiple myeloma and time to treatment failure (TTF) after high-dose melphalan and stem cell support (HDT), overall survival (OS) and efficacy of the anti-angiogenic drug thalidomide. Thalidomide 276-287 vascular endothelial growth factor A Homo sapiens 78-82 22139971-9 2012 In conclusion, the haplotype ACG in the VEGF gene may influence the efficacy of thalidomide in multiple myeloma. Thalidomide 80-91 vascular endothelial growth factor A Homo sapiens 40-44 22329352-5 2012 Consolidation thalidomide conferred an improved PFS in patients with normal FGFR3 expression (41 vs. 19 months, p =0.02), but there was no improvement in patients with up-regulated FGFR3 (31 vs. 29 months, p =0.76). Thalidomide 14-25 fibroblast growth factor receptor 3 Homo sapiens 76-81 23094815-4 2012 Greater success was achieved with thalidomide regimen (CTD: cyclophosphamide, thalidomide, dexamethasone, 10 cycles, 3/2009-1/2010) leading to reduction in the size of the hypervascularized lymph nodes (almost by 50%) as well as their radiopharmaceutical (fluorodeoxyglucose) uptake as seen on a combined positron emission tomography and computed tomography (PET/CT) scan imaging. Thalidomide 34-45 CTD Homo sapiens 55-58 23094815-4 2012 Greater success was achieved with thalidomide regimen (CTD: cyclophosphamide, thalidomide, dexamethasone, 10 cycles, 3/2009-1/2010) leading to reduction in the size of the hypervascularized lymph nodes (almost by 50%) as well as their radiopharmaceutical (fluorodeoxyglucose) uptake as seen on a combined positron emission tomography and computed tomography (PET/CT) scan imaging. Thalidomide 78-89 CTD Homo sapiens 55-58 22422151-0 2012 Oncogenic CUL4A determines the response to thalidomide treatment in prostate cancer. Thalidomide 43-54 cullin 4A Homo sapiens 10-15 22422151-2 2012 Here we demonstrate that CUL4A plays an oncogenic role in prostate cancer development and prostate cancer cells with higher level of CUL4A are particularly sensitive to thalidomide treatment. Thalidomide 169-180 cullin 4A Homo sapiens 25-30 22422151-2 2012 Here we demonstrate that CUL4A plays an oncogenic role in prostate cancer development and prostate cancer cells with higher level of CUL4A are particularly sensitive to thalidomide treatment. Thalidomide 169-180 cullin 4A Homo sapiens 133-138 22422151-6 2012 We further show that the sensitivity to thalidomide is positively correlated with CUL4A expression in a panel of prostate cell lines. Thalidomide 40-51 cullin 4A Homo sapiens 82-87 22422151-7 2012 Ectopic CUL4A expression greatly enhanced sensitivity to thalidomide, while its downregulation conferred resistance to this drug. Thalidomide 57-68 cullin 4A Homo sapiens 8-13 22422151-8 2012 Mechanistically, thalidomide decreased CUL4A in a time- and dose-dependent manner, consequently leading to inaction of ERK pathway. Thalidomide 17-28 cullin 4A Homo sapiens 39-44 22422151-8 2012 Mechanistically, thalidomide decreased CUL4A in a time- and dose-dependent manner, consequently leading to inaction of ERK pathway. Thalidomide 17-28 mitogen-activated protein kinase 1 Homo sapiens 119-122 22422151-10 2012 Our results offer the first evidence that CUL4A is a potential therapeutic target for prostate cancer and may serve as a biomarker for assessing prognosis of human prostate cancer and response to thalidomide treatment. Thalidomide 196-207 cullin 4A Homo sapiens 42-47 22731394-4 2012 In this study, we investigated the effects of a novel thalidomide-based TNF-alpha lowering drug, 3,6"-dithiothalidomide, on hippocampal progenitor cell proliferation, neurogenesis and, memory tasks after intracerebroventricular injection of beta-amyloid (Ass)(1-42) peptide. Thalidomide 54-65 tumor necrosis factor Homo sapiens 72-81 22527432-4 2012 RESULTS: IL17A, the hallmark of TH17, was consistently seen before and after thalidomide treatment, confirming the TH17 subset to be involved in ENL and potentially up-regulated by thalidomide. Thalidomide 181-192 interleukin 17A Homo sapiens 9-14 22498745-1 2012 In a randomized, phase 3 study, superior complete/near-complete response (CR/nCR) rates and extended progression-free survival were demonstrated with bortezomib-thalidomide-dexamethasone (VTD) versus thalidomide-dexamethasone (TD) as induction therapy before, and consolidation after, double autologous stem cell transplantation for newly diagnosed myeloma patients (intention-to-treat analysis; VTD, n = 236; TD, n = 238). Thalidomide 161-172 Neutrophil migration (granulocyte glycoprotein) Homo sapiens 77-80 22770990-0 2012 Novel thalidomide analogues from diamines inhibit pro-inflammatory cytokine production and CD80 expression while enhancing IL-10. Thalidomide 6-17 CD80 molecule Homo sapiens 91-95 22770990-0 2012 Novel thalidomide analogues from diamines inhibit pro-inflammatory cytokine production and CD80 expression while enhancing IL-10. Thalidomide 6-17 interleukin 10 Homo sapiens 123-128 22488443-5 2012 Given the high serum and plasma levels of VEGF observed in POEMS patients, the use of anti-angiogenetic drugs such as thalidomide and lenalidomide and other drugs with anti-VEGF and anti-TNF effect such as bortezomib have been considered to treat this syndrome. Thalidomide 118-129 vascular endothelial growth factor A Homo sapiens 42-46 22488443-5 2012 Given the high serum and plasma levels of VEGF observed in POEMS patients, the use of anti-angiogenetic drugs such as thalidomide and lenalidomide and other drugs with anti-VEGF and anti-TNF effect such as bortezomib have been considered to treat this syndrome. Thalidomide 118-129 vascular endothelial growth factor A Homo sapiens 173-177 22488443-5 2012 Given the high serum and plasma levels of VEGF observed in POEMS patients, the use of anti-angiogenetic drugs such as thalidomide and lenalidomide and other drugs with anti-VEGF and anti-TNF effect such as bortezomib have been considered to treat this syndrome. Thalidomide 118-129 tumor necrosis factor Homo sapiens 187-190 22098296-14 2012 Thalidomide suppressed bFGF-induced proliferation significantly and decreased VEGF expression, both at the protein and mRNA levels. Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 23-27 22098296-14 2012 Thalidomide suppressed bFGF-induced proliferation significantly and decreased VEGF expression, both at the protein and mRNA levels. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 78-82 22098296-15 2012 Thalidomide also inhibited HIF-1alpha in a dose-dependent manner (P < 0.05). Thalidomide 0-11 hypoxia inducible factor 1 subunit alpha Homo sapiens 27-37 22098296-17 2012 Thalidomide can suppress VEGF, either induced by HIF-1alpha or bFGF. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 25-29 22098296-17 2012 Thalidomide can suppress VEGF, either induced by HIF-1alpha or bFGF. Thalidomide 0-11 hypoxia inducible factor 1 subunit alpha Homo sapiens 49-59 22098296-17 2012 Thalidomide can suppress VEGF, either induced by HIF-1alpha or bFGF. Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 63-67 22855035-6 2012 RESULTS: The VEGF expressions in brucine- or thalidomide-treated mice were significantly reduced as compared with mice in the model group (P <0.01). Thalidomide 45-56 vascular endothelial growth factor A Mus musculus 13-17 22855035-9 2012 Further, VEGF expression was significantly increased in the low- and medium-dose brucine groups compared with the thalidomide group (P <0.05). Thalidomide 114-125 vascular endothelial growth factor A Mus musculus 9-13 21833519-0 2012 Clinical response to thalidomide and colchicine in two siblings with Behcet"s disease carrying a single mutated MEFV allele. Thalidomide 21-32 MEFV innate immuity regulator, pyrin Homo sapiens 112-116 22237514-8 2012 We discovered that the mechanism for thalidomide inhibiting adipogenesis was the down-regulation of PPARgamma, rather than C/EBPbeta and C/EBPdelta. Thalidomide 37-48 peroxisome proliferator activated receptor gamma Homo sapiens 100-109 22249337-15 2012 The combination of thalidomide (TNF-alpha inhibitor) and dipyrone (COX inhibitor) may be used as a potential therapeutic agent for the treatment of diabetic neuropathy. Thalidomide 19-30 tumor necrosis factor Rattus norvegicus 32-41 22883339-8 2012 CONCLUSION: Ang2, Notch1 and Dll4 may correlate with the pathogenesis of GIVM, while thalidomide can concentration-dependently down-regulate the expression of Ang2, Notch1 and Dll4, which may be one of the mechanism that thalidomide play a therapeutic role in GIVM. Thalidomide 85-96 angiopoietin 2 Homo sapiens 159-163 22883339-8 2012 CONCLUSION: Ang2, Notch1 and Dll4 may correlate with the pathogenesis of GIVM, while thalidomide can concentration-dependently down-regulate the expression of Ang2, Notch1 and Dll4, which may be one of the mechanism that thalidomide play a therapeutic role in GIVM. Thalidomide 85-96 notch receptor 1 Homo sapiens 165-171 22883339-8 2012 CONCLUSION: Ang2, Notch1 and Dll4 may correlate with the pathogenesis of GIVM, while thalidomide can concentration-dependently down-regulate the expression of Ang2, Notch1 and Dll4, which may be one of the mechanism that thalidomide play a therapeutic role in GIVM. Thalidomide 85-96 delta like canonical Notch ligand 4 Homo sapiens 176-180 22883339-8 2012 CONCLUSION: Ang2, Notch1 and Dll4 may correlate with the pathogenesis of GIVM, while thalidomide can concentration-dependently down-regulate the expression of Ang2, Notch1 and Dll4, which may be one of the mechanism that thalidomide play a therapeutic role in GIVM. Thalidomide 221-232 angiopoietin 2 Homo sapiens 159-163 22883339-8 2012 CONCLUSION: Ang2, Notch1 and Dll4 may correlate with the pathogenesis of GIVM, while thalidomide can concentration-dependently down-regulate the expression of Ang2, Notch1 and Dll4, which may be one of the mechanism that thalidomide play a therapeutic role in GIVM. Thalidomide 221-232 notch receptor 1 Homo sapiens 165-171 22883339-8 2012 CONCLUSION: Ang2, Notch1 and Dll4 may correlate with the pathogenesis of GIVM, while thalidomide can concentration-dependently down-regulate the expression of Ang2, Notch1 and Dll4, which may be one of the mechanism that thalidomide play a therapeutic role in GIVM. Thalidomide 221-232 delta like canonical Notch ligand 4 Homo sapiens 176-180 22425187-0 2012 The thalidomide analgesic effect is associated with differential TNF-alpha receptor expression in the dorsal horn of the spinal cord as studied in a rat model of neuropathic pain. Thalidomide 4-15 tumor necrosis factor Rattus norvegicus 65-74 22826791-2 2012 The common expression of vascular endothelial growth factor (VEGF) and its receptor in HEH provide a rationale for the reported use of antiangiogenic drugs, including bevacizumab, lenalidomide and thalidomide. Thalidomide 197-208 vascular endothelial growth factor A Homo sapiens 25-59 22826791-2 2012 The common expression of vascular endothelial growth factor (VEGF) and its receptor in HEH provide a rationale for the reported use of antiangiogenic drugs, including bevacizumab, lenalidomide and thalidomide. Thalidomide 197-208 vascular endothelial growth factor A Homo sapiens 61-65 22277195-3 2012 Our study was aimed to evaluate the therapeutic potential of a novel analog of thalidomide, 3,6"-dithiothalidomide (DT), an agent with anti-TNF-alpha activity, in a model of chronic neuroinflammation. Thalidomide 79-90 tumor necrosis factor Homo sapiens 140-149 22179948-12 2012 Administration of thalidomide (25 and 50 mg/kg, i.p), a TNF-alpha inhibitor, significantly prevented hyperglycemia-induced thermal hyperalgesia and mechanical allodynia and also attenuated the increase in serum and urinary nitrite concentration, as compared with untreated diabetic rats. Thalidomide 18-29 tumor necrosis factor Rattus norvegicus 56-65 22179948-14 2012 CONCLUSION: It may be concluded that thalidomide has a beneficial effect in neuropathic pain by decreasing cytokines (TNF-alpha) and nitric oxide level and may provide a novel promising therapeutic approach for managing painful diabetic neuropathy. Thalidomide 37-48 tumor necrosis factor Rattus norvegicus 118-127 22741020-7 2012 Treatment with thalidomide resulted in apoptosis of mouse breast cancer cells in a time- and dose-dependent manner as demonstrated by caspase-3 enzyme activity. Thalidomide 15-26 caspase 3 Mus musculus 134-143 21757252-0 2012 The predictive role of serum VEGF in an advanced malignant mesothelioma patient cohort treated with thalidomide alone or combined with cisplatin/gemcitabine. Thalidomide 100-111 vascular endothelial growth factor A Homo sapiens 29-33 22348778-10 2012 This suggests that thalidomide downregulates fibroblast growth factor 8 expression and induces limb malformation by binding to wild-type cereblon, inhibiting the function of the associated E3 ubiquitin ligase. Thalidomide 19-30 fibroblast growth factor 8 Homo sapiens 45-71 22348778-10 2012 This suggests that thalidomide downregulates fibroblast growth factor 8 expression and induces limb malformation by binding to wild-type cereblon, inhibiting the function of the associated E3 ubiquitin ligase. Thalidomide 19-30 Cbl proto-oncogene like 2 Homo sapiens 189-208 21638302-0 2012 Metabolism of thalidomide by human liver microsome cytochrome CYP2C19 is required for its antimyeloma and antiangiogenic activities in vitro. Thalidomide 14-25 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 62-69 21638302-7 2012 These effects were partially reversed by omeprazole (10 micromol/l), a potent inhibitor of CYP2C19, suggesting that CYP2C19 is required for thalidomide to exhibit its antimyeloma and antiangiogenic activities. Thalidomide 140-151 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 91-98 21638302-7 2012 These effects were partially reversed by omeprazole (10 micromol/l), a potent inhibitor of CYP2C19, suggesting that CYP2C19 is required for thalidomide to exhibit its antimyeloma and antiangiogenic activities. Thalidomide 140-151 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 116-123 22346253-15 2012 CONCLUSION: Thalidomide was found to be safe for advanced HCC patients, demonstrating anti-tumor activity including response, survival, and AFP decreases of greater than 50% from baseline. Thalidomide 12-23 alpha fetoprotein Homo sapiens 140-143 21883476-4 2012 In thalidomide- and bortezomid-based therapy, candidate genes include TNFA and genes involved in the nuclear factor kappa B pathway (NFKB2 and TRAF3), respectively. Thalidomide 3-14 tumor necrosis factor Homo sapiens 70-74 21883476-4 2012 In thalidomide- and bortezomid-based therapy, candidate genes include TNFA and genes involved in the nuclear factor kappa B pathway (NFKB2 and TRAF3), respectively. Thalidomide 3-14 nuclear factor kappa B subunit 2 Homo sapiens 133-138 21883476-4 2012 In thalidomide- and bortezomid-based therapy, candidate genes include TNFA and genes involved in the nuclear factor kappa B pathway (NFKB2 and TRAF3), respectively. Thalidomide 3-14 TNF receptor associated factor 3 Homo sapiens 143-148 22239529-5 2012 Results revealed that while thalidomide reduced IL-8 and nuclear factor kappa B (NF-kappaB) activity by 95% and 46% in Huh-7 cells, increasing concentrations of thalidomide correlated with a linear rise in HCV replication (17-fold at 200 mum). Thalidomide 28-39 C-X-C motif chemokine ligand 8 Homo sapiens 48-52 22239529-5 2012 Results revealed that while thalidomide reduced IL-8 and nuclear factor kappa B (NF-kappaB) activity by 95% and 46% in Huh-7 cells, increasing concentrations of thalidomide correlated with a linear rise in HCV replication (17-fold at 200 mum). Thalidomide 28-39 nuclear factor kappa B subunit 1 Homo sapiens 57-79 22239529-5 2012 Results revealed that while thalidomide reduced IL-8 and nuclear factor kappa B (NF-kappaB) activity by 95% and 46% in Huh-7 cells, increasing concentrations of thalidomide correlated with a linear rise in HCV replication (17-fold at 200 mum). Thalidomide 28-39 nuclear factor kappa B subunit 1 Homo sapiens 81-90 22239529-6 2012 The NF-kappaB inhibitors, wedelolactone and NF-kappaB activation inhibitor-1, which mimic the actions of thalidomide by preventing phosphorylation and activation of IkappaB kinase (IKK) and hence block NF-kappaB activity, increased HCV RNA by 18- and 19-fold, respectively. Thalidomide 105-116 nuclear factor kappa B subunit 1 Homo sapiens 4-13 22239529-6 2012 The NF-kappaB inhibitors, wedelolactone and NF-kappaB activation inhibitor-1, which mimic the actions of thalidomide by preventing phosphorylation and activation of IkappaB kinase (IKK) and hence block NF-kappaB activity, increased HCV RNA by 18- and 19-fold, respectively. Thalidomide 105-116 nuclear factor kappa B subunit 1 Homo sapiens 44-53 22239529-6 2012 The NF-kappaB inhibitors, wedelolactone and NF-kappaB activation inhibitor-1, which mimic the actions of thalidomide by preventing phosphorylation and activation of IkappaB kinase (IKK) and hence block NF-kappaB activity, increased HCV RNA by 18- and 19-fold, respectively. Thalidomide 105-116 nuclear factor kappa B subunit 1 Homo sapiens 44-53 22463096-8 2012 Thalidomide therapy (100 mg/day) provided reduction in the median D-dimer levels to 6.07 (4.71-10.21) mug/ml and increase in median fibrinogen concentration to 1.9 g/L; soluble fibrin monomers were unidentifiable. Thalidomide 0-11 fibrinogen beta chain Homo sapiens 132-142 22382316-0 2012 Influence of cytochrome P450 2C19 gene variations on pharmacokinetic parameters of thalidomide in Japanese patients. Thalidomide 83-94 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 13-33 21685944-6 2012 While the anti-angiogenic drug thalidomide inhibits HIF-1-dependent VEGF transcription in breast carcinoma cells, it does not prevent HIF-1alpha stabilization, but leads to aberrant localization of HIF-1alpha to the perinuclear compartments and surprisingly stimulates nuclear export of beta-arrestin1. Thalidomide 31-42 hypoxia inducible factor 1 subunit alpha Homo sapiens 52-57 21685944-6 2012 While the anti-angiogenic drug thalidomide inhibits HIF-1-dependent VEGF transcription in breast carcinoma cells, it does not prevent HIF-1alpha stabilization, but leads to aberrant localization of HIF-1alpha to the perinuclear compartments and surprisingly stimulates nuclear export of beta-arrestin1. Thalidomide 31-42 vascular endothelial growth factor A Homo sapiens 68-72 21685944-6 2012 While the anti-angiogenic drug thalidomide inhibits HIF-1-dependent VEGF transcription in breast carcinoma cells, it does not prevent HIF-1alpha stabilization, but leads to aberrant localization of HIF-1alpha to the perinuclear compartments and surprisingly stimulates nuclear export of beta-arrestin1. Thalidomide 31-42 hypoxia inducible factor 1 subunit alpha Homo sapiens 198-208 21685944-6 2012 While the anti-angiogenic drug thalidomide inhibits HIF-1-dependent VEGF transcription in breast carcinoma cells, it does not prevent HIF-1alpha stabilization, but leads to aberrant localization of HIF-1alpha to the perinuclear compartments and surprisingly stimulates nuclear export of beta-arrestin1. Thalidomide 31-42 arrestin beta 1 Homo sapiens 287-301 22382316-3 2012 MATERIALS AND METHODS: Variations in the CYP2C19 gene in 6 patients treated with thalidomide were analyzed. Thalidomide 81-92 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 41-48 22382316-6 2012 RESULTS: The frequencies of CYP2C19 PM and hetero EM (hetEM) genotypes in Japanese patients taking thalidomide were 2 (33.3%) and 4 (66.7%), respectively. Thalidomide 99-110 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 28-35 22382316-8 2012 CONCLUSIONS: This study provided new insights regarding the contribution of CYP2C19 gene variations to adverse responses to thalidomide. Thalidomide 124-135 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 76-83 22382316-9 2012 Genotyping of CYP2C19*2 and *3 can be considerably simplified by using KOD FX as a polymerase for prediction of adverse effects to thalidomide by the PCR-RFLP method. Thalidomide 131-142 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 14-21 22382316-10 2012 CYP2C19 PM patients tend to have high serum thalidomide concentrations. Thalidomide 44-55 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 0-7 21681114-14 2012 CONCLUSIONS: Thalidomide is a potentially effective rescue therapy for severe refractory CD in children who fail to respond to anti-TNF medications. Thalidomide 13-24 tumor necrosis factor Homo sapiens 132-135 21733935-4 2012 In this in vitro study, MM by itself alters the profibrinolytic potential of ECs decreasing the tissue plasminogen activator (t-PA) and increasing the plasminogen activator inhibitor 1 (PAI-1) levels which is potentiated by thalidomide. Thalidomide 224-235 serpin family E member 1 Homo sapiens 151-184 21733935-6 2012 Additionally, DF upregulated the t-PA and downregulated PAI-1 gene expression modulated by thalidomide. Thalidomide 91-102 serpin family E member 1 Homo sapiens 56-61 22650376-7 2012 In particular, thalidomide was reevaluated and attracted significant attention due to its selective inhibitory activity of tumor necrosis factor-alpha (TNF-alpha), which is a clinically important activity against serious diseases such as rheumatoid arthritis, Crohn"s disease, leprosy, AIDS, and various cancers. Thalidomide 15-26 tumor necrosis factor Homo sapiens 123-150 22650376-7 2012 In particular, thalidomide was reevaluated and attracted significant attention due to its selective inhibitory activity of tumor necrosis factor-alpha (TNF-alpha), which is a clinically important activity against serious diseases such as rheumatoid arthritis, Crohn"s disease, leprosy, AIDS, and various cancers. Thalidomide 15-26 tumor necrosis factor Homo sapiens 152-161 22650377-0 2012 TNF alpha signaling beholds thalidomide saga: a review of mechanistic role of TNF-alpha signaling under thalidomide. Thalidomide 28-39 tumor necrosis factor Homo sapiens 0-9 22650377-0 2012 TNF alpha signaling beholds thalidomide saga: a review of mechanistic role of TNF-alpha signaling under thalidomide. Thalidomide 28-39 tumor necrosis factor Homo sapiens 78-87 22650377-0 2012 TNF alpha signaling beholds thalidomide saga: a review of mechanistic role of TNF-alpha signaling under thalidomide. Thalidomide 104-115 tumor necrosis factor Homo sapiens 0-9 22650377-0 2012 TNF alpha signaling beholds thalidomide saga: a review of mechanistic role of TNF-alpha signaling under thalidomide. Thalidomide 104-115 tumor necrosis factor Homo sapiens 78-87 22650377-4 2012 Thalidomide, an inhibitor of TNF-alpha protein synthesis is readily capable of crossing the blood-brain barrier and thus thalidomide and its analogs are excellent candidates for use in determining the potential value of anti-TNF-alpha therapies in a variety of diseases. Thalidomide 0-11 tumor necrosis factor Homo sapiens 29-38 22650377-4 2012 Thalidomide, an inhibitor of TNF-alpha protein synthesis is readily capable of crossing the blood-brain barrier and thus thalidomide and its analogs are excellent candidates for use in determining the potential value of anti-TNF-alpha therapies in a variety of diseases. Thalidomide 0-11 tumor necrosis factor Homo sapiens 225-234 22650377-5 2012 Thalidomide blocks TNF-alpha expression by different possible mechanisms. Thalidomide 0-11 tumor necrosis factor Homo sapiens 19-28 22650377-6 2012 Down regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), an essential transcription factor for TNF and other cytokines under thalidomide treatment leads to reduction in the TNF-alpha expression. Thalidomide 159-170 tumor necrosis factor Homo sapiens 129-132 22650377-6 2012 Down regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), an essential transcription factor for TNF and other cytokines under thalidomide treatment leads to reduction in the TNF-alpha expression. Thalidomide 159-170 tumor necrosis factor Homo sapiens 207-216 22650377-7 2012 Additionally, myeloid differentiation factor 88 (MyD88), an adapter protein regulates the expression of TNF under thalidomide treatment. Thalidomide 114-125 MYD88 innate immune signal transduction adaptor Homo sapiens 14-47 22650377-7 2012 Additionally, myeloid differentiation factor 88 (MyD88), an adapter protein regulates the expression of TNF under thalidomide treatment. Thalidomide 114-125 MYD88 innate immune signal transduction adaptor Homo sapiens 49-54 22650377-7 2012 Additionally, myeloid differentiation factor 88 (MyD88), an adapter protein regulates the expression of TNF under thalidomide treatment. Thalidomide 114-125 tumor necrosis factor Homo sapiens 104-107 22650377-8 2012 Thalidomide treatment also leads to destruction of TNF-alpha mRNA thus, reducing the total expression of TNF-alpha protein. Thalidomide 0-11 tumor necrosis factor Homo sapiens 51-60 22650377-8 2012 Thalidomide treatment also leads to destruction of TNF-alpha mRNA thus, reducing the total expression of TNF-alpha protein. Thalidomide 0-11 tumor necrosis factor Homo sapiens 105-114 22650377-9 2012 Thalidomide also targets reactive oxygen species (ROS) and alpha(1)-acid glycoprotein (AGP) to regulate TNF-alpha. Thalidomide 0-11 tumor necrosis factor Homo sapiens 104-113 22650377-10 2012 In the present review, we discuss different possible mechanism that regulates TNF-alpha under thalidomide treatment. Thalidomide 94-105 tumor necrosis factor Homo sapiens 78-87 22650377-11 2012 Additionally, we suggest novel strategies for the future targeting combination therapies of thalidomide and its analogs with different other anti-inflammatory drug to curb TNF-alpha associated diseases. Thalidomide 92-103 tumor necrosis factor Homo sapiens 172-181 22949932-13 2012 This coincides with the clinical evidence of a response to thalidomide in patients with osteolytic LCH, given its reported ability to reduce TGF-beta 1 and FoxP3 cells. Thalidomide 59-70 transforming growth factor beta 1 Homo sapiens 141-151 22949932-13 2012 This coincides with the clinical evidence of a response to thalidomide in patients with osteolytic LCH, given its reported ability to reduce TGF-beta 1 and FoxP3 cells. Thalidomide 59-70 forkhead box P3 Homo sapiens 156-161 21876693-4 2012 Research on angiogenesis in general, and vascular endothelial growth factor in particular, is a major focus in biomedicine and has led to the clinical approval of several antiangiogenic agents including thalidomide, bevacizumab, sorafenib, sunitinib, pazopanib, temesirolimus, and everolimus. Thalidomide 203-214 vascular endothelial growth factor A Homo sapiens 41-75 22571700-2 2012 For a long period of time, the standard therapy for MDS was hematopoietic stem cell transplantation, however DNA methyltransferase inhibitors (DNMT inhibitors) including decitabine (DAC) and azacitidine (AZA), and lenalidomide, a derivative of thalidomide have been highlighted as new chemotherapeutic agents for MDS. Thalidomide 244-255 DNA methyltransferase 1 Homo sapiens 143-147 21860026-2 2011 We investigated the role of cereblon (CRBN), a primary teratogenic target of thalidomide, in the antimyeloma activity of IMiDs. Thalidomide 77-88 cereblon Homo sapiens 28-36 20800319-4 2011 Because of severe side effects or high costs, other agents, such as thalidomide or high-dose intravenous immunoglobulins, are reserved for severe recalcitrant CLE. Thalidomide 68-79 RNA transcription, translation and transport factor Homo sapiens 159-162 22333255-8 2011 However, in the thalidomide group, PFS and OS were extended only in MM of immunoglobulin G (IgG) and immunoglobulin A (IgA) but not in light-chain patients. Thalidomide 16-27 CD79a molecule Homo sapiens 101-117 22333255-8 2011 However, in the thalidomide group, PFS and OS were extended only in MM of immunoglobulin G (IgG) and immunoglobulin A (IgA) but not in light-chain patients. Thalidomide 16-27 CD79a molecule Homo sapiens 119-122 21465313-2 2011 Thalidomide is an anti-angiogenic agent with anti-TNF-alpha and anti-NF-kappaB activity. Thalidomide 0-11 tumor necrosis factor Homo sapiens 50-59 21465313-2 2011 Thalidomide is an anti-angiogenic agent with anti-TNF-alpha and anti-NF-kappaB activity. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 69-78 21465313-12 2011 Thalidomide treatment was associated with significant down-regulation of nuclear NF-kappaB expression levels in residual neoplastic cells and microenvironments of responsive tumors, but not in t(11;18)(q21;q21)-positive, thalidomide-refractory tumors. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 81-90 21860026-2 2011 We investigated the role of cereblon (CRBN), a primary teratogenic target of thalidomide, in the antimyeloma activity of IMiDs. Thalidomide 77-88 cereblon Homo sapiens 38-42 22011961-5 2011 The aim of this study was to evaluate the ability of pentoxifylline, forskolin, rolipram, and thalidomide to decrease in vitro production of TNF-alpha and IFN-gamma in cells of HTLV-1-infected subjects. Thalidomide 94-105 tumor necrosis factor Homo sapiens 141-150 22011961-5 2011 The aim of this study was to evaluate the ability of pentoxifylline, forskolin, rolipram, and thalidomide to decrease in vitro production of TNF-alpha and IFN-gamma in cells of HTLV-1-infected subjects. Thalidomide 94-105 interferon gamma Homo sapiens 155-164 22011961-11 2011 The minimum dose of thalidomide used (1 microM) inhibited TNF-alpha production but thalidomide did not inhibit IFN-gamma production even when the maximum dose (50 microM) was used. Thalidomide 20-31 tumor necrosis factor Homo sapiens 58-67 22011961-12 2011 All drugs had an in vitro inhibitory effect on TNF-alpha production and, with the exception of thalidomide, all of them also decreased IFN-gamma production. Thalidomide 95-106 interferon gamma Homo sapiens 135-144 21955427-10 2011 The reduction degree of Ang-2 after a 4-month treatment of thalidomide may offer values for evaluating its prognosis and efficacy. Thalidomide 59-70 angiopoietin 2 Homo sapiens 24-29 21784047-8 2011 Plasma levels of vascular endothelial growth factor were measured in the group given thalidomide. Thalidomide 85-96 vascular endothelial growth factor A Homo sapiens 17-51 21784047-12 2011 Levels of vascular endothelial growth factor were significantly reduced by thalidomide (P < .001). Thalidomide 75-86 vascular endothelial growth factor A Homo sapiens 10-44 21784047-14 2011 Mechanisms of thalidomide activity might involve vascular endothelial growth factor. Thalidomide 14-25 vascular endothelial growth factor A Homo sapiens 49-83 21347685-0 2011 Single nucleotide polymorphisms in the promoter region of the IL1B gene influence outcome in multiple myeloma patients treated with high-dose chemotherapy independently of relapse treatment with thalidomide and bortezomib. Thalidomide 195-206 interleukin 1 beta Homo sapiens 62-66 21702058-4 2011 A morphometric analysis showed that systemic administration of thalidomide protects neural cells in the ganglion cell layer (GCL) in a dose-dependent manner and significantly decreases the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells in GCL and in the inner nuclear layer (INL). Thalidomide 63-74 germ cell-less 1, spermatogenesis associated Rattus norvegicus 295-298 21702058-5 2011 ELISA showed that thalidomide significantly suppressed the elevation of TNF-alpha 6 and 24 hr after an NMDA injection. Thalidomide 18-29 tumor necrosis factor Rattus norvegicus 72-81 21702058-1 2011 Thalidomide, an inhibitor of tumor necrosis factor-alpha (TNF-alpha) production, has been indicated to be useful for many inflammatory and oncogenic diseases. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 29-56 21702058-1 2011 Thalidomide, an inhibitor of tumor necrosis factor-alpha (TNF-alpha) production, has been indicated to be useful for many inflammatory and oncogenic diseases. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 58-67 21702058-8 2011 Thalidomide suppressed the increased expressions of NF-kappaB p65, p-JNK, and p-p38 after NMDA injection. Thalidomide 0-11 synaptotagmin 1 Rattus norvegicus 62-65 21702058-4 2011 A morphometric analysis showed that systemic administration of thalidomide protects neural cells in the ganglion cell layer (GCL) in a dose-dependent manner and significantly decreases the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells in GCL and in the inner nuclear layer (INL). Thalidomide 63-74 germ cell-less 1, spermatogenesis associated Rattus norvegicus 125-128 21702058-9 2011 Immunohistochemical analysis showed that thalidomide attenuated NF-kappaB p65 immunoreactivity in the GCL induced by NMDA treatment. Thalidomide 41-52 synaptotagmin 1 Rattus norvegicus 74-77 21702058-9 2011 Immunohistochemical analysis showed that thalidomide attenuated NF-kappaB p65 immunoreactivity in the GCL induced by NMDA treatment. Thalidomide 41-52 germ cell-less 1, spermatogenesis associated Rattus norvegicus 102-105 21670663-12 2011 When Thal is given in combination with chemotherapies or Dex, endothelial cell and monocyte PCA may be induced through phosphatidylserine exposure, or TF expression. Thalidomide 5-9 coagulation factor III, tissue factor Homo sapiens 151-153 21890654-7 2011 Of note, in our study, combination therapy of bortezomib and thalidomide successfully improved the condition of the patient with polyneuropathy, organomegaly, endocrinopathy, M-protein and skin changes syndrome associated with multicentric Castleman"s disease, suggesting that the combination therapy may be an effective therapeutic strategy for the intractable polyneuropathy, organomegaly, endocrinopathy, M-protein and skin changes syndrome associated with multicentric Castleman"s disease. Thalidomide 61-72 myomesin 2 Homo sapiens 175-184 21890654-7 2011 Of note, in our study, combination therapy of bortezomib and thalidomide successfully improved the condition of the patient with polyneuropathy, organomegaly, endocrinopathy, M-protein and skin changes syndrome associated with multicentric Castleman"s disease, suggesting that the combination therapy may be an effective therapeutic strategy for the intractable polyneuropathy, organomegaly, endocrinopathy, M-protein and skin changes syndrome associated with multicentric Castleman"s disease. Thalidomide 61-72 myomesin 2 Homo sapiens 408-417 21400110-5 2011 RESULTS: alpha,beta-Amyrin and thalidomide significantly attenuated the cerulein-induced increase in tumor necrosis factor (TNF)-alpha, interleukin-6, lipase, amylase, MPO, and TBARS. Thalidomide 31-42 tumor necrosis factor Mus musculus 101-134 21941940-3 2011 Combination therapy showed additive effect of thalidomide which restored lipid peroxidation as well as reduced myeloperoxidase and TNF-a towards the normal levels. Thalidomide 46-57 myeloperoxidase Rattus norvegicus 111-126 21941940-3 2011 Combination therapy showed additive effect of thalidomide which restored lipid peroxidation as well as reduced myeloperoxidase and TNF-a towards the normal levels. Thalidomide 46-57 tumor necrosis factor Rattus norvegicus 131-136 21435719-4 2011 Finally, polymorphism in GSTT1 (rs4630) was associated with a lower frequency of thalidomide-induced peripheral neuropathy (p=0.04). Thalidomide 81-92 glutathione S-transferase theta 1 Homo sapiens 25-30 21724449-0 2011 Thalidomide induces apoptosis in human oral squamous cell carcinoma cell line with altered expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Thalidomide 0-11 TNF superfamily member 10 Homo sapiens 105-160 21724449-0 2011 Thalidomide induces apoptosis in human oral squamous cell carcinoma cell line with altered expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Thalidomide 0-11 TNF superfamily member 10 Homo sapiens 162-167 21913886-8 2011 The repression of the tumor necrosis factor-a (TNF-alpha) expression is the crucial factor of many of the anti-inflammatory properties of thalidomide. Thalidomide 138-149 tumor necrosis factor Homo sapiens 22-45 21913886-8 2011 The repression of the tumor necrosis factor-a (TNF-alpha) expression is the crucial factor of many of the anti-inflammatory properties of thalidomide. Thalidomide 138-149 tumor necrosis factor Homo sapiens 47-56 21511336-0 2011 Effects of thalidomide on long-term bone marrow cultures from patients with myelodysplastic syndromes: induction of IL-10 expression in the stromal layers. Thalidomide 11-22 interleukin 10 Homo sapiens 116-121 21511336-2 2011 We demonstrated that thalidomide induced an increase in granulocyte-macrophage colony forming unit numbers and in IL-10 expression. Thalidomide 21-32 interleukin 10 Homo sapiens 114-119 21511336-3 2011 Thalidomide also promoted a slight increase in IL-6, IL-1beta and TNF-alpha expression in the stromal layers. Thalidomide 0-11 interleukin 6 Homo sapiens 47-51 21511336-3 2011 Thalidomide also promoted a slight increase in IL-6, IL-1beta and TNF-alpha expression in the stromal layers. Thalidomide 0-11 interleukin 1 beta Homo sapiens 53-61 21511336-3 2011 Thalidomide also promoted a slight increase in IL-6, IL-1beta and TNF-alpha expression in the stromal layers. Thalidomide 0-11 tumor necrosis factor Homo sapiens 66-75 21511336-5 2011 Our results indicate a participation of thalidomide upon the hematopoietic microenvironment of patients with myelodysplastic syndromes, especially in the up regulation of IL-10. Thalidomide 40-51 interleukin 10 Homo sapiens 171-176 21400110-5 2011 RESULTS: alpha,beta-Amyrin and thalidomide significantly attenuated the cerulein-induced increase in tumor necrosis factor (TNF)-alpha, interleukin-6, lipase, amylase, MPO, and TBARS. Thalidomide 31-42 interleukin 6 Mus musculus 136-149 21400110-5 2011 RESULTS: alpha,beta-Amyrin and thalidomide significantly attenuated the cerulein-induced increase in tumor necrosis factor (TNF)-alpha, interleukin-6, lipase, amylase, MPO, and TBARS. Thalidomide 31-42 lipase, endothelial Mus musculus 151-157 21400110-5 2011 RESULTS: alpha,beta-Amyrin and thalidomide significantly attenuated the cerulein-induced increase in tumor necrosis factor (TNF)-alpha, interleukin-6, lipase, amylase, MPO, and TBARS. Thalidomide 31-42 myeloperoxidase Mus musculus 168-171 21693319-9 2011 Moreover, thalidomide significantly inhibited the production of VEGF and ICAM-1 in serum (P < .05). Thalidomide 10-21 vascular endothelial growth factor A Rattus norvegicus 64-68 21046104-2 2011 Here, we analyse the association between genetic variation in the genes RAI and CD3EAP and outcome of 348 myeloma patients treated with high-dose treatment (HDT), 146 patients treated with interferon-alpha (INF-alpha) as maintenance treatment, 177 patients treated with thalidomide, and 74 patients treated with bortezomib at relapse and address if the effects of polymorphisms in CD3EAP and RAI are modified by a functional polymorphism in NFKB1. Thalidomide 270-281 growth differentiation factor 1 Homo sapiens 72-75 21693319-9 2011 Moreover, thalidomide significantly inhibited the production of VEGF and ICAM-1 in serum (P < .05). Thalidomide 10-21 intercellular adhesion molecule 1 Rattus norvegicus 73-79 21514830-0 2011 Peroxisome proliferator-activated receptor agonists with phenethylphenylphthalimide skeleton derived from thalidomide-related liver X receptor antagonists: relationship between absolute configuration and subtype selectivity. Thalidomide 106-117 peroxisome proliferator activated receptor alpha Homo sapiens 0-42 21478015-0 2011 Non-competitive and selective dipeptidyl peptidase IV inhibitors with phenethylphenylphthalimide skeleton derived from thalidomide-related alpha-glucosidase inhibitors and liver X receptor antagonists. Thalidomide 119-130 sucrase-isomaltase Homo sapiens 139-156 21478015-1 2011 Novel dipeptidyl peptidase IV (DPP-IV) inhibitors with a phenethylphenylphthalimide skeleton were prepared based on alpha-glucosidase inhibitors and liver X receptor (LXR) antagonists derived from thalidomide. Thalidomide 197-208 dipeptidyl peptidase 4 Homo sapiens 6-29 21478015-1 2011 Novel dipeptidyl peptidase IV (DPP-IV) inhibitors with a phenethylphenylphthalimide skeleton were prepared based on alpha-glucosidase inhibitors and liver X receptor (LXR) antagonists derived from thalidomide. Thalidomide 197-208 dipeptidyl peptidase 4 Homo sapiens 31-37 21478015-0 2011 Non-competitive and selective dipeptidyl peptidase IV inhibitors with phenethylphenylphthalimide skeleton derived from thalidomide-related alpha-glucosidase inhibitors and liver X receptor antagonists. Thalidomide 119-130 dipeptidyl peptidase 4 Homo sapiens 30-53 22977536-0 2011 Thalidomide and irradiation combination therapy increases substance P levels in vitro. Thalidomide 0-11 tachykinin 1 Mus musculus 58-69 21207098-8 2011 We identified a thalidomide-binding protein, cereblon (CRBN), as a primary target for thalidomide teratogenicity. Thalidomide 16-27 cereblon Homo sapiens 45-53 21207098-8 2011 We identified a thalidomide-binding protein, cereblon (CRBN), as a primary target for thalidomide teratogenicity. Thalidomide 16-27 cereblon Homo sapiens 55-59 21207098-8 2011 We identified a thalidomide-binding protein, cereblon (CRBN), as a primary target for thalidomide teratogenicity. Thalidomide 86-97 cereblon Homo sapiens 45-53 21207098-8 2011 We identified a thalidomide-binding protein, cereblon (CRBN), as a primary target for thalidomide teratogenicity. Thalidomide 86-97 cereblon Homo sapiens 55-59 21207098-9 2011 Our data suggest that thalidomide initiates its teratogenic effects by binding to CRBN and inhibiting its ubiquitin ligase activity. Thalidomide 22-33 cereblon Homo sapiens 82-86 21207214-8 2011 The patient was treated according to the CTD protocol (cyclophosphamide, thalidomide, and dexamethasone) which was effective against the myeloma as well as the systemic sclerosis and patient achieved complete remission. Thalidomide 73-84 CTD Homo sapiens 41-44 21539454-8 2011 The expression of vascular endothelial growth factor (VEGF) mRNA and protein was suppressed after treatment with thalidomide alone in a dose-dependent manner. Thalidomide 113-124 vascular endothelial growth factor A Homo sapiens 18-52 21539454-8 2011 The expression of vascular endothelial growth factor (VEGF) mRNA and protein was suppressed after treatment with thalidomide alone in a dose-dependent manner. Thalidomide 113-124 vascular endothelial growth factor A Homo sapiens 54-58 21539454-9 2011 Synergistic suppressive effects on VEGF expression were observed after administration of thalidomide and X-ray exposure. Thalidomide 89-100 vascular endothelial growth factor A Homo sapiens 35-39 21539454-10 2011 In conclusion, thalidomide was able to enhance the radiosensitivity of TE1 cells in vitro, which could be closely related to its suppressive effects on the expression of VEGF in TE1 cells, but had no obvious effects on the cell cycle. Thalidomide 15-26 vascular endothelial growth factor A Homo sapiens 170-174 21614166-9 2011 Thalidomide treatment significantly ameliorated submesothelial thickening and angiogenesis, and decreased numbers of PCNA- and VEGF-expressing cells, myofibroblasts, and TGF-beta-positive cells. Thalidomide 0-11 proliferating cell nuclear antigen Mus musculus 117-121 21614166-9 2011 Thalidomide treatment significantly ameliorated submesothelial thickening and angiogenesis, and decreased numbers of PCNA- and VEGF-expressing cells, myofibroblasts, and TGF-beta-positive cells. Thalidomide 0-11 vascular endothelial growth factor A Mus musculus 127-131 21614166-9 2011 Thalidomide treatment significantly ameliorated submesothelial thickening and angiogenesis, and decreased numbers of PCNA- and VEGF-expressing cells, myofibroblasts, and TGF-beta-positive cells. Thalidomide 0-11 transforming growth factor, beta 1 Mus musculus 170-178 21159364-6 2011 RESULTS: At THL and LEN concentrations resembling those observed in myeloma patients in vivo and in the presence of tumor necrosis factor-alpha (TNFalpha) we observed significantly increased TF activity in human umbilical vein endothelial cells in vitro. Thalidomide 12-15 coagulation factor III, tissue factor Homo sapiens 191-193 21159364-10 2011 CONCLUSIONS: Our in vitro data support the hypothesis that THL and LEN induce a hypercoagulable state through increased endothelial TF expression. Thalidomide 59-62 coagulation factor III, tissue factor Homo sapiens 132-134 21215621-0 2011 Design, synthesis and biological evaluation of new thalidomide analogues as TNF-alpha and IL-6 production inhibitors. Thalidomide 51-62 tumor necrosis factor Homo sapiens 76-85 21215621-0 2011 Design, synthesis and biological evaluation of new thalidomide analogues as TNF-alpha and IL-6 production inhibitors. Thalidomide 51-62 interleukin 6 Homo sapiens 90-94 21215621-1 2011 Several thalidomide analogues were synthesized and compared to thalidomide and its more active analogue, lenalidomide, for their ability to inhibit the production of the pro-inflammatory cytokine tumour necrosis factor (TNF)-alpha and interleukin (IL)-6 by LPS-activated peripheral blood mononuclear cells (PBMCs). Thalidomide 8-19 tumor necrosis factor Homo sapiens 196-230 21215621-1 2011 Several thalidomide analogues were synthesized and compared to thalidomide and its more active analogue, lenalidomide, for their ability to inhibit the production of the pro-inflammatory cytokine tumour necrosis factor (TNF)-alpha and interleukin (IL)-6 by LPS-activated peripheral blood mononuclear cells (PBMCs). Thalidomide 8-19 interleukin 6 Homo sapiens 235-253 21215621-1 2011 Several thalidomide analogues were synthesized and compared to thalidomide and its more active analogue, lenalidomide, for their ability to inhibit the production of the pro-inflammatory cytokine tumour necrosis factor (TNF)-alpha and interleukin (IL)-6 by LPS-activated peripheral blood mononuclear cells (PBMCs). Thalidomide 63-74 tumor necrosis factor Homo sapiens 196-230 21215621-1 2011 Several thalidomide analogues were synthesized and compared to thalidomide and its more active analogue, lenalidomide, for their ability to inhibit the production of the pro-inflammatory cytokine tumour necrosis factor (TNF)-alpha and interleukin (IL)-6 by LPS-activated peripheral blood mononuclear cells (PBMCs). Thalidomide 63-74 interleukin 6 Homo sapiens 235-253 20846163-4 2010 We reported recently that two substances (ATRA and thalidomide) have preventive effects on pulmonary fibrosis by inhibiting IL-6-dependent proliferation and TGF-beta1-dependent transdifferentiation of lung fibroblasts. Thalidomide 51-62 interleukin 6 Mus musculus 124-128 21276247-3 2011 In the present study, we examined the effects of prednisone, thalidomide, cyclosporine A, and amitriptyline, drugs used in a variety of clinical conditions, on the production of TNF-alpha, IL-10, and IL-12 by purified epidermal Langerhans cells and peritoneal macrophages in BALB/c mice. Thalidomide 61-72 tumor necrosis factor Mus musculus 178-187 22003441-7 2011 Intriguingly, a new role for CRBN has been identified, i.e, the binding of immunomodulatory drugs (IMiDs), e.g. thalidomide, to CRBN has now been associated with teratogenicity and also the cytotoxicity of IMiDs, including lenalidomide, which are widely used to treat multiple myeloma patients. Thalidomide 112-123 cereblon Homo sapiens 29-33 22003441-7 2011 Intriguingly, a new role for CRBN has been identified, i.e, the binding of immunomodulatory drugs (IMiDs), e.g. thalidomide, to CRBN has now been associated with teratogenicity and also the cytotoxicity of IMiDs, including lenalidomide, which are widely used to treat multiple myeloma patients. Thalidomide 112-123 cereblon Homo sapiens 128-132 21792375-0 2011 Thalidomide Analogues Suppress Lipopolysaccharide-Induced Synthesis of TNF-alpha and Nitrite, an Intermediate of Nitric Oxide, in a Cellular Model of Inflammation. Thalidomide 0-11 tumor necrosis factor Homo sapiens 71-80 21792375-8 2011 Utilizing a RAW 264.7 cellular model of lipopolysaccharide-induced inflammation that induces high levels of TNF-alpha protein accompanied by a robust generation of nitrite, the properties of a series of thalidomide-based TNF-alpha synthesis inhibitors were evaluated to reduce the levels of both. Thalidomide 203-214 tumor necrosis factor Homo sapiens 108-117 21792375-8 2011 Utilizing a RAW 264.7 cellular model of lipopolysaccharide-induced inflammation that induces high levels of TNF-alpha protein accompanied by a robust generation of nitrite, the properties of a series of thalidomide-based TNF-alpha synthesis inhibitors were evaluated to reduce the levels of both. Thalidomide 203-214 tumor necrosis factor Homo sapiens 221-230 21792375-9 2011 Specific analogues of thalidomide effectively suppressed the generation of both TNF-alpha and nitrite at well-tolerated doses. Thalidomide 22-33 tumor necrosis factor Homo sapiens 80-89 21477797-0 2011 Thalidomide inhibits interferon-gamma-mediated nitric oxide production in mouse vascular endothelial cells. Thalidomide 0-11 interferon gamma Mus musculus 21-37 21477797-2 2011 The effect of thalidomide on interferon (IFN)-gamma induced nitric oxide (NO) production in mouse vascular endothelial cells was examined in order to elucidate the anti-angiogenic or anti-inflammatory action. Thalidomide 14-25 interferon gamma Mus musculus 29-51 21477797-3 2011 Thalidomide inhibited IFN-gamma-induced NO production in mouse END-D cells via reduced expression of an inducible type of NO synthase (iNOS) protein and mRNA. Thalidomide 0-11 interferon gamma Mus musculus 22-31 21477797-3 2011 Thalidomide inhibited IFN-gamma-induced NO production in mouse END-D cells via reduced expression of an inducible type of NO synthase (iNOS) protein and mRNA. Thalidomide 0-11 nitric oxide synthase 2, inducible Mus musculus 135-139 21477797-4 2011 Since thalidomide did not alter the cell surface expression of IFN-gamma receptor, the NO inhibition was suggested to be due to the impairment of IFN-gamma-induced intracellular event by thalidomide. Thalidomide 187-198 interferon gamma Mus musculus 146-155 21477797-5 2011 Thalidomide inhibited the phosphorylation of IRF1, which was required for the iNOS expression. Thalidomide 0-11 interferon regulatory factor 1 Mus musculus 45-49 21477797-5 2011 Thalidomide inhibited the phosphorylation of IRF1, which was required for the iNOS expression. Thalidomide 0-11 nitric oxide synthase 2, inducible Mus musculus 78-82 21477797-9 2011 Therefore, thalidomide was suggested to inhibit IFN-gamma-induced NO production via impaired STAT1 phosphorylation. Thalidomide 11-22 interferon gamma Mus musculus 48-57 21477797-9 2011 Therefore, thalidomide was suggested to inhibit IFN-gamma-induced NO production via impaired STAT1 phosphorylation. Thalidomide 11-22 signal transducer and activator of transcription 1 Mus musculus 93-98 20958261-6 2011 We then summarize data showing how thalidomide interferes with this signaling network: thalidomide inhibits the activity of the redox-sensitive transcription factor NF-kappaB, shifts the balance of fibroblast growth factors and bone morphogenetic proteins (Bmps) towards pro-apoptotic Bmps, and suppresses Wnt/beta-catenin- and Akt-dependent survival signaling in the limb bud. Thalidomide 35-46 nuclear factor kappa B subunit 1 Homo sapiens 165-174 20958261-6 2011 We then summarize data showing how thalidomide interferes with this signaling network: thalidomide inhibits the activity of the redox-sensitive transcription factor NF-kappaB, shifts the balance of fibroblast growth factors and bone morphogenetic proteins (Bmps) towards pro-apoptotic Bmps, and suppresses Wnt/beta-catenin- and Akt-dependent survival signaling in the limb bud. Thalidomide 35-46 catenin beta 1 Homo sapiens 310-331 20958261-6 2011 We then summarize data showing how thalidomide interferes with this signaling network: thalidomide inhibits the activity of the redox-sensitive transcription factor NF-kappaB, shifts the balance of fibroblast growth factors and bone morphogenetic proteins (Bmps) towards pro-apoptotic Bmps, and suppresses Wnt/beta-catenin- and Akt-dependent survival signaling in the limb bud. Thalidomide 87-98 nuclear factor kappa B subunit 1 Homo sapiens 165-174 20958261-6 2011 We then summarize data showing how thalidomide interferes with this signaling network: thalidomide inhibits the activity of the redox-sensitive transcription factor NF-kappaB, shifts the balance of fibroblast growth factors and bone morphogenetic proteins (Bmps) towards pro-apoptotic Bmps, and suppresses Wnt/beta-catenin- and Akt-dependent survival signaling in the limb bud. Thalidomide 87-98 catenin beta 1 Homo sapiens 310-331 20846163-4 2010 We reported recently that two substances (ATRA and thalidomide) have preventive effects on pulmonary fibrosis by inhibiting IL-6-dependent proliferation and TGF-beta1-dependent transdifferentiation of lung fibroblasts. Thalidomide 51-62 transforming growth factor, beta 1 Mus musculus 157-166 20594187-6 2010 Several drugs effective for the treatment of MM, including proteasome inhibitors, thalidomide, lenalidomide and arsenic trioxide, block NF-kappaB activation. Thalidomide 82-93 nuclear factor kappa B subunit 1 Homo sapiens 136-145 21211317-1 2010 OBJECTIVE: To investigate the influences of VEGF expression through the intervention of thalidomide in malignant transformation of hepatocytes. Thalidomide 88-99 vascular endothelial growth factor A Rattus norvegicus 44-48 21211317-12 2010 And the VEGF level of thalidomide group was lower than those in 2-FAA group. Thalidomide 22-33 vascular endothelial growth factor A Rattus norvegicus 8-12 21211317-13 2010 CONCLUSION: Thalidomide can inhibit the hepatic VEGF expression and arrest the development of rat hepatoma. Thalidomide 12-23 vascular endothelial growth factor A Rattus norvegicus 48-52 20858096-0 2010 High expression levels of the mammalian target of rapamycin inhibitor DEPTOR are predictive of response to thalidomide in myeloma. Thalidomide 107-118 DEP domain containing MTOR interacting protein Homo sapiens 70-76 20122728-0 2010 Role of the TNF-alpha promoter polymorphisms for development of multiple myeloma and clinical outcome in thalidomide plus dexamethasone. Thalidomide 105-116 tumor necrosis factor Homo sapiens 12-21 20858096-0 2010 High expression levels of the mammalian target of rapamycin inhibitor DEPTOR are predictive of response to thalidomide in myeloma. Thalidomide 107-118 mechanistic target of rapamycin kinase Homo sapiens 30-59 20709454-7 2010 Furthermore, systemic pre-treatment with the classically used drugs indomethacin, celecoxib, guanetidine, morphine, thalidomide or dexamethasone, also prevented IL-6-induced muscle pain. Thalidomide 116-127 interleukin 6 Mus musculus 161-165 20837031-1 2010 AIMS: Thalidomide is thought to prevent TNF-alpha production, and such mechanism could be useful in a spinally delivered drug approach for the control of peripheral inflammation. Thalidomide 6-17 tumor necrosis factor Rattus norvegicus 40-49 20705061-0 2010 PI3K/Akt signaling pathway is required for neuroprotection of thalidomide on hypoxic-ischemic cortical neurons in vitro. Thalidomide 62-73 AKT serine/threonine kinase 1 Rattus norvegicus 5-8 20705061-1 2010 Thalidomide, a derivative of glutamic acid, is used for immunomodulatory therapy in various diseases through inhibition of tumor necrotic factor-alpha (TNF-alpha) release. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 152-161 20705061-9 2010 Furthermore, we found that thalidomide protected neurons against apoptosis by decreasing CC3 and increasing Bcl-2 expression in a dose-dependent manner. Thalidomide 27-38 BCL2, apoptosis regulator Rattus norvegicus 108-113 20705061-10 2010 Meanwhile, we found that thalidomide induced p-Akt expression, which could be inhibited by PI3K specific inhibitor, LY294002. Thalidomide 25-36 AKT serine/threonine kinase 1 Rattus norvegicus 47-50 20705061-12 2010 In summary, thalidomide has anti-apoptotic effects on cortical neurons after OGD by modulating CC3 and Bcl-2 expression through activation of PI3K/Akt pathway. Thalidomide 12-23 BCL2, apoptosis regulator Rattus norvegicus 103-108 20705061-12 2010 In summary, thalidomide has anti-apoptotic effects on cortical neurons after OGD by modulating CC3 and Bcl-2 expression through activation of PI3K/Akt pathway. Thalidomide 12-23 AKT serine/threonine kinase 1 Rattus norvegicus 147-150 20837031-12 2010 thalidomide reduced the expression of CD11b/c and GFAP markers in the lumbar spinal cord. Thalidomide 0-11 integrin subunit alpha M Rattus norvegicus 38-43 20837031-12 2010 thalidomide reduced the expression of CD11b/c and GFAP markers in the lumbar spinal cord. Thalidomide 0-11 glial fibrillary acidic protein Rattus norvegicus 50-54 20601373-9 2010 On histological analysis, thalidomide treatment lowered total myocardial collagen content and the expression of TNF-alpha, IL-1beta, ICAM-1, and VCAM-1. Thalidomide 26-37 intercellular adhesion molecule 1 Rattus norvegicus 133-139 20923560-0 2010 Intraperitoneal injection of thalidomide attenuates bone cancer pain and decreases spinal tumor necrosis factor-alpha expression in a mouse model. Thalidomide 29-40 tumor necrosis factor Mus musculus 90-117 20923560-2 2010 Thalidomide has been shown to selectively inhibit TNF-alpha production. Thalidomide 0-11 tumor necrosis factor Mus musculus 50-59 20923560-4 2010 Therefore, in the present study, we investigated the effect of thalidomide on bone cancer-induced hyperalgesia and up-regulated expression of spinal TNF-alpha in a mouse model. Thalidomide 63-74 tumor necrosis factor Mus musculus 149-158 20923560-7 2010 Intraperitoneal injection of thalidomide (50 mg/kg), started at day 1 after surgery and once daily thereafter until day 7, attenuated bone cancer-evoked mechanical allodynia and thermal hyperalgesia as well as the up-regulation of TNF-alpha in the spinal cord. Thalidomide 29-40 tumor necrosis factor Mus musculus 231-240 20601373-9 2010 On histological analysis, thalidomide treatment lowered total myocardial collagen content and the expression of TNF-alpha, IL-1beta, ICAM-1, and VCAM-1. Thalidomide 26-37 tumor necrosis factor Rattus norvegicus 112-121 20601373-9 2010 On histological analysis, thalidomide treatment lowered total myocardial collagen content and the expression of TNF-alpha, IL-1beta, ICAM-1, and VCAM-1. Thalidomide 26-37 vascular cell adhesion molecule 1 Rattus norvegicus 145-151 20601373-9 2010 On histological analysis, thalidomide treatment lowered total myocardial collagen content and the expression of TNF-alpha, IL-1beta, ICAM-1, and VCAM-1. Thalidomide 26-37 interleukin 1 beta Rattus norvegicus 123-131 21222370-10 2010 Thalidomide is a drug with pleiotropic effects: it appears to downregulate production of TNF-alpha and other proinflammatory cytokines. Thalidomide 0-11 tumor necrosis factor Homo sapiens 89-98 20857659-0 2010 [Thalidomide--coverage by health insurance when used for treatment of ENL]. Thalidomide 1-12 MLLT1 super elongation complex subunit Homo sapiens 70-73 20627385-0 2010 The efficacy and safety of the low-thalidomide dose CTD (cyclophosphamide, thalidomide, dexamethasone) regimen in patients with multiple myeloma--a report by the Polish Myeloma Study Group. Thalidomide 35-46 CTD Homo sapiens 52-55 20627385-0 2010 The efficacy and safety of the low-thalidomide dose CTD (cyclophosphamide, thalidomide, dexamethasone) regimen in patients with multiple myeloma--a report by the Polish Myeloma Study Group. Thalidomide 75-86 CTD Homo sapiens 52-55 20627385-8 2010 The outpatient low-thalidomide dose CTD regimen is well tolerated and produces a significant response rate both in untreated and relapsing/resistant MM patients. Thalidomide 19-30 CTD Homo sapiens 36-39 20538468-4 2010 We identified potential novel anti-influenza agents from a screen of 34 synthesized phenethylphenylphthalimide analogs derived from thalidomide (PPT analogs). Thalidomide 132-143 tachykinin precursor 1 Homo sapiens 145-148 20421453-0 2010 XBP1s levels are implicated in the biology and outcome of myeloma mediating different clinical outcomes to thalidomide-based treatments. Thalidomide 107-118 X-box binding protein 1 Homo sapiens 0-4 20421453-6 2010 Furthermore, we show the beneficial therapeutic effects of thalidomide in patients with low XBP1s/u ratios. Thalidomide 59-70 X-box binding protein 1 Homo sapiens 92-96 20346742-1 2010 We document an EGFR mutation in a patient with papillary renal cell cancer with a history of multiple therapies, including interferon-alpha, interleukin-2, 5-fluorouracil, and interferon-alpha together with 13-cis-retinoic acid, to which floxuridine was later added, and thalidomide maintenance therapy for six years. Thalidomide 271-282 epidermal growth factor receptor Homo sapiens 15-19 20346742-1 2010 We document an EGFR mutation in a patient with papillary renal cell cancer with a history of multiple therapies, including interferon-alpha, interleukin-2, 5-fluorouracil, and interferon-alpha together with 13-cis-retinoic acid, to which floxuridine was later added, and thalidomide maintenance therapy for six years. Thalidomide 271-282 interleukin 2 Homo sapiens 141-154 20399910-6 2010 Remarkably, the levels of pro-inflammatory cytokines (IL-1alpha and TNF-alpha) were found to be decreased significantly (p<0.05) in thalidomide treated group but the levels of IL-10 were found to be significantly (p<0.05) elevated. Thalidomide 135-146 interleukin 1 alpha Mus musculus 54-63 20421453-7 2010 This study highlights the importance of XBP1 in myeloma and its significance as an independent prognostic marker and as a predictor of thalidomide response. Thalidomide 135-146 X-box binding protein 1 Homo sapiens 40-44 20438868-5 2010 As shown in our results, treatment of solid tumor-bearing mice with thalidomide 1 resulted in a significant reduction in tumor volume with 75.4% inhibition, a significant decrease in lactate dehydrogenase (LDH), ICAM-1 expression and NO. Thalidomide 68-79 intercellular adhesion molecule 1 Mus musculus 212-218 20399910-6 2010 Remarkably, the levels of pro-inflammatory cytokines (IL-1alpha and TNF-alpha) were found to be decreased significantly (p<0.05) in thalidomide treated group but the levels of IL-10 were found to be significantly (p<0.05) elevated. Thalidomide 135-146 tumor necrosis factor Mus musculus 68-77 20399910-6 2010 Remarkably, the levels of pro-inflammatory cytokines (IL-1alpha and TNF-alpha) were found to be decreased significantly (p<0.05) in thalidomide treated group but the levels of IL-10 were found to be significantly (p<0.05) elevated. Thalidomide 135-146 interleukin 10 Mus musculus 179-184 20616958-8 2010 Thalidomide inhibits the activity of basic fibroblast growth factor-2 (bFGF). Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 37-69 22966360-0 2010 Effect of thalidomide and arsenic trioxide on the release of tumor necrosis factor-alpha and vascular endothelial growth factor from the KG-1a human acute myelogenous leukemia cell line. Thalidomide 10-21 tumor necrosis factor Homo sapiens 61-88 22966360-0 2010 Effect of thalidomide and arsenic trioxide on the release of tumor necrosis factor-alpha and vascular endothelial growth factor from the KG-1a human acute myelogenous leukemia cell line. Thalidomide 10-21 vascular endothelial growth factor A Homo sapiens 93-127 22966360-2 2010 The current investigation was conducted in order to evaluate the effect of thalidomide either alone or in combination with arsenic trioxide on the release of tumor necrosis factor-alpha (TNF-alpha) and vascular endothelial growth factor (VEGF) from this cell line in an attempt to clarify its possible cytotoxic mechanism(s). Thalidomide 75-86 tumor necrosis factor Homo sapiens 158-185 22966360-2 2010 The current investigation was conducted in order to evaluate the effect of thalidomide either alone or in combination with arsenic trioxide on the release of tumor necrosis factor-alpha (TNF-alpha) and vascular endothelial growth factor (VEGF) from this cell line in an attempt to clarify its possible cytotoxic mechanism(s). Thalidomide 75-86 tumor necrosis factor Homo sapiens 187-196 22966360-2 2010 The current investigation was conducted in order to evaluate the effect of thalidomide either alone or in combination with arsenic trioxide on the release of tumor necrosis factor-alpha (TNF-alpha) and vascular endothelial growth factor (VEGF) from this cell line in an attempt to clarify its possible cytotoxic mechanism(s). Thalidomide 75-86 vascular endothelial growth factor A Homo sapiens 202-236 22966360-2 2010 The current investigation was conducted in order to evaluate the effect of thalidomide either alone or in combination with arsenic trioxide on the release of tumor necrosis factor-alpha (TNF-alpha) and vascular endothelial growth factor (VEGF) from this cell line in an attempt to clarify its possible cytotoxic mechanism(s). Thalidomide 75-86 vascular endothelial growth factor A Homo sapiens 238-242 22966360-6 2010 Results obtained indicate that the levels of TNF-alpha in the supernatant of KG-1a cell cultures incubated with thalidomide, arsenic trioxide, or combination were statistically lower than those observed in the supernatant of control cells (2.89, 5.07, 4.15 and 16.88 pg/ml, respectively). Thalidomide 112-123 tumor necrosis factor Homo sapiens 45-54 22966360-7 2010 However, the levels of VEGF in the supernatant of thalidomide-treated cells were statistically higher than those in the supernatant of control cells (69.61 vs. 11.48 pg/l). Thalidomide 50-61 vascular endothelial growth factor A Homo sapiens 23-27 20616958-8 2010 Thalidomide inhibits the activity of basic fibroblast growth factor-2 (bFGF). Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 71-75 20620488-9 2010 Both thalidomide and pentoxyphylline effectively reduced AST, LDH, TNF-alpha, and lipid peroxidation levels, as well as attenuated tissue edema and intestinal injury induced by I/R (P < .05). Thalidomide 5-16 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 57-60 20307255-2 2010 Thalidomide and IMiDs inhibit the cytokines tumour necrosis factor-alpha (TNF-alpha), interleukins (IL) 1-beta, 6, 12, and granulocyte macrophage-colony stimulating factor (GM-CSF). Thalidomide 0-11 tumor necrosis factor Homo sapiens 74-83 20307255-2 2010 Thalidomide and IMiDs inhibit the cytokines tumour necrosis factor-alpha (TNF-alpha), interleukins (IL) 1-beta, 6, 12, and granulocyte macrophage-colony stimulating factor (GM-CSF). Thalidomide 0-11 interleukin 1 beta Homo sapiens 86-110 20307255-2 2010 Thalidomide and IMiDs inhibit the cytokines tumour necrosis factor-alpha (TNF-alpha), interleukins (IL) 1-beta, 6, 12, and granulocyte macrophage-colony stimulating factor (GM-CSF). Thalidomide 0-11 colony stimulating factor 2 Homo sapiens 123-171 20307255-2 2010 Thalidomide and IMiDs inhibit the cytokines tumour necrosis factor-alpha (TNF-alpha), interleukins (IL) 1-beta, 6, 12, and granulocyte macrophage-colony stimulating factor (GM-CSF). Thalidomide 0-11 colony stimulating factor 2 Homo sapiens 173-179 20307255-5 2010 Thalidomide analogues have been used as inhibitors of alpha glucosidase and could be potential drugs for diabetes treatment. Thalidomide 0-11 sucrase-isomaltase Homo sapiens 54-71 20620488-9 2010 Both thalidomide and pentoxyphylline effectively reduced AST, LDH, TNF-alpha, and lipid peroxidation levels, as well as attenuated tissue edema and intestinal injury induced by I/R (P < .05). Thalidomide 5-16 tumor necrosis factor Rattus norvegicus 67-76 20419594-5 2010 In addition, GlaxoSmithKline plc is developing 256066, an inhaled formulation of a PDE4 inhibitor that has demonstrated efficacy in trials in asthma, and apremilast from Celgene Corp has been reported to be effective for the treatment of psoriasis. Thalidomide 170-177 heparan sulfate proteoglycan 2 Homo sapiens 29-32 20382589-8 2010 In the thalidomide-treated group, the morphologic changes generated only punctiform denaturation and necrosis at the early or middle stages, and nodular hyperplasia or a little atypical hyperplasia at the final stages, with the expression of NF-kappaB (X2=9.93, P<0.001) and VEGF (X2=8.024, P<0.001) lower than that in the 2-FAA group. Thalidomide 7-18 vascular endothelial growth factor A Rattus norvegicus 278-282 19713194-3 2010 Thalidomide has been shown to be anti-angiogenic via reduction of VEGF levels. Thalidomide 0-11 vascular endothelial growth factor A Mus musculus 66-70 20015241-2 2010 Increased concentration of hepatocyte growth factor (HGF) is associated with poor prognosis in patients treated with conventional chemotherapy or thalidomide. Thalidomide 146-157 hepatocyte growth factor Homo sapiens 27-51 20015241-2 2010 Increased concentration of hepatocyte growth factor (HGF) is associated with poor prognosis in patients treated with conventional chemotherapy or thalidomide. Thalidomide 146-157 hepatocyte growth factor Homo sapiens 53-56 19815837-5 2010 RESULTS: Thalidomide strongly inhibited gelatinase production by B-cell lines and primary myeloma cells in response to fibronectin, the most efficient gelatinase inducer identified in lymphoid cells. Thalidomide 9-20 fibronectin 1 Homo sapiens 119-130 20514789-1 2010 Thalidomide is anti-inflammatory under some conditions, yet has been reported to up-regulate Th1 (T helper 1) immunity measured by increased IL-2 (Interleukin-2) and gamma interferon. Thalidomide 0-11 interleukin 2 Mus musculus 141-145 20514789-1 2010 Thalidomide is anti-inflammatory under some conditions, yet has been reported to up-regulate Th1 (T helper 1) immunity measured by increased IL-2 (Interleukin-2) and gamma interferon. Thalidomide 0-11 interleukin 2 Mus musculus 147-160 20514789-6 2010 Inhibition of inducible nitric oxide synthase (iNOS) may prove to be an important target for the anti-inflammatory and anti-cancer effects of thalidomide and related immunomodulatory drugs (IMiDs). Thalidomide 142-153 nitric oxide synthase 2, inducible Mus musculus 14-45 20514789-6 2010 Inhibition of inducible nitric oxide synthase (iNOS) may prove to be an important target for the anti-inflammatory and anti-cancer effects of thalidomide and related immunomodulatory drugs (IMiDs). Thalidomide 142-153 nitric oxide synthase 2, inducible Mus musculus 47-51 20364125-5 2010 In mice heterozygous for a null mutation in the Eng gene (encoding endoglin), an experimental model of HHT, thalidomide treatment stimulated mural cell coverage and thus rescued vessel wall defects. Thalidomide 108-119 endoglin Mus musculus 48-51 20364125-5 2010 In mice heterozygous for a null mutation in the Eng gene (encoding endoglin), an experimental model of HHT, thalidomide treatment stimulated mural cell coverage and thus rescued vessel wall defects. Thalidomide 108-119 endoglin Mus musculus 67-75 20364125-6 2010 Thalidomide treatment increased platelet-derived growth factor-B (PDGF-B) expression in endothelial cells and stimulated mural cell activation. Thalidomide 0-11 platelet derived growth factor subunit B Homo sapiens 32-64 20364125-6 2010 Thalidomide treatment increased platelet-derived growth factor-B (PDGF-B) expression in endothelial cells and stimulated mural cell activation. Thalidomide 0-11 platelet derived growth factor subunit B Homo sapiens 66-72 20223979-5 2010 Thalidomide initiates its teratogenic effects by binding to CRBN and inhibiting the associated ubiquitin ligase activity. Thalidomide 0-11 cereblon Homo sapiens 60-64 19815837-6 2010 Thalidomide disrupted integrin-mediated signaling pathways involved in gelatinase induction and release, such as Src and MAP-kinase ERK activation, resulting in decreased cell motility and invasiveness. Thalidomide 0-11 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 113-116 19815837-7 2010 Unexpectedly, treatment with thalidomide elicited an increase in fibronectin-induced Akt phosphorylation through phosphoinositide 3-kinase-independent pathways since thalidomide decreased fibronectin-induced phosphoinositide 3-kinase phosphorylation and reversed the inhibition of Akt phosphorylation achieved by the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. Thalidomide 29-40 fibronectin 1 Homo sapiens 65-76 19815837-7 2010 Unexpectedly, treatment with thalidomide elicited an increase in fibronectin-induced Akt phosphorylation through phosphoinositide 3-kinase-independent pathways since thalidomide decreased fibronectin-induced phosphoinositide 3-kinase phosphorylation and reversed the inhibition of Akt phosphorylation achieved by the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. Thalidomide 29-40 AKT serine/threonine kinase 1 Homo sapiens 85-88 19815837-7 2010 Unexpectedly, treatment with thalidomide elicited an increase in fibronectin-induced Akt phosphorylation through phosphoinositide 3-kinase-independent pathways since thalidomide decreased fibronectin-induced phosphoinositide 3-kinase phosphorylation and reversed the inhibition of Akt phosphorylation achieved by the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. Thalidomide 29-40 fibronectin 1 Homo sapiens 188-199 19815837-7 2010 Unexpectedly, treatment with thalidomide elicited an increase in fibronectin-induced Akt phosphorylation through phosphoinositide 3-kinase-independent pathways since thalidomide decreased fibronectin-induced phosphoinositide 3-kinase phosphorylation and reversed the inhibition of Akt phosphorylation achieved by the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. Thalidomide 29-40 AKT serine/threonine kinase 1 Homo sapiens 281-284 19815837-7 2010 Unexpectedly, treatment with thalidomide elicited an increase in fibronectin-induced Akt phosphorylation through phosphoinositide 3-kinase-independent pathways since thalidomide decreased fibronectin-induced phosphoinositide 3-kinase phosphorylation and reversed the inhibition of Akt phosphorylation achieved by the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. Thalidomide 166-177 fibronectin 1 Homo sapiens 65-76 19815837-7 2010 Unexpectedly, treatment with thalidomide elicited an increase in fibronectin-induced Akt phosphorylation through phosphoinositide 3-kinase-independent pathways since thalidomide decreased fibronectin-induced phosphoinositide 3-kinase phosphorylation and reversed the inhibition of Akt phosphorylation achieved by the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. Thalidomide 166-177 AKT serine/threonine kinase 1 Homo sapiens 85-88 19815837-7 2010 Unexpectedly, treatment with thalidomide elicited an increase in fibronectin-induced Akt phosphorylation through phosphoinositide 3-kinase-independent pathways since thalidomide decreased fibronectin-induced phosphoinositide 3-kinase phosphorylation and reversed the inhibition of Akt phosphorylation achieved by the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. Thalidomide 166-177 fibronectin 1 Homo sapiens 188-199 19757088-0 2010 Anti-fibrotic effect of thalidomide through inhibiting TGF-beta-induced ERK1/2 pathways in bleomycin-induced lung fibrosis in mice. Thalidomide 24-35 transforming growth factor, beta 1 Mus musculus 55-63 19757088-0 2010 Anti-fibrotic effect of thalidomide through inhibiting TGF-beta-induced ERK1/2 pathways in bleomycin-induced lung fibrosis in mice. Thalidomide 24-35 mitogen-activated protein kinase 3 Mus musculus 72-78 19757088-5 2010 RESULTS: Thalidomide administration significantly inhibits TGF-beta1 mRNA expression in a dose-dependant manner following administration of IL-6 and IL-6R. Thalidomide 9-20 transforming growth factor, beta 1 Mus musculus 59-68 19815837-7 2010 Unexpectedly, treatment with thalidomide elicited an increase in fibronectin-induced Akt phosphorylation through phosphoinositide 3-kinase-independent pathways since thalidomide decreased fibronectin-induced phosphoinositide 3-kinase phosphorylation and reversed the inhibition of Akt phosphorylation achieved by the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. Thalidomide 166-177 AKT serine/threonine kinase 1 Homo sapiens 281-284 19815837-9 2010 The unexpected effects of thalidomide on Akt activation indicate the need for further studies to elucidate whether the interference with Akt downstream effects would synergize with the anti-tumor activity of thalidomide. Thalidomide 26-37 AKT serine/threonine kinase 1 Homo sapiens 41-44 20034904-1 2009 OBJECTIVE: To investigate the expression of B7 co-stimulatory molecules in human multiple myeloma (MM) and the immunoregulatory effects of thalidomide on B7.1 co-stimulator. Thalidomide 139-150 CD80 molecule Homo sapiens 154-158 19757088-5 2010 RESULTS: Thalidomide administration significantly inhibits TGF-beta1 mRNA expression in a dose-dependant manner following administration of IL-6 and IL-6R. Thalidomide 9-20 interleukin 6 Mus musculus 140-144 19757088-5 2010 RESULTS: Thalidomide administration significantly inhibits TGF-beta1 mRNA expression in a dose-dependant manner following administration of IL-6 and IL-6R. Thalidomide 9-20 interleukin 6 receptor, alpha Mus musculus 149-154 19757088-6 2010 In the analysis of BAL fluids, total BAL inflammatory cell counts, TGF-beta1, and IL-6 levels in thalidomide-treated mice were significantly reduced when compared with bleomycin-treated mice (p < 0.01, p < 0.01, and p < 0.001, respectively). Thalidomide 97-108 transforming growth factor, beta 1 Mus musculus 67-76 19757088-6 2010 In the analysis of BAL fluids, total BAL inflammatory cell counts, TGF-beta1, and IL-6 levels in thalidomide-treated mice were significantly reduced when compared with bleomycin-treated mice (p < 0.01, p < 0.01, and p < 0.001, respectively). Thalidomide 97-108 interleukin 6 Mus musculus 82-86 19757088-7 2010 Thalidomide inhibited total ERK1/2 and phospho-ERK1/2 expression after TGF-beta1 stimulation in the RT-PCR and western blotting. Thalidomide 0-11 mitogen-activated protein kinase 3 Mus musculus 28-34 19757088-7 2010 Thalidomide inhibited total ERK1/2 and phospho-ERK1/2 expression after TGF-beta1 stimulation in the RT-PCR and western blotting. Thalidomide 0-11 mitogen-activated protein kinase 3 Mus musculus 47-53 19757088-7 2010 Thalidomide inhibited total ERK1/2 and phospho-ERK1/2 expression after TGF-beta1 stimulation in the RT-PCR and western blotting. Thalidomide 0-11 transforming growth factor, beta 1 Mus musculus 71-80 19757088-8 2010 CONCLUSION: The results of our study suggest that the anti-fibrotic effect of thalidomide on lung fibrosis may be related to suppression of the TGF-beta1-induced ERK1/2 signaling pathway. Thalidomide 78-89 transforming growth factor, beta 1 Mus musculus 144-153 19757088-8 2010 CONCLUSION: The results of our study suggest that the anti-fibrotic effect of thalidomide on lung fibrosis may be related to suppression of the TGF-beta1-induced ERK1/2 signaling pathway. Thalidomide 78-89 mitogen-activated protein kinase 3 Mus musculus 162-168 19926820-1 2010 The glutamic acid derivative thalidomide is a transcriptional inhibitor of TNF-alpha but is also known to affect human blood vessels, which may underlie its teratogenicity. Thalidomide 29-40 tumor necrosis factor Homo sapiens 75-84 19926820-7 2010 Effects of thalidomide on NF-kappaB activation, cyclooxygenase (COX)-2, and inducible nitric oxide synthase (iNOS) expression in TNF-alpha/LPS-activated HIMEC were determined by RT-PCR and Western blotting. Thalidomide 11-22 tumor necrosis factor Homo sapiens 129-138 19926820-7 2010 Effects of thalidomide on NF-kappaB activation, cyclooxygenase (COX)-2, and inducible nitric oxide synthase (iNOS) expression in TNF-alpha/LPS-activated HIMEC were determined by RT-PCR and Western blotting. Thalidomide 11-22 interferon regulatory factor 6 Homo sapiens 139-142 19926820-11 2010 VEGF-induced HIMEC transmigration, growth, proliferation, tube formation, and Akt phosphorylation were significantly inhibited by thalidomide. Thalidomide 130-141 vascular endothelial growth factor A Homo sapiens 0-4 20522972-3 2010 BALB/c mice sensitized and challenged with ovalbumin (OVA) were treated orally with thalidomide (30, 100, or 300 mg/kg) or a vehicle. Thalidomide 84-95 serine (or cysteine) peptidase inhibitor, clade B, member 1, pseudogene Mus musculus 43-52 20522972-6 2010 Thalidomide inhibited the elevated levels of interleukin-5 (IL-5) and tumor necrosis factor-alpha (TNF-alpha) in BALF by OVA challenges. Thalidomide 0-11 interleukin 5 Mus musculus 45-58 20522972-6 2010 Thalidomide inhibited the elevated levels of interleukin-5 (IL-5) and tumor necrosis factor-alpha (TNF-alpha) in BALF by OVA challenges. Thalidomide 0-11 interleukin 5 Mus musculus 60-64 20522972-6 2010 Thalidomide inhibited the elevated levels of interleukin-5 (IL-5) and tumor necrosis factor-alpha (TNF-alpha) in BALF by OVA challenges. Thalidomide 0-11 tumor necrosis factor Mus musculus 70-97 20522972-6 2010 Thalidomide inhibited the elevated levels of interleukin-5 (IL-5) and tumor necrosis factor-alpha (TNF-alpha) in BALF by OVA challenges. Thalidomide 0-11 tumor necrosis factor Mus musculus 99-108 21048383-3 2010 The suppressive effect of thalidomide on the activation of the nuclear transcription factor NF-kappaB may explain these effects of thalidomide. Thalidomide 26-37 nuclear factor kappa B subunit 1 Homo sapiens 92-101 21048383-3 2010 The suppressive effect of thalidomide on the activation of the nuclear transcription factor NF-kappaB may explain these effects of thalidomide. Thalidomide 131-142 nuclear factor kappa B subunit 1 Homo sapiens 92-101 20306635-9 2009 Treatment with thalidomide showed a significant benefit compared to aspirin (RR 2.43; 95% CI 1.28 to 4.59). Thalidomide 15-26 ribonucleotide reductase regulatory subunit M2 Homo sapiens 77-81 20304198-9 2010 Therapy with either thalidomide or tamoxifen effectively maintained alpha-smooth muscle actin expression in the media, similar to uninjured arteries. Thalidomide 20-31 actin gamma 2, smooth muscle Rattus norvegicus 68-93 20034801-0 2010 New thalidomide analogues derived through Sonogashira or Suzuki reactions and their TNF expression inhibition profiles. Thalidomide 4-15 tumor necrosis factor Homo sapiens 84-87 20034801-2 2010 All thalidomide analogues were tested for their ability to inhibit the expression of the proinflammatory cytokine Tumor Necrosis Factor (TNF). Thalidomide 4-15 tumor necrosis factor Homo sapiens 114-135 20034801-2 2010 All thalidomide analogues were tested for their ability to inhibit the expression of the proinflammatory cytokine Tumor Necrosis Factor (TNF). Thalidomide 4-15 tumor necrosis factor Homo sapiens 137-140 20034801-4 2010 Several compounds either containing either an aryl-isobutyl or aryl-isopropoxy group were the most effective in inhibiting TNF expression, and were several times more active than thalidomide itself. Thalidomide 179-190 tumor necrosis factor Homo sapiens 123-126 20039400-4 2010 The aim of this study was to assess the efficacy of thalidomide in patients with severe EOS, based on etiology supporting an initial role of NF-kappaB in activation of this disease. Thalidomide 52-63 nuclear factor kappa B subunit 1 Homo sapiens 141-150 20039400-10 2010 CONCLUSION: The ability of thalidomide to improve clinical symptoms and laboratory findings in patients with EOS indicates a central role for NF-kappaB activity in this disorder. Thalidomide 27-38 nuclear factor kappa B subunit 1 Homo sapiens 142-151 20039400-11 2010 Inhibition of IKK might be a pharmacologic action by which thalidomide down-regulates NF-kappaB signaling. Thalidomide 59-70 nuclear factor kappa B subunit 1 Homo sapiens 86-95 20823570-11 2010 Pretreatment with quercetin or thalidomide significantly attenuated the severity of cerulein-induced acute pancreatitis as evidenced by effective reductions in the pancreatic wet weight/body weight ratio, biochemical indices, proinflammatory cytokines, myeloperoxidase activity, malondialdehyde formation, and an increase in antiinflammatory cytokine IL-10. Thalidomide 31-42 myeloperoxidase Mus musculus 253-268 20823570-11 2010 Pretreatment with quercetin or thalidomide significantly attenuated the severity of cerulein-induced acute pancreatitis as evidenced by effective reductions in the pancreatic wet weight/body weight ratio, biochemical indices, proinflammatory cytokines, myeloperoxidase activity, malondialdehyde formation, and an increase in antiinflammatory cytokine IL-10. Thalidomide 31-42 interleukin 10 Mus musculus 351-356 20190484-2 2010 Thus, we treated the patient with thalidomide plus dexamethasone, which brought about a marked improvement of systolic dysfunction, POEMS syndrome-related symptoms and the serum concentrations of vascular endothelial growth factor. Thalidomide 34-45 vascular endothelial growth factor A Homo sapiens 196-230 20034904-2 2009 METHODS: The immunoregulatory effects of thalidomide on the expression of B7-1 in human MM cell line was examined by detecting the changes in the expression of B7 co-stimulator on the cells using flow cytometry following the drug treatment. Thalidomide 41-52 CD80 molecule Homo sapiens 74-78 20034904-6 2009 Thalidomide can up-regulate the expression of B7-1 molecules on myeloma cells, which is probably one of the therapeutic mechanisms of thalidomide. Thalidomide 0-11 CD80 molecule Homo sapiens 46-50 20034904-6 2009 Thalidomide can up-regulate the expression of B7-1 molecules on myeloma cells, which is probably one of the therapeutic mechanisms of thalidomide. Thalidomide 134-145 CD80 molecule Homo sapiens 46-50 19843943-0 2009 Thalidomide inhibits activation of caspase-1. Thalidomide 0-11 caspase 1 Mus musculus 35-44 19458984-3 2009 We sought to investigate the effects of Thalidomide (Th), a potent anti-angiogenic drug on vasa vasorum (VV) neovascularization, vessel wall inflammation, and neointima formation in early experimental atherosclerosis. Thalidomide 40-51 DEAD-box helicase 4 Sus scrofa 91-95 19458984-8 2009 Treatment with Thalidomide preserved VV spatial density [2.7 +/- 0.3 (N), 6.4 +/- 0.7 (HC), 3.5 +/- 0.8 (HC + Th); p = ns HC + Th vs. N] and inhibited the expression of VEGF, TNF-alpha and LOX-1, but not NFkappabeta activity in the coronary vessel wall. Thalidomide 15-26 vascular endothelial growth factor A Sus scrofa 169-173 19458984-8 2009 Treatment with Thalidomide preserved VV spatial density [2.7 +/- 0.3 (N), 6.4 +/- 0.7 (HC), 3.5 +/- 0.8 (HC + Th); p = ns HC + Th vs. N] and inhibited the expression of VEGF, TNF-alpha and LOX-1, but not NFkappabeta activity in the coronary vessel wall. Thalidomide 15-26 tumor necrosis factor Sus scrofa 175-184 19458984-8 2009 Treatment with Thalidomide preserved VV spatial density [2.7 +/- 0.3 (N), 6.4 +/- 0.7 (HC), 3.5 +/- 0.8 (HC + Th); p = ns HC + Th vs. N] and inhibited the expression of VEGF, TNF-alpha and LOX-1, but not NFkappabeta activity in the coronary vessel wall. Thalidomide 15-26 oxidized low density lipoprotein receptor 1 Sus scrofa 189-194 19843943-5 2009 Thalidomide-treated cells also released less of other leaderless proteins, which require caspase-1 activity for their secretion. Thalidomide 0-11 caspase 1 Mus musculus 89-98 19843943-8 2009 The anti-inflammatory activity of thalidomide was also mediated via caspase-1 in mice. Thalidomide 34-45 caspase 1 Mus musculus 68-77 19843943-9 2009 These findings represent a novel mechanism by which thalidomide exerts its pharmacological activity and suggest that inhibition of the activity of IL-1 might represent a novel strategy to substitute thalidomide. Thalidomide 52-63 interleukin 1 complex Mus musculus 147-151 19843943-9 2009 These findings represent a novel mechanism by which thalidomide exerts its pharmacological activity and suggest that inhibition of the activity of IL-1 might represent a novel strategy to substitute thalidomide. Thalidomide 199-210 interleukin 1 complex Mus musculus 147-151 19639185-3 2009 Besides its well-documented anti-angiogenic effects, thalidomide therapy could result in a decrease in ABCB1 gene expression. Thalidomide 53-64 ATP binding cassette subfamily B member 1 Homo sapiens 103-108 19814869-12 2009 Thalidomide also provided significant protection against TNBS-induced colonic damage in terms of morphological and histological score and levels of lipid peroxidation, superoxide dismutase, catalase and glutathione peroxidase in colonic tissue. Thalidomide 0-11 catalase Rattus norvegicus 190-198 19954656-0 2009 [Effect of TNF-alpha gene polymorphism on outcome of thalidomide-based regimens for multiple myeloma]. Thalidomide 53-64 tumor necrosis factor Homo sapiens 11-20 19954656-1 2009 OBJECTIVE: To evaluate the effect of polymorphism at the -238 and -308 position of the TNF-alpha promotor region on the clinical outcome of thalidomide (Thal)-based regimens for the treatment of multiple myeloma (MM). Thalidomide 140-151 tumor necrosis factor Homo sapiens 87-96 19954656-1 2009 OBJECTIVE: To evaluate the effect of polymorphism at the -238 and -308 position of the TNF-alpha promotor region on the clinical outcome of thalidomide (Thal)-based regimens for the treatment of multiple myeloma (MM). Thalidomide 153-157 tumor necrosis factor Homo sapiens 87-96 19954656-8 2009 CONCLUSION: The TNF-alpha -238 polymorphic status is associated with a favorable clinical outcome in MM patients treated with thalidomide-based regimen. Thalidomide 126-137 tumor necrosis factor Homo sapiens 16-25 19922130-2 2009 We conducted a preliminary phase II trial of thalidomide, which reduces levels of TNF-alpha pre-transcriptionally and post-transcriptionally in vivo and has been shown to prolong disease duration and extend the lifespan of transgenic animal models of ALS. Thalidomide 45-56 tumor necrosis factor Homo sapiens 82-91 19922130-10 2009 In conclusion, treatment of ALS with the TNF-alpha inhibitor, thalidomide, does not appear to effectively modulate disease progression and can cause adverse effects. Thalidomide 62-73 tumor necrosis factor Homo sapiens 41-50 19394719-0 2009 Discovering a new analogue of thalidomide which may be used as a potent modulator of TNF-alpha production. Thalidomide 30-41 tumor necrosis factor Homo sapiens 85-94 19394719-1 2009 A new series of imide derivatives related to thalidomide were synthesized and evaluated as modulators of TNF-alpha production. Thalidomide 45-56 tumor necrosis factor Homo sapiens 105-114 19639185-8 2009 Conversely thalidomide and PGA induced down-regulation of VEGF gene expression in both drug-sensitive and -resistant myeloma cells. Thalidomide 11-22 vascular endothelial growth factor A Homo sapiens 58-62 19680075-7 2009 Thalidomide also caused a substantial reduction of 1) the rise in myeloperoxidase activity (mucosa); 2) the expression in the tissue of TNF-alpha, IL-1beta, transforming growth factor-beta, and vascular endothelial growth factor; 3) the increase in staining (immunohistochemistry) for nitrotyrosine and for poly(ADP ribose), as well as 4) the nuclear factor-kappaB activation caused by CG in the peritoneum. Thalidomide 0-11 myeloperoxidase Rattus norvegicus 66-81 21172267-1 2009 BACKGROUND: Thalidomide, one of whose activities is to inhibit Tumour Necrosis Factor (TNF)-alpha production, has been reported to be an effective treatment for refractory inflammatory bowel disease (IBD). Thalidomide 12-23 tumor necrosis factor Homo sapiens 63-97 21172267-2 2009 TNF-alpha driven production of matrix metalloproteinase (MMP)-3 by gut lamina propria mononuclear cells (LPMCs) is a major pathway of tissue injury in IBD; however the effect of thalidomide and newer more potent immunomodulatory derivatives on this pathway has not been studied. Thalidomide 178-189 tumor necrosis factor Homo sapiens 0-9 21172267-2 2009 TNF-alpha driven production of matrix metalloproteinase (MMP)-3 by gut lamina propria mononuclear cells (LPMCs) is a major pathway of tissue injury in IBD; however the effect of thalidomide and newer more potent immunomodulatory derivatives on this pathway has not been studied. Thalidomide 178-189 matrix metallopeptidase 3 Homo sapiens 31-63 19680075-7 2009 Thalidomide also caused a substantial reduction of 1) the rise in myeloperoxidase activity (mucosa); 2) the expression in the tissue of TNF-alpha, IL-1beta, transforming growth factor-beta, and vascular endothelial growth factor; 3) the increase in staining (immunohistochemistry) for nitrotyrosine and for poly(ADP ribose), as well as 4) the nuclear factor-kappaB activation caused by CG in the peritoneum. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 136-145 19680075-7 2009 Thalidomide also caused a substantial reduction of 1) the rise in myeloperoxidase activity (mucosa); 2) the expression in the tissue of TNF-alpha, IL-1beta, transforming growth factor-beta, and vascular endothelial growth factor; 3) the increase in staining (immunohistochemistry) for nitrotyrosine and for poly(ADP ribose), as well as 4) the nuclear factor-kappaB activation caused by CG in the peritoneum. Thalidomide 0-11 interleukin 1 beta Rattus norvegicus 147-155 19604271-7 2009 THD inhibited the up-regulation of HYP protein, pro-collagen III mRNA and alpha-SMA protein induced by TGF-beta1 in HFL-F cells, and additionally inhibited pro-collagen III mRNA expression on trans-differentiated MF. Thalidomide 0-3 actin alpha 2, smooth muscle, aorta Mus musculus 74-83 19604271-7 2009 THD inhibited the up-regulation of HYP protein, pro-collagen III mRNA and alpha-SMA protein induced by TGF-beta1 in HFL-F cells, and additionally inhibited pro-collagen III mRNA expression on trans-differentiated MF. Thalidomide 0-3 transforming growth factor, beta 1 Mus musculus 103-112 19604271-8 2009 THD reduced HYP synthesis in the lung tissues of BLM-treated mice at week 4, and slightly reduced the numbers of alpha-SMA-positive cells. Thalidomide 0-3 actin alpha 2, smooth muscle, aorta Mus musculus 113-122 19698226-5 2009 The combination of THD in concentration of 80 or 100 microg/ml with Dx in concentration of 4 microg/ml decreased the expression of IL-6, TNF-alpha and survivin, increased the expression of ES, while no influence on VEGF expression was found. Thalidomide 19-22 interleukin 6 Homo sapiens 131-135 19538513-5 2009 Further, thalidomide inhibited the MyD88 augmentation in lipopolysaccharide-stimulated cells, whereas it did not alter the expression of TIR domain-containing adaptor-inducing IFN-beta in the MyD88-independent pathway. Thalidomide 9-20 myeloid differentiation primary response gene 88 Mus musculus 35-40 19538513-6 2009 Thalidomide significantly inhibited the NO production in response to Pam(3)Cys, CpG DNA and imiquimod as MyD88-dependent Toll-like receptor (TLR) ligands, but not polyI:C as a MyD88-independent TLR ligand. Thalidomide 0-11 myeloid differentiation primary response gene 88 Mus musculus 105-110 19538513-6 2009 Thalidomide significantly inhibited the NO production in response to Pam(3)Cys, CpG DNA and imiquimod as MyD88-dependent Toll-like receptor (TLR) ligands, but not polyI:C as a MyD88-independent TLR ligand. Thalidomide 0-11 myeloid differentiation primary response gene 88 Mus musculus 176-181 19538513-7 2009 Therefore, thalidomide was suggested to inhibit lipopolysaccharide-induced NO production via downregulation of the MyD88-dependent signal pathway. Thalidomide 11-22 myeloid differentiation primary response gene 88 Mus musculus 115-120 19434479-2 2009 In this study we examined the effect of drugs interfering with TNF expression (thalidomide) and activity (infliximab) and compared it to that of a H(1)R histamine receptor antagonist (loratadine) in a model of histamine-induced rat hind-paw oedema formation. Thalidomide 79-90 tumor necrosis factor Rattus norvegicus 63-66 19663116-4 2009 We report a case of deep venous thrombosis (DVT) and pulmonary embolus (PE) following thalidomide use in a patient with leprosy (erythema nodosum leprosum, ENL) who was concurrently treated with prednisone, as well as a review of relevant literature. Thalidomide 86-97 MLLT1 super elongation complex subunit Homo sapiens 156-159 19698226-5 2009 The combination of THD in concentration of 80 or 100 microg/ml with Dx in concentration of 4 microg/ml decreased the expression of IL-6, TNF-alpha and survivin, increased the expression of ES, while no influence on VEGF expression was found. Thalidomide 19-22 tumor necrosis factor Homo sapiens 137-146 19698226-6 2009 It is concluded that THD combined with Dx shows the synergistic inhibitory effect on KM3 cells, they bring the effect resistant to multiple myeloma probably through down-regulating the expression of IL-6, TNF-alpha and survivin, and up-regulating the expression of ES in KM3 cell. Thalidomide 21-24 interleukin 6 Homo sapiens 199-203 19698226-6 2009 It is concluded that THD combined with Dx shows the synergistic inhibitory effect on KM3 cells, they bring the effect resistant to multiple myeloma probably through down-regulating the expression of IL-6, TNF-alpha and survivin, and up-regulating the expression of ES in KM3 cell. Thalidomide 21-24 tumor necrosis factor Homo sapiens 205-214 20021944-11 2009 The caspase-3 positivity of SMMC-7721 cells treated with thalidomide was increasing along with the increase of treatment time or drug concentration, but not in the control cells. Thalidomide 57-68 caspase 3 Homo sapiens 4-13 20021944-12 2009 The VEGF content in SMMC-7721 cells was lowering when thalidomide was used in an increasing concentration. Thalidomide 54-65 vascular endothelial growth factor A Homo sapiens 4-8 19822087-8 2009 Autocrinal TNF-alpha production and modulation of cadhelin expression to regulate cell adhesion might be involved in this inhibitory action of thalidomide. Thalidomide 143-154 tumor necrosis factor Homo sapiens 11-20 19569016-3 2009 The aim of our study was to examine the direct effect of various adiponectin concentrations applied separately or in combination with thalidomide on the growth of the murine endothelial cell line HECa 10 in 24- and 72-hour cell cultures. Thalidomide 134-145 hdc homolog, cell cycle regulator Mus musculus 196-200 20981323-6 2009 Treatment with thalidomide, an immunomodulatory agent, showed elevation of CCL5 by GEP but was not cytotoxic to CLL cells. Thalidomide 15-26 C-C motif chemokine ligand 5 Homo sapiens 75-79 18976811-0 2009 Thalidomide treatment down-regulates SDF-1alpha and CXCR4 expression in multiple myeloma patients. Thalidomide 0-11 C-X-C motif chemokine receptor 4 Homo sapiens 52-57 18976811-2 2009 Here, we show that the expression of SDF-1alpha and CXCR4 was significantly down-regulated in patients treated with thalidomide (n=10) as compared to newly diagnosed MM patients (n=31) and MM patients treated with other drugs (n=38). Thalidomide 116-127 C-X-C motif chemokine receptor 4 Homo sapiens 52-57 18976811-3 2009 SDF-1 alpha and CXCR4 expression was also significantly decreased in a RPMI 8226 cell line treated with 10 and 20micromol/L of thalidomide. Thalidomide 127-138 C-X-C motif chemokine receptor 4 Homo sapiens 16-21 18976811-4 2009 Our findings indicate that thalidomide therapy induces down-regulation of CXCR4 and its ligand SDF-1alpha in multiple myeloma. Thalidomide 27-38 C-X-C motif chemokine receptor 4 Homo sapiens 74-79 19954599-0 2009 [Effects of thalidomide on Annexin II gene regulation]. Thalidomide 12-23 annexin A2 Homo sapiens 27-37 19954599-1 2009 OBJECTIVE: To investigate the effect of thalidomide on Annexin II (AnxA2) gene regulation in multiple myeloma cell line RPMI8226 and human microvascular endothelial cell line HMEC-1 cells in vitro, and explore the potential mechanism of thrombosis induced by thalidomide. Thalidomide 40-51 annexin A2 Homo sapiens 55-65 19954599-1 2009 OBJECTIVE: To investigate the effect of thalidomide on Annexin II (AnxA2) gene regulation in multiple myeloma cell line RPMI8226 and human microvascular endothelial cell line HMEC-1 cells in vitro, and explore the potential mechanism of thrombosis induced by thalidomide. Thalidomide 40-51 annexin A2 Homo sapiens 67-72 19954599-3 2009 Real time quantitative PCR (RQ-PCR) was used to detect the influence of thalidomide at different concentration on the expression of AnxA2 mRNA, flow cytometry (FCM) and confocal microscopy were used to detect the cell surface protein level after the samples were stimulated with different concentrations of thalidomide. Thalidomide 72-83 annexin A2 Homo sapiens 132-137 19954599-6 2009 CONCLUSIONS: Thalidomide can inhibit the expression of AnxA2 mRNA and protein in RPMI8226 and HMEC-1 cells, which may be one of the mechanisms for the development of thrombosis induced by thalidomide in multiple myeloma patients. Thalidomide 13-24 annexin A2 Homo sapiens 55-60 19954599-6 2009 CONCLUSIONS: Thalidomide can inhibit the expression of AnxA2 mRNA and protein in RPMI8226 and HMEC-1 cells, which may be one of the mechanisms for the development of thrombosis induced by thalidomide in multiple myeloma patients. Thalidomide 188-199 annexin A2 Homo sapiens 55-60 19675744-2 2009 Thalidomide has been described as inhibitor of the fibroblast growth factor (FGF) and the vascular endothelial growth factor (VEGF). Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 90-124 19675744-2 2009 Thalidomide has been described as inhibitor of the fibroblast growth factor (FGF) and the vascular endothelial growth factor (VEGF). Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 126-130 19567134-15 2009 The expression level of phosphatase and tensin homolog deleted on chromosome ten (PTEN), which promoted autophagy by suppressing PI3K/Akt/mTOR signaling pathway, was elevated by thalidomide (thalidomide group: P = 0.000; combined group: P = 0.002). Thalidomide 178-189 phosphatase and tensin homolog Homo sapiens 82-86 19567134-15 2009 The expression level of phosphatase and tensin homolog deleted on chromosome ten (PTEN), which promoted autophagy by suppressing PI3K/Akt/mTOR signaling pathway, was elevated by thalidomide (thalidomide group: P = 0.000; combined group: P = 0.002). Thalidomide 178-189 AKT serine/threonine kinase 1 Homo sapiens 134-137 19567134-15 2009 The expression level of phosphatase and tensin homolog deleted on chromosome ten (PTEN), which promoted autophagy by suppressing PI3K/Akt/mTOR signaling pathway, was elevated by thalidomide (thalidomide group: P = 0.000; combined group: P = 0.002). Thalidomide 178-189 mechanistic target of rapamycin kinase Homo sapiens 138-142 19567134-15 2009 The expression level of phosphatase and tensin homolog deleted on chromosome ten (PTEN), which promoted autophagy by suppressing PI3K/Akt/mTOR signaling pathway, was elevated by thalidomide (thalidomide group: P = 0.000; combined group: P = 0.002). Thalidomide 191-202 phosphatase and tensin homolog Homo sapiens 82-86 19567134-15 2009 The expression level of phosphatase and tensin homolog deleted on chromosome ten (PTEN), which promoted autophagy by suppressing PI3K/Akt/mTOR signaling pathway, was elevated by thalidomide (thalidomide group: P = 0.000; combined group: P = 0.002). Thalidomide 191-202 AKT serine/threonine kinase 1 Homo sapiens 134-137 19567134-15 2009 The expression level of phosphatase and tensin homolog deleted on chromosome ten (PTEN), which promoted autophagy by suppressing PI3K/Akt/mTOR signaling pathway, was elevated by thalidomide (thalidomide group: P = 0.000; combined group: P = 0.002). Thalidomide 191-202 mechanistic target of rapamycin kinase Homo sapiens 138-142 19567134-17 2009 Contributing to the up-regulation of PTEN by thalidomide, the expression of autophagy associated protein-MAP1LC3 and Beclin1 was enhanced, which leads to a reinforced autophagy in the combined treatment of temozolomide and thalidomide in vitro. Thalidomide 45-56 phosphatase and tensin homolog Homo sapiens 37-41 19567134-17 2009 Contributing to the up-regulation of PTEN by thalidomide, the expression of autophagy associated protein-MAP1LC3 and Beclin1 was enhanced, which leads to a reinforced autophagy in the combined treatment of temozolomide and thalidomide in vitro. Thalidomide 45-56 beclin 1 Homo sapiens 117-124 19567134-17 2009 Contributing to the up-regulation of PTEN by thalidomide, the expression of autophagy associated protein-MAP1LC3 and Beclin1 was enhanced, which leads to a reinforced autophagy in the combined treatment of temozolomide and thalidomide in vitro. Thalidomide 223-234 phosphatase and tensin homolog Homo sapiens 37-41 19567134-17 2009 Contributing to the up-regulation of PTEN by thalidomide, the expression of autophagy associated protein-MAP1LC3 and Beclin1 was enhanced, which leads to a reinforced autophagy in the combined treatment of temozolomide and thalidomide in vitro. Thalidomide 223-234 beclin 1 Homo sapiens 117-124 19220680-6 2009 For the thalidomide treated group, despite the fact that MVD-CD34 of larger metastases was more than that of small metastases (P < 0.01), the MVD and BD were similar to those of small metastases respectively (P > 0.05). Thalidomide 8-19 CD34 antigen Mus musculus 61-65 19413671-3 2009 Thalidomide, which inhibited TNF-alpha effectively, has been used clinically in states of chronic TNF-alpha elevation with encouraging results. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 29-38 19467946-0 2009 Chronic thalidomide administration enhances vascular responsiveness to vasopressin in portal-systemic collaterals of bile duct-ligated rats. Thalidomide 8-19 arginine vasopressin Rattus norvegicus 71-82 19467946-2 2009 It has been shown that chronic thalidomide treatment decreases portal pressure, attenuates hyperdynamic circulation and inhibits vascular endothelial growth factor (VEGF) and tumor necrosis factor (TNF)-alpha in partially portal vein-ligated rats. Thalidomide 31-42 vascular endothelial growth factor A Rattus norvegicus 129-163 19467946-2 2009 It has been shown that chronic thalidomide treatment decreases portal pressure, attenuates hyperdynamic circulation and inhibits vascular endothelial growth factor (VEGF) and tumor necrosis factor (TNF)-alpha in partially portal vein-ligated rats. Thalidomide 31-42 vascular endothelial growth factor A Rattus norvegicus 165-169 19467946-2 2009 It has been shown that chronic thalidomide treatment decreases portal pressure, attenuates hyperdynamic circulation and inhibits vascular endothelial growth factor (VEGF) and tumor necrosis factor (TNF)-alpha in partially portal vein-ligated rats. Thalidomide 31-42 tumor necrosis factor Rattus norvegicus 175-208 19467946-11 2009 Compared with the control group, thalidomide-treated rats had significantly lower plasma VEGF levels (p < 0.001), accompanied by an insignificant reduction in plasma TNF-alpha levels (p > 0.05). Thalidomide 33-44 vascular endothelial growth factor A Rattus norvegicus 89-93 19467946-12 2009 The expressions of VEGF and TNF-alpha mRNA in the left adrenal veins of thalidomide-treated CBDL rats were not significantly changed compared with those of the control group. Thalidomide 72-83 vascular endothelial growth factor A Rattus norvegicus 19-23 19467946-12 2009 The expressions of VEGF and TNF-alpha mRNA in the left adrenal veins of thalidomide-treated CBDL rats were not significantly changed compared with those of the control group. Thalidomide 72-83 tumor necrosis factor Rattus norvegicus 28-37 19467946-14 2009 CONCLUSION: In cirrhotic rats, chronic thalidomide treatment improves the portal-systemic collateral vascular responsiveness to AVP, which was partly related to VEGF inhibition. Thalidomide 39-50 vascular endothelial growth factor A Rattus norvegicus 161-165 19413671-3 2009 Thalidomide, which inhibited TNF-alpha effectively, has been used clinically in states of chronic TNF-alpha elevation with encouraging results. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 98-107 19413671-11 2009 CONCLUSIONS: Thalidomide decreased the PVP and IHR through the attenuation of anandamide-induced constrictive response, decreasing the production of TNF-alpha, IL-6 and TXA(2) in the liver and the suppression of hepatic fibrogenesis of rats with biliary cirrhosis of this study. Thalidomide 13-24 tumor necrosis factor Rattus norvegicus 149-158 19413671-11 2009 CONCLUSIONS: Thalidomide decreased the PVP and IHR through the attenuation of anandamide-induced constrictive response, decreasing the production of TNF-alpha, IL-6 and TXA(2) in the liver and the suppression of hepatic fibrogenesis of rats with biliary cirrhosis of this study. Thalidomide 13-24 interleukin 6 Rattus norvegicus 160-164 19370684-2 2009 High levels of tumour necrosis factor-alpha (TNF-alpha) have been associated with the development of intestinal inflammation in Crohn"s disease.Thalidomide, has been demonstrated to have anti TNF-alpha properties in experimental and clinical studies. Thalidomide 144-155 tumor necrosis factor Homo sapiens 45-54 19050852-9 2009 Serum TNF-alpha level rose threefold after LPS treatment, but this rise was abolished by thalidomide. Thalidomide 89-100 tumor necrosis factor Rattus norvegicus 6-15 19370684-2 2009 High levels of tumour necrosis factor-alpha (TNF-alpha) have been associated with the development of intestinal inflammation in Crohn"s disease.Thalidomide, has been demonstrated to have anti TNF-alpha properties in experimental and clinical studies. Thalidomide 144-155 tumor necrosis factor Homo sapiens 192-201 19370685-2 2009 Thalidomide, a tumour necrosis factor-alpha (TNF-alpha) inhibitor and its analogue, lenalidomide, may have a role in the management of Crohn"s disease, but it is not clear whether it is an effective maintenance therapy. Thalidomide 0-11 tumor necrosis factor Homo sapiens 45-54 19271121-8 2009 Furthermore, thalidomide, a TNF-alpha antagonist, treatment abolished the LPS pretreatment-associated protective effects. Thalidomide 13-24 tumor necrosis factor Mus musculus 28-37 19222454-0 2009 Multifocal primary cutaneous CD30+ anaplastic large cell lymphoma responsive to thalidomide: the molecular mechanism and the clinical application. Thalidomide 80-91 TNF receptor superfamily member 8 Homo sapiens 29-33 19423468-7 2009 Treatment with thalidomide alone significantly (p<0.05) decreased interleukin-1 alpha (IL-1alpha), nitric oxide (NO) and malondialdehyde (MDA) levels in the serum without significantly (p<0.05) decreasing the bacterial count in blood. Thalidomide 15-26 interleukin 1 alpha Mus musculus 69-88 19423468-7 2009 Treatment with thalidomide alone significantly (p<0.05) decreased interleukin-1 alpha (IL-1alpha), nitric oxide (NO) and malondialdehyde (MDA) levels in the serum without significantly (p<0.05) decreasing the bacterial count in blood. Thalidomide 15-26 interleukin 1 alpha Mus musculus 90-99 19372569-4 2009 Curcumin also potentiated the apoptotic effects of thalidomide and bortezomib by down-regulating the constitutive activation of NF-kappaB and Akt, and this correlated with the suppression of NF-kappaB-regulated gene products, including cyclin D1, Bcl-xL, Bcl-2, TRAF1, cIAP-1, XIAP, survivin, and vascular endothelial growth factor. Thalidomide 51-62 AKT serine/threonine kinase 1 Homo sapiens 142-145 19372569-4 2009 Curcumin also potentiated the apoptotic effects of thalidomide and bortezomib by down-regulating the constitutive activation of NF-kappaB and Akt, and this correlated with the suppression of NF-kappaB-regulated gene products, including cyclin D1, Bcl-xL, Bcl-2, TRAF1, cIAP-1, XIAP, survivin, and vascular endothelial growth factor. Thalidomide 51-62 cyclin D1 Homo sapiens 236-245 19372569-4 2009 Curcumin also potentiated the apoptotic effects of thalidomide and bortezomib by down-regulating the constitutive activation of NF-kappaB and Akt, and this correlated with the suppression of NF-kappaB-regulated gene products, including cyclin D1, Bcl-xL, Bcl-2, TRAF1, cIAP-1, XIAP, survivin, and vascular endothelial growth factor. Thalidomide 51-62 BCL2 like 1 Homo sapiens 247-253 19372569-4 2009 Curcumin also potentiated the apoptotic effects of thalidomide and bortezomib by down-regulating the constitutive activation of NF-kappaB and Akt, and this correlated with the suppression of NF-kappaB-regulated gene products, including cyclin D1, Bcl-xL, Bcl-2, TRAF1, cIAP-1, XIAP, survivin, and vascular endothelial growth factor. Thalidomide 51-62 BCL2 apoptosis regulator Homo sapiens 255-260 19372569-4 2009 Curcumin also potentiated the apoptotic effects of thalidomide and bortezomib by down-regulating the constitutive activation of NF-kappaB and Akt, and this correlated with the suppression of NF-kappaB-regulated gene products, including cyclin D1, Bcl-xL, Bcl-2, TRAF1, cIAP-1, XIAP, survivin, and vascular endothelial growth factor. Thalidomide 51-62 TNF receptor associated factor 1 Homo sapiens 262-267 19372569-4 2009 Curcumin also potentiated the apoptotic effects of thalidomide and bortezomib by down-regulating the constitutive activation of NF-kappaB and Akt, and this correlated with the suppression of NF-kappaB-regulated gene products, including cyclin D1, Bcl-xL, Bcl-2, TRAF1, cIAP-1, XIAP, survivin, and vascular endothelial growth factor. Thalidomide 51-62 baculoviral IAP repeat containing 2 Homo sapiens 269-275 19372569-4 2009 Curcumin also potentiated the apoptotic effects of thalidomide and bortezomib by down-regulating the constitutive activation of NF-kappaB and Akt, and this correlated with the suppression of NF-kappaB-regulated gene products, including cyclin D1, Bcl-xL, Bcl-2, TRAF1, cIAP-1, XIAP, survivin, and vascular endothelial growth factor. Thalidomide 51-62 X-linked inhibitor of apoptosis Homo sapiens 277-281 19372569-4 2009 Curcumin also potentiated the apoptotic effects of thalidomide and bortezomib by down-regulating the constitutive activation of NF-kappaB and Akt, and this correlated with the suppression of NF-kappaB-regulated gene products, including cyclin D1, Bcl-xL, Bcl-2, TRAF1, cIAP-1, XIAP, survivin, and vascular endothelial growth factor. Thalidomide 51-62 vascular endothelial growth factor A Homo sapiens 297-331 19285019-2 2009 We evaluated the effects of thalidomide-related inhibition of TNF-alpha upon the hepatic microcirculation of cirrhosis in rats. Thalidomide 28-39 tumor necrosis factor Rattus norvegicus 62-71 19576118-9 2009 Real-time quantitative PCR and Western blot discovered that thalidomide inhibited the expression of VEGF/HIF-1alpha of HUVEC (P < 0.05). Thalidomide 60-71 vascular endothelial growth factor A Homo sapiens 100-104 19576118-9 2009 Real-time quantitative PCR and Western blot discovered that thalidomide inhibited the expression of VEGF/HIF-1alpha of HUVEC (P < 0.05). Thalidomide 60-71 hypoxia inducible factor 1 subunit alpha Homo sapiens 105-115 19576118-11 2009 CONCLUSIONS: Thalidomide can suppress the expression of HIF-1alpha and VEGF in HUVEC in vitro and then inhibit angiodysplasia, which may play a significant role in stopping the rebleeding in patients with recurrent gastrointestinal bleeding. Thalidomide 13-24 hypoxia inducible factor 1 subunit alpha Homo sapiens 56-66 19576118-11 2009 CONCLUSIONS: Thalidomide can suppress the expression of HIF-1alpha and VEGF in HUVEC in vitro and then inhibit angiodysplasia, which may play a significant role in stopping the rebleeding in patients with recurrent gastrointestinal bleeding. Thalidomide 13-24 vascular endothelial growth factor A Homo sapiens 71-75 19070887-7 2009 The findings suggest that a thalidomide-based regimen may overcome the poor prognosis associated with a cyclin D1-negative or fibroblast growth factor receptor 3-positive phenotype. Thalidomide 28-39 cyclin D1 Homo sapiens 104-113 19070887-7 2009 The findings suggest that a thalidomide-based regimen may overcome the poor prognosis associated with a cyclin D1-negative or fibroblast growth factor receptor 3-positive phenotype. Thalidomide 28-39 fibroblast growth factor receptor 3 Homo sapiens 126-161 19098937-0 2009 In vitro anticancer property of a novel thalidomide analogue through inhibition of NF-kappaB activation in HL-60 cells. Thalidomide 40-51 nuclear factor kappa B subunit 1 Homo sapiens 83-92 18937212-2 2009 Several inhibitors of NF-kappaB, like caffeic acid, captopril, curcumin, pyrrolidine dithiocarbamate, resveratrol, silymarin and thalidomide, have demonstrated antinecrotic, anticholestatic, antifibrotic and anticancer activities in the liver. Thalidomide 129-140 nuclear factor kappa B subunit 1 Homo sapiens 22-31 19167733-2 2009 MATERIALS AND METHODS: A total of 159 patients were enrolled in a double-blind randomized trial to determine if thalidomide can improve the efficacy of a gonadotropin-releasing hormone agonist in hormone responsive patients with an increasing prostate specific antigen after primary definitive therapy for prostate cancer. Thalidomide 112-123 gonadotropin releasing hormone 1 Homo sapiens 154-184 19167733-2 2009 MATERIALS AND METHODS: A total of 159 patients were enrolled in a double-blind randomized trial to determine if thalidomide can improve the efficacy of a gonadotropin-releasing hormone agonist in hormone responsive patients with an increasing prostate specific antigen after primary definitive therapy for prostate cancer. Thalidomide 112-123 kallikrein related peptidase 3 Homo sapiens 243-268 19167733-7 2009 RESULTS: During oral phase A the median time to prostate specific antigen progression was 15 months for the thalidomide group compared to 9.6 months on placebo (p = 0.21). Thalidomide 108-119 kallikrein related peptidase 3 Homo sapiens 48-73 19167733-8 2009 The median time to prostate specific antigen progression during oral phase B for the thalidomide group was 17.1 vs 6.6 months on placebo (p = 0.0002). Thalidomide 85-96 kallikrein related peptidase 3 Homo sapiens 19-44 19201823-6 2009 Thalidomide significantly inhibited the TNF-alpha production in response to palmitoyl-Cys(RS)-2,3-di(palmitoyloxy) propyl)-Ala-Gly-OH (Pam(3)Cys) as a MyD88-dependent TLR2 ligand. Thalidomide 0-11 toll-like receptor 2 Mus musculus 167-171 19201823-7 2009 Therefore, it is suggested that thalidomide might impair LPS signalling via down-regulation of MyD88 protein and mRNA and inhibit LPS-induced TNF-alpha production. Thalidomide 32-43 myeloid differentiation primary response gene 88 Mus musculus 95-100 19201823-7 2009 Therefore, it is suggested that thalidomide might impair LPS signalling via down-regulation of MyD88 protein and mRNA and inhibit LPS-induced TNF-alpha production. Thalidomide 32-43 tumor necrosis factor Mus musculus 142-151 19201823-8 2009 The putative mechanism of thalidomide-induced MyD88 down-regulation is discussed. Thalidomide 26-37 myeloid differentiation primary response gene 88 Mus musculus 46-51 19236757-0 2009 [Antiangiogenic activity of thalidomide in vitro mediated by cytochrome CYP2C19]. Thalidomide 28-39 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 72-79 19236757-7 2009 When omeprazole, a specific inhibitor of cytochrome CYP2C19, was added into the co-incubation mixture, the effects of thalidomide on cell proliferation ability, apoptosis, migration and tube formation decreased. Thalidomide 118-129 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 52-59 19236757-8 2009 It is concluded that effect of human hepatic cell microsome is required for thalidomide"s antiangiogenic activity in vitro and cytochrome CYP2C19 may be involved in the antiangiogenic effect of thalidomide. Thalidomide 194-205 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 138-145 19201823-0 2009 Thalidomide inhibits lipopolysaccharide-induced tumor necrosis factor-alpha production via down-regulation of MyD88 expression. Thalidomide 0-11 myeloid differentiation primary response gene 88 Mus musculus 110-115 19201823-1 2009 The effect of thalidomide on lipopolysaccharide (LPS)-induced tumor necrosis factor (TNF)-alpha production was studied by using RAW 264.7 murine macrophage-like cells. Thalidomide 14-25 tumor necrosis factor Mus musculus 62-95 19201823-2 2009 Thalidomide significantly inhibited LPS-induced TNF-alpha production. Thalidomide 0-11 tumor necrosis factor Mus musculus 48-57 19201823-3 2009 Thalidomide prevented the activation of nuclear factor (NF)-KB by down-regulating phosphorylation of inhibitory KB factor (IKB), and IKB kinase (IKK)-alpha and IKK-beta Moreover, thalidomide inhibited LPS-induced phosphorylation of AKT, p38 and stress-activated protein kinase (SAPK)/JNK. Thalidomide 0-11 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, beta Mus musculus 101-121 19201823-3 2009 Thalidomide prevented the activation of nuclear factor (NF)-KB by down-regulating phosphorylation of inhibitory KB factor (IKB), and IKB kinase (IKK)-alpha and IKK-beta Moreover, thalidomide inhibited LPS-induced phosphorylation of AKT, p38 and stress-activated protein kinase (SAPK)/JNK. Thalidomide 0-11 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, beta Mus musculus 123-126 19201823-3 2009 Thalidomide prevented the activation of nuclear factor (NF)-KB by down-regulating phosphorylation of inhibitory KB factor (IKB), and IKB kinase (IKK)-alpha and IKK-beta Moreover, thalidomide inhibited LPS-induced phosphorylation of AKT, p38 and stress-activated protein kinase (SAPK)/JNK. Thalidomide 0-11 conserved helix-loop-helix ubiquitous kinase Mus musculus 133-155 19201823-3 2009 Thalidomide prevented the activation of nuclear factor (NF)-KB by down-regulating phosphorylation of inhibitory KB factor (IKB), and IKB kinase (IKK)-alpha and IKK-beta Moreover, thalidomide inhibited LPS-induced phosphorylation of AKT, p38 and stress-activated protein kinase (SAPK)/JNK. Thalidomide 0-11 inhibitor of kappaB kinase beta Mus musculus 160-168 19201823-3 2009 Thalidomide prevented the activation of nuclear factor (NF)-KB by down-regulating phosphorylation of inhibitory KB factor (IKB), and IKB kinase (IKK)-alpha and IKK-beta Moreover, thalidomide inhibited LPS-induced phosphorylation of AKT, p38 and stress-activated protein kinase (SAPK)/JNK. Thalidomide 0-11 mitogen-activated protein kinase 14 Mus musculus 237-240 19201823-3 2009 Thalidomide prevented the activation of nuclear factor (NF)-KB by down-regulating phosphorylation of inhibitory KB factor (IKB), and IKB kinase (IKK)-alpha and IKK-beta Moreover, thalidomide inhibited LPS-induced phosphorylation of AKT, p38 and stress-activated protein kinase (SAPK)/JNK. Thalidomide 0-11 mitogen-activated protein kinase 8 Mus musculus 284-287 19201823-3 2009 Thalidomide prevented the activation of nuclear factor (NF)-KB by down-regulating phosphorylation of inhibitory KB factor (IKB), and IKB kinase (IKK)-alpha and IKK-beta Moreover, thalidomide inhibited LPS-induced phosphorylation of AKT, p38 and stress-activated protein kinase (SAPK)/JNK. Thalidomide 179-190 conserved helix-loop-helix ubiquitous kinase Mus musculus 133-155 19201823-4 2009 The expression of myeloid differentiation factor 88 (MyD88) protein and mRNA was markedly reduced in thalidomide-treated RAW 264.7 cells but there was no significant alteration in the expression of interleukin-1 receptor-associated kinase (IRAK) 1 and TNF receptor-associated factor (TRAF) 6 in the cells. Thalidomide 101-112 myeloid differentiation primary response gene 88 Mus musculus 18-51 19201823-4 2009 The expression of myeloid differentiation factor 88 (MyD88) protein and mRNA was markedly reduced in thalidomide-treated RAW 264.7 cells but there was no significant alteration in the expression of interleukin-1 receptor-associated kinase (IRAK) 1 and TNF receptor-associated factor (TRAF) 6 in the cells. Thalidomide 101-112 myeloid differentiation primary response gene 88 Mus musculus 53-58 19201823-4 2009 The expression of myeloid differentiation factor 88 (MyD88) protein and mRNA was markedly reduced in thalidomide-treated RAW 264.7 cells but there was no significant alteration in the expression of interleukin-1 receptor-associated kinase (IRAK) 1 and TNF receptor-associated factor (TRAF) 6 in the cells. Thalidomide 101-112 TNF receptor-associated factor 6 Mus musculus 252-291 19201823-6 2009 Thalidomide significantly inhibited the TNF-alpha production in response to palmitoyl-Cys(RS)-2,3-di(palmitoyloxy) propyl)-Ala-Gly-OH (Pam(3)Cys) as a MyD88-dependent TLR2 ligand. Thalidomide 0-11 tumor necrosis factor Mus musculus 40-49 19201823-6 2009 Thalidomide significantly inhibited the TNF-alpha production in response to palmitoyl-Cys(RS)-2,3-di(palmitoyloxy) propyl)-Ala-Gly-OH (Pam(3)Cys) as a MyD88-dependent TLR2 ligand. Thalidomide 0-11 myeloid differentiation primary response gene 88 Mus musculus 151-156 18687985-6 2009 Studies suggest that DKK1 activation in osteoblasts is the underlying cause of glucocorticoid- and estrogen deficiency-mediated osteoporosis, and at least partially underlies the teratogenic effects of thalidomide on limb development. Thalidomide 202-213 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 21-25 19098937-10 2009 CONCLUSION: A novel sugar-substituted thalidomide derivative, STA-35, is potent toward HL-60 cells in vitro and induces apoptosis by the suppression of NF-kappaB activation. Thalidomide 38-49 nuclear factor kappa B subunit 1 Homo sapiens 152-161 19067746-3 2009 In this study, the effect of different treatment regimens for MM on serum DKK-1 was evaluated and correlated with the response to treatment in 101 myeloma patients receiving bortezomib, thalidomide, lenalidomide, adriamycin and dexamethasone (AD) or high-dose chemotherapy (HDCT) followed by autologous stem cell transplantation (ASCT). Thalidomide 186-197 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 74-79 19754423-5 2009 Typical CYP1A2 substrates generally contain planar ring that can fit the narrow and planar active site of the enzyme, such as propranolol, clozapine, guanabenz, flutamide, imatinib, thalidomide, carbamazepine, lidocaine, theophylline, tacrine, tizanidine, zolpidem, riluzole, zileuton, and leflunomide. Thalidomide 182-193 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 8-14 18948377-0 2009 Drug interactions of thalidomide with midazolam and cyclosporine A: heterotropic cooperativity of human cytochrome P450 3A5. Thalidomide 21-32 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 104-123 18948377-8 2009 Enhanced midazolam hydroxylation by thalidomide was also seen in liver microsomal samples harboring the CYP3A5*1 allele. Thalidomide 36-47 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 104-110 19689289-10 2009 Moreover, we examine recent reports providing important insights into the understanding of efficacy of thalidomide and its analogs, as TNF-alpha activity inhibitories, especially in therapies of several inflammatory diseases within the nervous system. Thalidomide 103-114 tumor necrosis factor Homo sapiens 135-144 19067746-7 2009 Thalidomide led to a non-significant decrease in DKK-1 (1705 pg/mL vs. 1269 pg/mL, P = 0.081). Thalidomide 0-11 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 49-54 18691254-0 2008 Survival and outcome of blastoid variant myeloma following treatment with the novel thalidomide containing regime DT-PACE. Thalidomide 84-95 furin, paired basic amino acid cleaving enzyme Homo sapiens 117-121 18957334-7 2009 In addition, the systemic pretreatment with the preferential inhibitor of TNFalpha synthesis thalidomide (25 and 50 mg/kg, p.o) promoted a marked reduction of the first and second phases of formalin-evoked nociception. Thalidomide 93-104 tumor necrosis factor Rattus norvegicus 74-82 18621563-5 2009 The immunomodulatory effects of thalidomide are at least partially mediated through its ability to down-regulate the pathogenic over-production of tumor necrosis factor-alpha (TNF-alpha). Thalidomide 32-43 tumor necrosis factor Homo sapiens 147-174 18621563-5 2009 The immunomodulatory effects of thalidomide are at least partially mediated through its ability to down-regulate the pathogenic over-production of tumor necrosis factor-alpha (TNF-alpha). Thalidomide 32-43 tumor necrosis factor Homo sapiens 176-185 18621563-8 2009 Thalidomide has been shown to down-regulate the production of TNF-alpha. Thalidomide 0-11 tumor necrosis factor Homo sapiens 62-71 18367123-1 2009 OBJECTIVE: To assess the efficacy of granulocyte macrophage colony-stimulating factor (GM-CSF) in combination with thalidomide on prostate-specific antigen (PSA) reduction in hormone-naive prostate carcinoma (HNPC) patients with rising PSA levels after definitive local treatment. Thalidomide 115-126 kallikrein related peptidase 3 Homo sapiens 130-161 18367123-1 2009 OBJECTIVE: To assess the efficacy of granulocyte macrophage colony-stimulating factor (GM-CSF) in combination with thalidomide on prostate-specific antigen (PSA) reduction in hormone-naive prostate carcinoma (HNPC) patients with rising PSA levels after definitive local treatment. Thalidomide 115-126 kallikrein related peptidase 3 Homo sapiens 157-160 19152168-0 2009 Thalidomide inhibits leukemia cell invasion and migration by upregulation of early growth response gene 1. Thalidomide 0-11 early growth response 1 Homo sapiens 77-105 19152168-2 2009 In this study, 10 microM thalidomide treatment markedly increased the expression of the early growth response gene 1 (Egr-1) at both mRNA and protein levels in HL-60 leukemic cells. Thalidomide 25-36 early growth response 1 Homo sapiens 88-116 19152168-2 2009 In this study, 10 microM thalidomide treatment markedly increased the expression of the early growth response gene 1 (Egr-1) at both mRNA and protein levels in HL-60 leukemic cells. Thalidomide 25-36 early growth response 1 Homo sapiens 118-123 19152168-5 2009 Our data indicated thalidomide could suppress leukemia cell invasion and migration by upregulation of Egr-1, suggesting a novel mechanism of thalidomide in the treatment of leukemia. Thalidomide 19-30 early growth response 1 Homo sapiens 102-107 19152168-5 2009 Our data indicated thalidomide could suppress leukemia cell invasion and migration by upregulation of Egr-1, suggesting a novel mechanism of thalidomide in the treatment of leukemia. Thalidomide 141-152 early growth response 1 Homo sapiens 102-107 19152168-6 2009 Further investigations are needed to explore the detailed mechanism of Egr-1 induction by thalidomide and the downstream pathways involved in the regulation of leukemia cell invasion. Thalidomide 90-101 early growth response 1 Homo sapiens 71-76 18799726-5 2008 Among lenalidomide + dexamethasone-treated patients, ORR was higher in thalidomide-naive versus thalidomide-exposed patients (P = .04), with longer median TTP (P = .04) and PFS (P = .02). Thalidomide 71-82 ZFP36 ring finger protein Homo sapiens 155-158 18799726-11 2008 Although prior thalidomide may have contributed to inferior TTP and PFS compared with thalidomide-naive patients, these parameters remained superior compared with placebo + dexamethasone; similar benefits compared with placebo + dexamethasone were not evident for thalidomide-exposed patients in terms of overall survival. Thalidomide 15-26 ZFP36 ring finger protein Homo sapiens 60-63 18436450-6 2008 A promising approach is the pharmacological inhibition of tumor angiogenesis with anti-vascular endothelial growth factor (VEGF) agents, such as bevacizumab, cyclooxygenase-2 inhibitors (celecoxib), thalidomide and others. Thalidomide 199-210 vascular endothelial growth factor A Homo sapiens 82-121 18436450-6 2008 A promising approach is the pharmacological inhibition of tumor angiogenesis with anti-vascular endothelial growth factor (VEGF) agents, such as bevacizumab, cyclooxygenase-2 inhibitors (celecoxib), thalidomide and others. Thalidomide 199-210 vascular endothelial growth factor A Homo sapiens 123-127 19052974-8 2008 The number of blood mDC1 in patients taking thalidomide was also significantly higher than at relapse. Thalidomide 44-55 mediator of DNA damage checkpoint 1 Mus musculus 20-24 18983651-9 2008 CONCLUSION: Thalidomide inhibits vasculogenic mimicry channel and mosaic vessels formation in melanoma through the regulation of vasculogenic factors, and it can induce necrosis of melanoma cells, which may be related with the NF-kappaB signaling pathway. Thalidomide 12-23 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 227-236 19099634-0 2008 [Efficacy of different thalidomide regimens for patients with multiple myeloma and its relationship with TNF-alpha level]. Thalidomide 23-34 tumor necrosis factor Homo sapiens 105-114 19099634-1 2008 The study was aimed to investigate the clinical efficacy and adverse reactions of different thalidomide regimens in the treatment of multiple myeloma (MM), and to explore the relationship between efficacy of thalidomide and serum level of TNF-alpha in MM patients. Thalidomide 208-219 tumor necrosis factor Homo sapiens 239-248 19099634-6 2008 At the same time, serum levels of TNF-alpha in 30 cases of MM treated with thalidomide (15 cases effective and 15 cases ineffective) before and after treatment were detected by double-antibody sandwich enzyme-linked immunosorbent assay (ELISA) and were compared with the clinical efficacy. Thalidomide 75-86 tumor necrosis factor Homo sapiens 34-43 19099634-10 2008 At the same time, it was found that the serum levels of TNF-alpha in ineffective group treated with thalidomide were 44.7+/-5.7 pg/ml and 46.3+/-4.0 pg/ml before and after thalidomide treatment, and there was no significant difference (p>0.05). Thalidomide 100-111 tumor necrosis factor Homo sapiens 56-65 19099634-10 2008 At the same time, it was found that the serum levels of TNF-alpha in ineffective group treated with thalidomide were 44.7+/-5.7 pg/ml and 46.3+/-4.0 pg/ml before and after thalidomide treatment, and there was no significant difference (p>0.05). Thalidomide 172-183 tumor necrosis factor Homo sapiens 56-65 19099634-14 2008 Serum level of TNF-alpha is an indicator for finding effects of thalidomide, and plays a role in the pathogenesis of MM. Thalidomide 64-75 tumor necrosis factor Homo sapiens 15-24 18662673-2 2008 Thalidomide inhibited EGF-induced cell growth in mouse and human monocytic leukemia cells, RAW 264.7, U937 and THP-1. Thalidomide 0-11 GLI family zinc finger 2 Homo sapiens 111-116 18469028-0 2008 Thalidomide reduces serum VEGF levels and improves peripheral neuropathy in POEMS syndrome. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 26-30 18662673-3 2008 Thalidomide inhibited EGF-induced phosphorylation of extracellular signal regulated kinase (ERK) 1/2, but not p38 and stress-activated protein kinase (SAPK)/JNK. Thalidomide 0-11 mitogen-activated protein kinase 1 Homo sapiens 53-100 18662673-4 2008 The phosphorylation of MEK1/2 and Raf at Ser 338 as the upstream molecules of ERK 1/2 was also prevented by thalidomide. Thalidomide 108-119 mitogen-activated protein kinase kinase 1 Homo sapiens 23-29 18662673-4 2008 The phosphorylation of MEK1/2 and Raf at Ser 338 as the upstream molecules of ERK 1/2 was also prevented by thalidomide. Thalidomide 108-119 zinc fingers and homeoboxes 2 Homo sapiens 34-37 18662673-4 2008 The phosphorylation of MEK1/2 and Raf at Ser 338 as the upstream molecules of ERK 1/2 was also prevented by thalidomide. Thalidomide 108-119 mitogen-activated protein kinase 3 Homo sapiens 78-85 18662673-6 2008 Thalidomide inhibited the Ras activation induced by lipopolysaccharide (LPS) and vascular endothelial growth factor (VEGF) as well as EGF. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 81-115 18662673-6 2008 Thalidomide inhibited the Ras activation induced by lipopolysaccharide (LPS) and vascular endothelial growth factor (VEGF) as well as EGF. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 117-121 18197140-2 2008 We tested the neuroprotective effects of thalidomide, an immunomodulatory agent that inhibits TNF-alpha production, which have not been investigated so far. Thalidomide 41-52 tumor necrosis factor Mus musculus 94-103 18854284-0 2008 Serum C-reactive protein at diagnosis and response to therapy is the most powerful factor predicting outcome of multiple myeloma treated with thalidomide/ anthracycline-based therapy. Thalidomide 142-153 C-reactive protein Homo sapiens 6-24 18854284-9 2008 CONCLUSION: Serum C-reactive protein before therapy and response after therapy are the only factors useful in identifying patients benefiting from anthracycline/thalidomide-based therapy. Thalidomide 161-172 C-reactive protein Homo sapiens 18-36 18701263-15 2008 TNFalpha levels in both plasma and brain were reduced following thalidomide treatment (p<0.002) in arthritis group. Thalidomide 64-75 tumor necrosis factor Rattus norvegicus 0-8 18701263-19 2008 Thalidomide was also effective in reducing TNFalpha levels thus contributing to its antiepileptic activity. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 43-51 19176055-0 2008 [Effect of cytochrome CYP2C19 on the anti-myeloma activity of thalidomide in vitro]. Thalidomide 62-73 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 22-29 18389516-8 2008 Our structural development studies on thalidomide are reviewed focusing on recent development of tubulin polymerization inhibitors, alpha-glucosidase inhibitors, and nuclear liver X receptors antagonists. Thalidomide 38-49 sucrase-isomaltase Homo sapiens 132-149 18787470-13 2008 The serum concentrations of vascular endothelial growth factor and endostatin increased significantly (P=0.039 and P=0.024, respectively) after 3 months of thalidomide treatment. Thalidomide 156-167 vascular endothelial growth factor A Homo sapiens 28-62 18787470-13 2008 The serum concentrations of vascular endothelial growth factor and endostatin increased significantly (P=0.039 and P=0.024, respectively) after 3 months of thalidomide treatment. Thalidomide 156-167 collagen type XVIII alpha 1 chain Homo sapiens 67-77 18723357-2 2008 We designed and prepared a new C(60) fullerene hybrid bearing thalidomide as a potential double-action anti-inflammatory agent, capable of simultaneous inhibition of LPS-induced NO and TNF-alpha production. Thalidomide 62-73 tumor necrosis factor Homo sapiens 185-194 18662682-4 2008 The animals, which received thalidomide alone orally or in combination with augmentin, 30 min prior to bacterial instillation into the lungs via intranasal route, showed significant (P<0.05) decrease in neutrophil influx into the lungs and there was significant (P<0.05) decrease in the production of malondialdehyde, nitric oxide and myeloperoxidase activity. Thalidomide 28-39 myeloperoxidase Mus musculus 341-356 18670095-0 2008 Effect of thalidomide on endotoxin-induced decreases in activity and expression of hepatic cytochrome P450 3A2. Thalidomide 10-21 cytochrome P450, family 3, subfamily a, polypeptide 2 Rattus norvegicus 91-110 18720539-8 2008 Expression of TM was significantly lower in the endothelial cells (EC) of the radiation group than in the control and thalidomide groups (P<0.001). Thalidomide 118-129 thrombomodulin Rattus norvegicus 14-16 18720539-10 2008 The intensity of VEGF expression in capillaries was greater in the radiation group than in the control group and was also attenuated by thalidomide (P=0.003). Thalidomide 136-147 vascular endothelial growth factor A Rattus norvegicus 17-21 18670095-1 2008 Thalidomide has been reported to inhibit the production of tumor necrosis factor-alpha (TNF-alpha) and nitric oxide (NO) that are involved in the down-regulation of hepatic cytochrome P450 (CYP) induced by endotoxin. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 59-86 18670095-1 2008 Thalidomide has been reported to inhibit the production of tumor necrosis factor-alpha (TNF-alpha) and nitric oxide (NO) that are involved in the down-regulation of hepatic cytochrome P450 (CYP) induced by endotoxin. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 88-97 18670095-1 2008 Thalidomide has been reported to inhibit the production of tumor necrosis factor-alpha (TNF-alpha) and nitric oxide (NO) that are involved in the down-regulation of hepatic cytochrome P450 (CYP) induced by endotoxin. Thalidomide 0-11 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 173-188 18670095-1 2008 Thalidomide has been reported to inhibit the production of tumor necrosis factor-alpha (TNF-alpha) and nitric oxide (NO) that are involved in the down-regulation of hepatic cytochrome P450 (CYP) induced by endotoxin. Thalidomide 0-11 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 190-193 18670095-2 2008 In the present study, we investigated the effects of thalidomide on endotoxin-induced decreases in the activity and expression of hepatic CYP3A2 in rats. Thalidomide 53-64 cytochrome P450, family 3, subfamily a, polypeptide 2 Rattus norvegicus 138-144 18670095-7 2008 Western blot analysis revealed that thalidomide had no effect on the protein levels of hepatic CYP3A2, whereas it enhanced the down-regulation of hepatic CYP3A2 by endotoxin. Thalidomide 36-47 cytochrome P450, family 3, subfamily a, polypeptide 2 Rattus norvegicus 154-160 18670095-9 2008 The present findings suggest that thalidomide enhances endotoxin-induced decreases in the activity and expression of hepatic CYP3A2. Thalidomide 34-45 cytochrome P450, family 3, subfamily a, polypeptide 2 Rattus norvegicus 125-131 18525301-3 2008 This study assessed whether GM-CSF would improve the response rate of MRCC patients to the thalidomide plus IL-2 regimen. Thalidomide 91-102 colony stimulating factor 2 Homo sapiens 28-34 18687660-0 2008 The G-rich promoter and G-rich coding sequence of basic fibroblast growth factor are the targets of thalidomide in glioma. Thalidomide 100-111 fibroblast growth factor 2 Homo sapiens 50-80 18687660-4 2008 Using U-87 MG cell lines, we found that thalidomide, especially when encapsulated in a liposome, down-regulated the transcription and translation of the FGF-2 gene by interacting with G-rich regions present in the promoter and the internal ribosome entry site of its transcript at concentrations much lower than therapeutic serum concentrations. Thalidomide 40-51 fibroblast growth factor 2 Homo sapiens 153-158 18687660-5 2008 Thalidomide treatment also dramatically suppressed the anchorage-independent growth of U-87 MG and other glioma cells by over a thousand fold without affecting its anchorage-dependent growth, which may be accomplished by knocking down endogenous bFGF expression in these cells. Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 246-250 18687660-7 2008 Our data suggest that by targeting the G-rich regions of bFGF, thalidomide (at 0.1 microg/mL) can reduce cellular bFGF levels and affect tumor anchorage-independent growth, the hallmark of tumorigenicity. Thalidomide 63-74 fibroblast growth factor 2 Homo sapiens 57-61 18687660-7 2008 Our data suggest that by targeting the G-rich regions of bFGF, thalidomide (at 0.1 microg/mL) can reduce cellular bFGF levels and affect tumor anchorage-independent growth, the hallmark of tumorigenicity. Thalidomide 63-74 fibroblast growth factor 2 Homo sapiens 114-118 18381653-6 2008 Furthermore thalidomide, which prevents TNFalpha production, was without effect on hypoxic suppression of EAAT-1 but prevented hypoxic suppression of EAAT-2. Thalidomide 12-23 tumor necrosis factor Homo sapiens 40-48 18381653-6 2008 Furthermore thalidomide, which prevents TNFalpha production, was without effect on hypoxic suppression of EAAT-1 but prevented hypoxic suppression of EAAT-2. Thalidomide 12-23 solute carrier family 1 member 2 Homo sapiens 150-156 18671617-9 2008 Thalidomide may be an ideal therapy for life-threatening hemangiomas because it inhibits new blood vessel formation by antagonizing both the bFGF and VEGF pathways and has a more acceptable toxicity profile than other agents. Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 141-145 18671617-9 2008 Thalidomide may be an ideal therapy for life-threatening hemangiomas because it inhibits new blood vessel formation by antagonizing both the bFGF and VEGF pathways and has a more acceptable toxicity profile than other agents. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 150-154 18598295-10 2008 Treatment with thalidomide restored malondialdehyde as well as reduction of myeloperoxidase and tumour necrosis factor-alpha towards normal levels. Thalidomide 15-26 myeloperoxidase Rattus norvegicus 76-91 18536579-5 2008 Moreover, several drugs that are effective against multiple myeloma, including bortezomib, thalidomide, lenalidomide and arsenic trioxide, have been found to block activation of NF-kappaB. Thalidomide 91-102 nuclear factor kappa B subunit 1 Homo sapiens 178-187 18549624-8 2008 The population of CD3(+) T, CD4(+) T, CD8(+) T, NK and CD4(+)CD25(+) Treg significantly increased in patients with positive response to thalidomide, but the population of CD8(+) T remained unchanged. Thalidomide 136-147 CD4 molecule Homo sapiens 55-58 18343940-7 2008 Thalidomide (200 mg/Kg) was administered daily intraperitoneally to control and ROP mice in two protocols: (1) from P12 to P16, and (2) from P11 to P15 . Thalidomide 0-11 polymerase (DNA-directed), delta 4 Mus musculus 116-119 18343940-7 2008 Thalidomide (200 mg/Kg) was administered daily intraperitoneally to control and ROP mice in two protocols: (1) from P12 to P16, and (2) from P11 to P15 . Thalidomide 0-11 cyclin dependent kinase inhibitor 2A Mus musculus 123-126 18343940-7 2008 Thalidomide (200 mg/Kg) was administered daily intraperitoneally to control and ROP mice in two protocols: (1) from P12 to P16, and (2) from P11 to P15 . Thalidomide 0-11 S100 calcium binding protein A10 (calpactin) Mus musculus 141-144 18343940-7 2008 Thalidomide (200 mg/Kg) was administered daily intraperitoneally to control and ROP mice in two protocols: (1) from P12 to P16, and (2) from P11 to P15 . Thalidomide 0-11 cyclin dependent kinase inhibitor 2B Mus musculus 148-151 18549624-0 2008 [Effects of thalidomide on CD4(+)CD25(+) T regulatory cells in patients with multiple myeloma]. Thalidomide 12-23 CD4 molecule Homo sapiens 27-30 18549624-1 2008 The study was purposed to explore the changes of CD4(+)CD25(+) T regulatory cells in patients with multiple myeloma before and (MM) after treatment with thalidomide so as to provide evidences for effective immunotherapy. Thalidomide 153-164 CD4 molecule Homo sapiens 49-52 18549624-9 2008 It is concluded that the significant increase of CD4(+)CD25(+) regulatory T cells in peripheral blood of patients with MM is concerned with the MM pathogenesis; thalidomide may exert its anti-MM effects by down-regulating CD4(+)CD25(+) Treg. Thalidomide 161-172 CD4 molecule Homo sapiens 49-52 18549624-6 2008 The population of CD4(+)CD25(+ high) Treg cells in patients with response to thalidomide was significantly decreased (p < 0.01), but the population of these cells in patients without response not changed significantly (p > 0.05), as compared with patients before treatment. Thalidomide 77-88 CD4 molecule Homo sapiens 18-21 18549624-8 2008 The population of CD3(+) T, CD4(+) T, CD8(+) T, NK and CD4(+)CD25(+) Treg significantly increased in patients with positive response to thalidomide, but the population of CD8(+) T remained unchanged. Thalidomide 136-147 CD4 molecule Homo sapiens 28-31 18549624-8 2008 The population of CD3(+) T, CD4(+) T, CD8(+) T, NK and CD4(+)CD25(+) Treg significantly increased in patients with positive response to thalidomide, but the population of CD8(+) T remained unchanged. Thalidomide 136-147 CD8a molecule Homo sapiens 38-41 18549624-9 2008 It is concluded that the significant increase of CD4(+)CD25(+) regulatory T cells in peripheral blood of patients with MM is concerned with the MM pathogenesis; thalidomide may exert its anti-MM effects by down-regulating CD4(+)CD25(+) Treg. Thalidomide 161-172 CD4 molecule Homo sapiens 222-225 18325608-0 2008 Restraining tumor necrosis factor-alpha by thalidomide prevents the amyloid beta-induced impairment of recognition memory in mice. Thalidomide 43-54 tumor necrosis factor Mus musculus 12-39 19040069-1 2008 OBJECTIVE: To evaluate the effects of four agents including cyclophosphamide, thalidomide, total glucosides of peony (TGP) and simvastatin on cell-proliferation and endothelin-1 secretion of human endothelial cells (ECs). Thalidomide 78-89 endothelin 1 Homo sapiens 165-177 18325608-8 2008 Daily treatment with thalidomide (20 mg/kg), a preferential degrader of TNF-alpha mRNA, or i.c.v.-injection of an anti-TNF-alpha antibody (10 etag/mouse) prevented the nitration of proteins in the hippocampus and the impairment of recognition memory induced by Abeta(25-35) or Abeta(1-40) in mice. Thalidomide 21-32 tumor necrosis factor Mus musculus 72-81 18325608-8 2008 Daily treatment with thalidomide (20 mg/kg), a preferential degrader of TNF-alpha mRNA, or i.c.v.-injection of an anti-TNF-alpha antibody (10 etag/mouse) prevented the nitration of proteins in the hippocampus and the impairment of recognition memory induced by Abeta(25-35) or Abeta(1-40) in mice. Thalidomide 21-32 amyloid beta (A4) precursor protein Mus musculus 261-266 18953944-1 2008 OBJECTIVE: To investigate the release and intracellular localization of high mobility group box chromosomal protein 1 (HMGB1) in the peripheral blood monocytes of rheumatoid arthritis (RA) patients and the inhibitive effect of thalidomide. Thalidomide 227-238 structure specific recognition protein 1 Homo sapiens 72-95 18953944-10 2008 Thalidomide (40 microg/ml) could inhibit the release of HMGB1 in the monocytes from RA patients stimulated with TNFalpha (P < 0.05). Thalidomide 0-11 high mobility group box 1 Homo sapiens 56-61 18953944-10 2008 Thalidomide (40 microg/ml) could inhibit the release of HMGB1 in the monocytes from RA patients stimulated with TNFalpha (P < 0.05). Thalidomide 0-11 tumor necrosis factor Homo sapiens 112-120 18418038-5 2008 We extend this model by presenting new data demonstrating an involvement of the transcription factors Tbx5 and Sall4 in thalidomide-induced molecular pathology. Thalidomide 120-131 T-box transcription factor 5 Homo sapiens 102-106 18418038-5 2008 We extend this model by presenting new data demonstrating an involvement of the transcription factors Tbx5 and Sall4 in thalidomide-induced molecular pathology. Thalidomide 120-131 spalt like transcription factor 4 Homo sapiens 111-116 18953944-12 2008 Treatment with TNFalpha (100 ng/ml) for 24 hours resulted in a cytoplasmic translocation of HMGB1, which was inhibited significantly by thalidomide. Thalidomide 136-147 tumor necrosis factor Homo sapiens 15-23 18953944-1 2008 OBJECTIVE: To investigate the release and intracellular localization of high mobility group box chromosomal protein 1 (HMGB1) in the peripheral blood monocytes of rheumatoid arthritis (RA) patients and the inhibitive effect of thalidomide. Thalidomide 227-238 high mobility group box 1 Homo sapiens 119-124 18953944-12 2008 Treatment with TNFalpha (100 ng/ml) for 24 hours resulted in a cytoplasmic translocation of HMGB1, which was inhibited significantly by thalidomide. Thalidomide 136-147 high mobility group box 1 Homo sapiens 92-97 17786465-1 2008 BACKGROUND: To evaluate the effects of thalidomide treatment on the temporal course of TNF-alpha, VEGF production and the histopathological changes in ischemia/reperfusion (I/R) injured guinea pigs retina. Thalidomide 39-50 tumor necrosis factor Cavia porcellus 87-96 18953944-13 2008 CONCLUSION: TNFalpha induces the release and cytoplasmic translocation of HMGB1 in the peripheral blood monocytes of RA patients and thalidomide inhibits the release and translocation of HMGB1. Thalidomide 133-144 high mobility group box 1 Homo sapiens 74-79 18953944-13 2008 CONCLUSION: TNFalpha induces the release and cytoplasmic translocation of HMGB1 in the peripheral blood monocytes of RA patients and thalidomide inhibits the release and translocation of HMGB1. Thalidomide 133-144 high mobility group box 1 Homo sapiens 187-192 18034203-7 2008 These results suggest that thalidomide tends to decrease TNF-alpha receptor levels, CD8/CD11b+ T cells and natural killer cells in early treatment and increases CD4+CD45RO+ memory T and gammadelta+ T cells later in BD. Thalidomide 27-38 CD8a molecule Homo sapiens 84-87 18034203-7 2008 These results suggest that thalidomide tends to decrease TNF-alpha receptor levels, CD8/CD11b+ T cells and natural killer cells in early treatment and increases CD4+CD45RO+ memory T and gammadelta+ T cells later in BD. Thalidomide 27-38 integrin subunit alpha M Homo sapiens 88-93 18034203-7 2008 These results suggest that thalidomide tends to decrease TNF-alpha receptor levels, CD8/CD11b+ T cells and natural killer cells in early treatment and increases CD4+CD45RO+ memory T and gammadelta+ T cells later in BD. Thalidomide 27-38 CD4 molecule Homo sapiens 161-164 18343126-2 2008 On this basis, we have already developed novel LXR antagonists based upon alpha-glucosidase inhibitors derived from thalidomide. Thalidomide 116-127 sucrase-isomaltase Homo sapiens 74-91 18070197-1 2008 OBJECTIVE: To investigate how thalidomide confers its survival benefit in prostate cancer, by assessing its effect on circulating endothelial cells (CECs) and progenitors (CEPs) in a combined therapy of thalidomide and chemotherapy drugs in a human prostate cancer xenograft model, as in clinical trials patients treated with both thalidomide and docetaxel had a >50% decrease in prostate-specific antigen (PSA) levels and longer median overall survival than those treated with docetaxel monotherapy. Thalidomide 30-41 kallikrein related peptidase 3 Homo sapiens 383-414 17473922-4 2008 Thalidomide is an anti-inflammatory agent that potentiates the anti-tumour activity of DMXAA but decreases induction of TNF in plasma. Thalidomide 0-11 tumor necrosis factor Mus musculus 120-123 17473922-17 2008 Co-administration of thalidomide increases the effective dose of DMXAA by reducing clearance but also, by inhibiting production of circulating TNF, reduces the host toxicity of DMXAA. Thalidomide 21-32 tumor necrosis factor Mus musculus 143-146 17786465-1 2008 BACKGROUND: To evaluate the effects of thalidomide treatment on the temporal course of TNF-alpha, VEGF production and the histopathological changes in ischemia/reperfusion (I/R) injured guinea pigs retina. Thalidomide 39-50 Vegfa Cavia porcellus 98-102 17786465-8 2008 The thalidomide/ischemia group retinal VEGF level was significantly lower versus the ischemia group. Thalidomide 4-15 Vegfa Cavia porcellus 39-43 17786465-10 2008 Also, the retinal TNF-alpha level was significantly lower in the thalidomide/ischemia group versus the ischemia group. Thalidomide 65-76 tumor necrosis factor Cavia porcellus 18-27 17786465-13 2008 CONCLUSION: Thalidomide treatment decreases PMNL infiltration, retinal edema, VEGF, and TNF-alpha synthesis following I/R injury to the guinea pig retina. Thalidomide 12-23 Vegfa Cavia porcellus 78-82 17786465-13 2008 CONCLUSION: Thalidomide treatment decreases PMNL infiltration, retinal edema, VEGF, and TNF-alpha synthesis following I/R injury to the guinea pig retina. Thalidomide 12-23 tumor necrosis factor Cavia porcellus 88-97 18291259-13 2008 The expression of mRNA for Fas, caspase-3, and Bax of cholangiocarcinoma in the thalidomide-treated rats was greater than for saline-treated rats. Thalidomide 80-91 caspase 3 Rattus norvegicus 32-41 18291259-13 2008 The expression of mRNA for Fas, caspase-3, and Bax of cholangiocarcinoma in the thalidomide-treated rats was greater than for saline-treated rats. Thalidomide 80-91 BCL2 associated X, apoptosis regulator Rattus norvegicus 47-50 18291259-15 2008 The expression of TNFalpha and TGFbeta1 of soleus muscles for thalidomide-treated rats was less than for saline-treated rats. Thalidomide 62-73 tumor necrosis factor Rattus norvegicus 18-26 18291259-15 2008 The expression of TNFalpha and TGFbeta1 of soleus muscles for thalidomide-treated rats was less than for saline-treated rats. Thalidomide 62-73 transforming growth factor, beta 1 Rattus norvegicus 31-39 18291259-16 2008 CONCLUSIONS: Using our rat cholangiocarcinoma model, we demonstrated that thalidomide inhibited tumor growth and was associated with a decrease in expression of reduced eIF4E and VEGF expression; in addition, thalidomide preserved fast-twitch skeletal muscle fibers and was associated with decreased expression of TNFalpha and TGFbeta1. Thalidomide 74-85 eukaryotic translation initiation factor 4E Rattus norvegicus 169-174 18291259-16 2008 CONCLUSIONS: Using our rat cholangiocarcinoma model, we demonstrated that thalidomide inhibited tumor growth and was associated with a decrease in expression of reduced eIF4E and VEGF expression; in addition, thalidomide preserved fast-twitch skeletal muscle fibers and was associated with decreased expression of TNFalpha and TGFbeta1. Thalidomide 74-85 vascular endothelial growth factor A Rattus norvegicus 179-183 18291259-16 2008 CONCLUSIONS: Using our rat cholangiocarcinoma model, we demonstrated that thalidomide inhibited tumor growth and was associated with a decrease in expression of reduced eIF4E and VEGF expression; in addition, thalidomide preserved fast-twitch skeletal muscle fibers and was associated with decreased expression of TNFalpha and TGFbeta1. Thalidomide 74-85 tumor necrosis factor Rattus norvegicus 314-322 18291259-16 2008 CONCLUSIONS: Using our rat cholangiocarcinoma model, we demonstrated that thalidomide inhibited tumor growth and was associated with a decrease in expression of reduced eIF4E and VEGF expression; in addition, thalidomide preserved fast-twitch skeletal muscle fibers and was associated with decreased expression of TNFalpha and TGFbeta1. Thalidomide 74-85 transforming growth factor, beta 1 Rattus norvegicus 327-335 17964176-0 2008 Thalidomide inhibition of perturbed vasculature and glial-derived tumor necrosis factor-alpha in an animal model of inflamed Alzheimer"s disease brain. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 66-93 17618052-0 2008 Thalidomide decreases the plasma levels of IL-1 and TNF following burn injury: is it a new drug for modulation of systemic inflammatory response. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 52-55 17618052-3 2008 Thalidomide has been shown to decrease the secretion of TNF from phagocytic cells, therefore suppression of TNF and IL-1 production from activated phagocytic cells might be a successful treatment modality for prevention of systemic inflammatory response following severe burn. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 56-59 17618052-3 2008 Thalidomide has been shown to decrease the secretion of TNF from phagocytic cells, therefore suppression of TNF and IL-1 production from activated phagocytic cells might be a successful treatment modality for prevention of systemic inflammatory response following severe burn. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 108-111 17618052-4 2008 To address this issue, we aimed to show whether thalidomide treatment decreased or suppressed plasma levels of TNF and IL-1 following burn in rats. Thalidomide 48-59 tumor necrosis factor Rattus norvegicus 111-114 17618052-7 2008 Thalidomide treatment decreased TNF and IL-1 significantly in both experimental groups at both the points (P<0.05). Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 32-35 17618052-8 2008 Although in this study we just showed inhibitory effect of thalidomide on plasma the level of TNF and IL-1, we speculate that thalidomide may have modulatory effect on the systemic inflammatory response after burn by decreasing plasma levels of TNF and IL-1. Thalidomide 59-70 tumor necrosis factor Rattus norvegicus 94-97 17618052-8 2008 Although in this study we just showed inhibitory effect of thalidomide on plasma the level of TNF and IL-1, we speculate that thalidomide may have modulatory effect on the systemic inflammatory response after burn by decreasing plasma levels of TNF and IL-1. Thalidomide 126-137 tumor necrosis factor Rattus norvegicus 245-248 17964176-4 2008 Thalidomide inhibition of the glial-derived proinflammatory/angiogenic factor TNF-alpha (tumor necrosis factor-alpha) in Abeta(1-42)-injected brain and also in vitro from peptide-activated human microglia could underly the changes in vascular processes. Thalidomide 0-11 tumor necrosis factor Homo sapiens 89-116 17964176-6 2008 Our findings suggest altered cerebral vasculature as an integral component of inflammatory responses with thalidomide an effective inhibitor of gliosis, vascular changes and TNF-alpha leading to neuroprotection in an animal model of inflamed AD brain. Thalidomide 106-117 tumor necrosis factor Homo sapiens 174-183 17964176-2 2008 We report the anti-angiogenic and anti-inflammatory compound, thalidomide, to significantly inhibit peptide-induced vascular changes including endothelial cell proliferation (marker rat endothelial cell antigen-1, RECA-1), angiogenic activity (marker laminin) and leakiness of blood-brain barrier (BBB, marker albumin). Thalidomide 62-73 albumin Rattus norvegicus 310-317 17964176-4 2008 Thalidomide inhibition of the glial-derived proinflammatory/angiogenic factor TNF-alpha (tumor necrosis factor-alpha) in Abeta(1-42)-injected brain and also in vitro from peptide-activated human microglia could underly the changes in vascular processes. Thalidomide 0-11 tumor necrosis factor Homo sapiens 78-87 18178729-0 2008 Thalidomide induces limb anomalies by PTEN stabilization, Akt suppression, and stimulation of caspase-dependent cell death. Thalidomide 0-11 phosphatase and tensin homolog Homo sapiens 38-42 18668392-0 2008 Thalidomide suppressed IL-1beta while enhancing TNF-alpha and IL-10, when cells in whole blood were stimulated with lipopolysaccharide. Thalidomide 0-11 interleukin 1 beta Homo sapiens 23-31 18668392-0 2008 Thalidomide suppressed IL-1beta while enhancing TNF-alpha and IL-10, when cells in whole blood were stimulated with lipopolysaccharide. Thalidomide 0-11 tumor necrosis factor Homo sapiens 48-57 18668392-0 2008 Thalidomide suppressed IL-1beta while enhancing TNF-alpha and IL-10, when cells in whole blood were stimulated with lipopolysaccharide. Thalidomide 0-11 interleukin 10 Homo sapiens 62-67 18668392-3 2008 Thalidomide"s ability to inhibit tumor necrosis factor alpha (TNF-alpha) in vitro has been proposed as a partial explanation of its effective treatment of ENL. Thalidomide 0-11 tumor necrosis factor Homo sapiens 33-60 18668392-3 2008 Thalidomide"s ability to inhibit tumor necrosis factor alpha (TNF-alpha) in vitro has been proposed as a partial explanation of its effective treatment of ENL. Thalidomide 0-11 tumor necrosis factor Homo sapiens 62-71 18668392-4 2008 In in vitro assays, thalidomide can enhance or suppress TNF-alpha. Thalidomide 20-31 tumor necrosis factor Homo sapiens 56-65 18668392-7 2008 Thalidomide suppressed interleukin 1 beta (IL-1beta) (p = 0.007), and it enhanced TNF-alpha (p = 0.007) and interleukin 10 (IL-10) (p = 0.031). Thalidomide 0-11 interleukin 1 beta Homo sapiens 23-41 18668392-7 2008 Thalidomide suppressed interleukin 1 beta (IL-1beta) (p = 0.007), and it enhanced TNF-alpha (p = 0.007) and interleukin 10 (IL-10) (p = 0.031). Thalidomide 0-11 interleukin 1 beta Homo sapiens 43-51 18668392-7 2008 Thalidomide suppressed interleukin 1 beta (IL-1beta) (p = 0.007), and it enhanced TNF-alpha (p = 0.007) and interleukin 10 (IL-10) (p = 0.031). Thalidomide 0-11 tumor necrosis factor Homo sapiens 82-91 18668392-7 2008 Thalidomide suppressed interleukin 1 beta (IL-1beta) (p = 0.007), and it enhanced TNF-alpha (p = 0.007) and interleukin 10 (IL-10) (p = 0.031). Thalidomide 0-11 interleukin 10 Homo sapiens 108-122 18668392-7 2008 Thalidomide suppressed interleukin 1 beta (IL-1beta) (p = 0.007), and it enhanced TNF-alpha (p = 0.007) and interleukin 10 (IL-10) (p = 0.031). Thalidomide 0-11 interleukin 10 Homo sapiens 124-129 17638016-8 2008 Treatment 1 h before IFS injection with etoricoxib, indomethacin, thalidomide, and pentoxifylline reduced COX-2 expression and some macroscopic and microscopic parameters in IFS-induced HC. Thalidomide 66-77 prostaglandin-endoperoxide synthase 2 Rattus norvegicus 106-111 18178729-0 2008 Thalidomide induces limb anomalies by PTEN stabilization, Akt suppression, and stimulation of caspase-dependent cell death. Thalidomide 0-11 AKT serine/threonine kinase 1 Homo sapiens 58-61 18178729-3 2008 Here we demonstrate in primary human embryonic cells and in the chicken embryo that thalidomide-induced signaling through bone morphogenetic proteins (Bmps) protects active PTEN from proteasomal degradation, resulting in suppression of Akt signaling. Thalidomide 84-95 phosphatase and tensin homolog Gallus gallus 173-177 18178729-3 2008 Here we demonstrate in primary human embryonic cells and in the chicken embryo that thalidomide-induced signaling through bone morphogenetic proteins (Bmps) protects active PTEN from proteasomal degradation, resulting in suppression of Akt signaling. Thalidomide 84-95 AKT serine/threonine kinase 1 Homo sapiens 236-239 18178729-5 2008 Most importantly, thalidomide-induced limb deformities and microphthalmia in chicken embryos could be rescued by a pharmacological PTEN inhibitor as well as by insulin, a stimulant of Akt signaling. Thalidomide 18-29 phosphatase and tensin homolog Gallus gallus 131-135 18178729-5 2008 Most importantly, thalidomide-induced limb deformities and microphthalmia in chicken embryos could be rescued by a pharmacological PTEN inhibitor as well as by insulin, a stimulant of Akt signaling. Thalidomide 18-29 insulin Gallus gallus 160-167 18178729-5 2008 Most importantly, thalidomide-induced limb deformities and microphthalmia in chicken embryos could be rescued by a pharmacological PTEN inhibitor as well as by insulin, a stimulant of Akt signaling. Thalidomide 18-29 AKT serine/threonine kinase 1 Homo sapiens 184-187 18178729-6 2008 We therefore conclude that perturbation of PTEN/Akt signaling and stimulation of caspase activity is central to the teratogenic effects of thalidomide. Thalidomide 139-150 phosphatase and tensin homolog Homo sapiens 43-47 18178729-6 2008 We therefore conclude that perturbation of PTEN/Akt signaling and stimulation of caspase activity is central to the teratogenic effects of thalidomide. Thalidomide 139-150 AKT serine/threonine kinase 1 Homo sapiens 48-51 18246603-3 2008 Recently, it has been shown that TNF-[alpha] may be a novel target for the treatment of acute pancreatitis; but the role of thalidomide, an immunomodulatory agent that inhibits TNF-(alpha) and angiogenesis, has not been investigated so far. Thalidomide 124-135 tumor necrosis factor Mus musculus 177-188 17851074-0 2007 Synthesis and TNF expression inhibitory properties of new thalidomide analogues derived via Heck cross coupling. Thalidomide 58-69 tumor necrosis factor Homo sapiens 14-17 17710103-8 2007 CONCLUSION: Thalidomide is an immunomodulatory agent that acts to inhibit production of tumor-necrosis factor-alpha (TNF-alpha), an important mediator in CNS inflammation, by enhancing TNF-alpha mRNA degradation. Thalidomide 12-23 tumor necrosis factor Homo sapiens 88-115 17710103-8 2007 CONCLUSION: Thalidomide is an immunomodulatory agent that acts to inhibit production of tumor-necrosis factor-alpha (TNF-alpha), an important mediator in CNS inflammation, by enhancing TNF-alpha mRNA degradation. Thalidomide 12-23 tumor necrosis factor Homo sapiens 117-126 17710103-8 2007 CONCLUSION: Thalidomide is an immunomodulatory agent that acts to inhibit production of tumor-necrosis factor-alpha (TNF-alpha), an important mediator in CNS inflammation, by enhancing TNF-alpha mRNA degradation. Thalidomide 12-23 tumor necrosis factor Homo sapiens 185-194 17916085-0 2007 Downregulation of neuropilin-1 in patients with acute myeloid leukemia treated with thalidomide. Thalidomide 84-95 neuropilin 1 Homo sapiens 18-30 17916085-5 2007 Under thalidomide treatment, a marked difference in the course of NRP-1 expression between responders and non-responders was observed, however, without a statistical significance (P = 0.071) being reached. Thalidomide 6-17 neuropilin 1 Homo sapiens 66-71 17916085-6 2007 Additionally, a significant correlation of the NRP-1 expression level to microvessel density could be detected under treatment with thalidomide (P = 0.018). Thalidomide 132-143 neuropilin 1 Homo sapiens 47-52 17916085-8 2007 Additionally, our results suggest that thalidomide-induced antileukemic properties might at least in part be mediated by NRP-1 downregulation. Thalidomide 39-50 neuropilin 1 Homo sapiens 121-126 17557286-8 2007 Furthermore, we could show that antiangiogenic effects of thalidomide are related to tumor-cell derived factors including vascular endothelial growth factor, basic fibroblast growth factor, hepatocyte growth factor and Il-8 some known and with, granulocyte colony stimulating growth factor and granulocyte macrophage colony stimulating growth factor, some new target molecules of thalidomide. Thalidomide 58-69 fibroblast growth factor 2 Mus musculus 158-188 17534579-6 2007 Cisplatin combined with thalidomide caused a significant decrease in vascular endothelial growth factor (VEGF) levels by 73% in intracranial tumors (P < 0.05) and by 50% in subcutaneous tumors (P < 0.05) and caused the level of active hepatic growth factor (a-HGF) to double in both the subcutaneous and intracranial tumors (P < 0.05), suggesting this treatment altered the vasculature in these tumors. Thalidomide 24-35 vascular endothelial growth factor A Rattus norvegicus 69-103 17534579-6 2007 Cisplatin combined with thalidomide caused a significant decrease in vascular endothelial growth factor (VEGF) levels by 73% in intracranial tumors (P < 0.05) and by 50% in subcutaneous tumors (P < 0.05) and caused the level of active hepatic growth factor (a-HGF) to double in both the subcutaneous and intracranial tumors (P < 0.05), suggesting this treatment altered the vasculature in these tumors. Thalidomide 24-35 vascular endothelial growth factor A Rattus norvegicus 105-109 18050580-5 2007 Thalidomide"s mechanism of action in RAUs still is not understood completely, but it appears to be mediated by inhibition of the pro-inflammatory cytokine tumor necrosis factor alpha (TNF-alpha). Thalidomide 0-11 tumor necrosis factor Homo sapiens 155-182 18050580-5 2007 Thalidomide"s mechanism of action in RAUs still is not understood completely, but it appears to be mediated by inhibition of the pro-inflammatory cytokine tumor necrosis factor alpha (TNF-alpha). Thalidomide 0-11 tumor necrosis factor Homo sapiens 184-193 17964516-0 2007 Thalidomide suppressed interleukin-6 but not tumor necrosis factor-alpha in volunteers with experimental endotoxemia. Thalidomide 0-11 interleukin 6 Homo sapiens 23-36 17964516-1 2007 An early rationale for using thalidomide to treat erythema nodosum leprosum had been based on some reports that it suppresses tumor necrosis factor-alpha (TNF-alpha). Thalidomide 29-40 tumor necrosis factor Homo sapiens 126-153 17964516-1 2007 An early rationale for using thalidomide to treat erythema nodosum leprosum had been based on some reports that it suppresses tumor necrosis factor-alpha (TNF-alpha). Thalidomide 29-40 tumor necrosis factor Homo sapiens 155-164 17964516-2 2007 However, in vivo and in vitro studies have yielded variable results, having shown that thalidomide can either enhance or suppress TNF-alpha. Thalidomide 87-98 tumor necrosis factor Homo sapiens 130-139 17964516-11 2007 Using the area under the dose response curve (AUC(0 to 24) h), thalidomide reduced the AUC for IL-6 by 56%, for IL-8 by 30%, and TNF-alpha by 32%. Thalidomide 63-74 interleukin 6 Homo sapiens 95-99 17964516-11 2007 Using the area under the dose response curve (AUC(0 to 24) h), thalidomide reduced the AUC for IL-6 by 56%, for IL-8 by 30%, and TNF-alpha by 32%. Thalidomide 63-74 C-X-C motif chemokine ligand 8 Homo sapiens 112-116 17964516-11 2007 Using the area under the dose response curve (AUC(0 to 24) h), thalidomide reduced the AUC for IL-6 by 56%, for IL-8 by 30%, and TNF-alpha by 32%. Thalidomide 63-74 tumor necrosis factor Homo sapiens 129-138 17964516-12 2007 In this model, thalidomide did not suppress TNF-alpha or IL-8, but it did suppress IL-6 at 4-h postinfusion with lipopolysaccharide (P=0.004), at 6 h (P=0.014), at 12 h (P=0.001), and at 16 h (P=0.012). Thalidomide 15-26 interleukin 6 Homo sapiens 83-87 17620452-0 2007 Thalidomide induces gamma-globin gene expression through increased reactive oxygen species-mediated p38 MAPK signaling and histone H4 acetylation in adult erythropoiesis. Thalidomide 0-11 mitogen-activated protein kinase 14 Homo sapiens 100-103 17620452-1 2007 Although thalidomide has been shown to improve anemia in some patients with myelodysplastic syndromes and stimulates erythropoietin in patients with multiple myeloma, thalidomide"s specific effects on gamma-globin gene expression during erythroid differentiation have not been studied. Thalidomide 9-20 erythropoietin Homo sapiens 117-131 17557286-8 2007 Furthermore, we could show that antiangiogenic effects of thalidomide are related to tumor-cell derived factors including vascular endothelial growth factor, basic fibroblast growth factor, hepatocyte growth factor and Il-8 some known and with, granulocyte colony stimulating growth factor and granulocyte macrophage colony stimulating growth factor, some new target molecules of thalidomide. Thalidomide 58-69 chemokine (C-X-C motif) ligand 15 Mus musculus 219-223 17635852-7 2007 Furthermore, thalidomide increased renal expression of endothelial NO synthase and decreased tumor necrosis factor-alpha and renal inflammation but did not decrease oxidative stress. Thalidomide 13-24 tumor necrosis factor Sus scrofa 93-120 17620452-1 2007 Although thalidomide has been shown to improve anemia in some patients with myelodysplastic syndromes and stimulates erythropoietin in patients with multiple myeloma, thalidomide"s specific effects on gamma-globin gene expression during erythroid differentiation have not been studied. Thalidomide 167-178 erythropoietin Homo sapiens 117-131 17620452-5 2007 Western blot analysis demonstrated that thalidomide activated the p38 mitogen-activated protein kinase (MAPK) signaling pathway in a time- and dose-dependent manner and increased histone H4 acetylation. Thalidomide 40-51 mitogen-activated protein kinase 14 Homo sapiens 66-102 17620452-6 2007 Pretreatment of cells with the antioxidant enzyme catalase and the intracellular hydroxyl scavenger dimethylthiourea (DMTU) abrogated the thalidomide-induced p38 MAPK activation and histone H4 acetylation. Thalidomide 138-149 mitogen-activated protein kinase 14 Homo sapiens 158-161 17620452-8 2007 These data indicate that thalidomide induces increased expression of the gamma-globin gene via ROS-dependent activation of the p38 MAPK signaling pathway and histone H4 acetylation. Thalidomide 25-36 mitogen-activated protein kinase 14 Homo sapiens 127-130 17514648-3 2007 The purpose was to test if thalidomide can inhibit 13-cis RA-induced HOXB7 expression and whether thalidomide may enhance the antiproliferative effect of 13-cis RA in U343MG glioblastoma cells. Thalidomide 27-38 homeobox B7 Homo sapiens 69-74 18399167-0 2007 [Effect of CYP2C19 gene polymorphism on efficacy of thalidomide-based regimens for the treatment of multiple myeloma]. Thalidomide 52-63 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 11-18 18399167-8 2007 CONCLUSION: CYP2C19 genotype has no difference between MM patients and healthy person, but exhibits an effect on the treatment efficacy of thalidomide for MM. Thalidomide 139-150 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 12-19 17514648-4 2007 Quantitative real-time PCR showed significant inhibition of 13-cis RA-induced HOXB7 expression by thalidomide with IC(50) approximately 0.1-0.2 microg/ml when given simultaneously with 13-cis RA but not when administered 18 h later (p < 0.0001). Thalidomide 98-109 homeobox B7 Homo sapiens 78-83 17514648-7 2007 It is concluded that thalidomide inhibits the RA-induced HOXB7 expression in glioblastoma cells and that 13-cis RA/thalidomide combinations can in principle enhance cytotoxicity. Thalidomide 21-32 homeobox B7 Homo sapiens 57-62 17724360-2 2007 SUMMARY: Lenalidomide is an analogue of thalidomide and has been shown to be more potent than thalidomide in the stimulation of T-cell, interleukin-2, and interferon-gamma production. Thalidomide 94-105 interleukin 2 Homo sapiens 136-149 17724360-2 2007 SUMMARY: Lenalidomide is an analogue of thalidomide and has been shown to be more potent than thalidomide in the stimulation of T-cell, interleukin-2, and interferon-gamma production. Thalidomide 94-105 interferon gamma Homo sapiens 155-171 17666363-0 2007 Polymorphisms of CYP2C19 gene are associated with the efficacy of thalidomide based regimens in multiple myeloma. Thalidomide 66-77 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 17-24 17908040-10 2007 However, thalidomide, a small molecule drug, can inhibit TNF-alpha protein synthesis and, unlike larger molecules, is readily capable of crossing the blood-brain barrier. Thalidomide 9-20 tumor necrosis factor Homo sapiens 57-66 17908040-11 2007 Thus thalidomide and its analogs are excellent candidate agents for use in determining the potential value of anti-TNF-alpha therapies in a variety of diseases underpinned by inflammation within the nervous system. Thalidomide 5-16 tumor necrosis factor Homo sapiens 115-124 17908040-13 2007 Additionally, we consider the utilization of thalidomide-derived agents as anti-TNF-alpha therapeutics in the setting of neuroinflammation. Thalidomide 45-56 tumor necrosis factor Homo sapiens 80-89 17666363-5 2007 For the first time, our primary data suggested that the polymorphisms of CYP2C19 gene are associated with the efficacy of thalidomide based regimens in MM. Thalidomide 122-133 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 73-80 17597196-13 2007 Treatment with thalidomide was associated with a significant decrease in plasma bFGF (p=0.008) and serum sEPCR (p=0.006), but not in plasma VEGF. Thalidomide 15-26 fibroblast growth factor 2 Homo sapiens 80-84 17547733-0 2007 Deep vein thrombosis occurring on treatment of patients receiving thalidomide with erythropoietin. Thalidomide 66-77 erythropoietin Homo sapiens 83-97 17581613-4 2007 Among those mobilized with granulocyte-colony stimulating factor (G-CSF) alone, there was a significant decrease in total CD34(+) cells collected (P<0.001), average daily collection (P<0.001), day 1 collection (P<0.001) and increased number of aphereses (P=0.004) in patients treated with lenalidomide compared to those receiving dexamethasone, thalidomide-dexamethasone or VAD. Thalidomide 354-365 colony stimulating factor 3 Homo sapiens 27-64 17581613-4 2007 Among those mobilized with granulocyte-colony stimulating factor (G-CSF) alone, there was a significant decrease in total CD34(+) cells collected (P<0.001), average daily collection (P<0.001), day 1 collection (P<0.001) and increased number of aphereses (P=0.004) in patients treated with lenalidomide compared to those receiving dexamethasone, thalidomide-dexamethasone or VAD. Thalidomide 354-365 colony stimulating factor 3 Homo sapiens 66-71 17498949-1 2007 Alpha-glucosidase inhibitors with a chlorinated phthalimide or a thiophthalimide skeleton, derived from thalidomide, were found to possess liver X receptor (LXR) antagonistic activity. Thalidomide 104-115 sucrase-isomaltase Homo sapiens 0-17 17437439-13 2007 The combination of estramustine, docetaxel and thalidomide is an advantageous treatment in pre-clinical models of prostate cancer and is a safe, tolerable and active regimen in patients with AIPC. Thalidomide 47-58 PDZ domain containing 2 Homo sapiens 191-195 17255354-5 2007 Despite its role as a tumor suppressor and mediator of apoptosis in other cell types including osteoblasts, our data suggest that DKK1, a stress-responsive gene in MM, does not mediate apoptotic signaling, is not activated by TP53, and its forced overexpression could not inhibit cell growth or sensitize MM cells to apoptosis following treatment with thalidomide or lenalidomide. Thalidomide 352-363 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 130-134 17579094-5 2007 Thalidomide (Thal) has been used in the treatment of multiple myeloma through the inhibitory effect on IL-6-dependent cell growth and angiogenesis. Thalidomide 0-11 interleukin 6 Mus musculus 103-107 17579094-5 2007 Thalidomide (Thal) has been used in the treatment of multiple myeloma through the inhibitory effect on IL-6-dependent cell growth and angiogenesis. Thalidomide 0-4 interleukin 6 Mus musculus 103-107 17352720-4 2007 Thalidomide, a tumor necrosis factor-alpha antagonist, has been suggested as an alternative treatment option for recalcitrant prurigo nodularis. Thalidomide 0-11 tumor necrosis factor Homo sapiens 15-42 17940990-13 2007 Due to the progression of the disease, a thalidomide was used in therapy (it is anti-VEGF agent). Thalidomide 41-52 vascular endothelial growth factor A Homo sapiens 85-89 17283219-6 2007 Thalidomide-induced cell death was dramatically reduced in HEFs and in embryonic limb buds by the use of inhibitors against Bmps, Dkk1, and Gsk3beta, a beta-catenin antagonist acting downstream of Dkk1 in the Wnt pathway. Thalidomide 0-11 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 130-134 17283219-6 2007 Thalidomide-induced cell death was dramatically reduced in HEFs and in embryonic limb buds by the use of inhibitors against Bmps, Dkk1, and Gsk3beta, a beta-catenin antagonist acting downstream of Dkk1 in the Wnt pathway. Thalidomide 0-11 glycogen synthase kinase 3 beta Homo sapiens 140-148 17283219-6 2007 Thalidomide-induced cell death was dramatically reduced in HEFs and in embryonic limb buds by the use of inhibitors against Bmps, Dkk1, and Gsk3beta, a beta-catenin antagonist acting downstream of Dkk1 in the Wnt pathway. Thalidomide 0-11 catenin beta 1 Homo sapiens 152-164 17283219-6 2007 Thalidomide-induced cell death was dramatically reduced in HEFs and in embryonic limb buds by the use of inhibitors against Bmps, Dkk1, and Gsk3beta, a beta-catenin antagonist acting downstream of Dkk1 in the Wnt pathway. Thalidomide 0-11 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 197-201 17283219-7 2007 Most interestingly, blocking of Dkk1 or Gsk3beta dramatically counteracts thalidomide-induced limb truncations and microphthalmia. Thalidomide 74-85 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 32-36 17283219-7 2007 Most interestingly, blocking of Dkk1 or Gsk3beta dramatically counteracts thalidomide-induced limb truncations and microphthalmia. Thalidomide 74-85 glycogen synthase kinase 3 beta Homo sapiens 40-48 17283219-8 2007 From this, we conclude that perturbing of Bmp/Dkk1/Wnt signaling is central to the teratogenic effects of thalidomide. Thalidomide 106-117 bone morphogenetic protein 1 Homo sapiens 42-45 17283219-8 2007 From this, we conclude that perturbing of Bmp/Dkk1/Wnt signaling is central to the teratogenic effects of thalidomide. Thalidomide 106-117 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 46-50 17409565-0 2007 Hydrolyzed metabolites of thalidomide: synthesis and TNF-alpha production-inhibitory activity. Thalidomide 26-37 tumor necrosis factor Homo sapiens 53-62 17409565-1 2007 Putative hydrolyzed metabolites of thalidomide were prepared and characterized, and their inhibitory activity on tumor necrosis factor (TNF)-alpha production in the human monocytic leukemia cell line THP-1 was evaluated. Thalidomide 35-46 tumor necrosis factor Homo sapiens 113-146 17409565-1 2007 Putative hydrolyzed metabolites of thalidomide were prepared and characterized, and their inhibitory activity on tumor necrosis factor (TNF)-alpha production in the human monocytic leukemia cell line THP-1 was evaluated. Thalidomide 35-46 GLI family zinc finger 2 Homo sapiens 200-205 17283219-0 2007 Thalidomide induces limb deformities by perturbing the Bmp/Dkk1/Wnt signaling pathway. Thalidomide 0-11 bone morphogenetic protein 1 Homo sapiens 55-58 17283219-0 2007 Thalidomide induces limb deformities by perturbing the Bmp/Dkk1/Wnt signaling pathway. Thalidomide 0-11 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 59-63 17465495-0 2007 Therapeutic and prophylactic thalidomide in TNBS-induced colitis: synergistic effects on TNF-alpha, IL-12 and VEGF production. Thalidomide 29-40 tumor necrosis factor Homo sapiens 89-98 17465495-0 2007 Therapeutic and prophylactic thalidomide in TNBS-induced colitis: synergistic effects on TNF-alpha, IL-12 and VEGF production. Thalidomide 29-40 vascular endothelial growth factor A Homo sapiens 110-114 17465495-2 2007 Thalidomide has been reported to downregulate the expression of tumor necrosis factor alpha (TNF-alpha), IL-12, and vascular endothelial growth factor (VEGF), hallmarks of intestinal inflammation in Crohnos disease (CD). Thalidomide 0-11 tumor necrosis factor Homo sapiens 64-91 17465495-2 2007 Thalidomide has been reported to downregulate the expression of tumor necrosis factor alpha (TNF-alpha), IL-12, and vascular endothelial growth factor (VEGF), hallmarks of intestinal inflammation in Crohnos disease (CD). Thalidomide 0-11 tumor necrosis factor Homo sapiens 93-102 17465495-2 2007 Thalidomide has been reported to downregulate the expression of tumor necrosis factor alpha (TNF-alpha), IL-12, and vascular endothelial growth factor (VEGF), hallmarks of intestinal inflammation in Crohnos disease (CD). Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 116-150 17465495-2 2007 Thalidomide has been reported to downregulate the expression of tumor necrosis factor alpha (TNF-alpha), IL-12, and vascular endothelial growth factor (VEGF), hallmarks of intestinal inflammation in Crohnos disease (CD). Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 152-156 17465495-10 2007 RESULTS: Significant reduction in the inflammatory score and in the percentage of VEGF+ cells was observed in the group treated with thalidomide compared with animals not treated with thalidomide. Thalidomide 133-144 vascular endothelial growth factor A Homo sapiens 82-86 17465495-11 2007 Both TNF-alpha and IL-12 levels were significantly reduced among TNBS induced colitis animals treated with thalidomide compared with animals that did not receive thalidomide. Thalidomide 107-118 tumor necrosis factor Homo sapiens 5-14 17465495-12 2007 TNF-alpha levels were also significantly reduced among the animals receiving thalidomide prophylaxis compared with untreated animals with TNBS-induced colitis. Thalidomide 77-88 tumor necrosis factor Homo sapiens 0-9 17465495-14 2007 CONCLUSION: Thalidomide significantly attenuates TNBS-induced colitis by inhibiting the intestinal production of TNF-alpha, IL-12, and VEGF. Thalidomide 12-23 tumor necrosis factor Homo sapiens 113-122 17465495-14 2007 CONCLUSION: Thalidomide significantly attenuates TNBS-induced colitis by inhibiting the intestinal production of TNF-alpha, IL-12, and VEGF. Thalidomide 12-23 vascular endothelial growth factor A Homo sapiens 135-139 17367589-5 2007 Thalidomide was found to be effective in animal models by inhibiting TNF-alpha production. Thalidomide 0-11 tumor necrosis factor Homo sapiens 69-78 17208222-5 2007 These data suggest that one of the molecular mechanisms of thalidomide may be destabilization of COX-2 mRNA through inhibition of cytoplasmic shuttling of HuR and p38 MAPK. Thalidomide 59-70 mitogen-activated protein kinase 14 Homo sapiens 163-166 17383526-0 2007 Effects of geldanamycin and thalidomide on the Th1/Th2 cytokine balance in mice subjected to operative trauma. Thalidomide 28-39 negative elongation factor complex member C/D, Th1l Mus musculus 47-50 17383526-0 2007 Effects of geldanamycin and thalidomide on the Th1/Th2 cytokine balance in mice subjected to operative trauma. Thalidomide 28-39 heart and neural crest derivatives expressed 2 Mus musculus 51-54 17383526-13 2007 Geldanamycin and thalidomide improved the Th1/Th2 imbalance independently after major operative injury. Thalidomide 17-28 negative elongation factor complex member C/D, Th1l Mus musculus 42-45 17383526-13 2007 Geldanamycin and thalidomide improved the Th1/Th2 imbalance independently after major operative injury. Thalidomide 17-28 heart and neural crest derivatives expressed 2 Mus musculus 46-49 17208222-0 2007 Thalidomide destabilizes cyclooxygenase-2 mRNA by inhibiting p38 mitogen-activated protein kinase and cytoplasmic shuttling of HuR. Thalidomide 0-11 prostaglandin-endoperoxide synthase 2 Homo sapiens 25-41 17258708-8 2007 Intraperitoneal injection of thalidomide, an inhibitor of TNF-alpha synthesis, prevented p38 activation in DRG and spinal cord. Thalidomide 29-40 tumor necrosis factor Homo sapiens 58-67 17208222-0 2007 Thalidomide destabilizes cyclooxygenase-2 mRNA by inhibiting p38 mitogen-activated protein kinase and cytoplasmic shuttling of HuR. Thalidomide 0-11 mitogen-activated protein kinase 14 Homo sapiens 61-97 17208222-0 2007 Thalidomide destabilizes cyclooxygenase-2 mRNA by inhibiting p38 mitogen-activated protein kinase and cytoplasmic shuttling of HuR. Thalidomide 0-11 ELAV like RNA binding protein 1 Homo sapiens 127-130 17208222-1 2007 We investigated the effect of thalidomide on transcriptional and post-transcriptional cyclooxygenase-2 (COX-2) expression, including a pathway leading to COX-2 mRNA destabilization. Thalidomide 30-41 prostaglandin-endoperoxide synthase 2 Homo sapiens 86-102 17208222-1 2007 We investigated the effect of thalidomide on transcriptional and post-transcriptional cyclooxygenase-2 (COX-2) expression, including a pathway leading to COX-2 mRNA destabilization. Thalidomide 30-41 prostaglandin-endoperoxide synthase 2 Homo sapiens 104-109 17208222-2 2007 We found that thalidomide inhibited the interleukin-1beta (IL-1beta)-mediated induction of COX-2 protein and mRNA in Caco-2 cells. Thalidomide 14-25 interleukin 1 beta Homo sapiens 40-57 17208222-2 2007 We found that thalidomide inhibited the interleukin-1beta (IL-1beta)-mediated induction of COX-2 protein and mRNA in Caco-2 cells. Thalidomide 14-25 interleukin 1 beta Homo sapiens 59-67 17208222-2 2007 We found that thalidomide inhibited the interleukin-1beta (IL-1beta)-mediated induction of COX-2 protein and mRNA in Caco-2 cells. Thalidomide 14-25 prostaglandin-endoperoxide synthase 2 Homo sapiens 91-96 17208222-5 2007 These data suggest that one of the molecular mechanisms of thalidomide may be destabilization of COX-2 mRNA through inhibition of cytoplasmic shuttling of HuR and p38 MAPK. Thalidomide 59-70 prostaglandin-endoperoxide synthase 2 Homo sapiens 97-102 17208222-5 2007 These data suggest that one of the molecular mechanisms of thalidomide may be destabilization of COX-2 mRNA through inhibition of cytoplasmic shuttling of HuR and p38 MAPK. Thalidomide 59-70 ELAV like RNA binding protein 1 Homo sapiens 155-158 17327701-12 2007 The MTD was 100 mg thalidomide. Thalidomide 19-30 metallothionein 1E Homo sapiens 4-7 17562330-6 2007 The TNF antagonist thalidomide suppressed ADCC whereas pentoxifylline proved to be ineffective. Thalidomide 19-30 tumor necrosis factor Homo sapiens 4-7 17258708-8 2007 Intraperitoneal injection of thalidomide, an inhibitor of TNF-alpha synthesis, prevented p38 activation in DRG and spinal cord. Thalidomide 29-40 mitogen-activated protein kinase 14 Homo sapiens 89-92 17295769-3 2007 The aim of the study was to evaluate the effectiveness of thalidomide, an immunomodulator that exerts an inhibitory action on TNF-alpha by enhancing mRNA degradation, in reducing post-ERCP pancreatitis in a rat model. Thalidomide 58-69 tumor necrosis factor Rattus norvegicus 126-135 17223105-3 2007 Moreover, the actions of thalidomide were assessed on the cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) protein expression in retinal tissue. Thalidomide 25-36 cytochrome c oxidase II, mitochondrial Rattus norvegicus 58-80 17223105-3 2007 Moreover, the actions of thalidomide were assessed on the cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) protein expression in retinal tissue. Thalidomide 25-36 nitric oxide synthase 2 Rattus norvegicus 85-116 17223105-3 2007 Moreover, the actions of thalidomide were assessed on the cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) protein expression in retinal tissue. Thalidomide 25-36 nitric oxide synthase 2 Rattus norvegicus 118-122 17223105-7 2007 The oral pre-treatment with thalidomide decreased, in a dose-dependent manner, the number of inflammatory cells, the protein concentration, and the levels of IL-1beta and TNF-alpha in the AqH. Thalidomide 28-39 interleukin 1 beta Rattus norvegicus 158-166 17223105-7 2007 The oral pre-treatment with thalidomide decreased, in a dose-dependent manner, the number of inflammatory cells, the protein concentration, and the levels of IL-1beta and TNF-alpha in the AqH. Thalidomide 28-39 tumor necrosis factor Rattus norvegicus 171-180 17295769-7 2007 The prophylactic use of thalidomide significantly reduced serum amylase, pancreatic edema and the histologic grade of pancreatitis accompanied by a decrease in mRNA expression of TNF-alpha in the pancreatic tissue. Thalidomide 24-35 tumor necrosis factor Rattus norvegicus 179-188 17295769-9 2007 The mechanism of the protective effects of thalidomide seems to be the reduction of expression of TNF-alpha mRNA in pancreatic tissue. Thalidomide 43-54 tumor necrosis factor Rattus norvegicus 98-107 17268092-0 2007 Thalidomide analogs from diamines: Synthesis and evaluation as inhibitors of TNF-alpha production. Thalidomide 0-11 tumor necrosis factor Homo sapiens 77-86 17223105-9 2007 Furthermore, oral (50 mg/kg) and local (5%) thalidomide treatment also reduced expression of the pro-inflammatory proteins COX-2 and iNOS in the posterior segment of the eye. Thalidomide 44-55 cytochrome c oxidase II, mitochondrial Rattus norvegicus 123-128 17223105-9 2007 Furthermore, oral (50 mg/kg) and local (5%) thalidomide treatment also reduced expression of the pro-inflammatory proteins COX-2 and iNOS in the posterior segment of the eye. Thalidomide 44-55 nitric oxide synthase 2 Rattus norvegicus 133-137 17223105-10 2007 Thalidomide exhibited marked preventive and curative ocular effects in EIU in rats, a property that might be associated with its ability to inhibit the production of inflammatory cytokines and the expression of COX-2 and iNOS. Thalidomide 0-11 cytochrome c oxidase II, mitochondrial Rattus norvegicus 211-216 17223105-10 2007 Thalidomide exhibited marked preventive and curative ocular effects in EIU in rats, a property that might be associated with its ability to inhibit the production of inflammatory cytokines and the expression of COX-2 and iNOS. Thalidomide 0-11 nitric oxide synthase 2 Rattus norvegicus 221-225 16568139-2 2007 OBJECTIVES: To evaluate the effects of thalidomide on spinal cord ischemia/reperfusion injury via reduced TNF-alpha production. Thalidomide 39-50 tumor necrosis factor Oryctolagus cuniculus 106-115 16815546-2 2007 Thalidomide has been shown to inhibit production of TNF-alpha. Thalidomide 0-11 tumor necrosis factor Homo sapiens 52-61 17503883-6 2007 Although thalidomide has been regarded as an anti-TNF drug, it was found to increase plasma levels of TNFalpha in a placebo-controlled study in patients with HF, suggesting that other mechanisms may contribute to its beneficial effects. Thalidomide 9-20 tumor necrosis factor Homo sapiens 50-53 17208262-0 2007 Differential surface expression of CD18 and CD44 by neutrophils in bone marrow and spleen contributed to the neutrophilia in thalidomide-treated female B6C3F1 mice. Thalidomide 125-136 integrin beta 2 Mus musculus 35-39 17208262-0 2007 Differential surface expression of CD18 and CD44 by neutrophils in bone marrow and spleen contributed to the neutrophilia in thalidomide-treated female B6C3F1 mice. Thalidomide 125-136 CD44 antigen Mus musculus 44-48 17208262-7 2007 Flow cytometric analysis of the surface expression of adhesion molecules suggested that Thd treatment for either 14 or 28 days decreased the surface expression of either CD18 or CD44 by bone marrow neutrophils. Thalidomide 88-91 integrin beta 2 Mus musculus 170-174 17208262-7 2007 Flow cytometric analysis of the surface expression of adhesion molecules suggested that Thd treatment for either 14 or 28 days decreased the surface expression of either CD18 or CD44 by bone marrow neutrophils. Thalidomide 88-91 CD44 antigen Mus musculus 178-182 17208262-8 2007 On the other hand, the surface expression of both CD18 and CD44 by splenic neutrophils was increased following Thd treatment for 28 days but not for 14 days. Thalidomide 111-114 integrin beta 2 Mus musculus 50-54 17208262-8 2007 On the other hand, the surface expression of both CD18 and CD44 by splenic neutrophils was increased following Thd treatment for 28 days but not for 14 days. Thalidomide 111-114 CD44 antigen Mus musculus 59-63 17208262-10 2007 It was possible that decreased surface expressions of CD18 and CD44 facilitated neutrophils" release from the bone marrow; increased surface expressions of CD44 and CD18 by splenic neutrophils after 28 days of Thd treatment increased their ability to remain in the periphery. Thalidomide 210-213 integrin beta 2 Mus musculus 54-58 17208262-10 2007 It was possible that decreased surface expressions of CD18 and CD44 facilitated neutrophils" release from the bone marrow; increased surface expressions of CD44 and CD18 by splenic neutrophils after 28 days of Thd treatment increased their ability to remain in the periphery. Thalidomide 210-213 CD44 antigen Mus musculus 63-67 17208262-10 2007 It was possible that decreased surface expressions of CD18 and CD44 facilitated neutrophils" release from the bone marrow; increased surface expressions of CD44 and CD18 by splenic neutrophils after 28 days of Thd treatment increased their ability to remain in the periphery. Thalidomide 210-213 CD44 antigen Mus musculus 156-160 17208262-10 2007 It was possible that decreased surface expressions of CD18 and CD44 facilitated neutrophils" release from the bone marrow; increased surface expressions of CD44 and CD18 by splenic neutrophils after 28 days of Thd treatment increased their ability to remain in the periphery. Thalidomide 210-213 integrin beta 2 Mus musculus 165-169 17208262-11 2007 Taken together, Thd treatment increased neutrophils in female B6C3F1 mice, at least partially, through differentially modulating the surface expression of CD18 and CD44 by the neutrophils in the bone marrow and spleen. Thalidomide 16-19 integrin beta 2 Mus musculus 155-159 17208262-11 2007 Taken together, Thd treatment increased neutrophils in female B6C3F1 mice, at least partially, through differentially modulating the surface expression of CD18 and CD44 by the neutrophils in the bone marrow and spleen. Thalidomide 16-19 CD44 antigen Mus musculus 164-168 17174718-2 2007 This study was designed to investigate the effects of thalidomide on expressions of NF-kappaB, IkappaB and intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule (VCAM-1) in established rat liver cirrhosis. Thalidomide 54-65 intercellular adhesion molecule 1 Rattus norvegicus 107-140 17174718-2 2007 This study was designed to investigate the effects of thalidomide on expressions of NF-kappaB, IkappaB and intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule (VCAM-1) in established rat liver cirrhosis. Thalidomide 54-65 intercellular adhesion molecule 1 Rattus norvegicus 142-148 17503883-6 2007 Although thalidomide has been regarded as an anti-TNF drug, it was found to increase plasma levels of TNFalpha in a placebo-controlled study in patients with HF, suggesting that other mechanisms may contribute to its beneficial effects. Thalidomide 9-20 tumor necrosis factor Homo sapiens 102-110 18257406-4 2007 The newest trend in the treatment of this multi-organ granulomatose disease of unknown origin is the use of TNF inhibitors--pentoxifyllin, infliximab, adalimumab, leflunomid, thalidomid. Thalidomide 175-185 tumor necrosis factor Homo sapiens 108-111 17106197-2 2007 Thalidomide has recently shown to improve LVEF in chronic heart failure patients, accompanied by a marked decrease in plasma levels of tumor necrosis factor alpha (TNF-alpha). Thalidomide 0-11 tumor necrosis factor Homo sapiens 135-162 17106197-2 2007 Thalidomide has recently shown to improve LVEF in chronic heart failure patients, accompanied by a marked decrease in plasma levels of tumor necrosis factor alpha (TNF-alpha). Thalidomide 0-11 tumor necrosis factor Homo sapiens 164-173 17106197-7 2007 TNF-alpha levels were initially of 5.88 +/- 0.9 and 6.49 +/- 1.82 vs. 6.32 +/- 1.6 and 7.94 +/- 3.8 pg/ml during follow-up, for thalidomide and control groups, respectively. Thalidomide 128-139 tumor necrosis factor Homo sapiens 0-9 17106197-9 2007 CONCLUSION: In conclusion, although there is a large amount of information supporting a direct relationship between TNF-alpha and worsening of symptoms and prognosis in patients with HF and recently, the beneficial effect on thalidomide treatment has been suggested, these preliminary observations should be confirmed in a larger prospective study, specially trying to clarify the action mechanisms. Thalidomide 225-236 tumor necrosis factor Homo sapiens 116-125 17852116-3 2007 Our group were the first to demonstrate activity of thalidomide in RCC, believed to be in part related to the modulation of tumor necrosis factor (TNF-a), a cytokine secreted by RCC with a number of tumor promoting properties. Thalidomide 52-63 tumor necrosis factor Homo sapiens 124-145 17852116-3 2007 Our group were the first to demonstrate activity of thalidomide in RCC, believed to be in part related to the modulation of tumor necrosis factor (TNF-a), a cytokine secreted by RCC with a number of tumor promoting properties. Thalidomide 52-63 tumor necrosis factor Homo sapiens 147-152 17139107-0 2006 Mono- and dihydroxylated metabolites of thalidomide: synthesis and TNF-alpha production-inhibitory activity. Thalidomide 40-51 tumor necrosis factor Homo sapiens 67-76 17369076-4 2007 Furthermore, vascular endothelial growth factor (VEGF) and beta fibroblast growth factor (bFGF) secretion and cellular response are suppressed by thalidomide, thus antagonizing neoangiogenesis and altering the bone marrow stromal microenvironment in hematologic malignancies. Thalidomide 146-157 vascular endothelial growth factor A Homo sapiens 13-47 17369076-4 2007 Furthermore, vascular endothelial growth factor (VEGF) and beta fibroblast growth factor (bFGF) secretion and cellular response are suppressed by thalidomide, thus antagonizing neoangiogenesis and altering the bone marrow stromal microenvironment in hematologic malignancies. Thalidomide 146-157 vascular endothelial growth factor A Homo sapiens 49-53 17369076-4 2007 Furthermore, vascular endothelial growth factor (VEGF) and beta fibroblast growth factor (bFGF) secretion and cellular response are suppressed by thalidomide, thus antagonizing neoangiogenesis and altering the bone marrow stromal microenvironment in hematologic malignancies. Thalidomide 146-157 fibroblast growth factor 2 Homo sapiens 90-94 17369076-5 2007 The thalidomide analogs, lenalidomide (CC-5013; Revlimid) and CC-4047 (Actimid), have enhanced potency as inhibitors of TNF-alpha and other inflammatory cytokines, as well as greater capacity to promote T-cell activation and suppress angiogenesis. Thalidomide 4-15 tumor necrosis factor Homo sapiens 120-129 16333818-3 2007 Thalidomide inhibits TNF-alpha production by affecting the gene promoter as well as other anti-cytokine effects. Thalidomide 0-11 tumor necrosis factor Homo sapiens 21-30 17164777-0 2007 Therapeutic angiogenesis inhibits or rescues chemotherapy-induced peripheral neuropathy: taxol- and thalidomide-induced injury of vasa nervorum is ameliorated by VEGF. Thalidomide 100-111 vascular endothelial growth factor A Homo sapiens 162-166 17139107-1 2006 Mono- and dihydroxylated metabolites of thalidomide were efficiently prepared and characterized, and their inhibitory activity on tumor necrosis factor (TNF)-alpha production in the human monocytic leukemia cell line THP-1 was evaluated. Thalidomide 40-51 tumor necrosis factor Homo sapiens 130-163 17139107-1 2006 Mono- and dihydroxylated metabolites of thalidomide were efficiently prepared and characterized, and their inhibitory activity on tumor necrosis factor (TNF)-alpha production in the human monocytic leukemia cell line THP-1 was evaluated. Thalidomide 40-51 GLI family zinc finger 2 Homo sapiens 217-222 17139107-2 2006 5,N-Dihydroxythalidomide was a much more potent TNF-alpha production inhibitor than thalidomide. Thalidomide 13-24 tumor necrosis factor Homo sapiens 48-57 16549389-7 2006 (iii) Thalidomide appeared to have immunostimulatory effects on the myocardium as evident by increased MIP-1alpha gene expression (p<0.05). Thalidomide 6-17 C-C motif chemokine ligand 3 Rattus norvegicus 103-113 17145842-0 2006 A novel anticancer effect of thalidomide: inhibition of intercellular adhesion molecule-1-mediated cell invasion and metastasis through suppression of nuclear factor-kappaB. Thalidomide 29-40 intercellular adhesion molecule 1 Homo sapiens 56-89 17145842-12 2006 Thalidomide also suppressed tumor necrosis factor-alpha-induced ICAM-1 expression through inhibition of nuclear factor-kappaB binding to the ICAM-1 promoter. Thalidomide 0-11 tumor necrosis factor Homo sapiens 28-55 17145842-12 2006 Thalidomide also suppressed tumor necrosis factor-alpha-induced ICAM-1 expression through inhibition of nuclear factor-kappaB binding to the ICAM-1 promoter. Thalidomide 0-11 intercellular adhesion molecule 1 Homo sapiens 64-70 17145842-12 2006 Thalidomide also suppressed tumor necrosis factor-alpha-induced ICAM-1 expression through inhibition of nuclear factor-kappaB binding to the ICAM-1 promoter. Thalidomide 0-11 intercellular adhesion molecule 1 Homo sapiens 141-147 16549389-8 2006 (iv) Treating HF rats with thalidomide reduced myocardial collagen content, as assessed by markedly decreased levels of hydroxyproline ( approximately 40% reduction; p<0.05), accompanied by lower TGF-beta(1) gene expression (p<0.05). Thalidomide 27-38 transforming growth factor, beta 1 Rattus norvegicus 199-210 17099831-15 2006 CONCLUSIONS: We conclude that outpatient administration of thalidomide/IL-2 is feasible in patients with heavily pretreated and progressive RCC who desire further active treatment. Thalidomide 59-70 interleukin 2 Homo sapiens 71-75 17204188-3 2006 The results showed that thalidomide enhanced the proliferations of the CD8+ T, NK cells in PHA-stimulated PBMNC from healthy volunteers, increased the secretion of IL-6 significantly, and decreased the secretion of IFN-gamma, and the secretions of IL-2 and IL-10 were not affected. Thalidomide 24-35 interleukin 6 Homo sapiens 164-168 17204188-3 2006 The results showed that thalidomide enhanced the proliferations of the CD8+ T, NK cells in PHA-stimulated PBMNC from healthy volunteers, increased the secretion of IL-6 significantly, and decreased the secretion of IFN-gamma, and the secretions of IL-2 and IL-10 were not affected. Thalidomide 24-35 interferon gamma Homo sapiens 215-224 17204188-3 2006 The results showed that thalidomide enhanced the proliferations of the CD8+ T, NK cells in PHA-stimulated PBMNC from healthy volunteers, increased the secretion of IL-6 significantly, and decreased the secretion of IFN-gamma, and the secretions of IL-2 and IL-10 were not affected. Thalidomide 24-35 interleukin 2 Homo sapiens 248-252 17204188-3 2006 The results showed that thalidomide enhanced the proliferations of the CD8+ T, NK cells in PHA-stimulated PBMNC from healthy volunteers, increased the secretion of IL-6 significantly, and decreased the secretion of IFN-gamma, and the secretions of IL-2 and IL-10 were not affected. Thalidomide 24-35 interleukin 10 Homo sapiens 257-262 17204188-5 2006 It is concluded that thalidomide preferentially enhances the proliferations of CD8+ T, NK cells in PHA-stimulated PBMNC from healthy volunteers, and enhances the cytotoxic activity of PBMNC by increasing the secretion of IL-6 significantly, in short, thalidomide can exert anti-myeloma effects by increasing cellular immune function. Thalidomide 21-32 interleukin 6 Homo sapiens 221-225 17204188-5 2006 It is concluded that thalidomide preferentially enhances the proliferations of CD8+ T, NK cells in PHA-stimulated PBMNC from healthy volunteers, and enhances the cytotoxic activity of PBMNC by increasing the secretion of IL-6 significantly, in short, thalidomide can exert anti-myeloma effects by increasing cellular immune function. Thalidomide 251-262 interleukin 6 Homo sapiens 221-225 17162660-6 2006 In thalidomide-treated mice, TNF-alpha RNA levels were reduced in the SCN. Thalidomide 3-14 tumor necrosis factor Mus musculus 29-38 17195429-2 2006 Thalidomide (Thalomid, Celgene Corp., Summit, NJ), an angiogenesis inhibitor and immunomodulator that reduces tumor necrosis factor-alpha, is used for the treatment of multiple myeloma. Thalidomide 0-11 tumor necrosis factor Homo sapiens 110-137 17195429-2 2006 Thalidomide (Thalomid, Celgene Corp., Summit, NJ), an angiogenesis inhibitor and immunomodulator that reduces tumor necrosis factor-alpha, is used for the treatment of multiple myeloma. Thalidomide 13-21 tumor necrosis factor Homo sapiens 110-137 16965441-0 2006 Increased mortality in toxic epidermal necrolysis with thalidomide: corroborating or exonerating the pathogenetic role of TNF-alpha? Thalidomide 55-66 tumor necrosis factor Homo sapiens 122-131 16843662-1 2006 In the present study, we describe a new 3D-QSAR analysis of 42 previously reported thalidomide analogues, with the ability to modulate the pro-inflammatory cytokine TNFalpha, by using comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA). Thalidomide 83-94 tumor necrosis factor Homo sapiens 165-173 16815871-0 2006 A mechanistic study on reduced toxicity of irinotecan by coadministered thalidomide, a tumor necrosis factor-alpha inhibitor. Thalidomide 72-83 tumor necrosis factor Rattus norvegicus 87-114 16837501-0 2006 Thalidomide reduces IL-18, IL-8 and TNF-alpha release from alveolar macrophages in interstitial lung disease. Thalidomide 0-11 interleukin 18 Homo sapiens 20-25 16837501-0 2006 Thalidomide reduces IL-18, IL-8 and TNF-alpha release from alveolar macrophages in interstitial lung disease. Thalidomide 0-11 C-X-C motif chemokine ligand 8 Homo sapiens 27-31 16837501-0 2006 Thalidomide reduces IL-18, IL-8 and TNF-alpha release from alveolar macrophages in interstitial lung disease. Thalidomide 0-11 tumor necrosis factor Homo sapiens 36-45 16837501-2 2006 The efficacy of thalidomide treatment in sarcoidosis with lupus pernio is thought to be due to inhibition of tumour necrosis factor (TNF)-alpha. Thalidomide 16-27 tumor necrosis factor Homo sapiens 109-143 16837501-5 2006 In sarcoidosis and HP patients, thalidomide induced a dose-dependent, partial suppression of lipopolysacchride (LPS)-stimulated TNF-alpha, IL-12p40 and IL-18 release. Thalidomide 32-43 tumor necrosis factor Homo sapiens 128-137 16837501-5 2006 In sarcoidosis and HP patients, thalidomide induced a dose-dependent, partial suppression of lipopolysacchride (LPS)-stimulated TNF-alpha, IL-12p40 and IL-18 release. Thalidomide 32-43 interleukin 18 Homo sapiens 152-157 16837501-6 2006 At the highest thalidomide concentration (0.1 mM), LPS-stimulated IL-8 production was also suppressed. Thalidomide 15-26 C-X-C motif chemokine ligand 8 Homo sapiens 66-70 16837501-7 2006 In IPF patients, although spontaneous production of TNF-alpha, IL-12p40, IL-18 and IL-8 was lower than in sarcoidosis and HP patients, with LPS stimulation the cytokines were significantly elevated and also partially inhibited by thalidomide. Thalidomide 230-241 tumor necrosis factor Homo sapiens 52-61 16837501-7 2006 In IPF patients, although spontaneous production of TNF-alpha, IL-12p40, IL-18 and IL-8 was lower than in sarcoidosis and HP patients, with LPS stimulation the cytokines were significantly elevated and also partially inhibited by thalidomide. Thalidomide 230-241 C-X-C motif chemokine ligand 8 Homo sapiens 83-87 16837501-8 2006 In conclusion, thalidomide has the potential to improve the therapeutic regimens for sarcoidosis, hypersensitivity pneumonitis and idiopathic pulmonary fibrosis by reducing tumour necrosis factor-alpha, interleukin-12p40, interleukin-18 and interleukin-8 production. Thalidomide 15-26 interleukin 12B Homo sapiens 203-220 16837501-8 2006 In conclusion, thalidomide has the potential to improve the therapeutic regimens for sarcoidosis, hypersensitivity pneumonitis and idiopathic pulmonary fibrosis by reducing tumour necrosis factor-alpha, interleukin-12p40, interleukin-18 and interleukin-8 production. Thalidomide 15-26 interleukin 18 Homo sapiens 222-236 16837501-8 2006 In conclusion, thalidomide has the potential to improve the therapeutic regimens for sarcoidosis, hypersensitivity pneumonitis and idiopathic pulmonary fibrosis by reducing tumour necrosis factor-alpha, interleukin-12p40, interleukin-18 and interleukin-8 production. Thalidomide 15-26 C-X-C motif chemokine ligand 8 Homo sapiens 241-254 16815871-14 2006 Thalidomide and PGA also significantly inhibited P-glycoprotein (PgP/MDR1), multidrug resistance-associated protein (MRP1)- and MRP2-mediated CPT-11 and SN-38 transport in MDCKII cells. Thalidomide 0-11 PGP Canis lupus familiaris 65-68 16815871-14 2006 Thalidomide and PGA also significantly inhibited P-glycoprotein (PgP/MDR1), multidrug resistance-associated protein (MRP1)- and MRP2-mediated CPT-11 and SN-38 transport in MDCKII cells. Thalidomide 0-11 ATP binding cassette subfamily B member 1 Canis lupus familiaris 69-73 16815871-14 2006 Thalidomide and PGA also significantly inhibited P-glycoprotein (PgP/MDR1), multidrug resistance-associated protein (MRP1)- and MRP2-mediated CPT-11 and SN-38 transport in MDCKII cells. Thalidomide 0-11 ATP binding cassette subfamily C member 1 Canis lupus familiaris 76-121 16815871-14 2006 Thalidomide and PGA also significantly inhibited P-glycoprotein (PgP/MDR1), multidrug resistance-associated protein (MRP1)- and MRP2-mediated CPT-11 and SN-38 transport in MDCKII cells. Thalidomide 0-11 ATP binding cassette subfamily C member 2 Canis lupus familiaris 128-132 16846617-3 2006 The purpose of this study was to examine the effects of thalidomide on secretion of prolactin (PRL) and vascular endothelial growth factor (VEGF), cell proliferation, apoptosis and angiogenesis within the anterior pituitary gland in long-term diethylstilboestrol (DES)-treated male F344 rats in vivo and in vitro. Thalidomide 56-67 prolactin Rattus norvegicus 84-93 16846617-5 2006 On the other hand, simultaneous treatment of F344 rats with thalidomide for the last 15 days of the experiment attenuated the stimulatory effect of DES on PRL and VEGF secretion. Thalidomide 60-71 prolactin Rattus norvegicus 155-158 16846617-5 2006 On the other hand, simultaneous treatment of F344 rats with thalidomide for the last 15 days of the experiment attenuated the stimulatory effect of DES on PRL and VEGF secretion. Thalidomide 60-71 vascular endothelial growth factor A Rattus norvegicus 163-167 16846617-8 2006 In addition, thalidomide (10(-4) to 10(-6) M) inhibited cell proliferation, prolactin and VEGF secretion from rat pituitary prolactinoma cells cultured in vitro. Thalidomide 13-24 prolactin Rattus norvegicus 76-85 16846617-8 2006 In addition, thalidomide (10(-4) to 10(-6) M) inhibited cell proliferation, prolactin and VEGF secretion from rat pituitary prolactinoma cells cultured in vitro. Thalidomide 13-24 vascular endothelial growth factor A Rattus norvegicus 90-94 16923192-14 2006 CONCLUSION: Results suggest that exposure to valproate, thalidomide or alcohol during the first trimester of pregnancy may change the ultrastructure of the placenta by altering the migration of trophoblast cells and this effect may be mediated by drug- or alcohol-induced changes in the expression levels of beta1 and alpha5 integrin subunits. Thalidomide 56-67 potassium calcium-activated channel subfamily M regulatory beta subunit 1 Homo sapiens 308-324 17031469-1 2006 INTRODUCTION: Thalidomide is a derivative of glutamic acid with anti-angiogenic, anti-inflammatory, immunomodulatory and anti-cancer properties that was found to inhibit the production of tumor necrosis factor alpha in vitro, stimulate reactive oxygen species production, and inhibit vascular endothelial growth factor receptor in acute leukemias. Thalidomide 14-25 tumor necrosis factor Homo sapiens 188-215 16995741-3 2006 Thalidomide was chosen as the skeleton on which to base the new lead series, as it moderately inhibits MMP-3, is antiangiogenic, and lends itself easily to structural modifications. Thalidomide 0-11 matrix metallopeptidase 3 Homo sapiens 103-108 16877307-0 2006 Preparation and in vitro analysis of microcapsule thalidomide formulation for targeted suppression of TNF-alpha. Thalidomide 50-61 tumor necrosis factor Mus musculus 102-111 16877307-2 2006 Thalidomide has been shown to decrease this inflammation by the suppression of TNF-alpha secretion. Thalidomide 0-11 tumor necrosis factor Mus musculus 79-88 16675114-8 2006 Intraperitoneal injection of thalidomide, an inhibitor of TNF-alpha synthesis, started at 2h before surgery, blocked mechanical allodynia and thermal hyperalgesia. Thalidomide 29-40 tumor necrosis factor Rattus norvegicus 58-67 16765039-12 2006 It is concluded that thalidomide shows single-agent activity in patients with heavily pre-treated GCT, AFP elevations and slowly progressive disease. Thalidomide 21-32 alpha fetoprotein Homo sapiens 103-106 17013434-2 2006 Thalidomide exerts its effects by decreasing circulating CD4 positive T-cells and stimulating CD8 positive T-cells, by increasing the number of Natural Killer cells and T-helper 2 cells. Thalidomide 0-11 CD4 molecule Homo sapiens 57-60 16616857-0 2006 Thalidomide alters c-MYB and PIM-1 signaling in K-562 cells. Thalidomide 0-11 MYB proto-oncogene, transcription factor Homo sapiens 19-24 16616857-0 2006 Thalidomide alters c-MYB and PIM-1 signaling in K-562 cells. Thalidomide 0-11 Pim-1 proto-oncogene, serine/threonine kinase Homo sapiens 29-34 16616857-3 2006 We hypothesize that thalidomide acts by interfering with the c-Myb signaling pathway. Thalidomide 20-31 MYB proto-oncogene, transcription factor Homo sapiens 61-66 16616857-6 2006 Cells exposed to thalidomide (40 microg/ml) had significantly decreased c-Myb activity. Thalidomide 17-28 MYB proto-oncogene, transcription factor Homo sapiens 72-77 16616857-7 2006 Pre-incubation of cells with the anti-oxidative enzyme catalase (1600 units/ml), prevented thalidomide-induced decreased c-Myb activity, suggesting a role for ROS in the c-Myb signaling pathway. Thalidomide 91-102 MYB proto-oncogene, transcription factor Homo sapiens 121-126 16616857-7 2006 Pre-incubation of cells with the anti-oxidative enzyme catalase (1600 units/ml), prevented thalidomide-induced decreased c-Myb activity, suggesting a role for ROS in the c-Myb signaling pathway. Thalidomide 91-102 MYB proto-oncogene, transcription factor Homo sapiens 170-175 16616857-9 2006 Western blot analysis on thalidomide exposed cells showed a decrease in both Pim-1 protein expression and phosphorylated c-Myb protein expression, suggesting that the decrease in Pim-1 and the amount of phosphorylated c-Myb protein may be responsible for the observed decreases in c-Myb activity. Thalidomide 25-36 Pim-1 proto-oncogene, serine/threonine kinase Homo sapiens 77-82 16616857-9 2006 Western blot analysis on thalidomide exposed cells showed a decrease in both Pim-1 protein expression and phosphorylated c-Myb protein expression, suggesting that the decrease in Pim-1 and the amount of phosphorylated c-Myb protein may be responsible for the observed decreases in c-Myb activity. Thalidomide 25-36 MYB proto-oncogene, transcription factor Homo sapiens 121-126 16616857-9 2006 Western blot analysis on thalidomide exposed cells showed a decrease in both Pim-1 protein expression and phosphorylated c-Myb protein expression, suggesting that the decrease in Pim-1 and the amount of phosphorylated c-Myb protein may be responsible for the observed decreases in c-Myb activity. Thalidomide 25-36 Pim-1 proto-oncogene, serine/threonine kinase Homo sapiens 179-184 16616857-9 2006 Western blot analysis on thalidomide exposed cells showed a decrease in both Pim-1 protein expression and phosphorylated c-Myb protein expression, suggesting that the decrease in Pim-1 and the amount of phosphorylated c-Myb protein may be responsible for the observed decreases in c-Myb activity. Thalidomide 25-36 MYB proto-oncogene, transcription factor Homo sapiens 218-223 16616857-9 2006 Western blot analysis on thalidomide exposed cells showed a decrease in both Pim-1 protein expression and phosphorylated c-Myb protein expression, suggesting that the decrease in Pim-1 and the amount of phosphorylated c-Myb protein may be responsible for the observed decreases in c-Myb activity. Thalidomide 25-36 MYB proto-oncogene, transcription factor Homo sapiens 218-223 16616857-10 2006 Together these results demonstrate that thalidomide affects c-Myb signaling, in part, through increased ROS production. Thalidomide 40-51 MYB proto-oncogene, transcription factor Homo sapiens 60-65 16702985-9 2006 A noteworthy decrease in inflammatory and nociceptive responses caused by carrageenan was also observed when mice were previously treated with the preferential inhibitor of TNF-alpha synthesis, thalidomide. Thalidomide 194-205 tumor necrosis factor Mus musculus 173-182 16714224-0 2006 The neutropenia induced by the thalidomide analogue CC-4047 in patients with multiple myeloma is associated with an increased percentage of neutrophils bearing CD64. Thalidomide 31-42 Fc gamma receptor Ia Homo sapiens 160-164 17013434-2 2006 Thalidomide exerts its effects by decreasing circulating CD4 positive T-cells and stimulating CD8 positive T-cells, by increasing the number of Natural Killer cells and T-helper 2 cells. Thalidomide 0-11 CD8a molecule Homo sapiens 94-97 17013434-5 2006 It has been demonstrated that thalidomide inhibits TNFalpha, IL-5, IL-6, IL-8, IL-12 production and increases production of IL-2, IL-10 and INFgamma. Thalidomide 30-41 tumor necrosis factor Homo sapiens 51-59 17013434-5 2006 It has been demonstrated that thalidomide inhibits TNFalpha, IL-5, IL-6, IL-8, IL-12 production and increases production of IL-2, IL-10 and INFgamma. Thalidomide 30-41 interleukin 5 Homo sapiens 61-65 17013434-5 2006 It has been demonstrated that thalidomide inhibits TNFalpha, IL-5, IL-6, IL-8, IL-12 production and increases production of IL-2, IL-10 and INFgamma. Thalidomide 30-41 interleukin 6 Homo sapiens 67-71 17013434-5 2006 It has been demonstrated that thalidomide inhibits TNFalpha, IL-5, IL-6, IL-8, IL-12 production and increases production of IL-2, IL-10 and INFgamma. Thalidomide 30-41 C-X-C motif chemokine ligand 8 Homo sapiens 73-77 17013434-5 2006 It has been demonstrated that thalidomide inhibits TNFalpha, IL-5, IL-6, IL-8, IL-12 production and increases production of IL-2, IL-10 and INFgamma. Thalidomide 30-41 interleukin 2 Homo sapiens 124-128 17013434-5 2006 It has been demonstrated that thalidomide inhibits TNFalpha, IL-5, IL-6, IL-8, IL-12 production and increases production of IL-2, IL-10 and INFgamma. Thalidomide 30-41 interleukin 10 Homo sapiens 130-135 17013434-6 2006 Moreover thalidomide plays an important role in inhibition of VEGF and FGF-2 mediated angiogenesis. Thalidomide 9-20 vascular endothelial growth factor A Homo sapiens 62-66 17013434-6 2006 Moreover thalidomide plays an important role in inhibition of VEGF and FGF-2 mediated angiogenesis. Thalidomide 9-20 fibroblast growth factor 2 Homo sapiens 71-76 16775497-6 2006 Serum levels of vascular endothelial growth factor (VEGF) correlate with disease activity in human IBD and fall with the use of steroids, thalidomide, or infliximab. Thalidomide 138-149 vascular endothelial growth factor A Homo sapiens 16-50 16740776-0 2006 Importance of the stress kinase p38alpha in mediating the direct cytotoxic effects of the thalidomide analogue, CPS49, in cancer cells and endothelial cells. Thalidomide 90-101 mitogen-activated protein kinase 14 Homo sapiens 32-40 16775497-6 2006 Serum levels of vascular endothelial growth factor (VEGF) correlate with disease activity in human IBD and fall with the use of steroids, thalidomide, or infliximab. Thalidomide 138-149 vascular endothelial growth factor A Homo sapiens 52-56 16136308-6 2006 In this study, we investigated thalidomide"s potential to cause drug-drug interactions on the level of P-gp. Thalidomide 31-42 ATP binding cassette subfamily B member 1 Homo sapiens 103-107 16840205-1 2006 Thalidomide is a drug with anti-angiogenic, anti-inflammatory, immunomodulatory and anti-cancer properties that were found to inhibit the production of TNF-alpha in vitro, stimulate reactive oxygen species production, and inhibit VEGFR in acute leukemias. Thalidomide 0-11 tumor necrosis factor Homo sapiens 152-161 16840205-1 2006 Thalidomide is a drug with anti-angiogenic, anti-inflammatory, immunomodulatory and anti-cancer properties that were found to inhibit the production of TNF-alpha in vitro, stimulate reactive oxygen species production, and inhibit VEGFR in acute leukemias. Thalidomide 0-11 kinase insert domain receptor Homo sapiens 230-235 16136308-8 2006 A human leukaemia cell line over-expressing MDR1 (CCRF-CEM/MDR1) was used to measure uptake of rhodamine 123, a P-gp substrate, in the presence of thalidomide. Thalidomide 147-158 ATP binding cassette subfamily B member 1 Homo sapiens 44-48 16136308-8 2006 A human leukaemia cell line over-expressing MDR1 (CCRF-CEM/MDR1) was used to measure uptake of rhodamine 123, a P-gp substrate, in the presence of thalidomide. Thalidomide 147-158 ATP binding cassette subfamily B member 1 Homo sapiens 50-63 16136308-16 2006 Therefore, P-gp-related drug-drug interactions with thalidomide are not likely. Thalidomide 52-63 ATP binding cassette subfamily B member 1 Homo sapiens 11-15 16724931-2 2006 Recent clinical studies indicate that thalidomide, a known tumor necrosis factor-alpha inhibitor, ameliorated the toxicities induced by CPT-11. Thalidomide 38-49 tumor necrosis factor Rattus norvegicus 59-86 16680017-4 2006 When compared with DNBS-treated mice, thalidomide-treated (200 mg/kg orally starting 30 min after the administration of DNBS) mice subjected to DNBS-induced colitis experienced a significantly reduced rate of the extent and severity of the histological signs of colon injury associated with a significant reduction of plasma and colon tumor necrosis factor alpha levels. Thalidomide 38-49 tumor necrosis factor Mus musculus 335-362 16597438-3 2006 Furthermore, thalidomide pretreatment significantly reduced the concentration of tumor necrosis factor-alpha (TNF-alpha, -68.4%) in the exudate of zymosan-injected joints, but not those of interleukin-1beta, interleukin-6, CINC-1 or interleukin-10. Thalidomide 13-24 tumor necrosis factor Rattus norvegicus 81-108 16597438-3 2006 Furthermore, thalidomide pretreatment significantly reduced the concentration of tumor necrosis factor-alpha (TNF-alpha, -68.4%) in the exudate of zymosan-injected joints, but not those of interleukin-1beta, interleukin-6, CINC-1 or interleukin-10. Thalidomide 13-24 tumor necrosis factor Rattus norvegicus 110-119 16597438-3 2006 Furthermore, thalidomide pretreatment significantly reduced the concentration of tumor necrosis factor-alpha (TNF-alpha, -68.4%) in the exudate of zymosan-injected joints, but not those of interleukin-1beta, interleukin-6, CINC-1 or interleukin-10. Thalidomide 13-24 interleukin 6 Rattus norvegicus 208-221 16597438-3 2006 Furthermore, thalidomide pretreatment significantly reduced the concentration of tumor necrosis factor-alpha (TNF-alpha, -68.4%) in the exudate of zymosan-injected joints, but not those of interleukin-1beta, interleukin-6, CINC-1 or interleukin-10. Thalidomide 13-24 C-X-C motif chemokine ligand 1 Rattus norvegicus 223-229 16597438-3 2006 Furthermore, thalidomide pretreatment significantly reduced the concentration of tumor necrosis factor-alpha (TNF-alpha, -68.4%) in the exudate of zymosan-injected joints, but not those of interleukin-1beta, interleukin-6, CINC-1 or interleukin-10. Thalidomide 13-24 interleukin 10 Rattus norvegicus 233-247 16086096-9 2006 CONCLUSION: This study demonstrates antitumor activity of combination IFN-alpha/capecitabine/thalidomide in MRCC. Thalidomide 93-104 interferon alpha 1 Homo sapiens 70-79 16597438-4 2006 The expression of TNF-alpha, determined by immunohistochemical staining, in synovial tissues obtained from articular joints injected with zymosan was also inhibited by thalidomide pretreatment. Thalidomide 168-179 tumor necrosis factor Rattus norvegicus 18-27 16597438-6 2006 In conclusion, the anti-nociceptive activity of thalidomide in zymosan-induced articular incapacitation is associated with the inhibition of TNF-alpha by resident synovial cells. Thalidomide 48-59 tumor necrosis factor Rattus norvegicus 141-150 16604421-3 2006 Thalidomide, alpha-N-phthalidoglutarimide, (C(13)H(10)N(2))(4), has been shown to have immunomodulatory and anti-inflammatory properties, possibly mediated through its anti-TNF-alpha effect. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 173-182 16604421-8 2006 Thalidomide (100-800 nM) concentration-dependently inhibited NFkappaB transcriptional activity induced by TNF-alpha, including IKKalpha expression and IkappaBalpha phosphorylation in HSC-T6 cells. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 106-115 16604421-8 2006 Thalidomide (100-800 nM) concentration-dependently inhibited NFkappaB transcriptional activity induced by TNF-alpha, including IKKalpha expression and IkappaBalpha phosphorylation in HSC-T6 cells. Thalidomide 0-11 component of inhibitor of nuclear factor kappa B kinase complex Rattus norvegicus 127-135 16604421-8 2006 Thalidomide (100-800 nM) concentration-dependently inhibited NFkappaB transcriptional activity induced by TNF-alpha, including IKKalpha expression and IkappaBalpha phosphorylation in HSC-T6 cells. Thalidomide 0-11 NFKB inhibitor alpha Rattus norvegicus 151-163 16604421-9 2006 In addition, thalidomide also suppressed TGF-beta1-induced alpha-SMA expression and collagen deposition in HSC-T6 cells. Thalidomide 13-24 transforming growth factor, beta 1 Rattus norvegicus 41-50 16604421-9 2006 In addition, thalidomide also suppressed TGF-beta1-induced alpha-SMA expression and collagen deposition in HSC-T6 cells. Thalidomide 13-24 actin gamma 2, smooth muscle Rattus norvegicus 59-68 16604421-12 2006 Immunohistochemical double staining results showed that alpha-SMA- and NFkappaB-positive cells were decreased in the livers from DMN rats receiving either thalidomide or silymarin treatment. Thalidomide 155-166 actin gamma 2, smooth muscle Rattus norvegicus 56-65 16604421-13 2006 In addition, real-time PCR analysis indicated that hepatic mRNA expressions of TGF-beta1, alpha-SMA, collagen 1alpha2, TNF-alpha and iNOS genes were attenuated by thalidomide treatment. Thalidomide 163-174 transforming growth factor, beta 1 Rattus norvegicus 79-88 16604421-13 2006 In addition, real-time PCR analysis indicated that hepatic mRNA expressions of TGF-beta1, alpha-SMA, collagen 1alpha2, TNF-alpha and iNOS genes were attenuated by thalidomide treatment. Thalidomide 163-174 actin gamma 2, smooth muscle Rattus norvegicus 90-99 16604421-13 2006 In addition, real-time PCR analysis indicated that hepatic mRNA expressions of TGF-beta1, alpha-SMA, collagen 1alpha2, TNF-alpha and iNOS genes were attenuated by thalidomide treatment. Thalidomide 163-174 tumor necrosis factor Rattus norvegicus 119-128 16604421-14 2006 In conclusion, our results showed that thalidomide inhibited activation of HSC-T6 cells by TNF-alpha and ameliorated liver fibrosis in DMN-intoxicated rats. Thalidomide 39-50 tumor necrosis factor Rattus norvegicus 91-100 16680017-7 2006 In particular, we have observed that thalidomide treatment resulted in a significant reduction of the following: (1) the degree of colon injury, (2) the alteration of zonula occludens 1 and occludin localization (immunohistochemistry), and (3) intestinal permeability caused by DNBS in the colon. Thalidomide 37-48 occludin Mus musculus 190-198 16900926-3 2006 We therefore assessed the effect of adjunctive thalidomide, a tumor necrosis factor alpha-modulating drug, in intractable intracranial tuberculosis that did not respond to standard medical and surgical therapy. Thalidomide 47-58 tumor necrosis factor Homo sapiens 62-89 16310808-0 2006 Effect of thalidomide affecting VEGF secretion, cell migration, adhesion and capillary tube formation of human endothelial EA.hy 926 cells. Thalidomide 10-21 vascular endothelial growth factor A Homo sapiens 32-36 16310808-6 2006 Thalidomide at the concentrations of 0.01 microM and 10 microM inhibited VEGF secretion into supernatants, decreased the number of formed capillary tubes and increased cell adhesion to collagen. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 73-77 16310808-9 2006 We conclude that anti-angiogenic action of thalidomide is due to direct inhibitory action on VEGF secretion and capillary microvessel formation as well as immunomodulatory influence on EA.hy 926 cells migration and adhesion. Thalidomide 43-54 vascular endothelial growth factor A Homo sapiens 93-97 16475152-1 2006 BACKGROUND: The purpose of the study was to determine, in a Phase I/II study, the efficacy and safety profile of thalidomide with interleukin-2 (IL-2) in patients with metastatic renal cell carcinoma (MRCC). Thalidomide 113-124 interleukin 2 Homo sapiens 145-149 16523333-14 2006 In most patients, the BCL-2/BAX ratio was lower in cell cultures supplemented with mixture of lovastatin and thalidomide in comparison with cell cultures supplemented with lovastatin or thalidomide alone. Thalidomide 109-120 BCL2 apoptosis regulator Homo sapiens 22-27 16523333-14 2006 In most patients, the BCL-2/BAX ratio was lower in cell cultures supplemented with mixture of lovastatin and thalidomide in comparison with cell cultures supplemented with lovastatin or thalidomide alone. Thalidomide 109-120 BCL2 associated X, apoptosis regulator Homo sapiens 28-31 16523333-14 2006 In most patients, the BCL-2/BAX ratio was lower in cell cultures supplemented with mixture of lovastatin and thalidomide in comparison with cell cultures supplemented with lovastatin or thalidomide alone. Thalidomide 186-197 BCL2 apoptosis regulator Homo sapiens 22-27 16523333-14 2006 In most patients, the BCL-2/BAX ratio was lower in cell cultures supplemented with mixture of lovastatin and thalidomide in comparison with cell cultures supplemented with lovastatin or thalidomide alone. Thalidomide 186-197 BCL2 associated X, apoptosis regulator Homo sapiens 28-31 16386767-5 2006 A similar inhibition of the mechanical hypernociception was observed when treating mice with the TNFalpha synthesis inhibitor thalidomide (50 mg/kg, s.c.), either at the time of the surgery or 4 days after. Thalidomide 126-137 tumor necrosis factor Mus musculus 97-105 16219350-1 2006 We investigated the therapeutic activity of recombinant erythropoietin (r-EPO) in association with thalidomide in 30 patients with myelodysplastic syndromes (MDS), previously treated with r-EPO (n.15, group A) or thalidomide (n.15, group B) as single agents, respectively, without any significant benefit on their anemia. Thalidomide 99-110 erythropoietin Homo sapiens 56-70 16729912-1 2006 PURPOSE: The antiangiogenic and immunomodulatory effects of thalidomide induce responses in patients with androgen-independent prostate cancer (AIPC). Thalidomide 60-71 PDZ domain containing 2 Homo sapiens 144-148 16729912-19 2006 CONCLUSION: The combined dosing of paclitaxel (100 mg/m(2) weekly), doxorubicin (20 mg/m(2) weekly), and thalidomide (300 mg daily) is tolerated by men with AIPC and merits continued phase II study. Thalidomide 105-116 PDZ domain containing 2 Homo sapiens 157-161 16514283-6 2006 Cytokine study showed that the effect of thalidomide was not dependent on IL-12; however, it up-regulated IFN-gamma and down-regulated IL-10 production. Thalidomide 41-52 interferon gamma Mus musculus 106-115 16531263-0 2006 Stimulation of erythropoiesis by thalidomide in multiple myeloma patients: its influence on FasL, TRAIL and their receptors on erythroblasts. Thalidomide 33-44 Fas ligand Homo sapiens 92-96 16531263-0 2006 Stimulation of erythropoiesis by thalidomide in multiple myeloma patients: its influence on FasL, TRAIL and their receptors on erythroblasts. Thalidomide 33-44 TNF superfamily member 10 Homo sapiens 98-103 16531263-3 2006 The expression of FasL, Fas, TNF-related apoptosis-inducing ligand (TRAIL) and TRAIL-R1 on erythroblasts decreased significantly during thalidomide treatment. Thalidomide 136-147 Fas ligand Homo sapiens 18-22 16531263-3 2006 The expression of FasL, Fas, TNF-related apoptosis-inducing ligand (TRAIL) and TRAIL-R1 on erythroblasts decreased significantly during thalidomide treatment. Thalidomide 136-147 TNF superfamily member 10 Homo sapiens 29-66 16531263-3 2006 The expression of FasL, Fas, TNF-related apoptosis-inducing ligand (TRAIL) and TRAIL-R1 on erythroblasts decreased significantly during thalidomide treatment. Thalidomide 136-147 TNF superfamily member 10 Homo sapiens 68-73 16531263-3 2006 The expression of FasL, Fas, TNF-related apoptosis-inducing ligand (TRAIL) and TRAIL-R1 on erythroblasts decreased significantly during thalidomide treatment. Thalidomide 136-147 TNF receptor superfamily member 10a Homo sapiens 79-87 16531263-4 2006 Additional in vitro studies revealed that the apoptosis of erythroblasts and the expression of FasL, TRAIL, TRAIL-R1 and TRAIL-R2 was lower in cultures with thalidomide than in control cultures. Thalidomide 157-168 Fas ligand Homo sapiens 95-99 16531263-4 2006 Additional in vitro studies revealed that the apoptosis of erythroblasts and the expression of FasL, TRAIL, TRAIL-R1 and TRAIL-R2 was lower in cultures with thalidomide than in control cultures. Thalidomide 157-168 TNF superfamily member 10 Homo sapiens 101-106 16531263-4 2006 Additional in vitro studies revealed that the apoptosis of erythroblasts and the expression of FasL, TRAIL, TRAIL-R1 and TRAIL-R2 was lower in cultures with thalidomide than in control cultures. Thalidomide 157-168 TNF receptor superfamily member 10a Homo sapiens 108-116 16531263-4 2006 Additional in vitro studies revealed that the apoptosis of erythroblasts and the expression of FasL, TRAIL, TRAIL-R1 and TRAIL-R2 was lower in cultures with thalidomide than in control cultures. Thalidomide 157-168 TNF receptor superfamily member 10b Homo sapiens 121-129 16510725-5 2006 We tested the neuroprotective effect of thalidomide and its analog lenalidomide, pharmacological agents that inhibit the expression of TNF-alpha and other cytokines by destabilizing their mRNA. Thalidomide 40-51 tumor necrosis factor Mus musculus 135-144 16514283-6 2006 Cytokine study showed that the effect of thalidomide was not dependent on IL-12; however, it up-regulated IFN-gamma and down-regulated IL-10 production. Thalidomide 41-52 interleukin 10 Mus musculus 135-140 16490847-2 2006 Accordingly, TNF-alpha inhibitors, such as thalidomide, infliximab (Remicade), adalimumab (Humira), and etanercept (Enbrel), have been used with success in the treatment of autoimmune disorders, including psoriasis, rheumatoid arthritis, inflammatory bowel diseases, and lymphoproliferative disorders. Thalidomide 43-54 tumor necrosis factor Homo sapiens 13-22 16421420-5 2006 RESULTS: Treatment with temozolomide and thalidomide was associated with an objective biochemical (chromogranin A) response rate of 40%, and a radiologic response rate of 25% (45% among pancreatic endocrine tumors, 33% among pheochromocytomas, and 7% among carcinoid tumors). Thalidomide 41-52 chromogranin A Homo sapiens 99-113 16484869-4 2006 Thalidomide has been shown to have antiangiogenic and immunomodulatory effects, including the inhibition of vascular endothelial growth factor, basic fibroblast growth factor and tumor necrosis factor alpha. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 108-142 16454849-1 2006 Immunomodulation of thalidomide is represented by the antiinflammatory effect through inhibition of tumor necrosis factor alpha and costimulatory effect on human CD8+ T cells. Thalidomide 20-31 tumor necrosis factor Homo sapiens 100-127 16454849-4 2006 Tumor necrosis factor-alpha in vitro production in mononuclear cells decreased with thalidomide in all the subjects (p = 0.028). Thalidomide 84-95 tumor necrosis factor Homo sapiens 0-27 16454849-9 2006 In conclusion, thalidomide was able to reduce liver enzymes in six out of eight patients with chronic hepatitis C and to reduce tumor necrosis factor alpha production, representing a promising new approach for the treatment of HCV infection. Thalidomide 15-26 tumor necrosis factor Homo sapiens 128-155 16327979-8 2006 We further demonstrated that the expression of angiogenic factors, such as vascular endothelial cell growth factor (VEGF) and basic fibroblastic growth factor (bFGF), were inhibited by THD. Thalidomide 185-188 vascular endothelial growth factor A Rattus norvegicus 116-120 16344495-7 2006 B cell numbers and expression of the adhesion molecule L-sel also declined with thalidomide. Thalidomide 80-91 selectin L Homo sapiens 55-60 16997792-0 2006 Effect of thalidomide on the expression of TNF-alpha m-RNA and synthesis of TNF-alpha in cells from leprosy patients with reversal reaction. Thalidomide 10-21 tumor necrosis factor Homo sapiens 43-52 16997792-0 2006 Effect of thalidomide on the expression of TNF-alpha m-RNA and synthesis of TNF-alpha in cells from leprosy patients with reversal reaction. Thalidomide 10-21 tumor necrosis factor Homo sapiens 76-85 16997792-5 2006 We assessed thalidomide"s effect on TNF-alpha in RR. Thalidomide 12-23 tumor necrosis factor Homo sapiens 36-45 16997792-7 2006 Thalidomide suppressed TNF-alpha, but when some RR patients" cells were stimulated with AFB, it enhanced TNF-alpha. Thalidomide 0-11 tumor necrosis factor Homo sapiens 23-32 16327979-8 2006 We further demonstrated that the expression of angiogenic factors, such as vascular endothelial cell growth factor (VEGF) and basic fibroblastic growth factor (bFGF), were inhibited by THD. Thalidomide 185-188 fibroblast growth factor 2 Rattus norvegicus 126-158 16327979-8 2006 We further demonstrated that the expression of angiogenic factors, such as vascular endothelial cell growth factor (VEGF) and basic fibroblastic growth factor (bFGF), were inhibited by THD. Thalidomide 185-188 fibroblast growth factor 2 Rattus norvegicus 160-164 16943688-1 2006 BACKGROUND/AIMS: Thalidomide inhibited tumor necrosis factor-alpha (TNF-alpha) effectively in many trials. Thalidomide 17-28 tumor necrosis factor Rattus norvegicus 39-66 17047296-3 2006 Liver histopathology and immunohistochemistry were significantly improved and the expressions of ICAM-1, VCAM-1, E-selectin, and TNF-alpha mRNA and protein were decreased significantly in rats treated with a high dose of thalidomide. Thalidomide 221-232 intercellular adhesion molecule 1 Rattus norvegicus 97-103 17047296-3 2006 Liver histopathology and immunohistochemistry were significantly improved and the expressions of ICAM-1, VCAM-1, E-selectin, and TNF-alpha mRNA and protein were decreased significantly in rats treated with a high dose of thalidomide. Thalidomide 221-232 vascular cell adhesion molecule 1 Rattus norvegicus 105-111 17047296-3 2006 Liver histopathology and immunohistochemistry were significantly improved and the expressions of ICAM-1, VCAM-1, E-selectin, and TNF-alpha mRNA and protein were decreased significantly in rats treated with a high dose of thalidomide. Thalidomide 221-232 selectin E Rattus norvegicus 113-123 17047296-3 2006 Liver histopathology and immunohistochemistry were significantly improved and the expressions of ICAM-1, VCAM-1, E-selectin, and TNF-alpha mRNA and protein were decreased significantly in rats treated with a high dose of thalidomide. Thalidomide 221-232 tumor necrosis factor Rattus norvegicus 129-138 17047296-5 2006 These results indicate that thalidomide exerts its effect on the downregulation of adhesion molecules via TNF-alpha signaling pathway to inhibit liver fibrosis. Thalidomide 28-39 tumor necrosis factor Rattus norvegicus 106-115 16943688-0 2006 Thalidomide in rat liver cirrhosis: blockade of tumor necrosis factor-alpha via inhibition of degradation of an inhibitor of nuclear factor-kappaB. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 48-75 16943688-1 2006 BACKGROUND/AIMS: Thalidomide inhibited tumor necrosis factor-alpha (TNF-alpha) effectively in many trials. Thalidomide 17-28 tumor necrosis factor Rattus norvegicus 68-77 16943688-7 2006 CONCLUSION: Thalidomide downregulates NF-kappaB-induced TNF-alpha and activates hepatic stellate cells (HSC) via inhibition of IkappaB degradation to prevent liver cirrhosis. Thalidomide 12-23 tumor necrosis factor Rattus norvegicus 56-65 16299286-4 2005 Intraperitoneal injection of a TNF inhibitor, thalidomide (TH), reduced the number of exfoliated chambers, while indomethacin had no effect. Thalidomide 46-57 tumor necrosis factor Mus musculus 31-34 16324117-4 2005 The suppression of LTP by Abeta was absent in mutant mice null for TNF receptor type 1 (TNF-R1) and was prevented by the inhibitors of TNFalpha, infliximab and TNF peptide antagonist, and by the inhibitor of TNFalpha production, thalidomide. Thalidomide 229-240 tumor necrosis factor Mus musculus 208-216 16299286-4 2005 Intraperitoneal injection of a TNF inhibitor, thalidomide (TH), reduced the number of exfoliated chambers, while indomethacin had no effect. Thalidomide 59-61 tumor necrosis factor Mus musculus 31-34 16301340-5 2005 (3) Thalidomide also induced a decrease in total neutrophil count and an increase in plasma levels of tumor necrosis factor-alpha, suggesting both proinflammatory and antiinflammatory effects. Thalidomide 4-15 tumor necrosis factor Homo sapiens 102-129 16051743-2 2005 Thalidomide is a first-generation immuno-modulating agent that down-regulates TNF-alpha and VEGF. Thalidomide 0-11 tumor necrosis factor Homo sapiens 78-87 16051743-2 2005 Thalidomide is a first-generation immuno-modulating agent that down-regulates TNF-alpha and VEGF. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 92-96 16249352-0 2005 Thalidomide in advanced hepatocellular carcinoma with optional low-dose interferon-alpha2a upon progression. Thalidomide 0-11 interferon alpha 2 Homo sapiens 72-90 16039549-8 2005 A similar inhibition of B(1) receptor-mediated paw edema was observed when mice were treated with thalidomide (30 mg/kg, s.c.) [corrected] a drug known for reducing TNFalpha synthesis, 5 min prior to LPS administration. Thalidomide 98-109 tumor necrosis factor Mus musculus 165-173 16288038-3 2005 We examined whether thalidomide is able to suppress the expression of uPAR mRNA and protein in human ovarian cancer cell line HRA and human chondrosarcoma cell line HCS-2/8. Thalidomide 20-31 plasminogen activator, urokinase receptor Homo sapiens 70-74 16288038-7 2005 We conclude that thalidomide down-regulates constitutive and TGF-beta1-stimulated uPAR mRNA and protein expression possibly through suppression of NF-kappaB activation. Thalidomide 17-28 transforming growth factor beta 1 Homo sapiens 61-70 16288038-7 2005 We conclude that thalidomide down-regulates constitutive and TGF-beta1-stimulated uPAR mRNA and protein expression possibly through suppression of NF-kappaB activation. Thalidomide 17-28 plasminogen activator, urokinase receptor Homo sapiens 82-86 16288038-7 2005 We conclude that thalidomide down-regulates constitutive and TGF-beta1-stimulated uPAR mRNA and protein expression possibly through suppression of NF-kappaB activation. Thalidomide 17-28 nuclear factor kappa B subunit 1 Homo sapiens 147-156 16079895-0 2005 The combination of intermediate doses of thalidomide with dexamethasone is an effective treatment for patients with refractory/relapsed multiple myeloma and normalizes abnormal bone remodeling, through the reduction of sRANKL/osteoprotegerin ratio. Thalidomide 41-52 TNF receptor superfamily member 11b Homo sapiens 226-241 16079895-6 2005 Thal/Dex administration resulted in a significant reduction of sRANKL/OPG ratio, and bone resorption. Thalidomide 0-4 TNF receptor superfamily member 11b Homo sapiens 70-73 16079895-13 2005 These results suggest that the combination of intermediate dose of Thal/Dex is effective in patients with refractory/relapsed myeloma and improves abnormal bone remodeling through the reduction of sRANKL/OPG ratio. Thalidomide 67-71 TNF receptor superfamily member 11b Homo sapiens 204-207 16241859-3 2005 In the present study, we investigated the effects of one of the IMiDs, CC-4047 (Actimid, Celgene, Warren, NJ), on synthesis of IL-2 protein and mRNA and on the activity and expression of transcription factors. Thalidomide 89-96 interleukin 2 Homo sapiens 127-131 16086360-4 2005 SALL4 mutations may also cause acro-renal-ocular syndrome (AROS), which differs from DRRS by the presence of structural eye anomalies, and phenotypes similar to thalidomide embryopathy and Holt-Oram syndrome (HOS). Thalidomide 161-172 spalt like transcription factor 4 Homo sapiens 0-5 16344099-5 2005 First established as agents with antiangiogenic properties, thalidomide and IMiDs inhibit the production of interleukin (IL)-6, which is a growth factor for the proliferation of myeloma cells. Thalidomide 60-71 interleukin 6 Homo sapiens 108-126 16091059-0 2005 alpha-fetoprotein response predicts survival benefits of thalidomide in advanced hepatocellular carcinoma. Thalidomide 57-68 alpha fetoprotein Homo sapiens 0-17 18360564-0 2005 Therapeutic effects of thalidomide in myeloma are associated with the expression of fibroblast growth factor receptor 3. Thalidomide 23-34 fibroblast growth factor receptor 3 Homo sapiens 84-119 18360564-5 2005 FGFR3 transfected U266 clones revealed increased FGFR3 expression, but resulted in decreased DNA synthesis and increased apoptosis under Thal treatment. Thalidomide 137-141 fibroblast growth factor receptor 3 Homo sapiens 0-5 16091059-2 2005 AIMS: To evaluate the clinical implications of tumour marker (alpha-fetoprotein) response in advanced hepatocellular carcinoma patients with thalidomide treatment. Thalidomide 141-152 alpha fetoprotein Homo sapiens 62-79 16091059-10 2005 CONCLUSION: alpha-fetoprotein response can more accurately reflect the biological response of advanced hepatocellular carcinoma to thalidomide therapy than radiographic response. Thalidomide 131-142 alpha fetoprotein Homo sapiens 12-29 16061867-1 2005 Preclinical data indicates that cyclooxygenase-2 (COX-2) inhibition impairs plasma cell growth and potentially synergizes with thalidomide. Thalidomide 127-138 prostaglandin-endoperoxide synthase 2 Homo sapiens 32-48 16061867-1 2005 Preclinical data indicates that cyclooxygenase-2 (COX-2) inhibition impairs plasma cell growth and potentially synergizes with thalidomide. Thalidomide 127-138 prostaglandin-endoperoxide synthase 2 Homo sapiens 50-55 16061867-12 2005 Future studies should use newer COX-2 inhibitors with thalidomide, or their respective derivatives. Thalidomide 54-65 prostaglandin-endoperoxide synthase 2 Homo sapiens 32-37 15789431-6 2005 Furthermore, the effect of LPS was blocked by thalidomide, an inhibitor of TNF-alpha synthesis. Thalidomide 46-57 tumor necrosis factor Rattus norvegicus 75-84 15982930-7 2005 Thalidomide reduced COX-2 expression accompanied by a decrease of bcl-2 protein, TNFalpha, VEGF, GSH and an increased cytochrome c, but had no effect on that of COX-1, in MCF-7 and HL-60. Thalidomide 0-11 mitochondrially encoded cytochrome c oxidase II Homo sapiens 20-25 16098027-5 2005 We also measured clearly reduced levels of tumor necrosis factor-alpha in the thalidomide-treated group. Thalidomide 78-89 tumor necrosis factor Homo sapiens 43-70 15920492-0 2005 Long-term thalidomide therapy resulted in lack of mdr1 gene expression in a patient with primary resistant multiple myeloma. Thalidomide 10-21 ATP binding cassette subfamily B member 1 Homo sapiens 50-54 15982930-0 2005 Thalidomide inhibits growth of tumors through COX-2 degradation independent of antiangiogenesis. Thalidomide 0-11 mitochondrially encoded cytochrome c oxidase II Homo sapiens 46-51 15982930-7 2005 Thalidomide reduced COX-2 expression accompanied by a decrease of bcl-2 protein, TNFalpha, VEGF, GSH and an increased cytochrome c, but had no effect on that of COX-1, in MCF-7 and HL-60. Thalidomide 0-11 BCL2 apoptosis regulator Homo sapiens 66-71 15982930-7 2005 Thalidomide reduced COX-2 expression accompanied by a decrease of bcl-2 protein, TNFalpha, VEGF, GSH and an increased cytochrome c, but had no effect on that of COX-1, in MCF-7 and HL-60. Thalidomide 0-11 tumor necrosis factor Homo sapiens 81-89 15982930-7 2005 Thalidomide reduced COX-2 expression accompanied by a decrease of bcl-2 protein, TNFalpha, VEGF, GSH and an increased cytochrome c, but had no effect on that of COX-1, in MCF-7 and HL-60. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 91-95 15982930-7 2005 Thalidomide reduced COX-2 expression accompanied by a decrease of bcl-2 protein, TNFalpha, VEGF, GSH and an increased cytochrome c, but had no effect on that of COX-1, in MCF-7 and HL-60. Thalidomide 0-11 cytochrome c, somatic Homo sapiens 118-130 15982930-9 2005 In our mouse xenograft model of OVCAR-3 and HCT-8, we found that thalidomide could decrease intratumoral microvessel density in both tumors; it exerted antitumor effects only on OVCAR-3 expressing COX-2 but did not on HCT-8 not expressing COX-2. Thalidomide 65-76 cytochrome c oxidase II, mitochondrial Mus musculus 197-202 15982930-9 2005 In our mouse xenograft model of OVCAR-3 and HCT-8, we found that thalidomide could decrease intratumoral microvessel density in both tumors; it exerted antitumor effects only on OVCAR-3 expressing COX-2 but did not on HCT-8 not expressing COX-2. Thalidomide 65-76 cytochrome c oxidase II, mitochondrial Mus musculus 239-244 15982930-10 2005 Effect of thalidomide on COX-1 and COX-2 in vivo was consistent with that of in vitro. Thalidomide 10-21 mitochondrially encoded cytochrome c oxidase I Homo sapiens 25-30 15982930-10 2005 Effect of thalidomide on COX-1 and COX-2 in vivo was consistent with that of in vitro. Thalidomide 10-21 mitochondrially encoded cytochrome c oxidase II Homo sapiens 35-40 15982930-11 2005 These results demonstrated that thalidomide might inhibit growth of tumors through COX-2 degradation independent of antiangiogenesis. Thalidomide 32-43 mitochondrially encoded cytochrome c oxidase II Homo sapiens 83-88 15986355-8 2005 Interestingly, thalidomide and NF-kappaB small interfering RNA down-regulated cFLIP and Bcl-x(L) expression levels and sensitized BD activated T cells to CD95-induced apoptosis. Thalidomide 15-26 BCL2 like 1 Homo sapiens 88-96 15741222-0 2005 Thalidomide-induced antiangiogenic action is mediated by ceramide through depletion of VEGF receptors, and is antagonized by sphingosine-1-phosphate. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 87-91 15741222-3 2005 Thalidomide induces the defect of major blood vessels, which is demonstrated by their morphologic loss and confirmed by the depletion of vascular endothelial growth factor (VEGF) receptors such as neuropilin-1 and Flk-1. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 137-171 15741222-3 2005 Thalidomide induces the defect of major blood vessels, which is demonstrated by their morphologic loss and confirmed by the depletion of vascular endothelial growth factor (VEGF) receptors such as neuropilin-1 and Flk-1. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 173-177 15741222-3 2005 Thalidomide induces the defect of major blood vessels, which is demonstrated by their morphologic loss and confirmed by the depletion of vascular endothelial growth factor (VEGF) receptors such as neuropilin-1 and Flk-1. Thalidomide 0-11 neuropilin 1 Homo sapiens 197-209 15741222-3 2005 Thalidomide induces the defect of major blood vessels, which is demonstrated by their morphologic loss and confirmed by the depletion of vascular endothelial growth factor (VEGF) receptors such as neuropilin-1 and Flk-1. Thalidomide 0-11 kinase insert domain receptor Homo sapiens 214-219 15741222-4 2005 Transient increase of ceramide content through activation of neutral sphingomyelinase (nSMase) precedes thalidomide-induced vascular defect in the embryos. Thalidomide 104-115 sphingomyelin phosphodiesterase 2 Homo sapiens 61-85 15741222-4 2005 Transient increase of ceramide content through activation of neutral sphingomyelinase (nSMase) precedes thalidomide-induced vascular defect in the embryos. Thalidomide 104-115 sphingomyelin phosphodiesterase 2 Homo sapiens 87-93 15741222-6 2005 The blockade of ceramide generation by antisense morpholino oligonucleotides for nSMase prevents thalidomide-induced ceramide generation and vascular defect. Thalidomide 97-108 sphingomyelin phosphodiesterase 2 Homo sapiens 81-87 15741222-7 2005 In contrast to ceramide, sphingosine-1-phosphate (S1P) inhibits nSMase-dependent ceramide generation and restores thalidomide-induced embryonic vascular defect with an increase of expression of VEGF receptors. Thalidomide 114-125 vascular endothelial growth factor A Homo sapiens 194-198 15741222-8 2005 In human umbilical vein endothelial cells (HUVECs), thalidomide-induced inhibition of cell growth, generation of ceramide through nSMase, and depletion of VEGF receptors are restored to the control levels by pretreatment with S1P. Thalidomide 52-63 sphingomyelin phosphodiesterase 2 Homo sapiens 130-136 15741222-8 2005 In human umbilical vein endothelial cells (HUVECs), thalidomide-induced inhibition of cell growth, generation of ceramide through nSMase, and depletion of VEGF receptors are restored to the control levels by pretreatment with S1P. Thalidomide 52-63 vascular endothelial growth factor A Homo sapiens 155-159 16297005-4 2005 Opiate receptor antagonists, antidepressants, antiepileptics, and thalidomide are currently available therapeutic options that have benefited many patients with variegated sources of itch. Thalidomide 66-77 itchy E3 ubiquitin protein ligase Homo sapiens 183-187 15986355-8 2005 Interestingly, thalidomide and NF-kappaB small interfering RNA down-regulated cFLIP and Bcl-x(L) expression levels and sensitized BD activated T cells to CD95-induced apoptosis. Thalidomide 15-26 Fas cell surface death receptor Homo sapiens 154-158 16019550-1 2005 Thalidomide is thought to have anti-angiogenic and immunomodulatory properties, including suppression of tumor necrosis factor-alpha, effects on interleukins and interferons, down-regulation of some cell adhesion molecules, and changes in the proportion of lymphocyte subsets. Thalidomide 0-11 tumor necrosis factor Homo sapiens 105-132 16019550-11 2005 Although the CD4/CD8 ratio declined from 1.6 to 1.0 during 3 months of thalidomide treatment, this had no statistical significance (P = 0.1). Thalidomide 71-82 CD4 molecule Homo sapiens 13-16 16019550-11 2005 Although the CD4/CD8 ratio declined from 1.6 to 1.0 during 3 months of thalidomide treatment, this had no statistical significance (P = 0.1). Thalidomide 71-82 CD8a molecule Homo sapiens 17-20 15817259-3 2005 It is recognised that thalidomide decreases TNF production and may modulate several functions of the immune system. Thalidomide 22-33 tumor necrosis factor Mus musculus 44-47 16078585-6 2005 RESULTS: With respect to TNBS model, in the control, prednisone-treatment and thalidomide-treatment groups, the macroscopic and microscopic scores were 6.33 +/- 1.03, 1.67 +/- 0.82, 2.00 +/- 0.89 and 7.33 +/- 1.03, 2.67s +/- 0.82, 3.17 +/- 0.75 respectively; the expression levels of NF-kappaB P65 and TNF-alpha mRNA in the three groups were 62.45 +/- 12.38, 23.62 +/- 8.54, 34.18 +/- 9.65 and 12.42 +/- 4.63, 9.86 +/- 3.29, 4.35 +/- 1.74 respectively; the levels of TNF-alpha, IL-4, IFN-gamma were 540.32 +/- 80.76, 94.58 +/- 14.45, 486.18 +/- 68.47; 396.53 +/- 92.42, 78.45 +/- 12.69, 347.56 +/- 82.94; and 385.68 +/- 88.57, 123.68 +/- 38.15, 378.27 +/- 90.65 respectively. Thalidomide 78-89 tumor necrosis factor Rattus norvegicus 302-311 16078585-6 2005 RESULTS: With respect to TNBS model, in the control, prednisone-treatment and thalidomide-treatment groups, the macroscopic and microscopic scores were 6.33 +/- 1.03, 1.67 +/- 0.82, 2.00 +/- 0.89 and 7.33 +/- 1.03, 2.67s +/- 0.82, 3.17 +/- 0.75 respectively; the expression levels of NF-kappaB P65 and TNF-alpha mRNA in the three groups were 62.45 +/- 12.38, 23.62 +/- 8.54, 34.18 +/- 9.65 and 12.42 +/- 4.63, 9.86 +/- 3.29, 4.35 +/- 1.74 respectively; the levels of TNF-alpha, IL-4, IFN-gamma were 540.32 +/- 80.76, 94.58 +/- 14.45, 486.18 +/- 68.47; 396.53 +/- 92.42, 78.45 +/- 12.69, 347.56 +/- 82.94; and 385.68 +/- 88.57, 123.68 +/- 38.15, 378.27 +/- 90.65 respectively. Thalidomide 78-89 tumor necrosis factor Rattus norvegicus 467-476 16078585-6 2005 RESULTS: With respect to TNBS model, in the control, prednisone-treatment and thalidomide-treatment groups, the macroscopic and microscopic scores were 6.33 +/- 1.03, 1.67 +/- 0.82, 2.00 +/- 0.89 and 7.33 +/- 1.03, 2.67s +/- 0.82, 3.17 +/- 0.75 respectively; the expression levels of NF-kappaB P65 and TNF-alpha mRNA in the three groups were 62.45 +/- 12.38, 23.62 +/- 8.54, 34.18 +/- 9.65 and 12.42 +/- 4.63, 9.86 +/- 3.29, 4.35 +/- 1.74 respectively; the levels of TNF-alpha, IL-4, IFN-gamma were 540.32 +/- 80.76, 94.58 +/- 14.45, 486.18 +/- 68.47; 396.53 +/- 92.42, 78.45 +/- 12.69, 347.56 +/- 82.94; and 385.68 +/- 88.57, 123.68 +/- 38.15, 378.27 +/- 90.65 respectively. Thalidomide 78-89 interleukin 4 Rattus norvegicus 478-482 16078585-6 2005 RESULTS: With respect to TNBS model, in the control, prednisone-treatment and thalidomide-treatment groups, the macroscopic and microscopic scores were 6.33 +/- 1.03, 1.67 +/- 0.82, 2.00 +/- 0.89 and 7.33 +/- 1.03, 2.67s +/- 0.82, 3.17 +/- 0.75 respectively; the expression levels of NF-kappaB P65 and TNF-alpha mRNA in the three groups were 62.45 +/- 12.38, 23.62 +/- 8.54, 34.18 +/- 9.65 and 12.42 +/- 4.63, 9.86 +/- 3.29, 4.35 +/- 1.74 respectively; the levels of TNF-alpha, IL-4, IFN-gamma were 540.32 +/- 80.76, 94.58 +/- 14.45, 486.18 +/- 68.47; 396.53 +/- 92.42, 78.45 +/- 12.69, 347.56 +/- 82.94; and 385.68 +/- 88.57, 123.68 +/- 38.15, 378.27 +/- 90.65 respectively. Thalidomide 78-89 interferon gamma Rattus norvegicus 484-493 16078585-7 2005 The results indicated that thalidomide treatment significantly reduced colonic inflammation, suppressed NF-kappaB activation,enhanced TNF-alpha mRNA degradation, inhibited the synthesis of the TNF-alpha, IEN-gamma and increased the production of IL-4. Thalidomide 27-38 tumor necrosis factor Rattus norvegicus 134-143 16078585-7 2005 The results indicated that thalidomide treatment significantly reduced colonic inflammation, suppressed NF-kappaB activation,enhanced TNF-alpha mRNA degradation, inhibited the synthesis of the TNF-alpha, IEN-gamma and increased the production of IL-4. Thalidomide 27-38 tumor necrosis factor Rattus norvegicus 193-202 16078585-7 2005 The results indicated that thalidomide treatment significantly reduced colonic inflammation, suppressed NF-kappaB activation,enhanced TNF-alpha mRNA degradation, inhibited the synthesis of the TNF-alpha, IEN-gamma and increased the production of IL-4. Thalidomide 27-38 interleukin 4 Rattus norvegicus 246-250 15797874-7 2005 Several inhibitors of the NF-kappaB signaling pathway, including dexamethasone, thalidomide, and a proteasome inhibitor, bortezomib, showed inhibitory effects on LPS-induced GADD45beta expression as indicated by a decrease of the luciferase activity. Thalidomide 80-91 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 26-35 15797874-7 2005 Several inhibitors of the NF-kappaB signaling pathway, including dexamethasone, thalidomide, and a proteasome inhibitor, bortezomib, showed inhibitory effects on LPS-induced GADD45beta expression as indicated by a decrease of the luciferase activity. Thalidomide 80-91 growth arrest and DNA-damage-inducible 45 beta Mus musculus 174-184 15897810-12 2005 Thalidomide also caused a substantial reduction of the rise in myeloperoxidase activity (mucosa), in the increase in the tissue levels of TNF-alpha, IL-1beta, and VEGF, in the increase in staining (immunohistochemistry) for nitrotyrosine and for poly (ADP ribose), as well as in the upregulation of intercellular adhesion molecule-1 caused by DNBS in the colon. Thalidomide 0-11 myeloperoxidase Mus musculus 63-78 15897810-12 2005 Thalidomide also caused a substantial reduction of the rise in myeloperoxidase activity (mucosa), in the increase in the tissue levels of TNF-alpha, IL-1beta, and VEGF, in the increase in staining (immunohistochemistry) for nitrotyrosine and for poly (ADP ribose), as well as in the upregulation of intercellular adhesion molecule-1 caused by DNBS in the colon. Thalidomide 0-11 tumor necrosis factor Mus musculus 138-147 15897810-12 2005 Thalidomide also caused a substantial reduction of the rise in myeloperoxidase activity (mucosa), in the increase in the tissue levels of TNF-alpha, IL-1beta, and VEGF, in the increase in staining (immunohistochemistry) for nitrotyrosine and for poly (ADP ribose), as well as in the upregulation of intercellular adhesion molecule-1 caused by DNBS in the colon. Thalidomide 0-11 interleukin 1 beta Mus musculus 149-157 15897810-12 2005 Thalidomide also caused a substantial reduction of the rise in myeloperoxidase activity (mucosa), in the increase in the tissue levels of TNF-alpha, IL-1beta, and VEGF, in the increase in staining (immunohistochemistry) for nitrotyrosine and for poly (ADP ribose), as well as in the upregulation of intercellular adhesion molecule-1 caused by DNBS in the colon. Thalidomide 0-11 vascular endothelial growth factor A Mus musculus 163-167 15897810-12 2005 Thalidomide also caused a substantial reduction of the rise in myeloperoxidase activity (mucosa), in the increase in the tissue levels of TNF-alpha, IL-1beta, and VEGF, in the increase in staining (immunohistochemistry) for nitrotyrosine and for poly (ADP ribose), as well as in the upregulation of intercellular adhesion molecule-1 caused by DNBS in the colon. Thalidomide 0-11 intercellular adhesion molecule 1 Mus musculus 299-332 15915823-9 2005 Agents that inhibit NF-kappaB activation, such as resveratrol, thalidomide, ibuprofen, eicosapentaenoic acid, and beta-hydroxy-beta-methylbutyrate, are effective in the preservation of skeletal muscle mass in cachexia. Thalidomide 63-74 nuclear factor kappa B subunit 1 Homo sapiens 20-29 15687330-0 2005 Thalidomide inhibits tumor necrosis factor-alpha-induced interleukin-8 expression in endometriotic stromal cells, possibly through suppression of nuclear factor-kappaB activation. Thalidomide 0-11 C-X-C motif chemokine ligand 8 Homo sapiens 57-70 16146335-4 2005 Thalidomide and IMiDs inhibit the cytokines tumor necrosis factor-alpha (TNF-alpha), interleukins (IL) 1beta, 6, 12, and granulocyte macrophage-colony stimulating factor (GM-CSF). Thalidomide 0-11 colony stimulating factor 2 Homo sapiens 171-177 15618473-7 2005 In vitro exposure of ECs to thalidomide or its derivative CC-5013 inhibited gene expression of the receptors for transforming growth factor-beta and thrombin. Thalidomide 28-39 transforming growth factor beta 1 Homo sapiens 113-144 15618473-7 2005 In vitro exposure of ECs to thalidomide or its derivative CC-5013 inhibited gene expression of the receptors for transforming growth factor-beta and thrombin. Thalidomide 28-39 coagulation factor II, thrombin Homo sapiens 149-157 16536004-5 2005 Corticosteroids are still the first-line treatment, but alternative therapy with anti-TNF agents, like pentoxifylline, thalidomide and anti-TNF monoclonal antibodies become more interesting, especially in refractory sarcoidosis. Thalidomide 119-130 tumor necrosis factor Homo sapiens 86-89 15858091-3 2005 Evaluation of response to thalidomide treatment was based on findings at computed tomography (CT) and change in serum alpha-fetoprotein level. Thalidomide 26-37 alpha fetoprotein Homo sapiens 118-135 16146335-4 2005 Thalidomide and IMiDs inhibit the cytokines tumor necrosis factor-alpha (TNF-alpha), interleukins (IL) 1beta, 6, 12, and granulocyte macrophage-colony stimulating factor (GM-CSF). Thalidomide 0-11 tumor necrosis factor Homo sapiens 44-71 16146335-4 2005 Thalidomide and IMiDs inhibit the cytokines tumor necrosis factor-alpha (TNF-alpha), interleukins (IL) 1beta, 6, 12, and granulocyte macrophage-colony stimulating factor (GM-CSF). Thalidomide 0-11 tumor necrosis factor Homo sapiens 73-82 16146335-4 2005 Thalidomide and IMiDs inhibit the cytokines tumor necrosis factor-alpha (TNF-alpha), interleukins (IL) 1beta, 6, 12, and granulocyte macrophage-colony stimulating factor (GM-CSF). Thalidomide 0-11 interleukin 1 beta Homo sapiens 85-108 16146335-7 2005 Thalidomide stimulates the Th-1 response increasing IFN-gamma levels while CC-4047 increased IL-2 as well. Thalidomide 0-11 interferon gamma Homo sapiens 52-61 15683845-0 2005 LASSBio-468: a new achiral thalidomide analogue which modulates TNF-alpha and NO production and inhibits endotoxic shock and arthritis in an animal model. Thalidomide 27-38 tumor necrosis factor Rattus norvegicus 64-73 16146335-4 2005 Thalidomide and IMiDs inhibit the cytokines tumor necrosis factor-alpha (TNF-alpha), interleukins (IL) 1beta, 6, 12, and granulocyte macrophage-colony stimulating factor (GM-CSF). Thalidomide 0-11 colony stimulating factor 2 Homo sapiens 121-169 15683845-2 2005 This compound was planned as an N-substituted phthalimide derivate, structurally designed as a hybrid of thalidomide and aryl sulfonamides, which were previously described as tumor necrosis factor-alpha (TNF-alpha) and PDE4 inhibitors. Thalidomide 105-116 tumor necrosis factor Rattus norvegicus 175-202 15683845-2 2005 This compound was planned as an N-substituted phthalimide derivate, structurally designed as a hybrid of thalidomide and aryl sulfonamides, which were previously described as tumor necrosis factor-alpha (TNF-alpha) and PDE4 inhibitors. Thalidomide 105-116 tumor necrosis factor Rattus norvegicus 204-213 15840315-8 2005 (2) Expression of VEGF mRNA level in Thd group (55 +/- 9) and Thd + CTX group (26 +/- 7) was significantly lower than that in control group (79 +/- 7, P < 0.01). Thalidomide 37-40 vascular endothelial growth factor A Mus musculus 18-22 15840315-8 2005 (2) Expression of VEGF mRNA level in Thd group (55 +/- 9) and Thd + CTX group (26 +/- 7) was significantly lower than that in control group (79 +/- 7, P < 0.01). Thalidomide 62-65 vascular endothelial growth factor A Mus musculus 18-22 15840315-9 2005 Serum VEGF level in Thd group, [(29 +/- 10) pg/ml] and Thd + CTX group [(12 +/- 6) pg/ml] was significantly lower than that in control group, [(71 +/- 16) pg/ml, P < 0.01]. Thalidomide 20-23 vascular endothelial growth factor A Mus musculus 6-10 16264875-0 2005 Does the combination of rituximab and thalidomide influence the long-term perspectives of advanced-stage MCL? Thalidomide 38-49 C-type lectin domain family 4 member D Homo sapiens 105-108 15654911-2 2005 Thalidomide is an immunomodulator agent of inflammatory cytokines including TNF-alpha. Thalidomide 0-11 tumor necrosis factor Homo sapiens 76-85 15638853-3 2005 Specifically, we have reported that thalidomide increased serum interleukin-2 (IL-2) levels and natural killer (NK) cell numbers in the peripheral blood of responding MM patients. Thalidomide 36-47 interleukin 2 Homo sapiens 64-77 15638853-3 2005 Specifically, we have reported that thalidomide increased serum interleukin-2 (IL-2) levels and natural killer (NK) cell numbers in the peripheral blood of responding MM patients. Thalidomide 36-47 interleukin 2 Homo sapiens 79-83 15723633-6 2005 The suppressive effect of thalidomide on the activation of the nuclear transcription factor NF-(kappa)B may explain these effects of thalidomide. Thalidomide 133-144 nuclear factor kappa B subunit 1 Homo sapiens 92-103 15723633-9 2005 Thalidomide, on the other hand, has been shown to selectively suppress NF-(kappa)B activation induced by inflammatory mediators. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 71-82 15723633-3 2005 So far, investigations into the mode of action of thalidomide have focused on lymphocytes and vascular endothelial cells and have shown that this agent inhibits the production of tumor necrosis factor (TNF)-alpha and is an inhibitor of tumor angiogenesis. Thalidomide 50-61 tumor necrosis factor Homo sapiens 179-212 15569010-4 2004 In this work we describe the effect that thalidomide had on TNF-alpha sera levels and on IL-4- and IFN gamma (IFNgamma)-producing lymphocytes of actinic prurigo (AP) patients. Thalidomide 41-52 interferon gamma Homo sapiens 110-118 15723633-6 2005 The suppressive effect of thalidomide on the activation of the nuclear transcription factor NF-(kappa)B may explain these effects of thalidomide. Thalidomide 26-37 nuclear factor kappa B subunit 1 Homo sapiens 92-103 15628770-13 2004 The downregulation of bFGF, VEGF, and maybe some other angiogenesis stimulators, is one of the antiangiogenic mechanisms of thalidomide. Thalidomide 124-135 fibroblast growth factor 2 Homo sapiens 22-26 15628770-13 2004 The downregulation of bFGF, VEGF, and maybe some other angiogenesis stimulators, is one of the antiangiogenic mechanisms of thalidomide. Thalidomide 124-135 vascular endothelial growth factor A Homo sapiens 28-32 15569010-4 2004 In this work we describe the effect that thalidomide had on TNF-alpha sera levels and on IL-4- and IFN gamma (IFNgamma)-producing lymphocytes of actinic prurigo (AP) patients. Thalidomide 41-52 tumor necrosis factor Homo sapiens 60-69 15569010-10 2004 CONCLUSIONS: Our findings confirm that AP is a disease that has an immunological component and that thalidomide clinical efficacy is exerted not only through inhibition of TNFalpha synthesis, but also through modulation of INFgamma-producing CD3+ cells. Thalidomide 100-111 tumor necrosis factor Homo sapiens 172-180 15569010-4 2004 In this work we describe the effect that thalidomide had on TNF-alpha sera levels and on IL-4- and IFN gamma (IFNgamma)-producing lymphocytes of actinic prurigo (AP) patients. Thalidomide 41-52 interleukin 4 Homo sapiens 89-93 15569010-4 2004 In this work we describe the effect that thalidomide had on TNF-alpha sera levels and on IL-4- and IFN gamma (IFNgamma)-producing lymphocytes of actinic prurigo (AP) patients. Thalidomide 41-52 interferon gamma Homo sapiens 99-108 15345321-1 2004 Thalidomide has shown to inhibit, selectively and mainly the cytokine tumor necrosis factor-alpha (TNF-alpha), thus, thalidomide has inhibitory consequences on other cytokines; this is ascribed as an immunomodulatory effect. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 70-97 15345321-1 2004 Thalidomide has shown to inhibit, selectively and mainly the cytokine tumor necrosis factor-alpha (TNF-alpha), thus, thalidomide has inhibitory consequences on other cytokines; this is ascribed as an immunomodulatory effect. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 99-108 15345321-1 2004 Thalidomide has shown to inhibit, selectively and mainly the cytokine tumor necrosis factor-alpha (TNF-alpha), thus, thalidomide has inhibitory consequences on other cytokines; this is ascribed as an immunomodulatory effect. Thalidomide 117-128 tumor necrosis factor Rattus norvegicus 70-97 15345321-1 2004 Thalidomide has shown to inhibit, selectively and mainly the cytokine tumor necrosis factor-alpha (TNF-alpha), thus, thalidomide has inhibitory consequences on other cytokines; this is ascribed as an immunomodulatory effect. Thalidomide 117-128 tumor necrosis factor Rattus norvegicus 99-108 15377478-0 2004 Recombinant human erythropoietin and the risk of thrombosis in patients receiving thalidomide for multiple myeloma. Thalidomide 82-93 erythropoietin Homo sapiens 18-32 15333517-7 2004 VEGF-induced angiogenesis was inhibited after treatment with combretastatin and thalidomide. Thalidomide 80-91 vascular endothelial growth factor A Homo sapiens 0-4 15464241-9 2004 Expression of TNFalpha, TGFbeta1, TIMP-1, and TIMP-2 was decreased in thalidomide-treated rats compared to those treated with vehicles. Thalidomide 70-81 tumor necrosis factor Rattus norvegicus 14-22 15464241-9 2004 Expression of TNFalpha, TGFbeta1, TIMP-1, and TIMP-2 was decreased in thalidomide-treated rats compared to those treated with vehicles. Thalidomide 70-81 transforming growth factor, beta 1 Rattus norvegicus 24-32 15464241-9 2004 Expression of TNFalpha, TGFbeta1, TIMP-1, and TIMP-2 was decreased in thalidomide-treated rats compared to those treated with vehicles. Thalidomide 70-81 TIMP metallopeptidase inhibitor 1 Rattus norvegicus 34-40 15464241-9 2004 Expression of TNFalpha, TGFbeta1, TIMP-1, and TIMP-2 was decreased in thalidomide-treated rats compared to those treated with vehicles. Thalidomide 70-81 TIMP metallopeptidase inhibitor 2 Rattus norvegicus 46-52 15464241-10 2004 Finally, the expression of TNFalpha and TGFbeta1 mRNA of Kupffer"s cells derived from rats treated with thalidomide were much lower than those treated with vehicle. Thalidomide 104-115 tumor necrosis factor Rattus norvegicus 27-35 15464241-10 2004 Finally, the expression of TNFalpha and TGFbeta1 mRNA of Kupffer"s cells derived from rats treated with thalidomide were much lower than those treated with vehicle. Thalidomide 104-115 transforming growth factor, beta 1 Rattus norvegicus 40-48 15464241-11 2004 CONCLUSIONS: Thalidomide salvages lethal hepatic necroinflammation, accelerates recovery from cirrhosis in rats, and works by suppressing of TNFalpha and TGFbeta1 production of Kupffer"s cells. Thalidomide 13-24 tumor necrosis factor Rattus norvegicus 141-149 15464241-11 2004 CONCLUSIONS: Thalidomide salvages lethal hepatic necroinflammation, accelerates recovery from cirrhosis in rats, and works by suppressing of TNFalpha and TGFbeta1 production of Kupffer"s cells. Thalidomide 13-24 transforming growth factor, beta 1 Rattus norvegicus 154-162 15235749-0 2004 Modification of thrombomodulin plasma levels in refractory myeloma patients during treatment with thalidomide and dexamethasone. Thalidomide 98-109 thrombomodulin Homo sapiens 16-30 15547876-3 2004 Thalidomide is an immunodulatory and antiangiogenic drug; the most pronounced effect of this drug is the inhibition of tumor necrosis factor-alpha (TNF-alpha ) production. Thalidomide 0-11 tumor necrosis factor Homo sapiens 119-146 15547876-3 2004 Thalidomide is an immunodulatory and antiangiogenic drug; the most pronounced effect of this drug is the inhibition of tumor necrosis factor-alpha (TNF-alpha ) production. Thalidomide 0-11 tumor necrosis factor Homo sapiens 148-157 15205358-0 2004 The effect of thalidomide on non-small cell lung cancer (NSCLC) cell lines: possible involvement in the PPARgamma pathway. Thalidomide 14-25 peroxisome proliferator activated receptor gamma Mus musculus 104-113 15205358-8 2004 At the molecular level, thalidomide increased peroxisome proliferator-activated receptor gamma (PPARgamma) protein dose-dependently, and peroxisome proliferator response element activity. Thalidomide 24-35 peroxisome proliferator activated receptor gamma Mus musculus 46-94 15205358-8 2004 At the molecular level, thalidomide increased peroxisome proliferator-activated receptor gamma (PPARgamma) protein dose-dependently, and peroxisome proliferator response element activity. Thalidomide 24-35 peroxisome proliferator activated receptor gamma Mus musculus 96-105 15205358-10 2004 In our mouse xenograft model of lung cancer, we found that intratumoral thalidomide caused a 64% decrease in tumor growth; moreover, tumors from the thalidomide-treated mice expressed higher PPARgamma, than tumors from control mice. Thalidomide 72-83 peroxisome proliferator activated receptor gamma Mus musculus 191-200 15205358-10 2004 In our mouse xenograft model of lung cancer, we found that intratumoral thalidomide caused a 64% decrease in tumor growth; moreover, tumors from the thalidomide-treated mice expressed higher PPARgamma, than tumors from control mice. Thalidomide 149-160 peroxisome proliferator activated receptor gamma Mus musculus 191-200 15205358-11 2004 This study shows the antitumor activity of thalidomide against LCC tumors and suggests a model in which thalidomide exerts its antitumor effects on LCC cells through the induction of PPARgamma and subsequent downstream signaling. Thalidomide 43-54 peroxisome proliferator activated receptor gamma Mus musculus 183-192 15205358-11 2004 This study shows the antitumor activity of thalidomide against LCC tumors and suggests a model in which thalidomide exerts its antitumor effects on LCC cells through the induction of PPARgamma and subsequent downstream signaling. Thalidomide 104-115 peroxisome proliferator activated receptor gamma Mus musculus 183-192 15205358-12 2004 To our knowledge, this is the first study to show a link between thalidomide and PPARgamma. Thalidomide 65-76 peroxisome proliferator activated receptor gamma Mus musculus 81-90 15290170-0 2004 Thalidomide upregulates macrophage inflammatory protein-1alpha in a herpes simplex virus-induced Behcet"s disease-like animal model. Thalidomide 0-11 chemokine (C-C motif) ligand 3 Mus musculus 24-62 15290170-9 2004 Among these, TNFalpha, MIP-1alpha, perforin and Fas were influenced by thalidomide treatment. Thalidomide 71-82 tumor necrosis factor Mus musculus 13-21 15290170-9 2004 Among these, TNFalpha, MIP-1alpha, perforin and Fas were influenced by thalidomide treatment. Thalidomide 71-82 chemokine (C-C motif) ligand 3 Mus musculus 23-33 15290170-10 2004 These results suggest that thalidomide can attenuate HSV-induced BD-like symptoms in mice through the downregulation of TNFalpha (P < 0.005) and the upregulation of MIP-1alpha (P < 0.005), perforin (P < 0.05) and FasR (P < 0.1). Thalidomide 27-38 tumor necrosis factor Mus musculus 120-128 15290170-10 2004 These results suggest that thalidomide can attenuate HSV-induced BD-like symptoms in mice through the downregulation of TNFalpha (P < 0.005) and the upregulation of MIP-1alpha (P < 0.005), perforin (P < 0.05) and FasR (P < 0.1). Thalidomide 27-38 chemokine (C-C motif) ligand 3 Mus musculus 168-178 15377478-1 2004 Among 199 patients treated with thalidomide for multiple myeloma, four thromboses occurred in 49 cases during erythropoietin therapy (prevalence 8.1%; annual rate 7.25%), and another 14 events occurred in patients not on erythropoietin (9.3%; 7.56%). Thalidomide 32-43 erythropoietin Homo sapiens 110-124 15446566-7 2004 Thalidomide decreased the stability of TNF-mRNA and COX-2 mRNA. Thalidomide 0-11 tumor necrosis factor Homo sapiens 39-42 15446566-7 2004 Thalidomide decreased the stability of TNF-mRNA and COX-2 mRNA. Thalidomide 0-11 mitochondrially encoded cytochrome c oxidase II Homo sapiens 52-57 15329836-0 2004 Mutations in SALL4 in malformed father and daughter postulated previously due to reflect mutagenesis by thalidomide. Thalidomide 104-115 spalt like transcription factor 4 Homo sapiens 13-18 15261258-1 2004 Thalidomide shows moderate inhibitory activity toward neuronal nitric oxide synthase (nNOS) and inducible NOS (iNOS), but not toward endothelial NOS (eNOS). Thalidomide 0-11 nitric oxide synthase 1 Homo sapiens 54-84 15261258-1 2004 Thalidomide shows moderate inhibitory activity toward neuronal nitric oxide synthase (nNOS) and inducible NOS (iNOS), but not toward endothelial NOS (eNOS). Thalidomide 0-11 nitric oxide synthase 1 Homo sapiens 86-90 15261258-1 2004 Thalidomide shows moderate inhibitory activity toward neuronal nitric oxide synthase (nNOS) and inducible NOS (iNOS), but not toward endothelial NOS (eNOS). Thalidomide 0-11 nitric oxide synthase 2 Homo sapiens 96-109 15261258-1 2004 Thalidomide shows moderate inhibitory activity toward neuronal nitric oxide synthase (nNOS) and inducible NOS (iNOS), but not toward endothelial NOS (eNOS). Thalidomide 0-11 nitric oxide synthase 2 Homo sapiens 111-115 15288591-5 2004 High amounts of TNF-alpha were detected in the exudates, which was inhibited by dexamethasone, pentoxifylline and thalidomide. Thalidomide 114-125 tumor necrosis factor Rattus norvegicus 16-25 15288591-6 2004 These results suggest a specific role for TNF-alpha in this model, and the ability of pentoxifylline and thalidomide to inhibit both neutrophil influx and TNF-alpha release may have therapeutic implications in arthritis. Thalidomide 105-116 tumor necrosis factor Rattus norvegicus 155-164 15261445-3 2004 Clinical trials have recently been initiated to evaluate the anti-tumour activity of the VEGF inhibitors SU5416, bevacizumab and thalidomide. Thalidomide 129-140 vascular endothelial growth factor A Homo sapiens 89-93 15598423-3 2004 In the present study, we have investigated the effects of thalidomide, CC-5013 and CC-4047 on the expression of COX-2 by stimulated PBMC. Thalidomide 58-69 mitochondrially encoded cytochrome c oxidase II Homo sapiens 112-117 15598423-4 2004 Our results show that thalidomide and IMiDs inhibited the expression of COX-2 but not the COX-1 protein in LPS-TNF-alpha and IL-1beta stimulated PBMC and shortened the half-life of COX-2 mRNA in a dose-dependent manner. Thalidomide 22-33 mitochondrially encoded cytochrome c oxidase II Homo sapiens 72-77 15598423-4 2004 Our results show that thalidomide and IMiDs inhibited the expression of COX-2 but not the COX-1 protein in LPS-TNF-alpha and IL-1beta stimulated PBMC and shortened the half-life of COX-2 mRNA in a dose-dependent manner. Thalidomide 22-33 tumor necrosis factor Homo sapiens 111-120 15598423-4 2004 Our results show that thalidomide and IMiDs inhibited the expression of COX-2 but not the COX-1 protein in LPS-TNF-alpha and IL-1beta stimulated PBMC and shortened the half-life of COX-2 mRNA in a dose-dependent manner. Thalidomide 22-33 interleukin 1 beta Homo sapiens 125-133 15598423-4 2004 Our results show that thalidomide and IMiDs inhibited the expression of COX-2 but not the COX-1 protein in LPS-TNF-alpha and IL-1beta stimulated PBMC and shortened the half-life of COX-2 mRNA in a dose-dependent manner. Thalidomide 22-33 mitochondrially encoded cytochrome c oxidase II Homo sapiens 181-186 15203277-0 2004 Arsenic trioxide and thalidomide combination produces multi-lineage hematological responses in myelodysplastic syndromes patients, particularly in those with high pre-therapy EVI1 expression. Thalidomide 21-32 MDS1 and EVI1 complex locus Homo sapiens 175-179 15221409-0 2004 Anti-angiogenic effects of thalidomide: expression of apoptosis-inducible active-caspase-3 in a three-dimensional collagen gel culture of aorta. Thalidomide 27-38 caspase 3 Homo sapiens 81-90 15221409-7 2004 Taken together with earlier findings, our new results indicate that the thalidomide-induced inhibition of angiogenesis involves apoptosis in addition to the suppression of TNF-alpha and inhibition of cell migration from aorta explants, i.e., the factors important for capillarogenesis. Thalidomide 72-83 tumor necrosis factor Homo sapiens 172-181 15242253-1 2004 Celgene, in collaboration with the National Cancer Institute, is developing CC-5013, the lead compound in a series of thalidomide derivatives that inhibit TNFalpha overproduction, for the potential treatment of hematological and solid tumor cancers and inflammatory diseases. Thalidomide 0-7 tumor necrosis factor Homo sapiens 155-163 15226321-5 2004 RESULTS: After a median potential follow-up time of 26.4 months, the proportion of patients with a greater than 50% decline in PSA was higher in the docetaxel/thalidomide group (53% in the combined group, 37% in docetaxel-alone arm). Thalidomide 159-170 kallikrein related peptidase 3 Homo sapiens 127-130 15226321-9 2004 CONCLUSION: In this randomized phase II trial, the addition of thalidomide to docetaxel resulted in an encouraging PSA decline rate and overall median survival rate in patients with metastatic AIPC. Thalidomide 63-74 kallikrein related peptidase 3 Homo sapiens 115-118 15217957-10 2004 CPS45, CPS49, and thalidomide significantly reduced PC3 tumor platelet-derived growth factor (PDGF)-AA levels by 58-82% (P < 0.05). Thalidomide 18-29 chromobox 8 Homo sapiens 52-55 15244383-12 2004 Analysis of biomarker assays did not identify any biomarker associated with greater response, but a significant increase in levels of soluble intercellular adhesion molecule 1, IL-2, and interferon gamma was seen with thalidomide therapy. Thalidomide 218-229 intercellular adhesion molecule 1 Homo sapiens 142-175 15244383-12 2004 Analysis of biomarker assays did not identify any biomarker associated with greater response, but a significant increase in levels of soluble intercellular adhesion molecule 1, IL-2, and interferon gamma was seen with thalidomide therapy. Thalidomide 218-229 interleukin 2 Homo sapiens 177-181 15244383-12 2004 Analysis of biomarker assays did not identify any biomarker associated with greater response, but a significant increase in levels of soluble intercellular adhesion molecule 1, IL-2, and interferon gamma was seen with thalidomide therapy. Thalidomide 218-229 interferon gamma Homo sapiens 187-203 15069691-0 2004 Thalidomide reduces serum C-reactive protein and interleukin-6 and induces response to IL-2 in a fraction of metastatic renal cell cancer patients who failed IL-2-based therapy. Thalidomide 0-11 C-reactive protein Homo sapiens 26-44 15069691-0 2004 Thalidomide reduces serum C-reactive protein and interleukin-6 and induces response to IL-2 in a fraction of metastatic renal cell cancer patients who failed IL-2-based therapy. Thalidomide 0-11 interleukin 6 Homo sapiens 49-62 15069691-0 2004 Thalidomide reduces serum C-reactive protein and interleukin-6 and induces response to IL-2 in a fraction of metastatic renal cell cancer patients who failed IL-2-based therapy. Thalidomide 0-11 interleukin 2 Homo sapiens 87-91 15069691-0 2004 Thalidomide reduces serum C-reactive protein and interleukin-6 and induces response to IL-2 in a fraction of metastatic renal cell cancer patients who failed IL-2-based therapy. Thalidomide 0-11 interleukin 2 Homo sapiens 158-162 15069691-3 2004 We used thalidomide to treat patients with cancer-induced cachexia and noted that the drug significantly reduced serum levels of CRP and IL-6 to normal or near normal levels in a substantial fraction of patients. Thalidomide 8-19 C-reactive protein Homo sapiens 129-132 15069691-3 2004 We used thalidomide to treat patients with cancer-induced cachexia and noted that the drug significantly reduced serum levels of CRP and IL-6 to normal or near normal levels in a substantial fraction of patients. Thalidomide 8-19 interleukin 6 Homo sapiens 137-141 15069691-4 2004 We tested whether thalidomide might potentiate the response of patients with renal cell carcinoma to IL-2. Thalidomide 18-29 interleukin 2 Homo sapiens 101-105 15069691-9 2004 Reduction of serum CRP or IL-6 levels with thalidomide may enhance the responsiveness of renal cell carcinoma to IL-2. Thalidomide 43-54 C-reactive protein Homo sapiens 19-22 15069691-9 2004 Reduction of serum CRP or IL-6 levels with thalidomide may enhance the responsiveness of renal cell carcinoma to IL-2. Thalidomide 43-54 interleukin 6 Homo sapiens 26-30 15069691-9 2004 Reduction of serum CRP or IL-6 levels with thalidomide may enhance the responsiveness of renal cell carcinoma to IL-2. Thalidomide 43-54 interleukin 2 Homo sapiens 113-117 15069691-11 2004 It is possible that the thalidomide-induced normalization of serum acute phase proteins might improve the response of other types of malignancy to IL-2 or other immune-based therapies. Thalidomide 24-35 interleukin 2 Homo sapiens 147-151 15242253-1 2004 Celgene, in collaboration with the National Cancer Institute, is developing CC-5013, the lead compound in a series of thalidomide derivatives that inhibit TNFalpha overproduction, for the potential treatment of hematological and solid tumor cancers and inflammatory diseases. Thalidomide 118-129 tumor necrosis factor Homo sapiens 155-163 15222121-4 2004 Thalidomide has been shown to have antiangiogenic and immunomodulatory effects, including the inhibition of vascular endothelial growth factor, basic fibroblast growth factor and tumor necrosis factor alpha. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 108-142 15138204-10 2004 Treatment with thalidomide, dexamethasone, or fucoidin reduced gastric damage and MPO activity induced by indomethacin. Thalidomide 15-26 myeloperoxidase Mus musculus 82-85 15060740-13 2004 TNF mediates the effect of DMXAA on thalidomide pharmacokinetics but not that of cyclophosphamide. Thalidomide 36-47 tumor necrosis factor Mus musculus 0-3 15149630-1 2004 Thalidomide has antiangiogenic and immunomodulatory effects, mediated by several cytokines such as vascular endothelial growth factor (VEGF), fibroblastic growth factor (FGF-2), hepatocyte growth factor (HGF), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha). Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 99-133 15149630-1 2004 Thalidomide has antiangiogenic and immunomodulatory effects, mediated by several cytokines such as vascular endothelial growth factor (VEGF), fibroblastic growth factor (FGF-2), hepatocyte growth factor (HGF), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha). Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 170-175 15149630-1 2004 Thalidomide has antiangiogenic and immunomodulatory effects, mediated by several cytokines such as vascular endothelial growth factor (VEGF), fibroblastic growth factor (FGF-2), hepatocyte growth factor (HGF), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha). Thalidomide 0-11 hepatocyte growth factor Homo sapiens 178-202 15149630-1 2004 Thalidomide has antiangiogenic and immunomodulatory effects, mediated by several cytokines such as vascular endothelial growth factor (VEGF), fibroblastic growth factor (FGF-2), hepatocyte growth factor (HGF), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha). Thalidomide 0-11 hepatocyte growth factor Homo sapiens 204-207 15149630-1 2004 Thalidomide has antiangiogenic and immunomodulatory effects, mediated by several cytokines such as vascular endothelial growth factor (VEGF), fibroblastic growth factor (FGF-2), hepatocyte growth factor (HGF), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha). Thalidomide 0-11 interleukin 6 Homo sapiens 210-223 15149630-1 2004 Thalidomide has antiangiogenic and immunomodulatory effects, mediated by several cytokines such as vascular endothelial growth factor (VEGF), fibroblastic growth factor (FGF-2), hepatocyte growth factor (HGF), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha). Thalidomide 0-11 interleukin 6 Homo sapiens 225-229 15149630-1 2004 Thalidomide has antiangiogenic and immunomodulatory effects, mediated by several cytokines such as vascular endothelial growth factor (VEGF), fibroblastic growth factor (FGF-2), hepatocyte growth factor (HGF), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha). Thalidomide 0-11 tumor necrosis factor Homo sapiens 235-262 15149630-1 2004 Thalidomide has antiangiogenic and immunomodulatory effects, mediated by several cytokines such as vascular endothelial growth factor (VEGF), fibroblastic growth factor (FGF-2), hepatocyte growth factor (HGF), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha). Thalidomide 0-11 tumor necrosis factor Homo sapiens 264-273 15149630-1 2004 Thalidomide has antiangiogenic and immunomodulatory effects, mediated by several cytokines such as vascular endothelial growth factor (VEGF), fibroblastic growth factor (FGF-2), hepatocyte growth factor (HGF), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha). Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 135-139 15010523-11 2004 This is a possible mechanism of action with thalidomide reducing both tumor necrosis factor and the neutrophil migration. Thalidomide 44-55 tumor necrosis factor Homo sapiens 70-91 15086397-6 2004 In addition IgG1 antimycobacterial antibodies can reverse the effect of TNF-alpha blockers such as pentoxifylline and thalidomide. Thalidomide 118-129 tumor necrosis factor Homo sapiens 72-81 15182131-4 2004 Considering the involvement of IL-6 in Castleman"s disease we treated the patient with thalidomide obtaining the remission of the nephrotic syndrome. Thalidomide 87-98 interleukin 6 Homo sapiens 31-35 15179032-9 2004 Thalidomide was found to accelerate the degradation of TNF-alpha mRNA, but had little effect on IL-1beta or IL-8. Thalidomide 0-11 tumor necrosis factor Homo sapiens 55-64 14988093-6 2004 In addition, arterial macrophage infiltration and local expressions of tumor necrosis factor alpha (TNF-alpha) and basic fibroblast growth factor (bFGF) in the injured arteries as measured by immunohistochemistry and immunoblot analysis were significantly reduced by thalidomide treatment. Thalidomide 267-278 tumor necrosis factor Rattus norvegicus 71-98 14988093-6 2004 In addition, arterial macrophage infiltration and local expressions of tumor necrosis factor alpha (TNF-alpha) and basic fibroblast growth factor (bFGF) in the injured arteries as measured by immunohistochemistry and immunoblot analysis were significantly reduced by thalidomide treatment. Thalidomide 267-278 fibroblast growth factor 2 Rattus norvegicus 115-145 14988093-7 2004 Serum TNF-alpha, measured by ELISA, was also significantly reduced in the thalidomide-treated animals compared with controls after injury (856+/-213 versus 449+/-68 pg/mL on day 3, P=0.001; 129+/-34 versus 63+/-18 pg/mL on day 14, P=0.001), and we observed a good positive correlation between the serum TNF-alpha levels and the severity of neointimal growth. Thalidomide 74-85 tumor necrosis factor Rattus norvegicus 6-15 14988093-7 2004 Serum TNF-alpha, measured by ELISA, was also significantly reduced in the thalidomide-treated animals compared with controls after injury (856+/-213 versus 449+/-68 pg/mL on day 3, P=0.001; 129+/-34 versus 63+/-18 pg/mL on day 14, P=0.001), and we observed a good positive correlation between the serum TNF-alpha levels and the severity of neointimal growth. Thalidomide 74-85 tumor necrosis factor Rattus norvegicus 303-312 15136218-0 2004 A low serum level of soluble tumor necrosis factor receptor p55 predicts response to thalidomide in advanced multiple myeloma. Thalidomide 85-96 tumor necrosis factor Homo sapiens 29-50 15136218-0 2004 A low serum level of soluble tumor necrosis factor receptor p55 predicts response to thalidomide in advanced multiple myeloma. Thalidomide 85-96 TNF receptor superfamily member 1A Homo sapiens 60-63 15136218-1 2004 BACKGROUND AND OBJECTIVES: Thalidomide modulates the production of tumor necrosis factor (TNF-alpha). Thalidomide 27-38 tumor necrosis factor Homo sapiens 67-88 15136218-1 2004 BACKGROUND AND OBJECTIVES: Thalidomide modulates the production of tumor necrosis factor (TNF-alpha). Thalidomide 27-38 tumor necrosis factor Homo sapiens 90-99 15136218-6 2004 RESULTS: The pre-treatment serum level of soluble TNFR p55 in thalidomide responders was significantly lower than that in non-responders (median 1.75 ng/mL (range 1.19-2.84) vs. 2.79 ng/mL (1.36-5.51), p=0.004). Thalidomide 62-73 TNF receptor superfamily member 1A Homo sapiens 50-54 15136218-6 2004 RESULTS: The pre-treatment serum level of soluble TNFR p55 in thalidomide responders was significantly lower than that in non-responders (median 1.75 ng/mL (range 1.19-2.84) vs. 2.79 ng/mL (1.36-5.51), p=0.004). Thalidomide 62-73 TNF receptor superfamily member 1A Homo sapiens 55-58 15136218-10 2004 INTERPRETATION AND CONCLUSIONS: We conclude that soluble TNFR p55 is an adverse prognostic factor in myeloma patients with relapsed or refractory disease treated with thalidomide. Thalidomide 167-178 TNF receptor superfamily member 1A Homo sapiens 57-61 15136218-10 2004 INTERPRETATION AND CONCLUSIONS: We conclude that soluble TNFR p55 is an adverse prognostic factor in myeloma patients with relapsed or refractory disease treated with thalidomide. Thalidomide 167-178 TNF receptor superfamily member 1A Homo sapiens 62-65 15218967-4 2004 The myeloma cells decreased by thalidomide therapy were mature (MPC-1(+)/CD49e(+)) and intermediate (MPC-1(+)/CD49e(-)) types. Thalidomide 31-42 mitochondrial pyruvate carrier 1 Homo sapiens 64-72 15218967-4 2004 The myeloma cells decreased by thalidomide therapy were mature (MPC-1(+)/CD49e(+)) and intermediate (MPC-1(+)/CD49e(-)) types. Thalidomide 31-42 integrin subunit alpha 5 Homo sapiens 73-78 15218967-4 2004 The myeloma cells decreased by thalidomide therapy were mature (MPC-1(+)/CD49e(+)) and intermediate (MPC-1(+)/CD49e(-)) types. Thalidomide 31-42 mitochondrial pyruvate carrier 1 Homo sapiens 101-109 15218967-4 2004 The myeloma cells decreased by thalidomide therapy were mature (MPC-1(+)/CD49e(+)) and intermediate (MPC-1(+)/CD49e(-)) types. Thalidomide 31-42 integrin subunit alpha 5 Homo sapiens 110-115 15179032-0 2004 The effects of thalidomide on the stimulation of NF-kappaB activity and TNF-alpha production by lipopolysaccharide in a human colonic epithelial cell line. Thalidomide 15-26 nuclear factor kappa B subunit 1 Homo sapiens 49-58 15179032-0 2004 The effects of thalidomide on the stimulation of NF-kappaB activity and TNF-alpha production by lipopolysaccharide in a human colonic epithelial cell line. Thalidomide 15-26 tumor necrosis factor Homo sapiens 72-81 15179032-1 2004 The immunomodulatory and anti-inflammatory effects of thalidomide are associated with inhibition of TNF-alpha levels. Thalidomide 54-65 tumor necrosis factor Homo sapiens 100-109 15179032-2 2004 However, the mechanism by which thalidomide reduces TNF-alpha production remains elusive. Thalidomide 32-43 tumor necrosis factor Homo sapiens 52-61 15179032-4 2004 We tested whether thalidomide acts through inhibiting NF-kappaB activity. Thalidomide 18-29 nuclear factor kappa B subunit 1 Homo sapiens 54-63 15140182-2 2004 Here, male TNF-alpha (-/-) deficient mice and C57bL/6 mice were treated with MPTP (4 x 15 mg/kg, 24 h intervals) and in one series, thalidomide was administered to inhibit TNF-alpha synthesis. Thalidomide 132-143 tumor necrosis factor Mus musculus 172-181 15179032-10 2004 These observations suggest that the immunomodulatory effect of thalidomide in colonic epithelial cells is associated with inhibition of TNF-alpha. Thalidomide 63-74 tumor necrosis factor Homo sapiens 136-145 15067094-0 2004 Thalidomide induces apoptosis in human monocytes by using a cytochrome c-dependent pathway. Thalidomide 0-11 cytochrome c, somatic Homo sapiens 60-72 15086832-8 2004 The concentration of M-protein in the serum or urine of seven of the 11 patients was reduced by at least 30% after thalidomide treatment, and MVD in the BM decreased in three of these seven cases in response to thalidomide. Thalidomide 115-126 myomesin 2 Homo sapiens 21-30 15086832-8 2004 The concentration of M-protein in the serum or urine of seven of the 11 patients was reduced by at least 30% after thalidomide treatment, and MVD in the BM decreased in three of these seven cases in response to thalidomide. Thalidomide 211-222 myomesin 2 Homo sapiens 21-30 15086832-9 2004 Increased plasma concentrations of basic fibroblast growth factor (FGF-2) and vascular endothelial growth factor (VEGF) were observed in all 11 cases before thalidomide administration and both levels were reduced after treatment with thalidomide. Thalidomide 157-168 fibroblast growth factor 2 Homo sapiens 67-72 15086832-9 2004 Increased plasma concentrations of basic fibroblast growth factor (FGF-2) and vascular endothelial growth factor (VEGF) were observed in all 11 cases before thalidomide administration and both levels were reduced after treatment with thalidomide. Thalidomide 157-168 vascular endothelial growth factor A Homo sapiens 114-118 15086832-9 2004 Increased plasma concentrations of basic fibroblast growth factor (FGF-2) and vascular endothelial growth factor (VEGF) were observed in all 11 cases before thalidomide administration and both levels were reduced after treatment with thalidomide. Thalidomide 234-245 fibroblast growth factor 2 Homo sapiens 67-72 15086832-9 2004 Increased plasma concentrations of basic fibroblast growth factor (FGF-2) and vascular endothelial growth factor (VEGF) were observed in all 11 cases before thalidomide administration and both levels were reduced after treatment with thalidomide. Thalidomide 234-245 vascular endothelial growth factor A Homo sapiens 78-112 15086832-9 2004 Increased plasma concentrations of basic fibroblast growth factor (FGF-2) and vascular endothelial growth factor (VEGF) were observed in all 11 cases before thalidomide administration and both levels were reduced after treatment with thalidomide. Thalidomide 234-245 vascular endothelial growth factor A Homo sapiens 114-118 15086832-11 2004 It seems that thalidomide is effective in the treatment of MM through the impairment of angiogenesis by decreasing FGF-2 and VEGF production. Thalidomide 14-25 fibroblast growth factor 2 Homo sapiens 115-120 15086832-11 2004 It seems that thalidomide is effective in the treatment of MM through the impairment of angiogenesis by decreasing FGF-2 and VEGF production. Thalidomide 14-25 vascular endothelial growth factor A Homo sapiens 125-129 15067094-6 2004 Monocyte apoptosis required the activation of caspases, as combined stimulation by thalidomide together with the broad caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone markedly prevented monocyte cell death. Thalidomide 83-94 caspase 9 Homo sapiens 46-54 15016759-5 2004 After three months of thalidomide therapy, serum levels of vascular endothelial growth factor were strongly suppressed compared with pretreatment levels. Thalidomide 22-33 vascular endothelial growth factor A Homo sapiens 59-93 15064811-1 2004 Thalidomide is a selective inhibitor of tumor necrosis factor-alpha (TNF-alpha), a cytokine involved in mycobacterial death mechanisms. Thalidomide 0-11 tumor necrosis factor Mus musculus 69-78 15056977-0 2004 Thalidomide as a nitric oxide synthase inhibitor and its structural development. Thalidomide 0-11 nitric oxide synthase 2 Homo sapiens 17-38 15056977-1 2004 Thalidomide has been found to exhibit weak nitric oxide synthase (NOS)-inhibitory activity. Thalidomide 0-11 nitric oxide synthase 2 Homo sapiens 43-64 15163089-3 2004 Thalidomide, a potent tumor necrosis factor alpha inhibitor, was well tolerated and possibly showed some clinical benefit in children with tuberculous meningitis during a pilot study. Thalidomide 0-11 tumor necrosis factor Homo sapiens 22-49 14995997-10 2004 These findings suggest that thalidomide may be procoagulant, not by enhancing doxorubicin-mediated endothelial cell injury, but by altering the expression of PAR-1 on injured endothelium and resulting in endothelial dysfunction, which may explain hypercoagulability in patients treated with chemotherapy followed by thalidomide. Thalidomide 28-39 coagulation factor II thrombin receptor Homo sapiens 158-163 15190511-4 2004 Thalidomide, a drug with anti-TNF, antiangiogenic, and immunomodulatory activities, and other agents with anti-TNF effects, such as pentoxifylline, etanercept, and infliximab, have produced hematologic improvement in 20% to 40% of patients. Thalidomide 0-11 tumor necrosis factor Homo sapiens 30-33 14995997-10 2004 These findings suggest that thalidomide may be procoagulant, not by enhancing doxorubicin-mediated endothelial cell injury, but by altering the expression of PAR-1 on injured endothelium and resulting in endothelial dysfunction, which may explain hypercoagulability in patients treated with chemotherapy followed by thalidomide. Thalidomide 316-327 coagulation factor II thrombin receptor Homo sapiens 158-163 14680461-1 2004 Thalidomide has several targets and mechanisms of action: a hypnosedative effect, several immunomodulatory properties with an effect on the production of TNF-alpha and the balance between the different lymphocyte subsets and an antiangiogenic action. Thalidomide 0-11 tumor necrosis factor Homo sapiens 154-163 15663335-7 2004 Thalidomide has been demonstrated to suppress tumor necrosis factor (TNF) release, which may be important at both the initial and chronic phases of the inflammation of sarcoidosis and appears to be crucial as part of the initial granulomatous response. Thalidomide 0-11 tumor necrosis factor Homo sapiens 46-67 15663335-7 2004 Thalidomide has been demonstrated to suppress tumor necrosis factor (TNF) release, which may be important at both the initial and chronic phases of the inflammation of sarcoidosis and appears to be crucial as part of the initial granulomatous response. Thalidomide 0-11 tumor necrosis factor Homo sapiens 69-72 15190675-0 2004 Thalidomide-based TNF-alpha inhibitors for neurodegenerative diseases. Thalidomide 0-11 tumor necrosis factor Homo sapiens 18-27 15190675-5 2004 As its clinical value in treating ENL derives from its TNF-alpha inhibitory activity, thalidomide was chosen for structural modification for the discovery of novel and more potent isosteric analogues with appropriate lipophilicity to insure high brain penetration. Thalidomide 86-97 tumor necrosis factor Homo sapiens 55-64 15581054-2 2004 Using an in vivo tumor model, we report that thalidomide (100 mg kg(-1)day(-1)) or clotrimazole (120 mg kg(-1) day(-1)), inhibit blood vessel formation (determined by hemoglobin content), leukocyte recruitment [myeloperoxidase (MPO) activity; N-acetylglucosa-minidase (NAG) activity], and vascular endothelial growth factor production. Thalidomide 45-56 myeloperoxidase Mus musculus 211-226 15581054-2 2004 Using an in vivo tumor model, we report that thalidomide (100 mg kg(-1)day(-1)) or clotrimazole (120 mg kg(-1) day(-1)), inhibit blood vessel formation (determined by hemoglobin content), leukocyte recruitment [myeloperoxidase (MPO) activity; N-acetylglucosa-minidase (NAG) activity], and vascular endothelial growth factor production. Thalidomide 45-56 myeloperoxidase Mus musculus 228-231 15581054-2 2004 Using an in vivo tumor model, we report that thalidomide (100 mg kg(-1)day(-1)) or clotrimazole (120 mg kg(-1) day(-1)), inhibit blood vessel formation (determined by hemoglobin content), leukocyte recruitment [myeloperoxidase (MPO) activity; N-acetylglucosa-minidase (NAG) activity], and vascular endothelial growth factor production. Thalidomide 45-56 alpha-N-acetylglucosaminidase (Sanfilippo disease IIIB) Mus musculus 243-267 15581054-2 2004 Using an in vivo tumor model, we report that thalidomide (100 mg kg(-1)day(-1)) or clotrimazole (120 mg kg(-1) day(-1)), inhibit blood vessel formation (determined by hemoglobin content), leukocyte recruitment [myeloperoxidase (MPO) activity; N-acetylglucosa-minidase (NAG) activity], and vascular endothelial growth factor production. Thalidomide 45-56 alpha-N-acetylglucosaminidase (Sanfilippo disease IIIB) Mus musculus 269-272 15581054-5 2004 Thalidomide was able to reduce the inflammatory reaction (MPO and NAG activities) by 50% to 70%, but was unable to delay tumor development. Thalidomide 0-11 myeloperoxidase Mus musculus 58-61 15581054-5 2004 Thalidomide was able to reduce the inflammatory reaction (MPO and NAG activities) by 50% to 70%, but was unable to delay tumor development. Thalidomide 0-11 alpha-N-acetylglucosaminidase (Sanfilippo disease IIIB) Mus musculus 66-69 14962263-3 2004 Current data suggest that the action of thalidomide is related to several different mechanisms, including suppression of tumor necrosis factor, effects on basic fibroblast growth factor, vascular endothelial growth factor, interleukins and interferons, downregulation of selected cell surface adhesion molecules, and changes in the lymphocyte subsets. Thalidomide 40-51 vascular endothelial growth factor A Homo sapiens 187-221 15479647-7 2004 However, in 2 cases in which the mean blood concentration of thalidomide was 2 microg/mL or higher, an M-protein-reducing effect was not obtained. Thalidomide 61-72 myomesin 2 Homo sapiens 103-112 15025228-1 2004 BACKGROUND: Pentoxifylline (PTX) and thalidomide (TLD) have been shown to inhibit cytokine synthesis, mainly tumor necrosis factor (TNF)-alpha, in different inflammatory conditions. Thalidomide 37-48 tumor necrosis factor Rattus norvegicus 109-142 15167912-12 2004 Additionally, a dramatic decrease in Bcl-2 expression with s-thalidomide suggests a possible enhancement of cytotoxic effect if combined with other cytotoxic agents. Thalidomide 59-72 BCL2 apoptosis regulator Homo sapiens 37-42 29539102-1 2004 BACKGROUND: Pentoxifylline (PTX) and thalidomide (TLD) have been shown to inhibit cytokine synthesis, mainly tumor necrosis factor (TNF)-alpha, in different inflammatory conditions. Thalidomide 37-48 tumor necrosis factor Rattus norvegicus 109-142 14613324-0 2003 Thiothalidomides: novel isosteric analogues of thalidomide with enhanced TNF-alpha inhibitory activity. Thalidomide 4-15 tumor necrosis factor Homo sapiens 73-82 14693056-2 2003 The aim of this research was to observe the effect of thalidomide on tumor homograft growth in mouse H22 model and to investigate the mechanisms involved and its curative possibility to hepatoma. Thalidomide 54-65 histocompatibility 22 Mus musculus 101-104 14693056-15 2003 CONCLUSION: Thalidomide can significantly induce apoptosis and inhibit angiogenesis in mouse H22 model when it is used at the beginning of carcinogenesis, but it has no obvious anti-angiogenic efficacy on the grown tumor. Thalidomide 12-23 histocompatibility 22 Mus musculus 93-96 14693056-16 2003 Thalidomide cannot inhibit VEGF mRNA expression of grafted H22 tumor in mouse. Thalidomide 0-11 vascular endothelial growth factor A Mus musculus 27-31 14693056-16 2003 Thalidomide cannot inhibit VEGF mRNA expression of grafted H22 tumor in mouse. Thalidomide 0-11 histocompatibility 22 Mus musculus 59-62 14598030-8 2003 Thalidomide, but not rosiglitazone, was associated with the inhibition of basement membrane thickening and the blockade of the increase of VEGF in ocular fluid, thus representing a potential therapeutic drug for the prevention of diabetic retinopathy. Thalidomide 0-11 vascular endothelial growth factor A Rattus norvegicus 139-143 14612937-13 2003 Thalidomide induced co-option of host vasculature, an effect noted previously after VEGF blockade. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 84-88 15303463-11 2004 TNF-alpha inhibitors: infliximab, CDP 571, etanercept, onercept, CNI- 1493 and thalidomide. Thalidomide 79-90 tumor necrosis factor Homo sapiens 0-9 14679006-6 2003 Dexamethasone and immunomodulatory derivatives of thalidomide (IMiDs), but not proteasome inhibitor PS-341, augmented MM cell apoptosis triggered by LPAAT-beta inhibitors. Thalidomide 50-61 1-acylglycerol-3-phosphate O-acyltransferase 2 Homo sapiens 149-159 14617101-0 2003 Thalidomide inhibits UVB-induced mouse keratinocyte apoptosis by both TNF-alpha-dependent and TNF-alpha-independent pathways. Thalidomide 0-11 tumor necrosis factor Mus musculus 70-79 14617101-0 2003 Thalidomide inhibits UVB-induced mouse keratinocyte apoptosis by both TNF-alpha-dependent and TNF-alpha-independent pathways. Thalidomide 0-11 tumor necrosis factor Mus musculus 94-103 14617101-4 2003 PURPOSE: To test our hypothesis that thalidomide may antagonize TNF-alpha production in the skin, we used a mouse model for acute ultraviolet-B (UVB) exposure, a known stimulus for inducing this cytokine. Thalidomide 37-48 tumor necrosis factor Mus musculus 64-73 14617101-7 2003 RNase protection assays confirmed that high (400 mg/kg), but not low (100 mg/kg), doses of thalidomide inhibited the UVB-induced increase in steady-state TNF-alpha mRNA. Thalidomide 91-102 tumor necrosis factor Mus musculus 154-163 14617101-9 2003 The antiapoptotic effects of thalidomide can be reversed by the addition of exogenous recombinant mouse TNF-alpha and hence reconstituting UVB-induced programmed cell death. Thalidomide 29-40 tumor necrosis factor Mus musculus 104-113 14613324-3 2003 In this regard, oral thalidomide is clinically valuable in the treatment of erythema nodosum leprosum (ENL) and multiple myeloma and effectively reduces tumor necrosis factor-alpha (TNF-alpha) levels and angiogenesis in vivo. Thalidomide 21-32 tumor necrosis factor Homo sapiens 153-180 14613324-3 2003 In this regard, oral thalidomide is clinically valuable in the treatment of erythema nodosum leprosum (ENL) and multiple myeloma and effectively reduces tumor necrosis factor-alpha (TNF-alpha) levels and angiogenesis in vivo. Thalidomide 21-32 tumor necrosis factor Homo sapiens 182-191 14613324-7 2003 Two isosteric analogues of thalidomide, compounds 15 and 16, that mostly reflect the parent compound, together with the simple structure, dithioglutarimide 19, potently inhibited TNF-alpha secretion, compared to thalidomide, 1. Thalidomide 27-38 tumor necrosis factor Homo sapiens 179-188 14616984-0 2003 Serum MUC-1 as a marker of disease status in multiple myeloma patients receiving thalidomide. Thalidomide 81-92 mucin 1, cell surface associated Homo sapiens 6-11 12930142-2 2003 Synthesis and antiangiogenic activity of the teratogenic and TNFalpha-modulatory thalidomide analogue 2-(2,6-dioxopiperidine-3-yl)phthalimidine. Thalidomide 81-92 tumor necrosis factor Rattus norvegicus 61-69 14682394-0 2003 Exposure to the anti-TNF-alpha drug thalidomide induces apoptotic cell death in human T leukemic cells. Thalidomide 36-47 tumor necrosis factor Homo sapiens 21-30 14682394-2 2003 The controversial drug thalidomide is anti-inflammatory, anti-angiogenic and a selective inhibitor of TNF-alpha that is being studied as a treatment for HIV-1-related disorders, immune disorders and cancer. Thalidomide 23-34 tumor necrosis factor Homo sapiens 102-111 14682394-6 2003 Low dose thalidomide treatment of human T leukemic cells exhibited rapid increases in caspase-3 activity, annexin V-FITC binding and DNA disintegration that is characteristic of apoptosis. Thalidomide 9-20 caspase 3 Homo sapiens 86-95 14682394-6 2003 Low dose thalidomide treatment of human T leukemic cells exhibited rapid increases in caspase-3 activity, annexin V-FITC binding and DNA disintegration that is characteristic of apoptosis. Thalidomide 9-20 annexin A5 Homo sapiens 106-115 14505639-1 2003 Thalidomide, (1), has made a remarkable comeback from its days of a sedative with teratogenic properties due to its ability to selectively inhibit TNF-alpha, a key pro-inflammatory cytokine and its clinical benefit in the treatment of cancer. Thalidomide 0-11 tumor necrosis factor Homo sapiens 147-156 14505639-6 2003 On the other hand, 4 was non-cytotoxic at the tested concentrations and was found to be 830-fold more potent than thalidomide as TNF-alpha inhibitor. Thalidomide 114-125 tumor necrosis factor Homo sapiens 129-138 14531913-0 2003 Plasma levels of tumour necrosis factor alpha and interleukin-6 predict progression-free survival following thalidomide therapy in patients with previously untreated multiple myeloma. Thalidomide 108-119 interleukin 6 Homo sapiens 50-63 14558594-2 2003 The results show that thalidomide inhibits endotelial cell proliferation induced by bFGF and VEGF, more so if the cells are grown on vitronectin; moreover, treatment with thalidomide reduces the release of MMP-2 and IL-8 by endothelial cells, suggesting a further pathway for the antiangiogenic activity of drug. Thalidomide 22-33 fibroblast growth factor 2 Homo sapiens 84-88 14558594-2 2003 The results show that thalidomide inhibits endotelial cell proliferation induced by bFGF and VEGF, more so if the cells are grown on vitronectin; moreover, treatment with thalidomide reduces the release of MMP-2 and IL-8 by endothelial cells, suggesting a further pathway for the antiangiogenic activity of drug. Thalidomide 22-33 vascular endothelial growth factor A Homo sapiens 93-97 14558594-2 2003 The results show that thalidomide inhibits endotelial cell proliferation induced by bFGF and VEGF, more so if the cells are grown on vitronectin; moreover, treatment with thalidomide reduces the release of MMP-2 and IL-8 by endothelial cells, suggesting a further pathway for the antiangiogenic activity of drug. Thalidomide 22-33 vitronectin Homo sapiens 133-144 14558594-2 2003 The results show that thalidomide inhibits endotelial cell proliferation induced by bFGF and VEGF, more so if the cells are grown on vitronectin; moreover, treatment with thalidomide reduces the release of MMP-2 and IL-8 by endothelial cells, suggesting a further pathway for the antiangiogenic activity of drug. Thalidomide 22-33 matrix metallopeptidase 2 Homo sapiens 206-211 14558594-2 2003 The results show that thalidomide inhibits endotelial cell proliferation induced by bFGF and VEGF, more so if the cells are grown on vitronectin; moreover, treatment with thalidomide reduces the release of MMP-2 and IL-8 by endothelial cells, suggesting a further pathway for the antiangiogenic activity of drug. Thalidomide 22-33 C-X-C motif chemokine ligand 8 Homo sapiens 216-220 14558594-2 2003 The results show that thalidomide inhibits endotelial cell proliferation induced by bFGF and VEGF, more so if the cells are grown on vitronectin; moreover, treatment with thalidomide reduces the release of MMP-2 and IL-8 by endothelial cells, suggesting a further pathway for the antiangiogenic activity of drug. Thalidomide 171-182 fibroblast growth factor 2 Homo sapiens 84-88 14558594-2 2003 The results show that thalidomide inhibits endotelial cell proliferation induced by bFGF and VEGF, more so if the cells are grown on vitronectin; moreover, treatment with thalidomide reduces the release of MMP-2 and IL-8 by endothelial cells, suggesting a further pathway for the antiangiogenic activity of drug. Thalidomide 171-182 vascular endothelial growth factor A Homo sapiens 93-97 14558594-2 2003 The results show that thalidomide inhibits endotelial cell proliferation induced by bFGF and VEGF, more so if the cells are grown on vitronectin; moreover, treatment with thalidomide reduces the release of MMP-2 and IL-8 by endothelial cells, suggesting a further pathway for the antiangiogenic activity of drug. Thalidomide 171-182 vitronectin Homo sapiens 133-144 14558594-2 2003 The results show that thalidomide inhibits endotelial cell proliferation induced by bFGF and VEGF, more so if the cells are grown on vitronectin; moreover, treatment with thalidomide reduces the release of MMP-2 and IL-8 by endothelial cells, suggesting a further pathway for the antiangiogenic activity of drug. Thalidomide 171-182 matrix metallopeptidase 2 Homo sapiens 206-211 14558594-2 2003 The results show that thalidomide inhibits endotelial cell proliferation induced by bFGF and VEGF, more so if the cells are grown on vitronectin; moreover, treatment with thalidomide reduces the release of MMP-2 and IL-8 by endothelial cells, suggesting a further pathway for the antiangiogenic activity of drug. Thalidomide 171-182 C-X-C motif chemokine ligand 8 Homo sapiens 216-220 14577465-10 2003 In the Dhar-stimulated cells from the healthy staff thalidomide significantly suppressed TNF-alpha (P < 0.05). Thalidomide 52-63 tumor necrosis factor Homo sapiens 89-98 14577465-11 2003 A mixed effect was seen within and between the other groups, but there was a trend for thalidomide to suppress TNF-alpha induced by the M. leprae, Dhar and PPD antigens. Thalidomide 87-98 tumor necrosis factor Homo sapiens 111-120 14708607-0 2003 Thalidomide inhibits tumor necrosis factor-alpha production and antigen presentation by Langerhans cells. Thalidomide 0-11 tumor necrosis factor Mus musculus 21-48 14708607-2 2003 Thalidomide is believed to exert its anti-inflammatory effects, at least in part, by inhibiting tumor necrosis factor-alpha (TNF-alpha) production by monocytes. Thalidomide 0-11 tumor necrosis factor Mus musculus 96-123 14708607-2 2003 Thalidomide is believed to exert its anti-inflammatory effects, at least in part, by inhibiting tumor necrosis factor-alpha (TNF-alpha) production by monocytes. Thalidomide 0-11 tumor necrosis factor Mus musculus 125-134 14708607-5 2003 Using the murine epidermis-derived dendritic cell lines, XS106A from A/J mice and XS52 from BALB/c mice as surrogates for LC, we found that thalidomide inhibited TNF-alpha production in a concentration-dependent manner. Thalidomide 140-151 tumor necrosis factor Mus musculus 162-171 14708607-6 2003 Northern blot analysis revealed that thalidomide significantly decreased the peak-induced mRNA level of TNF-alpha in XS106A cells and XS52 cells. Thalidomide 37-48 tumor necrosis factor Mus musculus 104-113 14708607-8 2003 TNF-alpha production was reduced by 67.7% at a thalidomide concentration of 200 microg per mL. Thalidomide 47-58 tumor necrosis factor Mus musculus 0-9 14708607-9 2003 Thalidomide also had a profound inhibitory effect on the ability of LC to present antigen to a responsive TH1 clone. Thalidomide 0-11 negative elongation factor complex member C/D, Th1l Mus musculus 106-109 14708607-10 2003 Thalidomide inhibits TNF-alpha production and the antigen-presenting ability of epidermal LCs. Thalidomide 0-11 tumor necrosis factor Mus musculus 21-30 14551295-12 2003 CONCLUSION: The combination of frequently dosed IFN-alpha-2b and low-dose thalidomide is feasible and active in advanced RCC, but the clinical benefit may remain small compared with that of IFN alone. Thalidomide 74-85 interferon alpha 1 Homo sapiens 190-193 12867497-0 2003 Cyp1A1-mediated activation of thalidomide and suppression of embryo fibroblast proliferation. Thalidomide 30-41 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 0-6 12960498-2 2003 Tumor necrosis factor (TNF)-alpha is produced exclusively by the monocyte-macrophage lineage, which is mostly made up of Kupffer cells, and thalidomide has been shown to reduce TNF-alpha production from macrophages. Thalidomide 140-151 tumor necrosis factor Rattus norvegicus 177-186 12960498-12 2003 In isolated Kupffer cells, LPS-induced increases in [Ca2+]i and TNF-alpha production were suppressed by treatment with thalidomide. Thalidomide 119-130 tumor necrosis factor Rattus norvegicus 64-73 12960498-15 2003 CONCLUSIONS: Thalidomide prevents LPS-induced liver injury via mechanisms dependent on the suppression of TNF-alpha production from Kupffer cells. Thalidomide 13-24 tumor necrosis factor Rattus norvegicus 106-115 12736723-5 2003 In this report, we show that thalidomide downregulated the mRNA and protein expression of basic fibroblast growth factor (bFGF) in the human lung adenocarcinoma cell line A549, and that the effect of thalidomide was time and concentration-dependent. Thalidomide 29-40 fibroblast growth factor 2 Homo sapiens 122-126 12869646-7 2003 Significant reduction of EGFP was demonstrated in cells transfected with hTERT reporter gene and treated with racemic and (S)-Thd. Thalidomide 122-129 telomerase reverse transcriptase Homo sapiens 73-78 12878503-1 2003 Thalidomide, an agent which inhibits biosynthesis of tumor necrosis factor alpha (TNF-alpha) and which is used to treat a variety of chronic inflammatory conditions, was investigated as therapy for experimental sepsis. Thalidomide 0-11 tumor necrosis factor Homo sapiens 82-91 12878503-10 2003 Pretreatment of human monocytes with thalidomide prevented the secretion of TNF-alpha and IL-1beta but not that of IL-6. Thalidomide 37-48 tumor necrosis factor Homo sapiens 76-85 12878503-10 2003 Pretreatment of human monocytes with thalidomide prevented the secretion of TNF-alpha and IL-1beta but not that of IL-6. Thalidomide 37-48 interleukin 1 beta Homo sapiens 90-98 12878503-11 2003 It is concluded that thalidomide exerts a considerable anti-inflammatory effect by preventing evolution to sepsis and by decreasing the level of production of TNF-alpha and therefore deserves to be further evaluated in research for the therapy of sepsis. Thalidomide 21-32 tumor necrosis factor Homo sapiens 159-168 12819930-13 2003 Hypothetical target events/molecules of thalidomide include tumor necrosis factor-alpha production, nuclear androgen receptor, cyclooxygenases, aminopeptidases, and alpha-glucosidase. Thalidomide 40-51 sucrase-isomaltase Homo sapiens 165-182 12843316-0 2003 Mutations at the SALL4 locus on chromosome 20 result in a range of clinically overlapping phenotypes, including Okihiro syndrome, Holt-Oram syndrome, acro-renal-ocular syndrome, and patients previously reported to represent thalidomide embryopathy. Thalidomide 224-235 spalt like transcription factor 4 Homo sapiens 17-22 12843316-4 2003 We identified a novel SALL4 mutation in one family where the father was originally thought to have thalidomide embryopathy and had a daughter with a similar phenotype. Thalidomide 99-110 spalt like transcription factor 4 Homo sapiens 22-27 12843316-6 2003 Our results show that some cases of "thalidomide embryopathy" might be the result of SALL4 mutations, resulting in an increased risk for similarly affected offspring. Thalidomide 37-48 spalt like transcription factor 4 Homo sapiens 85-90 12736723-5 2003 In this report, we show that thalidomide downregulated the mRNA and protein expression of basic fibroblast growth factor (bFGF) in the human lung adenocarcinoma cell line A549, and that the effect of thalidomide was time and concentration-dependent. Thalidomide 29-40 fibroblast growth factor 2 Homo sapiens 90-120 12736723-6 2003 In contrast, the expression of vascular endothelial growth factor (VEGF) by thalidomide in these cells was in two phases. Thalidomide 76-87 vascular endothelial growth factor A Homo sapiens 31-65 12736723-6 2003 In contrast, the expression of vascular endothelial growth factor (VEGF) by thalidomide in these cells was in two phases. Thalidomide 76-87 vascular endothelial growth factor A Homo sapiens 67-71 12736723-7 2003 The mRNA and protein expression of VEGF was increased in the lung cancer cells treated with 0.6-6 microg/ml thalidomide, while higher concentrations of thalidomide decreased VEGF levels significantly in these cells. Thalidomide 108-119 vascular endothelial growth factor A Homo sapiens 35-39 12736723-7 2003 The mRNA and protein expression of VEGF was increased in the lung cancer cells treated with 0.6-6 microg/ml thalidomide, while higher concentrations of thalidomide decreased VEGF levels significantly in these cells. Thalidomide 152-163 vascular endothelial growth factor A Homo sapiens 174-178 12789488-5 2003 The mechanism of action of thalidomide is probably based on its immunomodulatory effect, namely the suppression of production of tumor necrosis factor alpha and the modulation of interleukins. Thalidomide 27-38 tumor necrosis factor Homo sapiens 129-156 12649301-0 2003 Enhancement of cytokine production and AP-1 transcriptional activity in T cells by thalidomide-related immunomodulatory drugs. Thalidomide 83-94 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 39-43 12642474-0 2003 Thalidomide-induced suppression of embryo fibroblast proliferation requires CYP1A1-mediated activation. Thalidomide 0-11 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 76-82 12894531-0 2003 Thalidomide down-regulates the expression of VEGF and bFGF in cisplatin-resistant human lung carcinoma cells. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 45-49 12894531-0 2003 Thalidomide down-regulates the expression of VEGF and bFGF in cisplatin-resistant human lung carcinoma cells. Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 54-58 12894531-4 2003 In the current study, we showed that thalidomide suppressed the expression of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in cisplatin-resistant human A549DDP lung carcinoma cells. Thalidomide 37-48 vascular endothelial growth factor A Homo sapiens 78-112 12894531-4 2003 In the current study, we showed that thalidomide suppressed the expression of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in cisplatin-resistant human A549DDP lung carcinoma cells. Thalidomide 37-48 vascular endothelial growth factor A Homo sapiens 114-118 12894531-4 2003 In the current study, we showed that thalidomide suppressed the expression of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in cisplatin-resistant human A549DDP lung carcinoma cells. Thalidomide 37-48 fibroblast growth factor 2 Homo sapiens 124-154 12894531-4 2003 In the current study, we showed that thalidomide suppressed the expression of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in cisplatin-resistant human A549DDP lung carcinoma cells. Thalidomide 37-48 fibroblast growth factor 2 Homo sapiens 156-160 12894531-5 2003 The mRNA levels of VEGF and bFGF were markedly decreased in the A549DDP cells treated with the therapeutic concentrations of thalidomide (0.6-6 micrograms/ml), as determined by RT-PCR analysis. Thalidomide 125-136 vascular endothelial growth factor A Homo sapiens 19-23 12894531-5 2003 The mRNA levels of VEGF and bFGF were markedly decreased in the A549DDP cells treated with the therapeutic concentrations of thalidomide (0.6-6 micrograms/ml), as determined by RT-PCR analysis. Thalidomide 125-136 fibroblast growth factor 2 Homo sapiens 28-32 12894531-6 2003 Consistent with these results, thalidomide also significantly reduced the protein levels of VEGF and bFGF in these cells in a dose- and time-dependent manner. Thalidomide 31-42 vascular endothelial growth factor A Homo sapiens 92-96 12894531-6 2003 Consistent with these results, thalidomide also significantly reduced the protein levels of VEGF and bFGF in these cells in a dose- and time-dependent manner. Thalidomide 31-42 fibroblast growth factor 2 Homo sapiens 101-105 12752784-4 2003 Inhibition of TNFalpha with thalidomide or a rat polyclonal anti-TNFalpha antibody attenuated carrageenan-induced hyperalgesia and prevented priming. Thalidomide 28-39 tumor necrosis factor Rattus norvegicus 14-22 12642474-8 2003 These results strongly suggest the involvement of CYP1A1 in the thalidomide-induced suppression of embryo fibroblast proliferation. Thalidomide 64-75 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 50-56 17041438-8 2003 There are rat experiments showing that thalidomide improves neuropathic pain, probably by selectively blocking tumor necrosis factor-alpha production in activated macrophages. Thalidomide 39-50 tumor necrosis factor Rattus norvegicus 111-138 12644816-2 2003 Thalidomide blocks the activity of angiogenic agents including bFGF, VEGF and IL-6. Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 63-67 12720152-2 2003 Although its molecular mechanisms of action have not yet been elucidated, thalidomide and the IMiDs affect a variety of cytokines and inflammatory mediators including tumor necrosis factor-alpha (TNFalpha), interleukin (IL)-1beta, interferon gamma (IFNgamma), IL-6, IL-10, IL-12, and COX-2 and angiogenesis factors such as vascular endothelial growth factor (VEGF) and its receptor. Thalidomide 74-85 tumor necrosis factor Homo sapiens 167-194 12720152-2 2003 Although its molecular mechanisms of action have not yet been elucidated, thalidomide and the IMiDs affect a variety of cytokines and inflammatory mediators including tumor necrosis factor-alpha (TNFalpha), interleukin (IL)-1beta, interferon gamma (IFNgamma), IL-6, IL-10, IL-12, and COX-2 and angiogenesis factors such as vascular endothelial growth factor (VEGF) and its receptor. Thalidomide 74-85 tumor necrosis factor Homo sapiens 196-204 12720152-2 2003 Although its molecular mechanisms of action have not yet been elucidated, thalidomide and the IMiDs affect a variety of cytokines and inflammatory mediators including tumor necrosis factor-alpha (TNFalpha), interleukin (IL)-1beta, interferon gamma (IFNgamma), IL-6, IL-10, IL-12, and COX-2 and angiogenesis factors such as vascular endothelial growth factor (VEGF) and its receptor. Thalidomide 74-85 interleukin 1 beta Homo sapiens 207-229 12720152-2 2003 Although its molecular mechanisms of action have not yet been elucidated, thalidomide and the IMiDs affect a variety of cytokines and inflammatory mediators including tumor necrosis factor-alpha (TNFalpha), interleukin (IL)-1beta, interferon gamma (IFNgamma), IL-6, IL-10, IL-12, and COX-2 and angiogenesis factors such as vascular endothelial growth factor (VEGF) and its receptor. Thalidomide 74-85 interferon gamma Homo sapiens 231-247 12720152-2 2003 Although its molecular mechanisms of action have not yet been elucidated, thalidomide and the IMiDs affect a variety of cytokines and inflammatory mediators including tumor necrosis factor-alpha (TNFalpha), interleukin (IL)-1beta, interferon gamma (IFNgamma), IL-6, IL-10, IL-12, and COX-2 and angiogenesis factors such as vascular endothelial growth factor (VEGF) and its receptor. Thalidomide 74-85 interferon gamma Homo sapiens 249-257 12720152-2 2003 Although its molecular mechanisms of action have not yet been elucidated, thalidomide and the IMiDs affect a variety of cytokines and inflammatory mediators including tumor necrosis factor-alpha (TNFalpha), interleukin (IL)-1beta, interferon gamma (IFNgamma), IL-6, IL-10, IL-12, and COX-2 and angiogenesis factors such as vascular endothelial growth factor (VEGF) and its receptor. Thalidomide 74-85 interleukin 6 Homo sapiens 260-264 12720152-2 2003 Although its molecular mechanisms of action have not yet been elucidated, thalidomide and the IMiDs affect a variety of cytokines and inflammatory mediators including tumor necrosis factor-alpha (TNFalpha), interleukin (IL)-1beta, interferon gamma (IFNgamma), IL-6, IL-10, IL-12, and COX-2 and angiogenesis factors such as vascular endothelial growth factor (VEGF) and its receptor. Thalidomide 74-85 interleukin 10 Homo sapiens 266-271 12720152-2 2003 Although its molecular mechanisms of action have not yet been elucidated, thalidomide and the IMiDs affect a variety of cytokines and inflammatory mediators including tumor necrosis factor-alpha (TNFalpha), interleukin (IL)-1beta, interferon gamma (IFNgamma), IL-6, IL-10, IL-12, and COX-2 and angiogenesis factors such as vascular endothelial growth factor (VEGF) and its receptor. Thalidomide 74-85 mitochondrially encoded cytochrome c oxidase II Homo sapiens 284-289 12720152-2 2003 Although its molecular mechanisms of action have not yet been elucidated, thalidomide and the IMiDs affect a variety of cytokines and inflammatory mediators including tumor necrosis factor-alpha (TNFalpha), interleukin (IL)-1beta, interferon gamma (IFNgamma), IL-6, IL-10, IL-12, and COX-2 and angiogenesis factors such as vascular endothelial growth factor (VEGF) and its receptor. Thalidomide 74-85 vascular endothelial growth factor A Homo sapiens 323-357 12720152-2 2003 Although its molecular mechanisms of action have not yet been elucidated, thalidomide and the IMiDs affect a variety of cytokines and inflammatory mediators including tumor necrosis factor-alpha (TNFalpha), interleukin (IL)-1beta, interferon gamma (IFNgamma), IL-6, IL-10, IL-12, and COX-2 and angiogenesis factors such as vascular endothelial growth factor (VEGF) and its receptor. Thalidomide 74-85 vascular endothelial growth factor A Homo sapiens 359-363 12644816-2 2003 Thalidomide blocks the activity of angiogenic agents including bFGF, VEGF and IL-6. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 69-73 12644816-2 2003 Thalidomide blocks the activity of angiogenic agents including bFGF, VEGF and IL-6. Thalidomide 0-11 interleukin 6 Homo sapiens 78-82 12874961-0 2003 Thalidomide inhibits early atherogenesis in apoE-deficient mice. Thalidomide 0-11 apolipoprotein E Mus musculus 44-48 12566301-0 2003 A novel subclass of thalidomide analogue with anti-solid tumor activity in which caspase-dependent apoptosis is associated with altered expression of bcl-2 family proteins. Thalidomide 20-31 B cell leukemia/lymphoma 2 Mus musculus 150-155 12749503-9 2003 RESULTS: Thalidomide is thought to exert its therapeutic effect through the modulation of cytokines, particularly tumor necrosis factor-alpha. Thalidomide 9-20 tumor necrosis factor Homo sapiens 114-141 12525736-0 2003 Phase I study of thalidomide for the treatment of plexiform neurofibroma in neurofibromatosis 1. Thalidomide 17-28 neurofibromin 1 Homo sapiens 76-95 12525736-1 2003 An open-label phase I trial of thalidomide (TL) in 20 patients with neurofibromatosis 1 (NF1) treated symptomatic plexiform neurofibroma (PNF). Thalidomide 31-42 neurofibromin 1 Homo sapiens 68-87 12525736-1 2003 An open-label phase I trial of thalidomide (TL) in 20 patients with neurofibromatosis 1 (NF1) treated symptomatic plexiform neurofibroma (PNF). Thalidomide 31-42 neurofibromin 1 Homo sapiens 89-92 12660941-0 2003 Thalidomide and a thalidomide analogue drug costimulate virus-specific CD8+ T cells in vitro. Thalidomide 0-11 CD8a molecule Homo sapiens 71-74 12660941-0 2003 Thalidomide and a thalidomide analogue drug costimulate virus-specific CD8+ T cells in vitro. Thalidomide 18-29 CD8a molecule Homo sapiens 71-74 12660941-3 2003 The recent finding that thalidomide acts as a T cell costimulator suggested that this drug may boost antiviral CD8(+) T cell responses. Thalidomide 24-35 CD8a molecule Homo sapiens 111-114 12660941-4 2003 In this in vitro study, in a human autologous CD8(+) T cell/dendritic cell (DC) coculture system, thalidomide and a potent thalidomide analogue were shown to enhance virus-specific CD8(+) T cell cytokine production and cytotoxic activity. Thalidomide 98-109 CD8a molecule Homo sapiens 46-49 12660941-4 2003 In this in vitro study, in a human autologous CD8(+) T cell/dendritic cell (DC) coculture system, thalidomide and a potent thalidomide analogue were shown to enhance virus-specific CD8(+) T cell cytokine production and cytotoxic activity. Thalidomide 98-109 CD8a molecule Homo sapiens 181-184 12660941-4 2003 In this in vitro study, in a human autologous CD8(+) T cell/dendritic cell (DC) coculture system, thalidomide and a potent thalidomide analogue were shown to enhance virus-specific CD8(+) T cell cytokine production and cytotoxic activity. Thalidomide 123-134 CD8a molecule Homo sapiens 181-184 12548578-4 2003 For example, immunomodulatory derivatives of thalidomide (IMiDs) and the proteasome inhibitor PS-341 both induce apoptosis of MM cell lines and patient cells refractory to melphalan, doxorubicin, and dexamethasone; abrogate MM cell binding to fibronectin and BMSCs and related protection against immune- and drug-induced apoptosis; block production of cytokines which promote MM cell growth, survival, drug resistance, and migration; inhibit angiogenesis; and stimulate host anti-tumor immunity. Thalidomide 45-56 fibronectin 1 Homo sapiens 243-254 15040702-4 2003 Thalidomide (TH) and systemic steroids (S), both TNFa production inhibitors, are the two current effective drugs for the management of ENL. Thalidomide 0-11 tumor necrosis factor Homo sapiens 49-53 12874961-3 2003 To clarify the role of this cytokine, we administered thalidomide, a compound known to inhibit TNF-alpha production, to homozygous apolipoprotein E-deficient (apoE-/-) mice in order to examine the effect of thalidomide on the development of early atherosclerotic lesions. Thalidomide 54-65 tumor necrosis factor Mus musculus 95-104 12533164-1 2003 BACKGROUND: Thalidomide is an anti-inflammatory agent and an immunomodulator that inhibits the production of tumor necrosis factor alpha. Thalidomide 12-23 tumor necrosis factor Homo sapiens 109-136 12874961-7 2003 Thus, thalidomide is capable of inhibiting the early development of atherosclerosis, presumably by inhibition of TNF-alpha secretion. Thalidomide 6-17 tumor necrosis factor Mus musculus 113-122 12771492-5 2003 Thalidomide reduces the activity of tumor necrosis factor (TNF) alpha by accelerating the degradation of its mRNA. Thalidomide 0-11 tumor necrosis factor Homo sapiens 36-69 12613280-4 2003 Thalidomide mainly reduces tumor necrosis factor (TNF)-alpha production by macrophages, and its TNF alpha antagonist properties explain the beneficial effects in several TNF alpha-associated complications of severe diseases. Thalidomide 0-11 tumor necrosis factor Homo sapiens 27-60 12613280-4 2003 Thalidomide mainly reduces tumor necrosis factor (TNF)-alpha production by macrophages, and its TNF alpha antagonist properties explain the beneficial effects in several TNF alpha-associated complications of severe diseases. Thalidomide 0-11 tumor necrosis factor Homo sapiens 170-179 12560891-0 2003 Thalidomide blocking of particle-induced TNFalpha release in vitro. Thalidomide 0-11 tumor necrosis factor Homo sapiens 41-49 12469200-4 2003 In this study, we demonstrate increased expression and production levels of VEGF in myeloma compared to non-myelomatous hematological lines, resistance to hypoxia and enhancement of VEGF-A production by hypoxia in myeloma, and direct growth inhibition of myeloma cells due to apoptosis and G1 arrest caused by TNFalpha upregulation induced by thalidomide. Thalidomide 343-354 vascular endothelial growth factor A Homo sapiens 76-80 12560891-9 2003 Thalidomide treatment was found to inhibit TNFalpha production in a dose-dependent manner in human macrophages exposed to Ti particles. Thalidomide 0-11 tumor necrosis factor Homo sapiens 43-51 14657598-10 2003 Another 6 of the 42 patients with elevated alpha-FP levels before treatment had a more than 50% decrease in their alpha-FP levels after thalidomide treatment. Thalidomide 136-147 alpha fetoprotein Homo sapiens 43-51 14657598-10 2003 Another 6 of the 42 patients with elevated alpha-FP levels before treatment had a more than 50% decrease in their alpha-FP levels after thalidomide treatment. Thalidomide 136-147 alpha fetoprotein Homo sapiens 114-122 12422154-9 2002 CONCLUSIONS: In this pilot study, therapy with thalidomide in patients with CHF resulted in decreased TNF-alpha levels and increased cardiac performance. Thalidomide 47-58 tumor necrosis factor Homo sapiens 102-111 14649541-2 2003 Thalidomide inhibits TNF induction by DMXAA and also potentiates its antitumor activity. Thalidomide 0-11 tumor necrosis factor Mus musculus 21-24 12520740-5 2002 Change of CD20 expression on the myeloma cells were measured by flow cytometer after and before myeloma cells were treated with thalidomide. Thalidomide 128-139 keratin 20 Homo sapiens 10-14 12520740-7 2002 CONCLUSION: Thalidomide could enhance the inhibition of Mabthera on colony formation of MM patients" myeloma cells, which is related to that thalidomide enhances CD20 antigen expression of myeloma cells. Thalidomide 12-23 keratin 20 Homo sapiens 162-166 12520740-7 2002 CONCLUSION: Thalidomide could enhance the inhibition of Mabthera on colony formation of MM patients" myeloma cells, which is related to that thalidomide enhances CD20 antigen expression of myeloma cells. Thalidomide 141-152 keratin 20 Homo sapiens 162-166 16088654-9 2002 Among the new anti-tumor necrosis factor drugs are thalidomide and infliximab, an antibody to tumor necrosis factor. Thalidomide 51-62 tumor necrosis factor Homo sapiens 19-40 16985953-6 2003 We performed a phase II trial of docetaxel with or without thalidomide in patients with AIPC and demonstrated encouraging response rates with combination therapy. Thalidomide 59-70 PDZ domain containing 2 Homo sapiens 88-92 15058168-2 2003 The main mechanism of action of thalidomide in these diseases is probably based on the suppression of tumor necrosis factor-alpha and the modulation of interleukins. Thalidomide 32-43 tumor necrosis factor Homo sapiens 102-129 12490072-10 2002 Thalidomide has no effect on tumor growth, but is also associated with increased VEGF and p53 expression. Thalidomide 0-11 transformation related protein 53, pseudogene Mus musculus 90-93 12485902-0 2002 Thalidomide suppresses the interleukin 1beta-induced NFkappaB signaling pathway in colon cancer cells. Thalidomide 0-11 interleukin 1 beta Homo sapiens 27-44 12485902-0 2002 Thalidomide suppresses the interleukin 1beta-induced NFkappaB signaling pathway in colon cancer cells. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 53-61 12485902-3 2002 We investigated the action mechanism of thalidomide through the NFkappaB pathway. Thalidomide 40-51 nuclear factor kappa B subunit 1 Homo sapiens 64-72 12485902-4 2002 Thalidomide inhibited interleukin (IL) 1beta-induced NFkappaB transcriptional activation and IL-8 production in Caco-2 colon cancer cells. Thalidomide 0-11 interleukin 1 beta Homo sapiens 22-44 12485902-4 2002 Thalidomide inhibited interleukin (IL) 1beta-induced NFkappaB transcriptional activation and IL-8 production in Caco-2 colon cancer cells. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 53-61 12485902-4 2002 Thalidomide inhibited interleukin (IL) 1beta-induced NFkappaB transcriptional activation and IL-8 production in Caco-2 colon cancer cells. Thalidomide 0-11 C-X-C motif chemokine ligand 8 Homo sapiens 93-97 12485902-5 2002 In addition, thalidomide suppressed NFkappaB nuclear translocation, IkappaB degradation, and NFkappaB-inducing kinase (NIK)-induced NFkappaB transcriptional activation. Thalidomide 13-24 nuclear factor kappa B subunit 1 Homo sapiens 36-44 12485902-5 2002 In addition, thalidomide suppressed NFkappaB nuclear translocation, IkappaB degradation, and NFkappaB-inducing kinase (NIK)-induced NFkappaB transcriptional activation. Thalidomide 13-24 nuclear factor kappa B subunit 1 Homo sapiens 93-101 12485902-5 2002 In addition, thalidomide suppressed NFkappaB nuclear translocation, IkappaB degradation, and NFkappaB-inducing kinase (NIK)-induced NFkappaB transcriptional activation. Thalidomide 13-24 nuclear factor kappa B subunit 1 Homo sapiens 93-101 12485902-6 2002 These results suggest that the molecular target of the effects of thalidomide may be IkappaB phosphorylation by IkappaB kinase (IKK), whose activation follows NIK activation and precedes IkappaB degradation in the NFkappaB pathway. Thalidomide 66-77 nuclear factor kappa B subunit 1 Homo sapiens 214-222 12463469-4 2002 Inhibition of NF-kappaB and/or TNFalpha is a putative mechanism for thalidomide efficacy in AS. Thalidomide 68-79 nuclear factor kappa B subunit 1 Homo sapiens 14-23 12463469-4 2002 Inhibition of NF-kappaB and/or TNFalpha is a putative mechanism for thalidomide efficacy in AS. Thalidomide 68-79 tumor necrosis factor Homo sapiens 31-39 12398921-1 2002 Thalidomide has been shown to reduce the production of tumor necrosis factor-alpha (TNF-alpha), a cytokine with deleterious pathophysiologic effects in various diseases. Thalidomide 0-11 tumor necrosis factor Mus musculus 55-82 12398921-1 2002 Thalidomide has been shown to reduce the production of tumor necrosis factor-alpha (TNF-alpha), a cytokine with deleterious pathophysiologic effects in various diseases. Thalidomide 0-11 tumor necrosis factor Mus musculus 84-93 12398921-2 2002 In search of thalidomide analogues with improved TNF-alpha inhibiting properties, 5-ethyl-1-phenyl-5-(3,4,5,6-tetrafluorophthalimido)barbituric acid (TFBA) was found to be superior to thalidomide. Thalidomide 13-24 tumor necrosis factor Mus musculus 49-58 12642692-0 2002 Pharmacogenetic associations of CYP2C19 genotype with in vivo metabolisms and pharmacological effects of thalidomide. Thalidomide 105-116 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 32-39 12642692-10 2002 The role of CYP2C19 polymorphism in thalidomide treatments remains to be elucidated. Thalidomide 36-47 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 12-19 12490072-6 2002 RESULTS: At 6 weeks, VEGF-mAb-treated tumors were smaller (1603 +/- 296 mm(3)) than either the thalidomide-treated (6007 +/- 1498 mm(3); p = 0.008) or the control groups (4040 +/- 831 mm(3); p = 0.014) and the VEGF-mAb-treated animals maintained greater weight (30.4 +/- 0.84 g at week 6 versus thalidomide-treated, 26.0 +/- 0.94 g, p = 0.003; and control, 25.8 +/- 0.78 g, p = 0.001 animals). Thalidomide 295-306 vascular endothelial growth factor A Mus musculus 21-25 12490072-10 2002 Thalidomide has no effect on tumor growth, but is also associated with increased VEGF and p53 expression. Thalidomide 0-11 vascular endothelial growth factor A Mus musculus 81-85 12358938-0 2002 Thalidomide treatment of relapsed multiple myeloma patients and changes in circulating VEGF and bFGF. Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 96-100 12242718-11 2002 Intravenous cotreatment with PBN and thalidomide (gavage) in rabbits restored normal patterns and localization of Twist, Fgf-8, and Fgf-10 expression. Thalidomide 37-48 fibroblast growth factor 10 Oryctolagus cuniculus 132-138 12296856-1 2002 Thalidomide (Thd) is clinically useful in a number of conditions where its efficacy is probably related to its anti-TNF-alpha activity. Thalidomide 0-11 tumor necrosis factor Homo sapiens 116-125 12296856-1 2002 Thalidomide (Thd) is clinically useful in a number of conditions where its efficacy is probably related to its anti-TNF-alpha activity. Thalidomide 13-16 tumor necrosis factor Homo sapiens 116-125 12296856-3 2002 However, in contrast to their known suppressive effects during inflammatory stimuli, the effects of Thd/IMiDs on TNF-alpha and TNF receptors (TNFRs) during T cell co-stimulation are not known. Thalidomide 100-103 tumor necrosis factor Homo sapiens 113-122 12296856-3 2002 However, in contrast to their known suppressive effects during inflammatory stimuli, the effects of Thd/IMiDs on TNF-alpha and TNF receptors (TNFRs) during T cell co-stimulation are not known. Thalidomide 100-103 tumor necrosis factor Homo sapiens 113-116 12296856-5 2002 We found that co-stimulation of PBMC with Thd/IMiDs, but not CC-3052, prevented alphaCD3-induced T cell surface expression of TNFR2 and thereby reduced soluble TNFR2 (sTNFR2) levels. Thalidomide 42-45 TNF receptor superfamily member 1B Homo sapiens 126-131 12296856-5 2002 We found that co-stimulation of PBMC with Thd/IMiDs, but not CC-3052, prevented alphaCD3-induced T cell surface expression of TNFR2 and thereby reduced soluble TNFR2 (sTNFR2) levels. Thalidomide 42-45 TNF receptor superfamily member 1B Homo sapiens 160-165 12296856-7 2002 The extent of co-stimulation by Thd/IMiDs (assessed by CD69/CD25 expression and IL-2/sIL-2Ralpha production) was similar for CD4+ and CD8+ T lymphocytes and correlated with TNFR2 inhibition. Thalidomide 32-35 CD69 molecule Homo sapiens 55-59 12296856-7 2002 The extent of co-stimulation by Thd/IMiDs (assessed by CD69/CD25 expression and IL-2/sIL-2Ralpha production) was similar for CD4+ and CD8+ T lymphocytes and correlated with TNFR2 inhibition. Thalidomide 32-35 interleukin 2 receptor subunit alpha Homo sapiens 60-64 12296856-7 2002 The extent of co-stimulation by Thd/IMiDs (assessed by CD69/CD25 expression and IL-2/sIL-2Ralpha production) was similar for CD4+ and CD8+ T lymphocytes and correlated with TNFR2 inhibition. Thalidomide 32-35 interleukin 2 Homo sapiens 80-84 12296856-7 2002 The extent of co-stimulation by Thd/IMiDs (assessed by CD69/CD25 expression and IL-2/sIL-2Ralpha production) was similar for CD4+ and CD8+ T lymphocytes and correlated with TNFR2 inhibition. Thalidomide 32-35 CD4 molecule Homo sapiens 125-128 12296856-7 2002 The extent of co-stimulation by Thd/IMiDs (assessed by CD69/CD25 expression and IL-2/sIL-2Ralpha production) was similar for CD4+ and CD8+ T lymphocytes and correlated with TNFR2 inhibition. Thalidomide 32-35 CD8a molecule Homo sapiens 134-137 12296856-7 2002 The extent of co-stimulation by Thd/IMiDs (assessed by CD69/CD25 expression and IL-2/sIL-2Ralpha production) was similar for CD4+ and CD8+ T lymphocytes and correlated with TNFR2 inhibition. Thalidomide 32-35 TNF receptor superfamily member 1B Homo sapiens 173-178 12296856-10 2002 Together, these results suggest a possible role for TNF-mediated events during co-stimulation and contrast with the TNF inhibitory effects of Thd and its analogues during inflammatory stimuli. Thalidomide 142-145 tumor necrosis factor Homo sapiens 116-119 12296856-0 2002 Thalidomide and its analogues have distinct and opposing effects on TNF-alpha and TNFR2 during co-stimulation of both CD4(+) and CD8(+) T cells. Thalidomide 0-11 tumor necrosis factor Homo sapiens 68-77 12296856-0 2002 Thalidomide and its analogues have distinct and opposing effects on TNF-alpha and TNFR2 during co-stimulation of both CD4(+) and CD8(+) T cells. Thalidomide 0-11 TNF receptor superfamily member 1B Homo sapiens 82-87 12296856-0 2002 Thalidomide and its analogues have distinct and opposing effects on TNF-alpha and TNFR2 during co-stimulation of both CD4(+) and CD8(+) T cells. Thalidomide 0-11 CD4 molecule Homo sapiens 118-121 12296856-0 2002 Thalidomide and its analogues have distinct and opposing effects on TNF-alpha and TNFR2 during co-stimulation of both CD4(+) and CD8(+) T cells. Thalidomide 0-11 CD8a molecule Homo sapiens 129-132 12242718-9 2002 In situ hybridization showed a preferential decrease in Twist, Fgf-8, and Fgf-10 expression after thalidomide treatment (400 mg/kg per day) in rabbit embryos. Thalidomide 98-109 fibroblast growth factor 8 Oryctolagus cuniculus 63-68 12242718-9 2002 In situ hybridization showed a preferential decrease in Twist, Fgf-8, and Fgf-10 expression after thalidomide treatment (400 mg/kg per day) in rabbit embryos. Thalidomide 98-109 fibroblast growth factor 10 Oryctolagus cuniculus 74-80 12242718-11 2002 Intravenous cotreatment with PBN and thalidomide (gavage) in rabbits restored normal patterns and localization of Twist, Fgf-8, and Fgf-10 expression. Thalidomide 37-48 fibroblast growth factor 8 Oryctolagus cuniculus 121-126 12482346-13 2002 CONCLUSION: MVD and VEGF concentration were decreased obviously by thalidomide treatment. Thalidomide 67-78 vascular endothelial growth factor A Homo sapiens 20-24 12484444-3 2002 On the other hand, when the culture was treated with thalidomide plus cytochrome P-450, both types of thalidomides significantly inhibited angiogenesis. Thalidomide 102-114 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 70-86 12484444-7 2002 The inhibition of angiogenesis by thalidomide and cytochrome P-450 takes place through a suppression of TNF-alpha and involves the metabolism of the thalidomide. Thalidomide 34-45 tumor necrosis factor Mus musculus 104-113 12484444-7 2002 The inhibition of angiogenesis by thalidomide and cytochrome P-450 takes place through a suppression of TNF-alpha and involves the metabolism of the thalidomide. Thalidomide 149-160 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 50-66 12368119-12 2002 It also indicates the role of TNF-alpha upon the pathophysiology of this event based on the inhibitory action of DEXA, TAL and pentoxifylline, and on TNF-alpha secretion induced by cholera toxin. Thalidomide 119-122 tumor necrosis factor Rattus norvegicus 30-39 12411981-2 2002 Thalidomide has anti-inflammatory properties and selectively inhibits TNF. Thalidomide 0-11 tumor necrosis factor Homo sapiens 70-73 12411981-7 2002 RESULTS: Thalidomide and thalidomide analogues inhibited LPS-induced TNF biosynthesis in cardiac myocytes in a dose-dependent manner. Thalidomide 9-20 tumor necrosis factor Homo sapiens 69-72 12411981-7 2002 RESULTS: Thalidomide and thalidomide analogues inhibited LPS-induced TNF biosynthesis in cardiac myocytes in a dose-dependent manner. Thalidomide 25-36 tumor necrosis factor Homo sapiens 69-72 12411981-8 2002 Thalidomide analogues had a greater inhibitory effect on TNF production than did thalidomide. Thalidomide 0-11 tumor necrosis factor Homo sapiens 57-60 12200397-0 2002 Polymorphisms of the tumor necrosis factor-alpha gene promoter predict for outcome after thalidomide therapy in relapsed and refractory multiple myeloma. Thalidomide 89-100 tumor necrosis factor Homo sapiens 21-48 12200397-1 2002 Thalidomide (Thal) is a drug with antiangiogenic, anti-inflammatory, and immunomodulatory properties that was found to inhibit the production of tumor necrosis factor-alpha (TNF-alpha) in vitro. Thalidomide 0-11 tumor necrosis factor Homo sapiens 145-172 12200397-1 2002 Thalidomide (Thal) is a drug with antiangiogenic, anti-inflammatory, and immunomodulatory properties that was found to inhibit the production of tumor necrosis factor-alpha (TNF-alpha) in vitro. Thalidomide 0-11 tumor necrosis factor Homo sapiens 174-183 12200397-1 2002 Thalidomide (Thal) is a drug with antiangiogenic, anti-inflammatory, and immunomodulatory properties that was found to inhibit the production of tumor necrosis factor-alpha (TNF-alpha) in vitro. Thalidomide 0-4 tumor necrosis factor Homo sapiens 145-172 12200397-1 2002 Thalidomide (Thal) is a drug with antiangiogenic, anti-inflammatory, and immunomodulatory properties that was found to inhibit the production of tumor necrosis factor-alpha (TNF-alpha) in vitro. Thalidomide 0-4 tumor necrosis factor Homo sapiens 174-183 12200397-2 2002 We studied single nucleotide polymorphisms at positions -308 and -238 of the TNF-alpha gene promoter and measured the corresponding TNF-alpha cytokine levels in 81 patients (pts) with refractory and relapsed multiple myeloma (MM) who were treated with Thal. Thalidomide 252-256 tumor necrosis factor Homo sapiens 77-86 12380944-0 2002 PSA response to thalidomide in patients with advanced prostate cancer. Thalidomide 16-27 aminopeptidase puromycin sensitive Homo sapiens 0-3 12412207-2 2002 More interestingly, thalidomide has shown the ability to suppress tumor necrosis factor alpha (TNF alpha) production and to modify the expression of TNF alpha induced adhesion molecules on endothelial cells and on human leukocytes. Thalidomide 20-31 tumor necrosis factor Homo sapiens 66-93 12412207-2 2002 More interestingly, thalidomide has shown the ability to suppress tumor necrosis factor alpha (TNF alpha) production and to modify the expression of TNF alpha induced adhesion molecules on endothelial cells and on human leukocytes. Thalidomide 20-31 tumor necrosis factor Homo sapiens 95-104 12412207-2 2002 More interestingly, thalidomide has shown the ability to suppress tumor necrosis factor alpha (TNF alpha) production and to modify the expression of TNF alpha induced adhesion molecules on endothelial cells and on human leukocytes. Thalidomide 20-31 tumor necrosis factor Homo sapiens 149-158 12359057-11 2002 After thalidomide treatment, plasma vascular endothelial growth factor (VEGF) level was significantly reduced in responders (P = 0.025), but not in non-responders (P = 0.37). Thalidomide 6-17 vascular endothelial growth factor A Homo sapiens 36-70 12359057-11 2002 After thalidomide treatment, plasma vascular endothelial growth factor (VEGF) level was significantly reduced in responders (P = 0.025), but not in non-responders (P = 0.37). Thalidomide 6-17 vascular endothelial growth factor A Homo sapiens 72-76 12359057-12 2002 Reduction of plasma VEGF level might be an important indicator for anti-myeloma effect of thalidomide. Thalidomide 90-101 vascular endothelial growth factor A Homo sapiens 20-24 12349951-15 2002 Increased production of IL-2 by activated T cells may be a mechanism through which thalidomide exerts its immunomodulatory effects. Thalidomide 83-94 interleukin 2 Homo sapiens 24-28 12190008-4 2002 Thalidomide inhibits TNF alpha production, has T-cell costimulatory properties and modulates the expression of cell surface molecules on leukocytes in vivo. Thalidomide 0-11 tumor necrosis factor Homo sapiens 21-30 12190011-12 2002 Thalidomide plus dexamethasone was given to 9 patients who did not respond to thalidomide alone, and in 6 of them a reduction of M-protein was then observed. Thalidomide 0-11 myomesin 2 Homo sapiens 129-138 12167383-2 2002 AIMS: We have tried to antagonise the detrimental effects of TNFalpha on skeletal muscle apoptosis, by using thalidomide, a drug that inhibits its biosynthesis. Thalidomide 109-120 tumor necrosis factor Homo sapiens 61-69 12209719-2 2002 Thalidomide inhibits basic fibroblast growth factor and vascular endothelial growth factor (VEGF)-induced angiogenesis. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 56-90 12209719-2 2002 Thalidomide inhibits basic fibroblast growth factor and vascular endothelial growth factor (VEGF)-induced angiogenesis. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 92-96 12240951-2 2002 To determine whether the N-alkyl analogs of the thalidomide are active and stable, their stabilities in buffer and their abilities to inhibit tumor necrosis factor alpha (TNF-alpha) in vitro in human peripheral blood mononuclear cell cultures were investigated. Thalidomide 48-59 tumor necrosis factor Homo sapiens 142-169 12240951-2 2002 To determine whether the N-alkyl analogs of the thalidomide are active and stable, their stabilities in buffer and their abilities to inhibit tumor necrosis factor alpha (TNF-alpha) in vitro in human peripheral blood mononuclear cell cultures were investigated. Thalidomide 48-59 tumor necrosis factor Homo sapiens 171-180 12240951-7 2002 Like thalidomide, the N-alkyl analogs in this series inhibit an average of 60% of the TNF-alpha synthesis in lipopolysaccharide-stimulated peripheral blood mononuclear cell cultures. Thalidomide 5-16 tumor necrosis factor Homo sapiens 86-95 12371635-0 2002 Successful treatment of familial Mediterranean fever attacks with thalidomide in a colchicine resistant patient. Thalidomide 66-77 MEFV innate immuity regulator, pyrin Homo sapiens 24-52 12105857-0 2002 Thalidomide prevents alcoholic liver injury in rats through suppression of Kupffer cell sensitization and TNF-alpha production. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 106-115 12105857-2 2002 Thalidomide has been shown to suppress TNF-alpha production from macrophages. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 39-48 12105857-12 2002 Furthermore, thalidomide abolished the LPS-induced increase in CD14 expression and [Ca2+]i elevation in KCs. Thalidomide 13-24 CD14 molecule Rattus norvegicus 63-67 12105857-14 2002 Moreover, thalidomide reduced the LPS-induced TNF-alpha production by KCs by decreasing TNF-alpha messenger RNA. Thalidomide 10-21 tumor necrosis factor Rattus norvegicus 46-55 12105857-14 2002 Moreover, thalidomide reduced the LPS-induced TNF-alpha production by KCs by decreasing TNF-alpha messenger RNA. Thalidomide 10-21 tumor necrosis factor Rattus norvegicus 88-97 12105857-15 2002 CONCLUSIONS: These results collectively indicate that thalidomide prevents alcoholic liver injury through suppression of TNF-alpha production and abolishment of KC sensitization. Thalidomide 54-65 tumor necrosis factor Rattus norvegicus 121-130 12349951-0 2002 Thalidomide can costimulate or suppress CD4+ cells" ability to incorporate [H3]-thymidine--dependence on the primary stimulant. Thalidomide 0-11 CD4 molecule Homo sapiens 40-43 12349951-1 2002 Thalidomide is a drug that can enhance mitogen- and antigen-stimulated cells" ability to synthesize IL-2. Thalidomide 0-11 interleukin 2 Homo sapiens 100-104 12349951-2 2002 To assess if thalidomide could concomitantly enhance the synthesis of IFN-gamma and incorporation of [H3]-thymidine, peripheral blood mononuclear cells (PBMC) were incubated in the presence or absence of thalidomide and staphylococcal enterotoxin A (SEA), anti-CD3, Con-A or PHA. Thalidomide 13-24 interferon gamma Homo sapiens 70-79 12349951-5 2002 Regardless of the mitogen used to stimulate the PBMC, the thalidomide-treated PBMC produced more IL-2 than controls. Thalidomide 58-69 interleukin 2 Homo sapiens 97-101 12349951-6 2002 Thalidomide enhanced IFN-gamma synthesis in the Con-A and anti-CD3-stimulated PBMC. Thalidomide 0-11 interferon gamma Homo sapiens 21-30 11994441-0 2002 Protective antitumor immunity induced by a costimulatory thalidomide analog in conjunction with whole tumor cell vaccination is mediated by increased Th1-type immunity. Thalidomide 57-68 negative elongation factor complex member C/D Homo sapiens 150-153 12349951-9 2002 In the thalidomide-treated SEA-stimulated CD4+ and CD8+ cells, thalidomide acted as a costimulant to enhance the synthesis of IL-2. Thalidomide 7-18 CD4 molecule Homo sapiens 42-45 12349951-9 2002 In the thalidomide-treated SEA-stimulated CD4+ and CD8+ cells, thalidomide acted as a costimulant to enhance the synthesis of IL-2. Thalidomide 7-18 interleukin 2 Homo sapiens 126-130 12349951-9 2002 In the thalidomide-treated SEA-stimulated CD4+ and CD8+ cells, thalidomide acted as a costimulant to enhance the synthesis of IL-2. Thalidomide 63-74 CD4 molecule Homo sapiens 42-45 12349951-9 2002 In the thalidomide-treated SEA-stimulated CD4+ and CD8+ cells, thalidomide acted as a costimulant to enhance the synthesis of IL-2. Thalidomide 63-74 interleukin 2 Homo sapiens 126-130 12349951-10 2002 In the anti-CD3-stimulated thalidomide-treated cultures of PBMC enriched for CD4+ cells, thalidomide acted as a costimulant to enhance the incorporation [H3]-thymidine. Thalidomide 27-38 CD4 molecule Homo sapiens 77-80 12349951-10 2002 In the anti-CD3-stimulated thalidomide-treated cultures of PBMC enriched for CD4+ cells, thalidomide acted as a costimulant to enhance the incorporation [H3]-thymidine. Thalidomide 89-100 CD4 molecule Homo sapiens 77-80 12349951-11 2002 Thalidomide cooperated with all of the mitogens to enhance T-cell synthesis of IL-2. Thalidomide 0-11 interleukin 2 Homo sapiens 79-83 12349951-13 2002 The SEA-stimulated cells targeted by thalidomide to suppress incorporation of [H3]-thymidine were CD4+. Thalidomide 37-48 CD4 molecule Homo sapiens 98-101 12349951-14 2002 CD4+ cells stimulated with anti-CD3 were enhanced by thalidomide in their ability to synthesize IL-2 and to incorporate [H3]-thymidine. Thalidomide 53-64 CD4 molecule Homo sapiens 0-3 12349951-14 2002 CD4+ cells stimulated with anti-CD3 were enhanced by thalidomide in their ability to synthesize IL-2 and to incorporate [H3]-thymidine. Thalidomide 53-64 interleukin 2 Homo sapiens 96-100 12010810-2 2002 Several established or novel anti-multiple myeloma (anti-MM) agents, such as dexamethasone, thalidomide, and proteasome inhibitors (PS-341), inhibit NF-kappaB activity as part of their diverse actions. Thalidomide 92-103 nuclear factor kappa B subunit 1 Homo sapiens 149-158 12060642-1 2002 PURPOSE: This research investigated the biotransformation of thalidomide by cytochrome P-450 (CYP). Thalidomide 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 12060642-1 2002 PURPOSE: This research investigated the biotransformation of thalidomide by cytochrome P-450 (CYP). Thalidomide 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 12060642-2 2002 EXPERIMENTAL DESIGN: We used liver microsomes from humans and/or animals and the recombinant specific CYP isozymes to investigate CYP-mediated metabolism of thalidomide. Thalidomide 157-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 12060642-2 2002 EXPERIMENTAL DESIGN: We used liver microsomes from humans and/or animals and the recombinant specific CYP isozymes to investigate CYP-mediated metabolism of thalidomide. Thalidomide 157-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-133 12060642-11 2002 CONCLUSIONS: We discovered that the polymorphic enzyme CYP2C19 is responsible for 5- and 5"-hydroxylation of thalidomide in humans. Thalidomide 109-120 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 55-62 12060642-12 2002 In rats, thalidomide was hydroxylated extensively by CYP2C6 as well as the sex-specific enzyme CYP2C11. Thalidomide 9-20 cytochrome P450, family 2, subfamily C, polypeptide 6, variant 1 Rattus norvegicus 53-59 12060642-12 2002 In rats, thalidomide was hydroxylated extensively by CYP2C6 as well as the sex-specific enzyme CYP2C11. Thalidomide 9-20 cytochrome P450, subfamily 2, polypeptide 11 Rattus norvegicus 95-102 11994441-8 2002 Our results are the first to demonstrate that a costimulatory thalidomide analog can prime protective, long-lasting, tumor-specific, Th1-type responses in vivo and further support their ongoing clinical development as novel anti-cancer agents. Thalidomide 62-73 negative elongation factor complex member C/D Homo sapiens 133-136 12046682-1 2002 Thalidomide--removed from widespread clinical use by 1962 because of severe teratogenicity--has anti-angiogenic and immunomodulatory effects, including the inhibition of TNF alpha. Thalidomide 0-11 tumor necrosis factor Homo sapiens 170-179 11972510-11 2002 Patients with PTMM and/or markedly increased circulating CD34+ cell counts might be susceptible to thalidomide-induced thrombocytosis. Thalidomide 99-110 CD34 molecule Homo sapiens 57-61 11909713-1 2002 Thalidomide showed cyclooxygenase (COX)-1/2 inhibitory activity with a potency comparable to that of aspirin. Thalidomide 0-11 mitochondrially encoded cytochrome c oxidase I Homo sapiens 19-41 11956484-8 2002 of the selective TNF-alpha synthesis inhibitor thalidomide or 200 mg./kg. Thalidomide 47-58 tumor necrosis factor Mus musculus 17-26 12515619-0 2002 [Influence of thalidomide on interleukin-6 and its transmission in multiple myeloma patients]. Thalidomide 14-25 interleukin 6 Homo sapiens 29-42 12515619-1 2002 OBJECTIVE: To evaluate the mechanism and influence of thalidomide on interleukin-6 (IL-6), IL-6 receptor (IL-6R) and its transmitting chain in multiple myeloma patients. Thalidomide 54-65 interleukin 6 Homo sapiens 69-82 12515619-1 2002 OBJECTIVE: To evaluate the mechanism and influence of thalidomide on interleukin-6 (IL-6), IL-6 receptor (IL-6R) and its transmitting chain in multiple myeloma patients. Thalidomide 54-65 interleukin 6 Homo sapiens 84-88 12515619-1 2002 OBJECTIVE: To evaluate the mechanism and influence of thalidomide on interleukin-6 (IL-6), IL-6 receptor (IL-6R) and its transmitting chain in multiple myeloma patients. Thalidomide 54-65 interleukin 6 receptor Homo sapiens 91-104 12515619-1 2002 OBJECTIVE: To evaluate the mechanism and influence of thalidomide on interleukin-6 (IL-6), IL-6 receptor (IL-6R) and its transmitting chain in multiple myeloma patients. Thalidomide 54-65 interleukin 6 receptor Homo sapiens 106-111 12515619-3 2002 RESULTS: Serum level of IL-6 in multiple myeloma patients was 564.8 +/- 319.4 ng/L, with a positive rate on the myeloma cells of 33.6% before oral 200 mg/d thalidomide. Thalidomide 156-167 interleukin 6 Homo sapiens 24-28 12515619-8 2002 CONCLUSION: Reduction of serum level of IL-6 in multiple myeloma patients and decrease in IL-6R expression on the myeloma cells and IL-6R beta mRNA occur on D14 after oral 400 mg/d thalidomide. Thalidomide 181-192 interleukin 6 Homo sapiens 40-44 12515619-8 2002 CONCLUSION: Reduction of serum level of IL-6 in multiple myeloma patients and decrease in IL-6R expression on the myeloma cells and IL-6R beta mRNA occur on D14 after oral 400 mg/d thalidomide. Thalidomide 181-192 interleukin 6 receptor Homo sapiens 90-95 12515619-8 2002 CONCLUSION: Reduction of serum level of IL-6 in multiple myeloma patients and decrease in IL-6R expression on the myeloma cells and IL-6R beta mRNA occur on D14 after oral 400 mg/d thalidomide. Thalidomide 181-192 interleukin 6 cytokine family signal transducer Homo sapiens 132-142 12515619-10 2002 The antitumor mechanism of thalidomide may be related to reduction of IL-6 serum level in multiple myeloma patients and decrease in IL-6R expression on the myeloma cells and IL-6R beta mRNA. Thalidomide 27-38 interleukin 6 Homo sapiens 70-74 12515619-10 2002 The antitumor mechanism of thalidomide may be related to reduction of IL-6 serum level in multiple myeloma patients and decrease in IL-6R expression on the myeloma cells and IL-6R beta mRNA. Thalidomide 27-38 interleukin 6 receptor Homo sapiens 132-137 12515619-10 2002 The antitumor mechanism of thalidomide may be related to reduction of IL-6 serum level in multiple myeloma patients and decrease in IL-6R expression on the myeloma cells and IL-6R beta mRNA. Thalidomide 27-38 interleukin 6 cytokine family signal transducer Homo sapiens 174-184 11909713-2 2002 Structural development studies of thalidomide resulted in potent COX-1/2 inhibitors, and COX-1-selective and COX-2-selective inhibitors. Thalidomide 34-45 mitochondrially encoded cytochrome c oxidase I Homo sapiens 65-70 11909713-2 2002 Structural development studies of thalidomide resulted in potent COX-1/2 inhibitors, and COX-1-selective and COX-2-selective inhibitors. Thalidomide 34-45 mitochondrially encoded cytochrome c oxidase I Homo sapiens 89-94 11884428-0 2002 Thalidomide suppresses NF-kappa B activation induced by TNF and H2O2, but not that activated by ceramide, lipopolysaccharides, or phorbol ester. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 23-33 11995671-5 2002 In this case report, we present a patient with MRS that was treated with thalidomide because of the identification of tumor necrosis factor a in the lesion by immunohistochemical analysis. Thalidomide 73-84 tumor necrosis factor Homo sapiens 118-139 11942970-9 2002 There was also a trend to increased expression of FGF-2 and tumour necrosis factor-alpha in thalidomide-treated tumours. Thalidomide 92-103 fibroblast growth factor 2 Homo sapiens 50-55 11940485-13 2002 Plasma cells from patients who responded favorably to thalidomide secreted a significantly lower amount of VEGF than plasma cells from resistant patients (126.45 165 pg/mL vs 227.11 70 pg/mL, p=0.04). Thalidomide 54-65 vascular endothelial growth factor A Homo sapiens 107-111 11884428-2 2002 Due to the central role of NF-kappaB in these responses, we postulated that thalidomide mediates its effects through suppression of NF-kappaB activation. Thalidomide 76-87 nuclear factor kappa B subunit 1 Homo sapiens 27-36 11884428-2 2002 Due to the central role of NF-kappaB in these responses, we postulated that thalidomide mediates its effects through suppression of NF-kappaB activation. Thalidomide 76-87 nuclear factor kappa B subunit 1 Homo sapiens 132-141 11884428-0 2002 Thalidomide suppresses NF-kappa B activation induced by TNF and H2O2, but not that activated by ceramide, lipopolysaccharides, or phorbol ester. Thalidomide 0-11 tumor necrosis factor Homo sapiens 56-59 11884428-3 2002 We investigated the effects of thalidomide on NF-kappaB activation induced by various inflammatory agents in Jurkat cells. Thalidomide 31-42 nuclear factor kappa B subunit 1 Homo sapiens 46-55 11884428-4 2002 The treatment of these cells with thalidomide suppressed TNF-induced NF-kappaB activation, with optimum effect occurring at 50 microg/ml thalidomide. Thalidomide 34-45 tumor necrosis factor Homo sapiens 57-60 11788559-3 2002 Suppression of tumour necrosis factor alpha (TNF-alpha) by thalidomide has been suggested as a possible mechanism. Thalidomide 59-70 tumor necrosis factor Homo sapiens 45-54 11884428-4 2002 The treatment of these cells with thalidomide suppressed TNF-induced NF-kappaB activation, with optimum effect occurring at 50 microg/ml thalidomide. Thalidomide 34-45 nuclear factor kappa B subunit 1 Homo sapiens 69-78 11884428-4 2002 The treatment of these cells with thalidomide suppressed TNF-induced NF-kappaB activation, with optimum effect occurring at 50 microg/ml thalidomide. Thalidomide 137-148 tumor necrosis factor Homo sapiens 57-60 11884428-4 2002 The treatment of these cells with thalidomide suppressed TNF-induced NF-kappaB activation, with optimum effect occurring at 50 microg/ml thalidomide. Thalidomide 137-148 nuclear factor kappa B subunit 1 Homo sapiens 69-78 11884428-8 2002 Thalidomide abolished the NF-kappaB-dependent reporter gene expression activated by overexpression of TNFR1, TNFR-associated factor-2, and NF-kappaB-inducing kinase, but not that activated by the p65 subunit of NF-kappaB. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 26-35 11884428-8 2002 Thalidomide abolished the NF-kappaB-dependent reporter gene expression activated by overexpression of TNFR1, TNFR-associated factor-2, and NF-kappaB-inducing kinase, but not that activated by the p65 subunit of NF-kappaB. Thalidomide 0-11 TNF receptor superfamily member 1A Homo sapiens 102-107 11884428-8 2002 Thalidomide abolished the NF-kappaB-dependent reporter gene expression activated by overexpression of TNFR1, TNFR-associated factor-2, and NF-kappaB-inducing kinase, but not that activated by the p65 subunit of NF-kappaB. Thalidomide 0-11 nuclear factor kappa B subunit 1 Homo sapiens 139-148 11884428-9 2002 Overall, our results clearly demonstrate that thalidomide suppresses NF-kappaB activation specifically induced by TNF and H(2)O(2) and that this may contribute to its role in suppression of proliferation, inflammation, angiogenesis, and the immune system. Thalidomide 46-57 nuclear factor kappa B subunit 1 Homo sapiens 69-78 11884428-9 2002 Overall, our results clearly demonstrate that thalidomide suppresses NF-kappaB activation specifically induced by TNF and H(2)O(2) and that this may contribute to its role in suppression of proliferation, inflammation, angiogenesis, and the immune system. Thalidomide 46-57 tumor necrosis factor Homo sapiens 114-117 11905505-0 2002 Management of erythema nodosum leprosum by thalidomide: thalidomide analogues inhibit M. leprae-induced TNFalpha production in vitro. Thalidomide 56-67 tumor necrosis factor Homo sapiens 104-112 11905505-2 2002 Although the active molecules responsible for the diverse therapeutic activities of the drug and the sequence of reactions triggered inside the cells remain unclear, it was demonstrated that thalidomide (THAL) inhibits TNFalpha mRNA expression and protein production by stimulated monocytes and activated T lymphocytes. Thalidomide 191-202 tumor necrosis factor Homo sapiens 219-227 11905505-2 2002 Although the active molecules responsible for the diverse therapeutic activities of the drug and the sequence of reactions triggered inside the cells remain unclear, it was demonstrated that thalidomide (THAL) inhibits TNFalpha mRNA expression and protein production by stimulated monocytes and activated T lymphocytes. Thalidomide 204-208 tumor necrosis factor Homo sapiens 219-227 11905505-4 2002 It has been reported that thalidomide as well as some its analogues decrease M. leprae-induced TNFalpha and IL-12 mRNA in vitro. Thalidomide 26-37 tumor necrosis factor Homo sapiens 95-103 11905505-6 2002 The present data further support thalidomide"s effects on TNFa synthesis and the growing need to search for new specific TNFalpha inhibitors (non-teratogenic compounds) that might be potentially used in clinical disorders such as leprosy. Thalidomide 33-44 tumor necrosis factor Homo sapiens 58-62 11788559-11 2002 Production of TNF-alpha and IL-12 decreased during treatment with thalidomide: LPMC (TNF-alpha: 42.3 (8.3) pg/ml v 16.4 (6.3); IL-12: 9.7 (3.3) v 5.0 (2.5); p<0.04) and PBMC (TNF-alpha: 62.8 (14.6) v 22.5 (9.2); p<0.02). Thalidomide 66-77 tumor necrosis factor Homo sapiens 14-23 11788559-11 2002 Production of TNF-alpha and IL-12 decreased during treatment with thalidomide: LPMC (TNF-alpha: 42.3 (8.3) pg/ml v 16.4 (6.3); IL-12: 9.7 (3.3) v 5.0 (2.5); p<0.04) and PBMC (TNF-alpha: 62.8 (14.6) v 22.5 (9.2); p<0.02). Thalidomide 66-77 tumor necrosis factor Homo sapiens 85-94 11788559-11 2002 Production of TNF-alpha and IL-12 decreased during treatment with thalidomide: LPMC (TNF-alpha: 42.3 (8.3) pg/ml v 16.4 (6.3); IL-12: 9.7 (3.3) v 5.0 (2.5); p<0.04) and PBMC (TNF-alpha: 62.8 (14.6) v 22.5 (9.2); p<0.02). Thalidomide 66-77 tumor necrosis factor Homo sapiens 85-94 11788559-13 2002 Culturing of LPMC with thalidomide showed a dose dependent decrease in TNF-alpha and IL-12 production. Thalidomide 23-34 tumor necrosis factor Homo sapiens 71-80 11788559-14 2002 CONCLUSION: The clinical effects of thalidomide in Crohn"s disease may be mediated by reduction of both TNF-alpha and IL-12. Thalidomide 36-47 tumor necrosis factor Homo sapiens 104-113 12513842-2 2002 The level of interleukin-6 (IL-6) autosecreted by myeloma cells was tested by IL-6-dependent cell line when myeloma cells were treated with thalidomide at 200 microgram/ml, and in the same concentration of thalidomide the expression of IL-6 receptor were tested by flow cytometry. Thalidomide 140-151 interleukin 6 Homo sapiens 13-26 12513842-2 2002 The level of interleukin-6 (IL-6) autosecreted by myeloma cells was tested by IL-6-dependent cell line when myeloma cells were treated with thalidomide at 200 microgram/ml, and in the same concentration of thalidomide the expression of IL-6 receptor were tested by flow cytometry. Thalidomide 140-151 interleukin 6 Homo sapiens 28-32 12513842-8 2002 Inhibiting tumor cells secreting level of IL-6 and reducing the expression of IL-6 receptor on myeloma cell surface is one of the mechanisms for thalidomide to remedy multiple myeloma patients Thalidomide 145-156 interleukin 6 Homo sapiens 42-46 12513842-8 2002 Inhibiting tumor cells secreting level of IL-6 and reducing the expression of IL-6 receptor on myeloma cell surface is one of the mechanisms for thalidomide to remedy multiple myeloma patients Thalidomide 145-156 interleukin 6 Homo sapiens 78-82 12513842-2 2002 The level of interleukin-6 (IL-6) autosecreted by myeloma cells was tested by IL-6-dependent cell line when myeloma cells were treated with thalidomide at 200 microgram/ml, and in the same concentration of thalidomide the expression of IL-6 receptor were tested by flow cytometry. Thalidomide 206-217 interleukin 6 Homo sapiens 13-26 12513842-2 2002 The level of interleukin-6 (IL-6) autosecreted by myeloma cells was tested by IL-6-dependent cell line when myeloma cells were treated with thalidomide at 200 microgram/ml, and in the same concentration of thalidomide the expression of IL-6 receptor were tested by flow cytometry. Thalidomide 206-217 interleukin 6 Homo sapiens 28-32 12513842-5 2002 After treatment with thalidomide at 200 microgram/ml, the concentrations of IL-6 secreted by myeloma cells were (148.5 +/- 96.7) microgram/ml, and the levels of IL-6 receptor expressed on the cell surface were 16.7% and 20.2% in untreated and relapsed or refractory patients, respectively, and those were significantly lower than those levels in the cells before exposure to thalidomide. Thalidomide 21-32 interleukin 6 Homo sapiens 76-80 12513842-5 2002 After treatment with thalidomide at 200 microgram/ml, the concentrations of IL-6 secreted by myeloma cells were (148.5 +/- 96.7) microgram/ml, and the levels of IL-6 receptor expressed on the cell surface were 16.7% and 20.2% in untreated and relapsed or refractory patients, respectively, and those were significantly lower than those levels in the cells before exposure to thalidomide. Thalidomide 21-32 interleukin 6 Homo sapiens 161-165 12513842-5 2002 After treatment with thalidomide at 200 microgram/ml, the concentrations of IL-6 secreted by myeloma cells were (148.5 +/- 96.7) microgram/ml, and the levels of IL-6 receptor expressed on the cell surface were 16.7% and 20.2% in untreated and relapsed or refractory patients, respectively, and those were significantly lower than those levels in the cells before exposure to thalidomide. Thalidomide 375-386 interleukin 6 Homo sapiens 161-165 11809002-3 2001 Recent reports of original pharmacological properties including modulation of cytokine production (mainly reduced TNF-alpha production) and inhibition of angiogenesis have led to the suggestion that thalidomide may be useful in some inflammatory and neoplastic conditions. Thalidomide 199-210 tumor necrosis factor Homo sapiens 114-123 11734114-3 2002 Current evidence indicates that thalidomide reduces the activity of the inflammatory cytokine tumor necrosis factor-alpha by accelerating the degradation of its messenger RNA. Thalidomide 32-43 tumor necrosis factor Homo sapiens 94-121 11705847-3 2001 The induction of Cox-2 protein and mRNA by LPS was also suppressed by thalidomide. Thalidomide 70-81 prostaglandin-endoperoxide synthase 2 Mus musculus 17-22 11753617-13 2001 Finally, thalidomide (100 microM) and its immunomodulatory analog IMiD1-CC4047 (1 microM) decreased the upregulation of IL-6 and VEGF secretion in cultures of BMSCs, MM cells and co-cultures of BMSCs with MM cells. Thalidomide 9-20 interleukin 6 Homo sapiens 120-124 11753617-13 2001 Finally, thalidomide (100 microM) and its immunomodulatory analog IMiD1-CC4047 (1 microM) decreased the upregulation of IL-6 and VEGF secretion in cultures of BMSCs, MM cells and co-cultures of BMSCs with MM cells. Thalidomide 9-20 vascular endothelial growth factor A Homo sapiens 129-133 11838958-0 2001 Down-regulation of cell adhesion molecules LFA-1 and ICAM-1 after in vitro treatment with the anti-TNF-alpha agent thalidomide. Thalidomide 115-126 integrin subunit alpha L Homo sapiens 43-48 11838958-0 2001 Down-regulation of cell adhesion molecules LFA-1 and ICAM-1 after in vitro treatment with the anti-TNF-alpha agent thalidomide. Thalidomide 115-126 intercellular adhesion molecule 1 Homo sapiens 53-59 11838958-0 2001 Down-regulation of cell adhesion molecules LFA-1 and ICAM-1 after in vitro treatment with the anti-TNF-alpha agent thalidomide. Thalidomide 115-126 tumor necrosis factor Homo sapiens 99-108 11838958-1 2001 The tumor necrosis factor-alpha (TNF-alpha) inhibitor thalidomide is known to be a potent modulator of host immunity, a potential treatment for autoimmune disorders such as rheumatoid arthritis (RA) and a treatment for complications of HIV-1 infection. Thalidomide 54-65 tumor necrosis factor Homo sapiens 33-42 11730158-2 2001 The effects of thalidomide are at least partly mediated by down-regulation of tumour necrosis factor (TNF)-alpha, a potent proinflammatory cytokine. Thalidomide 15-26 tumor necrosis factor Homo sapiens 78-112 11705847-0 2001 Thalidomide and its analogues inhibit lipopolysaccharide-mediated Iinduction of cyclooxygenase-2. Thalidomide 0-11 prostaglandin-endoperoxide synthase 2 Mus musculus 80-96 11705847-1 2001 We investigated the effect of thalidomide, a compound with immunomodulatory and antiangiogenic properties, on lipopolysaccharide (LPS)-mediated induction of cyclooxygenase-2 (Cox-2) and prostaglandin (PG) biosynthesis in murine macrophages. Thalidomide 30-41 prostaglandin-endoperoxide synthase 2 Mus musculus 157-173 11705847-1 2001 We investigated the effect of thalidomide, a compound with immunomodulatory and antiangiogenic properties, on lipopolysaccharide (LPS)-mediated induction of cyclooxygenase-2 (Cox-2) and prostaglandin (PG) biosynthesis in murine macrophages. Thalidomide 30-41 prostaglandin-endoperoxide synthase 2 Mus musculus 175-180 11705847-5 2001 Thalidomide decreased the stability of Cox-2 mRNA. Thalidomide 0-11 prostaglandin-endoperoxide synthase 2 Mus musculus 39-44 11705847-6 2001 A series of structural analogues of thalidomide also inhibited LPS-mediated induction of Cox-2 and PGE(2) synthesis. Thalidomide 36-47 prostaglandin-endoperoxide synthase 2 Mus musculus 89-94 11504824-8 2001 Thalidomide resulted in a nonsignificant decrease of CRP and SAA, but the concentrations of other inflammatory mediators, including urine neopterin, remained unchanged. Thalidomide 0-11 C-reactive protein Homo sapiens 53-56 11598844-4 2001 Thalidomide stimulated CD8+ T cells in peripheral blood, which increased expression of the activation markers CD38 and human leukocyte antigen DR and of the memory cell marker CD45RO. Thalidomide 0-11 CD8a molecule Homo sapiens 23-26 11598844-4 2001 Thalidomide stimulated CD8+ T cells in peripheral blood, which increased expression of the activation markers CD38 and human leukocyte antigen DR and of the memory cell marker CD45RO. Thalidomide 0-11 CD38 molecule Homo sapiens 110-114 11598844-5 2001 The frequency of HIV gag-specific CD8+ T cells in peripheral blood increased in 3 of 4 children who were evaluated during treatment with thalidomide. Thalidomide 137-148 CD8a molecule Homo sapiens 34-37 11592764-1 2001 Thalidomide is reported to suppress levels of several cytokines, angiogenic and growth factors including TNF-alpha, basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6). Thalidomide 0-11 tumor necrosis factor Homo sapiens 105-114 11592764-1 2001 Thalidomide is reported to suppress levels of several cytokines, angiogenic and growth factors including TNF-alpha, basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6). Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 116-146 11592764-1 2001 Thalidomide is reported to suppress levels of several cytokines, angiogenic and growth factors including TNF-alpha, basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6). Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 148-152 11592764-1 2001 Thalidomide is reported to suppress levels of several cytokines, angiogenic and growth factors including TNF-alpha, basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6). Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 155-189 11592764-1 2001 Thalidomide is reported to suppress levels of several cytokines, angiogenic and growth factors including TNF-alpha, basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6). Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 191-195 11592764-1 2001 Thalidomide is reported to suppress levels of several cytokines, angiogenic and growth factors including TNF-alpha, basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6). Thalidomide 0-11 interleukin 6 Homo sapiens 201-214 11592764-1 2001 Thalidomide is reported to suppress levels of several cytokines, angiogenic and growth factors including TNF-alpha, basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6). Thalidomide 0-11 interleukin 6 Homo sapiens 216-220 11555579-0 2001 High plasma basic fibroblast growth factor concentration is associated with response to thalidomide in progressive multiple myeloma. Thalidomide 88-99 fibroblast growth factor 2 Homo sapiens 12-42 11722460-5 2001 One possible predisposing factor in AP may be tumour necrosis factor (TNF)alpha as suggested by the good response of AP to the TNFalpha inhibitor thalidomide, and by the involvement of this cytokine in many immune responses. Thalidomide 146-157 tumor necrosis factor Homo sapiens 70-79 11722460-5 2001 One possible predisposing factor in AP may be tumour necrosis factor (TNF)alpha as suggested by the good response of AP to the TNFalpha inhibitor thalidomide, and by the involvement of this cytokine in many immune responses. Thalidomide 146-157 tumor necrosis factor Homo sapiens 127-135 11574978-2 2001 Thalidomide, a non-barbiturate hypnotic, also has an anti-inflammatory effect, presumably by suppressing the production of tumor necrosis factor alpha. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 123-150 11574978-9 2001 RESULTS: Colonic wet weight, lesion area, myeloperoxidase activity and serum levels of TNF alpha were significantly lower (P < 0.05) in the treatment group (iodoacetamide + thalidomide) than the control group (iodoacetamide only). Thalidomide 176-187 tumor necrosis factor Rattus norvegicus 87-96 11720008-0 2001 Potent inhibition of cytokine production from intestinal lamina propria T cells by phosphodiesterase-4 inhibitory thalidomide analogues. Thalidomide 114-125 phosphodiesterase 4A Homo sapiens 83-102 11720008-2 2001 Thalidomide influences cytokine production by leukocytes, inhibiting macrophage TNF-alpha, and is beneficial in treating Crohn"s disease. Thalidomide 0-11 tumor necrosis factor Homo sapiens 80-89 11504824-8 2001 Thalidomide resulted in a nonsignificant decrease of CRP and SAA, but the concentrations of other inflammatory mediators, including urine neopterin, remained unchanged. Thalidomide 0-11 serum amyloid A1 cluster Homo sapiens 61-64 11685731-4 2001 In a recent phase II trial of thalidomide in AIPC, 28% of patients achieved a prostate-specific antigen (PSA) decrease of >40%. Thalidomide 30-41 PDZ domain containing 2 Homo sapiens 45-49 11498529-11 2001 According to RT - PCR analysis, both thalidomide and supidimide also significantly reduced vascular cell adhesion molecule-1 mRNA expression in the affected part of the descending colon. Thalidomide 37-48 vascular cell adhesion molecule 1 Rattus norvegicus 91-124 11448901-13 2001 Because our preclinical studies had shown that thalidomide increases PSA secretion, we believe that the magnitude of PSA decline seen in our trial justifies further study. Thalidomide 47-58 kallikrein related peptidase 3 Homo sapiens 69-72 11418482-4 2001 It was demonstrated that Thal/IMiDs do not induce T-cell proliferation alone but act as costimulators to trigger proliferation of anti-CD3-stimulated T cells from patients with MM, accompanied by an increase in interferon-gamma and IL-2 secretion. Thalidomide 25-29 interferon gamma Homo sapiens 211-227 11418482-4 2001 It was demonstrated that Thal/IMiDs do not induce T-cell proliferation alone but act as costimulators to trigger proliferation of anti-CD3-stimulated T cells from patients with MM, accompanied by an increase in interferon-gamma and IL-2 secretion. Thalidomide 25-29 interleukin 2 Homo sapiens 232-236 11418482-6 2001 A role for natural killer (NK)- and LAK-cell-mediated killing is suggested because IL-2-primed peripheral blood mononuclear cells (PBMCs) treated with Thal/IMiDs demonstrated significantly increased lysis of MM cell lines. Thalidomide 151-155 alpha kinase 1 Homo sapiens 36-39 11418482-6 2001 A role for natural killer (NK)- and LAK-cell-mediated killing is suggested because IL-2-primed peripheral blood mononuclear cells (PBMCs) treated with Thal/IMiDs demonstrated significantly increased lysis of MM cell lines. Thalidomide 151-155 interleukin 2 Homo sapiens 83-87 11448901-13 2001 Because our preclinical studies had shown that thalidomide increases PSA secretion, we believe that the magnitude of PSA decline seen in our trial justifies further study. Thalidomide 47-58 kallikrein related peptidase 3 Homo sapiens 117-120 11302825-1 2001 Ingestion of thalidomide was associated with a reduction in the upregulation of the granulocyte activation marker CD11b and a reduced capacity to release elastase and lactoferrin after stimulation with lipopolysaccharide or lipoteichoic acid. Thalidomide 13-24 integrin subunit alpha M Homo sapiens 114-119 11411542-1 2001 Previous studies of alpha-glucosidase inhibitors derived from thalidomide revealed that 4,5,6,7-tetrachloro-N-alkylphthalimide derivatives are superior lead compounds. Thalidomide 62-73 sucrase-isomaltase Homo sapiens 20-37 12113124-4 2001 Thalidomide has antiangiogenic and immunomodulatory properties and is an effective inhibitor of TNF-alpha. Thalidomide 0-11 tumor necrosis factor Homo sapiens 96-105 11297551-0 2001 Inhibition of NF-kappa B activity by thalidomide through suppression of IkappaB kinase activity. Thalidomide 37-48 nuclear factor kappa B subunit 1 Homo sapiens 14-24 11297551-6 2001 We show here that thalidomide can block NF-kappaB activation through a mechanism that involves the inhibition of activity of the IkappaB kinase. Thalidomide 18-29 nuclear factor kappa B subunit 1 Homo sapiens 40-49 11297551-7 2001 Consistent with the observed inhibition of NF-kappaB, thalidomide blocked the cytokine-induced expression of NF-kappaB-regulated genes such as those encoding interleukin-8, TRAF1, and c-IAP2. Thalidomide 54-65 nuclear factor kappa B subunit 1 Homo sapiens 43-52 11297551-7 2001 Consistent with the observed inhibition of NF-kappaB, thalidomide blocked the cytokine-induced expression of NF-kappaB-regulated genes such as those encoding interleukin-8, TRAF1, and c-IAP2. Thalidomide 54-65 nuclear factor kappa B subunit 1 Homo sapiens 109-118 11297551-7 2001 Consistent with the observed inhibition of NF-kappaB, thalidomide blocked the cytokine-induced expression of NF-kappaB-regulated genes such as those encoding interleukin-8, TRAF1, and c-IAP2. Thalidomide 54-65 C-X-C motif chemokine ligand 8 Homo sapiens 158-171 11297551-7 2001 Consistent with the observed inhibition of NF-kappaB, thalidomide blocked the cytokine-induced expression of NF-kappaB-regulated genes such as those encoding interleukin-8, TRAF1, and c-IAP2. Thalidomide 54-65 TNF receptor associated factor 1 Homo sapiens 173-178 11297551-7 2001 Consistent with the observed inhibition of NF-kappaB, thalidomide blocked the cytokine-induced expression of NF-kappaB-regulated genes such as those encoding interleukin-8, TRAF1, and c-IAP2. Thalidomide 54-65 baculoviral IAP repeat containing 3 Homo sapiens 184-190 11297551-8 2001 These data indicate that the therapeutic potential for thalidomide may be based on its ability to block NF-kappaB activation through suppression of IkappaB kinase activity. Thalidomide 55-66 nuclear factor kappa B subunit 1 Homo sapiens 104-113 11321389-3 2001 Thalidomide selectively reduces TNF-alpha production by inflammatory cells. Thalidomide 0-11 tumor necrosis factor Homo sapiens 32-41 11165754-9 2001 Thalidomide also lowered remarkably VEGF expression (P<0.01) and CD31 (P<0.01) as well as VEGF mRNA (P<0.05), but it did not show statistically significant inhibitory effect on the tumor growth. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 36-40 11165754-9 2001 Thalidomide also lowered remarkably VEGF expression (P<0.01) and CD31 (P<0.01) as well as VEGF mRNA (P<0.05), but it did not show statistically significant inhibitory effect on the tumor growth. Thalidomide 0-11 platelet and endothelial cell adhesion molecule 1 Homo sapiens 68-72 11165754-9 2001 Thalidomide also lowered remarkably VEGF expression (P<0.01) and CD31 (P<0.01) as well as VEGF mRNA (P<0.05), but it did not show statistically significant inhibitory effect on the tumor growth. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 96-100 11417421-5 2001 Thalidomide, a potent inhibitor of tumour necrosis factor alpha (TNF-alpha), was added to the treatment regimen and resulted in marked clinical improvement with resolution of the abscess within 4 months. Thalidomide 0-11 tumor necrosis factor Homo sapiens 65-74 11354662-0 2001 Aza analogues of thalidomide: synthesis and evaluation as inhibitors of tumor necrosis factor-alpha production in vitro. Thalidomide 17-28 tumor necrosis factor Homo sapiens 72-99 11354662-2 2001 These 5-phthalimidopyrimidines as well as phthalimido-2,4-difluorobenzenes were designed as analogues of thalidomide, a well known inhibitor of TNF-alpha production. Thalidomide 105-116 tumor necrosis factor Homo sapiens 144-153 11604049-4 2001 Effects on tumour necrosis factor-alpha (TNFalpha) release play an important role in the ability of thalidomide to affect the immune system. Thalidomide 100-111 tumor necrosis factor Homo sapiens 41-49 11604049-5 2001 Alteration of synthesis and release of cytokines such as interleukin (IL)-1, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12 and interferon-gamma is involved in the complex mechanisms of thalidomide. Thalidomide 176-187 interferon gamma Homo sapiens 118-134 11604049-11 2001 However, despite the promising findings of thalidomide at the molecular level, namely its anti-TNFalpha properties and its intercalation with DNA, and activity in clinical trials, there is still a great need for more intensive research. Thalidomide 43-54 tumor necrosis factor Homo sapiens 95-103 11116278-9 2001 Finally, the TNF-alpha targeting drugs, namely thalidomide and the anti-TNF-alpha mAb, infliximab, have given promising results in the treatment of severe and/or refractory AS patients, however further controlled studies are required. Thalidomide 47-58 tumor necrosis factor Homo sapiens 13-22 11204273-6 2001 Treatment of explants with the angiogenesis inhibitor endostatin, the chemokine IP-10, and the thalidomide derivative phatolyl glutamic acid (PG-acid) resulted in an inhibition of the formation of PECAM-1 positive tubelike structures of 48.8% (+/- 4%), 50.2% (+/- 12%), and 80.8% (+/- 3%), respectively. Thalidomide 95-106 platelet/endothelial cell adhesion molecule 1 Mus musculus 197-204 11367517-0 2001 Differential effect of thalidomide and dexamethasone on the transcription factor NF-kappa B. Thalidomide 23-34 nuclear factor kappa B subunit 1 Homo sapiens 81-91 11367517-2 2001 In vitro studies have shown that thalidomide inhibits tumour necrosis factor alpha (TNF-alpha) mRNA expression and protein production by mitogen-stimulated macrophages and activated T cells. Thalidomide 33-44 tumor necrosis factor Homo sapiens 84-93 11367517-4 2001 In the present investigations, we have examined the influence of thalidomide on nuclear levels of NF-kappa B in human peripheral blood mononuclear cells (PBMC) following activation with mitogen or phorbol myristate acetate (PMA)/ionophore. Thalidomide 65-76 nuclear factor kappa B subunit 1 Homo sapiens 98-108 11367517-13 2001 These observations suggest that thalidomide does not act directly on NF-kappa B and therefore inhibits TNF-alpha production through another independent mechanism. Thalidomide 32-43 tumor necrosis factor Homo sapiens 103-112 11068114-0 2000 Thalidomide treatment in chronic constrictive neuropathy decreases endoneurial tumor necrosis factor-alpha, increases interleukin-10 and has long-term effects on spinal cord dorsal horn met-enkephalin. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 79-106 11123102-0 2000 The Th1/Th2 paradigm in the pathogenesis of scleroderma, and its modulation by thalidomide. Thalidomide 79-90 negative elongation factor complex member C/D Homo sapiens 4-7 11123102-5 2000 The immune modulating effects of the drug thalidomide on Th1 and Th2 immunity are also described, along with the drug"s potential application to disease processes like scleroderma. Thalidomide 42-53 negative elongation factor complex member C/D Homo sapiens 57-60 11121122-7 2000 An inhibitory effect on interleukin-10 mRNA was likewise seen after thalidomide and pentoxifylline, but not subsequent to prednisone treatment. Thalidomide 68-79 interleukin 10 Homo sapiens 24-38 11204669-4 2000 Clinical experience with thalidomide (Thalomid), which inhibits angiogenesis induced by both vascular endothelial growth factor and basic fibroblast growth factor, has included observation of "clinical improvement" in patients with androgen-independent prostate cancer and anecdotal responses in patients with metastatic disease refractory to chemotherapy. Thalidomide 25-36 vascular endothelial growth factor A Homo sapiens 93-127 11204669-4 2000 Clinical experience with thalidomide (Thalomid), which inhibits angiogenesis induced by both vascular endothelial growth factor and basic fibroblast growth factor, has included observation of "clinical improvement" in patients with androgen-independent prostate cancer and anecdotal responses in patients with metastatic disease refractory to chemotherapy. Thalidomide 38-46 vascular endothelial growth factor A Homo sapiens 93-127 11079045-5 2000 Thalidomide, an oral agent with anti-TNF effects, shows promise in non-controlled experience. Thalidomide 0-11 tumor necrosis factor Homo sapiens 37-40 11068114-0 2000 Thalidomide treatment in chronic constrictive neuropathy decreases endoneurial tumor necrosis factor-alpha, increases interleukin-10 and has long-term effects on spinal cord dorsal horn met-enkephalin. Thalidomide 0-11 interleukin 10 Rattus norvegicus 118-132 11068114-2 2000 Since thalidomide mainly inhibits tumor necrosis factor alpha (TNF-alpha) synthesis with less well defined effects on other cytokines, we investigated the effect of the drug on the expression of the proinflammatory cytokines TNF-alpha, interleukin-1beta (IL-1beta) and interleukin 6 (IL-6), and of the anti-inflammatory cytokine interleukin-10 (IL-10) in the lesioned rat sciatic nerve. Thalidomide 6-17 tumor necrosis factor Rattus norvegicus 34-61 11068114-2 2000 Since thalidomide mainly inhibits tumor necrosis factor alpha (TNF-alpha) synthesis with less well defined effects on other cytokines, we investigated the effect of the drug on the expression of the proinflammatory cytokines TNF-alpha, interleukin-1beta (IL-1beta) and interleukin 6 (IL-6), and of the anti-inflammatory cytokine interleukin-10 (IL-10) in the lesioned rat sciatic nerve. Thalidomide 6-17 tumor necrosis factor Rattus norvegicus 63-72 11068114-3 2000 The increase of endoneurial TNF-alpha during the first week after CCI was reduced after thalidomide treatment, as shown with immunohistochemistry and enzyme-linked-immunosorbent assay. Thalidomide 88-99 tumor necrosis factor Rattus norvegicus 28-37 11068114-5 2000 Recruitment of epineurial IL-10 immunoreactive macrophages as well as the recovery of injury-induced depletion of endoneurial IL-10-IR was enhanced by thalidomide treatment. Thalidomide 151-162 interleukin 10 Rattus norvegicus 26-31 11068114-5 2000 Recruitment of epineurial IL-10 immunoreactive macrophages as well as the recovery of injury-induced depletion of endoneurial IL-10-IR was enhanced by thalidomide treatment. Thalidomide 151-162 interleukin 10 Rattus norvegicus 126-131 11027451-5 2000 Histologic comparison of skin biopsies showed changes in skin fibrosis and an increase in epidermal and dermal infiltrating CD8(+) T cells with thalidomide treatment. Thalidomide 144-155 CD8a molecule Homo sapiens 124-127 11027451-8 2000 When SSc PBMCs activated by anti-CD3 mAb were exposed to thalidomide, increases in both production of IL-2, IL-3, GM-CSF, and IFN-gamma and T cell expression of CD40L were observed. Thalidomide 57-68 interferon gamma Homo sapiens 126-135 11027451-6 2000 In thalidomide-treated SSc patients, plasma levels of IL-12 and TNF-alpha increased, while plasma IL-5 and IL-10 levels remained unchanged. Thalidomide 3-14 tumor necrosis factor Homo sapiens 64-73 11027451-8 2000 When SSc PBMCs activated by anti-CD3 mAb were exposed to thalidomide, increases in both production of IL-2, IL-3, GM-CSF, and IFN-gamma and T cell expression of CD40L were observed. Thalidomide 57-68 CD40 ligand Homo sapiens 161-166 11027451-6 2000 In thalidomide-treated SSc patients, plasma levels of IL-12 and TNF-alpha increased, while plasma IL-5 and IL-10 levels remained unchanged. Thalidomide 3-14 interleukin 5 Homo sapiens 98-102 11027451-6 2000 In thalidomide-treated SSc patients, plasma levels of IL-12 and TNF-alpha increased, while plasma IL-5 and IL-10 levels remained unchanged. Thalidomide 3-14 interleukin 10 Homo sapiens 107-112 11027451-8 2000 When SSc PBMCs activated by anti-CD3 mAb were exposed to thalidomide, increases in both production of IL-2, IL-3, GM-CSF, and IFN-gamma and T cell expression of CD40L were observed. Thalidomide 57-68 interleukin 2 Homo sapiens 102-106 11027451-8 2000 When SSc PBMCs activated by anti-CD3 mAb were exposed to thalidomide, increases in both production of IL-2, IL-3, GM-CSF, and IFN-gamma and T cell expression of CD40L were observed. Thalidomide 57-68 interleukin 3 Homo sapiens 108-112 11027451-8 2000 When SSc PBMCs activated by anti-CD3 mAb were exposed to thalidomide, increases in both production of IL-2, IL-3, GM-CSF, and IFN-gamma and T cell expression of CD40L were observed. Thalidomide 57-68 colony stimulating factor 2 Homo sapiens 114-120 10952569-5 2000 In all six volunteers ingestion of thalidomide was associated with enhanced SEB- and anti-CD3/CD28-induced production of the Th1 cytokine gamma interferon (P < 0.05) and a decrease in the level of anti-CD3/CD28-induced interleukin-5 (IL-5) production (P < 0.05). Thalidomide 35-46 interleukin 5 Homo sapiens 222-235 10952569-5 2000 In all six volunteers ingestion of thalidomide was associated with enhanced SEB- and anti-CD3/CD28-induced production of the Th1 cytokine gamma interferon (P < 0.05) and a decrease in the level of anti-CD3/CD28-induced interleukin-5 (IL-5) production (P < 0.05). Thalidomide 35-46 interleukin 5 Homo sapiens 237-241 10952569-3 2000 To obtain insight into the effect of thalidomide on the capacity of lymphocytes to produce Th1 and Th2 cytokines, six healthy volunteers received an oral dose (400 mg) of thalidomide. Thalidomide 37-48 negative elongation factor complex member C/D Homo sapiens 91-94 10952569-8 2000 Thus, a single oral dose of thalidomide causes a Th1-type response in healthy humans. Thalidomide 28-39 negative elongation factor complex member C/D Homo sapiens 49-52 10952569-5 2000 In all six volunteers ingestion of thalidomide was associated with enhanced SEB- and anti-CD3/CD28-induced production of the Th1 cytokine gamma interferon (P < 0.05) and a decrease in the level of anti-CD3/CD28-induced interleukin-5 (IL-5) production (P < 0.05). Thalidomide 35-46 CD28 molecule Homo sapiens 94-98 10952569-5 2000 In all six volunteers ingestion of thalidomide was associated with enhanced SEB- and anti-CD3/CD28-induced production of the Th1 cytokine gamma interferon (P < 0.05) and a decrease in the level of anti-CD3/CD28-induced interleukin-5 (IL-5) production (P < 0.05). Thalidomide 35-46 negative elongation factor complex member C/D Homo sapiens 125-128 10961786-7 2000 Levels of tumor necrosis factor-alpha in the cerebrospinal fluid decreased markedly during thalidomide therapy. Thalidomide 91-102 tumor necrosis factor Homo sapiens 10-37 10952569-5 2000 In all six volunteers ingestion of thalidomide was associated with enhanced SEB- and anti-CD3/CD28-induced production of the Th1 cytokine gamma interferon (P < 0.05) and a decrease in the level of anti-CD3/CD28-induced interleukin-5 (IL-5) production (P < 0.05). Thalidomide 35-46 CD28 molecule Homo sapiens 209-213 10937517-3 2000 RESULTS: Thalidomide reduced insulin-stimulated glucose uptake by 31% (from 27.7 to 19.2 pmol x kg(-1) x min(-1), P < 0.05) compared with the prestudy and by 21% (from 24.2 to 19.2 pmol x kg(-1) x min(-1), P < 0.05) compared with placebo. Thalidomide 9-20 insulin Homo sapiens 29-36 10937517-4 2000 Thalidomide also reduced insulin-stimulated GS by 48% (from 14.1 to 8.2 micromol x kg(-1) x min(-1), P < 0.05) compared with the prestudy and by 40% (from 13.6 to 8.2 micromol x kg(-1) x min(-1), P < 0.5) compared with placebo. Thalidomide 0-11 insulin Homo sapiens 25-32 10937517-6 2000 CONCLUSIONS: We conclude that thalidomide increased insulin resistance in obese patients with type 2 diabetes by inhibiting insulin-stimulated GS and that patients taking thalidomide should be monitored for possible deterioration in their glucose tolerance. Thalidomide 30-41 insulin Homo sapiens 52-59 10937517-6 2000 CONCLUSIONS: We conclude that thalidomide increased insulin resistance in obese patients with type 2 diabetes by inhibiting insulin-stimulated GS and that patients taking thalidomide should be monitored for possible deterioration in their glucose tolerance. Thalidomide 30-41 insulin Homo sapiens 124-131 10971513-9 2000 The results suggest a role for thalidomide analogue CC-3052 in reducing persistent activation of the TNF-alpha system in HIV without markedly impairing neutrophil viability. Thalidomide 31-42 tumor necrosis factor Homo sapiens 101-110 10950803-4 2000 Thalidomide treatment was found to cause potent and selective inhibition of IL-8 production in a dose-responsive manner. Thalidomide 0-11 C-X-C motif chemokine ligand 8 Homo sapiens 76-80 10950803-6 2000 In addition, thalidomide treatment of lipopolysaccharide-stimulated microglia inhibited the activation of protein NF-kappaB, a transcription factor known to be important for IL-8 production. Thalidomide 13-24 nuclear factor kappa B subunit 1 Homo sapiens 114-123 10950803-6 2000 In addition, thalidomide treatment of lipopolysaccharide-stimulated microglia inhibited the activation of protein NF-kappaB, a transcription factor known to be important for IL-8 production. Thalidomide 13-24 C-X-C motif chemokine ligand 8 Homo sapiens 174-178 10950803-7 2000 These results suggest thalidomide could have a therapeutic role in acute bacterial meningitis through inhibition of IL-8-mediated neutrophil chemotaxis. Thalidomide 22-33 C-X-C motif chemokine ligand 8 Homo sapiens 116-120 10850856-0 2000 Modulation of hyperoxia-induced TNF-alpha expression in the newborn rat lung by thalidomide and dexamethasone. Thalidomide 80-91 tumor necrosis factor Rattus norvegicus 32-41 10850856-7 2000 In the lungs of animals treated with either dexamethasone or thalidomide, TNF-alpha mRNA levels were reduced, while only dexamethasone treatment also reduced IL-6 mRNA levels. Thalidomide 61-72 tumor necrosis factor Rattus norvegicus 74-83 10850856-7 2000 In the lungs of animals treated with either dexamethasone or thalidomide, TNF-alpha mRNA levels were reduced, while only dexamethasone treatment also reduced IL-6 mRNA levels. Thalidomide 61-72 interleukin 6 Rattus norvegicus 158-162 10915104-0 2000 Tumor necrosis factor-alpha increased production during thalidomide treatment in patients with tuberculosis and human immunodeficiency virus coinfection. Thalidomide 56-67 tumor necrosis factor Homo sapiens 0-27 10799645-2 2000 We propose that thalidomide affects the following pathway during development: insulin-like growth factor I (IGF-I) and fibroblast growth factor 2 (FGF-2) stimulation of the transcription of alphav and beta3 integrin subunit genes. Thalidomide 16-27 insulin like growth factor 1 Homo sapiens 78-106 10799645-2 2000 We propose that thalidomide affects the following pathway during development: insulin-like growth factor I (IGF-I) and fibroblast growth factor 2 (FGF-2) stimulation of the transcription of alphav and beta3 integrin subunit genes. Thalidomide 16-27 insulin like growth factor 1 Homo sapiens 108-113 10799645-2 2000 We propose that thalidomide affects the following pathway during development: insulin-like growth factor I (IGF-I) and fibroblast growth factor 2 (FGF-2) stimulation of the transcription of alphav and beta3 integrin subunit genes. Thalidomide 16-27 fibroblast growth factor 2 Homo sapiens 119-145 10799645-2 2000 We propose that thalidomide affects the following pathway during development: insulin-like growth factor I (IGF-I) and fibroblast growth factor 2 (FGF-2) stimulation of the transcription of alphav and beta3 integrin subunit genes. Thalidomide 16-27 fibroblast growth factor 2 Homo sapiens 147-152 10799645-2 2000 We propose that thalidomide affects the following pathway during development: insulin-like growth factor I (IGF-I) and fibroblast growth factor 2 (FGF-2) stimulation of the transcription of alphav and beta3 integrin subunit genes. Thalidomide 16-27 eukaryotic translation elongation factor 1 beta 2 pseudogene 2 Homo sapiens 201-206 10799645-7 2000 Intercalation into G-rich promoter regions of DNA may explain why certain thalidomide analogs are not teratogenic while retaining their anti-tumor necrosis factor-alpha (TNF-alpha) activity, and suggests that we look elsewhere to explain the action of thalidomide on TNF-alpha. Thalidomide 74-85 tumor necrosis factor Homo sapiens 170-179 10816156-2 2000 After receiving thalidomide treatment, serum tumor necrosis factor (TNF) and soluble TNF receptor II levels normalized, his constitutional and gastrointestinal symptoms diminished, and the mass lesion shrunk. Thalidomide 16-27 tumor necrosis factor Homo sapiens 45-66 11003055-0 2000 A possible role of N-cadherin in thalidomide teratogenicity. Thalidomide 33-44 cadherin 2 Homo sapiens 19-29 11003055-1 2000 Several experimental findings indicate that the adhesion molecule N-cadherin participates in distinct processes of embryogenesis that spatiotemporarily correlate with high sensitivity to thalidomide. Thalidomide 187-198 cadherin 2 Homo sapiens 66-76 11003055-2 2000 Therefore, we suppose that thalidomide might interfere with N-cadherin-mediated interactions. Thalidomide 27-38 cadherin 2 Homo sapiens 60-70 11003055-3 2000 This hypothesis is supported by protein-ligand docking studies simulating and characterizing the binding of thalidomide to N-cadherin molecules. Thalidomide 108-119 cadherin 2 Homo sapiens 123-133 11003055-4 2000 Thalidomide was found to bind at the N-terminal domain of N-cadherin mimicking a tryptophan residue which is critical for the homodimerization of the adhesion molecule. Thalidomide 0-11 cadherin 2 Homo sapiens 58-68 11003055-5 2000 Based on these results, we suggest that thalidomide might disturb cellular recognition and migration processes in morphogenesis by interaction with N-cadherin. Thalidomide 40-51 cadherin 2 Homo sapiens 148-158 10816156-2 2000 After receiving thalidomide treatment, serum tumor necrosis factor (TNF) and soluble TNF receptor II levels normalized, his constitutional and gastrointestinal symptoms diminished, and the mass lesion shrunk. Thalidomide 16-27 tumor necrosis factor Homo sapiens 68-71 10816156-2 2000 After receiving thalidomide treatment, serum tumor necrosis factor (TNF) and soluble TNF receptor II levels normalized, his constitutional and gastrointestinal symptoms diminished, and the mass lesion shrunk. Thalidomide 16-27 tumor necrosis factor Homo sapiens 85-88 10849134-2 2000 The pharmacologic action involves the downregulation of the pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) and inhibition of basic fibroblast growth factor (bFGF)-induced angiogenesis; however, not much is known about thalidomide"s effect on immunologic parameters in lupus erythematosus (LE). Thalidomide 237-248 fibroblast growth factor 2 Homo sapiens 176-180 11249506-3 2000 Other investigational drugs that also inhibit TNF-alpha activity include new "humanised" anti-TNF-alpha antibodies (CDP571), thalidomide, new analogues of thalidomide, and TNF-alpha receptor fusion proteins. Thalidomide 125-136 tumor necrosis factor Homo sapiens 46-55 11249506-3 2000 Other investigational drugs that also inhibit TNF-alpha activity include new "humanised" anti-TNF-alpha antibodies (CDP571), thalidomide, new analogues of thalidomide, and TNF-alpha receptor fusion proteins. Thalidomide 155-166 tumor necrosis factor Homo sapiens 46-55 10781782-2 2000 Current evidence indicates that thalidomide reduces the activity of the inflammatory cytokine tumor necrosis factor (TNF)-alpha by accelerating the degradation of its messenger RNA. Thalidomide 32-43 tumor necrosis factor Homo sapiens 94-127 10825407-5 2000 The principle therapeutic action of thalidomide appears to be reduction of TNF-alpha levels. Thalidomide 36-47 tumor necrosis factor Homo sapiens 75-84 11558112-8 2000 Once the anti-inflammatory, immuno-modulatory, anti-TNF-alpha and anti-angiogenic properties of thalidomide were discovered, it was also tried in AIDS and related wasting, apthous ulcers, microsporidiosis and Kaposi"s sarcoma. Thalidomide 96-107 tumor necrosis factor Homo sapiens 52-61 10784439-1 2000 Previous studies on the structural development of tumor necrosis factor alpha (TNF-alpha) production regulators derived from thalidomide (N-alpha-phthalimidoglutarimide) revealed that a hydrophobic substituent at the nitrogen atom of the phthalimide ring is critical for potent activity. Thalidomide 125-136 tumor necrosis factor Homo sapiens 50-77 18034532-5 2000 Thalidomide produces partial inhibition of tumour necrosis factor-alpha production in vivo but recent data reveals that it can also act as a co-stimulatory molecule for T cell activation in vitro, resulting in increased production of interleukin-2 and interferon-gamma. Thalidomide 0-11 interleukin 2 Homo sapiens 234-247 18034532-5 2000 Thalidomide produces partial inhibition of tumour necrosis factor-alpha production in vivo but recent data reveals that it can also act as a co-stimulatory molecule for T cell activation in vitro, resulting in increased production of interleukin-2 and interferon-gamma. Thalidomide 0-11 interferon gamma Homo sapiens 252-268 10784439-1 2000 Previous studies on the structural development of tumor necrosis factor alpha (TNF-alpha) production regulators derived from thalidomide (N-alpha-phthalimidoglutarimide) revealed that a hydrophobic substituent at the nitrogen atom of the phthalimide ring is critical for potent activity. Thalidomide 125-136 tumor necrosis factor Homo sapiens 79-88 10720640-10 2000 The demonstration that thalidomide is able to inhibit inflammatory hyperalgesia in rats and the writhing nociceptive response in mice suggests that these analgesic effects seem to be a consequence of the inhibition of TNF-alpha production, and indicates the need for investigations on the possibility of the use of thalidomide for the treatment of pain refractory to classical non-narcotic analgesics. Thalidomide 23-34 tumor necrosis factor Mus musculus 218-227 10720640-2 2000 Thalidomide has been shown to selectively inhibit TNF-alpha production. Thalidomide 0-11 tumor necrosis factor Mus musculus 50-59 10647875-8 2000 The TNF-alpha secreted by the same cells incubated with thalidomide and SEA was not significantly different from that secreted by the cells incubated with SEA alone. Thalidomide 56-67 tumor necrosis factor Homo sapiens 4-13 10720640-5 2000 injections of carrageenin or bradykinin, which act by stimulating TNF-alpha release, but not responses to TNF-alpha or prostaglandin E(2), were inhibited in a dose-dependent manner by pretreatment of the animals with thalidomide. Thalidomide 217-228 tumor necrosis factor Mus musculus 66-75 10720640-8 2000 Moreover, the thalidomide pretreatment also reduced the TNF-alpha mRNA levels in the peritoneal cells induced by injection of zymosan in mice. Thalidomide 14-25 tumor necrosis factor Mus musculus 56-65 10720518-1 2000 Thalidomide, which inhibits monocyte tumor necrosis factor (TNF)-alpha production and costimulates T cells, was tested for immune modulation in patients with human immunodeficiency virus (HIV) infection and tuberculosis (TB) in a placebo-controlled study. Thalidomide 0-11 tumor necrosis factor Homo sapiens 37-70 10720518-2 2000 Thalidomide therapy resulted in increased levels of plasma interleukin (IL)-2 receptor, soluble CD8, interferon-gamma, and IL-12, indicating immune stimulation. Thalidomide 0-11 CD8a molecule Homo sapiens 96-99 10720518-2 2000 Thalidomide therapy resulted in increased levels of plasma interleukin (IL)-2 receptor, soluble CD8, interferon-gamma, and IL-12, indicating immune stimulation. Thalidomide 0-11 interferon gamma Homo sapiens 101-117 10720518-4 2000 Thalidomide treatment increased CD4+ and CD8+ T cell counts and lymphocyte proliferation to purified protein derivative. Thalidomide 0-11 CD8a molecule Homo sapiens 41-44 10720518-7 2000 Thalidomide-induced increased viral replication in CD4+ T cells was abrogated by adding back autologous CD8+ T cells. Thalidomide 0-11 CD8a molecule Homo sapiens 104-107 10661908-1 2000 We propose that thalidomide affects the following pathway during limb development: Growth factors (FGF-2 and IGF-I) attach to receptors on limb bud mesenchymal cells and initiate some second messenger system (perhaps SP-1), which activates alphav and beta3 integrin subunit genes. Thalidomide 16-27 fibroblast growth factor 2 Homo sapiens 99-104 10661908-1 2000 We propose that thalidomide affects the following pathway during limb development: Growth factors (FGF-2 and IGF-I) attach to receptors on limb bud mesenchymal cells and initiate some second messenger system (perhaps SP-1), which activates alphav and beta3 integrin subunit genes. Thalidomide 16-27 insulin like growth factor 1 Homo sapiens 109-114 10647875-0 2000 Thalidomide"s ability to augment the synthesis of IL-2 in vitro in HIV-infected patients is associated with the percentage of CD4+ cells in their blood. Thalidomide 0-11 interleukin 2 Homo sapiens 50-54 10647875-0 2000 Thalidomide"s ability to augment the synthesis of IL-2 in vitro in HIV-infected patients is associated with the percentage of CD4+ cells in their blood. Thalidomide 0-11 CD4 molecule Homo sapiens 126-129 10647875-4 2000 We investigated the effect of thalidomide on the production of IL-2 and TNF-alpha by staphylococcal enterotoxin A (SEA) stimulated peripheral blood mononuclear cells (PBMC) from HIV-infected patients. Thalidomide 30-41 interleukin 2 Homo sapiens 63-67 10647875-4 2000 We investigated the effect of thalidomide on the production of IL-2 and TNF-alpha by staphylococcal enterotoxin A (SEA) stimulated peripheral blood mononuclear cells (PBMC) from HIV-infected patients. Thalidomide 30-41 tumor necrosis factor Homo sapiens 72-81 10647875-7 2000 The PBMC incubated with thalidomide and SEA produced significantly more IL-2 than those incubated with SEA alone. Thalidomide 24-35 interleukin 2 Homo sapiens 72-76 10732751-2 2000 Thalidomide inhibits the processing of mRNA encoding peptide molecules including tumour necrosis factor-alpha (TNF-alpha) and the angiogenic factor vascular endothelial growth factor (VEGF). Thalidomide 0-11 tumor necrosis factor Homo sapiens 111-120 10732751-2 2000 Thalidomide inhibits the processing of mRNA encoding peptide molecules including tumour necrosis factor-alpha (TNF-alpha) and the angiogenic factor vascular endothelial growth factor (VEGF). Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 184-188 10647875-9 2000 The amount of IL-2 produced in the thalidomide and SEA treated cultures was directly correlated with the percentage of CD4+ cells in blood, and inversely correlated with the percentage of CD8+ cells in blood. Thalidomide 35-46 interleukin 2 Homo sapiens 14-18 10647875-9 2000 The amount of IL-2 produced in the thalidomide and SEA treated cultures was directly correlated with the percentage of CD4+ cells in blood, and inversely correlated with the percentage of CD8+ cells in blood. Thalidomide 35-46 CD4 molecule Homo sapiens 119-122 10647875-10 2000 No statistically significant correlations were found when comparing the amount of TNF-alpha produced in the thalidomide and SEA treated cultures with the percentage of CD4+ or CD8+ cells in the blood. Thalidomide 108-119 tumor necrosis factor Homo sapiens 82-91 10647875-11 2000 Thalidomide can act, in vitro, as an additional stimulant to augment the synthesis of IL-2 in HIV-infected patients. Thalidomide 0-11 interleukin 2 Homo sapiens 86-90 10635613-13 1999 Thalidomide, another suppressor of TNF production, significantly increased the death rate when tested in a double-blind placebo controlled trial in patients with early TEN. Thalidomide 0-11 tumor necrosis factor Homo sapiens 35-38 10721827-1 2000 The effect of thalidomide on itch was studied in 11 patients with chronic pruritus from psoriasis, eczema, nodular prurigo, senile pruritus and primary biliary cirrhosis. Thalidomide 14-25 itchy E3 ubiquitin protein ligase Homo sapiens 29-33 10721827-2 2000 Itch, assessed subjectively by the patients on a 10 cm line and measured objectively as nocturnal scratch movement, was decreased by thalidomide 200 mg on the 2 nights it was given. Thalidomide 133-144 itchy E3 ubiquitin protein ligase Homo sapiens 0-4 11188934-3 2000 Thalidomide has been shown to block the activity of angiogenic substances like bFGF, VEGF and interleukin 6. Thalidomide 0-11 fibroblast growth factor 2 Homo sapiens 79-83 11188934-3 2000 Thalidomide has been shown to block the activity of angiogenic substances like bFGF, VEGF and interleukin 6. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 85-89 11188934-3 2000 Thalidomide has been shown to block the activity of angiogenic substances like bFGF, VEGF and interleukin 6. Thalidomide 0-11 interleukin 6 Homo sapiens 94-107 10632777-2 1999 Thalidomide, which inhibits TNF, is an effective drug, but has severe side-effects in pregnant women. Thalidomide 0-11 tumor necrosis factor Homo sapiens 28-31 10540197-2 1999 Thalidomide, and to a greater extent the new thalidomide analogue CC1069, inhibit monocyte TNF-alpha production both in vitro and in vivo. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 91-100 10540197-2 1999 Thalidomide, and to a greater extent the new thalidomide analogue CC1069, inhibit monocyte TNF-alpha production both in vitro and in vivo. Thalidomide 45-56 tumor necrosis factor Rattus norvegicus 91-100 10540197-13 1999 However, the development of rat AA can be blocked by the thalidomide analogue CC1069, which is an efficient inhibitor of TNF-alpha production and IL-2 in vitro. Thalidomide 57-68 tumor necrosis factor Rattus norvegicus 121-130 10540197-13 1999 However, the development of rat AA can be blocked by the thalidomide analogue CC1069, which is an efficient inhibitor of TNF-alpha production and IL-2 in vitro. Thalidomide 57-68 interleukin 2 Rattus norvegicus 146-150 10870669-1 2000 The potential therapeutic utility of thalidomide (Thd), an effective inhibitor of tumor necrosis factor (TNF)-alpha in vitro, was investigated in cynomolgus monkeys (Macaca fascicularis) at 10 months after infection with simian immunodeficiency virus (SIV). Thalidomide 37-48 tumor necrosis factor Macaca fascicularis 82-115 10870669-1 2000 The potential therapeutic utility of thalidomide (Thd), an effective inhibitor of tumor necrosis factor (TNF)-alpha in vitro, was investigated in cynomolgus monkeys (Macaca fascicularis) at 10 months after infection with simian immunodeficiency virus (SIV). Thalidomide 50-53 tumor necrosis factor Macaca fascicularis 82-115 10606973-0 2000 The thalidomide analogue CC-3052 inhibits HIV-1 and tumour necrosis factor-alpha (TNF-alpha) expression in acutely and chronically infected cells in vitro. Thalidomide 4-15 tumor necrosis factor Homo sapiens 82-91 10711629-0 2000 Metabolism of thalidomide in human microsomes, cloned human cytochrome P-450 isozymes, and Hansen"s disease patients. Thalidomide 14-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 10711629-1 2000 Previous in vitro studies in rat microsomal preparations suggested that thalidomide is metabolized by the cytochrome P450 system (CYP). Thalidomide 72-83 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 130-133 10711629-2 2000 In this study, we examined the extent of thalidomide metabolism by preparations of pooled human microsomes, microsomes containing cloned human CYP isozymes (CYPIA2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4), and Hansen"s disease patients. Thalidomide 41-52 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 143-146 10711629-2 2000 In this study, we examined the extent of thalidomide metabolism by preparations of pooled human microsomes, microsomes containing cloned human CYP isozymes (CYPIA2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4), and Hansen"s disease patients. Thalidomide 41-52 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 157-163 10711629-2 2000 In this study, we examined the extent of thalidomide metabolism by preparations of pooled human microsomes, microsomes containing cloned human CYP isozymes (CYPIA2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4), and Hansen"s disease patients. Thalidomide 41-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 10711629-3 2000 Results indicated that thalidomide was a poor substrate for CYP isozymes. Thalidomide 23-34 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 60-63 10607376-7 2000 Results obtained indicated that thalidomide was predominantly activated by P-450 isozyne CYP2E1, although weak cross-specificity between CYP1A1/A2 may have existed. Thalidomide 32-43 cytochrome P450 family 2 subfamily E member 1 L homeolog Xenopus laevis 89-95 10607376-7 2000 Results obtained indicated that thalidomide was predominantly activated by P-450 isozyne CYP2E1, although weak cross-specificity between CYP1A1/A2 may have existed. Thalidomide 32-43 cytochrome P450 family 1 subfamily A member 1 L homeolog Xenopus laevis 137-143 10651384-8 1999 Thalidomide may have antiretroviral effects as a result of its primarily inhibitory effects on the production of tumour necrosis factor alpha (TNFalpha). Thalidomide 0-11 tumor necrosis factor Homo sapiens 143-151 10651384-9 1999 TNFalpha induces expression of HIV from chronically infected cell lines by stimulating a cellular transcription factor, and blocking of TNFalpha-stimulated HIV replication by thalidomide has been shown in vitro and ex vivo. Thalidomide 175-186 tumor necrosis factor Homo sapiens 136-144 10579967-1 1999 BACKGROUND & AIMS: Inhibition of tumor necrosis factor is a proposed mechanism for the anti-inflammatory properties of thalidomide. Thalidomide 123-134 tumor necrosis factor Homo sapiens 37-58 10579968-1 1999 BACKGROUND & AIMS: Thalidomide decreases production of tumor necrosis factor alpha, a proinflammatory cytokine associated with Crohn"s disease (CD). Thalidomide 23-34 tumor necrosis factor Homo sapiens 59-86 10499831-0 1999 Inhibition of TNF-alpha synthesis with thalidomide for prevention of acute exacerbations and altering the natural history of multiple sclerosis. Thalidomide 39-50 tumor necrosis factor Homo sapiens 14-23 10353884-0 1999 Protein kinase C-dependent effects on leukocyte migration of thalidomide. Thalidomide 61-72 proline rich transmembrane protein 2 Homo sapiens 0-16 10353884-9 1999 In a cell-free assay, inhibition of PKC activation by bisindolylmaleimide could be reversed by thalidomide, indicating direct interactions of thalidomide with PKC. Thalidomide 95-106 proline rich transmembrane protein 2 Homo sapiens 36-39 10353884-9 1999 In a cell-free assay, inhibition of PKC activation by bisindolylmaleimide could be reversed by thalidomide, indicating direct interactions of thalidomide with PKC. Thalidomide 95-106 proline rich transmembrane protein 2 Homo sapiens 159-162 10353884-9 1999 In a cell-free assay, inhibition of PKC activation by bisindolylmaleimide could be reversed by thalidomide, indicating direct interactions of thalidomide with PKC. Thalidomide 142-153 proline rich transmembrane protein 2 Homo sapiens 36-39 10353884-9 1999 In a cell-free assay, inhibition of PKC activation by bisindolylmaleimide could be reversed by thalidomide, indicating direct interactions of thalidomide with PKC. Thalidomide 142-153 proline rich transmembrane protein 2 Homo sapiens 159-162 10571725-9 1999 Thalidomide did, however, induce a 2-4-fold increase in keratinocyte-derived interleukin-8, a pro-migratory cellular autocrine factor. Thalidomide 0-11 C-X-C motif chemokine ligand 8 Homo sapiens 77-90 10480630-8 1999 When purified T cells from HIV-infected individuals were stimulated by immobilized anti-CD3 in the presence of thalidomide, a costimulatory effect of the drug was observed, resulting in increased production of IL-2 and IFN-gamma, and increased T cell-proliferative responses. Thalidomide 111-122 interleukin 2 Homo sapiens 210-214 10480630-8 1999 When purified T cells from HIV-infected individuals were stimulated by immobilized anti-CD3 in the presence of thalidomide, a costimulatory effect of the drug was observed, resulting in increased production of IL-2 and IFN-gamma, and increased T cell-proliferative responses. Thalidomide 111-122 interferon gamma Homo sapiens 219-228 10384139-0 1999 Differential cytokine modulation and T cell activation by two distinct classes of thalidomide analogues that are potent inhibitors of TNF-alpha. Thalidomide 82-93 tumor necrosis factor Homo sapiens 134-143 10384139-2 1999 Our previous work has shown thalidomide to be a relatively selective inhibitor of TNF-alpha production in vivo and in vitro. Thalidomide 28-39 tumor necrosis factor Homo sapiens 82-91 10384139-4 1999 To develop thalidomide analogues with increased anti-TNF-alpha activity and reduced or absent toxicities, novel TNF-alpha inhibitors were designed and synthesized. Thalidomide 11-22 tumor necrosis factor Homo sapiens 53-62 10384139-4 1999 To develop thalidomide analogues with increased anti-TNF-alpha activity and reduced or absent toxicities, novel TNF-alpha inhibitors were designed and synthesized. Thalidomide 11-22 tumor necrosis factor Homo sapiens 112-121 10499831-4 1999 There is evidence that thalidomide is an agent which blocks production of TNF-alpha by a mechanism different from other agents. Thalidomide 23-34 tumor necrosis factor Homo sapiens 74-83 10424439-1 1999 Both thalidomide and dexamethasone have been shown to inhibit the production of tumour necrosis factor alpha (TNF-alpha), but little is known of their cellular selectivity. Thalidomide 5-16 tumor necrosis factor Homo sapiens 110-119 10386948-1 1999 Thalidomide, (1), is a known inhibitor of TNF-alpha release in LPS stimulated human PBMC. Thalidomide 0-11 tumor necrosis factor Homo sapiens 42-51 10386948-2 1999 Herein we describe the TNF-alpha inhibitory activity of amino substituted analogs of thalidomide (1) and its isoindolin-1-one analog, EM-12 (2). Thalidomide 85-96 tumor necrosis factor Homo sapiens 23-32 10437657-0 1999 Inducer-specific bidirectional regulation of endothelial interleukin-8 production by thalidomide. Thalidomide 85-96 C-X-C motif chemokine ligand 8 Homo sapiens 57-70 10437657-6 1999 Upon stimulation with TNFalpha or LPS, HUVEC produced increased amounts of eIL-8 and THD affected this process in a bidirectional manner, with augmentation of TNFalpha- and inhibition of LPS-effects. Thalidomide 85-88 tumor necrosis factor Homo sapiens 22-30 10437657-6 1999 Upon stimulation with TNFalpha or LPS, HUVEC produced increased amounts of eIL-8 and THD affected this process in a bidirectional manner, with augmentation of TNFalpha- and inhibition of LPS-effects. Thalidomide 85-88 tumor necrosis factor Homo sapiens 159-167 10424439-2 1999 Inhibition of monocyte TNF-alpha expression has been implicated in the clinical efficacy of thalidomide, and it has been suggested that the drug modulates only monocyte-derived cytokines. Thalidomide 92-103 tumor necrosis factor Homo sapiens 23-32 10424439-4 1999 In the present investigations we have examined the influence of both thalidomide and dexamethasone on mitogen-induced elaboration of TNF-alpha by CD3+ peripheral blood mononuclear cells (PBMC) and the T cell line MOLT-4. Thalidomide 69-80 tumor necrosis factor Homo sapiens 133-142 10424439-8 1999 Thalidomide was found to inhibit PBMC-derived TNF-alpha, but not IL-2. Thalidomide 0-11 tumor necrosis factor Homo sapiens 46-55 10424439-10 1999 Thalidomide and dexamethasone both suppressed intracellular levels of TNF-alpha in CD3+ PBMC, reducing percentages of double positive staining cells by 28 and 52%, respectively, compared with controls. Thalidomide 0-11 tumor necrosis factor Homo sapiens 70-79 10424439-11 1999 In addition, TNF-alpha production by CD3- PBMC was inhibited by 31% by thalidomide and by 47% by dexamethasone. Thalidomide 71-82 tumor necrosis factor Homo sapiens 13-22 10424439-13 1999 TNF-alpha secretion by PMA/ionophore activated MOLT-4 cells was reduced by 80% following thalidomide treatment and close to background levels following dexamethasone treatment. Thalidomide 89-100 tumor necrosis factor Homo sapiens 0-9 10336480-11 1999 A variety of well characterized angiogenesis inhibitors (including angiostatin, fumagillin, 2-methoxy estradiol, transforming growth factor-beta, and thalidomide) effectively blocked VEGF-induced down-regulation of caveolin-1 as seen by immunoblotting and immunofluorescence microscopy. Thalidomide 150-161 vascular endothelial growth factor A Homo sapiens 183-187 10424439-14 1999 To verify that thalidomide was acting selectively to down-regulate TNF-alpha, IL-2 production by MOLT-4 cells was also measured and found to be unaffected by the drug. Thalidomide 15-26 tumor necrosis factor Homo sapiens 67-76 10336480-11 1999 A variety of well characterized angiogenesis inhibitors (including angiostatin, fumagillin, 2-methoxy estradiol, transforming growth factor-beta, and thalidomide) effectively blocked VEGF-induced down-regulation of caveolin-1 as seen by immunoblotting and immunofluorescence microscopy. Thalidomide 150-161 caveolin 1 Homo sapiens 215-225 10424439-14 1999 To verify that thalidomide was acting selectively to down-regulate TNF-alpha, IL-2 production by MOLT-4 cells was also measured and found to be unaffected by the drug. Thalidomide 15-26 interleukin 2 Homo sapiens 78-82 10424439-16 1999 These observations suggest that thalidomide, in addition to its known inhibitory effect on monocyte-derived TNF-alpha, is capable also of down-regulating T cell-derived TNF-alpha in a direct and selective manner. Thalidomide 32-43 tumor necrosis factor Homo sapiens 108-117 10424439-16 1999 These observations suggest that thalidomide, in addition to its known inhibitory effect on monocyte-derived TNF-alpha, is capable also of down-regulating T cell-derived TNF-alpha in a direct and selective manner. Thalidomide 32-43 tumor necrosis factor Homo sapiens 169-178 10424439-17 1999 In addition, the inhibition of intracellular levels of TNF-alpha strengthens the evidence that the inhibitory effect of thalidomide is at the level of transcription and/or translation and does not reduce cellular TNF-alpha secretion. Thalidomide 120-131 tumor necrosis factor Homo sapiens 55-64 10080541-7 1999 Thalidomide, a drug known to inhibit TNF-alpha synthesis on monocytes, also exerted an inhibitory effect on TNF-alpha secretion in neutrophils. Thalidomide 0-11 tumor necrosis factor Homo sapiens 37-46 10318940-3 1999 Survival was associated with inhibition of tumor necrosis factor alpha (TNF-alpha) production by thalidomide. Thalidomide 97-108 tumor necrosis factor Mus musculus 72-81 10360649-4 1999 Co-administration of thalidomide with DMXAA increased anti-tumour activity and increased intra-tumoural TNF-alpha production approximately tenfold over that obtained with DMXAA alone. Thalidomide 21-32 tumor necrosis factor Mus musculus 104-113 10360649-5 1999 Thalidomide increased splenic TNF-alpha production slightly but significantly decreased serum and hepatic levels of TNF-alpha induced with DMXAA. Thalidomide 0-11 tumor necrosis factor Mus musculus 30-39 10360649-5 1999 Thalidomide increased splenic TNF-alpha production slightly but significantly decreased serum and hepatic levels of TNF-alpha induced with DMXAA. Thalidomide 0-11 tumor necrosis factor Mus musculus 116-125 10360649-7 1999 Splenic TNF-alpha activity induced with LPS was slightly increased with thalidomide, but serum and liver TNF-alpha levels were suppressed. Thalidomide 72-83 tumor necrosis factor Mus musculus 8-17 10360649-7 1999 Splenic TNF-alpha activity induced with LPS was slightly increased with thalidomide, but serum and liver TNF-alpha levels were suppressed. Thalidomide 72-83 toll-like receptor 4 Mus musculus 40-43 10360649-10 1999 The increase in the anti-tumour action by thalidomide in combination with DMXAA corresponded to an increase in intra-tumoural TNF-alpha production. Thalidomide 42-53 tumor necrosis factor Mus musculus 126-135 10360649-11 1999 Co-administration of thalidomide may represent a novel approach to improving selective intra-tumoural TNF-alpha production and anti-tumour efficacy of DMXAA. Thalidomide 21-32 tumor necrosis factor Mus musculus 102-111 10229222-0 1999 Thalidomide on the comeback trail. Thalidomide 0-11 TNF superfamily member 10 Homo sapiens 28-33 10080541-7 1999 Thalidomide, a drug known to inhibit TNF-alpha synthesis on monocytes, also exerted an inhibitory effect on TNF-alpha secretion in neutrophils. Thalidomide 0-11 tumor necrosis factor Homo sapiens 108-117 9780198-1 1998 The immunomodulatory drug thalidomide has been shown to be clinically useful in a number of situations due to its ability to inhibit TNF-alpha synthesis. Thalidomide 26-37 tumor necrosis factor Homo sapiens 133-142 10077449-1 1999 Recently, we developed novel tumor necrosis factor (TNF)-alpha production regulators with a phthalimide skeleton derived from thalidomide. Thalidomide 126-137 tumor necrosis factor Mus musculus 29-62 10357564-0 1999 Thalidomide up-regulates prostate-specific antigen secretion from LNCaP cells. Thalidomide 0-11 kallikrein related peptidase 3 Homo sapiens 25-50 10357564-9 1999 Thalidomide-treated LNCaP cells demonstrated increased PSA/cell levels at all concentrations tested compared to untreated control cells. Thalidomide 0-11 kallikrein related peptidase 3 Homo sapiens 55-58 10357564-11 1999 Comparison of cDNA expression arrays hybridized with thalidomide-treated LNCaP cDNA probes suggests that thalidomide may up- or downregulate expression of angiogenesis-related genes, i.e., vitronectin, but these differential effects require further verification. Thalidomide 105-116 vitronectin Homo sapiens 189-200 10357564-12 1999 Thalidomide over a range of doses has demonstrated nontoxic, cytostatic activity in LNCaP cells and significant upregulation of LNCaP cell PSA secretion in vitro. Thalidomide 0-11 kallikrein related peptidase 3 Homo sapiens 139-142 10357564-13 1999 Furthermore, preliminary data from cDNA nucleic acid arrays of thalidomide-treated LNCaP cells suggest that thalidomide upregulates a potential angiogenic modulatory protein, the vitronectin precursor, which may eventually link thalidomide"s antiangiogenic activity with modulation of angiogenic vascular integrin pathways. Thalidomide 63-74 vitronectin Homo sapiens 179-190 10357564-13 1999 Furthermore, preliminary data from cDNA nucleic acid arrays of thalidomide-treated LNCaP cells suggest that thalidomide upregulates a potential angiogenic modulatory protein, the vitronectin precursor, which may eventually link thalidomide"s antiangiogenic activity with modulation of angiogenic vascular integrin pathways. Thalidomide 108-119 vitronectin Homo sapiens 179-190 10357564-13 1999 Furthermore, preliminary data from cDNA nucleic acid arrays of thalidomide-treated LNCaP cells suggest that thalidomide upregulates a potential angiogenic modulatory protein, the vitronectin precursor, which may eventually link thalidomide"s antiangiogenic activity with modulation of angiogenic vascular integrin pathways. Thalidomide 108-119 vitronectin Homo sapiens 179-190 9894629-8 1999 Thalidomide was shown to be effective for treating oral and genital ulcers and follicular lesions in Behcet"s syndrome; severe refractory Behcet"s syndrome uveitis responded to treatment with IFN-alpha. Thalidomide 0-11 interferon alpha 1 Homo sapiens 192-201 9859910-7 1998 Meanwhile, thalidomide has been shown to possess a range of biologic actions, including inhibition of tumor necrosis factor alpha, possibly relevant to its clinical efficacy. Thalidomide 11-22 tumor necrosis factor Homo sapiens 102-129 10408702-7 1999 When we combined sulindac with thalidomide, we found a significantly increased inhibition of bFGF- or VEGF-induced corneal neovascularization (by 63% or 74% respectively) compared with either agent alone (P < 0.01). Thalidomide 31-42 vascular endothelial growth factor A Oryctolagus cuniculus 102-106 15991946-3 1998 Meanwhile, immunomodulatory and anti-inflammatory activities of thalidomide, particularly its inhibition of tumour necrosis factor alpha (TNF-alpha), have been identified and elucidated. Thalidomide 64-75 tumor necrosis factor Homo sapiens 138-147 9877451-3 1998 TNF-alpha production in IL-1beta-stimulated or PMA-stimulated hepatocyte cultures was not altered following the addition of dihydrocortisone (< or = 1 microg/ml), dibutyryl-cAMP (db-cAMP, < or = 100 micromol/l), adenosine (< or = 1 mmol/l), thalidomide (< or = 25 microg/ml), or cyclosporine (< or = 300 ng/ml). Thalidomide 250-261 tumor necrosis factor Homo sapiens 0-9 9843104-3 1998 Thalidomide is a potent inhibitor of TNF-alpha action. Thalidomide 0-11 tumor necrosis factor Homo sapiens 37-46 9843104-11 1998 Plasma TNF-alpha concentration was higher in the thalidomide group than the placebo group on day 2, though the difference was not significant (Wilcoxon rank-sum test p=0.07). Thalidomide 49-60 tumor necrosis factor Homo sapiens 7-16 9843104-12 1998 INTERPRETATION: Even though few patients were included, our data suggest that thalidomide is detrimental in TEN, possibly because of a paradoxical enhancement of TNF-alpha production. Thalidomide 78-89 tumor necrosis factor Homo sapiens 162-171 9891501-3 1998 Unexpectedly, the drug thalidomide, while reducing the serum TNF response to DMXAA, potentiates its antitumour effect. Thalidomide 23-34 tumor necrosis factor Mus musculus 61-64 10022026-7 1998 Thalidomide altered expression of bcl-2 (p < 0.011) and tissue PSA (p < 0.002). Thalidomide 0-11 BCL2, apoptosis regulator Rattus norvegicus 34-39 10022026-7 1998 Thalidomide altered expression of bcl-2 (p < 0.011) and tissue PSA (p < 0.002). Thalidomide 0-11 aminopeptidase puromycin sensitive Rattus norvegicus 66-69 9873600-1 1998 N-Phthaloyl 3-amino-3-arylpropionic acid analogs of thalidomide that are potent inhibitors of tumor necrosis factor-alpha are reported. Thalidomide 52-63 tumor necrosis factor Homo sapiens 94-121 9780198-3 1998 Recently, structural modifications of thalidomide have been designed enabling greatly enhanced anti-TNF-alpha activity in LPS-treated mice. Thalidomide 38-49 tumor necrosis factor Mus musculus 100-109 9692223-3 1998 The data indicates that the TNF-alpha production enhancing activity of thalidomide derivatives depends on both the electronic-state of substituents at the fused benzene ring and the stereochemistry of the glutarimide moiety. Thalidomide 71-82 tumor necrosis factor Homo sapiens 28-37 9790310-5 1998 This clinical effect was associated with increased numbers of inflammatory cells in sciatic nerve sections and elevated numbers of interferon-gamma (IFN-gamma) mRNA-expressing cells among lymph node mononuclear cells from thalidomide-treated EAN rats on day 17 postimmunization, i.e. at the peak of clinical EAN. Thalidomide 222-233 interferon gamma Rattus norvegicus 131-147 9790310-5 1998 This clinical effect was associated with increased numbers of inflammatory cells in sciatic nerve sections and elevated numbers of interferon-gamma (IFN-gamma) mRNA-expressing cells among lymph node mononuclear cells from thalidomide-treated EAN rats on day 17 postimmunization, i.e. at the peak of clinical EAN. Thalidomide 222-233 interferon gamma Rattus norvegicus 149-158 9703279-2 1998 Our previous studies have shown that thalidomide, a potent inhibitor of TNF biosynthesis that has numerous biological effects, including inhibition of tumour angiogenesis, unexpectedly augments the anti-tumour response in mice to DMXAA. Thalidomide 37-48 tumor necrosis factor Mus musculus 72-75 18465584-0 1998 Thalidomide Celgene Corp. Thalidomide, in development by Celgene, inhibits the effects of elevated TNFalpha and may consequently be of use in a range of diseases including cachexia, bacterial meningitis, rheumatoid arthritis, septic shock, AIDS, tuberculosis, multiple sclerosis, ulcerative colitis, graft-versus-host disease and systemic lupus erythematosus. Thalidomide 0-11 tumor necrosis factor Homo sapiens 99-107 18465584-0 1998 Thalidomide Celgene Corp. Thalidomide, in development by Celgene, inhibits the effects of elevated TNFalpha and may consequently be of use in a range of diseases including cachexia, bacterial meningitis, rheumatoid arthritis, septic shock, AIDS, tuberculosis, multiple sclerosis, ulcerative colitis, graft-versus-host disease and systemic lupus erythematosus. Thalidomide 26-37 tumor necrosis factor Homo sapiens 99-107 18465584-0 1998 Thalidomide Celgene Corp. Thalidomide, in development by Celgene, inhibits the effects of elevated TNFalpha and may consequently be of use in a range of diseases including cachexia, bacterial meningitis, rheumatoid arthritis, septic shock, AIDS, tuberculosis, multiple sclerosis, ulcerative colitis, graft-versus-host disease and systemic lupus erythematosus. Thalidomide 12-19 tumor necrosis factor Homo sapiens 99-107 9646092-0 1998 Thalidomide inhibits corneal angiogenesis induced by vascular endothelial growth factor. Thalidomide 0-11 vascular endothelial growth factor A Oryctolagus cuniculus 53-87 9562600-5 1998 The regulatory effect of thalidomide on TNF-alpha production has been found to be bidirectional, depending on both the cell-type and the TNF-alpha production-inducer; i.e., thalidomide possesses both enhancing and inhibiting activities on TNF-alpha production. Thalidomide 25-36 tumor necrosis factor Homo sapiens 40-49 9562600-5 1998 The regulatory effect of thalidomide on TNF-alpha production has been found to be bidirectional, depending on both the cell-type and the TNF-alpha production-inducer; i.e., thalidomide possesses both enhancing and inhibiting activities on TNF-alpha production. Thalidomide 25-36 tumor necrosis factor Homo sapiens 137-146 9562600-5 1998 The regulatory effect of thalidomide on TNF-alpha production has been found to be bidirectional, depending on both the cell-type and the TNF-alpha production-inducer; i.e., thalidomide possesses both enhancing and inhibiting activities on TNF-alpha production. Thalidomide 25-36 tumor necrosis factor Homo sapiens 137-146 9562600-5 1998 The regulatory effect of thalidomide on TNF-alpha production has been found to be bidirectional, depending on both the cell-type and the TNF-alpha production-inducer; i.e., thalidomide possesses both enhancing and inhibiting activities on TNF-alpha production. Thalidomide 173-184 tumor necrosis factor Homo sapiens 40-49 9776474-6 1998 Analysis of supernatants by enzyme-linked immunosorbent assay (ELISA) showed that thalidomide caused a dose-dependent inhibition of the pro-inflammatory cytokines interleukin 6 (IL-6) and tumour necrosis factor alpha (TNF-alpha), maximally reducing production by 20 (P < 0.05) and 30% (P < 0.01), respectively, compared with controls. Thalidomide 82-93 interleukin 6 Homo sapiens 163-176 9776474-6 1998 Analysis of supernatants by enzyme-linked immunosorbent assay (ELISA) showed that thalidomide caused a dose-dependent inhibition of the pro-inflammatory cytokines interleukin 6 (IL-6) and tumour necrosis factor alpha (TNF-alpha), maximally reducing production by 20 (P < 0.05) and 30% (P < 0.01), respectively, compared with controls. Thalidomide 82-93 interleukin 6 Homo sapiens 178-182 9776474-6 1998 Analysis of supernatants by enzyme-linked immunosorbent assay (ELISA) showed that thalidomide caused a dose-dependent inhibition of the pro-inflammatory cytokines interleukin 6 (IL-6) and tumour necrosis factor alpha (TNF-alpha), maximally reducing production by 20 (P < 0.05) and 30% (P < 0.01), respectively, compared with controls. Thalidomide 82-93 tumor necrosis factor Homo sapiens 218-227 9776474-10 1998 Reverse transcription-polymerase chain reaction (RT-PCR) analyses showed that thalidomide inhibited selectively the expression of TNF-alpha and IL-6 mRNA, whereas dexamethasone inhibited mRNA levels of all cytokines examined. Thalidomide 78-89 tumor necrosis factor Homo sapiens 130-139 9776474-10 1998 Reverse transcription-polymerase chain reaction (RT-PCR) analyses showed that thalidomide inhibited selectively the expression of TNF-alpha and IL-6 mRNA, whereas dexamethasone inhibited mRNA levels of all cytokines examined. Thalidomide 78-89 interleukin 6 Homo sapiens 144-148 9776474-12 1998 Conversely, thalidomide selectively inhibits the production of IL-6 and TNF-alpha. Thalidomide 12-23 interleukin 6 Homo sapiens 63-67 9776474-12 1998 Conversely, thalidomide selectively inhibits the production of IL-6 and TNF-alpha. Thalidomide 12-23 tumor necrosis factor Homo sapiens 72-81 9657054-0 1998 Thalidomide promotes the release of tumor necrosis factor-alpha (TNF-alpha) and lethality by lipopolysaccharide in mice. Thalidomide 0-11 tumor necrosis factor Mus musculus 36-63 9657054-0 1998 Thalidomide promotes the release of tumor necrosis factor-alpha (TNF-alpha) and lethality by lipopolysaccharide in mice. Thalidomide 0-11 tumor necrosis factor Mus musculus 65-74 9657054-1 1998 We investigated the in vivo effects of thalidomide on the production of tumor necrosis factor-alpha (TNF-alpha). Thalidomide 39-50 tumor necrosis factor Mus musculus 72-99 9657054-1 1998 We investigated the in vivo effects of thalidomide on the production of tumor necrosis factor-alpha (TNF-alpha). Thalidomide 39-50 tumor necrosis factor Mus musculus 101-110 9657054-3 1998 When thalidomide (1, 3, or 6 mg/kg) was administered intraperitoneally 3 h before the injection of LPS (0.3 mg/kg, i. p.), thalidomide markedly enhanced LPS-induced TNF-alpha release in a dose-dependent manner. Thalidomide 5-16 tumor necrosis factor Mus musculus 165-174 9657054-3 1998 When thalidomide (1, 3, or 6 mg/kg) was administered intraperitoneally 3 h before the injection of LPS (0.3 mg/kg, i. p.), thalidomide markedly enhanced LPS-induced TNF-alpha release in a dose-dependent manner. Thalidomide 123-134 tumor necrosis factor Mus musculus 165-174 9657054-6 1998 We conclude that thalidomide enhances in vivo TNF-alpha secretion and the lethality of LPS in mice. Thalidomide 17-28 tumor necrosis factor Mus musculus 46-55 9562600-5 1998 The regulatory effect of thalidomide on TNF-alpha production has been found to be bidirectional, depending on both the cell-type and the TNF-alpha production-inducer; i.e., thalidomide possesses both enhancing and inhibiting activities on TNF-alpha production. Thalidomide 173-184 tumor necrosis factor Homo sapiens 137-146 9562600-5 1998 The regulatory effect of thalidomide on TNF-alpha production has been found to be bidirectional, depending on both the cell-type and the TNF-alpha production-inducer; i.e., thalidomide possesses both enhancing and inhibiting activities on TNF-alpha production. Thalidomide 173-184 tumor necrosis factor Homo sapiens 137-146 9562600-6 1998 Structural modification of thalidomide aiming at the creation of superior TNF-alpha production-regulators has afforded a number of phenyl- and benzylphthalimide analogs possessing more potent activity than thalidomide itself. Thalidomide 27-38 tumor necrosis factor Homo sapiens 74-83 9562600-6 1998 Structural modification of thalidomide aiming at the creation of superior TNF-alpha production-regulators has afforded a number of phenyl- and benzylphthalimide analogs possessing more potent activity than thalidomide itself. Thalidomide 206-217 tumor necrosis factor Homo sapiens 74-83 9646092-5 1998 We therefore investigated the effect of thalidomide on corneal angiogenesis induced by vascular endothelial growth factor (VEGF), which has a special role among angiogenic growth factors. Thalidomide 40-51 vascular endothelial growth factor A Oryctolagus cuniculus 87-121 9646092-5 1998 We therefore investigated the effect of thalidomide on corneal angiogenesis induced by vascular endothelial growth factor (VEGF), which has a special role among angiogenic growth factors. Thalidomide 40-51 vascular endothelial growth factor A Oryctolagus cuniculus 123-127 9646092-10 1998 CONCLUSIONS: Thalidomide has a significant antiangiogenic effect against VEGF-induced neovasclar growth. Thalidomide 13-24 vascular endothelial growth factor A Oryctolagus cuniculus 73-77 9690766-4 1998 Evidence is presented here suggesting that the embryonic action of thalidomide was mediated by TNF-alpha, since the drug is a powerful inhibitor of the synthesis of this cytokine. Thalidomide 67-78 tumor necrosis factor Homo sapiens 95-104 9607928-0 1998 Thalidomide costimulates primary human T lymphocytes, preferentially inducing proliferation, cytokine production, and cytotoxic responses in the CD8+ subset. Thalidomide 0-11 CD8a molecule Homo sapiens 145-148 9607928-1 1998 The efficacy of thalidomide (alpha-phthalimido-glutarimide) therapy in leprosy patients with erythema nodosum leprosum is thought to be due to inhibition of tumor necrosis factor alpha. Thalidomide 16-27 tumor necrosis factor Homo sapiens 157-184 9607928-1 1998 The efficacy of thalidomide (alpha-phthalimido-glutarimide) therapy in leprosy patients with erythema nodosum leprosum is thought to be due to inhibition of tumor necrosis factor alpha. Thalidomide 29-58 tumor necrosis factor Homo sapiens 157-184 9607928-3 1998 We show that thalidomide is a potent costimulator of primary human T cells in vitro, synergizing with stimulation via the T cell receptor complex to increase interleukin 2-mediated T cell proliferation and interferon gamma production. Thalidomide 13-24 interleukin 2 Homo sapiens 158-171 9516386-11 1998 Specific inhibition of TNF-alpha using neutralizing antibody or alpha-N-phthalimidoglutarimide (thalidomide) resulted in reversal of the hypoxia-evoked responses. Thalidomide 64-94 tumor necrosis factor Homo sapiens 23-32 9516386-11 1998 Specific inhibition of TNF-alpha using neutralizing antibody or alpha-N-phthalimidoglutarimide (thalidomide) resulted in reversal of the hypoxia-evoked responses. Thalidomide 96-107 tumor necrosis factor Homo sapiens 23-32 9607834-6 1998 When thalidomide treatment was combined with antibiotics, there was a marked reduction in TNF-alpha levels, leukocytosis, and brain pathology. Thalidomide 5-16 tumor necrosis factor Oryctolagus cuniculus 90-99 9623511-5 1998 TNF levels were slightly decreased after 1 month of thalidomide treatment and significantly decreased after 2 months of treatment. Thalidomide 52-63 tumor necrosis factor Homo sapiens 0-3 9623511-6 1998 Serum IFN-gamma was significantly lower in patients at the onset of ENL and was increased after 1 and 2 months of thalidomide treatment. Thalidomide 114-125 interferon gamma Homo sapiens 6-15 9598899-1 1998 OBJECTIVE: Thalidomide has been described as an inhibitor of both angiogenesis (which may account for its teratogenic effects on limb bud formation) and tumor necrosis factor-alpha (TNF-alpha) production. Thalidomide 11-22 tumor necrosis factor Rattus norvegicus 153-180 9598899-1 1998 OBJECTIVE: Thalidomide has been described as an inhibitor of both angiogenesis (which may account for its teratogenic effects on limb bud formation) and tumor necrosis factor-alpha (TNF-alpha) production. Thalidomide 11-22 tumor necrosis factor Rattus norvegicus 182-191 9493761-17 1998 Examples of human teratogens that are substrates for CYP enzymes include thalidomide, phenytoin, ethanol, and several hormonal agents. Thalidomide 73-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 9507696-0 1998 5"-Substituted thalidomide analogs as modulators of TNF-alpha. Thalidomide 15-26 tumor necrosis factor Homo sapiens 52-61 9625384-2 1998 Pentoxyfylline and thalidomide are inhibitors of TNF-alpha that have been tried as rational therapeutic interventions in cachexia. Thalidomide 19-30 tumor necrosis factor Homo sapiens 49-58 9502617-2 1998 In addition, thalidomide has been shown to be an inhibitor of angiogenesis induced by basic fibroblast growth factor (bFGF). Thalidomide 13-24 fibroblast growth factor 2 Cavia porcellus 86-116 9502617-2 1998 In addition, thalidomide has been shown to be an inhibitor of angiogenesis induced by basic fibroblast growth factor (bFGF). Thalidomide 13-24 fibroblast growth factor 2 Cavia porcellus 118-122 9486396-12 1998 The amount of TNF-alpha mRNA was increased in the tissue during ENL compared with before the reaction, and decreased thereafter following treatment for reaction (either PTX or thalidomide). Thalidomide 176-187 tumor necrosis factor Homo sapiens 14-23 9430254-6 1997 However, a nonsignificant trend toward inhibition of mitogen-induced TNF-alpha production was observed in the thalidomide arm. Thalidomide 110-121 tumor necrosis factor Homo sapiens 69-78 9430254-7 1997 The lack of systemic effect and the lower tolerance of thalidomide (at this dose) in asymptomatic patients highlights the need for pharmacokinetic analysis to address possible absorption problems and the need for more potent and less toxic TNF-alpha inhibitors to be developed for use in this type of study. Thalidomide 55-66 tumor necrosis factor Homo sapiens 240-249 9402031-4 1997 In the present study, we investigated in vitro the effects of thalidomide (THD) on activation of endothelial cells for enhanced transmigration of neutrophils by lipopolysaccharide (LPS), tumor necrosis factor-alpha (TNF), and interleukin-1 (IL-1). Thalidomide 75-78 tumor necrosis factor Homo sapiens 187-214 9367395-0 1997 Thalidomide and derivatives: immunological investigations of tumour necrosis factor-alpha (TNF-alpha) inhibition suggest drugs capable of selective gene regulation. Thalidomide 0-11 tumor necrosis factor Homo sapiens 91-100 9402031-6 1997 Treatment of HUVEC with THD in combination with LPS, TNF, and IL-1, respectively, antagonized LPS-activated transmigration of neutrophils but stimulated the effects of TNF and IL-1. Thalidomide 24-27 tumor necrosis factor Homo sapiens 53-56 9402031-6 1997 Treatment of HUVEC with THD in combination with LPS, TNF, and IL-1, respectively, antagonized LPS-activated transmigration of neutrophils but stimulated the effects of TNF and IL-1. Thalidomide 24-27 tumor necrosis factor Homo sapiens 168-171 9402031-6 1997 Treatment of HUVEC with THD in combination with LPS, TNF, and IL-1, respectively, antagonized LPS-activated transmigration of neutrophils but stimulated the effects of TNF and IL-1. Thalidomide 24-27 interleukin 1 alpha Homo sapiens 176-180 9496438-10 1997 The LD50 of LPS in this model was increased by thalidomide pre-treatment from 150 micrograms to 300 micrograms in 72 h. Thus, at otherwise lethal doses of LPS, thalidomide treatment was found to protect animals from death. Thalidomide 47-58 toll-like receptor 4 Mus musculus 12-15 9496438-10 1997 The LD50 of LPS in this model was increased by thalidomide pre-treatment from 150 micrograms to 300 micrograms in 72 h. Thus, at otherwise lethal doses of LPS, thalidomide treatment was found to protect animals from death. Thalidomide 160-171 toll-like receptor 4 Mus musculus 12-15 9496438-1 1997 Thalidomide has been shown to selectively inhibit TNF-alpha production in vitro by lipopolysaccharide (LPS)-stimulated monocytes. Thalidomide 0-11 tumor necrosis factor Mus musculus 50-59 9496438-10 1997 The LD50 of LPS in this model was increased by thalidomide pre-treatment from 150 micrograms to 300 micrograms in 72 h. Thus, at otherwise lethal doses of LPS, thalidomide treatment was found to protect animals from death. Thalidomide 160-171 toll-like receptor 4 Mus musculus 155-158 9496438-1 1997 Thalidomide has been shown to selectively inhibit TNF-alpha production in vitro by lipopolysaccharide (LPS)-stimulated monocytes. Thalidomide 0-11 toll-like receptor 4 Mus musculus 103-106 9496438-3 1997 Using a mouse model of LPS-induced shock, we investigated the effects of thalidomide on the production of TNF-alpha and other cytokines and on animal survival. Thalidomide 73-84 tumor necrosis factor Mus musculus 106-115 9294124-8 1997 Consistent with these findings, thalidomide (which antagonizes TNF action) attenuates c-src induction by LPS. Thalidomide 32-43 tumor necrosis factor Mus musculus 63-66 9496438-5 1997 Administration of 200 mg/kg thalidomide to mice before LPS challenge modified the profile of LPS-induced cytokine secretion. Thalidomide 28-39 toll-like receptor 4 Mus musculus 55-58 9496438-5 1997 Administration of 200 mg/kg thalidomide to mice before LPS challenge modified the profile of LPS-induced cytokine secretion. Thalidomide 28-39 toll-like receptor 4 Mus musculus 93-96 9496438-8 1997 Thalidomide induced a two-fold increase in serum IL-10 levels. Thalidomide 0-11 interleukin 10 Mus musculus 49-54 9197379-3 1997 The thalidomide analogs were previously shown to inhibit TNF-alpha production in vitro at much lower concentrations than thalidomide. Thalidomide 4-15 tumor necrosis factor Homo sapiens 57-66 15989668-0 1997 TNF-alpha antagonists: monoclonal antibodies, soluble receptors, thalidomide and other novel approaches. Thalidomide 65-76 tumor necrosis factor Homo sapiens 0-9 9248860-6 1997 However, neutrophils obtained from HIV-infected patients treated with thalidomide showed reduced expression of CD16, a surrogate marker for apoptosis of neutrophils. Thalidomide 70-81 Fc gamma receptor IIIa Homo sapiens 111-115 9278924-2 1997 Studies in marmosets and humans indicate that thalidomide, and a teratogenic analogue, decrease the expression of beta integrin subunits, most notably beta 3 and the beta 2 produced by leukocytes. Thalidomide 46-57 potassium calcium-activated channel subfamily M regulatory beta subunit 2 Homo sapiens 166-172 9278924-3 1997 Since integrins are crucial for cell-matrix interactions, and the beta 2 integrins of leukocytes mediate adhesion to endothelium, it is reasonable to postulate that thalidomide inhibits cell migration in susceptible species, and that this accounts for its anti-inflammatory, anti-angiogenic, and teratogenic activity. Thalidomide 165-176 potassium calcium-activated channel subfamily M regulatory beta subunit 2 Homo sapiens 66-72 9301478-1 1997 Thalidomide, when administered orally, is an inhibitor of angiogenesis in the basic fibroblast growth factor (bFGF)-induced rabbit cornea micropocket assay. Thalidomide 0-11 fibroblast growth factor 2 Mus musculus 110-114 9301478-2 1997 We now show in the mouse that thalidomide given intraperitoneally but not orally significantly inhibits bFGF-induced and vascular endothelial growth factor (VEGF)-induced corneal neovascularization. Thalidomide 30-41 fibroblast growth factor 2 Mus musculus 104-108 9301478-2 1997 We now show in the mouse that thalidomide given intraperitoneally but not orally significantly inhibits bFGF-induced and vascular endothelial growth factor (VEGF)-induced corneal neovascularization. Thalidomide 30-41 vascular endothelial growth factor A Mus musculus 121-155 9301478-2 1997 We now show in the mouse that thalidomide given intraperitoneally but not orally significantly inhibits bFGF-induced and vascular endothelial growth factor (VEGF)-induced corneal neovascularization. Thalidomide 30-41 vascular endothelial growth factor A Mus musculus 157-161 9301478-3 1997 We further demonstrate that this inhibition is independent from thalidomide"s ability to suppress tumor necrosis factor-alpha (TNF-alpha) production. Thalidomide 64-75 tumor necrosis factor Mus musculus 98-125 9301478-3 1997 We further demonstrate that this inhibition is independent from thalidomide"s ability to suppress tumor necrosis factor-alpha (TNF-alpha) production. Thalidomide 64-75 tumor necrosis factor Mus musculus 127-136 9301478-4 1997 Experiments examining thalidomide"s enantiomers reveal-that the S(-)-enantiomer has the strongest antiangiogenic activity in VEGF-induced and bFGF-induced corneal neovascularization. Thalidomide 22-33 vascular endothelial growth factor A Mus musculus 125-129 9301478-4 1997 Experiments examining thalidomide"s enantiomers reveal-that the S(-)-enantiomer has the strongest antiangiogenic activity in VEGF-induced and bFGF-induced corneal neovascularization. Thalidomide 22-33 fibroblast growth factor 2 Mus musculus 142-146 9142912-10 1997 Inhibition of TNF-alpha synthesis/release with pentoxifylline, thalidomide, dexamethasone, or immunoneutralization of TNF-alpha (with anti-TNF-alpha) was found to significantly reduce the severity of ethanol-induced gastric mucosal damage in cirrhotic rats. Thalidomide 63-74 tumor necrosis factor Rattus norvegicus 14-23 9456687-8 1997 Thalidomide, which inhibits TNF alpha production, is effective treatment for RAU. Thalidomide 0-11 tumor necrosis factor Homo sapiens 28-37 9154767-12 1997 With thalidomide treatment there were unexpected increases in the plasma concentrations of TNF-alpha and soluble TNF-alpha receptors. Thalidomide 5-16 tumor necrosis factor Homo sapiens 91-100 9154767-12 1997 With thalidomide treatment there were unexpected increases in the plasma concentrations of TNF-alpha and soluble TNF-alpha receptors. Thalidomide 5-16 tumor necrosis factor Homo sapiens 113-122 9439767-1 1997 Thalidomide (Thd) has been shown to have interesting immunosuppressive properties and strong action against TNF-alpha. Thalidomide 0-11 tumor necrosis factor Homo sapiens 108-117 9439767-1 1997 Thalidomide (Thd) has been shown to have interesting immunosuppressive properties and strong action against TNF-alpha. Thalidomide 13-16 tumor necrosis factor Homo sapiens 108-117 9439767-4 1997 At different concentrations, both Thd and H-Thd alone inhibited the lymphocyte proliferation induced by alloantigen (MLR), mitogens (Con A, PWM) and superantigen (SEB) with an activity of 50-75% that of CsA, however, in some tests, immunosuppressive potency of H-Thd was shown to be higher than that of Thd. Thalidomide 34-37 SET binding protein 1 Homo sapiens 163-166 9439767-4 1997 At different concentrations, both Thd and H-Thd alone inhibited the lymphocyte proliferation induced by alloantigen (MLR), mitogens (Con A, PWM) and superantigen (SEB) with an activity of 50-75% that of CsA, however, in some tests, immunosuppressive potency of H-Thd was shown to be higher than that of Thd. Thalidomide 44-47 SET binding protein 1 Homo sapiens 163-166 9099468-3 1997 The effect of dexamethasone was most pronounced on PHA-induced T-cell proliferation and IFN-gamma secretion, whereas the effect of thalidomide was most pronounced on BPM-induced T-cell proliferation and IFN-gamma secretion. Thalidomide 131-142 interferon gamma Rattus norvegicus 203-212 9666962-8 1997 Thalidomide treatment resulted in a significant reduction in tumor necrosis factor-alpha, interleukin 6 (IL-6) and IL-10 protein levels (blood) and mRNA expression (lungs). Thalidomide 0-11 tumor necrosis factor Mus musculus 61-88 9129992-0 1997 Hydrolysis of thalidomide abrogates its ability to enhance mononuclear cell synthesis of IL-2 as well as its ability to suppress the synthesis of TNF-alpha. Thalidomide 14-25 interleukin 2 Homo sapiens 89-93 9129992-0 1997 Hydrolysis of thalidomide abrogates its ability to enhance mononuclear cell synthesis of IL-2 as well as its ability to suppress the synthesis of TNF-alpha. Thalidomide 14-25 tumor necrosis factor Homo sapiens 146-155 9129992-4 1997 Unhydrolyzed thalidomide at 4.0 micrograms/ml consistently enhanced the synthesis of IL-2 in SEA-stimulated cells, and suppressed the synthesis of TNF-alpha in LPS-stimulated cells; whereas, hydrolyzed thalidomide had no enhancing effect on SEA stimulated-cell synthesis of IL-2 or suppressive effect on LPS stimulated-cell synthesis of TNF-alpha. Thalidomide 13-24 interleukin 2 Homo sapiens 85-89 9129992-4 1997 Unhydrolyzed thalidomide at 4.0 micrograms/ml consistently enhanced the synthesis of IL-2 in SEA-stimulated cells, and suppressed the synthesis of TNF-alpha in LPS-stimulated cells; whereas, hydrolyzed thalidomide had no enhancing effect on SEA stimulated-cell synthesis of IL-2 or suppressive effect on LPS stimulated-cell synthesis of TNF-alpha. Thalidomide 13-24 tumor necrosis factor Homo sapiens 147-156 9129992-4 1997 Unhydrolyzed thalidomide at 4.0 micrograms/ml consistently enhanced the synthesis of IL-2 in SEA-stimulated cells, and suppressed the synthesis of TNF-alpha in LPS-stimulated cells; whereas, hydrolyzed thalidomide had no enhancing effect on SEA stimulated-cell synthesis of IL-2 or suppressive effect on LPS stimulated-cell synthesis of TNF-alpha. Thalidomide 13-24 interleukin 2 Homo sapiens 274-278 9129992-4 1997 Unhydrolyzed thalidomide at 4.0 micrograms/ml consistently enhanced the synthesis of IL-2 in SEA-stimulated cells, and suppressed the synthesis of TNF-alpha in LPS-stimulated cells; whereas, hydrolyzed thalidomide had no enhancing effect on SEA stimulated-cell synthesis of IL-2 or suppressive effect on LPS stimulated-cell synthesis of TNF-alpha. Thalidomide 13-24 tumor necrosis factor Homo sapiens 337-346 9129992-5 1997 These findings demonstrate that thalidomide"s ability in vitro to enhance IL-2 and to suppress TNF-alpha in stimulated cells is dependent on the intact molecule and underscore the necessity to employ thalidomide under appropriate physicochemical conditions. Thalidomide 32-43 interleukin 2 Homo sapiens 74-78 9129992-5 1997 These findings demonstrate that thalidomide"s ability in vitro to enhance IL-2 and to suppress TNF-alpha in stimulated cells is dependent on the intact molecule and underscore the necessity to employ thalidomide under appropriate physicochemical conditions. Thalidomide 32-43 tumor necrosis factor Homo sapiens 95-104 9084226-6 1997 Thalidomide is a specific inhibitor of TNF-alpha production, and this effect has been shown to be useful for the treatment of various immunodiseases. Thalidomide 0-11 tumor necrosis factor Homo sapiens 39-48 9084226-8 1997 We also found that in a optically active phthalimide analogues of thalidomide the inducer specific bi-directional regulation of TNF-alpha production is separated. Thalidomide 66-77 tumor necrosis factor Homo sapiens 128-137 9068814-3 1997 This study evaluated the effect of thalidomide on lipopolysaccharide (LPS)-induced TNF-alpha production by human alveolar macrophages obtained from patients with tuberculosis and a group of other diseases associated with macrophage activation. Thalidomide 35-46 tumor necrosis factor Homo sapiens 83-92 9068814-6 1997 Similarly, TNF-alpha mRNA as measured by in situ hybridization was increased following incubation with LPS (P < 0.05), but this increase was prevented by addition of thalidomide (P < 0.05) or dexamethasone (P < 0.05). Thalidomide 169-180 tumor necrosis factor Homo sapiens 11-20 9068814-7 1997 The ability of thalidomide to reduce LPS-induced TNF-alpha production by alveolar macrophages was the same when cells from patients with tuberculosis (a disease associated with TNF-alpha production) and cells from patients with the other conditions were compared. Thalidomide 15-26 tumor necrosis factor Homo sapiens 49-58 9068814-7 1997 The ability of thalidomide to reduce LPS-induced TNF-alpha production by alveolar macrophages was the same when cells from patients with tuberculosis (a disease associated with TNF-alpha production) and cells from patients with the other conditions were compared. Thalidomide 15-26 tumor necrosis factor Homo sapiens 177-186 9068814-8 1997 The ability of thalidomide to reduce TNF-alpha production by human alveolar macrophages from patients with active lung disease suggests that thalidomide and its analogues may have potential as drugs to reduce TNF-alpha production in disease. Thalidomide 15-26 tumor necrosis factor Homo sapiens 37-46 9068814-8 1997 The ability of thalidomide to reduce TNF-alpha production by human alveolar macrophages from patients with active lung disease suggests that thalidomide and its analogues may have potential as drugs to reduce TNF-alpha production in disease. Thalidomide 15-26 tumor necrosis factor Homo sapiens 209-218 9068814-8 1997 The ability of thalidomide to reduce TNF-alpha production by human alveolar macrophages from patients with active lung disease suggests that thalidomide and its analogues may have potential as drugs to reduce TNF-alpha production in disease. Thalidomide 141-152 tumor necrosis factor Homo sapiens 37-46 9068814-8 1997 The ability of thalidomide to reduce TNF-alpha production by human alveolar macrophages from patients with active lung disease suggests that thalidomide and its analogues may have potential as drugs to reduce TNF-alpha production in disease. Thalidomide 141-152 tumor necrosis factor Homo sapiens 209-218 9666962-8 1997 Thalidomide treatment resulted in a significant reduction in tumor necrosis factor-alpha, interleukin 6 (IL-6) and IL-10 protein levels (blood) and mRNA expression (lungs). Thalidomide 0-11 interleukin 6 Mus musculus 90-103 9666962-8 1997 Thalidomide treatment resulted in a significant reduction in tumor necrosis factor-alpha, interleukin 6 (IL-6) and IL-10 protein levels (blood) and mRNA expression (lungs). Thalidomide 0-11 interleukin 6 Mus musculus 105-109 9666962-8 1997 Thalidomide treatment resulted in a significant reduction in tumor necrosis factor-alpha, interleukin 6 (IL-6) and IL-10 protein levels (blood) and mRNA expression (lungs). Thalidomide 0-11 interleukin 10 Mus musculus 115-120 8806653-0 1996 Cell type-/inducer-specific bidirectional regulation by thalidomide and phenylphthalimides of tumor necrosis factor-alpha production and its enantio-dependence. Thalidomide 56-67 tumor necrosis factor Homo sapiens 94-121 8931784-1 1996 OBJECTIVE: To evaluate the efficacy of thalidomide in treating wasting syndrome in patients with advanced HIV disease, and to assess the effects of thalidomide on circulating CD4+ T cells, and on HIV viral burden in peripheral blood mononuclear cells (PBMC). Thalidomide 148-159 CD4 molecule Homo sapiens 175-178 8976641-2 1996 Since pentoxifylline and thalidomide inhibit endotoxin induced TNF production in vitro, these drugs were tested in an open study in rheumatoid arthritis patients to assess toxicity, the effect on TNF production, and the antiarthritic effects. Thalidomide 25-36 tumor necrosis factor Homo sapiens 63-66 8976641-8 1996 CONCLUSIONS: Although pentoxifylline/thalidomide reduced the production capacity of TNF, the benefit/side effects ratio was poor due to multiple adverse effects, while clinical observation suggests limited efficacy. Thalidomide 37-48 tumor necrosis factor Homo sapiens 84-87 8755512-0 1996 Binding of thalidomide to alpha1-acid glycoprotein may be involved in its inhibition of tumor necrosis factor alpha production. Thalidomide 11-22 tumor necrosis factor Homo sapiens 88-115 8862260-12 1996 Thalidomide reduces the frequency of IFN-gamma-induced ENL, but also eliminates the IFN-gamma-induced bacillary clearance. Thalidomide 0-11 interferon gamma Homo sapiens 37-46 8862260-12 1996 Thalidomide reduces the frequency of IFN-gamma-induced ENL, but also eliminates the IFN-gamma-induced bacillary clearance. Thalidomide 0-11 interferon gamma Homo sapiens 84-93 8755512-2 1996 The immunomodulatory activity of thalidomide has been ascribed to the selective inhibition of tumor necrosis factor alpha from monocytes. Thalidomide 33-44 tumor necrosis factor Homo sapiens 94-121 8755512-7 1996 We show that the binding of thalidomide photoaffinity label to authentic human AGP is competed with both thalidomide and the nonradioactive photoaffinity label at concentrations comparable to those required for inhibition of production of tumor necrosis factor alpha from human monocytes, suggesting that AGP may be involved in the immunomodulatory activity of thalidomide. Thalidomide 28-39 tumor necrosis factor Homo sapiens 239-266 8675185-11 1996 Thalidomide inhibits TNF synthesis and reduces NO production, blunts the development of the hyperdynamic circulation, and decreases portal pressure in PVL-operated rats. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 21-24 8827720-0 1996 Selection of novel analogs of thalidomide with enhanced tumor necrosis factor alpha inhibitory activity. Thalidomide 30-41 tumor necrosis factor Homo sapiens 56-83 8827720-2 1996 Recently, thalidomide (alpha-N-phthalimidoglutarimide) was shown to partially inhibit monocyte TNF alpha production (by 50-70%) both in vivo and in vitro. Thalidomide 10-21 tumor necrosis factor Homo sapiens 95-104 8827720-2 1996 Recently, thalidomide (alpha-N-phthalimidoglutarimide) was shown to partially inhibit monocyte TNF alpha production (by 50-70%) both in vivo and in vitro. Thalidomide 23-53 tumor necrosis factor Homo sapiens 95-104 8827720-4 1996 MATERIALS AND METHODS: Three structural analogues of thalidomide were selected for study based on increased activity against TNF alpha production. Thalidomide 53-64 tumor necrosis factor Homo sapiens 125-134 8827720-7 1996 RESULTS: The new compounds (two esters and one amide) showed increased inhibition of TNF alpha production by LPS-stimulated human monocytes, relative to the parent drug thalidomide. Thalidomide 169-180 tumor necrosis factor Homo sapiens 85-94 8827720-10 1996 CONCLUSIONS: Analogs of thalidomide designed to better inhibit TNF alpha production in vitro have correspondingly greater efficacy in vivo. Thalidomide 24-35 tumor necrosis factor Homo sapiens 63-72 8675185-0 1996 Thalidomide inhibits tumor necrosis factor alpha, decreases nitric oxide synthesis, and ameliorates the hyperdynamic circulatory syndrome in portal-hypertensive rats. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 21-48 8675185-3 1996 Thalidomide selectively inhibits TNF production by enhancing messenger RNA degradation. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 33-36 8702405-0 1996 Inducer-specific bidirectional regulation by thalidomide and phenylphthalimides of tumor necrosis factor-alpha production. Thalidomide 45-56 tumor necrosis factor Homo sapiens 83-110 8702405-1 1996 Regulation by thalidomide [N(alpha)-phthalimidoglutarimide] of tumor necrosis factor (TNF)-alpha production was found to be inducer-specific. Thalidomide 14-25 tumor necrosis factor Homo sapiens 63-96 8702405-2 1996 Thalidomide enhances TNF-alpha production by human leukemia HL-60 cells induced with 12-O-tetradecanoylphorbol 13-acetate (TPA), while it inhibits TNF-alpha production induced with okadaic acid (OA) in the same cell line. Thalidomide 0-11 tumor necrosis factor Homo sapiens 21-30 8702405-2 1996 Thalidomide enhances TNF-alpha production by human leukemia HL-60 cells induced with 12-O-tetradecanoylphorbol 13-acetate (TPA), while it inhibits TNF-alpha production induced with okadaic acid (OA) in the same cell line. Thalidomide 0-11 tumor necrosis factor Homo sapiens 147-156 8843588-6 1996 Stimulated lymphocytes secreted significantly more IL-2 in the presence of microsomal-treated thalidomide than did controls. Thalidomide 94-105 interleukin 2 Rattus norvegicus 51-55 8661187-0 1996 Thalidomide inhibits TNF response and increases survival following endotoxin injection in rats. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 21-24 8661187-4 1996 Thalidomide, shown in vitro to selectively inhibit TNF production, has been used clinically in states of chronic TNF elevation with encouraging results. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 51-54 8661187-4 1996 Thalidomide, shown in vitro to selectively inhibit TNF production, has been used clinically in states of chronic TNF elevation with encouraging results. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 113-116 8661187-12 1996 Thalidomide treated rats had lower TNF levels at all time points (P = <0.01 at 90 and 120 min), with the inhibition being dose dependent. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 35-38 8661187-14 1996 In conclusion, we found that thalidomide administration leads to increased survival following acute endotoxemia, which may be due to the observed TNF inhibition. Thalidomide 29-40 tumor necrosis factor Rattus norvegicus 146-149 8843588-13 1996 In untransplanted rats given thalidomide a decrease of CD4 positive T cells was detected on days 3 and 5. Thalidomide 29-40 Cd4 molecule Rattus norvegicus 55-58 8627072-4 1996 MGC formation was blocked by anti-CD14 and anti-CD18 antibodies and by thalidomide, a potent inhibitor of tumor necrosis factor-alpha (TNF-alpha) production my microglia. Thalidomide 71-82 tumor necrosis factor Sus scrofa 135-144 8627072-4 1996 MGC formation was blocked by anti-CD14 and anti-CD18 antibodies and by thalidomide, a potent inhibitor of tumor necrosis factor-alpha (TNF-alpha) production my microglia. Thalidomide 71-82 tumor necrosis factor Sus scrofa 106-133 8603261-2 1996 Thalidomide inhibits the synthesis of TNF-alpha both in vitro and in vivo and may have clinical usefulness. Thalidomide 0-11 tumor necrosis factor Homo sapiens 38-47 8861747-3 1996 THD is capable of changing the density of tumor necrosis factor alpha (TNFalpha) induced ICAM-1 (CD54), VCAM-1 (CD106) and E-selectin antigens on HUVEC. Thalidomide 0-3 tumor necrosis factor Homo sapiens 42-69 8861747-3 1996 THD is capable of changing the density of tumor necrosis factor alpha (TNFalpha) induced ICAM-1 (CD54), VCAM-1 (CD106) and E-selectin antigens on HUVEC. Thalidomide 0-3 tumor necrosis factor Homo sapiens 71-79 8861747-3 1996 THD is capable of changing the density of tumor necrosis factor alpha (TNFalpha) induced ICAM-1 (CD54), VCAM-1 (CD106) and E-selectin antigens on HUVEC. Thalidomide 0-3 intercellular adhesion molecule 1 Homo sapiens 89-95 8728022-1 1996 The present study investigates synergistic effects of the TNF-alpha inhibitor thalidomide and the poly(ADP-ribose) polymerase (PARP)-inhibitor nicotinic acid amide (NAA) in male DBA/1 hybird mice suffering from type II collagen-induced arthritis. Thalidomide 78-89 tumor necrosis factor Mus musculus 58-67 8603261-6 1996 In patients with concomitant HIV-1 and tuberculosis infections, thalidomide therapy was associated with a reduction in both plasma TNF-alpha levels and HIV-1 levels. Thalidomide 64-75 tumor necrosis factor Homo sapiens 131-140 8861747-3 1996 THD is capable of changing the density of tumor necrosis factor alpha (TNFalpha) induced ICAM-1 (CD54), VCAM-1 (CD106) and E-selectin antigens on HUVEC. Thalidomide 0-3 intercellular adhesion molecule 1 Homo sapiens 97-101 8861747-3 1996 THD is capable of changing the density of tumor necrosis factor alpha (TNFalpha) induced ICAM-1 (CD54), VCAM-1 (CD106) and E-selectin antigens on HUVEC. Thalidomide 0-3 vascular cell adhesion molecule 1 Homo sapiens 104-110 8861747-3 1996 THD is capable of changing the density of tumor necrosis factor alpha (TNFalpha) induced ICAM-1 (CD54), VCAM-1 (CD106) and E-selectin antigens on HUVEC. Thalidomide 0-3 vascular cell adhesion molecule 1 Homo sapiens 112-117 8603261-10 1996 The results of this pilot study suggest that thalidomide may have a clinical role in enhancing weight gain and possibly reducing TNF-alpha and HIV-1 levels in patients with HIV-1 and concomitant mycobacterial infections. Thalidomide 45-56 tumor necrosis factor Homo sapiens 129-138 8861747-3 1996 THD is capable of changing the density of tumor necrosis factor alpha (TNFalpha) induced ICAM-1 (CD54), VCAM-1 (CD106) and E-selectin antigens on HUVEC. Thalidomide 0-3 selectin E Homo sapiens 123-133 8683039-2 1996 In cultures of LPS stimulated human mononuclear cells enriched for adherent cells and in cultures of LPS stimulated human monocytes of the cell line-THP-1, thalidomide enhanced the synthesis of TNF-alpha. Thalidomide 156-167 tumor necrosis factor Homo sapiens 194-203 8861747-4 1996 Furthermore, modulation of L-selectin (CD62L) by THD can be demonstrated on human leukocytes in vitro. Thalidomide 49-52 selectin L Homo sapiens 27-37 8861747-4 1996 Furthermore, modulation of L-selectin (CD62L) by THD can be demonstrated on human leukocytes in vitro. Thalidomide 49-52 selectin L Homo sapiens 39-44 8683039-3 1996 When cultures of unfractionated peripheral blood mononuclear cells were stimulated with LPS, thalidomide decreased the synthesis of TNF-alpha. Thalidomide 93-104 tumor necrosis factor Homo sapiens 132-141 8683039-0 1996 Thalidomide can be either agonistic or antagonistic to LPS evoked synthesis of TNF-alpha by mononuclear cells. Thalidomide 0-11 tumor necrosis factor Homo sapiens 79-88 8683039-1 1996 The effect if thalidomide on tumor necrosis factor alpha (TNF-alpha) produced in vitro by lipopolysaccharide (LPS) stimulated human cells was investigated. Thalidomide 14-25 tumor necrosis factor Homo sapiens 29-56 8683039-4 1996 Depending on the type of cells stimulated with LPS in vitro, thalidomide, at concentrations achieved in vivo, can either enhance or suppress the synthesis of TNF-alpha. Thalidomide 61-72 tumor necrosis factor Homo sapiens 158-167 8683039-1 1996 The effect if thalidomide on tumor necrosis factor alpha (TNF-alpha) produced in vitro by lipopolysaccharide (LPS) stimulated human cells was investigated. Thalidomide 14-25 tumor necrosis factor Homo sapiens 58-67 8538367-3 1996 Treatment with low doses (20 or as little as 1 mg/kg body weight) of a highly teratogenic derivative (EM12) of thalidomide, the most notorious human teratogen, triggers a dramatic and statistically highly significant down-regulation of several surface adhesion receptors (e.g. CD11a/CD18, CD49d/CD29, CD61, etc.) Thalidomide 111-122 integrin subunit beta 1 Felis catus 295-299 8900028-0 1996 Enantioselective inhibition of TNF-alpha release by thalidomide and thalidomide-analogues. Thalidomide 52-63 tumor necrosis factor Homo sapiens 31-40 8900028-0 1996 Enantioselective inhibition of TNF-alpha release by thalidomide and thalidomide-analogues. Thalidomide 68-79 tumor necrosis factor Homo sapiens 31-40 8900028-2 1996 The effect of the pure enantiomers of the thalidomide-analogues and also of the enantiomers of thalidomide on release of TNF-alpha was tested in vitro by using stimulated peripheral mononuclear blood cells. Thalidomide 95-106 tumor necrosis factor Homo sapiens 121-130 8900028-3 1996 Both enantiomers of thalidomide inhibited the release of TNF-alpha equally well at low concentrations (5 and 12.5 micrograms/ml) but at higher concentrations (25 and 50 micrograms 50 micrograms/ml) there was a weak but statistically significant selectivity towards the (-)-(S)-enantiomer. Thalidomide 20-31 tumor necrosis factor Homo sapiens 57-66 8900028-5 1996 The (S)-enantiomers of the thalidomide-analogues differed in their inhibitory potency from (-)-(S)-thalidomide suggesting that the introduction of the methyl-group increases the TNF-alpha-inhibitory activity while the reduction of one of the carbonyl-functions in the glutarimide-moiety to a methylene-group decreases activity. Thalidomide 27-38 tumor necrosis factor Homo sapiens 178-187 8900028-5 1996 The (S)-enantiomers of the thalidomide-analogues differed in their inhibitory potency from (-)-(S)-thalidomide suggesting that the introduction of the methyl-group increases the TNF-alpha-inhibitory activity while the reduction of one of the carbonyl-functions in the glutarimide-moiety to a methylene-group decreases activity. Thalidomide 91-110 tumor necrosis factor Homo sapiens 178-187 7590113-4 1995 A 2.1-fold increase of GOT and a 1.9-fold increase of GPT activities (P < 0.001) were observed in mice treated perorally with 500 mg/kg of AAP plus 150 mg/kg of thalidomide (Thal). Thalidomide 164-175 glutamic pyruvic transaminase, soluble Mus musculus 54-57 8745000-6 1995 ICAM-1 expression was markedly reduced after (+)-thalidomide injection. Thalidomide 45-60 intercellular adhesion molecule 1 Mus musculus 0-6 8745000-7 1995 Thalidomide and dexamethasone pretreatment reduced Mac-1 expression on perivascular infiltrated granulocytes. Thalidomide 0-11 integrin alpha M Mus musculus 51-56 8655287-0 1995 Thalidomide increases the synthesis of IL-2 in cultures of human mononuclear cells stimulated with Concanavalin-A, Staphylococcal enterotoxin A, and purified protein derivative. Thalidomide 0-11 interleukin 2 Homo sapiens 39-43 8655287-1 1995 Thalidomide significantly increases the quantity of extracellular IL-2 in cultures of human mononuclear cells stimulated with mitogens or antigen. Thalidomide 0-11 interleukin 2 Homo sapiens 66-70 8655287-2 1995 Cells from 7 donors exposed for 2 h to 4.0 micrograms/ml of thalidomide and stimulated for 16-18 h with 20 micrograms/ml of Concanavalin-A (Con-A) averaged producing 187 +/- 49% more IL-2 than cells stimulated with Con-A alone. Thalidomide 60-71 interleukin 2 Homo sapiens 183-187 8655287-3 1995 In similar experimental procedures and comparisons the pg/ml of IL-2 secreted by thalidomide-treated cells from five donors stimulated with 50 ng/ml of Staphylococcal enterotoxin A (SEA) increased by 159 +/- 32%, and the pg/ml of IL-2 secreted by thalidomide-treated cells from 2 donors stimulated with 5.0 micrograms/ml of purified protein derivative of Mycobacterium tuberculosis increased by 120 +/- 4%. Thalidomide 81-92 interleukin 2 Homo sapiens 64-68 8655287-3 1995 In similar experimental procedures and comparisons the pg/ml of IL-2 secreted by thalidomide-treated cells from five donors stimulated with 50 ng/ml of Staphylococcal enterotoxin A (SEA) increased by 159 +/- 32%, and the pg/ml of IL-2 secreted by thalidomide-treated cells from 2 donors stimulated with 5.0 micrograms/ml of purified protein derivative of Mycobacterium tuberculosis increased by 120 +/- 4%. Thalidomide 81-92 interleukin 2 Homo sapiens 230-234 8655287-3 1995 In similar experimental procedures and comparisons the pg/ml of IL-2 secreted by thalidomide-treated cells from five donors stimulated with 50 ng/ml of Staphylococcal enterotoxin A (SEA) increased by 159 +/- 32%, and the pg/ml of IL-2 secreted by thalidomide-treated cells from 2 donors stimulated with 5.0 micrograms/ml of purified protein derivative of Mycobacterium tuberculosis increased by 120 +/- 4%. Thalidomide 247-258 interleukin 2 Homo sapiens 64-68 8655287-4 1995 Thalidomide also significantly increases the quantity of intracellular IL-2 in cells stimulated with mitogens. Thalidomide 0-11 interleukin 2 Homo sapiens 71-75 8655287-5 1995 Cells exposed to thalidomide and stimulated with Con-A had an increase in intracellular IL-2 of 130% after 8 h and 157% after 12 h in culture; cells stimulated with SEA had an increase in intracellular IL-2 of 120% after 8 h and 182% after 12 h in culture. Thalidomide 17-28 interleukin 2 Homo sapiens 88-92 8655287-5 1995 Cells exposed to thalidomide and stimulated with Con-A had an increase in intracellular IL-2 of 130% after 8 h and 157% after 12 h in culture; cells stimulated with SEA had an increase in intracellular IL-2 of 120% after 8 h and 182% after 12 h in culture. Thalidomide 17-28 interleukin 2 Homo sapiens 202-206 8634679-3 1995 In the present study, we investigated the effect of thalidomide (alpha-N-pthalimidoglutarimide), a selective inhibitor of TNF-alpha synthesis, on granuloma formation during BCG infection in Oncins France 1 (OF-1) mice. Thalidomide 52-63 tumor necrosis factor Mus musculus 122-131 8634679-6 1995 Serum TNF-alpha levels of thalidomide-treated mice were significantly lower (85%) than control mice on day 14 and remained lower (55%) on days 21 and 28. Thalidomide 26-37 tumor necrosis factor Mus musculus 6-15 8634679-8 1995 Daily administration of thalidomide for 21 to 28 days to the BCG-infected mice inhibited local TNF-alpha expression in well-developed granulomas. Thalidomide 24-35 tumor necrosis factor Mus musculus 95-104 7590113-7 1995 The Thal-induced exacerbation of AAP hepatotoxicity was completely inhibited by nicotinic acid amide, a selective inhibitor of poly(ADP-ribose) polymerase (PARP) (P < 0.0001), suggesting a possible influence of Thal on the hepatic metabolism of NAD-adenoribosylation. Thalidomide 4-8 poly (ADP-ribose) polymerase family, member 1 Mus musculus 127-154 7590113-7 1995 The Thal-induced exacerbation of AAP hepatotoxicity was completely inhibited by nicotinic acid amide, a selective inhibitor of poly(ADP-ribose) polymerase (PARP) (P < 0.0001), suggesting a possible influence of Thal on the hepatic metabolism of NAD-adenoribosylation. Thalidomide 4-8 poly (ADP-ribose) polymerase family, member 1 Mus musculus 156-160 7590113-7 1995 The Thal-induced exacerbation of AAP hepatotoxicity was completely inhibited by nicotinic acid amide, a selective inhibitor of poly(ADP-ribose) polymerase (PARP) (P < 0.0001), suggesting a possible influence of Thal on the hepatic metabolism of NAD-adenoribosylation. Thalidomide 214-218 poly (ADP-ribose) polymerase family, member 1 Mus musculus 127-154 7590113-7 1995 The Thal-induced exacerbation of AAP hepatotoxicity was completely inhibited by nicotinic acid amide, a selective inhibitor of poly(ADP-ribose) polymerase (PARP) (P < 0.0001), suggesting a possible influence of Thal on the hepatic metabolism of NAD-adenoribosylation. Thalidomide 214-218 poly (ADP-ribose) polymerase family, member 1 Mus musculus 156-160 8825912-2 1995 Since thalidomide has been shown to inhibit TNF-alpha production, experiments were carried out to determine whether the drug can modulate the inflammatory response to either lysates of H. influenzae (gram negative) or heat killed S. pneumoniae (gram positive) in rabbits. Thalidomide 6-17 tumor necrosis factor Oryctolagus cuniculus 44-53 7561198-0 1995 Thalidomide inhibits tumor necrosis factor-alpha production by lipopolysaccharide- and lipoarabinomannan-stimulated human microglial cells. Thalidomide 0-11 tumor necrosis factor Homo sapiens 21-48 7561198-2 1995 The effect of thalidomide on TNF-alpha production by microglia, the resident macrophages of the brain, was evaluated. Thalidomide 14-25 tumor necrosis factor Homo sapiens 29-38 7561198-3 1995 In primary human fetal microglial cell cultures stimulated with lipopolysaccharide or lipoarabinomannan, thalidomide inhibited TNF-alpha release in a dose-dependent manner. Thalidomide 105-116 tumor necrosis factor Homo sapiens 127-136 7561198-4 1995 The inhibitory effect of thalidomide was similar to that of dexamethasone, although expression of TNF-alpha mRNA in microglial cells was reduced only by thalidomide. Thalidomide 153-164 tumor necrosis factor Homo sapiens 98-107 8825912-5 1995 Thalidomide treatment reduces TNF-alpha production in both experimental systems, but has a greater effect on the more indolent gram positive inflammatory response in which peak TNF-alpha levels in the CSF are reduced by > 50%. Thalidomide 0-11 tumor necrosis factor Oryctolagus cuniculus 30-39 8825912-5 1995 Thalidomide treatment reduces TNF-alpha production in both experimental systems, but has a greater effect on the more indolent gram positive inflammatory response in which peak TNF-alpha levels in the CSF are reduced by > 50%. Thalidomide 0-11 tumor necrosis factor Oryctolagus cuniculus 177-186 8825912-7 1995 In meningeal inflammation induced by the Gram negative lysate, treatment with thalidomide results in only a 29% inhibition of TNF-alpha release into the CSF. Thalidomide 78-89 tumor necrosis factor Oryctolagus cuniculus 126-135 8845811-0 1995 Benzylphthalimides and phenethylphthalimides with thalidomide-like activity on the production of tumor necrosis factor alpha. Thalidomide 50-61 tumor necrosis factor Homo sapiens 97-124 8845811-1 1995 Benzylphthalimide analogs (P1P"s) and phenethylphthalimide analogs (P2P"s) have been found to exhibit thalidomide-like activity on the production of tumor necrosis factor (TNF)-alpha by the human leukemia cell line, HL-60, stimulated by 12-O-tetradecanoylphorbol-13-acetate (TPA). Thalidomide 102-113 tumor necrosis factor Homo sapiens 149-182 7558825-7 1995 Because thalidomide has been shown to inhibit TNF-alpha production, down-modulation of this cytokine seems to be a useful treatment strategy in Langerhans" cell granulomatosis. Thalidomide 8-19 tumor necrosis factor Homo sapiens 46-55 7635184-0 1995 Does thalidomide affect IL-2 response and production? Thalidomide 5-16 interleukin 2 Homo sapiens 24-28 7635184-2 1995 A common denominator in the pathogenesis of graft-vs.-host disease, graft rejection, reactional lepromatous leprosy, and autoimmune disorders modulated by thalidomide is the activation of T lymphocytes culminating in the synthesis of interleukin-2 (IL-2), the expression of high-affinity IL-2 receptors, and the induction of proliferation. Thalidomide 155-166 interleukin 2 Homo sapiens 234-247 7635184-2 1995 A common denominator in the pathogenesis of graft-vs.-host disease, graft rejection, reactional lepromatous leprosy, and autoimmune disorders modulated by thalidomide is the activation of T lymphocytes culminating in the synthesis of interleukin-2 (IL-2), the expression of high-affinity IL-2 receptors, and the induction of proliferation. Thalidomide 155-166 interleukin 2 Homo sapiens 249-253 7635184-2 1995 A common denominator in the pathogenesis of graft-vs.-host disease, graft rejection, reactional lepromatous leprosy, and autoimmune disorders modulated by thalidomide is the activation of T lymphocytes culminating in the synthesis of interleukin-2 (IL-2), the expression of high-affinity IL-2 receptors, and the induction of proliferation. Thalidomide 155-166 interleukin 2 Homo sapiens 288-292 7635184-3 1995 We investigated the effect of thalidomide on the production of IL-2 by the human leukemia cell line Jurkat through induction of IL-2 gene enhancer activity and through the presence of IL-2 in supernatants. Thalidomide 30-41 interleukin 2 Homo sapiens 63-67 7635184-4 1995 beta-galactosidase activity, encoded by a reporter lac z construct and controlled by a transcription factor in thalidomide-treated PMA- and ionomycin-stimulated Jurkat cells, was similar (97 +/- 1.33%; p > 0.1) to non-thalidomide-treated controls at all drug concentrations tested. Thalidomide 111-122 galactosidase beta 1 Homo sapiens 0-18 7635184-5 1995 IL-2 enhancer-driven beta-galactose activity of thalidomide-treated and stimulated cells was also similar to that of untreated controls (p > 0.2). Thalidomide 48-59 interleukin 2 Homo sapiens 0-4 7640215-0 1995 Effect of thalidomide on tumour necrosis factor production and anti-tumour activity induced by 5,6-dimethylxanthenone-4-acetic acid. Thalidomide 10-21 tumor necrosis factor Mus musculus 25-47 7640215-4 1995 Thalidomide at doses of between 8 and 250 mg kg-1 efficiently suppressed serum TNF activity in response to 5,6-MeXAA at its optimal TNF inducing dose of 55 mg kg-1. Thalidomide 0-11 tumor necrosis factor Mus musculus 79-82 7640215-4 1995 Thalidomide at doses of between 8 and 250 mg kg-1 efficiently suppressed serum TNF activity in response to 5,6-MeXAA at its optimal TNF inducing dose of 55 mg kg-1. Thalidomide 0-11 tumor necrosis factor Mus musculus 132-135 8527011-3 1995 Recent research has shown a decrease in tumour necrosis factor-alpha (TNF alpha) during thalidomide treatment in several settings. Thalidomide 88-99 tumor necrosis factor Homo sapiens 70-79 7593465-4 1995 Inhibitors of TNF synthesis, pentoxifylline (PTX) and thalidomide, inhibited TNF mRNA accumulation in LPS-activated monocytes and down-regulated DR mRNA but not DP or DQ mRNA. Thalidomide 54-65 tumor necrosis factor Homo sapiens 77-80 7742424-9 1995 The mechanism of the effect of thalidomide is uncertain, although recent studies have suggested that thalidomide selectively inhibits the production of tumor necrosis factor alpha. Thalidomide 101-112 tumor necrosis factor Homo sapiens 152-179 8521296-0 1995 Thalidomide treatment reduces tumor necrosis factor alpha production and enhances weight gain in patients with pulmonary tuberculosis. Thalidomide 0-11 tumor necrosis factor Homo sapiens 30-57 8521296-3 1995 Thalidomide (alpha-N-phthalimidoglutarimide) has recently been shown to suppress TNF alpha production by human monocytes in vitro and to reduce serum TNF alpha in leprosy patients. Thalidomide 0-11 tumor necrosis factor Homo sapiens 81-90 8521296-3 1995 Thalidomide (alpha-N-phthalimidoglutarimide) has recently been shown to suppress TNF alpha production by human monocytes in vitro and to reduce serum TNF alpha in leprosy patients. Thalidomide 0-11 tumor necrosis factor Homo sapiens 150-159 8521296-3 1995 Thalidomide (alpha-N-phthalimidoglutarimide) has recently been shown to suppress TNF alpha production by human monocytes in vitro and to reduce serum TNF alpha in leprosy patients. Thalidomide 13-43 tumor necrosis factor Homo sapiens 81-90 8521296-3 1995 Thalidomide (alpha-N-phthalimidoglutarimide) has recently been shown to suppress TNF alpha production by human monocytes in vitro and to reduce serum TNF alpha in leprosy patients. Thalidomide 13-43 tumor necrosis factor Homo sapiens 150-159 8521296-4 1995 We have therefore conducted a two-part placebo-controlled pilot study of thalidomide in patients with active tuberculosis to determine its effects on clinical response, immune reactivity, TNF alpha levels, and weight. Thalidomide 73-84 tumor necrosis factor Homo sapiens 188-197 8521296-9 1995 TNF alpha production was significantly reduced during thalidomide treatment while interferon-gamma (IFN gamma) production was enhanced. Thalidomide 54-65 tumor necrosis factor Homo sapiens 0-9 8521296-12 1995 Thalidomide treatment reduces TNF alpha production both in vivo and in vitro and is associated with an accelerated weight gain during the study period. Thalidomide 0-11 tumor necrosis factor Homo sapiens 30-39 7851006-0 1995 The immunosuppressive drug thalidomide induces T helper cell type 2 (Th2) and concomitantly inhibits Th1 cytokine production in mitogen- and antigen-stimulated human peripheral blood mononuclear cell cultures. Thalidomide 27-38 negative elongation factor complex member C/D Homo sapiens 101-104 7851006-3 1995 In a comparative study with the immunosuppressant cyclosporin A, we have demonstrated a potent and specific effect of thalidomide on cytokine production relating to the distinct Th1 and Th2 subsets. Thalidomide 118-129 negative elongation factor complex member C/D Homo sapiens 178-181 7851006-6 1995 Thalidomide switched this response from a Th1 to a Th2 type. Thalidomide 0-11 negative elongation factor complex member C/D Homo sapiens 42-45 7851006-7 1995 The effect was most pronounced at 1000 ng/ml thalidomide, where inhibition of IFN-gamma and enhancement of IL-4 production was maximal. Thalidomide 45-56 interferon gamma Homo sapiens 78-87 7851006-7 1995 The effect was most pronounced at 1000 ng/ml thalidomide, where inhibition of IFN-gamma and enhancement of IL-4 production was maximal. Thalidomide 45-56 interleukin 4 Homo sapiens 107-111 7844376-2 1995 Pentoxifylline, chlorpromazine, and thalidomide inhibit TNF-alpha production. Thalidomide 36-47 tumor necrosis factor Mus musculus 56-65 7851006-8 1995 In unstimulated cultures thalidomide alone induced IL-4 production. Thalidomide 25-36 interleukin 4 Homo sapiens 51-55 7851006-10 1995 Time course data from thalidomide-treated cultures revealed that the augmented IL-4 production diminished as the culture time increased, whereas IFN-gamma production was significantly increased. Thalidomide 22-33 interleukin 4 Homo sapiens 79-83 7851006-10 1995 Time course data from thalidomide-treated cultures revealed that the augmented IL-4 production diminished as the culture time increased, whereas IFN-gamma production was significantly increased. Thalidomide 22-33 interferon gamma Homo sapiens 145-154 7895307-3 1995 Among the investigated compounds, including thalidomide and methylthalidomide, N-adamantylphthalimide showed the most potent TNF-alpha production-enhancing activity. Thalidomide 44-55 tumor necrosis factor Homo sapiens 125-134 7851006-12 1995 The effects of thalidomide and related compounds may enhance our understanding of the mechanisms of T helper cell selection, offer the possibility of controlled therapeutic switching between Th1 and Th2 responses, and may lead to a rational approach for the treatment of some T cell-mediated immunological disorders. Thalidomide 15-26 negative elongation factor complex member C/D Homo sapiens 191-194 22514378-4 1995 Thalidomide, a drug used to treat reactional states in Hansen"s disease, has been reported to enhance degradation of tnf-alpha mrna. Thalidomide 0-11 tumor necrosis factor Homo sapiens 117-126 7830502-3 1995 Thalidomide (Thd) is capable of down-regulating the CD26 receptor on CD4+ lymphocytes after treatment of healthy volunteers. Thalidomide 0-11 dipeptidyl peptidase 4 Homo sapiens 52-56 7830502-3 1995 Thalidomide (Thd) is capable of down-regulating the CD26 receptor on CD4+ lymphocytes after treatment of healthy volunteers. Thalidomide 0-11 CD4 molecule Homo sapiens 69-72 7830502-3 1995 Thalidomide (Thd) is capable of down-regulating the CD26 receptor on CD4+ lymphocytes after treatment of healthy volunteers. Thalidomide 13-16 dipeptidyl peptidase 4 Homo sapiens 52-56 7830502-3 1995 Thalidomide (Thd) is capable of down-regulating the CD26 receptor on CD4+ lymphocytes after treatment of healthy volunteers. Thalidomide 13-16 CD4 molecule Homo sapiens 69-72 7516993-7 1994 After the first two doses of 7 mg Thd/kg body wt the CD18 (the common beta-chain of the beta 2-integrins) marker already decreased in surface density or was no longer detectable on granulocytes, monocytes and lymphocytes. Thalidomide 34-37 integrin subunit beta 2 Homo sapiens 53-57 7713571-6 1994 Both the toxic symptoms and the high levels of TNF-alpha production could be inhibited by thalidomide treatment, a drug we have shown reduces the half life of TNF-alpha mRNA. Thalidomide 90-101 tumor necrosis factor Homo sapiens 47-56 7713571-6 1994 Both the toxic symptoms and the high levels of TNF-alpha production could be inhibited by thalidomide treatment, a drug we have shown reduces the half life of TNF-alpha mRNA. Thalidomide 90-101 tumor necrosis factor Homo sapiens 159-168 7713571-7 1994 In preliminary clinical trials in tuberculosis patients we have attempted to use thalidomide to reduce TNF-alpha production and toxicities. Thalidomide 81-92 tumor necrosis factor Homo sapiens 103-112 8069968-1 1994 The rate of racemization of N(alpha)-phthalimidoglutarimide (thalidomide) was determined as its half life to be 566 min at pH 7.4/37 degrees C. This fast racemization of thalidomide resulted in no apparent difference between (S)- and (R)-forms of the compound on enhancing activity of phorbol ester-induced tumor necrosis factor (TNF)-alpha production by human leukemia HL-60 cells. Thalidomide 61-72 tumor necrosis factor Homo sapiens 307-340 8069968-1 1994 The rate of racemization of N(alpha)-phthalimidoglutarimide (thalidomide) was determined as its half life to be 566 min at pH 7.4/37 degrees C. This fast racemization of thalidomide resulted in no apparent difference between (S)- and (R)-forms of the compound on enhancing activity of phorbol ester-induced tumor necrosis factor (TNF)-alpha production by human leukemia HL-60 cells. Thalidomide 170-181 tumor necrosis factor Homo sapiens 307-340 8069968-3 1994 Only (S)-form of methylthalidomide, but not its (R)-form, elicited TNF-alpha production-enhancing effect, suggesting that the (S)-isomer of thalidomide would be the active form in terms of thalidomidal biological response modifying effects. Thalidomide 23-34 tumor necrosis factor Homo sapiens 67-76 8090535-3 1994 Thalidomide reduces polyclonal hypergammaglobulinemia, that is associated with a clinical and laboratorial improvement, in a dose dependent manner as well as TNF-alpha levels. Thalidomide 0-11 tumor necrosis factor Homo sapiens 158-167 8135786-0 1994 Enhancement of phorbol ester-induced production of tumor necrosis factor alpha by thalidomide. Thalidomide 82-93 tumor necrosis factor Homo sapiens 51-78 8135786-1 1994 The effect of thalidomide [racemic (DL-) form and optically pure (D- and L-) forms] on tumor necrosis factor (TNF) alpha production by human leukemia cell lines (HL-60, K562 and U937) stimulated with 12-O-tetradecanoylphorbol-13-acetate (TPA) was investigated. Thalidomide 14-25 tumor necrosis factor Homo sapiens 87-120 8135786-2 1994 Though thalidomide has been regarded as a specific inhibitor of TNF-alpha production, our study indicated that all forms of thalidomide enhanced (but did not inhibit) the TPA-induced TNF-alpha production by the human leukemia cell lines investigated. Thalidomide 124-135 tumor necrosis factor Homo sapiens 183-192 8135786-3 1994 The effects of thalidomide on TNF-alpha production might be cell type-specific. Thalidomide 15-26 tumor necrosis factor Homo sapiens 30-39 7516993-7 1994 After the first two doses of 7 mg Thd/kg body wt the CD18 (the common beta-chain of the beta 2-integrins) marker already decreased in surface density or was no longer detectable on granulocytes, monocytes and lymphocytes. Thalidomide 34-37 tubulin beta 4B class IVb Homo sapiens 88-94 7516993-12 1994 Other markers, such as the alpha chains of the beta 1 integrins CD49b (VLA alpha 2) and CD49d (VLA alpha 4) showed contrasting reactions to the Thd-treatment. Thalidomide 144-147 UDP-GlcNAc:betaGal beta-1,3-N-acetylglucosaminyltransferase 2 Homo sapiens 47-53 7516993-12 1994 Other markers, such as the alpha chains of the beta 1 integrins CD49b (VLA alpha 2) and CD49d (VLA alpha 4) showed contrasting reactions to the Thd-treatment. Thalidomide 144-147 integrin subunit alpha 2 Homo sapiens 64-69 7516993-12 1994 Other markers, such as the alpha chains of the beta 1 integrins CD49b (VLA alpha 2) and CD49d (VLA alpha 4) showed contrasting reactions to the Thd-treatment. Thalidomide 144-147 integrin subunit alpha 4 Homo sapiens 88-93 8327469-1 1993 Thalidomide, a selective inhibitor of tumor necrosis factor alpha (TNF-alpha) synthesis, suppresses the activation of latent human immunodeficiency virus type 1 (HIV-1) in a monocytoid (U1) line. Thalidomide 0-11 tumor necrosis factor Homo sapiens 67-76 8335978-3 1993 Patients with systemic ENL demonstrated the highest serum TNF alpha levels, which decreased significantly during thalidomide treatment. Thalidomide 113-124 tumor necrosis factor Homo sapiens 58-67 8335978-6 1993 Thalidomide therapy reduced not only serum TNF alpha levels and the clinical symptoms but also the dermal infiltration of polymorphonuclear leukocytes and T cells. Thalidomide 0-11 tumor necrosis factor Homo sapiens 43-52 8335978-8 1993 These results indicate that the thalidomide-induced alleviation of clinical symptoms of ENL was associated with a reduction of TNF alpha levels. Thalidomide 32-43 tumor necrosis factor Homo sapiens 127-136 8177242-0 1993 Inhibition of tumor necrosis factor-alpha by thalidomide in magnesium deficiency. Thalidomide 45-56 tumor necrosis factor Rattus norvegicus 14-41 8177242-3 1993 Thalidomide (1 mg/day) caused a complete inhibition of the increase in circulating tumor necrosis factor-alpha levels, without having an effect on interleukin 1. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 83-110 8327469-4 1993 Thalidomide inhibition of virus replication in the phorbol myristate acetate- and recombinant TNF-alpha-stimulated T-cell line ACH-2 is not observed. Thalidomide 0-11 tumor necrosis factor Homo sapiens 94-103 8327469-4 1993 Thalidomide inhibition of virus replication in the phorbol myristate acetate- and recombinant TNF-alpha-stimulated T-cell line ACH-2 is not observed. Thalidomide 0-11 acyl-CoA thioesterase 1 Homo sapiens 127-132 8327469-6 1993 These results suggest the use of thalidomide in a clinical setting to inhibit both virus replication and the TNF-alpha-induced systemic toxicity of HIV-1 and opportunistic infections. Thalidomide 33-44 tumor necrosis factor Homo sapiens 109-118 8452474-11 1993 (c) The most significant effect, reduction in the reactivity of CD2+, was detectable subsequent to daily oral doses as low as 10 mg Thd/kg or 1 mg EM12/kg bw. Thalidomide 132-135 T-cell surface antigen CD2 Callithrix jacchus 64-67 8496685-0 1993 Thalidomide exerts its inhibitory action on tumor necrosis factor alpha by enhancing mRNA degradation. Thalidomide 0-11 tumor necrosis factor Homo sapiens 44-71 8496685-1 1993 We have examined the mechanism of thalidomide inhibition of lipopolysaccharide (LPS)-induced tumor necrosis factor alpha (TNF-alpha) production and found that the drug enhances the degradation of TNF-alpha mRNA. Thalidomide 34-45 tumor necrosis factor Homo sapiens 93-120 8496685-1 1993 We have examined the mechanism of thalidomide inhibition of lipopolysaccharide (LPS)-induced tumor necrosis factor alpha (TNF-alpha) production and found that the drug enhances the degradation of TNF-alpha mRNA. Thalidomide 34-45 tumor necrosis factor Homo sapiens 122-131 8496685-1 1993 We have examined the mechanism of thalidomide inhibition of lipopolysaccharide (LPS)-induced tumor necrosis factor alpha (TNF-alpha) production and found that the drug enhances the degradation of TNF-alpha mRNA. Thalidomide 34-45 tumor necrosis factor Homo sapiens 196-205 8496685-6 1993 The selective inhibition of TNF-alpha production makes thalidomide an ideal candidate for the treatment of inflammatory conditions where TNF-alpha-induced toxicities are observed and where immunity must remain intact. Thalidomide 55-66 tumor necrosis factor Homo sapiens 28-37 8496685-6 1993 The selective inhibition of TNF-alpha production makes thalidomide an ideal candidate for the treatment of inflammatory conditions where TNF-alpha-induced toxicities are observed and where immunity must remain intact. Thalidomide 55-66 tumor necrosis factor Homo sapiens 137-146 8433015-8 1993 Cytokine treatment induced large amounts of tumor necrosis factor-alpha, which is toxic in this context but can be selectively down-regulated by thalidomide without interfering with other monocyte cytokines necessary for normal immune function. Thalidomide 145-156 tumor necrosis factor Homo sapiens 44-71 8452474-16 1993 (f) A rough estimate of the relative potency to reduce the CD2 receptor in the marmoset suggests EM12 to be five to ten times more potent than Thd. Thalidomide 143-146 T-cell surface antigen CD2 Callithrix jacchus 59-62 1466907-1 1992 Four agents, thalidomide, oxpentifylline, dexamethasone and a polyclonal anti-TNF-alpha antibody, were all shown by specific Elisa to block endogenous TNF-alpha production by Bacillus Calmette Guerin (BCG)-infected human monocyte-derived macrophages in in vitro culture. Thalidomide 13-24 tumor necrosis factor Homo sapiens 151-160 1588290-9 1992 Control of toxic symptoms with thalidomide was associated with a 50-80% reduction in agonist-stimulated monocyte TNF-alpha secretion. Thalidomide 31-42 tumor necrosis factor Homo sapiens 113-122 1588290-10 1992 IFN-gamma enhanced the monocyte release of TNF-alpha by 3-7.5-fold (agonist dependent) when added to patient"s cells in vitro, and this could be suppressed by the in vitro addition of 10 micrograms/ml of thalidomide. Thalidomide 204-215 interferon gamma Homo sapiens 0-9 1588290-10 1992 IFN-gamma enhanced the monocyte release of TNF-alpha by 3-7.5-fold (agonist dependent) when added to patient"s cells in vitro, and this could be suppressed by the in vitro addition of 10 micrograms/ml of thalidomide. Thalidomide 204-215 tumor necrosis factor Homo sapiens 43-52 15508726-3 2004 In the 1990s, the biological activities of thalidomide were determined to include the control of tumor necrosis factor-alpha production and inhibition of angiogenesis. Thalidomide 43-54 tumor necrosis factor Homo sapiens 97-124 1548069-6 1992 Treatment of ENL patients with thalidomide reduced TNF secretion by more than 90%. Thalidomide 31-42 tumor necrosis factor Homo sapiens 51-54 1569817-4 1992 Thalidomide has been shown to cause a decrease in the ratio of CD4+ to CD8+ lymphocytes in the blood of healthy males. Thalidomide 0-11 CD8a molecule Homo sapiens 71-74 2262716-1 1990 A clinical experience of using thalidomide in type-2 lepra reaction (ENL) in 90 male patients--57 with lepromatous leprosy (LL) and 33 with borderline lepromatous leprosy (BL)--is described. Thalidomide 31-42 MLLT1 super elongation complex subunit Homo sapiens 69-72 33773263-0 2021 In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties. Thalidomide 76-87 signal transducer and activator of transcription 3 Homo sapiens 142-147 1569817-0 1992 Thalidomide"s effectiveness in erythema nodosum leprosum is associated with a decrease in CD4+ cells in the peripheral blood. Thalidomide 0-11 CD4 molecule Homo sapiens 90-93 1569817-4 1992 Thalidomide has been shown to cause a decrease in the ratio of CD4+ to CD8+ lymphocytes in the blood of healthy males. Thalidomide 0-11 CD4 molecule Homo sapiens 63-66 1569817-6 1992 In this study, thalidomide"s effectiveness in halting chronic ENL and arresting a relapse into ENL was consistently associated with a decrease in the numbers of CD4+ lymphocytes in the blood of 2 male lepromatous leprosy patients. Thalidomide 15-26 CD4 molecule Homo sapiens 161-164 1997652-0 1991 Thalidomide selectively inhibits tumor necrosis factor alpha production by stimulated human monocytes. Thalidomide 0-11 tumor necrosis factor Homo sapiens 33-60 1997652-1 1991 Thalidomide selectively inhibits the production of human monocyte tumor necrosis factor alpha (TNF-alpha) when these cells are triggered with lipopolysaccharide and other agonists in culture. Thalidomide 0-11 tumor necrosis factor Homo sapiens 66-93 1997652-1 1991 Thalidomide selectively inhibits the production of human monocyte tumor necrosis factor alpha (TNF-alpha) when these cells are triggered with lipopolysaccharide and other agonists in culture. Thalidomide 0-11 tumor necrosis factor Homo sapiens 95-104 2397633-0 1990 Behcet"s disease, palmoplantar pustulosis and HLA-B27 treatment with thalidomide. Thalidomide 69-80 major histocompatibility complex, class I, B Homo sapiens 46-53 33763605-9 2021 Moreover, thalidomide altered the expression of the genes involved in TGF-beta signaling, limb formation, and multiple myeloma, which are related to thalidomide-induced malformations and medication. Thalidomide 10-21 transforming growth factor alpha Homo sapiens 70-78 33763605-9 2021 Moreover, thalidomide altered the expression of the genes involved in TGF-beta signaling, limb formation, and multiple myeloma, which are related to thalidomide-induced malformations and medication. Thalidomide 149-160 transforming growth factor alpha Homo sapiens 70-78 33777938-2 2021 We had previously demonstrated that cereblon (CRBN) is the target of thalidomide embryopathy and acts as a substrate receptor for the E3 ubiquitin ligase complex, Cullin-Ring ligase 4 (CRL4CRBN) in zebrafish and chicks. Thalidomide 69-80 cereblon Danio rerio 46-50 33777938-4 2021 Fetuses exposed to thalidomide in early pregnancy were at risk of neurodevelopmental disorders such as autism, suggesting that CRBN is involved in prenatal brain development. Thalidomide 19-30 cereblon Homo sapiens 127-131 33777938-7 2021 Accumulating evidence shows that CRBN is essential not only for the teratogenic effects but also for the therapeutic effects of thalidomide. Thalidomide 128-139 cereblon Homo sapiens 33-37 33777938-9 2021 Investigation of the molecular mechanisms underlying the therapeutic effects of thalidomide and its derivatives, CRBN E3 ligase modulators (CELMoDs), reveals that these modulators provide CRBN the ability to recognize neosubstrates depending on their structure. Thalidomide 80-91 cereblon Homo sapiens 113-117 33777938-9 2021 Investigation of the molecular mechanisms underlying the therapeutic effects of thalidomide and its derivatives, CRBN E3 ligase modulators (CELMoDs), reveals that these modulators provide CRBN the ability to recognize neosubstrates depending on their structure. Thalidomide 80-91 cereblon Homo sapiens 188-192 26159874-5 2015 The incidence of PM status by phenotype following administration of omeprazole/esomeprazole (known inhibitors of CYP2C19) was 10-fold higher than those who are genetically PMs in the general population, which could have critical clinical implications for personalizing medications primarily metabolized by CYP2C19, such as clopidogrel, PPI, cyclophosphamide, thalidomide, citalopram, clonazepam, diazepam, phenytoin, etc. Thalidomide 359-370 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 113-120 34704527-9 2022 EXPERT OPINION: The chemical space of CRBN ligands which is related to the classical IMiDs (thalidomide/lenalidomide/pomalidomide) is comprehensively covered by the current patent literature. Thalidomide 92-103 cereblon Homo sapiens 38-42 34932796-7 2021 Drinking alcohol and the fT3/fT4 ratio (<= 0.24) were independent risk factors, and thalidomide was an independent protective factor, for NCR in intestinal BS patients treated by IFX. Thalidomide 84-95 Neutrophil migration (granulocyte glycoprotein) Homo sapiens 138-141 34834536-8 2021 As all these chemotypes-thalidomide, IMiDs, CELMoDs and PROTACs-engage CRBN and modify its functions, we describe them here in aggregate as "CRBN-interacting small molecules" (CISMs). Thalidomide 24-35 cereblon Homo sapiens 71-75 34834536-8 2021 As all these chemotypes-thalidomide, IMiDs, CELMoDs and PROTACs-engage CRBN and modify its functions, we describe them here in aggregate as "CRBN-interacting small molecules" (CISMs). Thalidomide 24-35 cereblon Homo sapiens 141-145 34838332-3 2022 Here, we aimed to develop a MYC PROTAC based on the MYC-MAX dimerization inhibitor 10058-F4 derivative 28RH and Thalidomide, called MDEG-541. Thalidomide 112-123 MYC proto-oncogene, bHLH transcription factor Homo sapiens 28-31 34838332-3 2022 Here, we aimed to develop a MYC PROTAC based on the MYC-MAX dimerization inhibitor 10058-F4 derivative 28RH and Thalidomide, called MDEG-541. Thalidomide 112-123 MYC proto-oncogene, bHLH transcription factor Homo sapiens 52-55 34967303-11 2021 In vitro, compared with the HG group, MC apoptosis reduced dramatically with Thd treatment along with upregulation of Bcl-2 and downregulation of Bax. Thalidomide 77-80 B cell leukemia/lymphoma 2 Mus musculus 118-123 34967303-11 2021 In vitro, compared with the HG group, MC apoptosis reduced dramatically with Thd treatment along with upregulation of Bcl-2 and downregulation of Bax. Thalidomide 77-80 BCL2-associated X protein Mus musculus 146-149 34967303-12 2021 At the same time, ER stress markers GRP78, C/EBP homologous protein (CHOP), and Caspase-12 were also mitigated following the Thd treatment. Thalidomide 125-128 heat shock protein 5 Mus musculus 36-41 34967303-12 2021 At the same time, ER stress markers GRP78, C/EBP homologous protein (CHOP), and Caspase-12 were also mitigated following the Thd treatment. Thalidomide 125-128 DNA-damage inducible transcript 3 Mus musculus 43-67 34967303-12 2021 At the same time, ER stress markers GRP78, C/EBP homologous protein (CHOP), and Caspase-12 were also mitigated following the Thd treatment. Thalidomide 125-128 DNA-damage inducible transcript 3 Mus musculus 69-73 34967303-12 2021 At the same time, ER stress markers GRP78, C/EBP homologous protein (CHOP), and Caspase-12 were also mitigated following the Thd treatment. Thalidomide 125-128 caspase 12 Mus musculus 80-90 34967303-13 2021 CONCLUSION: The present study indicates that Thd might reduce the ER stress in DN via downregulating of GRP78, CHOP, and Caspase12 expression, ultimately mitigating MCs apoptosis. Thalidomide 45-48 heat shock protein 5 Mus musculus 104-109 34967303-13 2021 CONCLUSION: The present study indicates that Thd might reduce the ER stress in DN via downregulating of GRP78, CHOP, and Caspase12 expression, ultimately mitigating MCs apoptosis. Thalidomide 45-48 DNA-damage inducible transcript 3 Mus musculus 111-115 34967303-13 2021 CONCLUSION: The present study indicates that Thd might reduce the ER stress in DN via downregulating of GRP78, CHOP, and Caspase12 expression, ultimately mitigating MCs apoptosis. Thalidomide 45-48 caspase 12 Mus musculus 121-130 34944673-0 2021 Thalidomide Exerts Anti-Inflammatory Effects in Cutaneous Lupus by Inhibiting the IRF4/NF-kB and AMPK1/mTOR Pathways. Thalidomide 0-11 interferon regulatory factor 4 Homo sapiens 82-86 34944673-0 2021 Thalidomide Exerts Anti-Inflammatory Effects in Cutaneous Lupus by Inhibiting the IRF4/NF-kB and AMPK1/mTOR Pathways. Thalidomide 0-11 mechanistic target of rapamycin kinase Homo sapiens 103-107 34944673-5 2021 Integrating RNA-seq data, public databases, and literature, TMPS analysis generated mathematical models which predicted that thalidomide acts via two CRBN-CRL4A- (CRL4CRBN) dependent pathways: IRF4/NF-kB and AMPK1/mTOR. Thalidomide 125-136 cereblon Homo sapiens 150-154 34944673-5 2021 Integrating RNA-seq data, public databases, and literature, TMPS analysis generated mathematical models which predicted that thalidomide acts via two CRBN-CRL4A- (CRL4CRBN) dependent pathways: IRF4/NF-kB and AMPK1/mTOR. Thalidomide 125-136 interleukin 17 receptor B Homo sapiens 163-171 34944673-5 2021 Integrating RNA-seq data, public databases, and literature, TMPS analysis generated mathematical models which predicted that thalidomide acts via two CRBN-CRL4A- (CRL4CRBN) dependent pathways: IRF4/NF-kB and AMPK1/mTOR. Thalidomide 125-136 interferon regulatory factor 4 Homo sapiens 193-197 34944673-5 2021 Integrating RNA-seq data, public databases, and literature, TMPS analysis generated mathematical models which predicted that thalidomide acts via two CRBN-CRL4A- (CRL4CRBN) dependent pathways: IRF4/NF-kB and AMPK1/mTOR. Thalidomide 125-136 mechanistic target of rapamycin kinase Homo sapiens 214-218 34944673-7 2021 In vitro experiments confirmed the effect of thalidomide downregulating IRF4 in PBMCs and mTOR in keratinocytes, which converged in an NF-kB reduction that led to a resolution of the inflammatory lesion. Thalidomide 45-56 interferon regulatory factor 4 Homo sapiens 72-76 34944673-7 2021 In vitro experiments confirmed the effect of thalidomide downregulating IRF4 in PBMCs and mTOR in keratinocytes, which converged in an NF-kB reduction that led to a resolution of the inflammatory lesion. Thalidomide 45-56 mechanistic target of rapamycin kinase Homo sapiens 90-94 34764413-2 2021 Cereblon (CRBN) is a common direct target of thalidomide and related compounds and works as a Cullin Ring 4 E3 ubiquitin ligase (CRL4) with DDB1, CUL4, and ROC1. Thalidomide 45-56 cereblon Homo sapiens 10-14 34764413-2 2021 Cereblon (CRBN) is a common direct target of thalidomide and related compounds and works as a Cullin Ring 4 E3 ubiquitin ligase (CRL4) with DDB1, CUL4, and ROC1. Thalidomide 45-56 interleukin 17 receptor B Homo sapiens 129-133 34764413-2 2021 Cereblon (CRBN) is a common direct target of thalidomide and related compounds and works as a Cullin Ring 4 E3 ubiquitin ligase (CRL4) with DDB1, CUL4, and ROC1. Thalidomide 45-56 damage specific DNA binding protein 1 Homo sapiens 140-144 34764413-2 2021 Cereblon (CRBN) is a common direct target of thalidomide and related compounds and works as a Cullin Ring 4 E3 ubiquitin ligase (CRL4) with DDB1, CUL4, and ROC1. Thalidomide 45-56 ring-box 1 Homo sapiens 156-160 34764413-3 2021 The substrate specificity of CRL4CRBN is modulated by thalidomide-related compounds. Thalidomide 54-65 interleukin 17 receptor B Homo sapiens 29-37 34704527-10 2022 The promising area of research is in the identification of non-thalidomide-like chemotypes capable of binding to CRBN. Thalidomide 63-74 cereblon Homo sapiens 113-117 34455300-6 2021 Collectively, a substitution position, an alkyne-azide cyclization and a liker type significantly affect the ability of ANF-thalidomide conjugates in eliminating drug resistance of CYP1B1-expressing DU145 (DU145/CY) cells to docetaxel via targeted CYP1B1 degradation. Thalidomide 124-135 cytochrome P450 family 1 subfamily B member 1 Homo sapiens 181-187 34455300-6 2021 Collectively, a substitution position, an alkyne-azide cyclization and a liker type significantly affect the ability of ANF-thalidomide conjugates in eliminating drug resistance of CYP1B1-expressing DU145 (DU145/CY) cells to docetaxel via targeted CYP1B1 degradation. Thalidomide 124-135 cytochrome P450 family 1 subfamily B member 1 Homo sapiens 248-254 34085173-2 2021 The TNF-alpha inhibitor thalidomide is reported to be effective for inducing remission in pediatric Crohn"s disease (CD) and adults with refractory CD. Thalidomide 24-35 tumor necrosis factor Mus musculus 4-13 34520751-0 2021 Thalidomide suppresses angiogenesis and immune evasion via lncRNA FGD5-AS1/miR-454-3p/ZEB1 axis-mediated VEGFA expression and PD-1/PD-L1 checkpoint in NSCLC. Thalidomide 0-11 FYVE, RhoGEF and PH domain containing 5 Homo sapiens 66-70 34520751-0 2021 Thalidomide suppresses angiogenesis and immune evasion via lncRNA FGD5-AS1/miR-454-3p/ZEB1 axis-mediated VEGFA expression and PD-1/PD-L1 checkpoint in NSCLC. Thalidomide 0-11 prostaglandin D2 receptor Homo sapiens 71-74 34520751-0 2021 Thalidomide suppresses angiogenesis and immune evasion via lncRNA FGD5-AS1/miR-454-3p/ZEB1 axis-mediated VEGFA expression and PD-1/PD-L1 checkpoint in NSCLC. Thalidomide 0-11 microRNA 454 Homo sapiens 75-82 34520751-0 2021 Thalidomide suppresses angiogenesis and immune evasion via lncRNA FGD5-AS1/miR-454-3p/ZEB1 axis-mediated VEGFA expression and PD-1/PD-L1 checkpoint in NSCLC. Thalidomide 0-11 zinc finger E-box binding homeobox 1 Homo sapiens 86-90 34520751-0 2021 Thalidomide suppresses angiogenesis and immune evasion via lncRNA FGD5-AS1/miR-454-3p/ZEB1 axis-mediated VEGFA expression and PD-1/PD-L1 checkpoint in NSCLC. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 105-110 34520751-0 2021 Thalidomide suppresses angiogenesis and immune evasion via lncRNA FGD5-AS1/miR-454-3p/ZEB1 axis-mediated VEGFA expression and PD-1/PD-L1 checkpoint in NSCLC. Thalidomide 0-11 programmed cell death 1 Homo sapiens 126-130 34520751-0 2021 Thalidomide suppresses angiogenesis and immune evasion via lncRNA FGD5-AS1/miR-454-3p/ZEB1 axis-mediated VEGFA expression and PD-1/PD-L1 checkpoint in NSCLC. Thalidomide 0-11 CD274 molecule Homo sapiens 131-136 34520751-11 2021 RESULTS: Thalidomide reduced tumor growth and angiogenesis and increased CD8+ T cell ratio in a mouse model. Thalidomide 9-20 CD8a molecule Homo sapiens 73-76 34520751-14 2021 Thalidomide targeted FGD5-AS1 to exert its anti-tumor activity in NSCLC. Thalidomide 0-11 FYVE, RhoGEF and PH domain containing 5 Homo sapiens 21-25 34520751-14 2021 Thalidomide targeted FGD5-AS1 to exert its anti-tumor activity in NSCLC. Thalidomide 0-11 prostaglandin D2 receptor Homo sapiens 26-29 34520751-16 2021 Simultaneous overexpression of FGD5-AS1 and silencing of miR-454-3p reversed thalidomide-mediated anti-tumor effects in NSCLC. Thalidomide 77-88 FGD5 antisense RNA 1 Homo sapiens 31-39 34520751-16 2021 Simultaneous overexpression of FGD5-AS1 and silencing of miR-454-3p reversed thalidomide-mediated anti-tumor effects in NSCLC. Thalidomide 77-88 microRNA 454 Homo sapiens 57-64 34520751-17 2021 CONCLUSION: Thalidomide inhibits NSCLC angiogenesis and immune evasion via FGD5-AS1/miR-454-3p/ZEB1 axis-mediated regulation of VEGFA expression and PD-1/PD-L1 checkpoint. Thalidomide 12-23 FYVE, RhoGEF and PH domain containing 5 Homo sapiens 75-79 34520751-17 2021 CONCLUSION: Thalidomide inhibits NSCLC angiogenesis and immune evasion via FGD5-AS1/miR-454-3p/ZEB1 axis-mediated regulation of VEGFA expression and PD-1/PD-L1 checkpoint. Thalidomide 12-23 prostaglandin D2 receptor Homo sapiens 80-83 34085173-0 2021 Thalidomide Attenuates Colitis and Is Associated with the Suppression of M1 Macrophage Polarization by Targeting the Transcription Factor IRF5. Thalidomide 0-11 interferon regulatory factor 5 Mus musculus 138-142 34085173-11 2021 Mechanistically, we observed that thalidomide could increase epithelial cell self-renewal capacity and modulate M1/M2 polarization by decreasing M1 markers CD86 and CCR7 and increasing M2 protein signatures CD206 and Arg-1. Thalidomide 34-45 CD86 antigen Mus musculus 156-160 34520751-17 2021 CONCLUSION: Thalidomide inhibits NSCLC angiogenesis and immune evasion via FGD5-AS1/miR-454-3p/ZEB1 axis-mediated regulation of VEGFA expression and PD-1/PD-L1 checkpoint. Thalidomide 12-23 microRNA 454 Homo sapiens 84-91 34085173-11 2021 Mechanistically, we observed that thalidomide could increase epithelial cell self-renewal capacity and modulate M1/M2 polarization by decreasing M1 markers CD86 and CCR7 and increasing M2 protein signatures CD206 and Arg-1. Thalidomide 34-45 chemokine (C-C motif) receptor 7 Mus musculus 165-169 34520751-17 2021 CONCLUSION: Thalidomide inhibits NSCLC angiogenesis and immune evasion via FGD5-AS1/miR-454-3p/ZEB1 axis-mediated regulation of VEGFA expression and PD-1/PD-L1 checkpoint. Thalidomide 12-23 zinc finger E-box binding homeobox 1 Homo sapiens 95-99 34520751-17 2021 CONCLUSION: Thalidomide inhibits NSCLC angiogenesis and immune evasion via FGD5-AS1/miR-454-3p/ZEB1 axis-mediated regulation of VEGFA expression and PD-1/PD-L1 checkpoint. Thalidomide 12-23 vascular endothelial growth factor A Homo sapiens 128-133 34520751-17 2021 CONCLUSION: Thalidomide inhibits NSCLC angiogenesis and immune evasion via FGD5-AS1/miR-454-3p/ZEB1 axis-mediated regulation of VEGFA expression and PD-1/PD-L1 checkpoint. Thalidomide 12-23 programmed cell death 1 Homo sapiens 149-153 34520751-17 2021 CONCLUSION: Thalidomide inhibits NSCLC angiogenesis and immune evasion via FGD5-AS1/miR-454-3p/ZEB1 axis-mediated regulation of VEGFA expression and PD-1/PD-L1 checkpoint. Thalidomide 12-23 CD274 molecule Homo sapiens 154-159 34085173-11 2021 Mechanistically, we observed that thalidomide could increase epithelial cell self-renewal capacity and modulate M1/M2 polarization by decreasing M1 markers CD86 and CCR7 and increasing M2 protein signatures CD206 and Arg-1. Thalidomide 34-45 mannose receptor, C type 1 Mus musculus 207-212 34085173-11 2021 Mechanistically, we observed that thalidomide could increase epithelial cell self-renewal capacity and modulate M1/M2 polarization by decreasing M1 markers CD86 and CCR7 and increasing M2 protein signatures CD206 and Arg-1. Thalidomide 34-45 arginase, liver Mus musculus 217-222 34085173-12 2021 Thalidomide controls M1 macrophage polarization by targeting the transcription factor IRF5. Thalidomide 0-11 interferon regulatory factor 5 Mus musculus 86-90 34583995-3 2021 CRBN, a substrate receptor of CUL4-RBX1-DDB1-CRBN (CRL4CRBN) E3 ubiquitin ligase, is a direct target for thalidomide teratogenicity and antitumor activity of IMiDs (now known as Cereblon E3 ligase modulators: CELMoDs). Thalidomide 105-116 cereblon Homo sapiens 0-4 34339726-0 2021 Palladium (II) complex and thalidomide intercept angiogenic signaling via targeting FAK/Src and Erk/Akt/PLCgamma dependent autophagy pathways in human umbilical vein endothelial cells. Thalidomide 27-38 protein tyrosine kinase 2 Homo sapiens 84-87 34339726-0 2021 Palladium (II) complex and thalidomide intercept angiogenic signaling via targeting FAK/Src and Erk/Akt/PLCgamma dependent autophagy pathways in human umbilical vein endothelial cells. Thalidomide 27-38 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 88-91 34339726-0 2021 Palladium (II) complex and thalidomide intercept angiogenic signaling via targeting FAK/Src and Erk/Akt/PLCgamma dependent autophagy pathways in human umbilical vein endothelial cells. Thalidomide 27-38 mitogen-activated protein kinase 1 Homo sapiens 96-99 34339726-0 2021 Palladium (II) complex and thalidomide intercept angiogenic signaling via targeting FAK/Src and Erk/Akt/PLCgamma dependent autophagy pathways in human umbilical vein endothelial cells. Thalidomide 27-38 AKT serine/threonine kinase 1 Homo sapiens 100-103 34339726-2 2021 The viability was assessed after treatment with palladium (II) complex (1.56-100 muM) and thalidomide (0.1-400 muM) alone by using ATP assay for 48 h. Palladium (II) complex was found to inhibit growth statistically significant in a dose-dependent manner in HUVECs and promoted PARP-1 cleavage through the production of ROS. Thalidomide 90-101 poly(ADP-ribose) polymerase 1 Homo sapiens 278-284 34339726-8 2021 We also showed that being treated with thalidomide and palladium (II) complex inhibited phosphorylation of the signaling regulators downstream of the VEGFR2. Thalidomide 39-50 kinase insert domain receptor Homo sapiens 150-156 34339726-11 2021 Hence, we demonstrated that palladium (II) and thalidomide can induce cell death via the Erk/Akt/PLCgamma signaling pathway and that this pathway might be a novel mechanism. Thalidomide 47-58 mitogen-activated protein kinase 1 Homo sapiens 89-92 34339726-11 2021 Hence, we demonstrated that palladium (II) and thalidomide can induce cell death via the Erk/Akt/PLCgamma signaling pathway and that this pathway might be a novel mechanism. Thalidomide 47-58 AKT serine/threonine kinase 1 Homo sapiens 93-96 34583995-3 2021 CRBN, a substrate receptor of CUL4-RBX1-DDB1-CRBN (CRL4CRBN) E3 ubiquitin ligase, is a direct target for thalidomide teratogenicity and antitumor activity of IMiDs (now known as Cereblon E3 ligase modulators: CELMoDs). Thalidomide 105-116 ring-box 1 Homo sapiens 35-39 34583995-3 2021 CRBN, a substrate receptor of CUL4-RBX1-DDB1-CRBN (CRL4CRBN) E3 ubiquitin ligase, is a direct target for thalidomide teratogenicity and antitumor activity of IMiDs (now known as Cereblon E3 ligase modulators: CELMoDs). Thalidomide 105-116 damage specific DNA binding protein 1 Homo sapiens 40-44 34583995-3 2021 CRBN, a substrate receptor of CUL4-RBX1-DDB1-CRBN (CRL4CRBN) E3 ubiquitin ligase, is a direct target for thalidomide teratogenicity and antitumor activity of IMiDs (now known as Cereblon E3 ligase modulators: CELMoDs). Thalidomide 105-116 cereblon Homo sapiens 45-49 34583995-3 2021 CRBN, a substrate receptor of CUL4-RBX1-DDB1-CRBN (CRL4CRBN) E3 ubiquitin ligase, is a direct target for thalidomide teratogenicity and antitumor activity of IMiDs (now known as Cereblon E3 ligase modulators: CELMoDs). Thalidomide 105-116 interleukin 17 receptor B Homo sapiens 51-59 34641286-0 2021 Isotretinoin and Thalidomide Down-Regulate c-MYC Gene Expression and Modify Proteins Associated with Cancer in Hepatic Cells. Thalidomide 17-28 MYC proto-oncogene, bHLH transcription factor Homo sapiens 43-48 34312685-0 2021 Combined use of interferon alpha-1b, interleukin-2, and thalidomide to reverse the AML1-ETO fusion gene in acute myeloid leukemia. Thalidomide 56-67 RUNX family transcription factor 1 Homo sapiens 83-87 34312685-0 2021 Combined use of interferon alpha-1b, interleukin-2, and thalidomide to reverse the AML1-ETO fusion gene in acute myeloid leukemia. Thalidomide 56-67 RUNX1 partner transcriptional co-repressor 1 Homo sapiens 88-91 34312685-1 2021 This study aims to explore the effect of the ITI (interferon alpha-1b, thalidomide, and interleukin-2) regimen on the AML1-ETO fusion gene in patients with t(8;21) acute myeloid leukemia (AML) who were in hematologic remission but positive for the AML1-ETO fusion gene. Thalidomide 71-82 RUNX family transcription factor 1 Homo sapiens 118-122 34312685-1 2021 This study aims to explore the effect of the ITI (interferon alpha-1b, thalidomide, and interleukin-2) regimen on the AML1-ETO fusion gene in patients with t(8;21) acute myeloid leukemia (AML) who were in hematologic remission but positive for the AML1-ETO fusion gene. Thalidomide 71-82 RUNX1 partner transcriptional co-repressor 1 Homo sapiens 123-126 34312685-1 2021 This study aims to explore the effect of the ITI (interferon alpha-1b, thalidomide, and interleukin-2) regimen on the AML1-ETO fusion gene in patients with t(8;21) acute myeloid leukemia (AML) who were in hematologic remission but positive for the AML1-ETO fusion gene. Thalidomide 71-82 RUNX family transcription factor 1 Homo sapiens 248-252 34312685-1 2021 This study aims to explore the effect of the ITI (interferon alpha-1b, thalidomide, and interleukin-2) regimen on the AML1-ETO fusion gene in patients with t(8;21) acute myeloid leukemia (AML) who were in hematologic remission but positive for the AML1-ETO fusion gene. Thalidomide 71-82 RUNX1 partner transcriptional co-repressor 1 Homo sapiens 253-256 34390739-8 2021 Particularly, thalidomide specifically stimulated IL-10 and beta-endorphin expressions in microglia but not astrocytes or neurons. Thalidomide 14-25 interleukin 10 Rattus norvegicus 50-55 34390739-10 2021 Importantly, pretreatment with intrathecal injection of the microglial metabolic inhibitor minocycline, IL-10 antibody, beta-endorphin antiserum, and preferred or selective mu-opioid receptor antagonist naloxone or CTAP entirely blocked thalidomide gavage-induced mechanical antiallodynia. Thalidomide 237-248 interleukin 10 Rattus norvegicus 104-109 34390739-11 2021 Our results demonstrate that thalidomide, but not lenalidomide or pomalidomide, alleviates neuropathic pain, which is mediated by upregulation of spinal microglial IL-10/beta-endorphin expression, rather than downregulation of TNFalpha expression. Thalidomide 29-40 interleukin 10 Rattus norvegicus 164-169 34390739-0 2021 Thalidomide alleviates neuropathic pain through microglial IL-10/beta-endorphin signaling pathway. Thalidomide 0-11 interleukin 10 Rattus norvegicus 59-64 34390739-6 2021 Treatment with thalidomide, lenalidomide and pomalidomide reduced peripheral nerve injury-induced proinflammatory cytokines (TNFalpha, IL-1beta and IL-6) in the ipsilateral spinal cords of neuropathic rats and LPS-treated primary microglial cells. Thalidomide 15-26 tumor necrosis factor Rattus norvegicus 125-133 34390739-6 2021 Treatment with thalidomide, lenalidomide and pomalidomide reduced peripheral nerve injury-induced proinflammatory cytokines (TNFalpha, IL-1beta and IL-6) in the ipsilateral spinal cords of neuropathic rats and LPS-treated primary microglial cells. Thalidomide 15-26 interleukin 1 alpha Rattus norvegicus 135-143 34390739-6 2021 Treatment with thalidomide, lenalidomide and pomalidomide reduced peripheral nerve injury-induced proinflammatory cytokines (TNFalpha, IL-1beta and IL-6) in the ipsilateral spinal cords of neuropathic rats and LPS-treated primary microglial cells. Thalidomide 15-26 interleukin 6 Rattus norvegicus 148-152 34390739-7 2021 In contrast, treatment with thalidomide, but not lenalidomide or pomalidomide, stimulated spinal expressions of IL-10 and beta-endorphin in neuropathic rats. Thalidomide 28-39 interleukin 10 Rattus norvegicus 112-117 34584130-9 2021 Thalidomide treatment significantly reduced serum levels of hepatic enzymes and TNF-alpha, IL-1-beta, and IL-6. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 80-89 34584130-9 2021 Thalidomide treatment significantly reduced serum levels of hepatic enzymes and TNF-alpha, IL-1-beta, and IL-6. Thalidomide 0-11 interleukin 1 alpha Rattus norvegicus 91-100 34584130-9 2021 Thalidomide treatment significantly reduced serum levels of hepatic enzymes and TNF-alpha, IL-1-beta, and IL-6. Thalidomide 0-11 interleukin 6 Rattus norvegicus 106-110 34251884-6 2021 These data might have also implications for cancer, as frequently occurring mutations of MEKK1 might have an impact on genome stability and the therapeutic efficacy of CRL4-dependent immunomodulatory drugs such as thalidomide-derivatives. Thalidomide 214-225 mitogen-activated protein kinase kinase kinase 1 Homo sapiens 89-94 34251884-6 2021 These data might have also implications for cancer, as frequently occurring mutations of MEKK1 might have an impact on genome stability and the therapeutic efficacy of CRL4-dependent immunomodulatory drugs such as thalidomide-derivatives. Thalidomide 214-225 interleukin 17 receptor B Homo sapiens 168-172 34641286-5 2021 The objective of this work was to evaluate the effect of the drugs isotretinoin and thalidomide on c-MYC expression and cancer-related proteins in an HCC cellular model. Thalidomide 84-95 MYC proto-oncogene, bHLH transcription factor Homo sapiens 99-104 34641286-10 2021 We demonstrated that in HepG2 cells, isotretinoin and thalidomide reduced c-MYC mRNA expression levels, but this decrease in expression was linked to the regulation of P1 and P1-P2 c-MYC promoter activity in isotretinoin only. Thalidomide 54-65 MYC proto-oncogene, bHLH transcription factor Homo sapiens 74-79 34641286-13 2021 In conclusion, isotretinoin and thalidomide down-regulate c-MYC mRNA expression and this is partially due to P1 or P2 promoter activity, suggesting that these drugs could be promising options for modulating the expression of oncogenes and tumor suppressor genes in HCC. Thalidomide 32-43 MYC proto-oncogene, bHLH transcription factor Homo sapiens 58-63 34522548-4 2021 She had several stigmata of thalidomide embryopathy. Thalidomide 28-39 Src homology 2 domain containing E Homo sapiens 0-3 34530760-6 2021 The antiangiogenic and immunomodulatory drug thalidomide is a cereblon-based molecular glue that can induce the degradation of the IKAROS family transcription factors IKZF1 and IKZF3. Thalidomide 45-56 IKAROS family zinc finger 1 Homo sapiens 167-172 34530760-6 2021 The antiangiogenic and immunomodulatory drug thalidomide is a cereblon-based molecular glue that can induce the degradation of the IKAROS family transcription factors IKZF1 and IKZF3. Thalidomide 45-56 IKAROS family zinc finger 3 Homo sapiens 177-182 34530760-14 2021 DISCUSSION: The addition of thalidomide to pyrotinib is expected to increase the clinical benefit in advanced NSCLC patients with HER2 exon 20 insertions, and reduce the incidence of pyrotinib-related diarrhea. Thalidomide 28-39 erb-b2 receptor tyrosine kinase 2 Homo sapiens 130-134 34650759-0 2021 Effect of thalidomide combined with TP chemotherapy on serum VEGF and NRP-1 levels advanced esophageal cancer patients. Thalidomide 10-21 vascular endothelial growth factor A Homo sapiens 61-65 34650759-0 2021 Effect of thalidomide combined with TP chemotherapy on serum VEGF and NRP-1 levels advanced esophageal cancer patients. Thalidomide 10-21 neuropilin 1 Homo sapiens 70-75 34650759-1 2021 OBJECTIVE: This study aimed to investigate the effect of thalidomide combined with pacilitaxel plus cisplatin (TP) chemotherapy on serum vascular endothelial growth factor (VEGF) and neuropilin-1 (NRP-1) levels in advanced esophageal cancer patients. Thalidomide 57-68 vascular endothelial growth factor A Homo sapiens 137-171 34650759-1 2021 OBJECTIVE: This study aimed to investigate the effect of thalidomide combined with pacilitaxel plus cisplatin (TP) chemotherapy on serum vascular endothelial growth factor (VEGF) and neuropilin-1 (NRP-1) levels in advanced esophageal cancer patients. Thalidomide 57-68 vascular endothelial growth factor A Homo sapiens 173-177 34650759-1 2021 OBJECTIVE: This study aimed to investigate the effect of thalidomide combined with pacilitaxel plus cisplatin (TP) chemotherapy on serum vascular endothelial growth factor (VEGF) and neuropilin-1 (NRP-1) levels in advanced esophageal cancer patients. Thalidomide 57-68 neuropilin 1 Homo sapiens 183-195 34650759-1 2021 OBJECTIVE: This study aimed to investigate the effect of thalidomide combined with pacilitaxel plus cisplatin (TP) chemotherapy on serum vascular endothelial growth factor (VEGF) and neuropilin-1 (NRP-1) levels in advanced esophageal cancer patients. Thalidomide 57-68 neuropilin 1 Homo sapiens 197-202 34539435-2 2021 Thalidomide has been confirmed to inhibit angiogenesis by inhibiting the secretion of vascular endothelial growth factor (VEGF), but the exact mechanism by which thalidomide inhibits vascular proliferation during PD is still unclear. Thalidomide 0-11 vascular endothelial growth factor A Homo sapiens 122-126 34539435-2 2021 Thalidomide has been confirmed to inhibit angiogenesis by inhibiting the secretion of vascular endothelial growth factor (VEGF), but the exact mechanism by which thalidomide inhibits vascular proliferation during PD is still unclear. Thalidomide 162-173 vascular endothelial growth factor A Homo sapiens 122-126 34539435-3 2021 Here, the objective of the present study was to investigate whether the reduction in VEGF production by human peritoneal mesothelial cells (HPMCs) was controlled by thalidomide. Thalidomide 165-176 vascular endothelial growth factor A Homo sapiens 85-89 34539435-4 2021 Stimulation of HPMCs with IL-6 in combination with soluble IL-6 receptor (sIL-6R) promoted VEGF expression and secretion, but these effects were attenuated by thalidomide treatment through a transcriptional mechanism that involved signal transducer and activator of transcription 3 (STAT3) and SP4. Thalidomide 159-170 interleukin 6 Rattus norvegicus 26-30 34539435-4 2021 Stimulation of HPMCs with IL-6 in combination with soluble IL-6 receptor (sIL-6R) promoted VEGF expression and secretion, but these effects were attenuated by thalidomide treatment through a transcriptional mechanism that involved signal transducer and activator of transcription 3 (STAT3) and SP4. Thalidomide 159-170 vascular endothelial growth factor A Homo sapiens 91-95 34539435-4 2021 Stimulation of HPMCs with IL-6 in combination with soluble IL-6 receptor (sIL-6R) promoted VEGF expression and secretion, but these effects were attenuated by thalidomide treatment through a transcriptional mechanism that involved signal transducer and activator of transcription 3 (STAT3) and SP4. Thalidomide 159-170 signal transducer and activator of transcription 3 Rattus norvegicus 231-281 34539435-4 2021 Stimulation of HPMCs with IL-6 in combination with soluble IL-6 receptor (sIL-6R) promoted VEGF expression and secretion, but these effects were attenuated by thalidomide treatment through a transcriptional mechanism that involved signal transducer and activator of transcription 3 (STAT3) and SP4. Thalidomide 159-170 signal transducer and activator of transcription 3 Homo sapiens 283-288 34539435-4 2021 Stimulation of HPMCs with IL-6 in combination with soluble IL-6 receptor (sIL-6R) promoted VEGF expression and secretion, but these effects were attenuated by thalidomide treatment through a transcriptional mechanism that involved signal transducer and activator of transcription 3 (STAT3) and SP4. Thalidomide 159-170 Sp4 transcription factor Homo sapiens 294-297 34539435-5 2021 Conditioned medium from HPMCs cultured with thalidomide inhibited angiogenic endothelial tube formation, which could be further blocked by silencing SP4 and promoted by overexpressing SP4. Thalidomide 44-55 Sp4 transcription factor Homo sapiens 149-152 34539435-5 2021 Conditioned medium from HPMCs cultured with thalidomide inhibited angiogenic endothelial tube formation, which could be further blocked by silencing SP4 and promoted by overexpressing SP4. Thalidomide 44-55 Sp4 transcription factor Homo sapiens 184-187 34539435-7 2021 Taken together, these findings identify a novel mechanism that links thalidomide, STAT3/SP4 signaling, and angiogenesis in the peritoneal membrane. Thalidomide 69-80 Sp4 transcription factor Rattus norvegicus 88-91 34431670-3 2021 Employing thalidomide to recruit cereblon/cullin 4A E3 ligase and through the rigidification of the linker, we discovered highly potent AR degraders with good oral pharmacokinetic properties in mice with ARD-2128 being the best compound. Thalidomide 10-21 androgen receptor Mus musculus 136-138 34539435-0 2021 Thalidomide Suppresses Angiogenesis Through the Signal Transducer and Activator of Transcription 3/SP4 Signaling Pathway in the Peritoneal Membrane. Thalidomide 0-11 signal transducer and activator of transcription 3 Rattus norvegicus 48-98 34539435-0 2021 Thalidomide Suppresses Angiogenesis Through the Signal Transducer and Activator of Transcription 3/SP4 Signaling Pathway in the Peritoneal Membrane. Thalidomide 0-11 Sp4 transcription factor Rattus norvegicus 99-102 34512420-8 2021 Administration of TNF-alpha synthesis blocker, thalidomide (THL), immediately after SPS prevented increased TNF-alpha and development of allodynia and hyperalgesia. Thalidomide 47-58 tumor necrosis factor Homo sapiens 18-27 34512420-8 2021 Administration of TNF-alpha synthesis blocker, thalidomide (THL), immediately after SPS prevented increased TNF-alpha and development of allodynia and hyperalgesia. Thalidomide 47-58 tumor necrosis factor Homo sapiens 108-117 34512420-8 2021 Administration of TNF-alpha synthesis blocker, thalidomide (THL), immediately after SPS prevented increased TNF-alpha and development of allodynia and hyperalgesia. Thalidomide 60-63 tumor necrosis factor Homo sapiens 18-27 34512420-8 2021 Administration of TNF-alpha synthesis blocker, thalidomide (THL), immediately after SPS prevented increased TNF-alpha and development of allodynia and hyperalgesia. Thalidomide 60-63 tumor necrosis factor Homo sapiens 108-117 34512420-10 2021 THL also prevented SPS-induced increases in serum N/OFQ and reversed regional N/OFQ mRNA expression changes in the CNS. Thalidomide 0-3 prepronociceptin Homo sapiens 50-55 34512420-10 2021 THL also prevented SPS-induced increases in serum N/OFQ and reversed regional N/OFQ mRNA expression changes in the CNS. Thalidomide 0-3 prepronociceptin Homo sapiens 78-83 34150160-0 2021 The effect of TACE in combination with thalidomide-mediated adjuvant therapy on the levels of VEGF and bFGF in patients with hepatocellular carcinoma. Thalidomide 39-50 vascular endothelial growth factor A Homo sapiens 94-98 34128013-3 2021 To compare the structure of the bound CRBN thalidomide enantiomers with that of the isolated molecule, the rotational spectrum of laser-ablated thalidomide has been studied by chirp-pulsed Fourier transform microwave spectroscopy in supersonic jets complemented by theoretical computations. Thalidomide 43-54 cereblon Homo sapiens 38-42 34128013-9 2021 This is consistent with a less stable (R)-enantiomer and the known preference of (S)-thalidomide to bind CRBN, which starts the process leading to teratogenic effects. Thalidomide 81-96 cereblon Homo sapiens 105-109 34207079-4 2021 Here, we evaluated the expression of CRL4-CRBN complex proteins and their downstream targets with immunohistochemistry (IHC) staining in 130 bone marrow samples from MM patients treated with thalidomide or lenalidomide-based regimens. Thalidomide 191-202 interleukin 17 receptor B Homo sapiens 37-41 34207079-4 2021 Here, we evaluated the expression of CRL4-CRBN complex proteins and their downstream targets with immunohistochemistry (IHC) staining in 130 bone marrow samples from MM patients treated with thalidomide or lenalidomide-based regimens. Thalidomide 191-202 cereblon Homo sapiens 42-46 34306363-6 2021 The caspase 3, 8, and 9 immunoreactivities were increased in all treatment groups and TUNEL positive cells were the highest in the Wortmannin and Thalidomide group. Thalidomide 146-157 caspase 3 Mus musculus 4-13 34073624-4 2021 However, the effect of thalidomide on the function of CRBN as a negative regulator of AMPK through interaction with the alpha subunit remains unclear. Thalidomide 23-34 cereblon Homo sapiens 54-58 34249098-0 2021 Comparative Genomics Identifies Putative Interspecies Mechanisms Underlying Crbn-Sall4-Linked Thalidomide Embryopathy. Thalidomide 94-105 cereblon Mus musculus 76-80 34249098-0 2021 Comparative Genomics Identifies Putative Interspecies Mechanisms Underlying Crbn-Sall4-Linked Thalidomide Embryopathy. Thalidomide 94-105 spalt like transcription factor 4 Mus musculus 81-86 34249098-1 2021 The identification of thalidomide-Cereblon-induced SALL4 degradation has brought new understanding for thalidomide embryopathy (TE) differences across species. Thalidomide 22-33 spalt like transcription factor 4 Mus musculus 51-56 34249098-1 2021 The identification of thalidomide-Cereblon-induced SALL4 degradation has brought new understanding for thalidomide embryopathy (TE) differences across species. Thalidomide 103-114 spalt like transcription factor 4 Mus musculus 51-56 34249098-9 2021 In co-expression analysis, Crbn was a gene identified as a driver of the co-expression of other genes implicated in genetic, non-teratogenic, limb reduction defects (LRD), such as Tbx5, Esco2, Recql4, and Sall4; Crbn and Sall4 were shown to have a moderate co-expression correlation, which is affected after thalidomide exposure. Thalidomide 308-319 cereblon Mus musculus 27-31 34249098-9 2021 In co-expression analysis, Crbn was a gene identified as a driver of the co-expression of other genes implicated in genetic, non-teratogenic, limb reduction defects (LRD), such as Tbx5, Esco2, Recql4, and Sall4; Crbn and Sall4 were shown to have a moderate co-expression correlation, which is affected after thalidomide exposure. Thalidomide 308-319 T-box 5 Mus musculus 180-184 34249098-9 2021 In co-expression analysis, Crbn was a gene identified as a driver of the co-expression of other genes implicated in genetic, non-teratogenic, limb reduction defects (LRD), such as Tbx5, Esco2, Recql4, and Sall4; Crbn and Sall4 were shown to have a moderate co-expression correlation, which is affected after thalidomide exposure. Thalidomide 308-319 establishment of sister chromatid cohesion N-acetyltransferase 2 Mus musculus 186-191 34249098-9 2021 In co-expression analysis, Crbn was a gene identified as a driver of the co-expression of other genes implicated in genetic, non-teratogenic, limb reduction defects (LRD), such as Tbx5, Esco2, Recql4, and Sall4; Crbn and Sall4 were shown to have a moderate co-expression correlation, which is affected after thalidomide exposure. Thalidomide 308-319 RecQ protein-like 4 Mus musculus 193-199 34249098-9 2021 In co-expression analysis, Crbn was a gene identified as a driver of the co-expression of other genes implicated in genetic, non-teratogenic, limb reduction defects (LRD), such as Tbx5, Esco2, Recql4, and Sall4; Crbn and Sall4 were shown to have a moderate co-expression correlation, which is affected after thalidomide exposure. Thalidomide 308-319 spalt like transcription factor 4 Mus musculus 205-210 34249098-9 2021 In co-expression analysis, Crbn was a gene identified as a driver of the co-expression of other genes implicated in genetic, non-teratogenic, limb reduction defects (LRD), such as Tbx5, Esco2, Recql4, and Sall4; Crbn and Sall4 were shown to have a moderate co-expression correlation, which is affected after thalidomide exposure. Thalidomide 308-319 cereblon Mus musculus 212-216 34249098-9 2021 In co-expression analysis, Crbn was a gene identified as a driver of the co-expression of other genes implicated in genetic, non-teratogenic, limb reduction defects (LRD), such as Tbx5, Esco2, Recql4, and Sall4; Crbn and Sall4 were shown to have a moderate co-expression correlation, which is affected after thalidomide exposure. Thalidomide 308-319 spalt like transcription factor 4 Mus musculus 221-226 34150160-0 2021 The effect of TACE in combination with thalidomide-mediated adjuvant therapy on the levels of VEGF and bFGF in patients with hepatocellular carcinoma. Thalidomide 39-50 fibroblast growth factor 2 Homo sapiens 103-107 34150160-1 2021 OBJECTIVE: To investigate the efficacy of transcatheter arterial chemoembolization (TACE) combined with thalidomide-mediated adjuvant therapy on the expression levels of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in hepatocellular carcinoma (HCC) patients. Thalidomide 104-115 vascular endothelial growth factor A Homo sapiens 170-204 34150160-1 2021 OBJECTIVE: To investigate the efficacy of transcatheter arterial chemoembolization (TACE) combined with thalidomide-mediated adjuvant therapy on the expression levels of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in hepatocellular carcinoma (HCC) patients. Thalidomide 104-115 vascular endothelial growth factor A Homo sapiens 206-210 34150160-1 2021 OBJECTIVE: To investigate the efficacy of transcatheter arterial chemoembolization (TACE) combined with thalidomide-mediated adjuvant therapy on the expression levels of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in hepatocellular carcinoma (HCC) patients. Thalidomide 104-115 fibroblast growth factor 2 Homo sapiens 216-246 34150160-1 2021 OBJECTIVE: To investigate the efficacy of transcatheter arterial chemoembolization (TACE) combined with thalidomide-mediated adjuvant therapy on the expression levels of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in hepatocellular carcinoma (HCC) patients. Thalidomide 104-115 fibroblast growth factor 2 Homo sapiens 248-252 34150160-13 2021 CONCLUSION: TACE in combination with thalidomide-mediated adjuvant treatment has revealed a promising clinical outcome on HCC patients by downregulating the levels of VEGF and bFGF. Thalidomide 37-48 vascular endothelial growth factor A Homo sapiens 167-171 34150160-13 2021 CONCLUSION: TACE in combination with thalidomide-mediated adjuvant treatment has revealed a promising clinical outcome on HCC patients by downregulating the levels of VEGF and bFGF. Thalidomide 37-48 fibroblast growth factor 2 Homo sapiens 176-180 34853241-7 2021 These results suggest that metabolic activation of thalidomide may be dependent on rabbit liver enzymes just it was for cytochrome P450 enzymes in humanized-liver mice; in contrast, rodent livers predominantly mediate biotransformation of thalidomide to 5"-hydroxythalidomide. Thalidomide 51-62 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 120-135 34162559-0 2021 Thalidomide Reduces Activation of Murine Pancreatic Stellate Cells by Inhibiting the TGF-beta/Smad Pathway. Thalidomide 0-11 transforming growth factor alpha Mus musculus 85-93 34162559-0 2021 Thalidomide Reduces Activation of Murine Pancreatic Stellate Cells by Inhibiting the TGF-beta/Smad Pathway. Thalidomide 0-11 SMAD family member 7 Mus musculus 94-98 34162559-3 2021 We measured the effects of thalidomide on PSC activation by detecting the expression of alpha-SMA, collagen type I, and the TGF-beta/Smad pathway through quantitative real-time PCR and Western blot analysis. Thalidomide 27-38 actin alpha 2, smooth muscle, aorta Mus musculus 88-97 34162559-3 2021 We measured the effects of thalidomide on PSC activation by detecting the expression of alpha-SMA, collagen type I, and the TGF-beta/Smad pathway through quantitative real-time PCR and Western blot analysis. Thalidomide 27-38 transforming growth factor alpha Mus musculus 124-132 34162559-4 2021 RESULTS: Compared with TGF-beta1 alone, thalidomide significantly inhibited PSC activation by reducing alpha-SMA expression (P<0.05) and decreasing collagen type I deposition (P<0.05). Thalidomide 40-51 actin alpha 2, smooth muscle, aorta Mus musculus 103-112 34162559-5 2021 PSCs treated with thalidomide alone showed lower expression of alpha-SMA and collagen type I than those treated with thalidomide and TGF-beta1 at random order (P<0.01). Thalidomide 18-29 corneal susceptibility to P. aeruginosa Mus musculus 0-4 34162559-5 2021 PSCs treated with thalidomide alone showed lower expression of alpha-SMA and collagen type I than those treated with thalidomide and TGF-beta1 at random order (P<0.01). Thalidomide 18-29 actin alpha 2, smooth muscle, aorta Mus musculus 63-72 34162559-5 2021 PSCs treated with thalidomide alone showed lower expression of alpha-SMA and collagen type I than those treated with thalidomide and TGF-beta1 at random order (P<0.01). Thalidomide 117-128 actin alpha 2, smooth muscle, aorta Mus musculus 63-72 34162559-6 2021 Thalidomide downregulated TGF-beta1 and Smad3 and upregulated Smad7 (P<0.05). Thalidomide 0-11 transforming growth factor, beta 1 Mus musculus 26-35 34162559-6 2021 Thalidomide downregulated TGF-beta1 and Smad3 and upregulated Smad7 (P<0.05). Thalidomide 0-11 SMAD family member 3 Mus musculus 40-45 34162559-6 2021 Thalidomide downregulated TGF-beta1 and Smad3 and upregulated Smad7 (P<0.05). Thalidomide 0-11 SMAD family member 7 Mus musculus 62-67 34162559-7 2021 CONCLUSION: Thalidomide could repress PSC activation and alleviate fibrosis by regulating the TGF-beta/Smad pathway. Thalidomide 12-23 transforming growth factor alpha Mus musculus 94-102 34162559-7 2021 CONCLUSION: Thalidomide could repress PSC activation and alleviate fibrosis by regulating the TGF-beta/Smad pathway. Thalidomide 12-23 SMAD family member 7 Mus musculus 103-107 35413617-3 2022 It was found that thalidomide and its analogs could bind to CRBN E3 ubiquitin ligase and modulate CRBN. Thalidomide 18-29 cereblon Homo sapiens 98-102 35472743-2 2022 Food and Drug Administration (FDA)-approved immune-modulatory agent, thalidomide, can modulate the related proteins of upstream signaling pathway of programmed cell death-ligand 1 (PD-L1), including nuclear transcription factor kappaB (NF-kappaB), hypoxia inducible factor-1alpha (HIF-1alpha), epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), all acting as key antitumor target proteins. Thalidomide 69-80 CD274 molecule Homo sapiens 149-179 35472743-2 2022 Food and Drug Administration (FDA)-approved immune-modulatory agent, thalidomide, can modulate the related proteins of upstream signaling pathway of programmed cell death-ligand 1 (PD-L1), including nuclear transcription factor kappaB (NF-kappaB), hypoxia inducible factor-1alpha (HIF-1alpha), epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), all acting as key antitumor target proteins. Thalidomide 69-80 CD274 molecule Homo sapiens 181-186 35472743-2 2022 Food and Drug Administration (FDA)-approved immune-modulatory agent, thalidomide, can modulate the related proteins of upstream signaling pathway of programmed cell death-ligand 1 (PD-L1), including nuclear transcription factor kappaB (NF-kappaB), hypoxia inducible factor-1alpha (HIF-1alpha), epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), all acting as key antitumor target proteins. Thalidomide 69-80 nuclear factor kappa B subunit 1 Homo sapiens 236-245 35472743-2 2022 Food and Drug Administration (FDA)-approved immune-modulatory agent, thalidomide, can modulate the related proteins of upstream signaling pathway of programmed cell death-ligand 1 (PD-L1), including nuclear transcription factor kappaB (NF-kappaB), hypoxia inducible factor-1alpha (HIF-1alpha), epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), all acting as key antitumor target proteins. Thalidomide 69-80 hypoxia inducible factor 1 subunit alpha Homo sapiens 248-279 35472743-2 2022 Food and Drug Administration (FDA)-approved immune-modulatory agent, thalidomide, can modulate the related proteins of upstream signaling pathway of programmed cell death-ligand 1 (PD-L1), including nuclear transcription factor kappaB (NF-kappaB), hypoxia inducible factor-1alpha (HIF-1alpha), epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), all acting as key antitumor target proteins. Thalidomide 69-80 hypoxia inducible factor 1 subunit alpha Homo sapiens 281-291 35472743-2 2022 Food and Drug Administration (FDA)-approved immune-modulatory agent, thalidomide, can modulate the related proteins of upstream signaling pathway of programmed cell death-ligand 1 (PD-L1), including nuclear transcription factor kappaB (NF-kappaB), hypoxia inducible factor-1alpha (HIF-1alpha), epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), all acting as key antitumor target proteins. Thalidomide 69-80 epidermal growth factor receptor Homo sapiens 294-326 35472743-2 2022 Food and Drug Administration (FDA)-approved immune-modulatory agent, thalidomide, can modulate the related proteins of upstream signaling pathway of programmed cell death-ligand 1 (PD-L1), including nuclear transcription factor kappaB (NF-kappaB), hypoxia inducible factor-1alpha (HIF-1alpha), epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), all acting as key antitumor target proteins. Thalidomide 69-80 epidermal growth factor receptor Homo sapiens 328-332 35472743-2 2022 Food and Drug Administration (FDA)-approved immune-modulatory agent, thalidomide, can modulate the related proteins of upstream signaling pathway of programmed cell death-ligand 1 (PD-L1), including nuclear transcription factor kappaB (NF-kappaB), hypoxia inducible factor-1alpha (HIF-1alpha), epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), all acting as key antitumor target proteins. Thalidomide 69-80 signal transducer and activator of transcription 3 Homo sapiens 339-389 35472743-2 2022 Food and Drug Administration (FDA)-approved immune-modulatory agent, thalidomide, can modulate the related proteins of upstream signaling pathway of programmed cell death-ligand 1 (PD-L1), including nuclear transcription factor kappaB (NF-kappaB), hypoxia inducible factor-1alpha (HIF-1alpha), epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), all acting as key antitumor target proteins. Thalidomide 69-80 signal transducer and activator of transcription 3 Homo sapiens 391-396 35472743-6 2022 Fluorescent confocal and western blotting experiments proved that 6 significantly regulated NF-kappaB, EGFR, HIF-1alpha and phosphor-signal transducer and activator of transcription 3 (p-STAT3), and simultaneously inhibited PD-L1 expression to interrupt programmed cell death 1 (PD-1)/PD-L1 signaling pathway, suggesting a synergistic action of cisplatin and thalidomide. Thalidomide 359-370 signal transducer and activator of transcription 3 Homo sapiens 187-192 35628464-0 2022 Thalidomide Alleviates Pulmonary Fibrosis Induced by Silica in Mice by Inhibiting ER Stress and the TLR4-NF-kappaB Pathway. Thalidomide 0-11 toll-like receptor 4 Mus musculus 100-104 35384188-0 2022 Cutaneous involvement in DOCK8 related immunodeficiency syndrome responding to thalidomide. Thalidomide 79-90 dedicator of cytokinesis 8 Homo sapiens 25-30 35495588-0 2022 Targeting TNFalpha-mediated cytotoxicity using thalidomide after experimental cardiac arrest in rats: An exploratory study. Thalidomide 47-58 tumor necrosis factor Rattus norvegicus 10-18 35495588-2 2022 Thalidomide has been reported to be neuroprotective by selectively decreasing TNFalpha synthesis. Thalidomide 0-11 tumor necrosis factor Rattus norvegicus 78-86 35495588-3 2022 We hypothesized that thalidomide would decrease the systemic and organ-specific TNFalpha/cytokine response and biomarkers of injury in rats subjected to 10 min CA. Thalidomide 21-32 tumor necrosis factor Rattus norvegicus 80-88 35628464-0 2022 Thalidomide Alleviates Pulmonary Fibrosis Induced by Silica in Mice by Inhibiting ER Stress and the TLR4-NF-kappaB Pathway. Thalidomide 0-11 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 105-114 35628464-9 2022 Treating both silicotic mice and silica-stimulated MH-S cells with thalidomide inhibited ER stress and the TLR4-NF-kappaB pathway as well as the inflammatory response. Thalidomide 67-78 toll-like receptor 4 Mus musculus 107-111 35628464-9 2022 Treating both silicotic mice and silica-stimulated MH-S cells with thalidomide inhibited ER stress and the TLR4-NF-kappaB pathway as well as the inflammatory response. Thalidomide 67-78 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 112-121 34768284-5 2022 We validated the predictive role of MCT1 for IMiD-based maintenance in an independent cohort of patients receiving thalidomide (OS 83.6 months vs. NR in MCT1high vs. MCT1low; P=.03). Thalidomide 115-126 solute carrier family 16 member 1 Homo sapiens 36-40 35595613-6 2022 On the other hand, the efficiency of Thalidomide derivates in myelodysplastic syndromes (MDS), such as Lenalidomide, acted as the starting point for the development of targeted leukemia-associated protein degradation molecules. Thalidomide 37-48 ribosomal protein L34 Homo sapiens 177-204 35512188-3 2022 In a systematic analysis of 416 patients with t-MN and detailed prior exposure history, we found that TP53 mutations were significantly associated with prior treatment with thalidomide analogs, specifically lenalidomide. Thalidomide 173-184 tumor protein p53 Homo sapiens 102-106 35143878-7 2022 Most strikingly, burn injury-induced pain symptoms and the activation of astrocytes were significantly suppressed by TNF inhibitor Thalidomide. Thalidomide 131-142 tumor necrosis factor Mus musculus 117-120 34989322-0 2022 Esco2 and cohesin regulate CRL4 ubiquitin ligase ddb1 expression and thalidomide teratogenicity. Thalidomide 69-80 establishment of sister chromatid cohesion N-acetyltransferase 2 Homo sapiens 0-5 34989322-4 2022 Our results reveal that Esco2 and cohesin co-regulate the transcription of a component of CRL4 ubiquitin ligase through which thalidomide exerts teratogenic effects. Thalidomide 126-137 establishment of sister chromatid cohesion N-acetyltransferase 2 Homo sapiens 24-29 34989322-4 2022 Our results reveal that Esco2 and cohesin co-regulate the transcription of a component of CRL4 ubiquitin ligase through which thalidomide exerts teratogenic effects. Thalidomide 126-137 interleukin 17 receptor B Homo sapiens 90-94 35045330-1 2022 Cereblon (CRBN) mediates the teratogenic effect of thalidomide in zebrafish, chicken, and humans. Thalidomide 51-62 cereblon Danio rerio 10-14 35083901-8 2022 The TM/DX treatment affected the CD4+ T cell subsets without suppressing the total CD4+ T cell population. Thalidomide 4-6 CD4 molecule Homo sapiens 33-36 35083901-10 2022 Increase in cell counts and median fluorescence intensity on CD11c+CD85k+ with TM/DX were observed. Thalidomide 79-81 integrin subunit alpha X Homo sapiens 61-66 35083901-10 2022 Increase in cell counts and median fluorescence intensity on CD11c+CD85k+ with TM/DX were observed. Thalidomide 79-81 leukocyte immunoglobulin-like receptor, subfamily B, member 4A Mus musculus 67-72 35141236-9 2021 We found an association between polymorphisms in TLR1/rs4833095, TLR2/rs3804099, TLR4/rs1927914, and TLR6/rs5783810 with the dose variation of thalidomide in a time-dependent manner, i.e., the association with the genetic variant and the dose of the drug was different depending on the moment of the treatment evaluated. Thalidomide 143-154 toll like receptor 1 Homo sapiens 49-53 35141236-9 2021 We found an association between polymorphisms in TLR1/rs4833095, TLR2/rs3804099, TLR4/rs1927914, and TLR6/rs5783810 with the dose variation of thalidomide in a time-dependent manner, i.e., the association with the genetic variant and the dose of the drug was different depending on the moment of the treatment evaluated. Thalidomide 143-154 toll like receptor 2 Homo sapiens 65-69 35141236-9 2021 We found an association between polymorphisms in TLR1/rs4833095, TLR2/rs3804099, TLR4/rs1927914, and TLR6/rs5783810 with the dose variation of thalidomide in a time-dependent manner, i.e., the association with the genetic variant and the dose of the drug was different depending on the moment of the treatment evaluated. Thalidomide 143-154 toll like receptor 4 Homo sapiens 81-85 35141236-9 2021 We found an association between polymorphisms in TLR1/rs4833095, TLR2/rs3804099, TLR4/rs1927914, and TLR6/rs5783810 with the dose variation of thalidomide in a time-dependent manner, i.e., the association with the genetic variant and the dose of the drug was different depending on the moment of the treatment evaluated. Thalidomide 143-154 toll like receptor 6 Homo sapiens 101-105 34978798-1 2022 The thalidomide analogue lenalidomide (Len) is a clinical therapeutic that alters the substrate engagement of cereblon (CRBN), a substrate receptor for the CRL4 E3 ubiquitin ligase. Thalidomide 4-15 cereblon Homo sapiens 120-124 34978798-1 2022 The thalidomide analogue lenalidomide (Len) is a clinical therapeutic that alters the substrate engagement of cereblon (CRBN), a substrate receptor for the CRL4 E3 ubiquitin ligase. Thalidomide 4-15 interleukin 17 receptor B Homo sapiens 156-160 34978798-3 2022 pLen preserves the substrate degradation profile, phenotypic antiproliferative and immunomodulatory properties of Len, and enhances interactions with the thalidomide-binding domain of CRBN, as revealed by binding site mapping and molecular modeling. Thalidomide 154-165 cereblon Homo sapiens 184-188