PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 20103630-3 2010 Surface plasmon resonance revealed up to several thousand-fold increased affinities of TRAIL(mim/DR5)-receptor complexes on generation of divalent and trivalent molecules, the latter of which was achieved with a conformationally restricted adamantane core. Adamantane 240-250 TNF receptor superfamily member 10b Homo sapiens 97-100 20103630-5 2010 In tumor models derived from human embryonic kidney cells or primary foreskin fibroblasts, TRAIL(mim/DR5) peptides exerted a cancer cell-selective action that could synergize with resveratrol in a manner independent of p53. Resveratrol 180-191 TNF receptor superfamily member 10b Homo sapiens 101-104 19877156-8 2010 Concerning the molecular mechanism mediating the dexamethasone-suppression of the TRAIL activity in pre-osteoclasts, but not in leukemic cells, we found that dexamethasone induced a significant down-regulation of the surface levels of TRAIL-R2 in cells of the osteoclastic lineage but not in leukemic cells. Dexamethasone 49-62 TNF receptor superfamily member 10b Homo sapiens 235-243 19877156-8 2010 Concerning the molecular mechanism mediating the dexamethasone-suppression of the TRAIL activity in pre-osteoclasts, but not in leukemic cells, we found that dexamethasone induced a significant down-regulation of the surface levels of TRAIL-R2 in cells of the osteoclastic lineage but not in leukemic cells. Dexamethasone 158-171 TNF receptor superfamily member 10b Homo sapiens 235-243 19949310-5 2010 Western blot analyses revealed that CDODO-Me-12 and CDODO-Me-11 downregulated the levels of anti-apoptosis proteins, c-FLIP, XIAP and Mcl-1, without altering the protein levels of Bcl-2 and the death receptors DR4 and DR5. methyl 2-cyano-3,12-dioxooleana-1, 12-dien-30-oate 36-47 TNF receptor superfamily member 10b Homo sapiens 218-221 20113484-10 2010 Interestingly, knockdown of DR5, but not DR4, prevented GGTI298/TRAIL-induced IkappaBalpha and p-Akt reduction, suggesting that DR5 mediates reduction of IkappaBalpha and p-Akt induced by GGTI298/TRAIL. GGTI 298 56-63 TNF receptor superfamily member 10b Homo sapiens 28-31 20113484-10 2010 Interestingly, knockdown of DR5, but not DR4, prevented GGTI298/TRAIL-induced IkappaBalpha and p-Akt reduction, suggesting that DR5 mediates reduction of IkappaBalpha and p-Akt induced by GGTI298/TRAIL. GGTI 298 56-63 TNF receptor superfamily member 10b Homo sapiens 128-131 20113484-10 2010 Interestingly, knockdown of DR5, but not DR4, prevented GGTI298/TRAIL-induced IkappaBalpha and p-Akt reduction, suggesting that DR5 mediates reduction of IkappaBalpha and p-Akt induced by GGTI298/TRAIL. GGTI 298 188-195 TNF receptor superfamily member 10b Homo sapiens 28-31 20113484-10 2010 Interestingly, knockdown of DR5, but not DR4, prevented GGTI298/TRAIL-induced IkappaBalpha and p-Akt reduction, suggesting that DR5 mediates reduction of IkappaBalpha and p-Akt induced by GGTI298/TRAIL. GGTI 298 188-195 TNF receptor superfamily member 10b Homo sapiens 128-131 20037825-12 2009 The expression of DR4 instead of DR5 was significantly increased in the EGCG group. epigallocatechin gallate 72-76 TNF receptor superfamily member 10b Homo sapiens 33-36 19943170-6 2010 Salt supplementation substantially reduces the Cd-induced elevation of IAA oxidase activity, thereby maintaining auxin levels in Cd-stressed plants, as indicated by DR5::GUS expression. Salts 0-4 TNF receptor superfamily member 10b Homo sapiens 165-168 19943170-6 2010 Salt supplementation substantially reduces the Cd-induced elevation of IAA oxidase activity, thereby maintaining auxin levels in Cd-stressed plants, as indicated by DR5::GUS expression. Cadmium 47-49 TNF receptor superfamily member 10b Homo sapiens 165-168 20003459-0 2009 2-Deoxy-D-glucose enhances TRAIL-induced apoptosis in human melanoma cells through XBP-1-mediated up-regulation of TRAIL-R2. Deoxyglucose 0-17 TNF receptor superfamily member 10b Homo sapiens 115-123 20003459-9 2009 Treatment with 2-DG up-regulated TRAIL death receptors, in particular, TRAIL-R2, on the melanoma cell surface. Deoxyglucose 15-19 TNF receptor superfamily member 10b Homo sapiens 71-79 20003459-12 2009 Moreover, XBP-1, which is known to be transcriptionally regulated by ATF6 and functionally activated by IRE1 alpha, was found to play an important role in 2-DG-mediated transcriptional up-regulation of TRAIL-R2 in melanoma cells. Deoxyglucose 155-159 TNF receptor superfamily member 10b Homo sapiens 202-210 19039521-5 2009 Bortezomib enhanced the sensitivity of ovarian cancer cells and tissue explants to an apoptosis-inducing TRAIL receptor antibody by upregulating the TRAIL receptor DR5. Bortezomib 0-10 TNF receptor superfamily member 10b Homo sapiens 164-167 19903871-3 2009 We found that although AgNO(3) dramatically decreased root indole-3-acetic acid (IAA) responsiveness in inhibition of root elongation, promotion of DR5-beta-glucuronidase activity, and reduction of Aux/IAA protein levels, AVG had more mild effects. agno 23-27 TNF receptor superfamily member 10b Homo sapiens 148-151 19720557-0 2009 Esculetin enhances TRAIL-induced apoptosis through DR5 upregulation in human oral cancer SAS cells. esculetin 0-9 TNF receptor superfamily member 10b Homo sapiens 51-54 19720557-5 2009 Furthermore, treatment with esculetin significantly increased TRAIL-induced apoptosis in SAS cells and the TRAIL-sensitizing effect was blocked by DR5/Fc chimera protein. esculetin 28-37 TNF receptor superfamily member 10b Homo sapiens 147-150 19720557-6 2009 Our results indicate that esculetin enhances TRAIL-induced apoptosis primarily through upregulation of DR5. esculetin 26-35 TNF receptor superfamily member 10b Homo sapiens 103-106 19731037-3 2009 In this study, we demonstrate that both Dox and Dox-CPPs can increase the density of the TRAIL receptors DR4 and DR5 at the plasma membrane and moderately sensitize MDA-MB 231 cells to exogeneously added recombinant TRAIL, as has already been shown for other chemotherapeutic drugs. Doxorubicin 40-43 TNF receptor superfamily member 10b Homo sapiens 113-116 19903871-3 2009 We found that although AgNO(3) dramatically decreased root indole-3-acetic acid (IAA) responsiveness in inhibition of root elongation, promotion of DR5-beta-glucuronidase activity, and reduction of Aux/IAA protein levels, AVG had more mild effects. indoleacetic acid 59-79 TNF receptor superfamily member 10b Homo sapiens 148-151 19903871-3 2009 We found that although AgNO(3) dramatically decreased root indole-3-acetic acid (IAA) responsiveness in inhibition of root elongation, promotion of DR5-beta-glucuronidase activity, and reduction of Aux/IAA protein levels, AVG had more mild effects. indoleacetic acid 81-84 TNF receptor superfamily member 10b Homo sapiens 148-151 19734228-10 2009 We next demonstrated by surface plasmon resonance that phosphorothioate-modified CpG ODNs directly bound to either TRAIL or lexatumumab and then decreased their binding to DR5. Parathion 55-71 TNF receptor superfamily member 10b Homo sapiens 172-175 19666002-3 2009 Here, we investigated whether the apoptotic process triggered by apple procyanidins involving the up-regulation of TRAIL-death receptors DR4/DR5 at the cell surface was dependent on cell membrane lipid-raft formation. Proanthocyanidins 71-83 TNF receptor superfamily member 10b Homo sapiens 141-144 19734228-11 2009 Finally, NK cell lysis of a DR5-sensitive MM cell line (NCI-H929) through TRAIL was partially inhibited by phosphorothioate-modified CpG ODNs. Parathion 107-123 TNF receptor superfamily member 10b Homo sapiens 28-31 19638488-8 2009 None of the death receptors that commonly initiate the extrinsic pathway: FAS, TNF-R1, and TRAIL-R2 are found to be responsible for triggering the apoptosis cascade induced by DHA and EPA. Dihydroalprenolol 176-179 TNF receptor superfamily member 10b Homo sapiens 91-99 19682913-2 2009 We previously found that isoflavonoid enhanced TRAIL-induced apoptosis in TRAIL-resistant cells, which is achieved through up-regulation of death receptor 5 (DR5). isoflavonoid 25-37 TNF receptor superfamily member 10b Homo sapiens 140-156 19682913-2 2009 We previously found that isoflavonoid enhanced TRAIL-induced apoptosis in TRAIL-resistant cells, which is achieved through up-regulation of death receptor 5 (DR5). isoflavonoid 25-37 TNF receptor superfamily member 10b Homo sapiens 158-161 19682913-4 2009 Of the isolates, cardamomin (6), the most potent compound, enhanced the expressions of DR5 and DR4 and decreased the Bcl-xL level in TRAIL-resistant DLD1 cells. cardamonin 17-27 TNF receptor superfamily member 10b Homo sapiens 87-90 19654295-3 2009 We found that zerumbone potentiated TRAIL-induced apoptosis in human HCT116 colon cancer cells and that this correlated with the up-regulation of TRAIL death receptor (DR) 4 and DR5. zerumbone 14-23 TNF receptor superfamily member 10b Homo sapiens 178-181 19464267-5 2009 In addition, we revealed that treatment with DMNB, a specific inhibitor of DNA-PK, resulted in an increase of DR4/DR5 mRNA levels and their surface expression and a decrease of c-FLIP mRNA levels in K562 cells. 4,5-dimethoxy-2-nitrobenzaldehyde 45-49 TNF receptor superfamily member 10b Homo sapiens 114-117 19737941-0 2009 Suberoylanilide hydroxamic acid sensitizes human oral cancer cells to TRAIL-induced apoptosis through increase DR5 expression. Vorinostat 0-31 TNF receptor superfamily member 10b Homo sapiens 111-114 19737941-4 2009 Western blotting showed suberoylanilide hydroxamic acid increased Fas, Fas ligand, DR4, and DR5 protein expression and activated caspase-8 and caspase-9. Vorinostat 24-55 TNF receptor superfamily member 10b Homo sapiens 92-95 19737941-6 2009 Human recombinant DR5/Fc chimera protein but not Fas/Fc or DR4/Fc significantly inhibited apoptosis induced by suberoylanilide hydroxamic acid. Vorinostat 111-142 TNF receptor superfamily member 10b Homo sapiens 18-21 19737941-7 2009 These results suggest that suberoylanilide hydroxamic acid induces apoptosis mainly through activation of DR5/TRAIL death pathway. Vorinostat 27-58 TNF receptor superfamily member 10b Homo sapiens 106-109 19654295-11 2009 Overall, our results show that zerumbone can potentiate TRAIL-induced apoptosis through the reactive oxygen species-mediated activation of extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase leading to DR4 and DR5 induction and resulting in enhancement of the anticancer effects of TRAIL. zerumbone 31-40 TNF receptor superfamily member 10b Homo sapiens 241-244 19654295-11 2009 Overall, our results show that zerumbone can potentiate TRAIL-induced apoptosis through the reactive oxygen species-mediated activation of extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase leading to DR4 and DR5 induction and resulting in enhancement of the anticancer effects of TRAIL. Reactive Oxygen Species 92-115 TNF receptor superfamily member 10b Homo sapiens 241-244 19638488-8 2009 None of the death receptors that commonly initiate the extrinsic pathway: FAS, TNF-R1, and TRAIL-R2 are found to be responsible for triggering the apoptosis cascade induced by DHA and EPA. Eicosapentaenoic Acid 184-187 TNF receptor superfamily member 10b Homo sapiens 91-99 19343733-7 2009 In addition, increased expression of death receptor 5 protein was observed further supporting the proposed mechanism of apoptosis induction via the extrinsic pathway in 2-ME-exposed oesophageal carcinoma cells. 2-Methoxyestradiol 169-173 TNF receptor superfamily member 10b Homo sapiens 37-53 19468286-0 2009 An agonistic monoclonal antibody against DR5 induces ROS production, sustained JNK activation and Endo G release in Jurkat leukemia cells. Reactive Oxygen Species 53-56 TNF receptor superfamily member 10b Homo sapiens 41-44 19468286-1 2009 We have previously reported that AD5-10, a novel agonistic monoclonal antibody against DR5, possessed a strong cytotoxic activity in various tumor cells, via induction of caspase-dependent and -independent signaling pathways. ad5-10 33-39 TNF receptor superfamily member 10b Homo sapiens 87-90 19493960-6 2009 An analysis of the DR5 reporter in vam3-4 indicated that VAM3 is involved in the proper pattern formation of auxin maxima in the leaf primordium. vam3-4 35-41 TNF receptor superfamily member 10b Homo sapiens 19-22 19493960-6 2009 An analysis of the DR5 reporter in vam3-4 indicated that VAM3 is involved in the proper pattern formation of auxin maxima in the leaf primordium. vam3 57-61 TNF receptor superfamily member 10b Homo sapiens 19-22 19594141-2 2009 In particular, cardenolide glycosides (1 and 2) from T. peruviana were shown to have a significant reversal effect on TRAIL resistance in human gastric adenocarcinoma cells, and real-time PCR showed that thevefolin (2) enhanced mRNA expression of death receptor 4 (DR4) and DR5. cardenolide glycosides 15-37 TNF receptor superfamily member 10b Homo sapiens 274-277 19345731-0 2009 Withaferin A sensitizes TRAIL-induced apoptosis through reactive oxygen species-mediated up-regulation of death receptor 5 and down-regulation of c-FLIP. withaferin A 0-12 TNF receptor superfamily member 10b Homo sapiens 106-122 19345731-0 2009 Withaferin A sensitizes TRAIL-induced apoptosis through reactive oxygen species-mediated up-regulation of death receptor 5 and down-regulation of c-FLIP. Reactive Oxygen Species 56-79 TNF receptor superfamily member 10b Homo sapiens 106-122 19345731-5 2009 Interestingly, a Wit A-induced increase in ROS levels preceded the up-regulation of CHOP and DR5. Reactive Oxygen Species 43-46 TNF receptor superfamily member 10b Homo sapiens 93-96 19345731-6 2009 The involvement of ROS in CHOP-mediated DR5 up-regulation was confirmed by the result that pretreatment with an antioxidant, NAC or catalase, inhibited Wit A-induced up-regulation of both CHOP and DR5. Reactive Oxygen Species 19-22 TNF receptor superfamily member 10b Homo sapiens 40-43 19345731-6 2009 The involvement of ROS in CHOP-mediated DR5 up-regulation was confirmed by the result that pretreatment with an antioxidant, NAC or catalase, inhibited Wit A-induced up-regulation of both CHOP and DR5. Reactive Oxygen Species 19-22 TNF receptor superfamily member 10b Homo sapiens 197-200 20641892-11 2004 Using HGS-ETR2 labeled with radioactive indium ((111)In) ((111)In -EC-HGS-ETR2), the investigators determined that the activity of HGS-ETR2 was increased because the pretreatment with paclitaxel upregulated the expression of TRAIL-R2 in Colo205 cell xenograft tumors in nude mice. Paclitaxel 184-194 TNF receptor superfamily member 10b Homo sapiens 225-233 19272388-7 2009 DR5-enhanced green fluorescent protein internalization was dependent on a dileucine-based internalization motif. dileucine 74-83 TNF receptor superfamily member 10b Homo sapiens 0-3 20641185-11 2004 Using HGS-ETR2 labeled with radioactive technetium ((99m)Tc) ((99m)Tc-EC-HGS-ETR2), the investigators determined that the activity of HGS-ETR2 was increased because the pretreatment with paclitaxel upregulated the expression of TRAIL-R2 in Colo205 cell xenograft tumors in nude mice. Paclitaxel 187-197 TNF receptor superfamily member 10b Homo sapiens 228-236 19037087-0 2009 Rottlerin induces apoptosis via death receptor 5 (DR5) upregulation through CHOP-dependent and PKC delta-independent mechanism in human malignant tumor cells. rottlerin 0-9 TNF receptor superfamily member 10b Homo sapiens 32-48 19037087-7 2009 These results suggest that rottlerin induces upregulation of DR5 via PKC delta-independent pathway. rottlerin 27-36 TNF receptor superfamily member 10b Homo sapiens 61-64 19264955-9 2009 Additionally, fisetin caused an increase in the protein levels of cleaved caspase-8, Fas ligand, death receptor 5, and TNF-related apoptosis-inducing ligand, and the caspase-8 inhibitor Z-IETD-FMK suppressed fisetin-induced apoptosis and the activation of caspase-3. fisetin 14-21 TNF receptor superfamily member 10b Homo sapiens 97-113 19037087-0 2009 Rottlerin induces apoptosis via death receptor 5 (DR5) upregulation through CHOP-dependent and PKC delta-independent mechanism in human malignant tumor cells. rottlerin 0-9 TNF receptor superfamily member 10b Homo sapiens 50-53 19037087-9 2009 Thus, DR5-mediated apoptosis, which is induced by rottlerin alone or by the combined treatment with rottlerin and TRAIL, may offer a new therapeutic strategy against cancer. rottlerin 50-59 TNF receptor superfamily member 10b Homo sapiens 6-9 19037087-9 2009 Thus, DR5-mediated apoptosis, which is induced by rottlerin alone or by the combined treatment with rottlerin and TRAIL, may offer a new therapeutic strategy against cancer. rottlerin 100-109 TNF receptor superfamily member 10b Homo sapiens 6-9 19037087-3 2009 We found that treatment with rottlerin significantly induces DR5 expression both at its messenger RNA and protein levels. rottlerin 29-38 TNF receptor superfamily member 10b Homo sapiens 61-64 19037087-4 2009 Downregulation of DR5 expression with small-interfering RNA (siRNA) efficiently attenuated rottlerin-induced apoptosis, showing that the critical role of DR5 in this cell death. rottlerin 91-100 TNF receptor superfamily member 10b Homo sapiens 18-21 19037087-4 2009 Downregulation of DR5 expression with small-interfering RNA (siRNA) efficiently attenuated rottlerin-induced apoptosis, showing that the critical role of DR5 in this cell death. rottlerin 91-100 TNF receptor superfamily member 10b Homo sapiens 154-157 19037087-5 2009 Rottlerin-induced DR5 upregulation was accompanied by CCAAT/enhancer-binding protein-homologous protein (CHOP) protein expression and rottlerin-induced increase of DR5 promoter activity was diminished by mutation of a CHOP-binding site of DR5 promoter. rottlerin 0-9 TNF receptor superfamily member 10b Homo sapiens 18-21 19037087-5 2009 Rottlerin-induced DR5 upregulation was accompanied by CCAAT/enhancer-binding protein-homologous protein (CHOP) protein expression and rottlerin-induced increase of DR5 promoter activity was diminished by mutation of a CHOP-binding site of DR5 promoter. rottlerin 0-9 TNF receptor superfamily member 10b Homo sapiens 164-167 19037087-5 2009 Rottlerin-induced DR5 upregulation was accompanied by CCAAT/enhancer-binding protein-homologous protein (CHOP) protein expression and rottlerin-induced increase of DR5 promoter activity was diminished by mutation of a CHOP-binding site of DR5 promoter. rottlerin 0-9 TNF receptor superfamily member 10b Homo sapiens 164-167 19037087-5 2009 Rottlerin-induced DR5 upregulation was accompanied by CCAAT/enhancer-binding protein-homologous protein (CHOP) protein expression and rottlerin-induced increase of DR5 promoter activity was diminished by mutation of a CHOP-binding site of DR5 promoter. rottlerin 134-143 TNF receptor superfamily member 10b Homo sapiens 164-167 19037087-5 2009 Rottlerin-induced DR5 upregulation was accompanied by CCAAT/enhancer-binding protein-homologous protein (CHOP) protein expression and rottlerin-induced increase of DR5 promoter activity was diminished by mutation of a CHOP-binding site of DR5 promoter. rottlerin 134-143 TNF receptor superfamily member 10b Homo sapiens 164-167 19037087-6 2009 Although rottlerin is known to be as an inhibitor of novel isoforms of protein kinase C (PKC), specifically PKC delta, not only suppression of PKC delta expression by siRNA but also overexpression of wild-type-PKC delta or dominant-negative-PKC delta did not affect the rottlerin-mediated induction of DR5 in our study. rottlerin 9-18 TNF receptor superfamily member 10b Homo sapiens 302-305 19379558-3 2009 The results showed that both TRAIL and DR5 protein and mRNA expressions in NB4, U937 and Jurkat cells increased after treated with sodium butyrate (SB) and in time-dependent manner. Butyric Acid 131-146 TNF receptor superfamily member 10b Homo sapiens 39-42 19351839-4 2009 Pretreatment of several melanoma lines just before gamma-irradiation with the inhibitor of ATM kinase KU-55933 suppressed p53 and nuclear factor-kappaB (NF-kappaB) activation but notably increased radiation-induced DR5 surface expression, down-regulated cFLIP (caspase-8 inhibitor) levels, and substantially enhanced exogenous TRAIL-induced apoptosis. 2-morpholin-4-yl-6-thianthren-1-yl-pyran-4-one 102-110 TNF receptor superfamily member 10b Homo sapiens 215-218 19379558-3 2009 The results showed that both TRAIL and DR5 protein and mRNA expressions in NB4, U937 and Jurkat cells increased after treated with sodium butyrate (SB) and in time-dependent manner. Butyric Acid 148-150 TNF receptor superfamily member 10b Homo sapiens 39-42 19276256-9 2009 Cisplatin significantly increased TRAIL-R2 expression at both the mRNA and the protein levels. Cisplatin 0-9 TNF receptor superfamily member 10b Homo sapiens 34-42 19223550-3 2009 We provide evidence that LY30-induced increase in intracellular H(2)O(2) up-regulates the expression of TRAIL receptors (DR4 and DR5) in SHEP-1 cells by activating mitogen-activated protein kinases, resulting in a significant amplification of TRAIL-mediated caspase-8 processing and activity, cytosolic translocation of cytochrome c, and cell death. LY 303511 25-29 TNF receptor superfamily member 10b Homo sapiens 129-132 19223002-4 2009 Oxaliplatin promoted death receptor 4 (DR4) and death receptor 5 (DR5) clustering into aggregated lipid rafts, while the cholesterol-sequestering agent nystatin partially prevented lipid raft aggregation, DR4 and DR5 clustering, and reduced apoptosis. Oxaliplatin 0-11 TNF receptor superfamily member 10b Homo sapiens 48-64 19223002-4 2009 Oxaliplatin promoted death receptor 4 (DR4) and death receptor 5 (DR5) clustering into aggregated lipid rafts, while the cholesterol-sequestering agent nystatin partially prevented lipid raft aggregation, DR4 and DR5 clustering, and reduced apoptosis. Oxaliplatin 0-11 TNF receptor superfamily member 10b Homo sapiens 66-69 19276284-4 2009 DESIGN: Effect of cisplatin alone or in combination with rhTRAIL or rhTRAIL-DR5 on DR5 surface expression, apoptosis, and cell survival of A2780 was measured. Cisplatin 18-27 TNF receptor superfamily member 10b Homo sapiens 83-86 19276284-7 2009 RESULTS: Cisplatin strongly enhanced DR5 surface expression. Cisplatin 9-18 TNF receptor superfamily member 10b Homo sapiens 37-40 19276284-8 2009 Both rhTRAIL and rhTRAIL-DR5 in combination with cisplatin induced high levels of caspase-3 activation, apoptosis, and cell kill, with rhTRAIL-DR5 being most potent. Cisplatin 49-58 TNF receptor superfamily member 10b Homo sapiens 143-146 19276284-10 2009 Intraperitoneal administration of rhTRAIL-DR5 delayed A2780 tumor progression, reflected in a mean light reduction of 68.3% (P = 0.015), whereas rhTRAIL or rhTRAIL-DR5 plus cisplatin resulted in 85% (P = 0.003) and 97% (P = 0.002) reduction compared with A2780 tumor progression in vehicle-treated animals. Cisplatin 173-182 TNF receptor superfamily member 10b Homo sapiens 42-45 19276284-12 2009 CONCLUSION: rhTRAIL-DR5 was superior over rhTRAIL in vitro and in vivo against DR5-expressing ovarian cancer also in combination with cisplatin. Cisplatin 134-143 TNF receptor superfamily member 10b Homo sapiens 20-23 19223002-4 2009 Oxaliplatin promoted death receptor 4 (DR4) and death receptor 5 (DR5) clustering into aggregated lipid rafts, while the cholesterol-sequestering agent nystatin partially prevented lipid raft aggregation, DR4 and DR5 clustering, and reduced apoptosis. Nystatin 152-160 TNF receptor superfamily member 10b Homo sapiens 213-216 19223550-3 2009 We provide evidence that LY30-induced increase in intracellular H(2)O(2) up-regulates the expression of TRAIL receptors (DR4 and DR5) in SHEP-1 cells by activating mitogen-activated protein kinases, resulting in a significant amplification of TRAIL-mediated caspase-8 processing and activity, cytosolic translocation of cytochrome c, and cell death. Hydrogen Peroxide 64-72 TNF receptor superfamily member 10b Homo sapiens 129-132 19223550-4 2009 Involvement of the death receptors was further confirmed by the ability of blocking antibodies against DR4 and/or DR5 to inhibit LY30-induced TRAIL sensitization. LY 303511 129-133 TNF receptor superfamily member 10b Homo sapiens 114-117 19223550-6 2009 Finally, small interfering RNA-mediated gene silencing of JNK and ERK inhibited LY30-induced increase in surface expression of DR4 and DR5, respectively. LY 303511 80-84 TNF receptor superfamily member 10b Homo sapiens 135-138 19230044-9 2009 Cytometry analysis revealed higher levels of DR5 expression on the surfaces of brefeldin-A-untreated cells than on the surfaces of brefeldin-A-treated cells, but brefeldin A treatment did not affect the total DR5 expression levels. Brefeldin A 79-90 TNF receptor superfamily member 10b Homo sapiens 45-48 19212626-10 2009 DDP facilitated the induction of expression of DR4 and DR5 significantly in cell line (P<0.05), but 5-FU influenced only the expression of DR5 significantly. Fluorouracil 103-107 TNF receptor superfamily member 10b Homo sapiens 142-145 18852135-4 2008 In addition, curcumin treatment of Burkitt"s lymphoma cell lines also causes up-regulation of DR5; however, this up-regulation does not result in apoptosis. Curcumin 13-21 TNF receptor superfamily member 10b Homo sapiens 94-97 19124248-2 2009 On screening for compounds that enhance DR5 expression using a luciferase assay with DLD-1/SacI, we previously identified 4"-demethyltoxicarol isoflavone (1) isolated from the leaves of Millettia brandisiana. 4'-demethyltoxicarol isoflavone 122-153 TNF receptor superfamily member 10b Homo sapiens 40-43 19124248-7 2009 Furthermore, a MeOH extract of the bark of Ardisia colorata markedly enhanced DR5 activity in this screening system. Methanol 15-19 TNF receptor superfamily member 10b Homo sapiens 78-81 18978361-8 2009 RESULTS: Tunicamycin increases both DR5 mRNA (P < 0.005) and TRAIL-induced apoptosis (P < 0.0001) in ESC. Tunicamycin 9-20 TNF receptor superfamily member 10b Homo sapiens 36-39 18978361-10 2009 Transfection with DR5 siRNA suppressed the tunicamycin-induced increase in DR5 mRNA and abrogated the up-regulation of TRAIL-induced apoptosis by tunicamycin. Tunicamycin 43-54 TNF receptor superfamily member 10b Homo sapiens 18-21 18978361-10 2009 Transfection with DR5 siRNA suppressed the tunicamycin-induced increase in DR5 mRNA and abrogated the up-regulation of TRAIL-induced apoptosis by tunicamycin. Tunicamycin 43-54 TNF receptor superfamily member 10b Homo sapiens 75-78 18978361-10 2009 Transfection with DR5 siRNA suppressed the tunicamycin-induced increase in DR5 mRNA and abrogated the up-regulation of TRAIL-induced apoptosis by tunicamycin. Tunicamycin 146-157 TNF receptor superfamily member 10b Homo sapiens 18-21 18778787-0 2009 Nitric oxide sensitizes tumor cells to TRAIL-induced apoptosis via inhibition of the DR5 transcription repressor Yin Yang 1. Nitric Oxide 0-12 TNF receptor superfamily member 10b Homo sapiens 85-88 18778787-1 2009 Treatment of TRAIL-resistant tumor cells with the nitric oxide donor DETANONOate sensitizes the tumor cells to TRAIL-induced apoptosis concomitantly with DR5 upregulation. Nitric Oxide 50-62 TNF receptor superfamily member 10b Homo sapiens 154-157 18778787-1 2009 Treatment of TRAIL-resistant tumor cells with the nitric oxide donor DETANONOate sensitizes the tumor cells to TRAIL-induced apoptosis concomitantly with DR5 upregulation. 2,2'-(hydroxynitrosohydrazono)bis-ethanamine 69-80 TNF receptor superfamily member 10b Homo sapiens 154-157 18778787-2 2009 The mechanism of sensitization was examined based on the hypothesis that DETANONOate inhibits a transcription repressor Yin Yang 1 (YY1) that negatively regulates DR5 transcription. 2,2'-(hydroxynitrosohydrazono)bis-ethanamine 73-84 TNF receptor superfamily member 10b Homo sapiens 163-166 18778787-3 2009 Treatment of the prostate carcinoma cell lines with DETANONOate inhibited both NF-kappaB and YY1 DNA-binding activities concomitantly with upregulation of DR5 expression. 2,2'-(hydroxynitrosohydrazono)bis-ethanamine 52-63 TNF receptor superfamily member 10b Homo sapiens 155-158 20209078-7 2009 In Smac-deficient cells, although TG-induced DR5 upregulation is not affected, activation of caspases 3, 9 and 8 is affected. Thapsigargin 34-36 TNF receptor superfamily member 10b Homo sapiens 45-48 20419043-6 2009 Sulindac sulfide-induced apoptosis is coupled with upregulation of death receptor 5 (DR5), and activation of caspases 3, 9 and 8 in Smac-proficient cells. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 67-83 20419043-6 2009 Sulindac sulfide-induced apoptosis is coupled with upregulation of death receptor 5 (DR5), and activation of caspases 3, 9 and 8 in Smac-proficient cells. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 85-88 20419043-7 2009 In Smac-deficient cells, although sulindac sulfide-induced DR5 upregulation is not altered, activation of caspases 3, 9 and 8 is affected. sulindac sulfide 34-50 TNF receptor superfamily member 10b Homo sapiens 59-62 19000651-4 2008 Nelfinavir sensitized ovarian cancer cells to treatment with an apoptosis-inducing TRAIL receptor antibody due to upregulation of the TRAIL receptor DR5 as shown by RT-PCR and FACScan analysis. Nelfinavir 0-10 TNF receptor superfamily member 10b Homo sapiens 149-152 19074830-6 2008 Allopurinol up-regulated the expression of a proapoptotic TRAIL receptor, death receptor 5 (DR5). Allopurinol 0-11 TNF receptor superfamily member 10b Homo sapiens 74-90 19074830-6 2008 Allopurinol up-regulated the expression of a proapoptotic TRAIL receptor, death receptor 5 (DR5). Allopurinol 0-11 TNF receptor superfamily member 10b Homo sapiens 92-95 19074830-7 2008 Allopurinol increased DR5 protein, mRNA, and promoter activity. Allopurinol 0-11 TNF receptor superfamily member 10b Homo sapiens 22-25 19074830-8 2008 Using DR5 small interfering RNA (siRNA), we showed that allopurinol-mediated DR5 up-regulation contributed to the enhancement of TRAIL effect by allopurinol. Allopurinol 56-67 TNF receptor superfamily member 10b Homo sapiens 6-9 19074830-8 2008 Using DR5 small interfering RNA (siRNA), we showed that allopurinol-mediated DR5 up-regulation contributed to the enhancement of TRAIL effect by allopurinol. Allopurinol 56-67 TNF receptor superfamily member 10b Homo sapiens 77-80 19074830-8 2008 Using DR5 small interfering RNA (siRNA), we showed that allopurinol-mediated DR5 up-regulation contributed to the enhancement of TRAIL effect by allopurinol. Allopurinol 145-156 TNF receptor superfamily member 10b Homo sapiens 6-9 19074830-8 2008 Using DR5 small interfering RNA (siRNA), we showed that allopurinol-mediated DR5 up-regulation contributed to the enhancement of TRAIL effect by allopurinol. Allopurinol 145-156 TNF receptor superfamily member 10b Homo sapiens 77-80 19074830-9 2008 Furthermore, we examined the mechanism of allopurinol-mediated DR5 up-regulation. Allopurinol 42-53 TNF receptor superfamily member 10b Homo sapiens 63-66 19074830-10 2008 DR5 promoter activity induced by allopurinol was diminished by a mutation of a CAAT/enhancer binding protein homologous protein (CHOP)-binding site. Allopurinol 33-44 TNF receptor superfamily member 10b Homo sapiens 0-3 19074830-11 2008 In addition, allopurinol also increased CHOP expression, suggesting that allopurinol induced DR5 expression via CHOP. Allopurinol 13-24 TNF receptor superfamily member 10b Homo sapiens 93-96 19074830-11 2008 In addition, allopurinol also increased CHOP expression, suggesting that allopurinol induced DR5 expression via CHOP. Allopurinol 73-84 TNF receptor superfamily member 10b Homo sapiens 93-96 19010914-2 2008 The rationale for using such a combination is that perifosine was recently described to increase TRAIL-R2 receptor expression and decrease the cellular FLICE-inhibitory protein (cFLIP) in human lung cancer cell lines. perifosine 51-61 TNF receptor superfamily member 10b Homo sapiens 97-105 19010914-4 2008 Perifosine, at concentrations below IC(50), dephosphorylated Akt and increased TRAIL-R2 levels, as shown by Western blot, reverse transcription-PCR, and flow cytometric analysis. perifosine 0-10 TNF receptor superfamily member 10b Homo sapiens 79-87 19010914-7 2008 Up-regulation of TRAIL-R2 expression was dependent on a protein kinase Calpha/c-Jun-NH(2)-kinase 2/c-Jun signaling pathway activated by perifosine through reactive oxygen species production. perifosine 136-146 TNF receptor superfamily member 10b Homo sapiens 17-25 19010914-7 2008 Up-regulation of TRAIL-R2 expression was dependent on a protein kinase Calpha/c-Jun-NH(2)-kinase 2/c-Jun signaling pathway activated by perifosine through reactive oxygen species production. Reactive Oxygen Species 155-178 TNF receptor superfamily member 10b Homo sapiens 17-25 19010914-8 2008 Perifosine also synergized with TRAIL in primary AML cells displaying constitutive activation of the Akt pathway by inducing apoptosis, Akt dephosphorylation, TRAIL-R2 up-regulation, cFLIP-L and XIAP down-regulation, and c-Jun phosphorylation. perifosine 0-10 TNF receptor superfamily member 10b Homo sapiens 159-167 18974136-12 2008 Interestingly, baicalein induced reactive oxygen species (ROS) and a ROS scavenger prevented DR5 expression and TRAIL sensitization in PC3 but not SW480 cells. baicalein 15-24 TNF receptor superfamily member 10b Homo sapiens 93-96 18974136-12 2008 Interestingly, baicalein induced reactive oxygen species (ROS) and a ROS scavenger prevented DR5 expression and TRAIL sensitization in PC3 but not SW480 cells. Reactive Oxygen Species 69-72 TNF receptor superfamily member 10b Homo sapiens 93-96 18980980-6 2008 Sulforaphane-induced apoptosis in PC-3 cells correlated with the generation of intracellular reactive oxygen species (ROS), collapse of mitochondrial membrane potential, activation of caspase-3 and caspase-9, and up-regulation of DR4 and DR5. sulforaphane 0-12 TNF receptor superfamily member 10b Homo sapiens 238-241 18980980-10 2008 Sulforaphane up-regulated the expressions of TRAIL-R1/DR4, TRAIL-R2/DR5, Bax and Bak and inhibited the activation of nuclear factor-kappaB P13K/AKT and MEK/ERK pathways in tumor tissues. sulforaphane 0-12 TNF receptor superfamily member 10b Homo sapiens 59-67 18980980-10 2008 Sulforaphane up-regulated the expressions of TRAIL-R1/DR4, TRAIL-R2/DR5, Bax and Bak and inhibited the activation of nuclear factor-kappaB P13K/AKT and MEK/ERK pathways in tumor tissues. sulforaphane 0-12 TNF receptor superfamily member 10b Homo sapiens 68-71 18680719-4 2008 DR5 but not DR4 siRNA efficiently blocked apoptosis induced by the co-treatment with kaempferol and TRAIL, indicating that DR5 up-regulation by kaempferol helps to enhance TRAIL actions. kaempferol 85-95 TNF receptor superfamily member 10b Homo sapiens 0-3 18680719-4 2008 DR5 but not DR4 siRNA efficiently blocked apoptosis induced by the co-treatment with kaempferol and TRAIL, indicating that DR5 up-regulation by kaempferol helps to enhance TRAIL actions. kaempferol 85-95 TNF receptor superfamily member 10b Homo sapiens 123-126 18680719-4 2008 DR5 but not DR4 siRNA efficiently blocked apoptosis induced by the co-treatment with kaempferol and TRAIL, indicating that DR5 up-regulation by kaempferol helps to enhance TRAIL actions. kaempferol 144-154 TNF receptor superfamily member 10b Homo sapiens 0-3 18680719-4 2008 DR5 but not DR4 siRNA efficiently blocked apoptosis induced by the co-treatment with kaempferol and TRAIL, indicating that DR5 up-regulation by kaempferol helps to enhance TRAIL actions. kaempferol 144-154 TNF receptor superfamily member 10b Homo sapiens 123-126 18636537-6 2008 Additionally, the induction of death receptor 5 (DR5), which contributes to Adriamycin-induced cytotoxicity, was not affected by NF-kappaB blockage. Doxorubicin 76-86 TNF receptor superfamily member 10b Homo sapiens 31-47 18636537-6 2008 Additionally, the induction of death receptor 5 (DR5), which contributes to Adriamycin-induced cytotoxicity, was not affected by NF-kappaB blockage. Doxorubicin 76-86 TNF receptor superfamily member 10b Homo sapiens 49-52 18852139-4 2008 Both DIM-C-pPhBr and DIM-C-pPhF resembled the classic ER stress inducer thapsigargin in pancreatic cancer cells and activated ER stress markers, such as glucose-related protein 78 and the c-Jun NH(2) kinase pathway, resulting in the induction of CCAAT/enhancer-binding protein homologous protein, death receptor 5, and the extrinsic apoptotic pathway. dim-c-pphbr 5-16 TNF receptor superfamily member 10b Homo sapiens 297-313 18852139-4 2008 Both DIM-C-pPhBr and DIM-C-pPhF resembled the classic ER stress inducer thapsigargin in pancreatic cancer cells and activated ER stress markers, such as glucose-related protein 78 and the c-Jun NH(2) kinase pathway, resulting in the induction of CCAAT/enhancer-binding protein homologous protein, death receptor 5, and the extrinsic apoptotic pathway. dim-c-pphf 21-31 TNF receptor superfamily member 10b Homo sapiens 297-313 18852146-0 2008 15-deoxy-Delta12,14-prostaglandin J2 up-regulates death receptor 5 gene expression in HCT116 cells: involvement of reactive oxygen species and C/EBP homologous transcription factor gene transcription. 14-prostaglandin j2 17-36 TNF receptor superfamily member 10b Homo sapiens 50-66 18852146-1 2008 Although 15-deoxy-Delta(12,14)-prostaglandin J(2) (15dPGJ(2)) was reported to up-regulate death receptor 5 (DR5) protein expression and sensitize TRAIL-induced cytotoxicity, its action mechanism remains unclear. 15-deoxy-delta(12,14)-prostaglandin j 9-46 TNF receptor superfamily member 10b Homo sapiens 90-106 18852146-1 2008 Although 15-deoxy-Delta(12,14)-prostaglandin J(2) (15dPGJ(2)) was reported to up-regulate death receptor 5 (DR5) protein expression and sensitize TRAIL-induced cytotoxicity, its action mechanism remains unclear. 15-deoxy-delta(12,14)-prostaglandin j 9-46 TNF receptor superfamily member 10b Homo sapiens 108-111 18852146-6 2008 A mechanistic study showed the contributions of reactive oxygen species (ROS) and intracellular calcium in CHOP and DR5 gene up-regulation. Reactive Oxygen Species 48-71 TNF receptor superfamily member 10b Homo sapiens 116-119 18852146-6 2008 A mechanistic study showed the contributions of reactive oxygen species (ROS) and intracellular calcium in CHOP and DR5 gene up-regulation. Reactive Oxygen Species 73-76 TNF receptor superfamily member 10b Homo sapiens 116-119 18852146-6 2008 A mechanistic study showed the contributions of reactive oxygen species (ROS) and intracellular calcium in CHOP and DR5 gene up-regulation. Calcium 96-103 TNF receptor superfamily member 10b Homo sapiens 116-119 18852146-11 2008 In summary, the effect of up-regulation of DR5 by 15dPGJ(2) in colon cancer cells is independent of PPAR-gamma and p53 but relies on CHOP induction through gene transcription involving ROS and calcium. Reactive Oxygen Species 185-188 TNF receptor superfamily member 10b Homo sapiens 43-46 19174554-0 2009 Mapatumumab and lexatumumab induce apoptosis in TRAIL-R1 and TRAIL-R2 antibody-resistant NSCLC cell lines when treated in combination with bortezomib. Bortezomib 139-149 TNF receptor superfamily member 10b Homo sapiens 61-69 20209078-2 2009 We have previously reported that TG induces apoptosis by engaging the death receptor 5 (DR5) and the intrinsic pathways. Thapsigargin 33-35 TNF receptor superfamily member 10b Homo sapiens 70-86 20209078-2 2009 We have previously reported that TG induces apoptosis by engaging the death receptor 5 (DR5) and the intrinsic pathways. Thapsigargin 33-35 TNF receptor superfamily member 10b Homo sapiens 88-91 20209078-6 2009 For example, ER stress inducing agent TG upregulates DR5, and activates caspases 3, 9 and 8 in Smac-proficient cells. Thapsigargin 38-40 TNF receptor superfamily member 10b Homo sapiens 53-56 18980244-0 2008 Quercetin sensitizes human hepatoma cells to TRAIL-induced apoptosis via Sp1-mediated DR5 up-regulation and proteasome-mediated c-FLIPS down-regulation. Quercetin 0-9 TNF receptor superfamily member 10b Homo sapiens 86-89 18980244-5 2008 We found that quercetin treatment of HCC cells significantly up-regulated the mRNA and protein levels of DR5, a death receptor of TRAIL, in a transcription factor Sp1-dependent manner. Quercetin 14-23 TNF receptor superfamily member 10b Homo sapiens 105-108 18980244-7 2008 Finally, administration of small interfering RNA against DR5 or overexpression of c-FLIPS, but not c-FLIPL, significantly attenuated quercetin-stimulated TRAIL-induced apoptosis. Quercetin 133-142 TNF receptor superfamily member 10b Homo sapiens 57-60 18980244-8 2008 Collectively, these findings show that quercetin recovers TRAIL sensitivity in various HCC cells via up-regulation of DR5 and down-regulation of c-FLIPS. Quercetin 39-48 TNF receptor superfamily member 10b Homo sapiens 118-121 18852146-11 2008 In summary, the effect of up-regulation of DR5 by 15dPGJ(2) in colon cancer cells is independent of PPAR-gamma and p53 but relies on CHOP induction through gene transcription involving ROS and calcium. Calcium 193-200 TNF receptor superfamily member 10b Homo sapiens 43-46 18583568-3 2008 In HMCLs carrying a nonfunctional p53, cotreatment with PD strikingly elevates the (DR4 + DR5)/(DcR1 + DcR2) tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptors ratio and caspase-8 activation of ATO-treated cells. Arsenic Trioxide 223-226 TNF receptor superfamily member 10b Homo sapiens 90-93 18599488-0 2008 ABT-737 induces expression of the death receptor 5 and sensitizes human cancer cells to TRAIL-induced apoptosis. ABT-737 0-7 TNF receptor superfamily member 10b Homo sapiens 34-50 18794136-11 2008 Furthermore, salubrinal, an inhibitor of ER stress-induced apoptosis, inhibited JNK activation and up-regulation of CHOP and DR5 by CDDO-Me and protected cells from CDDO-Me-induced apoptosis. salubrinal 13-23 TNF receptor superfamily member 10b Homo sapiens 125-128 18794136-11 2008 Furthermore, salubrinal, an inhibitor of ER stress-induced apoptosis, inhibited JNK activation and up-regulation of CHOP and DR5 by CDDO-Me and protected cells from CDDO-Me-induced apoptosis. bardoxolone methyl 132-139 TNF receptor superfamily member 10b Homo sapiens 125-128 18794136-12 2008 Thus, ER stress seems to be important for CDDO-Me-induced JNK activation, CHOP and DR5 up-regulation, and apoptosis. bardoxolone methyl 42-49 TNF receptor superfamily member 10b Homo sapiens 83-86 18794136-13 2008 Collectively, we conclude that CDDO-Me triggers ER stress, leading to JNK-dependent, CHOP-mediated DR5 up-regulation and apoptosis. bardoxolone methyl 31-38 TNF receptor superfamily member 10b Homo sapiens 99-102 18794136-0 2008 Coupling of endoplasmic reticulum stress to CDDO-Me-induced up-regulation of death receptor 5 via a CHOP-dependent mechanism involving JNK activation. bardoxolone methyl 44-51 TNF receptor superfamily member 10b Homo sapiens 77-93 18794136-2 2008 We previously showed that CDDO-Me induces c-Jun NH(2)-terminal kinase (JNK)-dependent death receptor 5 (DR5) expression and augments death receptor-induced apoptosis. bardoxolone methyl 26-33 TNF receptor superfamily member 10b Homo sapiens 86-102 18794136-2 2008 We previously showed that CDDO-Me induces c-Jun NH(2)-terminal kinase (JNK)-dependent death receptor 5 (DR5) expression and augments death receptor-induced apoptosis. bardoxolone methyl 26-33 TNF receptor superfamily member 10b Homo sapiens 104-107 18794136-3 2008 The current study focused on addressing how CDDO-Me induces JNK-dependent DR5 expression. bardoxolone methyl 44-51 TNF receptor superfamily member 10b Homo sapiens 74-77 18794136-4 2008 Analysis of DR5 promoter regions defines that the CCAAT/enhancer binding protein homologous protein (CHOP) binding site is responsible for CDDO-Me-induced transactivation of the DR5 gene. bardoxolone methyl 139-146 TNF receptor superfamily member 10b Homo sapiens 12-15 18794136-4 2008 Analysis of DR5 promoter regions defines that the CCAAT/enhancer binding protein homologous protein (CHOP) binding site is responsible for CDDO-Me-induced transactivation of the DR5 gene. bardoxolone methyl 139-146 TNF receptor superfamily member 10b Homo sapiens 178-181 18794136-5 2008 Consistently, CDDO-Me induced DR5 expression and parallel CHOP up-regulation. bardoxolone methyl 14-21 TNF receptor superfamily member 10b Homo sapiens 30-33 18794136-6 2008 Blockade of CHOP up-regulation also abrogated CDDO-Me-induced DR5 expression. bardoxolone methyl 46-53 TNF receptor superfamily member 10b Homo sapiens 62-65 18794136-7 2008 These results indicate that CDDO-Me induces CHOP-dependent DR5 up-regulation. bardoxolone methyl 28-35 TNF receptor superfamily member 10b Homo sapiens 59-62 18599488-6 2008 Treatment with ABT-737 did not change the levels of c-FLIP, FADD, and caspase-8 but up-regulated the levels of the TRAIL receptor DR5. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 15-18 TNF receptor superfamily member 10b Homo sapiens 130-133 18599488-7 2008 DR5 up-regulation induced by ABT-737 treatment occurred through a transcriptional mechanism, and mutagenesis studies demonstrated that the NF-kappaB site found in the DR5 promoter was essential for the ability of ABT-737 to increase the levels of this mRNA. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 29-32 TNF receptor superfamily member 10b Homo sapiens 0-3 18599488-7 2008 DR5 up-regulation induced by ABT-737 treatment occurred through a transcriptional mechanism, and mutagenesis studies demonstrated that the NF-kappaB site found in the DR5 promoter was essential for the ability of ABT-737 to increase the levels of this mRNA. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 29-32 TNF receptor superfamily member 10b Homo sapiens 167-170 18599488-7 2008 DR5 up-regulation induced by ABT-737 treatment occurred through a transcriptional mechanism, and mutagenesis studies demonstrated that the NF-kappaB site found in the DR5 promoter was essential for the ability of ABT-737 to increase the levels of this mRNA. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 213-216 TNF receptor superfamily member 10b Homo sapiens 0-3 18599488-7 2008 DR5 up-regulation induced by ABT-737 treatment occurred through a transcriptional mechanism, and mutagenesis studies demonstrated that the NF-kappaB site found in the DR5 promoter was essential for the ability of ABT-737 to increase the levels of this mRNA. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 213-216 TNF receptor superfamily member 10b Homo sapiens 167-170 18534188-0 2008 Hydrogen peroxide enhances TRAIL-induced cell death through up-regulation of DR5 in human astrocytic cells. Hydrogen Peroxide 0-17 TNF receptor superfamily member 10b Homo sapiens 77-80 18544564-7 2008 PS-341 (VELCADE, bortezomib), a proteasome inhibitor used to treat human cancer, increases the levels of both HIF-2alpha and c-Myc and elevates the level of DR5 in renal cancer, sensitizing renal cancer cells to TRAIL therapy. Bortezomib 0-6 TNF receptor superfamily member 10b Homo sapiens 157-160 18544564-7 2008 PS-341 (VELCADE, bortezomib), a proteasome inhibitor used to treat human cancer, increases the levels of both HIF-2alpha and c-Myc and elevates the level of DR5 in renal cancer, sensitizing renal cancer cells to TRAIL therapy. Bortezomib 8-15 TNF receptor superfamily member 10b Homo sapiens 157-160 18544564-7 2008 PS-341 (VELCADE, bortezomib), a proteasome inhibitor used to treat human cancer, increases the levels of both HIF-2alpha and c-Myc and elevates the level of DR5 in renal cancer, sensitizing renal cancer cells to TRAIL therapy. Bortezomib 17-27 TNF receptor superfamily member 10b Homo sapiens 157-160 18698026-2 2008 EXPERIMENTAL DESIGN: The activity of perifosine in combination with TRAIL was evaluated with experiments testing the effect of perifosine on DR4/DR5 expression by the use of chimeric blocking antibodies, as well as siRNA. perifosine 37-47 TNF receptor superfamily member 10b Homo sapiens 145-148 18698026-2 2008 EXPERIMENTAL DESIGN: The activity of perifosine in combination with TRAIL was evaluated with experiments testing the effect of perifosine on DR4/DR5 expression by the use of chimeric blocking antibodies, as well as siRNA. perifosine 127-137 TNF receptor superfamily member 10b Homo sapiens 145-148 18698026-3 2008 RESULTS: DR4 and DR5 expression was induced by exposure to single-agent perifosine. perifosine 72-82 TNF receptor superfamily member 10b Homo sapiens 17-20 19568915-12 2008 Isoliquiritigenin increased the amount of DR5 protein among TRAIL receptors. isoliquiritigenin 0-17 TNF receptor superfamily member 10b Homo sapiens 42-45 18534188-7 2008 H(2)O(2)-mediated cell death was blocked upon treatment with DR5:Fc protein, a TRAIL-specific antagonistic protein. Hydrogen Peroxide 0-8 TNF receptor superfamily member 10b Homo sapiens 61-64 18460448-4 2008 DIM-C-pPhBr and DIM-C-pPhF not only induced CHOP but also activated death receptor 5 (CHOP dependent), cleavage of caspase 8 and poly (ADP ribose) polymerase (PARP) that is consistent with activation of the extrinsic pathway of apoptosis. dim-c-pphbr 0-11 TNF receptor superfamily member 10b Homo sapiens 68-84 18523266-0 2008 Proteasome inhibitors enhance TRAIL-induced apoptosis through the intronic regulation of DR5: involvement of NF-kappa B and reactive oxygen species-mediated p53 activation. Reactive Oxygen Species 124-147 TNF receptor superfamily member 10b Homo sapiens 89-92 18460448-4 2008 DIM-C-pPhBr and DIM-C-pPhF not only induced CHOP but also activated death receptor 5 (CHOP dependent), cleavage of caspase 8 and poly (ADP ribose) polymerase (PARP) that is consistent with activation of the extrinsic pathway of apoptosis. dim-c-pphf 16-26 TNF receptor superfamily member 10b Homo sapiens 68-84 18636153-6 2008 In addition, SAHA up-regulated the death receptor DR5, inducing the activation of caspase-8 with the consequent cleavage of Bid. Vorinostat 13-17 TNF receptor superfamily member 10b Homo sapiens 50-53 18304628-2 2008 The addition of TRAIL significantly potentiated the cytotoxic effect of CDDO-Im, through mechanisms involving the induction of TRAIL-R1/TRAIL-R2 and downmodulation of TRAIL-R3/TRAIL-R4. 1-(2-cyano-3,12-dioxooleana-1,9-dien-28-oyl) imidazole 72-79 TNF receptor superfamily member 10b Homo sapiens 136-144 18523266-13 2008 These findings reveal that proteasome inhibitor-mediated NF-kappaB and ROS-dependent p53 activation are contributed to intronic regulation of DR5 transcription, and resulted in the subsequent enhancement of TRAIL-induced apoptosis in human lung cancer cells. Reactive Oxygen Species 71-74 TNF receptor superfamily member 10b Homo sapiens 142-145 18374381-4 2008 KTA induced cancer cell death treatment by triggering mitochondrial and death receptor 5 (DR5) apoptotic pathways, but did not act on the Fas receptor. kotomolide A 0-3 TNF receptor superfamily member 10b Homo sapiens 72-88 18374381-4 2008 KTA induced cancer cell death treatment by triggering mitochondrial and death receptor 5 (DR5) apoptotic pathways, but did not act on the Fas receptor. kotomolide A 0-3 TNF receptor superfamily member 10b Homo sapiens 90-93 18193086-4 2008 Although dipyridamole upregulated both DR4 and DR5 and increased their cell surface expression, RNA interference revealed a preferential dependence on DR5. Dipyridamole 9-21 TNF receptor superfamily member 10b Homo sapiens 47-50 18549619-3 2008 The cytotoxic effect was measured by MTT assay; cell apoptosis rate was determined by flow cytometry after Annexin V/PI staining; the expression level of DR5 on surface of HL-60 cells treated with Ara-C at different concentrations for 24 hours was determined by flow cytometry. Cytarabine 197-202 TNF receptor superfamily member 10b Homo sapiens 154-157 18549619-6 2008 The apoptosis rate of HL-60 cells in Ara-C + rsTRAIL tandem given group was higher than that in Ara-C + rsTRAIL simultaneously given group, the expression level of DR5 on surface of HL-60 cells and intracellular activity of caspase-8 in Ara-C + rsTRAIL tandem given group were higher than those in rsTRAIL group. Cytarabine 37-42 TNF receptor superfamily member 10b Homo sapiens 164-167 18549619-6 2008 The apoptosis rate of HL-60 cells in Ara-C + rsTRAIL tandem given group was higher than that in Ara-C + rsTRAIL simultaneously given group, the expression level of DR5 on surface of HL-60 cells and intracellular activity of caspase-8 in Ara-C + rsTRAIL tandem given group were higher than those in rsTRAIL group. rstrail 45-52 TNF receptor superfamily member 10b Homo sapiens 164-167 18549619-6 2008 The apoptosis rate of HL-60 cells in Ara-C + rsTRAIL tandem given group was higher than that in Ara-C + rsTRAIL simultaneously given group, the expression level of DR5 on surface of HL-60 cells and intracellular activity of caspase-8 in Ara-C + rsTRAIL tandem given group were higher than those in rsTRAIL group. rstrail 104-111 TNF receptor superfamily member 10b Homo sapiens 164-167 18549619-6 2008 The apoptosis rate of HL-60 cells in Ara-C + rsTRAIL tandem given group was higher than that in Ara-C + rsTRAIL simultaneously given group, the expression level of DR5 on surface of HL-60 cells and intracellular activity of caspase-8 in Ara-C + rsTRAIL tandem given group were higher than those in rsTRAIL group. rstrail 104-111 TNF receptor superfamily member 10b Homo sapiens 164-167 18549619-6 2008 The apoptosis rate of HL-60 cells in Ara-C + rsTRAIL tandem given group was higher than that in Ara-C + rsTRAIL simultaneously given group, the expression level of DR5 on surface of HL-60 cells and intracellular activity of caspase-8 in Ara-C + rsTRAIL tandem given group were higher than those in rsTRAIL group. rstrail 104-111 TNF receptor superfamily member 10b Homo sapiens 164-167 18549619-7 2008 When HL-60 cells treated with 5 and 10 mg/L of Ara-C for 24 hours, the expression level of DR5 on surface of HL-60 cells was higher than that in control group. Cytarabine 47-52 TNF receptor superfamily member 10b Homo sapiens 91-94 18549619-9 2008 Ara-C can upregulate DR5 expression on the surface of HL-60 cells and enhance the effect of rsTRAIL-inducing apoptosis. Cytarabine 0-5 TNF receptor superfamily member 10b Homo sapiens 21-24 18549619-12 2008 The mechanism may correlate with up-regulation of the expression level of DR5 and/or caspase-8 in HL-60 cells by Ara-C. Cytarabine 113-118 TNF receptor superfamily member 10b Homo sapiens 74-77 18193086-7 2008 In addition, a transcriptional mechanism powered by CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP) induction was responsible for DR5 upregulation by dipyridamole. Dipyridamole 167-179 TNF receptor superfamily member 10b Homo sapiens 147-150 17957723-6 2008 DIM-C-pPhOCH(3) also activated the extrinsic apoptosis pathway through increased phosphorylation of c-jun N-terminal kinase which, in turn, activated C/EBP homologous transcription factor (CHOP) and death receptor 5 (DR5). dim-c-pphoch(3) 0-15 TNF receptor superfamily member 10b Homo sapiens 199-215 18187663-0 2008 Triptolide sensitizes AML cells to TRAIL-induced apoptosis via decrease of XIAP and p53-mediated increase of DR5. triptolide 0-10 TNF receptor superfamily member 10b Homo sapiens 109-112 18381960-1 2008 PURPOSE: This study was designed to evaluate the in vitro cytotoxicity and in vivo efficacy of TRA-8, a mouse monoclonal antibody that binds to the DR5 death receptor for tumor necrosis factor-related apoptosis-inducing ligand (also called Apo2L), alone and in combination with CPT-11, against human colon cancer cells and xenografts. Irinotecan 278-284 TNF receptor superfamily member 10b Homo sapiens 148-151 18483298-0 2008 Proteasome inhibitor PS-341 (VELCADE) induces stabilization of the TRAIL receptor DR5 mRNA through the 3"-untranslated region. Bortezomib 21-27 TNF receptor superfamily member 10b Homo sapiens 82-85 18483298-0 2008 Proteasome inhibitor PS-341 (VELCADE) induces stabilization of the TRAIL receptor DR5 mRNA through the 3"-untranslated region. Bortezomib 29-36 TNF receptor superfamily member 10b Homo sapiens 82-85 18483298-2 2008 PS-341 sensitizes prostate cancer cells to TRAIL-induced apoptosis by increasing TRAIL receptors (DR5), inhibiting protein degradation, and elevating DR5 mRNA. Bortezomib 0-6 TNF receptor superfamily member 10b Homo sapiens 98-101 18483298-2 2008 PS-341 sensitizes prostate cancer cells to TRAIL-induced apoptosis by increasing TRAIL receptors (DR5), inhibiting protein degradation, and elevating DR5 mRNA. Bortezomib 0-6 TNF receptor superfamily member 10b Homo sapiens 150-153 18483298-7 2008 In LNCaP cells treated with PS-341, small interfering RNA-mediated knockdown of HuR markedly decreases the half-life of DR5 mRNA, indicating that HuR is essential for mRNA stabilization. Bortezomib 28-34 TNF receptor superfamily member 10b Homo sapiens 120-123 17968315-0 2008 Celecoxib and a novel COX-2 inhibitor ON09310 upregulate death receptor 5 expression via GADD153/CHOP. Celecoxib 0-9 TNF receptor superfamily member 10b Homo sapiens 57-73 17968315-3 2008 Here we report that COX-2-selective celecoxib and a novel COX-2 inhibitor ON09310 upregulate death receptor 5 (DR5) and cooperate with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), the ligand for DR5, to induce apoptosis in COX-2-positive and -negative cancer cells. Celecoxib 36-45 TNF receptor superfamily member 10b Homo sapiens 93-109 17968315-3 2008 Here we report that COX-2-selective celecoxib and a novel COX-2 inhibitor ON09310 upregulate death receptor 5 (DR5) and cooperate with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), the ligand for DR5, to induce apoptosis in COX-2-positive and -negative cancer cells. Celecoxib 36-45 TNF receptor superfamily member 10b Homo sapiens 111-114 17968315-3 2008 Here we report that COX-2-selective celecoxib and a novel COX-2 inhibitor ON09310 upregulate death receptor 5 (DR5) and cooperate with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), the ligand for DR5, to induce apoptosis in COX-2-positive and -negative cancer cells. Celecoxib 36-45 TNF receptor superfamily member 10b Homo sapiens 215-218 17968315-3 2008 Here we report that COX-2-selective celecoxib and a novel COX-2 inhibitor ON09310 upregulate death receptor 5 (DR5) and cooperate with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), the ligand for DR5, to induce apoptosis in COX-2-positive and -negative cancer cells. ON09310 74-81 TNF receptor superfamily member 10b Homo sapiens 93-109 17968315-3 2008 Here we report that COX-2-selective celecoxib and a novel COX-2 inhibitor ON09310 upregulate death receptor 5 (DR5) and cooperate with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), the ligand for DR5, to induce apoptosis in COX-2-positive and -negative cancer cells. ON09310 74-81 TNF receptor superfamily member 10b Homo sapiens 111-114 17968315-3 2008 Here we report that COX-2-selective celecoxib and a novel COX-2 inhibitor ON09310 upregulate death receptor 5 (DR5) and cooperate with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), the ligand for DR5, to induce apoptosis in COX-2-positive and -negative cancer cells. ON09310 74-81 TNF receptor superfamily member 10b Homo sapiens 215-218 18413750-6 2008 Furthermore, the OSU-03012/EGFR inhibitor combination induced GADD153-mediated up-regulation of death receptor 5 expression and subsequent activation of the extrinsic apoptosis pathway. OSU 03012 17-26 TNF receptor superfamily member 10b Homo sapiens 96-112 18423123-8 2008 RESULTS: The positive rate of DR5 on SMMC-7721 cells was 95.0%. smmc 37-41 TNF receptor superfamily member 10b Homo sapiens 30-33 18187663-5 2008 Furthermore, triptolide increased DR5 levels in OCI-AML3, while the DR5 increase was blunted in p53-knockdown OCI-AML3 and p53-mutated U937 cells, confirming a role for p53 in the regulation of DR5. triptolide 13-23 TNF receptor superfamily member 10b Homo sapiens 34-37 18341587-7 2008 Transient knockdown of the transcription factor GADD153/C/EBP homologous protein and application of the synthetic c-Jun N-terminal kinase inhibitor SP600125 indicated that enhanced DR5 expression occurred independently of GADD153/C/EBP homologous protein, but required activation of the c-Jun N-terminal kinase/c-Jun signaling pathway. pyrazolanthrone 148-156 TNF receptor superfamily member 10b Homo sapiens 181-184 17957723-6 2008 DIM-C-pPhOCH(3) also activated the extrinsic apoptosis pathway through increased phosphorylation of c-jun N-terminal kinase which, in turn, activated C/EBP homologous transcription factor (CHOP) and death receptor 5 (DR5). dim-c-pphoch(3) 0-15 TNF receptor superfamily member 10b Homo sapiens 217-220 18164688-0 2008 Rosiglitazone promotes tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by reactive oxygen species-mediated up-regulation of death receptor 5 and down-regulation of c-FLIP. Rosiglitazone 0-13 TNF receptor superfamily member 10b Homo sapiens 150-166 18164688-9 2008 Taken together, the results of this study demonstrate that rosiglitazone enhances TRAIL-induced apoptosis in various cancer cells by ROS-mediated DR5 up-regulation and down-regulation of c-FLIPs. Rosiglitazone 59-72 TNF receptor superfamily member 10b Homo sapiens 146-149 18164688-9 2008 Taken together, the results of this study demonstrate that rosiglitazone enhances TRAIL-induced apoptosis in various cancer cells by ROS-mediated DR5 up-regulation and down-regulation of c-FLIPs. Reactive Oxygen Species 133-136 TNF receptor superfamily member 10b Homo sapiens 146-149 18222423-4 2008 Our observations indicated that resveratrol, a polyphenolic phytoalexin, decreased STAT3 and NF-kappaB activation, while activating JNK-cJun that finally suppressed expression of cFLIP and Bcl-xL proteins and increased sensitivity to exogenous TRAIL in DR5-positive melanomas. Resveratrol 32-43 TNF receptor superfamily member 10b Homo sapiens 253-256 18222423-5 2008 Interestingly, resveratrol did not increase surface expression of DR5 in human melanomas, while gamma-irradiation or sodium arsenite treatment substantially upregulated DR5 expression. sodium arsenite 117-132 TNF receptor superfamily member 10b Homo sapiens 169-172 18222423-6 2008 Hence, an initial increase in DR5 surface expression (either by gamma-irradiation or arsenite), and subsequent downregulation of antiapoptotic cFLIP and Bcl-xL (by resveratrol), appear to constitute an efficient approach to reactivate apoptotic death pathways in TRAIL-resistant human melanomas. arsenite 85-93 TNF receptor superfamily member 10b Homo sapiens 30-33 18164688-0 2008 Rosiglitazone promotes tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by reactive oxygen species-mediated up-regulation of death receptor 5 and down-regulation of c-FLIP. Reactive Oxygen Species 100-123 TNF receptor superfamily member 10b Homo sapiens 150-166 18164688-4 2008 We found that treatment with rosiglitazone significantly induces DR5 expression at both its mRNA and its protein levels, accompanying the generation of reactive oxygen species (ROS). Rosiglitazone 29-42 TNF receptor superfamily member 10b Homo sapiens 65-68 18164688-4 2008 We found that treatment with rosiglitazone significantly induces DR5 expression at both its mRNA and its protein levels, accompanying the generation of reactive oxygen species (ROS). Reactive Oxygen Species 152-175 TNF receptor superfamily member 10b Homo sapiens 65-68 18164688-4 2008 We found that treatment with rosiglitazone significantly induces DR5 expression at both its mRNA and its protein levels, accompanying the generation of reactive oxygen species (ROS). Reactive Oxygen Species 177-180 TNF receptor superfamily member 10b Homo sapiens 65-68 18164688-5 2008 Both treatment with DR5/Fc chimeric protein and silencing of DR5 expression using small interfering RNAs attenuated rosiglitazone plus TRAIL-induced apoptosis, showing the critical role of DR5 in this cell death. Rosiglitazone 116-129 TNF receptor superfamily member 10b Homo sapiens 61-64 18164688-5 2008 Both treatment with DR5/Fc chimeric protein and silencing of DR5 expression using small interfering RNAs attenuated rosiglitazone plus TRAIL-induced apoptosis, showing the critical role of DR5 in this cell death. Rosiglitazone 116-129 TNF receptor superfamily member 10b Homo sapiens 61-64 18164688-6 2008 Pretreatment with GSH significantly inhibited rosiglitazone-induced DR5 up-regulation and the cell death induced by the combined treatment with rosiglitazone and TRAIL, suggesting that ROS mediate rosiglitazone-induced DR5 up-regulation, contributing to TRAIL-mediated apoptosis. Glutathione 18-21 TNF receptor superfamily member 10b Homo sapiens 68-71 18164688-6 2008 Pretreatment with GSH significantly inhibited rosiglitazone-induced DR5 up-regulation and the cell death induced by the combined treatment with rosiglitazone and TRAIL, suggesting that ROS mediate rosiglitazone-induced DR5 up-regulation, contributing to TRAIL-mediated apoptosis. Glutathione 18-21 TNF receptor superfamily member 10b Homo sapiens 219-222 18164688-6 2008 Pretreatment with GSH significantly inhibited rosiglitazone-induced DR5 up-regulation and the cell death induced by the combined treatment with rosiglitazone and TRAIL, suggesting that ROS mediate rosiglitazone-induced DR5 up-regulation, contributing to TRAIL-mediated apoptosis. Rosiglitazone 46-59 TNF receptor superfamily member 10b Homo sapiens 68-71 18164688-6 2008 Pretreatment with GSH significantly inhibited rosiglitazone-induced DR5 up-regulation and the cell death induced by the combined treatment with rosiglitazone and TRAIL, suggesting that ROS mediate rosiglitazone-induced DR5 up-regulation, contributing to TRAIL-mediated apoptosis. Rosiglitazone 46-59 TNF receptor superfamily member 10b Homo sapiens 219-222 18164688-6 2008 Pretreatment with GSH significantly inhibited rosiglitazone-induced DR5 up-regulation and the cell death induced by the combined treatment with rosiglitazone and TRAIL, suggesting that ROS mediate rosiglitazone-induced DR5 up-regulation, contributing to TRAIL-mediated apoptosis. Rosiglitazone 144-157 TNF receptor superfamily member 10b Homo sapiens 219-222 18164688-6 2008 Pretreatment with GSH significantly inhibited rosiglitazone-induced DR5 up-regulation and the cell death induced by the combined treatment with rosiglitazone and TRAIL, suggesting that ROS mediate rosiglitazone-induced DR5 up-regulation, contributing to TRAIL-mediated apoptosis. Reactive Oxygen Species 185-188 TNF receptor superfamily member 10b Homo sapiens 68-71 18164688-6 2008 Pretreatment with GSH significantly inhibited rosiglitazone-induced DR5 up-regulation and the cell death induced by the combined treatment with rosiglitazone and TRAIL, suggesting that ROS mediate rosiglitazone-induced DR5 up-regulation, contributing to TRAIL-mediated apoptosis. Reactive Oxygen Species 185-188 TNF receptor superfamily member 10b Homo sapiens 219-222 18164688-6 2008 Pretreatment with GSH significantly inhibited rosiglitazone-induced DR5 up-regulation and the cell death induced by the combined treatment with rosiglitazone and TRAIL, suggesting that ROS mediate rosiglitazone-induced DR5 up-regulation, contributing to TRAIL-mediated apoptosis. Rosiglitazone 144-157 TNF receptor superfamily member 10b Homo sapiens 219-222 18006694-4 2008 The association of FADD with DR5 results in the formation of death-inducing signaling complex (DISC) to trigger the downstream apoptotic signaling cascade. fadd 19-23 TNF receptor superfamily member 10b Homo sapiens 29-32 18036820-0 2008 A novel sesquiterpenoid dimer parviflorene F induces apoptosis by up-regulating the expression of TRAIL-R2 and a caspase-dependent mechanism. sesquiterpenoid 8-23 TNF receptor superfamily member 10b Homo sapiens 98-106 18036820-0 2008 A novel sesquiterpenoid dimer parviflorene F induces apoptosis by up-regulating the expression of TRAIL-R2 and a caspase-dependent mechanism. parviflorene F 30-44 TNF receptor superfamily member 10b Homo sapiens 98-106 18022315-0 2008 RRR-gamma-tocopherol induces human breast cancer cells to undergo apoptosis via death receptor 5 (DR5)-mediated apoptotic signaling. Tocopherols 9-20 TNF receptor superfamily member 10b Homo sapiens 80-96 18022315-0 2008 RRR-gamma-tocopherol induces human breast cancer cells to undergo apoptosis via death receptor 5 (DR5)-mediated apoptotic signaling. Tocopherols 9-20 TNF receptor superfamily member 10b Homo sapiens 98-101 18022315-2 2008 gammaT was shown to induce cancer cells but not normal cells to undergo apoptosis, sensitize cancer cells to Tumor necrosis factor-Related Apoptosis-Inducing Ligand (TRAIL)-induced apoptosis, and increase death receptor 5 (DR5) mRNA, protein and cell surface expression. gamma-Tocopherol 0-6 TNF receptor superfamily member 10b Homo sapiens 205-221 18022315-2 2008 gammaT was shown to induce cancer cells but not normal cells to undergo apoptosis, sensitize cancer cells to Tumor necrosis factor-Related Apoptosis-Inducing Ligand (TRAIL)-induced apoptosis, and increase death receptor 5 (DR5) mRNA, protein and cell surface expression. gamma-Tocopherol 0-6 TNF receptor superfamily member 10b Homo sapiens 223-226 18022315-3 2008 Knockdown of DR5 attenuated gammaT-induced apoptosis. gamma-Tocopherol 28-34 TNF receptor superfamily member 10b Homo sapiens 13-16 18022315-5 2008 Thus, gammaT is a potent pro-apoptotic agent for human breast cancer cells inducing apoptosis via activation of DR5-mediated apoptotic pathway. gamma-Tocopherol 6-12 TNF receptor superfamily member 10b Homo sapiens 112-115 18177927-11 2008 IHC revealed an increase in DR5 expression in tumor slices treated with cisplatin for 24 h. IHC and Western blotting demonstrated TRA-8-induced cell death via apoptosis and activation of caspase 3 and 8. Cisplatin 72-81 TNF receptor superfamily member 10b Homo sapiens 28-31 18202802-0 2008 Induction of apoptosis by pectenotoxin-2 is mediated with the induction of DR4/DR5, Egr-1 and NAG-1, activation of caspases and modulation of the Bcl-2 family in p53-deficient Hep3B hepatocellular carcinoma cells. pectenotoxin 2 26-40 TNF receptor superfamily member 10b Homo sapiens 79-82 17703232-5 2007 Notably, disulfide-bonded fusion proteins of the THD of mTRAIL and mCD95L with a subdomain of the tenascin-C (TNC) oligomerization domain, which still assembled into trimers, efficiently interacted with their cognate cellular receptors and robustly stimulated CD95 and TRAILR2 signaling after secondary cross-linking. Disulfides 9-18 TNF receptor superfamily member 10b Homo sapiens 269-276 18097593-0 2008 Sp1 is involved in 8-chloro-adenosine-upregulated death receptor 5 expression in human hepatoma cells. 8-chloro 19-27 TNF receptor superfamily member 10b Homo sapiens 50-66 18097593-0 2008 Sp1 is involved in 8-chloro-adenosine-upregulated death receptor 5 expression in human hepatoma cells. Adenosine 28-37 TNF receptor superfamily member 10b Homo sapiens 50-66 18097593-4 2008 In the present study, we demonstrated by Western blot and real-time PCR that 8-Cl-Ado selectively up-regulated death receptor 5 (DR5), but not death receptor 4 (DR4), at both protein and RNA levels in human hepatoma cell line BEL-7402. 8-chloroadenosine 77-85 TNF receptor superfamily member 10b Homo sapiens 111-127 18097593-4 2008 In the present study, we demonstrated by Western blot and real-time PCR that 8-Cl-Ado selectively up-regulated death receptor 5 (DR5), but not death receptor 4 (DR4), at both protein and RNA levels in human hepatoma cell line BEL-7402. 8-chloroadenosine 77-85 TNF receptor superfamily member 10b Homo sapiens 129-132 18097593-6 2008 Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) confirmed that 8-Cl-Ado treatment facilitated transcription factor Sp1 binding to its cis-element -198/-189 in the DR5 promoter, suggesting that Sp1 is at least one of the 8-Cl-Ado-responsive transcription factors. 8-chloroadenosine 100-108 TNF receptor superfamily member 10b Homo sapiens 200-203 18097593-6 2008 Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) confirmed that 8-Cl-Ado treatment facilitated transcription factor Sp1 binding to its cis-element -198/-189 in the DR5 promoter, suggesting that Sp1 is at least one of the 8-Cl-Ado-responsive transcription factors. 8-cl 100-104 TNF receptor superfamily member 10b Homo sapiens 200-203 18097593-6 2008 Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) confirmed that 8-Cl-Ado treatment facilitated transcription factor Sp1 binding to its cis-element -198/-189 in the DR5 promoter, suggesting that Sp1 is at least one of the 8-Cl-Ado-responsive transcription factors. beta-apocarotenoid-14',13'-dioxygenase 105-108 TNF receptor superfamily member 10b Homo sapiens 200-203 18097593-8 2008 These data allowed us to draw a conclusion that 8-Cl-Ado-enhanced DR5 expression is regulated by Sp1 binding to the -198/-189 cis-element in DR5 promoter without affecting NF-kappaB activity in the hepatoma cells. 8-chloroadenosine 48-56 TNF receptor superfamily member 10b Homo sapiens 66-69 18097593-8 2008 These data allowed us to draw a conclusion that 8-Cl-Ado-enhanced DR5 expression is regulated by Sp1 binding to the -198/-189 cis-element in DR5 promoter without affecting NF-kappaB activity in the hepatoma cells. 8-chloroadenosine 48-56 TNF receptor superfamily member 10b Homo sapiens 141-144 17922852-0 2007 Low concentrations of doxorubicin sensitizes human solid cancer cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-receptor (R) 2-mediated apoptosis by inducing TRAIL-R2 expression. Doxorubicin 22-33 TNF receptor superfamily member 10b Homo sapiens 183-191 17922852-7 2007 Low concentrations of doxorubicin (0.1 and 1 microg/mL) significantly increased TRAIL-R2 expression at both the mRNA and protein levels. Doxorubicin 22-33 TNF receptor superfamily member 10b Homo sapiens 80-88 17922852-11 2007 These findings suggest that doxorubicin sensitizes solid cancer cells to TRAIL-R2-mediated apoptosis by inducing TRAIL-R2 expression, and that the combination treatment with lexatumumab and doxorubicin might be a promising targeted therapy for cancers, including RCC, prostate, bladder, and lung cancers. Doxorubicin 28-39 TNF receptor superfamily member 10b Homo sapiens 73-81 17922852-11 2007 These findings suggest that doxorubicin sensitizes solid cancer cells to TRAIL-R2-mediated apoptosis by inducing TRAIL-R2 expression, and that the combination treatment with lexatumumab and doxorubicin might be a promising targeted therapy for cancers, including RCC, prostate, bladder, and lung cancers. Doxorubicin 28-39 TNF receptor superfamily member 10b Homo sapiens 113-121 17922852-11 2007 These findings suggest that doxorubicin sensitizes solid cancer cells to TRAIL-R2-mediated apoptosis by inducing TRAIL-R2 expression, and that the combination treatment with lexatumumab and doxorubicin might be a promising targeted therapy for cancers, including RCC, prostate, bladder, and lung cancers. Doxorubicin 190-201 TNF receptor superfamily member 10b Homo sapiens 73-81 17922852-11 2007 These findings suggest that doxorubicin sensitizes solid cancer cells to TRAIL-R2-mediated apoptosis by inducing TRAIL-R2 expression, and that the combination treatment with lexatumumab and doxorubicin might be a promising targeted therapy for cancers, including RCC, prostate, bladder, and lung cancers. Doxorubicin 190-201 TNF receptor superfamily member 10b Homo sapiens 113-121 18172319-0 2008 Arsenic trioxide sensitizes human glioma cells, but not normal astrocytes, to TRAIL-induced apoptosis via CCAAT/enhancer-binding protein homologous protein-dependent DR5 up-regulation. Arsenic Trioxide 0-16 TNF receptor superfamily member 10b Homo sapiens 166-169 18172319-6 2008 The involvement of CHOP in this process was confirmed by siRNA-mediated CHOP suppression, which not only attenuated As(2)O(3)-induced DR5 up-regulation but also inhibited the As(2)O(3)-stimulated TRAIL-induced apoptosis. (2)o(3) 118-125 TNF receptor superfamily member 10b Homo sapiens 134-137 18172319-6 2008 The involvement of CHOP in this process was confirmed by siRNA-mediated CHOP suppression, which not only attenuated As(2)O(3)-induced DR5 up-regulation but also inhibited the As(2)O(3)-stimulated TRAIL-induced apoptosis. (2)o(3) 177-184 TNF receptor superfamily member 10b Homo sapiens 134-137 18172319-10 2008 In addition, As(2)O(3)-mediated up-regulation of CHOP and DR5, as well as partial proteolytic processing of procaspase-3 by TRAIL, was not induced in astrocytes. (2)o(3) 15-22 TNF receptor superfamily member 10b Homo sapiens 58-61 17703232-5 2007 Notably, disulfide-bonded fusion proteins of the THD of mTRAIL and mCD95L with a subdomain of the tenascin-C (TNC) oligomerization domain, which still assembled into trimers, efficiently interacted with their cognate cellular receptors and robustly stimulated CD95 and TRAILR2 signaling after secondary cross-linking. Thiamine 49-52 TNF receptor superfamily member 10b Homo sapiens 269-276 17982676-6 2007 Compared to 5-FU-treated TP53-proficient HCT116 cultures, 5-FU-treated TP53-depleted HCT116 cultures showed lack of CDKN1A induction, decreased apoptotic levels, decreased FAS and TNFRSF10B transcript levels and cleaved PARP protein levels, G1/S transition arrests, decreased CCND1 protein levels, and smaller intra-S phase arrests. Fluorouracil 58-62 TNF receptor superfamily member 10b Homo sapiens 180-189 18065493-7 2007 We show that TRAIL activates the canonical caspase-dependent pathway, whereas treatment with cycloheximide increases the sensitivity of MG-63 cells to TRAIL and anti-DR5 and can also sensitize hPOB-tert cells to both agents. Cycloheximide 93-106 TNF receptor superfamily member 10b Homo sapiens 166-169 17893044-7 2007 In addition, cells treated with hydrogen peroxide expressed less X-linked inhibitor of apoptosis protein (XIAP) and survivin which, like cFLIP(S), are short-half-life proteins with an antiapoptotic function while expression levels of DR5, caspases-8, -9, -3, and Bax are maintained. Hydrogen Peroxide 32-49 TNF receptor superfamily member 10b Homo sapiens 234-257 18006776-5 2007 DIM-C-pPhtBu also induced several common proapoptotic responses in ACHN and 786-0 cells, including increased expression of nonsteroidal anti-inflammatory drug-activated gene-1 and endoplasmic reticulum stress, which activates death receptor 5 and the extrinsic pathway of apoptosis. 1,1-bis(3'-indolyl)-1-(4-t-butylphenyl)methane 0-12 TNF receptor superfamily member 10b Homo sapiens 226-242 17689858-6 2007 These results indicate that caspase-3 is a key regulator of apoptosis in response to combined genistein and TRAIL in human gastric adenocarcinoma AGS cells through the activation of DR5 and mitochondrial dysfunction. Genistein 94-103 TNF receptor superfamily member 10b Homo sapiens 182-185 17652336-0 2007 Thapsigargin sensitizes human melanoma cells to TRAIL-induced apoptosis by up-regulation of TRAIL-R2 through the unfolded protein response. Thapsigargin 0-12 TNF receptor superfamily member 10b Homo sapiens 92-100 17652336-3 2007 We show in this study that the endoplasmic reticulum stress inducer, thapsigargin (TG), selectively up-regulated TRAIL-R2 and enhanced TRAIL-induced apoptosis in melanoma cells. Thapsigargin 69-81 TNF receptor superfamily member 10b Homo sapiens 113-121 17652336-3 2007 We show in this study that the endoplasmic reticulum stress inducer, thapsigargin (TG), selectively up-regulated TRAIL-R2 and enhanced TRAIL-induced apoptosis in melanoma cells. Thapsigargin 83-85 TNF receptor superfamily member 10b Homo sapiens 113-121 17684158-6 2007 To determine the underlying mechanism of the synergy between DMC and TRAIL, we have demonstrated that DMC induces a CCAAT/enhancer binding protein homologous protein-dependent expression of DR5, a major TRAIL receptor, and reduces the levels of cellular FLICE-inhibitory protein (c-FLIP) (both the long and short forms), key inhibitors of death receptor-mediated apoptosis, by facilitating c-FLIP degradation through a ubiquitin/proteasome-dependent mechanism. 2,5-dimethylcelecoxib 61-64 TNF receptor superfamily member 10b Homo sapiens 190-193 17684158-6 2007 To determine the underlying mechanism of the synergy between DMC and TRAIL, we have demonstrated that DMC induces a CCAAT/enhancer binding protein homologous protein-dependent expression of DR5, a major TRAIL receptor, and reduces the levels of cellular FLICE-inhibitory protein (c-FLIP) (both the long and short forms), key inhibitors of death receptor-mediated apoptosis, by facilitating c-FLIP degradation through a ubiquitin/proteasome-dependent mechanism. 2,5-dimethylcelecoxib 102-105 TNF receptor superfamily member 10b Homo sapiens 190-193 17684158-7 2007 It is noteworthy that enforced expression of c-FLIP or silencing of DR5 expression using DR5 small interfering RNA abrogated the enhanced effects on induction of apoptosis by the combination of DMC and TRAIL, indicating that both DR5 up-regulation and c-FLIP reduction contribute to cooperative induction of apoptosis by the combination of DMC and TRAIL. 2,5-dimethylcelecoxib 194-197 TNF receptor superfamily member 10b Homo sapiens 68-71 17684158-7 2007 It is noteworthy that enforced expression of c-FLIP or silencing of DR5 expression using DR5 small interfering RNA abrogated the enhanced effects on induction of apoptosis by the combination of DMC and TRAIL, indicating that both DR5 up-regulation and c-FLIP reduction contribute to cooperative induction of apoptosis by the combination of DMC and TRAIL. 2,5-dimethylcelecoxib 194-197 TNF receptor superfamily member 10b Homo sapiens 89-92 17684158-7 2007 It is noteworthy that enforced expression of c-FLIP or silencing of DR5 expression using DR5 small interfering RNA abrogated the enhanced effects on induction of apoptosis by the combination of DMC and TRAIL, indicating that both DR5 up-regulation and c-FLIP reduction contribute to cooperative induction of apoptosis by the combination of DMC and TRAIL. 2,5-dimethylcelecoxib 194-197 TNF receptor superfamily member 10b Homo sapiens 89-92 17684158-7 2007 It is noteworthy that enforced expression of c-FLIP or silencing of DR5 expression using DR5 small interfering RNA abrogated the enhanced effects on induction of apoptosis by the combination of DMC and TRAIL, indicating that both DR5 up-regulation and c-FLIP reduction contribute to cooperative induction of apoptosis by the combination of DMC and TRAIL. 2,5-dimethylcelecoxib 340-343 TNF receptor superfamily member 10b Homo sapiens 68-71 17684158-7 2007 It is noteworthy that enforced expression of c-FLIP or silencing of DR5 expression using DR5 small interfering RNA abrogated the enhanced effects on induction of apoptosis by the combination of DMC and TRAIL, indicating that both DR5 up-regulation and c-FLIP reduction contribute to cooperative induction of apoptosis by the combination of DMC and TRAIL. 2,5-dimethylcelecoxib 340-343 TNF receptor superfamily member 10b Homo sapiens 89-92 17684158-7 2007 It is noteworthy that enforced expression of c-FLIP or silencing of DR5 expression using DR5 small interfering RNA abrogated the enhanced effects on induction of apoptosis by the combination of DMC and TRAIL, indicating that both DR5 up-regulation and c-FLIP reduction contribute to cooperative induction of apoptosis by the combination of DMC and TRAIL. 2,5-dimethylcelecoxib 340-343 TNF receptor superfamily member 10b Homo sapiens 89-92 17684158-8 2007 Together, we conclude that DMC sensitizes human NSCLC cells to TRAIL-induced apoptosis via induction of DR5 and down-regulation of c-FLIP. 2,5-dimethylcelecoxib 27-30 TNF receptor superfamily member 10b Homo sapiens 104-107 17671697-0 2007 Regulation of DR-5 protein and mitochondrial transmembrane potential by gemcitabine, a possible mechanism of gemcitabine-enhanced TRAIL-induced apoptosis. gemcitabine 72-83 TNF receptor superfamily member 10b Homo sapiens 14-18 17548900-0 2007 Induction of death receptor 5 and suppression of survivin contribute to sensitization of TRAIL-induced cytotoxicity by quercetin in non-small cell lung cancer cells. Quercetin 119-128 TNF receptor superfamily member 10b Homo sapiens 13-29 17548900-3 2007 Quercetin increased expression of death receptor (DR) 5, whereas it had no effect on that of other components of the death-inducing signaling complex. Quercetin 0-9 TNF receptor superfamily member 10b Homo sapiens 34-55 17548900-7 2007 These results suggest the pathways for regulation of DR5 and survivin expression by quercetin are distinct. Quercetin 84-93 TNF receptor superfamily member 10b Homo sapiens 53-56 17548900-8 2007 Importantly, suppression of survivin-sensitized TRAIL-induced cell death and blockage of DR5 expression suppressed the synergistic cytotoxicity induced by quercetin and TRAIL co-treatment. Quercetin 155-164 TNF receptor superfamily member 10b Homo sapiens 89-92 17548900-9 2007 On the whole, our data show that quercetin sensitizes TRAIL-induced cytotoxicity in lung cancer cells through two independent pathways: induction of DR5 and suppression of survivin expression, which may underlie the mechanism of the lung cancer preventive activity of quercetin. Quercetin 33-42 TNF receptor superfamily member 10b Homo sapiens 149-152 17585340-0 2007 LY303511 amplifies TRAIL-induced apoptosis in tumor cells by enhancing DR5 oligomerization, DISC assembly, and mitochondrial permeabilization. LY 303511 0-8 TNF receptor superfamily member 10b Homo sapiens 71-74 17585340-7 2007 We link this execution signal to the ability of LY30 to downregulate cFLIP(S) and oligomerize DR5, thus facilitating the signaling of the death initiating signaling complex. ly30 48-52 TNF receptor superfamily member 10b Homo sapiens 94-97 17804742-0 2007 Silibinin sensitizes human glioma cells to TRAIL-mediated apoptosis via DR5 up-regulation and down-regulation of c-FLIP and survivin. Silybin 0-9 TNF receptor superfamily member 10b Homo sapiens 72-75 17804742-4 2007 Silibinin treatment up-regulated DR5, a death receptor of TRAIL, in a transcription factor CHOP-dependent manner. Silybin 0-9 TNF receptor superfamily member 10b Homo sapiens 33-36 17876056-5 2007 We report that quercetin enhanced TRAIL-induced apoptosis by causing the redistribution of DR4 and DR5 into lipid rafts. Quercetin 15-24 TNF receptor superfamily member 10b Homo sapiens 99-102 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 50-57 TNF receptor superfamily member 10b Homo sapiens 242-258 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 50-57 TNF receptor superfamily member 10b Homo sapiens 260-263 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 150-157 TNF receptor superfamily member 10b Homo sapiens 242-258 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 150-157 TNF receptor superfamily member 10b Homo sapiens 260-263 17786298-8 2007 We present, herein, one potential novel sensitizing agent, namely, nitric oxide (NO) that has been shown to sensitize TRAIL-resistant tumor cells to TRAIL apoptosis via its inhibitory effect on the transcription factors NF-kappaB and Yin Yang 1 (YY1), concomitantly with upregulation of DR5. Nitric Oxide 67-79 TNF receptor superfamily member 10b Homo sapiens 287-290 17875785-0 2007 Bortezomib sensitizes non-Hodgkin"s lymphoma cells to apoptosis induced by antibodies to tumor necrosis factor related apoptosis-inducing ligand (TRAIL) receptors TRAIL-R1 and TRAIL-R2. Bortezomib 0-10 TNF receptor superfamily member 10b Homo sapiens 176-184 17936227-9 2007 The DR5 cluster is characterized by a Glu in beta 9 and its polymorphism is connected with preferred anchors at beta 9 of the binding peptide, while the DQ3 cluster is characterized by Glu B86 and Leu B87, which allows the binding of large hydrophobic amino acids at p1 of the binding peptide. Glutamic Acid 38-41 TNF receptor superfamily member 10b Homo sapiens 4-7 17936227-9 2007 The DR5 cluster is characterized by a Glu in beta 9 and its polymorphism is connected with preferred anchors at beta 9 of the binding peptide, while the DQ3 cluster is characterized by Glu B86 and Leu B87, which allows the binding of large hydrophobic amino acids at p1 of the binding peptide. Glutamic Acid 185-188 TNF receptor superfamily member 10b Homo sapiens 4-7 17936227-9 2007 The DR5 cluster is characterized by a Glu in beta 9 and its polymorphism is connected with preferred anchors at beta 9 of the binding peptide, while the DQ3 cluster is characterized by Glu B86 and Leu B87, which allows the binding of large hydrophobic amino acids at p1 of the binding peptide. Leucine 197-200 TNF receptor superfamily member 10b Homo sapiens 4-7 17767167-4 2007 Biochemical analysis of DR5 identified several ectodomain O-(N-acetyl galactosamine-galactose-sialic acid) structures. o-(n-acetyl galactosamine-galactose 58-93 TNF receptor superfamily member 10b Homo sapiens 24-27 17767167-4 2007 Biochemical analysis of DR5 identified several ectodomain O-(N-acetyl galactosamine-galactose-sialic acid) structures. N-Acetylneuraminic Acid 94-105 TNF receptor superfamily member 10b Homo sapiens 24-27 17671697-0 2007 Regulation of DR-5 protein and mitochondrial transmembrane potential by gemcitabine, a possible mechanism of gemcitabine-enhanced TRAIL-induced apoptosis. gemcitabine 109-120 TNF receptor superfamily member 10b Homo sapiens 14-18 17671697-7 2007 In addition, the gemcitabine pretreatment up-regulated DR-5 and p53 protein expression in a time-dependent manner, which suggests the possible involvement of the p53 protein as a transcriptional factor for DR-5 up-regulation. gemcitabine 17-28 TNF receptor superfamily member 10b Homo sapiens 55-59 17671697-7 2007 In addition, the gemcitabine pretreatment up-regulated DR-5 and p53 protein expression in a time-dependent manner, which suggests the possible involvement of the p53 protein as a transcriptional factor for DR-5 up-regulation. gemcitabine 17-28 TNF receptor superfamily member 10b Homo sapiens 206-210 17671697-8 2007 Thus, we report our findings that gemcitabine enhanced the TRAIL-induced apoptosis and the apoptotic signals are mediated by DR-5-dependent pathway and mitochondrial pathway. gemcitabine 34-45 TNF receptor superfamily member 10b Homo sapiens 125-129 17671697-9 2007 Taken together, gemcitabine enhanced TRAIL-induced apoptosis via DR-5 up-regulation and lowering MTP, and suggest that gemcitabine may be used as a successful chemotherapeutic agent for ligand type tumor therapy combined with TRAIL. gemcitabine 16-27 TNF receptor superfamily member 10b Homo sapiens 65-69 17671697-9 2007 Taken together, gemcitabine enhanced TRAIL-induced apoptosis via DR-5 up-regulation and lowering MTP, and suggest that gemcitabine may be used as a successful chemotherapeutic agent for ligand type tumor therapy combined with TRAIL. gemcitabine 119-130 TNF receptor superfamily member 10b Homo sapiens 65-69 17699792-6 2007 EABE-induced apoptosis in HL-60 cells correlated with increased levels of reactive oxygen species, the death receptor 5 (DR5), and caspase activation and decreased levels of the mitochondrial membrane potential. ethacrynic acid butyl ester 0-4 TNF receptor superfamily member 10b Homo sapiens 103-119 17825219-7 2007 RESULTS: Human DR5 extracellular fragment gene was successfully amplified and high level expression was obtained in E.coli BL21 (DE3) induced by 0.1 mmol/L IPTG. Isopropyl Thiogalactoside 156-160 TNF receptor superfamily member 10b Homo sapiens 15-18 17825219-8 2007 The DR5 extracellular fragment protein was identified by SDS-PAGE and Western blot analysis. Sodium Dodecyl Sulfate 57-60 TNF receptor superfamily member 10b Homo sapiens 4-7 17699792-6 2007 EABE-induced apoptosis in HL-60 cells correlated with increased levels of reactive oxygen species, the death receptor 5 (DR5), and caspase activation and decreased levels of the mitochondrial membrane potential. ethacrynic acid butyl ester 0-4 TNF receptor superfamily member 10b Homo sapiens 121-124 17699792-7 2007 Pretreatment with antioxidants, either N-acetylcysteine or catalase, completely blocked EABE-induced apoptosis, H2O2 accumulation, and up-regulation of DR5 levels. Acetylcysteine 39-55 TNF receptor superfamily member 10b Homo sapiens 152-155 17699792-7 2007 Pretreatment with antioxidants, either N-acetylcysteine or catalase, completely blocked EABE-induced apoptosis, H2O2 accumulation, and up-regulation of DR5 levels. ethacrynic acid butyl ester 88-92 TNF receptor superfamily member 10b Homo sapiens 152-155 17520194-1 2007 Previous studies indicated that signalling via CD95 and DR5 is greatly enhanced by the formation of ceramide-enriched membrane platforms. Ceramides 100-108 TNF receptor superfamily member 10b Homo sapiens 56-59 17520194-4 2007 The latter served DR5 to cluster after application of very low doses of TRAIL in combination with doxorubicin. Doxorubicin 98-109 TNF receptor superfamily member 10b Homo sapiens 18-21 17520194-5 2007 Genetic deficiency of the ASM abrogated doxorubicin-induced ceramide release, as well as clustering of DR5 and apoptosis induced by the combined treatment of doxorubicin and TRAIL. Doxorubicin 158-169 TNF receptor superfamily member 10b Homo sapiens 103-106 17620439-6 2007 Enhanced surface expression of TRAIL-R2, but also TRAIL-R1, was associated with bortezomib treatment, which is likely to contribute to the increased processing of caspase-8 in the combination treatment. Bortezomib 80-90 TNF receptor superfamily member 10b Homo sapiens 31-39 17690453-0 2007 Tryptophol induces death receptor (DR) 5-mediated apoptosis in U937 cells. tryptophol 0-10 TNF receptor superfamily member 10b Homo sapiens 19-40 17690453-4 2007 In this study, we found that tryptophol enhances formation of a death-inducing signaling complex including death receptor (DR) 5. tryptophol 29-39 TNF receptor superfamily member 10b Homo sapiens 107-128 17690453-6 2007 These results indicate that tryptophol induces apoptosis through DR5 and that the resistance of PBL to tryptophol-induced apoptosis might be due to competition from DcR1. tryptophol 28-38 TNF receptor superfamily member 10b Homo sapiens 65-68 17470478-10 2007 Oleic acid treatment led to upregulation of the cognate TRAIL receptor death receptor 5 (DR5) but not death receptor 4 (DR4). Oleic Acid 0-10 TNF receptor superfamily member 10b Homo sapiens 71-87 17470478-10 2007 Oleic acid treatment led to upregulation of the cognate TRAIL receptor death receptor 5 (DR5) but not death receptor 4 (DR4). Oleic Acid 0-10 TNF receptor superfamily member 10b Homo sapiens 89-92 17470478-14 2007 CONCLUSION: Our results suggest that FFA induced hepatocyte steatosis sensitises to TRAIL by a DR5 mediated JNK dependent mechanism. Fatty Acids, Nonesterified 37-40 TNF receptor superfamily member 10b Homo sapiens 95-98 17604333-8 2007 Despite the activation of both caspase-8 and caspase-9, perifosine strikingly induced the expression of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor, death receptor 5, and down-regulated cellular FLICE-inhibitory protein (c-FLIP), an endogenous inhibitor of the extrinsic apoptotic pathway, with limited modulatory effects on the expression of other genes including Bcl-2, Bcl-X(L), PUMA, and survivin. perifosine 56-66 TNF receptor superfamily member 10b Homo sapiens 182-198 17604333-9 2007 Silencing of either caspase-8 or death receptor 5 attenuated perifosine-induced apoptosis. perifosine 61-71 TNF receptor superfamily member 10b Homo sapiens 33-49 17493934-0 2007 The farnesyltransferase inhibitor lonafarnib induces CCAAT/enhancer-binding protein homologous protein-dependent expression of death receptor 5, leading to induction of apoptosis in human cancer cells. lonafarnib 34-44 TNF receptor superfamily member 10b Homo sapiens 127-143 17493934-5 2007 In this study, we investigated the modulation of DR5 by the FTI lonafarnib and the involvement of DR5 up-regulation in FTI-induced apoptosis. lonafarnib 64-74 TNF receptor superfamily member 10b Homo sapiens 49-52 17493934-7 2007 Lonafarnib up-regulated DR5 expression, increased cell-surface DR5 distribution, and enhanced tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. lonafarnib 0-10 TNF receptor superfamily member 10b Homo sapiens 24-27 17493934-7 2007 Lonafarnib up-regulated DR5 expression, increased cell-surface DR5 distribution, and enhanced tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. lonafarnib 0-10 TNF receptor superfamily member 10b Homo sapiens 63-66 17493934-8 2007 Overexpression of a dominant-negative Fas-associated death domain mutant or silencing of DR5 expression using small interfering RNA attenuated lonafarnib-induced apoptosis. lonafarnib 143-153 TNF receptor superfamily member 10b Homo sapiens 89-92 17493934-9 2007 These results indicate a critical role of the DR5-mediated extrinsic apoptotic pathway in lonafarnib-induced apoptosis. lonafarnib 90-100 TNF receptor superfamily member 10b Homo sapiens 46-49 17493934-10 2007 By analyzing the DR5 promoter, we found that lonafarnib induced a CCAAT/enhancer-binding protein homologous protein (CHOP)-dependent transactivation of the DR5 promoter. lonafarnib 45-55 TNF receptor superfamily member 10b Homo sapiens 17-20 17493934-10 2007 By analyzing the DR5 promoter, we found that lonafarnib induced a CCAAT/enhancer-binding protein homologous protein (CHOP)-dependent transactivation of the DR5 promoter. lonafarnib 45-55 TNF receptor superfamily member 10b Homo sapiens 156-159 17493934-11 2007 Lonafarnib increased CHOP expression, whereas silencing of CHOP expression abrogated lonafarnib-induced DR5 expression. lonafarnib 85-95 TNF receptor superfamily member 10b Homo sapiens 104-107 17493934-12 2007 These results thus indicate that lonafarnib induces CHOP-dependent DR5 up-regulation. lonafarnib 33-43 TNF receptor superfamily member 10b Homo sapiens 67-70 17493934-13 2007 We conclude that CHOP-dependent DR5 up-regulation contributes to lonafarnib-induced apoptosis. lonafarnib 65-75 TNF receptor superfamily member 10b Homo sapiens 32-35 17510428-0 2007 The farnesyltransferase inhibitor R115777 up-regulates the expression of death receptor 5 and enhances TRAIL-induced apoptosis in human lung cancer cells. tipifarnib 34-41 TNF receptor superfamily member 10b Homo sapiens 73-89 17579120-11 2007 Halocynthiaxanthin markedly up-regulated a TRAIL receptor, death receptor 5 (DR5), among the death receptor-related genes, suggesting a possible mechanism for the combined effects. halocynthiaxanthin 0-18 TNF receptor superfamily member 10b Homo sapiens 59-75 17579120-11 2007 Halocynthiaxanthin markedly up-regulated a TRAIL receptor, death receptor 5 (DR5), among the death receptor-related genes, suggesting a possible mechanism for the combined effects. halocynthiaxanthin 0-18 TNF receptor superfamily member 10b Homo sapiens 77-80 17510429-0 2007 The proteasome inhibitor PS-341 (bortezomib) up-regulates DR5 expression leading to induction of apoptosis and enhancement of TRAIL-induced apoptosis despite up-regulation of c-FLIP and survivin expression in human NSCLC cells. Bortezomib 25-31 TNF receptor superfamily member 10b Homo sapiens 58-61 17510429-5 2007 Blockage of PS-341-induced DR5 up-regulation using DR5 small interfering RNA (siRNA) rendered cells less sensitive to apoptosis induced by either PS-341 or its combination with TRAIL, indicating that DR5 up-regulation mediates PS-341-induced apoptosis and enhancement of TRAIL-induced apoptosis in human NSCLC cells. Bortezomib 12-18 TNF receptor superfamily member 10b Homo sapiens 51-54 17510429-5 2007 Blockage of PS-341-induced DR5 up-regulation using DR5 small interfering RNA (siRNA) rendered cells less sensitive to apoptosis induced by either PS-341 or its combination with TRAIL, indicating that DR5 up-regulation mediates PS-341-induced apoptosis and enhancement of TRAIL-induced apoptosis in human NSCLC cells. Bortezomib 146-152 TNF receptor superfamily member 10b Homo sapiens 27-30 17510429-0 2007 The proteasome inhibitor PS-341 (bortezomib) up-regulates DR5 expression leading to induction of apoptosis and enhancement of TRAIL-induced apoptosis despite up-regulation of c-FLIP and survivin expression in human NSCLC cells. Bortezomib 33-43 TNF receptor superfamily member 10b Homo sapiens 58-61 17510429-5 2007 Blockage of PS-341-induced DR5 up-regulation using DR5 small interfering RNA (siRNA) rendered cells less sensitive to apoptosis induced by either PS-341 or its combination with TRAIL, indicating that DR5 up-regulation mediates PS-341-induced apoptosis and enhancement of TRAIL-induced apoptosis in human NSCLC cells. Bortezomib 146-152 TNF receptor superfamily member 10b Homo sapiens 27-30 17510429-3 2007 In the present study, we show that PS-341 induced caspase-8-dependent apoptosis, cooperated with TRAIL to induce apoptosis, and up-regulated death receptor 5 (DR5) expression in human non-small cell lung cancer (NSCLC) cells. Bortezomib 35-41 TNF receptor superfamily member 10b Homo sapiens 141-157 17510429-3 2007 In the present study, we show that PS-341 induced caspase-8-dependent apoptosis, cooperated with TRAIL to induce apoptosis, and up-regulated death receptor 5 (DR5) expression in human non-small cell lung cancer (NSCLC) cells. Bortezomib 35-41 TNF receptor superfamily member 10b Homo sapiens 159-162 17510429-4 2007 DR5 induction correlated with the ability of PS-341 to induce apoptosis. Bortezomib 45-51 TNF receptor superfamily member 10b Homo sapiens 0-3 17510429-5 2007 Blockage of PS-341-induced DR5 up-regulation using DR5 small interfering RNA (siRNA) rendered cells less sensitive to apoptosis induced by either PS-341 or its combination with TRAIL, indicating that DR5 up-regulation mediates PS-341-induced apoptosis and enhancement of TRAIL-induced apoptosis in human NSCLC cells. Bortezomib 12-18 TNF receptor superfamily member 10b Homo sapiens 27-30 17510429-5 2007 Blockage of PS-341-induced DR5 up-regulation using DR5 small interfering RNA (siRNA) rendered cells less sensitive to apoptosis induced by either PS-341 or its combination with TRAIL, indicating that DR5 up-regulation mediates PS-341-induced apoptosis and enhancement of TRAIL-induced apoptosis in human NSCLC cells. Bortezomib 12-18 TNF receptor superfamily member 10b Homo sapiens 51-54 17440103-8 2007 Finally, N-acetylcysteine, an inhibitor of ROS, inhibits sanguinarine-induced generation of ROS, up-regulation of DR5, Bax conformational changes, activation of caspase-3, and down-regulation of IAPs. Acetylcysteine 9-25 TNF receptor superfamily member 10b Homo sapiens 114-117 17440103-3 2007 Our data show that sanguinarine treatment of PEL cells results in up-regulation of death receptor 5 (DR5) expression via generation of reactive oxygen species (ROS) and causes activation of caspase-8 and truncation of Bid (tBid). sanguinarine 19-31 TNF receptor superfamily member 10b Homo sapiens 83-99 17440103-8 2007 Finally, N-acetylcysteine, an inhibitor of ROS, inhibits sanguinarine-induced generation of ROS, up-regulation of DR5, Bax conformational changes, activation of caspase-3, and down-regulation of IAPs. sanguinarine 57-69 TNF receptor superfamily member 10b Homo sapiens 114-117 17440103-3 2007 Our data show that sanguinarine treatment of PEL cells results in up-regulation of death receptor 5 (DR5) expression via generation of reactive oxygen species (ROS) and causes activation of caspase-8 and truncation of Bid (tBid). sanguinarine 19-31 TNF receptor superfamily member 10b Homo sapiens 101-104 17440103-3 2007 Our data show that sanguinarine treatment of PEL cells results in up-regulation of death receptor 5 (DR5) expression via generation of reactive oxygen species (ROS) and causes activation of caspase-8 and truncation of Bid (tBid). Reactive Oxygen Species 135-158 TNF receptor superfamily member 10b Homo sapiens 83-99 17440103-9 2007 Taken together, our findings suggest that sanguinarine is a potent inducer of apoptosis of PEL cells via up-regulation of DR5 and raise the possibility that this agent may be of value in the development of novel therapeutic approaches for the treatment of PEL. sanguinarine 42-54 TNF receptor superfamily member 10b Homo sapiens 122-125 17440103-3 2007 Our data show that sanguinarine treatment of PEL cells results in up-regulation of death receptor 5 (DR5) expression via generation of reactive oxygen species (ROS) and causes activation of caspase-8 and truncation of Bid (tBid). Reactive Oxygen Species 135-158 TNF receptor superfamily member 10b Homo sapiens 101-104 17440103-3 2007 Our data show that sanguinarine treatment of PEL cells results in up-regulation of death receptor 5 (DR5) expression via generation of reactive oxygen species (ROS) and causes activation of caspase-8 and truncation of Bid (tBid). Reactive Oxygen Species 160-163 TNF receptor superfamily member 10b Homo sapiens 83-99 17432717-9 2007 Expression of the death receptors DR5 and DR4 in two cell lines (A172 and U251) upregulated significantly when they were used in combination hTRAIL/TMZ treatment (p < 0.05 compared with baseline control), leading to activation of caspase-8 and caspase-3 (p < 0.05 compared with baseline control) and confirming an extrinsic apoptotic pathway. Temozolomide 148-151 TNF receptor superfamily member 10b Homo sapiens 34-37 17440103-3 2007 Our data show that sanguinarine treatment of PEL cells results in up-regulation of death receptor 5 (DR5) expression via generation of reactive oxygen species (ROS) and causes activation of caspase-8 and truncation of Bid (tBid). Reactive Oxygen Species 160-163 TNF receptor superfamily member 10b Homo sapiens 101-104 17409433-8 2007 Whereas treatment with R-flurbiprofen or ibuprofen resulted in a massive induction of p75(NTR) protein levels, the expression of Fas, p55(TNFR), DR3, DR4, DR5, and DR6 remained largely unchanged. Ibuprofen 41-50 TNF receptor superfamily member 10b Homo sapiens 155-158 17575157-0 2007 Tunicamycin sensitizes human melanoma cells to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by up-regulation of TRAIL-R2 via the unfolded protein response. Tunicamycin 0-11 TNF receptor superfamily member 10b Homo sapiens 141-149 17575157-3 2007 We show in this study that the endoplasmic reticulum (ER) stress inducer, tunicamycin, selectively up-regulated the cell surface expression of TRAIL-R2, but not other members of the TNF receptor family, and enhanced TRAIL-induced apoptosis in cultured melanoma cells and fresh melanoma isolates. Tunicamycin 74-85 TNF receptor superfamily member 10b Homo sapiens 143-151 17575157-4 2007 Tunicamycin-mediated sensitization of melanoma cells to TRAIL-induced apoptosis was associated with increased activation of the caspase cascade and reduction in mitochondrial membrane potential and was inhibited by a recombinant TRAIL-R2/Fc chimeric protein. Tunicamycin 0-11 TNF receptor superfamily member 10b Homo sapiens 229-237 17575157-9 2007 Moreover, the transcription factor CCAAT/enhancer-binding protein homologous protein seemed to be involved in the up-regulation of TRAIL-R2 by tunicamycin, but its role varied between different melanoma lines. Tunicamycin 143-154 TNF receptor superfamily member 10b Homo sapiens 131-139 16936710-6 2007 In addition, the increased expression of DR5 receptor after beta amyloid treatment was sustained by p53 transcriptional activity, as demonstrated by the data showing that the p53 inhibitor Pifithrin alpha prevented both beta amyloid-induced DR5 induction and cell death. pifithrin 189-204 TNF receptor superfamily member 10b Homo sapiens 41-44 17326159-6 2007 RNA interference and TRAIL receptor blockage experiments revealed that in bortezomib-treated hepatoma cells TRAIL-R1/TRAIL-R2 up-regulation, enhanced TRAIL DISC formation and cFLIPL down-regulation in addition to accumulation of Bak cooperatively sensitized for TRAIL. Bortezomib 74-84 TNF receptor superfamily member 10b Homo sapiens 117-125 17363481-0 2007 High-resolution single-photon emission computed tomography and X-ray computed tomography imaging of Tc-99m-labeled anti-DR5 antibody in breast tumor xenografts. Technetium 100-106 TNF receptor superfamily member 10b Homo sapiens 120-123 17273769-4 2007 In this study, we report that fenretinide induces death receptor 5 (DR5)/TRAIL-R2 up-regulation via the induction of the transcription factor CHOP in colon cancer cell lines. Fenretinide 30-41 TNF receptor superfamily member 10b Homo sapiens 73-81 17273769-5 2007 Fenretinide induced DR5 expression at protein and mRNA levels. Fenretinide 0-11 TNF receptor superfamily member 10b Homo sapiens 20-23 17273769-6 2007 Furthermore, fenretinide increased DR5 promoter activity and the enhanced activity decreased by mutation of the CHOP binding site. Fenretinide 13-24 TNF receptor superfamily member 10b Homo sapiens 35-38 17273769-10 2007 Fenretinide and TRAIL cooperatively activated caspase-3, -8, -10 and -9 and cleavage of Bid and PARP, and this activation was also blocked in the presence of DR5/Fc chimeric protein. Fenretinide 0-11 TNF receptor superfamily member 10b Homo sapiens 158-161 17273769-0 2007 Fenretinide up-regulates DR5/TRAIL-R2 expression via the induction of the transcription factor CHOP and combined treatment with fenretinide and TRAIL induces synergistic apoptosis in colon cancer cell lines. Fenretinide 0-11 TNF receptor superfamily member 10b Homo sapiens 25-28 17273769-0 2007 Fenretinide up-regulates DR5/TRAIL-R2 expression via the induction of the transcription factor CHOP and combined treatment with fenretinide and TRAIL induces synergistic apoptosis in colon cancer cell lines. Fenretinide 0-11 TNF receptor superfamily member 10b Homo sapiens 29-37 17273769-4 2007 In this study, we report that fenretinide induces death receptor 5 (DR5)/TRAIL-R2 up-regulation via the induction of the transcription factor CHOP in colon cancer cell lines. Fenretinide 30-41 TNF receptor superfamily member 10b Homo sapiens 50-66 17273769-4 2007 In this study, we report that fenretinide induces death receptor 5 (DR5)/TRAIL-R2 up-regulation via the induction of the transcription factor CHOP in colon cancer cell lines. Fenretinide 30-41 TNF receptor superfamily member 10b Homo sapiens 68-71 17363481-1 2007 A murine, apoptosis-inducing monoclonal antibody (mTRA-8) targeting human DR5 was radiolabeled with Tc-99m. mtra 50-54 TNF receptor superfamily member 10b Homo sapiens 74-77 17363481-1 2007 A murine, apoptosis-inducing monoclonal antibody (mTRA-8) targeting human DR5 was radiolabeled with Tc-99m. tc-99 100-105 TNF receptor superfamily member 10b Homo sapiens 74-77 17363481-4 2007 Scatchard assays showed specific and high binding affinity of Tc-99m-mTRA-8 to DR5; the killing efficacy of mTRA-8 was unchanged by Tc-99m labeling. tc-99m-mtra-8 62-75 TNF receptor superfamily member 10b Homo sapiens 79-82 17363481-4 2007 Scatchard assays showed specific and high binding affinity of Tc-99m-mTRA-8 to DR5; the killing efficacy of mTRA-8 was unchanged by Tc-99m labeling. mtra-8 69-75 TNF receptor superfamily member 10b Homo sapiens 79-82 17363481-4 2007 Scatchard assays showed specific and high binding affinity of Tc-99m-mTRA-8 to DR5; the killing efficacy of mTRA-8 was unchanged by Tc-99m labeling. tc-99 62-67 TNF receptor superfamily member 10b Homo sapiens 79-82 17286903-0 2007 [Enhancement of glioma cell apoptosis induced by anti-human DR5/DR4 monoclonal antibodies by sub-toxic dose of doxorubicin in human]. Doxorubicin 111-122 TNF receptor superfamily member 10b Homo sapiens 60-63 17105846-7 2007 Asoprisnil treatment significantly (P < 0.05) increased TRAIL, DR4, and DR5 contents in cultured leiomyoma cells in a dose-dependent manner with a cleavage of caspase-8, -7, and -3, and decreased X-linked chromosome-linked inhibitor of apoptosis protein contents. asoprisnil 0-10 TNF receptor superfamily member 10b Homo sapiens 75-78 17286903-7 2007 CONCLUSION: Subtoxic doxorubicin applied with antibodies caused higher cell death rate of glioma cells, which may be relevant to DR4/DR5, the release of cytochrome C and FLIP and Ca2+ concentration. Doxorubicin 21-32 TNF receptor superfamily member 10b Homo sapiens 133-136 17499001-8 2007 However, SAHA induces, in addition to p21(WAF1/CIP1) also re-expression of DR5. Vorinostat 9-13 TNF receptor superfamily member 10b Homo sapiens 75-78 17003375-7 2007 A Fas/CD95-deficient MM subline expressing DR4 and DR5 was resistant to edelfosine. ammonium ferrous sulfate 2-5 TNF receptor superfamily member 10b Homo sapiens 51-54 17849272-0 2007 The Effect of N-nitrosodimethylamine on TRAIL and DR5 expression in human neutrophils--preliminary study. Dimethylnitrosamine 14-36 TNF receptor superfamily member 10b Homo sapiens 50-53 17145888-6 2006 Treatment with As(2)O(3) plus BSO, but not with As(2)O(3) alone, increased the levels of death receptor (DR) 5 protein and caspase-8 cleavage. Arsenic Trioxide 15-24 TNF receptor superfamily member 10b Homo sapiens 89-110 17649723-6 2007 DR5 expression increased while P65 decreased as the doses of doxorubicin increased when KM3 cells treated with doxorubicin (0.5, 1.0, 2.0 and 4.0 microg/ml) plus 20 ng/ml TRAIL. Doxorubicin 61-72 TNF receptor superfamily member 10b Homo sapiens 0-3 17649723-6 2007 DR5 expression increased while P65 decreased as the doses of doxorubicin increased when KM3 cells treated with doxorubicin (0.5, 1.0, 2.0 and 4.0 microg/ml) plus 20 ng/ml TRAIL. Doxorubicin 111-122 TNF receptor superfamily member 10b Homo sapiens 0-3 17649723-7 2007 CONCLUSION: Increasing the expression of DR5 and nuclear transferring of P65 are the important molecular mechanism by which doxorubicin enhances TRAIL-inducing apoptosis of KM3 cells. Doxorubicin 124-135 TNF receptor superfamily member 10b Homo sapiens 41-44 17145853-3 2006 Our previous work has shown that celecoxib induces death receptor 5 expression, resulting in induction of apoptosis and enhancement of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in human lung cancer cells. Celecoxib 33-42 TNF receptor superfamily member 10b Homo sapiens 51-67 17143496-0 2007 Phytosphingosine in combination with TRAIL sensitizes cancer cells to TRAIL through synergistic up-regulation of DR4 and DR5. phytosphingosine 0-16 TNF receptor superfamily member 10b Homo sapiens 121-124 17143496-9 2007 These results indicate that phytosphingosine sensitizes cancer cells to TRAIL through the synergistic up-regulation of DR4 and DR5 in an NF-kappaB-dependent fashion resulting in caspase-8 activation and subsequent mitochondrial dysfunction. phytosphingosine 28-44 TNF receptor superfamily member 10b Homo sapiens 127-130 17070520-6 2006 Arsenite treatment upregulated surface levels of death receptors, TRAIL-R1 and TRAIL-R2, through increased translocation of these proteins from cytoplasm to the cell surface. arsenite 0-8 TNF receptor superfamily member 10b Homo sapiens 79-87 17145853-7 2006 Collectively, we conclude that celecoxib induces apoptosis in human lung cancer cells through activation of the extrinsic apoptotic pathway, primarily by induction of death receptor 5 and down-regulation of c-FLIP. Celecoxib 31-40 TNF receptor superfamily member 10b Homo sapiens 167-183 17145888-7 2006 The JNK inhibitor SP600125 inhibited the increase in DR5 protein and attenuated apoptosis induced by treatment with As(2)O(3) plus BSO. pyrazolanthrone 18-26 TNF receptor superfamily member 10b Homo sapiens 53-56 17257501-3 2006 Cytotoxicity exerted by FasL/anti-DR5 mAb on tumor cell lines was measured by MTT assay and the induced apoptosis was determined by agarose gel electrophoresis. monooxyethylene trimethylolpropane tristearate 78-81 TNF receptor superfamily member 10b Homo sapiens 34-37 16859704-4 2006 Despite great differences in overall size and structure, the DR5-binding peptides and antibodies shared a tripeptide motif, which was conserved within a disulfide-constrained loop of the peptides and the third complementarity determining region of the antibody heavy chains. tripeptide K-26 106-116 TNF receptor superfamily member 10b Homo sapiens 61-64 16435155-4 2006 TSA induced the transcription of TRAIL death receptor DR5 without affecting the transcription of DR4 and the decoy receptors; DcR1 and DcR2. trichostatin A 0-3 TNF receptor superfamily member 10b Homo sapiens 54-57 16571651-0 2006 Sulforaphane enhances TRAIL-induced apoptosis through the induction of DR5 expression in human osteosarcoma cells. sulforaphane 0-12 TNF receptor superfamily member 10b Homo sapiens 71-74 17148761-4 2006 Paclitaxel significantly increased in vivo expression of TRAIL-R1 and TRAIL-R2 in a time-dependent manner. Paclitaxel 0-10 TNF receptor superfamily member 10b Homo sapiens 70-78 17148761-5 2006 The imaging results were confirmed by immunoblots for steady-state protein levels (>20-fold increase in TRAIL-R1 and TRAIL-R2 levels in tumor xenografts by 48 h after paclitaxel administration). Paclitaxel 170-180 TNF receptor superfamily member 10b Homo sapiens 120-128 16613838-0 2006 Curcumin sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis through CHOP-independent DR5 upregulation. Curcumin 0-8 TNF receptor superfamily member 10b Homo sapiens 128-131 16613838-2 2006 In this study, we showed that curcumin, a plant product containing the phenolic phytochemical, is a potent enhancer of TRAIL-induced apoptosis through upregulation of DR5 expression. Curcumin 30-38 TNF receptor superfamily member 10b Homo sapiens 167-170 16613838-3 2006 Both treatment with DR5/Fc chimeric protein and silencing of DR5 expression using small interfering RNA (siRNA) attenuated curcumin plus TRAIL-induced apoptosis, showing that the critical role of DR5 in this cell death. Curcumin 123-131 TNF receptor superfamily member 10b Homo sapiens 20-23 16613838-3 2006 Both treatment with DR5/Fc chimeric protein and silencing of DR5 expression using small interfering RNA (siRNA) attenuated curcumin plus TRAIL-induced apoptosis, showing that the critical role of DR5 in this cell death. Curcumin 123-131 TNF receptor superfamily member 10b Homo sapiens 61-64 16613838-3 2006 Both treatment with DR5/Fc chimeric protein and silencing of DR5 expression using small interfering RNA (siRNA) attenuated curcumin plus TRAIL-induced apoptosis, showing that the critical role of DR5 in this cell death. Curcumin 123-131 TNF receptor superfamily member 10b Homo sapiens 61-64 16613838-6 2006 Taken together, the present study demonstrates that curcumin enhances TRAIL-induced apoptosis by CHOP-independent upregulation of DR5. Curcumin 52-60 TNF receptor superfamily member 10b Homo sapiens 130-133 16636669-3 2006 Ceramide-enriched membrane platforms serve to cluster DR5 upon stimulation. Ceramides 0-8 TNF receptor superfamily member 10b Homo sapiens 54-57 16636669-7 2006 Our data indicate that ceramide-enriched membrane platforms are required for the signaling of TRAIL-DR5 complexes under physiological conditions. Ceramides 23-31 TNF receptor superfamily member 10b Homo sapiens 100-103 16859704-4 2006 Despite great differences in overall size and structure, the DR5-binding peptides and antibodies shared a tripeptide motif, which was conserved within a disulfide-constrained loop of the peptides and the third complementarity determining region of the antibody heavy chains. Disulfides 153-162 TNF receptor superfamily member 10b Homo sapiens 61-64 16859704-5 2006 The X-ray crystal structure of an antibody in complex with DR5 revealed that the tripeptide motif is buried at the core of the interface, confirming its central role in antigen recognition. tripeptide K-26 81-91 TNF receptor superfamily member 10b Homo sapiens 59-62 16820931-8 2006 Flavopiridol up-regulated TRAIL, TRAIL-R1 and TRAIL-R2 in both cell lines. alvocidib 0-12 TNF receptor superfamily member 10b Homo sapiens 46-54 16550602-8 2006 Treatment with SAHA increased cell surface expression of DR5 but not DR4. Vorinostat 15-19 TNF receptor superfamily member 10b Homo sapiens 57-60 16891469-3 2006 Here, we report that 15d-PGJ(2) induces DR5 expression at both mRNA and protein levels, resulting in the synergistic sensitization of TRAIL-induced apoptosis in human neoplastic cells, such as Jurkat human leukemia cells or PC3 human prostate cancer cells. 15d-pgj 21-28 TNF receptor superfamily member 10b Homo sapiens 40-43 16923369-8 2006 Expression of DR5 protein was not detected in SKNDZ cells but an increased DR5 protein expression was found after treatment with adriamycin or etoposide. Doxorubicin 129-139 TNF receptor superfamily member 10b Homo sapiens 14-17 16923369-8 2006 Expression of DR5 protein was not detected in SKNDZ cells but an increased DR5 protein expression was found after treatment with adriamycin or etoposide. Doxorubicin 129-139 TNF receptor superfamily member 10b Homo sapiens 75-78 16923369-8 2006 Expression of DR5 protein was not detected in SKNDZ cells but an increased DR5 protein expression was found after treatment with adriamycin or etoposide. Etoposide 143-152 TNF receptor superfamily member 10b Homo sapiens 14-17 16923369-8 2006 Expression of DR5 protein was not detected in SKNDZ cells but an increased DR5 protein expression was found after treatment with adriamycin or etoposide. Etoposide 143-152 TNF receptor superfamily member 10b Homo sapiens 75-78 16753135-5 2006 DR5 but not DR4 engagement by TRAIL attenuated cellular ATP levels by robustly stimulating P-gp ATPase activity, and thus triggered P-gp-dependent apoptosis by depletion of the cellular ATP pool. Adenosine Triphosphate 56-59 TNF receptor superfamily member 10b Homo sapiens 0-3 16753135-5 2006 DR5 but not DR4 engagement by TRAIL attenuated cellular ATP levels by robustly stimulating P-gp ATPase activity, and thus triggered P-gp-dependent apoptosis by depletion of the cellular ATP pool. Adenosine Triphosphate 96-99 TNF receptor superfamily member 10b Homo sapiens 0-3 16699947-9 2006 Sulindac sulfide caused apoptosis, and induced expression of DR-5 and NAG-1 in Hep3B cells. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 61-65 16699949-3 2006 Herein, we have demonstrated that the induction of apoptosis by the proteasome inhibitors, MG-132 and PS-341 (bortezomib, Velcade), in primary CLL cells and the Burkitt lymphoma cell line, BJAB, is associated with up-regulation of TRAIL and its death receptors, DR4 and DR5. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 91-97 TNF receptor superfamily member 10b Homo sapiens 270-273 16699949-3 2006 Herein, we have demonstrated that the induction of apoptosis by the proteasome inhibitors, MG-132 and PS-341 (bortezomib, Velcade), in primary CLL cells and the Burkitt lymphoma cell line, BJAB, is associated with up-regulation of TRAIL and its death receptors, DR4 and DR5. Bortezomib 102-108 TNF receptor superfamily member 10b Homo sapiens 270-273 16699949-3 2006 Herein, we have demonstrated that the induction of apoptosis by the proteasome inhibitors, MG-132 and PS-341 (bortezomib, Velcade), in primary CLL cells and the Burkitt lymphoma cell line, BJAB, is associated with up-regulation of TRAIL and its death receptors, DR4 and DR5. Bortezomib 122-129 TNF receptor superfamily member 10b Homo sapiens 270-273 16699949-5 2006 MG-132 treatment increases binding of DR5 to the adaptor protein FADD, and causes caspase-8 activation and cleavage of pro-apoptotic BID. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-6 TNF receptor superfamily member 10b Homo sapiens 38-41 16699949-7 2006 MG-132 also increases apoptosis and DR5 expression in normal B-cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-6 TNF receptor superfamily member 10b Homo sapiens 36-39 16891471-6 2006 However, these cells did undergo apoptosis in response to another form of recombinant TRAIL, histidine-tagged TRAIL, suggesting differing contributions of DR4 and DR5 in the response to these two forms of TRAIL. Histidine 93-102 TNF receptor superfamily member 10b Homo sapiens 163-166 16891469-7 2006 DR5/Fc chimera protein, zVAD-fmk pancaspase inhibitor, and caspase-8 inhibitor efficiently blocked the activation of these apoptotic signal mediators and the induction of apoptotic cell death enhanced by cotreatment with 15d-PGJ(2) and TRAIL. 15d-pgj 221-228 TNF receptor superfamily member 10b Homo sapiens 0-3 16891469-8 2006 Moreover, a double-stranded small interfering RNA targeting DR5 gene, which suppressed DR5 up-regulation by 15d-PGJ(2), significantly attenuated apoptosis induced by cotreatment with 15d-PGJ(2) and TRAIL. 15d-pgj 108-115 TNF receptor superfamily member 10b Homo sapiens 60-63 16891469-8 2006 Moreover, a double-stranded small interfering RNA targeting DR5 gene, which suppressed DR5 up-regulation by 15d-PGJ(2), significantly attenuated apoptosis induced by cotreatment with 15d-PGJ(2) and TRAIL. 15d-pgj 108-115 TNF receptor superfamily member 10b Homo sapiens 87-90 16891469-8 2006 Moreover, a double-stranded small interfering RNA targeting DR5 gene, which suppressed DR5 up-regulation by 15d-PGJ(2), significantly attenuated apoptosis induced by cotreatment with 15d-PGJ(2) and TRAIL. 15d-pgj 183-190 TNF receptor superfamily member 10b Homo sapiens 60-63 16891469-8 2006 Moreover, a double-stranded small interfering RNA targeting DR5 gene, which suppressed DR5 up-regulation by 15d-PGJ(2), significantly attenuated apoptosis induced by cotreatment with 15d-PGJ(2) and TRAIL. 15d-pgj 183-190 TNF receptor superfamily member 10b Homo sapiens 87-90 16740726-5 2006 Sensitization to Apo2L/TRAIL mediated by cardiac glycosides was accompanied by up-regulation of death receptors 4 (DR4) and 5 (DR5) on both RNA and protein levels. Glycosides 49-59 TNF receptor superfamily member 10b Homo sapiens 127-130 17147249-0 2006 [Adriamycin enhances anti-human DR5 monoclonal antibody (mDRA-6) induced HL-60 cells apoptosis]. Doxorubicin 1-11 TNF receptor superfamily member 10b Homo sapiens 32-35 16740726-7 2006 Blocking of up-regulation of DR4 and DR5 alone significantly reduced cell death after combined treatment with cardiac glycosides and Apo2L/TRAIL. Glycosides 118-128 TNF receptor superfamily member 10b Homo sapiens 37-40 16740726-8 2006 Combined silencing of DR4 and DR5 abrogated the ability of cardiac glycosides and Apo2L/TRAIL to induce apoptosis in an additive manner. Glycosides 67-77 TNF receptor superfamily member 10b Homo sapiens 30-33 16740726-9 2006 To our knowledge, this is the first demonstration that glycosides up-regulate DR4 and DR5, thereby reverting the resistance of lung cancer cells to Apo2/TRAIL-induced apoptosis. Glycosides 55-65 TNF receptor superfamily member 10b Homo sapiens 86-89 16452234-0 2006 Sulforaphane sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-resistant hepatoma cells to TRAIL-induced apoptosis through reactive oxygen species-mediated up-regulation of DR5. sulforaphane 0-12 TNF receptor superfamily member 10b Homo sapiens 198-201 16582599-0 2006 Depletion of intracellular glutathione contributes to JNK-mediated death receptor 5 upregulation and apoptosis induction by the novel synthetic triterpenoid methyl-2-cyano-3, 12-dioxooleana-1, 9-dien-28-oate (CDDO-Me). Glutathione 27-38 TNF receptor superfamily member 10b Homo sapiens 67-83 16582599-0 2006 Depletion of intracellular glutathione contributes to JNK-mediated death receptor 5 upregulation and apoptosis induction by the novel synthetic triterpenoid methyl-2-cyano-3, 12-dioxooleana-1, 9-dien-28-oate (CDDO-Me). triterpenoid TP-222 144-156 TNF receptor superfamily member 10b Homo sapiens 67-83 16582599-0 2006 Depletion of intracellular glutathione contributes to JNK-mediated death receptor 5 upregulation and apoptosis induction by the novel synthetic triterpenoid methyl-2-cyano-3, 12-dioxooleana-1, 9-dien-28-oate (CDDO-Me). methyl-2-cyano-3, 12-dioxooleana-1, 9-dien-28-oate 157-207 TNF receptor superfamily member 10b Homo sapiens 67-83 16582599-2 2006 We previously demonstrated that CDDO-Me induces a c-Jun N-terminal kinase (JNK)-mediated DR5 expression and apoptosis. bardoxolone methyl 32-39 TNF receptor superfamily member 10b Homo sapiens 89-92 16582599-3 2006 This study revealed the mechanism by which CDDO-Me induces JNK activation and subsequent DR5 upregulation and apoptosis. bardoxolone methyl 43-50 TNF receptor superfamily member 10b Homo sapiens 89-92 16582599-4 2006 To determine whether CDDO-Me activates JNK and induces DR5 expression and apoptosis via oxidative stress by inducing the generation of reactive oxygen species (ROS), we examined the effects of various antioxidants on JNK activation, DR5 upregulation, and apoptosis induction by CDDO-Me. bardoxolone methyl 21-28 TNF receptor superfamily member 10b Homo sapiens 55-58 16582599-11 2006 Furthermore, these thiol antioxidants abrogated CDDO-Me-induced DR5 expression, whereas the GSH-depleting agent diethylmaleate also upregulated DR5 expression at concentrations that deplete intracellular GSH, demonstrating that GSH depletion can cause DR5 upregulation. Sulfhydryl Compounds 19-24 TNF receptor superfamily member 10b Homo sapiens 64-67 16582599-11 2006 Furthermore, these thiol antioxidants abrogated CDDO-Me-induced DR5 expression, whereas the GSH-depleting agent diethylmaleate also upregulated DR5 expression at concentrations that deplete intracellular GSH, demonstrating that GSH depletion can cause DR5 upregulation. bardoxolone methyl 48-55 TNF receptor superfamily member 10b Homo sapiens 64-67 16582599-11 2006 Furthermore, these thiol antioxidants abrogated CDDO-Me-induced DR5 expression, whereas the GSH-depleting agent diethylmaleate also upregulated DR5 expression at concentrations that deplete intracellular GSH, demonstrating that GSH depletion can cause DR5 upregulation. Glutathione 92-95 TNF receptor superfamily member 10b Homo sapiens 144-147 16582599-11 2006 Furthermore, these thiol antioxidants abrogated CDDO-Me-induced DR5 expression, whereas the GSH-depleting agent diethylmaleate also upregulated DR5 expression at concentrations that deplete intracellular GSH, demonstrating that GSH depletion can cause DR5 upregulation. Glutathione 92-95 TNF receptor superfamily member 10b Homo sapiens 144-147 16582599-12 2006 Collectively, we conclude that CDDO-Me activates the JNK pathway via depletion of intracellular GSH, leading to DR5 upregulation and induction of apoptosis. bardoxolone methyl 31-38 TNF receptor superfamily member 10b Homo sapiens 112-115 16582599-12 2006 Collectively, we conclude that CDDO-Me activates the JNK pathway via depletion of intracellular GSH, leading to DR5 upregulation and induction of apoptosis. Glutathione 96-99 TNF receptor superfamily member 10b Homo sapiens 112-115 16529749-0 2006 Alpha-tocopheryl succinate induces DR4 and DR5 expression by a p53-dependent route: implication for sensitisation of resistant cancer cells to TRAIL apoptosis. alpha-Tocopherol 0-26 TNF receptor superfamily member 10b Homo sapiens 43-46 16529749-2 2006 We show that alpha-TOS activates expression of DR4/DR5 in a p53-dependent manner and re-establishes sensitivity of resistant MM cells to TRAIL-mediated apoptosis, as documented in p53wt MM cells but not in their p53null counterparts. alpha-Tocopherol 13-22 TNF receptor superfamily member 10b Homo sapiens 51-54 16529749-4 2006 Treatment with sub-lethal doses of alpha-TOS restored expression of DR4 and DR5. alpha-Tocopherol 35-44 TNF receptor superfamily member 10b Homo sapiens 76-79 16332727-0 2006 3,3"-diindolylmethane (DIM) and its derivatives induce apoptosis in pancreatic cancer cells through endoplasmic reticulum stress-dependent upregulation of DR5. 3,3'-diindolylmethane 0-21 TNF receptor superfamily member 10b Homo sapiens 155-158 16332727-5 2006 The subsequent downstream effects of DIM/DIM-C-pPhtBu- and Tg-induced ER stress included CHOP-dependent induction of death receptor DR5 and subsequent cleavage of caspase 8, caspase 3, Bid and PARP. Thapsigargin 59-61 TNF receptor superfamily member 10b Homo sapiens 132-135 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 16434995-4 2006 Arsenic trioxide led to increased cell surface expression of DR5 (death receptor 5), inhibition of the serine/threonine kinase Akt and downregulation of the short isoform of FLIP (FLICE-inhibitory protein, FLIPS). Arsenic Trioxide 0-16 TNF receptor superfamily member 10b Homo sapiens 61-64 16434995-4 2006 Arsenic trioxide led to increased cell surface expression of DR5 (death receptor 5), inhibition of the serine/threonine kinase Akt and downregulation of the short isoform of FLIP (FLICE-inhibitory protein, FLIPS). Arsenic Trioxide 0-16 TNF receptor superfamily member 10b Homo sapiens 66-82 16452234-4 2006 We found that sulforaphane treatment significantly up-regulated mRNA and protein levels of DR5, a death receptor of TRAIL. sulforaphane 14-26 TNF receptor superfamily member 10b Homo sapiens 91-94 16452234-6 2006 Pretreatment with N-acetyl-l-cysteine and overexpression of catalase inhibited sulforaphane-induced up-regulation of DR5 and almost completely blocked the cotreatment-induced apoptosis. Acetylcysteine 18-37 TNF receptor superfamily member 10b Homo sapiens 117-120 16452234-6 2006 Pretreatment with N-acetyl-l-cysteine and overexpression of catalase inhibited sulforaphane-induced up-regulation of DR5 and almost completely blocked the cotreatment-induced apoptosis. sulforaphane 79-91 TNF receptor superfamily member 10b Homo sapiens 117-120 16452234-7 2006 Furthermore, the sulforaphane-mediated sensitization to TRAIL was efficiently reduced by administration of a blocking antibody or small interfering RNAs for DR5. sulforaphane 17-29 TNF receptor superfamily member 10b Homo sapiens 157-160 16452234-8 2006 These results collectively indicate that sulforaphane-induced generation of ROS and the subsequent up-regulation of DR5 are critical for triggering and amplifying TRAIL-induced apoptotic signaling. sulforaphane 41-53 TNF receptor superfamily member 10b Homo sapiens 116-119 16452234-0 2006 Sulforaphane sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-resistant hepatoma cells to TRAIL-induced apoptosis through reactive oxygen species-mediated up-regulation of DR5. Reactive Oxygen Species 148-171 TNF receptor superfamily member 10b Homo sapiens 198-201 16357153-3 2005 We have shown that COX-2 inhibition sensitizes human colon carcinoma cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by inducing clustering of the TRAIL receptor DR5 at the cell surface and the redistribution of the death-inducing signaling complex components (DR5, FADD, and procaspase-8) into cholesterol-rich and ceramide-rich domains known as caveolae. Cholesterol 338-349 TNF receptor superfamily member 10b Homo sapiens 205-208 16397266-6 2006 HDAC inhibition, especially with sodium butyrate and trichostatin A, led to increased TRAIL-R2 gene transcription that correlated with increased TRAIL-R2 surface expression. Butyric Acid 33-48 TNF receptor superfamily member 10b Homo sapiens 86-94 16397266-6 2006 HDAC inhibition, especially with sodium butyrate and trichostatin A, led to increased TRAIL-R2 gene transcription that correlated with increased TRAIL-R2 surface expression. Butyric Acid 33-48 TNF receptor superfamily member 10b Homo sapiens 145-153 16397266-6 2006 HDAC inhibition, especially with sodium butyrate and trichostatin A, led to increased TRAIL-R2 gene transcription that correlated with increased TRAIL-R2 surface expression. trichostatin A 53-67 TNF receptor superfamily member 10b Homo sapiens 86-94 16397266-6 2006 HDAC inhibition, especially with sodium butyrate and trichostatin A, led to increased TRAIL-R2 gene transcription that correlated with increased TRAIL-R2 surface expression. trichostatin A 53-67 TNF receptor superfamily member 10b Homo sapiens 145-153 16357153-0 2005 Cyclooxygenase-2 inhibition sensitizes human colon carcinoma cells to TRAIL-induced apoptosis through clustering of DR5 and concentrating death-inducing signaling complex components into ceramide-enriched caveolae. Ceramides 187-195 TNF receptor superfamily member 10b Homo sapiens 116-119 16357153-3 2005 We have shown that COX-2 inhibition sensitizes human colon carcinoma cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by inducing clustering of the TRAIL receptor DR5 at the cell surface and the redistribution of the death-inducing signaling complex components (DR5, FADD, and procaspase-8) into cholesterol-rich and ceramide-rich domains known as caveolae. Ceramides 359-367 TNF receptor superfamily member 10b Homo sapiens 205-208 15987718-0 2005 Curcumin sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through reactive oxygen species-mediated upregulation of death receptor 5 (DR5). Curcumin 0-8 TNF receptor superfamily member 10b Homo sapiens 159-175 15987718-0 2005 Curcumin sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through reactive oxygen species-mediated upregulation of death receptor 5 (DR5). Curcumin 0-8 TNF receptor superfamily member 10b Homo sapiens 177-180 16227396-7 2005 Given that sulindac sulfide activated caspase-8 and increased membrane death receptor (DR4 and DR5) protein levels, we evaluated its combination with the endogenous death receptor ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). sulindac sulfide 11-27 TNF receptor superfamily member 10b Homo sapiens 95-98 15987718-0 2005 Curcumin sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through reactive oxygen species-mediated upregulation of death receptor 5 (DR5). Reactive Oxygen Species 110-133 TNF receptor superfamily member 10b Homo sapiens 159-175 15987718-0 2005 Curcumin sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through reactive oxygen species-mediated upregulation of death receptor 5 (DR5). Reactive Oxygen Species 110-133 TNF receptor superfamily member 10b Homo sapiens 177-180 15987718-5 2005 We found that treatment with curcumin significantly induces death receptor 5 (DR5) expression both at its mRNA and protein levels, accompanying the generation of the reactive oxygen species (ROS). Curcumin 29-37 TNF receptor superfamily member 10b Homo sapiens 60-76 15987718-5 2005 We found that treatment with curcumin significantly induces death receptor 5 (DR5) expression both at its mRNA and protein levels, accompanying the generation of the reactive oxygen species (ROS). Curcumin 29-37 TNF receptor superfamily member 10b Homo sapiens 78-81 15987718-5 2005 We found that treatment with curcumin significantly induces death receptor 5 (DR5) expression both at its mRNA and protein levels, accompanying the generation of the reactive oxygen species (ROS). Reactive Oxygen Species 166-189 TNF receptor superfamily member 10b Homo sapiens 60-76 15987718-5 2005 We found that treatment with curcumin significantly induces death receptor 5 (DR5) expression both at its mRNA and protein levels, accompanying the generation of the reactive oxygen species (ROS). Reactive Oxygen Species 166-189 TNF receptor superfamily member 10b Homo sapiens 78-81 15987718-5 2005 We found that treatment with curcumin significantly induces death receptor 5 (DR5) expression both at its mRNA and protein levels, accompanying the generation of the reactive oxygen species (ROS). Reactive Oxygen Species 191-194 TNF receptor superfamily member 10b Homo sapiens 60-76 15987718-5 2005 We found that treatment with curcumin significantly induces death receptor 5 (DR5) expression both at its mRNA and protein levels, accompanying the generation of the reactive oxygen species (ROS). Reactive Oxygen Species 191-194 TNF receptor superfamily member 10b Homo sapiens 78-81 15987718-6 2005 Not only the pretreatment with N-acetylcystine but also the ectopic expression of peroxiredoxin II, an antioxidative protein, dramatically inhibited the apoptosis induced by curcumin and TRAIL in combination, blocking the curcumin-mediated DR5 upregulation. N-monoacetylcystine 31-46 TNF receptor superfamily member 10b Homo sapiens 240-243 15987718-6 2005 Not only the pretreatment with N-acetylcystine but also the ectopic expression of peroxiredoxin II, an antioxidative protein, dramatically inhibited the apoptosis induced by curcumin and TRAIL in combination, blocking the curcumin-mediated DR5 upregulation. Curcumin 174-182 TNF receptor superfamily member 10b Homo sapiens 240-243 15987718-7 2005 Taken together, the present study demonstrates that curcumin enhances TRAIL-induced apoptosis by ROS-mediated DR5 upregulation. Curcumin 52-60 TNF receptor superfamily member 10b Homo sapiens 110-113 15987718-7 2005 Taken together, the present study demonstrates that curcumin enhances TRAIL-induced apoptosis by ROS-mediated DR5 upregulation. Reactive Oxygen Species 97-100 TNF receptor superfamily member 10b Homo sapiens 110-113 16123594-4 2005 NFkappaB activation induces death receptor 5 (DR5) expression following DNA damaging agent etoposide treatment but not following growth factor EGF treatment. Etoposide 91-100 TNF receptor superfamily member 10b Homo sapiens 28-44 16123594-4 2005 NFkappaB activation induces death receptor 5 (DR5) expression following DNA damaging agent etoposide treatment but not following growth factor EGF treatment. Etoposide 91-100 TNF receptor superfamily member 10b Homo sapiens 46-49 16024638-4 2005 This effect was associated with the ability of CD437 to induce the expression of DR4 and DR5. CD 437 47-52 TNF receptor superfamily member 10b Homo sapiens 89-92 16098063-0 2005 Activity of selective fully human agonistic antibodies to the TRAIL death receptors TRAIL-R1 and TRAIL-R2 in primary and cultured lymphoma cells: induction of apoptosis and enhancement of doxorubicin- and bortezomib-induced cell death. Doxorubicin 188-199 TNF receptor superfamily member 10b Homo sapiens 97-105 16098063-0 2005 Activity of selective fully human agonistic antibodies to the TRAIL death receptors TRAIL-R1 and TRAIL-R2 in primary and cultured lymphoma cells: induction of apoptosis and enhancement of doxorubicin- and bortezomib-induced cell death. Bortezomib 205-215 TNF receptor superfamily member 10b Homo sapiens 97-105 16127151-5 2005 A specific role for p53 in TRAIL-R2-mediated apoptosis was indicated by the ability of p53 siRNA to significantly reduce RASF apoptosis and by the reduced apoptosis of RASFs bearing p53 mutations on treatment with anti-DR5 antibody or anti-DR5 antibody plus lactacystin. lactacystin 258-269 TNF receptor superfamily member 10b Homo sapiens 27-35 15963948-3 2005 Our previous studies have shown that luteolin, a naturally occurring flavonoid, induces the up-regulation of death receptor 5 (DR5), which is a receptor for TRAIL. Flavonoids 69-78 TNF receptor superfamily member 10b Homo sapiens 109-125 15963948-3 2005 Our previous studies have shown that luteolin, a naturally occurring flavonoid, induces the up-regulation of death receptor 5 (DR5), which is a receptor for TRAIL. Flavonoids 69-78 TNF receptor superfamily member 10b Homo sapiens 127-130 16024639-2 2005 In this study, we showed that tunicamycin, a naturally occurring antibiotic, is a potent enhancer of TRAIL-induced apoptosis through up-regulation of DR5 expression. Tunicamycin 30-41 TNF receptor superfamily member 10b Homo sapiens 150-153 16024639-4 2005 The tunicamycin-mediated enhancement of TRAIL-induced apoptosis was markedly blocked by a recombinant human DR5/Fc chimeric protein. Tunicamycin 4-15 TNF receptor superfamily member 10b Homo sapiens 108-111 16024639-5 2005 Tunicamycin and TRAIL cooperatively activated caspase-8, -10, -9, and -3 and Bid cleavage and this activation was also blocked in the presence of the DR5/Fc chimera. Tunicamycin 0-11 TNF receptor superfamily member 10b Homo sapiens 150-153 16024639-6 2005 Tunicamycin up-regulated DR5 expression at the mRNA and protein levels in a dose-dependent manner. Tunicamycin 0-11 TNF receptor superfamily member 10b Homo sapiens 25-28 16024639-7 2005 Furthermore, the tunicamycin-mediated sensitization to TRAIL was efficiently reduced by DR5 small interfering RNA, suggesting that the sensitization was mediated through induction of DR5 expression. Tunicamycin 17-28 TNF receptor superfamily member 10b Homo sapiens 88-91 16024639-7 2005 Furthermore, the tunicamycin-mediated sensitization to TRAIL was efficiently reduced by DR5 small interfering RNA, suggesting that the sensitization was mediated through induction of DR5 expression. Tunicamycin 17-28 TNF receptor superfamily member 10b Homo sapiens 183-186 16024639-8 2005 Tunicamycin increased DR5 promoter activity and this enhanced activity was diminished by mutation of a CHOP-binding site. Tunicamycin 0-11 TNF receptor superfamily member 10b Homo sapiens 22-25 16024639-9 2005 In addition, suppression of CHOP expression by small interfering RNA reduced the tunicamycin-mediated induction of DR5. Tunicamycin 81-92 TNF receptor superfamily member 10b Homo sapiens 115-118 16024639-10 2005 Of note, tunicamycin-mediated induction of CHOP and DR5 protein expression was not observed in normal human peripheral blood mononuclear cells. Tunicamycin 9-20 TNF receptor superfamily member 10b Homo sapiens 52-55 15994939-0 2005 Proteasome inhibitor MG132 induces death receptor 5 through CCAAT/enhancer-binding protein homologous protein. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 21-26 TNF receptor superfamily member 10b Homo sapiens 35-51 15994939-11 2005 Furthermore, CHOP small interfering RNA attenuated the DR5 up-regulation due to MG132. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 80-85 TNF receptor superfamily member 10b Homo sapiens 55-58 15994939-3 2005 In this study, we report that CCAAT/enhancer-binding protein homologous protein (CHOP) is a regulator of DR5 induction by proteasome inhibitor MG132. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 143-148 TNF receptor superfamily member 10b Homo sapiens 105-108 15994939-12 2005 These results indicate that the proteasome inhibitor MG132 induces DR5 expression through CHOP up-regulation. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 53-58 TNF receptor superfamily member 10b Homo sapiens 67-70 15994939-4 2005 MG132 induced DR5 expression at a protein and mRNA level in prostate cancer DU145 cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-5 TNF receptor superfamily member 10b Homo sapiens 14-17 15994939-5 2005 Furthermore, MG132 increased DR5 promoter activity. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 13-18 TNF receptor superfamily member 10b Homo sapiens 29-32 16082182-5 2005 Subsequent studies showed that combined treatment with bortezomib or MG132 resulted in an increase of death receptor (DR) 5 and Bik at protein levels but had no effects on protein levels of DR4, Bax, Bak, Bcl-2, Bcl-XL or Flice-inhibitory protein (FLIP). Bortezomib 55-65 TNF receptor superfamily member 10b Homo sapiens 102-123 15994939-6 2005 Using a series of deletion mutant plasmids containing DR5 promoters of various sizes, we found that MG132 stimulated the promoter activity via the region of -289 to -253. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 100-105 TNF receptor superfamily member 10b Homo sapiens 54-57 16082182-5 2005 Subsequent studies showed that combined treatment with bortezomib or MG132 resulted in an increase of death receptor (DR) 5 and Bik at protein levels but had no effects on protein levels of DR4, Bax, Bak, Bcl-2, Bcl-XL or Flice-inhibitory protein (FLIP). benzyloxycarbonylleucyl-leucyl-leucine aldehyde 69-74 TNF receptor superfamily member 10b Homo sapiens 102-123 15994939-9 2005 An electrophoretic mobility shift assay showed that CHOP directly bound to the MG132-responsive site on the DR5 promoter. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 79-84 TNF receptor superfamily member 10b Homo sapiens 108-111 16082182-7 2005 Blocking JNK activation with the JNK inhibitor SP600125 attenuated DR5 increase, but enhancement of apoptosis induction and increase of Bik protein were not affected. pyrazolanthrone 47-55 TNF receptor superfamily member 10b Homo sapiens 67-70 15930300-7 2005 From a mechanistic standpoint, we show that CDDO and CDDO-Im down-regulate the antiapoptotic protein c-FLIP(L), and up-regulate cell surface TRAIL receptors DR4 and DR5. 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid 44-48 TNF receptor superfamily member 10b Homo sapiens 165-168 15942663-4 2005 LY294002 increased Hep3B cell susceptibility to chemotherapy-induced apoptosis by enhancing the expression of DR4 and DR5 and the activation of caspase-8 and -3. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 0-8 TNF receptor superfamily member 10b Homo sapiens 118-121 15964798-4 2005 Herein, we determined that etoposide-induced DR5 expression requires the first intronic region of the DR5 gene. Etoposide 27-36 TNF receptor superfamily member 10b Homo sapiens 45-48 15964798-4 2005 Herein, we determined that etoposide-induced DR5 expression requires the first intronic region of the DR5 gene. Etoposide 27-36 TNF receptor superfamily member 10b Homo sapiens 102-105 16026644-2 2005 Low doses of SAHA or TSA enhanced the cytotoxic and apoptotic effects of TRAIL and upregulated the surface expression of TRAIL death receptors (DR4 and/or DR5). trichostatin A 21-24 TNF receptor superfamily member 10b Homo sapiens 155-158 15850806-11 2005 Deletion and mutation of the DR5, but not the IR6 element, abolished the atRA-mediated activity. Tretinoin 73-77 TNF receptor superfamily member 10b Homo sapiens 29-32 15850806-13 2005 In addition to the functional DR5, the region contains many other potential sequence elements that are required to maximize the atRA-mediated induction. Tretinoin 128-132 TNF receptor superfamily member 10b Homo sapiens 30-33 15930300-7 2005 From a mechanistic standpoint, we show that CDDO and CDDO-Im down-regulate the antiapoptotic protein c-FLIP(L), and up-regulate cell surface TRAIL receptors DR4 and DR5. 1-(2-cyano-3,12-dioxooleana-1,9-dien-28-oyl) imidazole 53-60 TNF receptor superfamily member 10b Homo sapiens 165-168 15956246-7 2005 Many gene expression changes observed following in vitro SN-38 exposure were also seen following in vivo administration of 10 or 15 mg/m(2) CPT-11; notably, proapoptotic changes included reduced transcription of survivin pathway-associated genes and increased transcription of death receptor 5. Irinotecan 57-62 TNF receptor superfamily member 10b Homo sapiens 277-293 15956246-7 2005 Many gene expression changes observed following in vitro SN-38 exposure were also seen following in vivo administration of 10 or 15 mg/m(2) CPT-11; notably, proapoptotic changes included reduced transcription of survivin pathway-associated genes and increased transcription of death receptor 5. Irinotecan 140-146 TNF receptor superfamily member 10b Homo sapiens 277-293 15767547-8 2005 The mRNAs of death receptors 4 and 5 (DR4 and DR5) were induced in leukemia cells undergoing apoptosis after boswellic acid acetate treatment. boswellic acid acetate 109-131 TNF receptor superfamily member 10b Homo sapiens 46-49 15339846-4 2005 In addition, CD40 ligation in vitro and in vivo induces CLL cells to express the proapoptotic protein, BH3 interacting domain death agonist (Bid), which can facilitate crosstalk between mitochondrial-dependent, apoptosis-inducing pathways and death receptors, such as death receptor 5 (DR5). BH 3 103-106 TNF receptor superfamily member 10b Homo sapiens 268-284 15339846-4 2005 In addition, CD40 ligation in vitro and in vivo induces CLL cells to express the proapoptotic protein, BH3 interacting domain death agonist (Bid), which can facilitate crosstalk between mitochondrial-dependent, apoptosis-inducing pathways and death receptors, such as death receptor 5 (DR5). BH 3 103-106 TNF receptor superfamily member 10b Homo sapiens 286-289 15806306-0 2005 IFN-gamma/JAK/STAT pathway-induced inhibition of DR4 and DR5 expression on endothelial cells is cancelled by cycloheximide-sensitive mechanism: novel finding of cycloheximide-regulating death receptor expression. Cycloheximide 109-122 TNF receptor superfamily member 10b Homo sapiens 57-60 15806306-6 2005 IFN-gamma/JAK/STAT-induced suppression was regulated by cycloheximide (CHX)-sensitive mechanism since the use of CHX mimicked the action of chemical inhibition of JAK in regard to DR4/DR5 expression as well as TRAIL-mediated endothelial cell apoptosis. Cycloheximide 56-69 TNF receptor superfamily member 10b Homo sapiens 184-187 15806306-6 2005 IFN-gamma/JAK/STAT-induced suppression was regulated by cycloheximide (CHX)-sensitive mechanism since the use of CHX mimicked the action of chemical inhibition of JAK in regard to DR4/DR5 expression as well as TRAIL-mediated endothelial cell apoptosis. Cycloheximide 71-74 TNF receptor superfamily member 10b Homo sapiens 184-187 15949263-5 2005 The expression of DR5 on MM and KM3 cells was up-regulated after chemotherapy and exposure to doxorubicin (P < 0. Doxorubicin 94-105 TNF receptor superfamily member 10b Homo sapiens 18-21 15949263-8 2005 Up-regulated DR5 on KM3 cells after incubating with doxorubicin and after chemotherapy suggests the cytotoxic agents might enhance the apoptosis of MM cells. Doxorubicin 52-63 TNF receptor superfamily member 10b Homo sapiens 13-16 15767547-9 2005 These data taken together suggest that boswellic acid acetate induces myeloid leukemia cell apoptosis through activation of caspase-8 by induced expression of DR4 and DR5, and that the activated caspase-8 either directly activates caspase-3 by cleavage or indirectly by cleaving Bid, which in turn decreases mitochondria membrane potential. boswellic acid acetate 39-61 TNF receptor superfamily member 10b Homo sapiens 167-170 15735044-6 2005 Fenretinide treatment also increased levels of the death receptor DR5 and caused mitochondrial membrane depolarization. Fenretinide 0-11 TNF receptor superfamily member 10b Homo sapiens 66-69 15802865-5 2005 The post-irradiation treatment of the cells with N-acetyl-L-cysteine (NAC) abolished the up-regulation of the expression of Fas and DR5 on the plasma membrane. Acetylcysteine 49-68 TNF receptor superfamily member 10b Homo sapiens 132-135 15735044-9 2005 We conclude that fenretinide induces apoptosis in all three histologic subtypes of meningioma and exerts diverse cellular effects, including DR5 up-regulation, modulation of retinoid receptor levels, and inhibition of IGF-I-induced proliferation. Fenretinide 17-28 TNF receptor superfamily member 10b Homo sapiens 141-144 15572759-10 2004 Celecoxib treatment induced the expression of death receptors, particularly that of DR5. Celecoxib 0-9 TNF receptor superfamily member 10b Homo sapiens 84-87 15748438-3 2005 The results showed that (1) TRAIL, DR4 and DR5 were highly expressed in both patient group and CR group, while the DcR1 and DcR2 were poorly expressed. Chromium 95-97 TNF receptor superfamily member 10b Homo sapiens 43-46 15748438-4 2005 (2) The level of DR5 expression in CR group was higher than that in patient group. Chromium 35-37 TNF receptor superfamily member 10b Homo sapiens 17-20 15748438-5 2005 (3) The level of DR5 was higher than DR4 in both patient group and CR group. Chromium 67-69 TNF receptor superfamily member 10b Homo sapiens 17-20 15897917-4 2005 TRAIL receptor-1 (DR4) and TRAIL receptor-2 (DR5) were upregulated under the treatment of doxorubicin and verified to be responsible for TRAIL-mediated signal transduction. Doxorubicin 90-101 TNF receptor superfamily member 10b Homo sapiens 45-48 15719269-4 2005 Our data show that gliomas preferentially express TRAIL R2 and that treatment with topotecan, a topoisomerase I inhibitor, significantly up-regulates its expression as detected by flow cytometry and western blotting. Topotecan 83-92 TNF receptor superfamily member 10b Homo sapiens 50-58 15572759-11 2004 Overexpression of a dominant negative Fas-associated death domain mutant, but not of BCL2, reduced the level of celecoxib-induced apoptosis, and silencing of DR5 expression by DR5 siRNA suppressed celecoxib-induced caspase 8 activation and apoptosis. Celecoxib 197-206 TNF receptor superfamily member 10b Homo sapiens 158-161 15572759-11 2004 Overexpression of a dominant negative Fas-associated death domain mutant, but not of BCL2, reduced the level of celecoxib-induced apoptosis, and silencing of DR5 expression by DR5 siRNA suppressed celecoxib-induced caspase 8 activation and apoptosis. Celecoxib 197-206 TNF receptor superfamily member 10b Homo sapiens 176-179 15322075-2 2004 We and others have reported previously that DR5 expression is up-regulated in thapsigargin (THG)-treated human cancer cells. Thapsigargin 78-90 TNF receptor superfamily member 10b Homo sapiens 44-47 15480430-0 2004 Redistribution of CD95, DR4 and DR5 in rafts accounts for the synergistic toxicity of resveratrol and death receptor ligands in colon carcinoma cells. Resveratrol 86-97 TNF receptor superfamily member 10b Homo sapiens 32-35 15322075-2 2004 We and others have reported previously that DR5 expression is up-regulated in thapsigargin (THG)-treated human cancer cells. Thapsigargin 92-95 TNF receptor superfamily member 10b Homo sapiens 44-47 15322075-6 2004 In addition to HCT116 cells, inhibition of CHOP or DR5 induction also attenuated THG-induced cell death in other cancer cell lines including LNCaP, A2780S, and DU145, indicating that CHOP and DR5 are critical for ER stress-mediated apoptosis in human carcinoma cells. Thapsigargin 81-84 TNF receptor superfamily member 10b Homo sapiens 192-195 15322075-3 2004 Here, we provide evidence that CHOP is involved in THG up-regulation of DR5, which is a critical step for ER stress-induced apoptosis in human cancer cells. Thapsigargin 51-54 TNF receptor superfamily member 10b Homo sapiens 72-75 15322075-4 2004 In human colon cancer HCT116 cells, knockdown of DR5 by siRNA blocked THG-induced Bax conformational change along with caspase-3 activation and cell death. Thapsigargin 70-73 TNF receptor superfamily member 10b Homo sapiens 49-52 15322075-5 2004 Moreover, inhibition of CHOP expression attenuated DR5 up-regulation and apoptosis induced by THG, whereas ectopic expression of DR5 restored the sensitivity of CHOP siRNA-transfected cells to THG-induced apoptosis. Thapsigargin 193-196 TNF receptor superfamily member 10b Homo sapiens 129-132 15322075-6 2004 In addition to HCT116 cells, inhibition of CHOP or DR5 induction also attenuated THG-induced cell death in other cancer cell lines including LNCaP, A2780S, and DU145, indicating that CHOP and DR5 are critical for ER stress-mediated apoptosis in human carcinoma cells. Thapsigargin 81-84 TNF receptor superfamily member 10b Homo sapiens 51-54 15334061-7 2004 Knockdown of FLIP expression in MM cell lines, by a FLIPsiRNA, re-established the normal response to apoptosis induced by Fas or DR4/DR5, which was blocked by pretreatment with the caspase-8 inhibitor z-IETD-fmk. benzyloxycarbonyl-isoleucyl-glutamyl-threonyl-aspartic acid fluoromethyl ketone 201-211 TNF receptor superfamily member 10b Homo sapiens 133-136 15492278-6 2004 In addition, some of the changes observed in SWB77 were opposite to those seen in methionine-dependent tumors, including expression of p21, TRAIL-R2, and TIEG. Methionine 82-92 TNF receptor superfamily member 10b Homo sapiens 140-148 15180942-7 2004 Moreover, temporal expressions of DR-4 and DR-5 induced by sulindac sulfide were similar to that of NAG-1. sulindac sulfide 59-75 TNF receptor superfamily member 10b Homo sapiens 43-47 15142888-0 2004 Sodium butyrate sensitizes TRAIL-mediated apoptosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon cancer cells. Butyric Acid 0-15 TNF receptor superfamily member 10b Homo sapiens 91-94 15492284-0 2004 c-Jun NH2-terminal kinase-mediated up-regulation of death receptor 5 contributes to induction of apoptosis by the novel synthetic triterpenoid methyl-2-cyano-3,12-dioxooleana-1, 9-dien-28-oate in human lung cancer cells. triterpenoid TP-222 130-142 TNF receptor superfamily member 10b Homo sapiens 52-68 15492284-0 2004 c-Jun NH2-terminal kinase-mediated up-regulation of death receptor 5 contributes to induction of apoptosis by the novel synthetic triterpenoid methyl-2-cyano-3,12-dioxooleana-1, 9-dien-28-oate in human lung cancer cells. methyl-2-cyano-3,12-dioxooleana-1, 9-dien-28-oate 143-192 TNF receptor superfamily member 10b Homo sapiens 52-68 15492284-4 2004 We found that CDDO-Me not only activated caspase-8 but also induced expression of DRs, particularly DR5, in a p53-independent mechanism. bardoxolone methyl 14-21 TNF receptor superfamily member 10b Homo sapiens 100-103 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 98-105 TNF receptor superfamily member 10b Homo sapiens 23-26 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 133-140 TNF receptor superfamily member 10b Homo sapiens 23-26 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 133-140 TNF receptor superfamily member 10b Homo sapiens 23-26 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 133-140 TNF receptor superfamily member 10b Homo sapiens 23-26 15142888-3 2004 In this study, we demonstrate for the first time that sodium butyrate selectively up-regulated DR5 but had no effect on the expression of the other TNF-alpha-related apoptosis-inducing ligand (TRAIL) receptor, DR4. Butyric Acid 54-69 TNF receptor superfamily member 10b Homo sapiens 95-98 15142888-4 2004 Sodium butyrate-induced expression of DR5 involves the putative Sp1 site within the DR5 promoter region. Butyric Acid 0-15 TNF receptor superfamily member 10b Homo sapiens 38-41 15374982-0 2004 Inducible silencing of KILLER/DR5 in vivo promotes bioluminescent colon tumor xenograft growth and confers resistance to chemotherapeutic agent 5-fluorouracil. Fluorouracil 144-158 TNF receptor superfamily member 10b Homo sapiens 23-33 15142888-4 2004 Sodium butyrate-induced expression of DR5 involves the putative Sp1 site within the DR5 promoter region. Butyric Acid 0-15 TNF receptor superfamily member 10b Homo sapiens 84-87 15142888-5 2004 Using a combination of the electrophoretic mobility shift assay and the luciferase reporter assay, we found that a specific Sp1 site (located at -195 bp relative to the transcription start site) is required for sodium butyrate-mediated activation of the DR5 promoter. Butyric Acid 211-226 TNF receptor superfamily member 10b Homo sapiens 254-257 15208660-3 2004 Here, we report that histone deacetylase inhibitors (HDACIs) such as trichostatin A (TSA), sodium butyrate, and suberoylanilide hydroxamic acid (SAHA) upregulated DR5 expression in various human malignant tumor cells. trichostatin A 69-83 TNF receptor superfamily member 10b Homo sapiens 163-166 15155747-11 2004 Interestingly, TRAIL-resistant cells were resensitized to TRAIL by tunicamycin pretreatment, which increased cell surface expression of DR4 and KILLER/DR5. Tunicamycin 67-78 TNF receptor superfamily member 10b Homo sapiens 144-154 15208660-3 2004 Here, we report that histone deacetylase inhibitors (HDACIs) such as trichostatin A (TSA), sodium butyrate, and suberoylanilide hydroxamic acid (SAHA) upregulated DR5 expression in various human malignant tumor cells. trichostatin A 85-88 TNF receptor superfamily member 10b Homo sapiens 163-166 15208660-3 2004 Here, we report that histone deacetylase inhibitors (HDACIs) such as trichostatin A (TSA), sodium butyrate, and suberoylanilide hydroxamic acid (SAHA) upregulated DR5 expression in various human malignant tumor cells. Butyric Acid 91-106 TNF receptor superfamily member 10b Homo sapiens 163-166 15208660-3 2004 Here, we report that histone deacetylase inhibitors (HDACIs) such as trichostatin A (TSA), sodium butyrate, and suberoylanilide hydroxamic acid (SAHA) upregulated DR5 expression in various human malignant tumor cells. Vorinostat 112-143 TNF receptor superfamily member 10b Homo sapiens 163-166 15208660-3 2004 Here, we report that histone deacetylase inhibitors (HDACIs) such as trichostatin A (TSA), sodium butyrate, and suberoylanilide hydroxamic acid (SAHA) upregulated DR5 expression in various human malignant tumor cells. Vorinostat 145-149 TNF receptor superfamily member 10b Homo sapiens 163-166 15201983-4 2004 We showed that the expression of death receptor DR5, but not DR4 was up-regulated and the decoy receptor DcR1 was down-regulated in the cells treated with 8-Cl-Ado and the recombinant soluble TRAIL (rsTRAIL, 95-281 a.a.). 8-chloroadenosine 155-163 TNF receptor superfamily member 10b Homo sapiens 48-51 15201983-8 2004 Our results provided evidence for the first time that 8-Cl-Ado sensitizes the human hepatoma cells BEL-7402 to rsTRAIL-induced apoptosis by up-regulating DR5 expression, inactivating the NF-kappaB activity, and signaling by the caspase-dependent and -independent pathway. 8-chloroadenosine 54-62 TNF receptor superfamily member 10b Homo sapiens 154-157 14739940-6 2004 Preincubation with E-17-beta completely prevented both TRAIL-induced DR4 and DR5 upregulation and apoptosis. e-17-beta 19-28 TNF receptor superfamily member 10b Homo sapiens 77-80 15077162-6 2004 This cytotoxicity was partially inhibited by addition of TRAILR2/Fc fusion protein, indicating that the secreted TRAIL mediates, at least in part, the apoptotic effect displayed by the supernatants of stimulated SK-N-MC cells. sk-n-mc 212-219 TNF receptor superfamily member 10b Homo sapiens 57-64 14691451-0 2004 Proteasome inhibitor MG132 upregulates death receptor 5 and cooperates with Apo2L/TRAIL to induce apoptosis in Bax-proficient and -deficient cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 21-26 TNF receptor superfamily member 10b Homo sapiens 39-55 14561739-0 2004 Bile acids up-regulate death receptor 5/TRAIL-receptor 2 expression via a c-Jun N-terminal kinase-dependent pathway involving Sp1. Bile Acids and Salts 0-10 TNF receptor superfamily member 10b Homo sapiens 23-39 14691451-5 2004 Here, we report that proteasome inhibitor MG132 upregulates Apo2L/TRAIL death receptor 5 expression in both Bax-proficient and -deficient HCT116 cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 42-47 TNF receptor superfamily member 10b Homo sapiens 72-88 14691451-8 2004 These results suggest that Bax is not absolutely required for death receptor 5-dependent apoptotic signals and MG132 by upregulating DR5 effectively cooperates with Apo2L/TRAIL to overcome Bax deficiency-induced resistance to Apo2L/TRAIL. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 111-116 TNF receptor superfamily member 10b Homo sapiens 133-136 14561739-1 2004 Bile acids up-regulate death receptor 5 (DR5)/TRAIL-receptor 2 (TRAIL-R2) expression thereby sensitizing hepatocytes to TRAIL-mediated apoptosis. Bile Acids and Salts 0-10 TNF receptor superfamily member 10b Homo sapiens 23-39 14561739-1 2004 Bile acids up-regulate death receptor 5 (DR5)/TRAIL-receptor 2 (TRAIL-R2) expression thereby sensitizing hepatocytes to TRAIL-mediated apoptosis. Bile Acids and Salts 0-10 TNF receptor superfamily member 10b Homo sapiens 41-44 14561739-1 2004 Bile acids up-regulate death receptor 5 (DR5)/TRAIL-receptor 2 (TRAIL-R2) expression thereby sensitizing hepatocytes to TRAIL-mediated apoptosis. Bile Acids and Salts 0-10 TNF receptor superfamily member 10b Homo sapiens 64-72 14561739-2 2004 However, the precise mechanism by which bile acids enhance DR5/TRAIL-R2 expression is unknown. Bile Acids and Salts 40-50 TNF receptor superfamily member 10b Homo sapiens 59-62 14561739-2 2004 However, the precise mechanism by which bile acids enhance DR5/TRAIL-R2 expression is unknown. Bile Acids and Salts 40-50 TNF receptor superfamily member 10b Homo sapiens 63-71 14561739-3 2004 Although several bile acids enhanced DR5/TRAIL-R2 expression, deoxycholic acid (DCA) was the most potent. Bile Acids and Salts 17-27 TNF receptor superfamily member 10b Homo sapiens 37-40 14561739-3 2004 Although several bile acids enhanced DR5/TRAIL-R2 expression, deoxycholic acid (DCA) was the most potent. Bile Acids and Salts 17-27 TNF receptor superfamily member 10b Homo sapiens 41-49 14561739-4 2004 DCA stimulated JNK activation and the JNK inhibitor SP600125 blocked DCA-induced DR5/TRAIL-R2 mRNA and protein expression. Deoxycholic Acid 0-3 TNF receptor superfamily member 10b Homo sapiens 81-84 14561739-4 2004 DCA stimulated JNK activation and the JNK inhibitor SP600125 blocked DCA-induced DR5/TRAIL-R2 mRNA and protein expression. Deoxycholic Acid 0-3 TNF receptor superfamily member 10b Homo sapiens 85-93 14561739-4 2004 DCA stimulated JNK activation and the JNK inhibitor SP600125 blocked DCA-induced DR5/TRAIL-R2 mRNA and protein expression. pyrazolanthrone 52-60 TNF receptor superfamily member 10b Homo sapiens 81-84 14561739-4 2004 DCA stimulated JNK activation and the JNK inhibitor SP600125 blocked DCA-induced DR5/TRAIL-R2 mRNA and protein expression. pyrazolanthrone 52-60 TNF receptor superfamily member 10b Homo sapiens 85-93 14561739-4 2004 DCA stimulated JNK activation and the JNK inhibitor SP600125 blocked DCA-induced DR5/TRAIL-R2 mRNA and protein expression. Deoxycholic Acid 69-72 TNF receptor superfamily member 10b Homo sapiens 81-84 14561739-4 2004 DCA stimulated JNK activation and the JNK inhibitor SP600125 blocked DCA-induced DR5/TRAIL-R2 mRNA and protein expression. Deoxycholic Acid 69-72 TNF receptor superfamily member 10b Homo sapiens 85-93 14561739-7 2004 JNK inhibition with SP600125 also blocked binding of Sp1 to the DR5/TRAIL-R2 promoter. pyrazolanthrone 20-28 TNF receptor superfamily member 10b Homo sapiens 64-67 14561739-7 2004 JNK inhibition with SP600125 also blocked binding of Sp1 to the DR5/TRAIL-R2 promoter. pyrazolanthrone 20-28 TNF receptor superfamily member 10b Homo sapiens 68-76 14561739-9 2004 In conclusion, Sp1 is a bile acid-responsive transcription factor that mediates DR5/TRAIL-R2 gene expression downstream of JNK. Bile Acids and Salts 24-33 TNF receptor superfamily member 10b Homo sapiens 80-83 14561739-9 2004 In conclusion, Sp1 is a bile acid-responsive transcription factor that mediates DR5/TRAIL-R2 gene expression downstream of JNK. Bile Acids and Salts 24-33 TNF receptor superfamily member 10b Homo sapiens 84-92 12904602-8 2003 Western blot and flow cytometric analyses revealed that doxorubicin significantly increased the expression of DR5, and somewhat up-regulated the expression of DR4 and DcR2. Doxorubicin 56-67 TNF receptor superfamily member 10b Homo sapiens 110-113 14500373-4 2003 The chemotherapeutic drugs (paclitaxel, vincristine, vinblastine, etoposide, camptothecin, and Adriamycin) induced death receptors (DRs) TRAIL receptor 1/DR4 and TRAIL receptor 2/DR5, and successive treatment with TRAIL resulted in apoptosis of both TRAIL-sensitive and -resistant cells. Paclitaxel 28-38 TNF receptor superfamily member 10b Homo sapiens 179-182 14720446-8 2003 Vp16 or Ara-C upregulated DR5 gene expression and augmented Apo2L-induced apoptosis in HL-60 cells. Cytarabine 8-13 TNF receptor superfamily member 10b Homo sapiens 26-29 14720446-10 2003 Ara-C or Vp16 upregulated DR5 gene expression and increased the sensitivity of HL-60 to Apo2L-induced cytotoxicity. Cytarabine 0-5 TNF receptor superfamily member 10b Homo sapiens 26-29 14520473-4 2003 We also show that the caspase-9-specific inhibitor z-LEHD-fmk inhibits drug-mediated apoptosis, leading to decreased PARP and caspase-3 cleavage, and reduced DR5 expression. benzyloxycarbonyl-leucyl-glutamyl-histidyl-aspartic acid fluoromethyl ketone 51-61 TNF receptor superfamily member 10b Homo sapiens 158-161 12811515-12 2003 Furthermore, the increase in TRAIL/Apo-2L sensitivity following topotecan treatment correlated with increased expression of TRAIL-R1 and TRAIL-R2 and decreased intracellular levels of the antiapoptotic protein survivin. Topotecan 64-73 TNF receptor superfamily member 10b Homo sapiens 137-145 12811515-14 2003 The sensitivity of DU-145 cells to TRAIL/Apo-2L was dramatically increased when combined with topotecan, suggesting that low-dose topotecan treatment to upregulate TRAIL-R1 and TRAIL-R2 and downregulate survivin, followed by TRAIL/Apo-2L administration, may be a viable therapy for treating cancer of the prostate. Topotecan 130-139 TNF receptor superfamily member 10b Homo sapiens 177-185 14500373-4 2003 The chemotherapeutic drugs (paclitaxel, vincristine, vinblastine, etoposide, camptothecin, and Adriamycin) induced death receptors (DRs) TRAIL receptor 1/DR4 and TRAIL receptor 2/DR5, and successive treatment with TRAIL resulted in apoptosis of both TRAIL-sensitive and -resistant cells. Vincristine 40-51 TNF receptor superfamily member 10b Homo sapiens 179-182 14500373-4 2003 The chemotherapeutic drugs (paclitaxel, vincristine, vinblastine, etoposide, camptothecin, and Adriamycin) induced death receptors (DRs) TRAIL receptor 1/DR4 and TRAIL receptor 2/DR5, and successive treatment with TRAIL resulted in apoptosis of both TRAIL-sensitive and -resistant cells. Vinblastine 53-64 TNF receptor superfamily member 10b Homo sapiens 179-182 14500373-4 2003 The chemotherapeutic drugs (paclitaxel, vincristine, vinblastine, etoposide, camptothecin, and Adriamycin) induced death receptors (DRs) TRAIL receptor 1/DR4 and TRAIL receptor 2/DR5, and successive treatment with TRAIL resulted in apoptosis of both TRAIL-sensitive and -resistant cells. Etoposide 66-75 TNF receptor superfamily member 10b Homo sapiens 179-182 14500373-4 2003 The chemotherapeutic drugs (paclitaxel, vincristine, vinblastine, etoposide, camptothecin, and Adriamycin) induced death receptors (DRs) TRAIL receptor 1/DR4 and TRAIL receptor 2/DR5, and successive treatment with TRAIL resulted in apoptosis of both TRAIL-sensitive and -resistant cells. Camptothecin 77-89 TNF receptor superfamily member 10b Homo sapiens 179-182 14500373-4 2003 The chemotherapeutic drugs (paclitaxel, vincristine, vinblastine, etoposide, camptothecin, and Adriamycin) induced death receptors (DRs) TRAIL receptor 1/DR4 and TRAIL receptor 2/DR5, and successive treatment with TRAIL resulted in apoptosis of both TRAIL-sensitive and -resistant cells. Doxorubicin 95-105 TNF receptor superfamily member 10b Homo sapiens 179-182 14500373-6 2003 TRAIL induces apoptosis in Adriamycin-resistant MCF7 cells already expressing high levels of death receptors DR4 and DR5. Doxorubicin 27-37 TNF receptor superfamily member 10b Homo sapiens 117-120 12838325-7 2003 Importantly, treatment of resistant cells with the chemotherapeutic agents doxorubicin, cisplatin and etoposide reversed the resistance to Apo2L/TRAIL, which was associated with drug-induced upregulation of mRNA encoding the death receptors DR4 and DR5. Doxorubicin 75-86 TNF receptor superfamily member 10b Homo sapiens 249-252 12902978-2 2003 PS-341 treatment by itself does not affect the levels of Bax, Bak, caspases 3 and 8, c-Flip or FADD, but elevates levels of TRAIL receptors DR4 and DR5. Bortezomib 0-6 TNF receptor superfamily member 10b Homo sapiens 148-151 12838325-7 2003 Importantly, treatment of resistant cells with the chemotherapeutic agents doxorubicin, cisplatin and etoposide reversed the resistance to Apo2L/TRAIL, which was associated with drug-induced upregulation of mRNA encoding the death receptors DR4 and DR5. Cisplatin 88-97 TNF receptor superfamily member 10b Homo sapiens 249-252 12838325-7 2003 Importantly, treatment of resistant cells with the chemotherapeutic agents doxorubicin, cisplatin and etoposide reversed the resistance to Apo2L/TRAIL, which was associated with drug-induced upregulation of mRNA encoding the death receptors DR4 and DR5. Etoposide 102-111 TNF receptor superfamily member 10b Homo sapiens 249-252 12835727-4 2003 DR4 expression was increased when Jurkat cells were treated with TRAIL, and DR5 expression increased after exposing Molt-4 cells to TRAIL plus a low dose of doxorubicin for 24 h. The expression of DR4 and DR5 in UCB mononuclear cells was unchanged after treatment with TRAIL, a low-dose doxorubicin, or TRAIL plus a low dose of doxorubicin. Doxorubicin 157-168 TNF receptor superfamily member 10b Homo sapiens 76-79 12835727-4 2003 DR4 expression was increased when Jurkat cells were treated with TRAIL, and DR5 expression increased after exposing Molt-4 cells to TRAIL plus a low dose of doxorubicin for 24 h. The expression of DR4 and DR5 in UCB mononuclear cells was unchanged after treatment with TRAIL, a low-dose doxorubicin, or TRAIL plus a low dose of doxorubicin. Doxorubicin 287-298 TNF receptor superfamily member 10b Homo sapiens 205-208 12835727-4 2003 DR4 expression was increased when Jurkat cells were treated with TRAIL, and DR5 expression increased after exposing Molt-4 cells to TRAIL plus a low dose of doxorubicin for 24 h. The expression of DR4 and DR5 in UCB mononuclear cells was unchanged after treatment with TRAIL, a low-dose doxorubicin, or TRAIL plus a low dose of doxorubicin. Doxorubicin 157-168 TNF receptor superfamily member 10b Homo sapiens 205-208 12835727-4 2003 DR4 expression was increased when Jurkat cells were treated with TRAIL, and DR5 expression increased after exposing Molt-4 cells to TRAIL plus a low dose of doxorubicin for 24 h. The expression of DR4 and DR5 in UCB mononuclear cells was unchanged after treatment with TRAIL, a low-dose doxorubicin, or TRAIL plus a low dose of doxorubicin. Doxorubicin 287-298 TNF receptor superfamily member 10b Homo sapiens 205-208 12684208-3 2003 Bile acid modulation of death-receptor signaling is multifactorial and includes trafficking of Fas to the cell surface, enhancing TRAIL-R2/DR5 expression, and suppression of function of cFLIP, an antiapoptotic protein modulating death-receptor function. Bile Acids and Salts 0-9 TNF receptor superfamily member 10b Homo sapiens 130-138 12763223-6 2003 Coincubation of TRAIL and doxorubicin led to the overexpression of TRAIL-R2 resulting in a synergistic effect of doxorubicin and TRAIL in TRAIL-sensitive cell lines and in the overcoming of TRAIL-resistance in all of the TRAIL-resistant cell lines, except HTB-91, which lacked caspase 8 expression. Doxorubicin 26-37 TNF receptor superfamily member 10b Homo sapiens 67-75 12763223-6 2003 Coincubation of TRAIL and doxorubicin led to the overexpression of TRAIL-R2 resulting in a synergistic effect of doxorubicin and TRAIL in TRAIL-sensitive cell lines and in the overcoming of TRAIL-resistance in all of the TRAIL-resistant cell lines, except HTB-91, which lacked caspase 8 expression. Doxorubicin 113-124 TNF receptor superfamily member 10b Homo sapiens 67-75 12684208-3 2003 Bile acid modulation of death-receptor signaling is multifactorial and includes trafficking of Fas to the cell surface, enhancing TRAIL-R2/DR5 expression, and suppression of function of cFLIP, an antiapoptotic protein modulating death-receptor function. Bile Acids and Salts 0-9 TNF receptor superfamily member 10b Homo sapiens 139-142 12584736-0 2003 Enhancement of TRAIL/Apo2L-mediated apoptosis by adriamycin through inducing DR4 and DR5 in renal cell carcinoma cells. Doxorubicin 49-59 TNF receptor superfamily member 10b Homo sapiens 85-88 12730681-0 2003 Effect of Bax deficiency on death receptor 5 and mitochondrial pathways during endoplasmic reticulum calcium pool depletion-induced apoptosis. Calcium 101-108 TNF receptor superfamily member 10b Homo sapiens 28-44 14716031-0 2003 Death receptor 5 and Bcl-2 protein expression as predictors of tumor response to gemcitabine and cisplatin in patients with advanced non-small-cell lung cancer. gemcitabine 81-92 TNF receptor superfamily member 10b Homo sapiens 0-16 12610517-6 2003 We show that genotoxic agents such as cisplatin, doxorubicin and camptothecin, in addition to UV radiation, can induce TRAIL-R2 on the cell surface of TRAIL receptor-negative tumour cells. Cisplatin 38-47 TNF receptor superfamily member 10b Homo sapiens 119-127 12610517-6 2003 We show that genotoxic agents such as cisplatin, doxorubicin and camptothecin, in addition to UV radiation, can induce TRAIL-R2 on the cell surface of TRAIL receptor-negative tumour cells. Doxorubicin 49-60 TNF receptor superfamily member 10b Homo sapiens 119-127 12610517-6 2003 We show that genotoxic agents such as cisplatin, doxorubicin and camptothecin, in addition to UV radiation, can induce TRAIL-R2 on the cell surface of TRAIL receptor-negative tumour cells. Camptothecin 65-77 TNF receptor superfamily member 10b Homo sapiens 119-127 12670894-6 2003 Caspase-8 processing is dependent on de novo protein synthesis; DR5 expression is strongly up-regulated by THG treatment. Thapsigargin 107-110 TNF receptor superfamily member 10b Homo sapiens 64-67 12569362-7 2003 A DR5 retinoic acid responsive element was observed in the LTR. Tretinoin 6-19 TNF receptor superfamily member 10b Homo sapiens 2-5 12543804-0 2003 2-methoxyestradiol up-regulates death receptor 5 and induces apoptosis through activation of the extrinsic pathway. 2-Methoxyestradiol 0-18 TNF receptor superfamily member 10b Homo sapiens 32-48 14716031-0 2003 Death receptor 5 and Bcl-2 protein expression as predictors of tumor response to gemcitabine and cisplatin in patients with advanced non-small-cell lung cancer. Cisplatin 97-106 TNF receptor superfamily member 10b Homo sapiens 0-16 12642685-3 2002 This resistance was determined by failure to form the TRAIL-DISC and by decreased TRAIL-R1 and TRAIL-R2 levels after androgen deprivation; the capacity of TRAIL to induce DISC formation was completely restored in the presence of DHT. Dihydrotestosterone 229-232 TNF receptor superfamily member 10b Homo sapiens 95-103 12390973-6 2002 A blocking experiment with monoclonal antibodies directed against TRAIL-R1 and TRAIL-R2 revealed that TRAIL-R1 is mainly involved in TRAIL-induced apoptosis of ahOL. ahol 160-164 TNF receptor superfamily member 10b Homo sapiens 79-87 12384541-9 2002 Tumors treated with CPT-11 showed increased membrane expression of DR5, suggesting that CPT-11 may increase sensitivity to TRAIL by up-regulation of DR5. Irinotecan 20-26 TNF receptor superfamily member 10b Homo sapiens 67-70 12388624-2 2002 We have recently demonstrated, however, that bile acids increase TRAIL-R2/DR5 expression in a human liver cell line and render these cells susceptible to TRAIL-mediated apoptosis. Bile Acids and Salts 45-55 TNF receptor superfamily member 10b Homo sapiens 65-73 12388624-2 2002 We have recently demonstrated, however, that bile acids increase TRAIL-R2/DR5 expression in a human liver cell line and render these cells susceptible to TRAIL-mediated apoptosis. Bile Acids and Salts 45-55 TNF receptor superfamily member 10b Homo sapiens 74-77 12384541-9 2002 Tumors treated with CPT-11 showed increased membrane expression of DR5, suggesting that CPT-11 may increase sensitivity to TRAIL by up-regulation of DR5. Irinotecan 20-26 TNF receptor superfamily member 10b Homo sapiens 149-152 12384541-9 2002 Tumors treated with CPT-11 showed increased membrane expression of DR5, suggesting that CPT-11 may increase sensitivity to TRAIL by up-regulation of DR5. Irinotecan 88-94 TNF receptor superfamily member 10b Homo sapiens 67-70 12384541-9 2002 Tumors treated with CPT-11 showed increased membrane expression of DR5, suggesting that CPT-11 may increase sensitivity to TRAIL by up-regulation of DR5. Irinotecan 88-94 TNF receptor superfamily member 10b Homo sapiens 149-152 12203115-2 2002 Here we report that two different NSAIDs, sulindac sulfide and SC-"236 engage the death receptor 5 (DR5) and mitochondrial pathways to mediate apoptosis in human colon cancer cells. sulindac sulfide 42-58 TNF receptor superfamily member 10b Homo sapiens 82-98 12207174-7 2002 Blockage of NFkappaB activation either by expression of dominant negative IkappaB or treatment with proteasome inhibitor lactacystin eliminates TRAIL induced DR5 expression. lactacystin 121-132 TNF receptor superfamily member 10b Homo sapiens 158-161 12203115-2 2002 Here we report that two different NSAIDs, sulindac sulfide and SC-"236 engage the death receptor 5 (DR5) and mitochondrial pathways to mediate apoptosis in human colon cancer cells. sulindac sulfide 42-58 TNF receptor superfamily member 10b Homo sapiens 100-103 12203115-2 2002 Here we report that two different NSAIDs, sulindac sulfide and SC-"236 engage the death receptor 5 (DR5) and mitochondrial pathways to mediate apoptosis in human colon cancer cells. 4-(5-(4-chlorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)benzenesulfonamide 63-70 TNF receptor superfamily member 10b Homo sapiens 82-98 12203115-2 2002 Here we report that two different NSAIDs, sulindac sulfide and SC-"236 engage the death receptor 5 (DR5) and mitochondrial pathways to mediate apoptosis in human colon cancer cells. 4-(5-(4-chlorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)benzenesulfonamide 63-70 TNF receptor superfamily member 10b Homo sapiens 100-103 12203115-3 2002 We show that sulindac sulfide and SC-"236-induced apoptosis is coupled with upregulation of DR5, caspase 8 activation and Bid cleavage. sulindac sulfide 13-29 TNF receptor superfamily member 10b Homo sapiens 92-95 12203115-3 2002 We show that sulindac sulfide and SC-"236-induced apoptosis is coupled with upregulation of DR5, caspase 8 activation and Bid cleavage. 4-(5-(4-chlorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)benzenesulfonamide 34-41 TNF receptor superfamily member 10b Homo sapiens 92-95 12203115-5 2002 We further show that sulindac sulfide and SC-"236-induced DR5 upregulation occurs independent of the COX inhibitory effects of these NSAIDs. sulindac sulfide 21-37 TNF receptor superfamily member 10b Homo sapiens 58-61 12203115-5 2002 We further show that sulindac sulfide and SC-"236-induced DR5 upregulation occurs independent of the COX inhibitory effects of these NSAIDs. 4-(5-(4-chlorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)benzenesulfonamide 42-49 TNF receptor superfamily member 10b Homo sapiens 58-61 11916527-1 2002 The relationship between p53 gene status and the expression of DR5 and Fas was evaluated as a function of sensitivity of 11 acute lymphoblastic leukemia cell lines to adriamycin, etoposide, vincristine, methotrexate and dexamethasone. Doxorubicin 167-177 TNF receptor superfamily member 10b Homo sapiens 63-66 12203115-7 2002 For example, sulindac sulfide upregulates DR5 in both Bax-deficient and proficient cells, but Apo2L/TRAIL efficiently potentiates sulindac sulfide-induced apoptosis as well as activation of caspase-8, -9 and -3 only in Bax-proficient cells. sulindac sulfide 13-29 TNF receptor superfamily member 10b Homo sapiens 42-45 12208739-6 2002 Sulindac sulfide treatment restored DR-5 expression and, when combined with TRAIL, reduced cell viability to a greater extent than did either drug alone. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 36-40 12034736-0 2002 Bisindolylmaleimide VIII enhances DR5-mediated apoptosis through the MKK4/JNK/p38 kinase and the mitochondrial pathways. bisindolylmaleimide 0-19 TNF receptor superfamily member 10b Homo sapiens 34-37 12432279-6 2002 Selenium upregulation of DR5 was coupled with caspase 8 activation and Bid cleavage thereby suggesting the existence of a potential cross-talk between the DR5 and the mitochondrial pathways. Selenium 0-8 TNF receptor superfamily member 10b Homo sapiens 155-158 12432279-7 2002 Thus, our results suggest that DR5 is specifically regulated by selenium and its activation may play an important role in selenium-mediated chemoprevention. Selenium 64-72 TNF receptor superfamily member 10b Homo sapiens 31-34 12432279-7 2002 Thus, our results suggest that DR5 is specifically regulated by selenium and its activation may play an important role in selenium-mediated chemoprevention. Selenium 122-130 TNF receptor superfamily member 10b Homo sapiens 31-34 12496481-6 2002 TRAIL receptor mRNAs (DR4, DR5, and DcR2) were induced by doxorubicin but these changes were not reflected at the protein level. Doxorubicin 58-69 TNF receptor superfamily member 10b Homo sapiens 27-30 12432279-0 2002 Death receptor 5 regulation during selenium-mediated apoptosis in human prostate cancer cells. Selenium 35-43 TNF receptor superfamily member 10b Homo sapiens 0-16 12432279-4 2002 Here we report that selenium-mediated apoptosis appears to involve membrane death receptor, DR5-dependent pathway in human prostate cancer cells. Selenium 20-28 TNF receptor superfamily member 10b Homo sapiens 92-95 12432279-5 2002 Selenium specifically upregulated DR5 expression but not that of DR4. Selenium 0-8 TNF receptor superfamily member 10b Homo sapiens 34-37 12432279-6 2002 Selenium upregulation of DR5 was coupled with caspase 8 activation and Bid cleavage thereby suggesting the existence of a potential cross-talk between the DR5 and the mitochondrial pathways. Selenium 0-8 TNF receptor superfamily member 10b Homo sapiens 25-28 11916527-1 2002 The relationship between p53 gene status and the expression of DR5 and Fas was evaluated as a function of sensitivity of 11 acute lymphoblastic leukemia cell lines to adriamycin, etoposide, vincristine, methotrexate and dexamethasone. Etoposide 179-188 TNF receptor superfamily member 10b Homo sapiens 63-66 11916527-1 2002 The relationship between p53 gene status and the expression of DR5 and Fas was evaluated as a function of sensitivity of 11 acute lymphoblastic leukemia cell lines to adriamycin, etoposide, vincristine, methotrexate and dexamethasone. Vincristine 190-201 TNF receptor superfamily member 10b Homo sapiens 63-66 11916527-1 2002 The relationship between p53 gene status and the expression of DR5 and Fas was evaluated as a function of sensitivity of 11 acute lymphoblastic leukemia cell lines to adriamycin, etoposide, vincristine, methotrexate and dexamethasone. Methotrexate 203-215 TNF receptor superfamily member 10b Homo sapiens 63-66 11916527-1 2002 The relationship between p53 gene status and the expression of DR5 and Fas was evaluated as a function of sensitivity of 11 acute lymphoblastic leukemia cell lines to adriamycin, etoposide, vincristine, methotrexate and dexamethasone. Dexamethasone 220-233 TNF receptor superfamily member 10b Homo sapiens 63-66 11802207-0 2002 TRAIL, FasL and a blocking anti-DR5 antibody augment paclitaxel-induced apoptosis in human non-small-cell lung cancer. Paclitaxel 53-63 TNF receptor superfamily member 10b Homo sapiens 32-35 11965535-0 2002 Endoplasmic reticulum calcium pool depletion-induced apoptosis is coupled with activation of the death receptor 5 pathway. Calcium 22-29 TNF receptor superfamily member 10b Homo sapiens 97-113 11965535-3 2002 In this study, we demonstrate for the first time that TG-mediated perturbations in Ca(2+) homeostasis are coupled with activation of the death receptor 5 (DR5)-dependent apoptotic pathway in human cancer cells. Thapsigargin 54-56 TNF receptor superfamily member 10b Homo sapiens 137-153 11965535-3 2002 In this study, we demonstrate for the first time that TG-mediated perturbations in Ca(2+) homeostasis are coupled with activation of the death receptor 5 (DR5)-dependent apoptotic pathway in human cancer cells. Thapsigargin 54-56 TNF receptor superfamily member 10b Homo sapiens 155-158 11965535-9 2002 The TG-induced increase in DR5 expression appeared to occur as a consequence of TG-induced endoplasmic reticulum (ER) Ca(2+) pool depletion. Thapsigargin 4-6 TNF receptor superfamily member 10b Homo sapiens 27-30 11802207-10 2002 The combination treatments, FasL+PA, TRAIL+PA or PA+anti-DR5 antibody, greatly enhance PA-apoptotic effect in most cell lines. Paclitaxel 49-52 TNF receptor superfamily member 10b Homo sapiens 57-60 12794242-8 2002 PMA also increased the expression of TRAIL receptors DR4 and DR5; this expression was inhibited by the PKC inhibitors GF109203x, Go6983, and Ro-31-8220 and the MEK inhibitor UO126, suggesting a role for conventional PKC isoforms and MEK in the regulation of TRAIL receptor expression. bisindolylmaleimide I 118-127 TNF receptor superfamily member 10b Homo sapiens 61-64 11803469-2 2002 We demonstrate that in H157 human lung carcinoma cells, etoposide and doxorubicin induce the NF-kappaB-dependent expression of both pro- and anti-apoptotic proteins including TRAIL and its death receptor, DR5, and IAPs. Etoposide 56-65 TNF receptor superfamily member 10b Homo sapiens 205-208 11803469-2 2002 We demonstrate that in H157 human lung carcinoma cells, etoposide and doxorubicin induce the NF-kappaB-dependent expression of both pro- and anti-apoptotic proteins including TRAIL and its death receptor, DR5, and IAPs. Doxorubicin 70-81 TNF receptor superfamily member 10b Homo sapiens 205-208 12174820-7 2002 p53 target genes such as p21WAF1/Cip1, and KILLER/DR5 were upregulated by CP-31398, but their expression did not correlate with arrest or apoptosis induction. CP 31398 74-82 TNF receptor superfamily member 10b Homo sapiens 43-53 12174820-8 2002 Combination of CP-31398 and TRAIL or chemotherapeutic agents enhanced cancer cell killing effect possibly through upregulation of p53-regulated genes such as KILLER/DR5. CP 31398 15-23 TNF receptor superfamily member 10b Homo sapiens 158-168 12794242-8 2002 PMA also increased the expression of TRAIL receptors DR4 and DR5; this expression was inhibited by the PKC inhibitors GF109203x, Go6983, and Ro-31-8220 and the MEK inhibitor UO126, suggesting a role for conventional PKC isoforms and MEK in the regulation of TRAIL receptor expression. 2-(1-(3-dimethylaminopropyl)-5-methoxyindol-3-yl)-3-(1H-indol-3-yl)maleimide 129-135 TNF receptor superfamily member 10b Homo sapiens 61-64 12794242-8 2002 PMA also increased the expression of TRAIL receptors DR4 and DR5; this expression was inhibited by the PKC inhibitors GF109203x, Go6983, and Ro-31-8220 and the MEK inhibitor UO126, suggesting a role for conventional PKC isoforms and MEK in the regulation of TRAIL receptor expression. U 0126 174-179 TNF receptor superfamily member 10b Homo sapiens 61-64 11498783-7 2001 Although the basal mRNA levels of these genes did not depend on the presence of p53, we observed a p53-dependent induction of all these targets in response to gamma-irradiation and a p53-independent regulation for p21 and KILLER/DR5 in response to dexamethasone. Dexamethasone 248-261 TNF receptor superfamily member 10b Homo sapiens 222-232 11677215-1 2001 BACKGROUND & AIMS: The KILLER/death receptor (DR)5 has been identified as a potent inducer of apoptosis, and mapped to chromosome 8p21-22, showing frequent allelic loss in gastric cancer. Adenosine Monophosphate 12-15 TNF receptor superfamily member 10b Homo sapiens 27-54 11507096-0 2001 The bile acid glycochenodeoxycholate induces trail-receptor 2/DR5 expression and apoptosis. Bile Acids and Salts 4-13 TNF receptor superfamily member 10b Homo sapiens 62-65 11507096-0 2001 The bile acid glycochenodeoxycholate induces trail-receptor 2/DR5 expression and apoptosis. Glycochenodeoxycholic Acid 14-36 TNF receptor superfamily member 10b Homo sapiens 62-65 11507096-8 2001 GCDC treatment increased expression of TRAIL-R2/DR5 mRNA and protein 10-fold while expression of TRAIL-R1 was unchanged. Glycochenodeoxycholic Acid 0-4 TNF receptor superfamily member 10b Homo sapiens 39-47 11507096-8 2001 GCDC treatment increased expression of TRAIL-R2/DR5 mRNA and protein 10-fold while expression of TRAIL-R1 was unchanged. Glycochenodeoxycholic Acid 0-4 TNF receptor superfamily member 10b Homo sapiens 48-51 11507096-9 2001 Furthermore, aggregation of TRAIL-R2/DR5, but not TRAIL-R1/DR4 was observed following GCDC treatment of the cells. Glycochenodeoxycholic Acid 86-90 TNF receptor superfamily member 10b Homo sapiens 28-36 11507096-9 2001 Furthermore, aggregation of TRAIL-R2/DR5, but not TRAIL-R1/DR4 was observed following GCDC treatment of the cells. Glycochenodeoxycholic Acid 86-90 TNF receptor superfamily member 10b Homo sapiens 37-40 11507096-10 2001 Induction of TRAIL-R2/DR5 expression and apoptosis by bile acids provides new insights into the mechanisms of hepatocyte apoptosis and the regulation of TRAIL-R2/DR5 expression. Bile Acids and Salts 54-64 TNF receptor superfamily member 10b Homo sapiens 13-21 11507096-10 2001 Induction of TRAIL-R2/DR5 expression and apoptosis by bile acids provides new insights into the mechanisms of hepatocyte apoptosis and the regulation of TRAIL-R2/DR5 expression. Bile Acids and Salts 54-64 TNF receptor superfamily member 10b Homo sapiens 22-25 11507096-10 2001 Induction of TRAIL-R2/DR5 expression and apoptosis by bile acids provides new insights into the mechanisms of hepatocyte apoptosis and the regulation of TRAIL-R2/DR5 expression. Bile Acids and Salts 54-64 TNF receptor superfamily member 10b Homo sapiens 153-161 11507096-10 2001 Induction of TRAIL-R2/DR5 expression and apoptosis by bile acids provides new insights into the mechanisms of hepatocyte apoptosis and the regulation of TRAIL-R2/DR5 expression. Bile Acids and Salts 54-64 TNF receptor superfamily member 10b Homo sapiens 162-165 11559570-0 2001 Sulindac sulfide-induced apoptosis involves death receptor 5 and the caspase 8-dependent pathway in human colon and prostate cancer cells. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 44-60 11559570-5 2001 Sulindac sulfide up-regulated DR5 and activated the proximal caspase 8 in various different colon and prostate cancer cell lines. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 30-33 11559570-6 2001 Sulindac sulfide specifically up-regulated the DR5 levels but had no effect on the levels of other DRs including DR4, Fas, and tumor necrosis factor receptor 1. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 47-50 11559570-9 2001 Our results indicated that by blocking the DR5-dependent apoptotic pathway, dominant-negative Fas-associated death domain did indeed inhibit sulindac sulfide-induced apoptosis. sulindac sulfide 141-157 TNF receptor superfamily member 10b Homo sapiens 43-46 11559570-10 2001 Furthermore, exogenous tumor necrosis factor-related apoptosis-inducing ligand, the ligand for DR5, also potentiated sulindac sulfide-induced apoptosis in all of the cell lines tested, thereby further supporting the involvement of DR5 in sulindac sulfide-induced apoptosis. sulindac sulfide 117-133 TNF receptor superfamily member 10b Homo sapiens 95-98 11559570-10 2001 Furthermore, exogenous tumor necrosis factor-related apoptosis-inducing ligand, the ligand for DR5, also potentiated sulindac sulfide-induced apoptosis in all of the cell lines tested, thereby further supporting the involvement of DR5 in sulindac sulfide-induced apoptosis. sulindac sulfide 117-133 TNF receptor superfamily member 10b Homo sapiens 231-234 11559570-10 2001 Furthermore, exogenous tumor necrosis factor-related apoptosis-inducing ligand, the ligand for DR5, also potentiated sulindac sulfide-induced apoptosis in all of the cell lines tested, thereby further supporting the involvement of DR5 in sulindac sulfide-induced apoptosis. sulindac sulfide 238-254 TNF receptor superfamily member 10b Homo sapiens 95-98 11559570-11 2001 Thus, our results demonstrate that sulindac sulfide also engages the membrane DR pathway involving DR5 and proximal caspase 8 to induce apoptosis. sulindac sulfide 35-51 TNF receptor superfamily member 10b Homo sapiens 99-102 11751478-7 2001 Adriamycin treatment modestly up-regulated Apo2L/TRAIL-R2 (DR5) and had no effect on the expression of Fas-associated death domain, c-FLIP, Bcl-2, Bcl(xL), Bax, and IAP family members (cIAP-1, cIAP-2, XIAP, and survivin). Doxorubicin 0-10 TNF receptor superfamily member 10b Homo sapiens 49-57 11751478-7 2001 Adriamycin treatment modestly up-regulated Apo2L/TRAIL-R2 (DR5) and had no effect on the expression of Fas-associated death domain, c-FLIP, Bcl-2, Bcl(xL), Bax, and IAP family members (cIAP-1, cIAP-2, XIAP, and survivin). Doxorubicin 0-10 TNF receptor superfamily member 10b Homo sapiens 59-62 11468181-7 2001 Dox up-regulated the expression of the TRAIL receptor death receptor 5 (DR5) and synergistically enhanced the effect of TRAIL not only against MM cells sensitive to, but also against those resistant to, Dex- or Dox-induced apoptosis. Doxorubicin 0-3 TNF receptor superfamily member 10b Homo sapiens 54-70 11468181-7 2001 Dox up-regulated the expression of the TRAIL receptor death receptor 5 (DR5) and synergistically enhanced the effect of TRAIL not only against MM cells sensitive to, but also against those resistant to, Dex- or Dox-induced apoptosis. Doxorubicin 0-3 TNF receptor superfamily member 10b Homo sapiens 72-75 11468181-7 2001 Dox up-regulated the expression of the TRAIL receptor death receptor 5 (DR5) and synergistically enhanced the effect of TRAIL not only against MM cells sensitive to, but also against those resistant to, Dex- or Dox-induced apoptosis. Dexamethasone 203-206 TNF receptor superfamily member 10b Homo sapiens 54-70 11468181-7 2001 Dox up-regulated the expression of the TRAIL receptor death receptor 5 (DR5) and synergistically enhanced the effect of TRAIL not only against MM cells sensitive to, but also against those resistant to, Dex- or Dox-induced apoptosis. Doxorubicin 211-214 TNF receptor superfamily member 10b Homo sapiens 54-70 11468181-7 2001 Dox up-regulated the expression of the TRAIL receptor death receptor 5 (DR5) and synergistically enhanced the effect of TRAIL not only against MM cells sensitive to, but also against those resistant to, Dex- or Dox-induced apoptosis. Doxorubicin 211-214 TNF receptor superfamily member 10b Homo sapiens 72-75 11212279-5 2001 Treatment with paclitaxel or taxotere increased DR4 and/or DR5 protein levels (up to 8-fold) without affecting the protein levels of DcR1 and DcR2, Apo-2L/TRAIL, Fas, or Fas ligand. Paclitaxel 15-25 TNF receptor superfamily member 10b Homo sapiens 59-62 11234890-9 2001 Cotreatment with STI-571 significantly enhanced Apo-2L/TRAIL-induced apoptosis (P < 0.01) as well as increased the processing of caspase-9 and -3 and XIAP, without affecting the levels of DR4, DR5, decoy receptors, or c-FLIP(L). Imatinib Mesylate 17-24 TNF receptor superfamily member 10b Homo sapiens 196-199 11139340-4 2001 beta-Lapachone, a topoisomerase inhibitor, increased KILLER/DR5 mRNA in colon cancer cell lines with wild-type p53 but not with mutant p53. beta-lapachone 0-14 TNF receptor superfamily member 10b Homo sapiens 53-63 11139340-5 2001 In contrast, betulinic acid, a novel chemotherapeutic compound, induced apoptosis and KILLER/DR5 mRNA in melanoma and glioblastoma cells through a p53-independent mechanism. betulinic acid 13-27 TNF receptor superfamily member 10b Homo sapiens 86-96 11139340-6 2001 The synthetic glucocorticoid dexamethasone elevated KILLER/DR5 mRNA in glioblastoma, ovarian cancer, and colon cancer cell lines with mutant p53 undergoing apoptosis, and this induction was inhibited by the transcriptional inhibitor actinomycin D. Dexamethasone 29-42 TNF receptor superfamily member 10b Homo sapiens 52-62 11139340-6 2001 The synthetic glucocorticoid dexamethasone elevated KILLER/DR5 mRNA in glioblastoma, ovarian cancer, and colon cancer cell lines with mutant p53 undergoing apoptosis, and this induction was inhibited by the transcriptional inhibitor actinomycin D. Dactinomycin 233-246 TNF receptor superfamily member 10b Homo sapiens 52-62 11212279-5 2001 Treatment with paclitaxel or taxotere increased DR4 and/or DR5 protein levels (up to 8-fold) without affecting the protein levels of DcR1 and DcR2, Apo-2L/TRAIL, Fas, or Fas ligand. Docetaxel 29-37 TNF receptor superfamily member 10b Homo sapiens 59-62 11139287-6 2000 Topotecan, a novel topoisomerase I inhibitor, induced upregulation of TRAIL-R2 as well as downregulation of TRAIL. Topotecan 0-9 TNF receptor superfamily member 10b Homo sapiens 70-78 11090076-6 2000 Treatment of human leukemic cells with etoposide, Ara-C, or doxorubicin increased DR5 but not DR4, Fas, DcR1, DcR2, Fas ligand, or Apo-2L levels. Doxorubicin 60-71 TNF receptor superfamily member 10b Homo sapiens 82-85 11090076-8 2000 These findings indicate that treatment with etoposide, Ara-C, or doxorubicin up-regulates DR5 levels in a p53-independent manner and sensitizes human acute leukemia cells to Apo-2L-induced apoptosis. Etoposide 44-53 TNF receptor superfamily member 10b Homo sapiens 90-93 11090076-8 2000 These findings indicate that treatment with etoposide, Ara-C, or doxorubicin up-regulates DR5 levels in a p53-independent manner and sensitizes human acute leukemia cells to Apo-2L-induced apoptosis. Cytarabine 55-60 TNF receptor superfamily member 10b Homo sapiens 90-93 11090076-8 2000 These findings indicate that treatment with etoposide, Ara-C, or doxorubicin up-regulates DR5 levels in a p53-independent manner and sensitizes human acute leukemia cells to Apo-2L-induced apoptosis. Doxorubicin 65-76 TNF receptor superfamily member 10b Homo sapiens 90-93 11212279-6 2001 Up-regulation of DR4 and DR5 was not preceded by the induction of their mRNA levels but was inhibited by cotreatment with cycloheximide. Cycloheximide 122-135 TNF receptor superfamily member 10b Homo sapiens 25-28 11212279-9 2001 These findings indicate that up-regulation of DR4 and DR5 protein levels by treatment with paclitaxel enhances subsequent Apo-2L/TRAIL-induced apoptosis of human prostate cancer cells. Paclitaxel 91-101 TNF receptor superfamily member 10b Homo sapiens 54-57 11090076-6 2000 Treatment of human leukemic cells with etoposide, Ara-C, or doxorubicin increased DR5 but not DR4, Fas, DcR1, DcR2, Fas ligand, or Apo-2L levels. Etoposide 39-48 TNF receptor superfamily member 10b Homo sapiens 82-85 11090076-6 2000 Treatment of human leukemic cells with etoposide, Ara-C, or doxorubicin increased DR5 but not DR4, Fas, DcR1, DcR2, Fas ligand, or Apo-2L levels. Cytarabine 50-55 TNF receptor superfamily member 10b Homo sapiens 82-85 10893485-1 2000 The cholecystokinin A receptor (CCK-AR) modulates CCK-stimulated dopamine release in the posterior nucleus accumbens, and its gene is mapped to 4p15.2-15.1 with the dopamine receptor 5 (DR5) gene. Dopamine 65-73 TNF receptor superfamily member 10b Homo sapiens 165-184 10930093-0 2000 Coordinated regulation of two TRAIL-R2/KILLER/DR5 mRNA isoforms by DNA damaging agents, serum and 17beta-estradiol in human breast cancer cells. Estradiol 98-114 TNF receptor superfamily member 10b Homo sapiens 30-38 10930093-0 2000 Coordinated regulation of two TRAIL-R2/KILLER/DR5 mRNA isoforms by DNA damaging agents, serum and 17beta-estradiol in human breast cancer cells. Estradiol 98-114 TNF receptor superfamily member 10b Homo sapiens 39-49 10930093-6 2000 Treatment of the p53-mutant T-47D breast cancer cell line with adriamycin also results in up-regulation of both isoforms, suggesting that adriamycin up-regulates TRAIL-R2/Killer/DR5 expression independent of functional p53. Doxorubicin 63-73 TNF receptor superfamily member 10b Homo sapiens 162-170 10930093-6 2000 Treatment of the p53-mutant T-47D breast cancer cell line with adriamycin also results in up-regulation of both isoforms, suggesting that adriamycin up-regulates TRAIL-R2/Killer/DR5 expression independent of functional p53. Doxorubicin 63-73 TNF receptor superfamily member 10b Homo sapiens 171-181 10930093-6 2000 Treatment of the p53-mutant T-47D breast cancer cell line with adriamycin also results in up-regulation of both isoforms, suggesting that adriamycin up-regulates TRAIL-R2/Killer/DR5 expression independent of functional p53. Doxorubicin 138-148 TNF receptor superfamily member 10b Homo sapiens 162-170 10930093-6 2000 Treatment of the p53-mutant T-47D breast cancer cell line with adriamycin also results in up-regulation of both isoforms, suggesting that adriamycin up-regulates TRAIL-R2/Killer/DR5 expression independent of functional p53. Doxorubicin 138-148 TNF receptor superfamily member 10b Homo sapiens 171-181 11002424-9 2000 Because CD437 induced the expression of DR4, DR5 and Fas, we examined the effects of combining CD437 and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and Fas ligand, respectively, in HPC cells. CD 437 8-13 TNF receptor superfamily member 10b Homo sapiens 45-48 10893485-1 2000 The cholecystokinin A receptor (CCK-AR) modulates CCK-stimulated dopamine release in the posterior nucleus accumbens, and its gene is mapped to 4p15.2-15.1 with the dopamine receptor 5 (DR5) gene. Dopamine 65-73 TNF receptor superfamily member 10b Homo sapiens 186-189 10693703-12 2000 Expression of TRAIL (a ligand for DR4 and DR5) was significantly lower in CCSK and RTK than in normal kidney (9.5+/-1.5% v. 56.1+/-10.1%; P = .01). ccsk 74-78 TNF receptor superfamily member 10b Homo sapiens 42-45 10777207-9 2000 In p53-wild-type but not -mutant or -null cell lines, doxorubicin treatment stabilized p53 protein, and increased specific binding to BS2 as revealed by EMSA, and upregulated the KILLER/DR5 promoter-luciferase reporter gene. Doxorubicin 54-65 TNF receptor superfamily member 10b Homo sapiens 179-189 10706092-3 2000 Here we show that the DNA-damaging chemotherapeutic drugs, cis-diamminedichloroplatinum(II) (CDDP) and etoposide, elicited increased expression of DR5 in human glioma cells. Cisplatin 59-91 TNF receptor superfamily member 10b Homo sapiens 147-150 10706092-3 2000 Here we show that the DNA-damaging chemotherapeutic drugs, cis-diamminedichloroplatinum(II) (CDDP) and etoposide, elicited increased expression of DR5 in human glioma cells. Cisplatin 93-97 TNF receptor superfamily member 10b Homo sapiens 147-150 10706092-3 2000 Here we show that the DNA-damaging chemotherapeutic drugs, cis-diamminedichloroplatinum(II) (CDDP) and etoposide, elicited increased expression of DR5 in human glioma cells. Etoposide 103-112 TNF receptor superfamily member 10b Homo sapiens 147-150 10706092-4 2000 Exposure of such cells in vitro to soluble human TRAIL in combination with CDDP or etoposide resulted in synergistic cell death that could be blocked by soluble TRAIL-neutralizing DR5-Fc or the caspase inhibitors, Z-Asp-CH2-DCB and CrmA. Cisplatin 75-79 TNF receptor superfamily member 10b Homo sapiens 180-183 10706092-4 2000 Exposure of such cells in vitro to soluble human TRAIL in combination with CDDP or etoposide resulted in synergistic cell death that could be blocked by soluble TRAIL-neutralizing DR5-Fc or the caspase inhibitors, Z-Asp-CH2-DCB and CrmA. Etoposide 83-92 TNF receptor superfamily member 10b Homo sapiens 180-183 10706092-4 2000 Exposure of such cells in vitro to soluble human TRAIL in combination with CDDP or etoposide resulted in synergistic cell death that could be blocked by soluble TRAIL-neutralizing DR5-Fc or the caspase inhibitors, Z-Asp-CH2-DCB and CrmA. benzyloxycarbonyl-Asp-CH2OC(O)-2,6-dichlorobenzene 214-227 TNF receptor superfamily member 10b Homo sapiens 180-183 10706092-4 2000 Exposure of such cells in vitro to soluble human TRAIL in combination with CDDP or etoposide resulted in synergistic cell death that could be blocked by soluble TRAIL-neutralizing DR5-Fc or the caspase inhibitors, Z-Asp-CH2-DCB and CrmA. crma 232-236 TNF receptor superfamily member 10b Homo sapiens 180-183 10676636-6 2000 Biochemical analysis of the signaling events induced by TRAIL revealed that PK could be sensitized for TRAIL and, similarly, for TRAIL-R1- and TRAIL-R2-specific apoptosis by pretreatment of the cells with cycloheximide (CHX). Cycloheximide 205-218 TNF receptor superfamily member 10b Homo sapiens 143-151 10676636-6 2000 Biochemical analysis of the signaling events induced by TRAIL revealed that PK could be sensitized for TRAIL and, similarly, for TRAIL-R1- and TRAIL-R2-specific apoptosis by pretreatment of the cells with cycloheximide (CHX). Cycloheximide 220-223 TNF receptor superfamily member 10b Homo sapiens 143-151 9311998-5 1997 TRAIL-R2 contains two extracellular cysteine-rich repeats, typical for TNF receptor (TNFR) family members, and a cytoplasmic death domain. Cysteine 36-44 TNF receptor superfamily member 10b Homo sapiens 0-8 10933923-2 2000 We previously identified the death domain containing proapoptotic TRAIL death receptor KILLER/DR5 (TRAIL-R2) as an upregulated transcript following exposure of cancer cells, with wild-type but not with mutant or degraded p53 proteins, to a cytotoxic dose of adriamycin. Doxorubicin 258-268 TNF receptor superfamily member 10b Homo sapiens 87-97 10933923-2 2000 We previously identified the death domain containing proapoptotic TRAIL death receptor KILLER/DR5 (TRAIL-R2) as an upregulated transcript following exposure of cancer cells, with wild-type but not with mutant or degraded p53 proteins, to a cytotoxic dose of adriamycin. Doxorubicin 258-268 TNF receptor superfamily member 10b Homo sapiens 99-107 10594023-4 2000 The mechanism of the synergistic effect results from the etoposide-mediated increase in the expression of the death receptors 4 (DR4) and 5 (DR5). Etoposide 57-66 TNF receptor superfamily member 10b Homo sapiens 141-144 10594023-5 2000 Inhibition of NF-kappaB activation by expression of kinase-inactive MEK kinase 1(MEKK1) or dominant-negative IkappaB (DeltaIkappaB) blocked the increase in DR4 and DR5 expression following etoposide treatment. Etoposide 189-198 TNF receptor superfamily member 10b Homo sapiens 164-167 9563466-1 1998 The death receptor (DR) KILLER/DR5 gene has recently been identified as a doxorubicin-regulated transcript that was also induced by exogenous wild-type p53 in p53-negative cells. Doxorubicin 74-85 TNF receptor superfamily member 10b Homo sapiens 24-34 9563466-6 1998 Just like other p53-regulated genes, ionizing radiation induction of KILLER/DR5 occurs in p53 wild-type cells, whereas methyl methanesulfonate regulation of KILLER/DR5 occurs in a p53-dependent and -independent manner. Methyl Methanesulfonate 119-142 TNF receptor superfamily member 10b Homo sapiens 157-167 10597242-6 1999 Inhibition of transcription by Actinomycin D blocks both KILLER/DR5 and p21 induction in cells undergoing p53-dependent apoptosis. Dactinomycin 31-44 TNF receptor superfamily member 10b Homo sapiens 57-67 10336890-6 1999 Despite the altered polarity, the DR5-bound heterodimer was able to recruit the nuclear receptor coactivator ACTR in a vitamin D-dependent fashion. Vitamin D 119-128 TNF receptor superfamily member 10b Homo sapiens 34-37 10327056-8 1999 These results suggest that CD437-induced apoptosis is more extensive in NSCLC cells that express wild-type p53, possibly due to the involvement of the p53 regulated genes Killer/DR5, and Bax although CD437 can also induce apoptosis by means of a p53-independent mechanism. CD 437 27-32 TNF receptor superfamily member 10b Homo sapiens 178-181 9856825-9 1998 Deletion of the DR1d or DR5 sites reduced retinoid-induced promoter activity by 63% and 27%, respectively. Retinoids 42-50 TNF receptor superfamily member 10b Homo sapiens 24-27 9486405-6 1998 All patients with pSS, with or without autoantibodies to Ro and La, were found to have at least one of the HLA-DRB1 types DR2, DR3 or DR5. pss 18-21 TNF receptor superfamily member 10b Homo sapiens 134-137 7654186-1 1995 Functional retinoic acid response elements (RAREs) have been described wherein the direct repeats are separated by 1, 2 or 5 bp (termed DR1, DR2 and DR5 respectively). Tretinoin 11-24 TNF receptor superfamily member 10b Homo sapiens 149-152 7565738-3 1995 The greatest RA-dependent transactivation, seen with DR15, was similar to that observed with the canonical DR5. Tretinoin 13-15 TNF receptor superfamily member 10b Homo sapiens 107-110 8139911-1 1994 The polymorphic nature of sequences which act as retinoic acid response elements (RAREs and RXREs) in transactivation assays in mammalian cells, suggests that elements consisting of a direct repetition of a half site motif, separated by 1 to 5 base pairs (DR1 to DR5), are targets for retinoic acid (RA) signalling. Tretinoin 82-84 TNF receptor superfamily member 10b Homo sapiens 263-266 7558913-1 1995 Susceptibility to developing CD is widely accepted to be primarily associated with a particular HLA-DQ alpha beta heterodimer encoded by the DQA1*0501 and DQB1*0201 alleles in cis position on the DR3,DQ2 haplotype or in trans position by DR5,DQ7/DR7,DQ2 heterozygotes. Cadmium 29-31 TNF receptor superfamily member 10b Homo sapiens 238-241 7558913-5 1995 Furthermore, our data also show that those carrying the genotype DR5,DQ7/DR7,DQ2 have a significantly increased risk of developing CD as compared to those that are non-DR7 positive, also carrying the CD-associated HLA-DQ alpha beta heterodimer. Cadmium 131-133 TNF receptor superfamily member 10b Homo sapiens 65-68 7558913-5 1995 Furthermore, our data also show that those carrying the genotype DR5,DQ7/DR7,DQ2 have a significantly increased risk of developing CD as compared to those that are non-DR7 positive, also carrying the CD-associated HLA-DQ alpha beta heterodimer. Cadmium 200-202 TNF receptor superfamily member 10b Homo sapiens 65-68 7706255-0 1995 Retinoic acid induction of human tissue-type plasminogen activator gene expression via a direct repeat element (DR5) located at -7 kilobases. Tretinoin 0-13 TNF receptor superfamily member 10b Homo sapiens 112-115 7706255-5 1995 A functional retinoic acid response element (RARE), consisting of a direct repeat of the GGGTCA motif spaced by 5 nucleotides (t-PA/DR5), was localized at -7.3 kilobases. Tretinoin 13-26 TNF receptor superfamily member 10b Homo sapiens 132-135 7706255-5 1995 A functional retinoic acid response element (RARE), consisting of a direct repeat of the GGGTCA motif spaced by 5 nucleotides (t-PA/DR5), was localized at -7.3 kilobases. gggtca 89-95 TNF receptor superfamily member 10b Homo sapiens 132-135 7706255-6 1995 The t-PA/DR5 element interacted with the heterodimer composed of retinoic acid receptor alpha and retinoid X receptor alpha in vitro, whereas its mutation abolished induction by RA in transient expression. Tretinoin 178-180 TNF receptor superfamily member 10b Homo sapiens 9-12 16287099-6 2006 MG132 massively induced TRAIL receptor DR4 and DR5 membrane expression in HeLa, whereas in SiHa only DR5 membrane expression was upregulated from almost undetectable to high levels. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-5 TNF receptor superfamily member 10b Homo sapiens 47-50 8409432-7 1993 Finally, bulk sequencing profiles contained signals from further putative anchor residues clustering in the NH2-terminal region:tyrosine, phenylalanine, leucine, isoleucine, and valine are enriched at positions 2 to 4 in DR1, DR5, and DR6, however, at positions 4 to 6 in DR3. Tyrosine 128-136 TNF receptor superfamily member 10b Homo sapiens 226-229 8409432-7 1993 Finally, bulk sequencing profiles contained signals from further putative anchor residues clustering in the NH2-terminal region:tyrosine, phenylalanine, leucine, isoleucine, and valine are enriched at positions 2 to 4 in DR1, DR5, and DR6, however, at positions 4 to 6 in DR3. Valine 178-184 TNF receptor superfamily member 10b Homo sapiens 226-229 8151585-0 1994 Association of HLA-DR5 with mucocutaneous lesions in patients with rheumatoid arthritis receiving gold sodium thiomalate. Gold Sodium Thiomalate 98-120 TNF receptor superfamily member 10b Homo sapiens 19-22 1993839-6 1991 For DR2 and DR3 the shortest peptide was residues 6-15, an additional serine (residue 5) was required for DR1 and DR7 and an aspartic acid (residue 4) for DR4, DR5, and DR6. Aspartic Acid 125-138 TNF receptor superfamily member 10b Homo sapiens 160-163 1899160-0 1991 HLA-DR1, -DR5, and -DR8 antigen disparities are associated with acute steroid-resistant rejection and poor kidney graft survival. Steroids 70-77 TNF receptor superfamily member 10b Homo sapiens 10-13 1673331-2 1991 The 285-fold doxorubicin-resistant colon adenocarcinoma subline, LoVo/DR5, was found to overexpress the mRNA for P-glycoprotein without the concomitant requirement of MDR1 gene amplification, suggesting that relatively high levels of P-glycoprotein mediated multiple drug resistance may occur by transcriptional activation of the gene. Doxorubicin 13-24 TNF receptor superfamily member 10b Homo sapiens 70-73 33767436-7 2021 We showed that CRISPR/Cas9 mediated silencing of DR5 could block Bortezomib-mediated re-sensitization, demonstrating its critical role. Bortezomib 65-75 TNF receptor superfamily member 10b Homo sapiens 49-52 26074143-0 2015 RAD001 (everolimus) enhances TRAIL cytotoxicity in human leukemic Jurkat T cells by upregulating DR5. Everolimus 8-18 TNF receptor superfamily member 10b Homo sapiens 97-100 26074143-3 2015 In the present study, we showed that the Akt/mTOR inhibitor RAD001 (everolimus) induced cell death in a dose-dependent manner and enhanced TRAIL-induced apoptosis in human leukemic Jurkat T cells, which show PI3K/Akt/mTOR pathway activation and basal expression levels of death receptor (DR) 5 (TRAIL-R2). Everolimus 68-78 TNF receptor superfamily member 10b Homo sapiens 272-293 26074143-3 2015 In the present study, we showed that the Akt/mTOR inhibitor RAD001 (everolimus) induced cell death in a dose-dependent manner and enhanced TRAIL-induced apoptosis in human leukemic Jurkat T cells, which show PI3K/Akt/mTOR pathway activation and basal expression levels of death receptor (DR) 5 (TRAIL-R2). Everolimus 68-78 TNF receptor superfamily member 10b Homo sapiens 295-303 16287099-14 2006 Our results indicate that not only DR4 and DR5 upregulation but also XIAP inactivation contribute to rhTRAIL sensitization by MG132 in cervical cancer cell lines. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 126-131 TNF receptor superfamily member 10b Homo sapiens 43-46 16287099-9 2006 Although p53 siRNA partially inhibited MG132-induced DR5 upregulation in HeLa and SiHa, no effect on rhTRAIL-induced apoptosis was observed. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 39-44 TNF receptor superfamily member 10b Homo sapiens 53-56 34926196-0 2021 Combination of oridonin and TRAIL induces apoptosis in uveal melanoma cells by upregulating DR5. oridonin 15-23 TNF receptor superfamily member 10b Homo sapiens 92-95 34288496-0 2022 Pyronaridine induces apoptosis in Non-small cell lung cancer cells by upregulating DR5 expression and inhibiting EGFR. pyronaridine 0-12 TNF receptor superfamily member 10b Homo sapiens 83-86 34288496-4 2022 The data showed that pyronaridine could upregulate the expression of TRAIL (TNF-related apoptosis-inducing ligand)-mediated DR5 (Death receptor 5) to promote cellular apoptosis. pyronaridine 21-33 TNF receptor superfamily member 10b Homo sapiens 124-127 34288496-4 2022 The data showed that pyronaridine could upregulate the expression of TRAIL (TNF-related apoptosis-inducing ligand)-mediated DR5 (Death receptor 5) to promote cellular apoptosis. pyronaridine 21-33 TNF receptor superfamily member 10b Homo sapiens 129-145 34926196-9 2021 The Western blot results showed that the protein expression levels of the DR5, a-caspase-3, and BAX increased, while the expression levels of the anti-apoptosis-related proteins XIAP and BCL-2 were suppressed when TRAIL and oridonin simultaneously administered to MUM-2B cells. oridonin 224-232 TNF receptor superfamily member 10b Homo sapiens 74-77 34926196-10 2021 Interfering the expression of DR5 gene in MUM-2B cells could reverse the inhibitory effect of oridonin and TRAIL on the proliferation and apoptosis induction of MUM-2B cells. oridonin 94-102 TNF receptor superfamily member 10b Homo sapiens 30-33 34926196-11 2021 CONCLUSION: The inhibitory effects of oridonin and TRAIL on MUM-2B cells are significantly enhanced when they were administered as a combined treatment, which may ascribe to up-regulation of DR5. oridonin 38-46 TNF receptor superfamily member 10b Homo sapiens 191-194 34681736-5 2021 ROS-induced endoplasmic reticulum (ER) stress by OSMI-1 not only upregulated CHOP-DR5 signaling but also activated Jun-N-terminal kinase (JNK), resulting in a decrease in Bcl2 and the release of cytochrome c from mitochondria. ros 0-3 TNF receptor superfamily member 10b Homo sapiens 82-85 34599545-0 2021 CSC-3436 sensitizes triple negative breast cancer cells to TRAIL-induced apoptosis through ROS-mediated p38/CHOP/death receptor 5 signaling pathways. csc-3436 0-8 TNF receptor superfamily member 10b Homo sapiens 113-129 34599545-0 2021 CSC-3436 sensitizes triple negative breast cancer cells to TRAIL-induced apoptosis through ROS-mediated p38/CHOP/death receptor 5 signaling pathways. Reactive Oxygen Species 91-94 TNF receptor superfamily member 10b Homo sapiens 113-129 34599545-4 2021 We found that CSC-3436 potentiated TRAIL-induced apoptosis in TRAIL-resistant TNBC cells and this correlated with the upregulation of death receptors (DR)-5 and down-regulation of decreased decoy receptor (DcR)-1 expression. csc-3436 14-22 TNF receptor superfamily member 10b Homo sapiens 134-156 34599545-7 2021 In addition, the induction of DR5 by CSC-3436 was found to be dependent on the modulation of reactive oxygen species (ROS)/p38/C/EBP-homologous protein (CHOP) signaling pathways. csc-3436 37-45 TNF receptor superfamily member 10b Homo sapiens 30-33 34599545-7 2021 In addition, the induction of DR5 by CSC-3436 was found to be dependent on the modulation of reactive oxygen species (ROS)/p38/C/EBP-homologous protein (CHOP) signaling pathways. Reactive Oxygen Species 93-116 TNF receptor superfamily member 10b Homo sapiens 30-33 34599545-7 2021 In addition, the induction of DR5 by CSC-3436 was found to be dependent on the modulation of reactive oxygen species (ROS)/p38/C/EBP-homologous protein (CHOP) signaling pathways. Reactive Oxygen Species 118-121 TNF receptor superfamily member 10b Homo sapiens 30-33 34599545-8 2021 Overall, our results indicated that CSC-3436 could potentiate the apoptotic effects of TRAIL through down-regulation of cell survival proteins and upregulation of DR5 via the ROS-mediated upregulation of CHOP protein. csc-3436 36-44 TNF receptor superfamily member 10b Homo sapiens 163-166 34599545-8 2021 Overall, our results indicated that CSC-3436 could potentiate the apoptotic effects of TRAIL through down-regulation of cell survival proteins and upregulation of DR5 via the ROS-mediated upregulation of CHOP protein. Reactive Oxygen Species 175-178 TNF receptor superfamily member 10b Homo sapiens 163-166 34711645-0 2021 Multimeric Anti-DR5 IgM Agonist Antibody IGM-8444 Is a Potent Inducer of Cancer Cell Apoptosis and Synergizes with Chemotherapy and BCL-2 Inhibitor ABT-199. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 148-151 TNF receptor superfamily member 10b Homo sapiens 16-19 34503423-7 2022 NU 9056 also induced apoptosis by upregulating DR4, DR5, and caspase 8 expressions. 1,2-bis(isothiazol-5-yl)disulfane 0-7 TNF receptor superfamily member 10b Homo sapiens 52-55 34659909-7 2021 Upon treatment with the imipridones, DIPG cell lines engaged CLpP/CLPX, the integrated stress response with ATF4 activation, and TRAIL death receptor 5 (DR5) induction. imipridones 24-35 TNF receptor superfamily member 10b Homo sapiens 153-156 34611476-8 2021 TRAILR2 was down-regulated by oxaliplatin and 5-FU, was not affected by CPT-11, and was increased by cisplatin. Oxaliplatin 30-41 TNF receptor superfamily member 10b Homo sapiens 0-7 34611476-8 2021 TRAILR2 was down-regulated by oxaliplatin and 5-FU, was not affected by CPT-11, and was increased by cisplatin. Fluorouracil 46-50 TNF receptor superfamily member 10b Homo sapiens 0-7 34611476-8 2021 TRAILR2 was down-regulated by oxaliplatin and 5-FU, was not affected by CPT-11, and was increased by cisplatin. Cisplatin 101-110 TNF receptor superfamily member 10b Homo sapiens 0-7 34246076-0 2021 EZH2i EPZ-6438 and HDACi vorinostat synergize with ONC201/TIC10 to activate integrated stress response, DR5, reduce H3K27 methylation, ClpX and promote apoptosis of multiple tumor types including DIPG. Uridine Diphosphate N-Acetylmuramic Acid 6-9 TNF receptor superfamily member 10b Homo sapiens 104-107 34575490-5 2021 The aim of the study was to fabricate Amph-PVP-based nanoparticles covalently conjugated with antitumor DR5-specific TRAIL (Tumor necrosis factor-related apoptosis-inducing ligand) variant DR5-B and to evaluate their in vitro cytotoxicity in 3D tumor spheroids. amph-pvp 38-46 TNF receptor superfamily member 10b Homo sapiens 104-107 34475055-7 2021 We also found that hyper-acetylation of histone by CBUD-1001 treatment leads to up-regulation of death receptor (DR) 5 in a dose- and time-dependent manner. cbud-1001 51-60 TNF receptor superfamily member 10b Homo sapiens 97-118 34475055-8 2021 Furthermore, we identified that enhanced sensitivity to TRAIL by combination with CBUD-1001 depends on the MAPK/CHOP axis, being a key mediator of DR5. cbud-1001 82-91 TNF receptor superfamily member 10b Homo sapiens 147-150 34475055-9 2021 CONCLUSION: A novel HDAC inhibitor CBUD-1001 sensitizes TRAIL-induced apoptosis via up-regulation of DR5, and that CBUD-1001 and TRAIL combination treatment offers an effective strategy to overcome TRAIL resistance in CRC cells. cbud-1001 35-44 TNF receptor superfamily member 10b Homo sapiens 101-104 34549714-0 2021 (Effect of DR5-mediated docetaxel-loaded lipid microbubble combined with ultrasoundtargeted microbubble destruction on HepG2 cell proliferation and apoptosis). Docetaxel 24-33 TNF receptor superfamily member 10b Homo sapiens 11-14 34549714-1 2021 OBJECTIVE: To investigate the effect of DR5-mediated docetaxel-targeted lipid microbubbles (MBs) combined with ultrasound-targeted microbubble destruction on apoptosis and expressions of Bcl-2, nuclear factor-kappaB(NF-kappaB), caspase-8, and DR5 in human HepG2 cells. Docetaxel 53-62 TNF receptor superfamily member 10b Homo sapiens 40-43 34549714-1 2021 OBJECTIVE: To investigate the effect of DR5-mediated docetaxel-targeted lipid microbubbles (MBs) combined with ultrasound-targeted microbubble destruction on apoptosis and expressions of Bcl-2, nuclear factor-kappaB(NF-kappaB), caspase-8, and DR5 in human HepG2 cells. Docetaxel 53-62 TNF receptor superfamily member 10b Homo sapiens 243-246 34582654-0 2021 Thymoquinone Crosstalks with DR5 to Sensitize TRAIL Resistance and Stimulate ROS-Mediated Cancer Apoptosis. thymoquinone 0-12 TNF receptor superfamily member 10b Homo sapiens 29-32 34582654-0 2021 Thymoquinone Crosstalks with DR5 to Sensitize TRAIL Resistance and Stimulate ROS-Mediated Cancer Apoptosis. Reactive Oxygen Species 77-80 TNF receptor superfamily member 10b Homo sapiens 29-32 34582654-6 2021 CONCLUSIONS: The synergistic influence between TQ which induced the DR5 and TRAIL, facilitating the connection between TRAIL and its receptors on the cancerous cell membrane. thymoquinone 47-49 TNF receptor superfamily member 10b Homo sapiens 68-71 34479917-7 2021 Furthermore, paclitaxel induced ROS-mediated DNA damage that triggered the activation of the extrinsic pathway of apoptosis via the up-regulation of death receptor (DR5) and caspase-8 activation. Paclitaxel 13-23 TNF receptor superfamily member 10b Homo sapiens 165-168 34479917-7 2021 Furthermore, paclitaxel induced ROS-mediated DNA damage that triggered the activation of the extrinsic pathway of apoptosis via the up-regulation of death receptor (DR5) and caspase-8 activation. Reactive Oxygen Species 32-35 TNF receptor superfamily member 10b Homo sapiens 165-168 34497804-4 2021 For functional analyses, we knocked down one of the cisplatin inducible lncRNAs, death receptor 5 antisense (DR5-AS) lncRNA, which resulted in a morphological change in HeLa cell shape without inducing any cell death. Cisplatin 52-61 TNF receptor superfamily member 10b Homo sapiens 81-97 34497804-4 2021 For functional analyses, we knocked down one of the cisplatin inducible lncRNAs, death receptor 5 antisense (DR5-AS) lncRNA, which resulted in a morphological change in HeLa cell shape without inducing any cell death. Cisplatin 52-61 TNF receptor superfamily member 10b Homo sapiens 109-112 34246076-0 2021 EZH2i EPZ-6438 and HDACi vorinostat synergize with ONC201/TIC10 to activate integrated stress response, DR5, reduce H3K27 methylation, ClpX and promote apoptosis of multiple tumor types including DIPG. Vorinostat 25-35 TNF receptor superfamily member 10b Homo sapiens 104-107 34187946-10 2021 In A549 and NCI-H2170 cells vanillin oxime exposure caused significant (p < 0.05) enhancement in CHOP and DR5 mRNA expression. Vanillin Oxime 28-42 TNF receptor superfamily member 10b Homo sapiens 106-109 34903991-6 2021 The result indicated that zebularine and TSA changed the expression level of the Bax, Bak, Bim Bcl-2, Bcl-xL, Mcl-1, DR4, DR5, FAS, FAS-L, TRAIL, DNA methyltransferase 1, 3a, and 3b, histone deacetylase inhibitors 1, 2, and 3 by which induced cell apoptosis and inhibit cell growth in all three cell lines. pyrimidin-2-one beta-ribofuranoside 26-36 TNF receptor superfamily member 10b Homo sapiens 122-125 34903991-6 2021 The result indicated that zebularine and TSA changed the expression level of the Bax, Bak, Bim Bcl-2, Bcl-xL, Mcl-1, DR4, DR5, FAS, FAS-L, TRAIL, DNA methyltransferase 1, 3a, and 3b, histone deacetylase inhibitors 1, 2, and 3 by which induced cell apoptosis and inhibit cell growth in all three cell lines. trichostatin A 41-44 TNF receptor superfamily member 10b Homo sapiens 122-125 34360606-12 2021 AP-1 also significantly enhanced the apoptotic effect of DOX after 24 h of treatment with significant upregulation of catalase, HTRA2/Omi, FADD together with DR5 and DR4 TRAIL-mediated apoptosis (p < 0.05), contributing to the antiproliferative activity of AP-1. Doxorubicin 57-60 TNF receptor superfamily member 10b Homo sapiens 158-161 34080659-6 2021 Genetic inhibitors of DR4 and DR5 significantly reduced amitriptyline-enhanced TRAIL-mediated apoptosis. Amitriptyline 56-69 TNF receptor superfamily member 10b Homo sapiens 30-33 34080659-8 2021 Blocking autophagy flux with the final stage autophagy inhibitor chloroquine (CQ) also upregulated DR4 and DR5 expression. Chloroquine 65-76 TNF receptor superfamily member 10b Homo sapiens 107-110 34080659-8 2021 Blocking autophagy flux with the final stage autophagy inhibitor chloroquine (CQ) also upregulated DR4 and DR5 expression. Chloroquine 78-80 TNF receptor superfamily member 10b Homo sapiens 107-110 34080659-11 2021 Overall, the present results contributed to understanding the mechanism responsible for the synergistic anticancer effect of amitriptyline and TRAIL and also presented a novel mechanism involved in DR4 and DR5 upregulation. Amitriptyline 125-138 TNF receptor superfamily member 10b Homo sapiens 206-209 35616096-4 2022 CPT MV could produce ROS in the targeted cells upon laser irradiation to improve death receptor (DR)-5 expression and trigger Cyt c release from mitochondria. ros 21-24 TNF receptor superfamily member 10b Homo sapiens 81-102 34195076-0 2021 Fangchinoline Inhibits Human Esophageal Cancer by Transactivating ATF4 to Trigger Both Noxa-Dependent Intrinsic and DR5-Dependent Extrinsic Apoptosis. fangchinoline 0-13 TNF receptor superfamily member 10b Homo sapiens 116-119 34195076-6 2021 Moreover, we found that FCL coordinatively triggered Noxa-dependent intrinsic apoptosis and DR5-dependent extrinsic apoptosis by transactivating ATF4, which is a novel mechanism. fangchinoline 24-27 TNF receptor superfamily member 10b Homo sapiens 92-95 34141868-6 2021 Metformin-upregulated TRAIL was secreted into conditioned medium (CM) and found to be functional, since the CM promoted TNBC cells undergoing apoptosis, which was abrogated by a recombinant TRAIL-R2-Fc chimera. Metformin 0-9 TNF receptor superfamily member 10b Homo sapiens 190-198 34141868-7 2021 Moreover, blockade of TRAIL binding to DR4/DR5 or specific knockdown of TRAIL expression significantly attenuated metformin-induced apoptosis. Metformin 114-123 TNF receptor superfamily member 10b Homo sapiens 43-46 34141870-5 2021 Oba01 utilizes anti-DR5 humanized monoclonal antibody (zaptuzumab) coupled via a cleavable linker to monomethyl auristatin E (MMAE). monomethyl auristatin E 101-124 TNF receptor superfamily member 10b Homo sapiens 20-23 34141870-5 2021 Oba01 utilizes anti-DR5 humanized monoclonal antibody (zaptuzumab) coupled via a cleavable linker to monomethyl auristatin E (MMAE). monomethyl auristatin E 126-130 TNF receptor superfamily member 10b Homo sapiens 20-23 35247515-8 2022 Together, the results herein reveal DDAs as the first small molecule, active site inhibitors of AGR2 and ERp44, and demonstrate roles for AGR2 and ERp44 in regulating the activity, stability, and localization of DR4 and DR5, and activation of Caspase 8. ddas 36-40 TNF receptor superfamily member 10b Homo sapiens 220-223 35039880-0 2022 (Corrigendum) 3-Bromopyruvate sensitizes human breast cancer cells to TRAIL-induced apoptosis via the phosphorylated AMPK-mediated upregulation of DR5. bromopyruvate 14-29 TNF receptor superfamily member 10b Homo sapiens 147-150 35568793-0 2022 20(s)-ginsenoside Rh2 promotes TRAIL-induced apoptosis by upregulating DR5 in human hepatocellular carcinoma cells. Ginsenoside Rh2 0-21 TNF receptor superfamily member 10b Homo sapiens 71-74 35568793-7 2022 At the same time, Rh2 can further enhance TRAIL-induced apoptosis by upregulating the death receptor 5, thereby significantly enhancing its anti-tumor effect. rh2 18-21 TNF receptor superfamily member 10b Homo sapiens 86-102 35149588-7 2022 Deletion of DR5 in cancer cells in vitro and in vivo abrogated the potentiating effects of CDK4/6 inhibitors on immune cytokine TNF-related apoptosis-inducing ligand (TRAIL), 5-fluorouracil (5-FU) chemotherapy, and anti-PD-1 immunotherapy. Fluorouracil 175-189 TNF receptor superfamily member 10b Homo sapiens 12-15 35149588-7 2022 Deletion of DR5 in cancer cells in vitro and in vivo abrogated the potentiating effects of CDK4/6 inhibitors on immune cytokine TNF-related apoptosis-inducing ligand (TRAIL), 5-fluorouracil (5-FU) chemotherapy, and anti-PD-1 immunotherapy. Fluorouracil 191-195 TNF receptor superfamily member 10b Homo sapiens 12-15 35455398-5 2022 Specifically, dehydrocrenatidine significantly increased the expression of extrinsic pathway components (FAS, DR5, FADD, and TRADD) as well as intrinsic pathway components (Bax and Bim L/S) in liver cancer cells. dehydrocrenatidine 14-32 TNF receptor superfamily member 10b Homo sapiens 110-113 35086954-7 2022 Subsequent MEDI3039 combination-screening of TRAIL-R2, caspase-8, FADD and BID knockout models with 60 compounds with varying mechanisms-of-action identified 2 inhibitor of apoptosis proteins (IAPs) that exhibited strong synergy with MEDI3039 that could reverse resistance only in BID-deleted models. medi3039 234-242 TNF receptor superfamily member 10b Homo sapiens 45-53 35141009-8 2022 Cariporide treatment also decreased CRC cell shed TRAIL-R2, TRAIL-R3, and PD-L1 which is relevant in the context of immunotherapy. cariporide 0-10 TNF receptor superfamily member 10b Homo sapiens 50-58 35077998-13 2022 In conclusion, we identified that the concomitant regulation of both antiapoptotic and proapoptotic factors and DR5 is an essential molecular mechanism for overcoming TRAIL resistance in SH-SY5Y and the combination of ERN-T and AZD5582 potentially constitutes a novel therapeutic strategy, which is highly effective and safe for neuroblastoma. ern-t 218-223 TNF receptor superfamily member 10b Homo sapiens 112-115 35077998-13 2022 In conclusion, we identified that the concomitant regulation of both antiapoptotic and proapoptotic factors and DR5 is an essential molecular mechanism for overcoming TRAIL resistance in SH-SY5Y and the combination of ERN-T and AZD5582 potentially constitutes a novel therapeutic strategy, which is highly effective and safe for neuroblastoma. N,N'-(2,2'-(hexa-2,4-diyne-1,6-diylbis(oxy))bis(2,3-dihydro-1H-indene-2,1-diyl))bis(1-(2-cyclohexyl-2-(2-(methylamino)propanamido)acetyl)pyrrolidine-2-carboxamide) 228-235 TNF receptor superfamily member 10b Homo sapiens 112-115 35200638-9 2022 (-)-Agelasidine A also increased apoptosis-associated proteins (DR4, DR5, FAS), which are related to extrinsic pathways. agelasidine A 4-17 TNF receptor superfamily member 10b Homo sapiens 69-72 35200638-13 2022 Moreover, 4-PBA, an ER stress inhibitor, could also abrogate (-)-agelasidine A-induced cell viability reduction, annexin V+ apoptosis, death receptor (DR4, DR5, FAS) expression, mitochondrial dysfunction, and cytochrome c release. 4-phenylbutylamine 10-15 TNF receptor superfamily member 10b Homo sapiens 156-159 35200638-13 2022 Moreover, 4-PBA, an ER stress inhibitor, could also abrogate (-)-agelasidine A-induced cell viability reduction, annexin V+ apoptosis, death receptor (DR4, DR5, FAS) expression, mitochondrial dysfunction, and cytochrome c release. agelasidine A 65-78 TNF receptor superfamily member 10b Homo sapiens 156-159 35047402-7 2021 We found that the combination of DHA-TF and the death receptor 5-specific TRAIL variant DHER leads to an increase in DR5 expression in all four TNBC cell lines, while higher cytotoxicity was observed in MDA-MB-231, and MDA-MB-436. artenimol 33-36 TNF receptor superfamily member 10b Homo sapiens 117-120 35047402-7 2021 We found that the combination of DHA-TF and the death receptor 5-specific TRAIL variant DHER leads to an increase in DR5 expression in all four TNBC cell lines, while higher cytotoxicity was observed in MDA-MB-231, and MDA-MB-436. dher 88-92 TNF receptor superfamily member 10b Homo sapiens 48-64 35047402-7 2021 We found that the combination of DHA-TF and the death receptor 5-specific TRAIL variant DHER leads to an increase in DR5 expression in all four TNBC cell lines, while higher cytotoxicity was observed in MDA-MB-231, and MDA-MB-436. dher 88-92 TNF receptor superfamily member 10b Homo sapiens 117-120 2715056-7 1989 A comparison of all the available protein sequences of DRB gene products showed that the first hypervariable region of DR3 and DR5 (and DRw6), and no other region, contains the sequence Glu9-Tyr-Ser-Thr-Ser13. Serine 195-198 TNF receptor superfamily member 10b Homo sapiens 127-130 2490151-5 1989 We found HLA-DR5 to be significantly overrepresented in the patients with RSA who aborted again after treatment with paternal mononuclear cell immunotherapy, compared with the incidence of this phenotype in a control population. rabbit sperm membrane autoantigen 74-77 TNF receptor superfamily member 10b Homo sapiens 13-16 2715056-7 1989 A comparison of all the available protein sequences of DRB gene products showed that the first hypervariable region of DR3 and DR5 (and DRw6), and no other region, contains the sequence Glu9-Tyr-Ser-Thr-Ser13. Threonine 199-202 TNF receptor superfamily member 10b Homo sapiens 127-130 2466297-3 1989 The binding of the CS peptides to DR5 or DRw6 proteins was then determined in a proliferation assay using two established DR5 or DRw6-restricted T-cell clones with specificity for the stimulator peptides as responder cells. Cesium 19-21 TNF receptor superfamily member 10b Homo sapiens 34-37 2466297-3 1989 The binding of the CS peptides to DR5 or DRw6 proteins was then determined in a proliferation assay using two established DR5 or DRw6-restricted T-cell clones with specificity for the stimulator peptides as responder cells. Cesium 19-21 TNF receptor superfamily member 10b Homo sapiens 122-125 2466297-4 1989 One of five CS peptides, comprising together about 50% of the CS protein sequence, was found to compete with the binding of the stimulator peptides to DR5 and DRw6. Cesium 12-14 TNF receptor superfamily member 10b Homo sapiens 151-154 2466297-5 1989 The CS peptide CS-(378-398), binding to DR5 and DRw6, was then shown to be able to induce primary in vitro responses of T cells from donors with DR5 and DRw6 haplotypes. Cesium 4-6 TNF receptor superfamily member 10b Homo sapiens 40-43 2466297-5 1989 The CS peptide CS-(378-398), binding to DR5 and DRw6, was then shown to be able to induce primary in vitro responses of T cells from donors with DR5 and DRw6 haplotypes. Cesium 4-6 TNF receptor superfamily member 10b Homo sapiens 145-148 2466297-5 1989 The CS peptide CS-(378-398), binding to DR5 and DRw6, was then shown to be able to induce primary in vitro responses of T cells from donors with DR5 and DRw6 haplotypes. Cesium 15-17 TNF receptor superfamily member 10b Homo sapiens 40-43 2466297-5 1989 The CS peptide CS-(378-398), binding to DR5 and DRw6, was then shown to be able to induce primary in vitro responses of T cells from donors with DR5 and DRw6 haplotypes. Cesium 15-17 TNF receptor superfamily member 10b Homo sapiens 145-148 33857626-0 2021 Sodium fluoride activates the extrinsic apoptosis via regulating NOX4/ROS-mediated p53/DR5 signaling pathway in lung cells both in vitro and in vivo. Sodium Fluoride 0-15 TNF receptor superfamily member 10b Homo sapiens 87-90 2445714-2 1987 The population distribution shows that each of the DRw52 associated specificities DR3, DR5, and DRw6 may occur with and without LB-Q1. lb-q1 128-133 TNF receptor superfamily member 10b Homo sapiens 87-90 3890265-6 1985 These studies document that the MB1 allodeterminant resides on DS molecules from this DR5 cell line and provide additional evidence that MB1 and MB3 are alleles of the same Class II antigen system. Deuterium 63-65 TNF receptor superfamily member 10b Homo sapiens 86-89 3935278-4 1985 This oligonucleotide hybridized only with the DNA from DR3 or DR5 individuals tested, even under stringent conditions of washing. Oligonucleotides 5-20 TNF receptor superfamily member 10b Homo sapiens 62-65 3871759-1 1985 Antiserum 8w670 which had been classified as anti-DR5 in the Workshop analysis was found by the direct binding test to react with about 40% of Ia molecules in an Ia preparation from LG38 cells (DR5,5) that was deleted of DR molecules and DR-like molecules we call BR and enriched in DC molecules by pretreatment with a rabbit antiserum raised against alpha-subunits of DR and BR molecules. Bromine 264-266 TNF receptor superfamily member 10b Homo sapiens 50-53 3871759-1 1985 Antiserum 8w670 which had been classified as anti-DR5 in the Workshop analysis was found by the direct binding test to react with about 40% of Ia molecules in an Ia preparation from LG38 cells (DR5,5) that was deleted of DR molecules and DR-like molecules we call BR and enriched in DC molecules by pretreatment with a rabbit antiserum raised against alpha-subunits of DR and BR molecules. Bromine 376-378 TNF receptor superfamily member 10b Homo sapiens 50-53 6328695-5 1984 The persons with HLA-DR5 had significantly higher anti-EBNA titers than those without DR5. N-tert-Butyl-N-ethylnitrosamine 55-59 TNF receptor superfamily member 10b Homo sapiens 21-24 33857626-0 2021 Sodium fluoride activates the extrinsic apoptosis via regulating NOX4/ROS-mediated p53/DR5 signaling pathway in lung cells both in vitro and in vivo. Reactive Oxygen Species 70-73 TNF receptor superfamily member 10b Homo sapiens 87-90 33857626-8 2021 Specifically, NOX4 knockdown inhibited NaF-induced the activation of p53/DR5 axis by reducing NOX4-derived ROS production. Reactive Oxygen Species 107-110 TNF receptor superfamily member 10b Homo sapiens 73-76 33885752-12 2021 Lenalidomide also synergized with HexaBody-DR5/DR5, but only via its immunomodulatory effects, presumably by enhancing the antibody-dependent cellular cytotoxicity activity of HexaBody-DR5/DR5. Lenalidomide 0-12 TNF receptor superfamily member 10b Homo sapiens 43-46 33942150-18 2021 Multivalent TRAIL polymers bind and activate DR4 and DR5 specifically and exclusively, triggering the signaling pathways with higher potency, and greater efficacy than TRAIL. Polymers 18-26 TNF receptor superfamily member 10b Homo sapiens 53-56 33885752-12 2021 Lenalidomide also synergized with HexaBody-DR5/DR5, but only via its immunomodulatory effects, presumably by enhancing the antibody-dependent cellular cytotoxicity activity of HexaBody-DR5/DR5. Lenalidomide 0-12 TNF receptor superfamily member 10b Homo sapiens 47-50 33885752-12 2021 Lenalidomide also synergized with HexaBody-DR5/DR5, but only via its immunomodulatory effects, presumably by enhancing the antibody-dependent cellular cytotoxicity activity of HexaBody-DR5/DR5. Lenalidomide 0-12 TNF receptor superfamily member 10b Homo sapiens 47-50 33885752-12 2021 Lenalidomide also synergized with HexaBody-DR5/DR5, but only via its immunomodulatory effects, presumably by enhancing the antibody-dependent cellular cytotoxicity activity of HexaBody-DR5/DR5. Lenalidomide 0-12 TNF receptor superfamily member 10b Homo sapiens 47-50 33898931-6 2021 Consistent with our earlier findings, the BMMSCs protected MM cells against KHYG-1 and DR5-agonistic antibodies by inducing resistance mechanisms that were largely abrogated by the small molecule FL118, an inhibitor of multiple antiapoptotic proteins including Survivin, Mcl-1, and XIAP. 7-ethyl-7-hydroxy-10H-1,3-Dioxolo(4,5-g)pyrano(3',4':6,7)indolizino(1,2-b)quinoline-8,11(7H,12H)-dione 196-201 TNF receptor superfamily member 10b Homo sapiens 87-90 33937072-9 2021 Taken together, our study showed that LCT-3d induced apoptosis via DR5-mediated mitochondrial apoptotic pathway in gastric cancer cells. N-[5-(4-Nitrophenyl)-1,3,4-thiadiazol-2-yl]acetamide 38-41 TNF receptor superfamily member 10b Homo sapiens 67-70 33894272-8 2021 KEY FINDINGS: Upon treatment with oleuropein, the expression of P21, P53, and TNFRSF10B increased while that of Bcl-2 and Mcl1 decreased. oleuropein 34-44 TNF receptor superfamily member 10b Homo sapiens 78-87 33937072-0 2021 LCT-3d Induces Oxidative Stress-Mediated Apoptosis by Upregulating Death Receptor 5 in Gastric Cancer Cells. N-[5-(4-Nitrophenyl)-1,3,4-thiadiazol-2-yl]acetamide 0-3 TNF receptor superfamily member 10b Homo sapiens 67-83 33841136-14 2021 DR5 upregulation induced by alternol required the production of reactive oxygen species (ROS). Alternol 28-36 TNF receptor superfamily member 10b Homo sapiens 0-3 33937072-4 2021 The up-regulation of Death receptor 5 (DR5) in MGC803 cells was observed with LCT-3d treatment. N-[5-(4-Nitrophenyl)-1,3,4-thiadiazol-2-yl]acetamide 78-81 TNF receptor superfamily member 10b Homo sapiens 21-37 33937072-4 2021 The up-regulation of Death receptor 5 (DR5) in MGC803 cells was observed with LCT-3d treatment. N-[5-(4-Nitrophenyl)-1,3,4-thiadiazol-2-yl]acetamide 78-81 TNF receptor superfamily member 10b Homo sapiens 39-42 33937072-5 2021 Knockdown of DR5 on MGC803 cells partially reversed the LCT-3d-induced mitochondrial apoptosis. N-[5-(4-Nitrophenyl)-1,3,4-thiadiazol-2-yl]acetamide 56-59 TNF receptor superfamily member 10b Homo sapiens 13-16 33937072-7 2021 The results demonstrate that the elevating ROS can up-regulate the expression of DR5, resulting in apoptosis via mitochondrial pathway. Reactive Oxygen Species 43-46 TNF receptor superfamily member 10b Homo sapiens 81-84 33916015-0 2021 Death Receptor 5 (TNFRSF10B) Is Upregulated and TRAIL Resistance Is Reversed in Hypoxia and Normoxia in Colorectal Cancer Cell Lines after Treatment with Skyrin, the Active Metabolite of Hypericum spp. skyrin 154-160 TNF receptor superfamily member 10b Homo sapiens 0-16 33916015-0 2021 Death Receptor 5 (TNFRSF10B) Is Upregulated and TRAIL Resistance Is Reversed in Hypoxia and Normoxia in Colorectal Cancer Cell Lines after Treatment with Skyrin, the Active Metabolite of Hypericum spp. skyrin 154-160 TNF receptor superfamily member 10b Homo sapiens 18-27 33921748-7 2021 Altogether, our findings establish ATF3 as a pro-apoptotic protein in arsenic-induced airway epithelial apoptosis through transcriptionally regulating DR5 and Bcl-xL, highlighting the potential of ATF3 as an early and sensitive biomarker for arsenic-caused lung injury. Arsenic 70-77 TNF receptor superfamily member 10b Homo sapiens 151-154 33841136-10 2021 Results: When the mechanisms were investigated, we discovered that alternol increased DR5 expression. Alternol 67-75 TNF receptor superfamily member 10b Homo sapiens 86-89 33841136-14 2021 DR5 upregulation induced by alternol required the production of reactive oxygen species (ROS). Reactive Oxygen Species 64-87 TNF receptor superfamily member 10b Homo sapiens 0-3 33841136-11 2021 DR5 knockdown by siRNA eliminated the enhanced effect of alternol on TRAIL-mediated apoptosis. Alternol 57-65 TNF receptor superfamily member 10b Homo sapiens 0-3 33841136-14 2021 DR5 upregulation induced by alternol required the production of reactive oxygen species (ROS). Reactive Oxygen Species 89-92 TNF receptor superfamily member 10b Homo sapiens 0-3 33841136-15 2021 Removing ROS inhibited the induction of DR5 and blocked the antiapoptotic proteins induced by alternol. Reactive Oxygen Species 9-12 TNF receptor superfamily member 10b Homo sapiens 40-43 33841136-16 2021 Conclusion: Taken together, our research suggested that alternol increased TRAIL-mediated apoptosis via inhibiting antiapoptotic proteins and upregulating DR5 levels via ROS generation and the CHOP pathway. Alternol 56-64 TNF receptor superfamily member 10b Homo sapiens 155-158 33676890-0 2021 Citrus-derived DHCP inhibits mitochondrial complex II to enhance TRAIL sensitivity via ROS-induced DR5 upregulation. dhcp 15-19 TNF receptor superfamily member 10b Homo sapiens 99-102 33737574-0 2021 Death agonist antibody against TRAILR2/DR5/TNFRSF10B enhances birinapant anti-tumor activity in HPV-positive head and neck squamous cell carcinomas. birinapant 62-72 TNF receptor superfamily member 10b Homo sapiens 31-38 33737574-0 2021 Death agonist antibody against TRAILR2/DR5/TNFRSF10B enhances birinapant anti-tumor activity in HPV-positive head and neck squamous cell carcinomas. birinapant 62-72 TNF receptor superfamily member 10b Homo sapiens 39-42 33737574-0 2021 Death agonist antibody against TRAILR2/DR5/TNFRSF10B enhances birinapant anti-tumor activity in HPV-positive head and neck squamous cell carcinomas. birinapant 62-72 TNF receptor superfamily member 10b Homo sapiens 43-52 33676890-0 2021 Citrus-derived DHCP inhibits mitochondrial complex II to enhance TRAIL sensitivity via ROS-induced DR5 upregulation. ros 87-90 TNF receptor superfamily member 10b Homo sapiens 99-102 33676890-7 2021 ROS significantly increase the expression of TRAIL death receptor 5 (DR5) via the p53 and C/EBP homologous protein pathways. ros 0-3 TNF receptor superfamily member 10b Homo sapiens 69-72 33385372-1 2021 In the present study, Death receptor-5 (DR5) antibody conjugated solid lipid nanoparticles (DR5-DAPT-SLNs) has been formulated for effective intracellular of gamma-secretase inhibitor, N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) to cancer cells. dapt 185-254 TNF receptor superfamily member 10b Homo sapiens 22-38 33385372-1 2021 In the present study, Death receptor-5 (DR5) antibody conjugated solid lipid nanoparticles (DR5-DAPT-SLNs) has been formulated for effective intracellular of gamma-secretase inhibitor, N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) to cancer cells. dapt 185-254 TNF receptor superfamily member 10b Homo sapiens 40-43 33385372-1 2021 In the present study, Death receptor-5 (DR5) antibody conjugated solid lipid nanoparticles (DR5-DAPT-SLNs) has been formulated for effective intracellular of gamma-secretase inhibitor, N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) to cancer cells. dapt 185-254 TNF receptor superfamily member 10b Homo sapiens 92-95 33385372-1 2021 In the present study, Death receptor-5 (DR5) antibody conjugated solid lipid nanoparticles (DR5-DAPT-SLNs) has been formulated for effective intracellular of gamma-secretase inhibitor, N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) to cancer cells. dapt 96-100 TNF receptor superfamily member 10b Homo sapiens 22-38 33385372-1 2021 In the present study, Death receptor-5 (DR5) antibody conjugated solid lipid nanoparticles (DR5-DAPT-SLNs) has been formulated for effective intracellular of gamma-secretase inhibitor, N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) to cancer cells. dapt 96-100 TNF receptor superfamily member 10b Homo sapiens 40-43 33385372-1 2021 In the present study, Death receptor-5 (DR5) antibody conjugated solid lipid nanoparticles (DR5-DAPT-SLNs) has been formulated for effective intracellular of gamma-secretase inhibitor, N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) to cancer cells. dapt 96-100 TNF receptor superfamily member 10b Homo sapiens 92-95 33385372-6 2021 DR5-DAPT-SLNs have higher cytotoxicity in MDA-MB231 cells compared to DAPT-SLNs (non-targeted) and the bulk drug. dapt 4-8 TNF receptor superfamily member 10b Homo sapiens 0-3 33385372-8 2021 The above results, therefore, demonstrate DR5 mediated uptake is responsible for improved cytotoxicity of DAPT. dapt 106-110 TNF receptor superfamily member 10b Homo sapiens 42-45 33385372-9 2021 In the in vivo anticancer study, DR5-DAPT-SLNs show greater tumor regression when compared to DAPT-SLNs and the bulk drug. dapt 37-41 TNF receptor superfamily member 10b Homo sapiens 33-36 33385372-10 2021 In conclusion, the results of the present study demonstrate that the DR5-DAPT-SLNs selectively target cancer cells and potentiate the anticancer efficacy of DAPT against TNBC cells. dapt 73-77 TNF receptor superfamily member 10b Homo sapiens 69-72 33385372-10 2021 In conclusion, the results of the present study demonstrate that the DR5-DAPT-SLNs selectively target cancer cells and potentiate the anticancer efficacy of DAPT against TNBC cells. dapt 157-161 TNF receptor superfamily member 10b Homo sapiens 69-72 33505114-12 2021 Furthermore, p53 knockdown attenuated the expression of PUMA and DR5 in TRAIL-induced CRC cells treated with encorafenib. encorafenib 109-120 TNF receptor superfamily member 10b Homo sapiens 65-68 33469674-5 2021 Combination treatment with VPA and MSCs-TRAIL enhanced the glioma therapeutic effect by increasing death receptor 5 and caspase activation. Valproic Acid 27-30 TNF receptor superfamily member 10b Homo sapiens 99-115 33092912-0 2021 Corrigendum to "FLLL12 induces apoptosis in lung cancer cells through a p53/p73-independent but death receptor 5-dependent pathway" [Canc. flll12 16-22 TNF receptor superfamily member 10b Homo sapiens 96-112 33505114-10 2021 It was also found that encorafenib exerted its promoting effects on cell apoptosis of CRC cells via the elevation of DR5. encorafenib 23-34 TNF receptor superfamily member 10b Homo sapiens 117-120 33037135-0 2021 Selective tumor cell apoptosis and tumor regression in CDH17-positive colorectal cancer models using BI 905711, a novel liver-sparing TRAILR2 agonist. bi 905711 101-110 TNF receptor superfamily member 10b Homo sapiens 134-141 33037135-3 2021 BI 905711 is a novel tetravalent bispecific antibody targeting both TRAILR2 and CDH17 and represents a novel liver-sparing TRAILR2 agonist specifically designed to overcome the disadvantages of previous strategies. bi 905711 0-9 TNF receptor superfamily member 10b Homo sapiens 68-75 33037135-3 2021 BI 905711 is a novel tetravalent bispecific antibody targeting both TRAILR2 and CDH17 and represents a novel liver-sparing TRAILR2 agonist specifically designed to overcome the disadvantages of previous strategies. bi 905711 0-9 TNF receptor superfamily member 10b Homo sapiens 123-130 33352782-5 2020 The G1-blocker hydroxyurea augmented BS-181-induced apoptosis by enhancing TRAIL/DR4/DR5 upregulation and c-FLIP down-regulation. Hydroxyurea 15-26 TNF receptor superfamily member 10b Homo sapiens 85-88 33296043-5 2020 Binding of 18F-C2Am to apoptotic and necrotic tumor cells was assessed in vitro, and also in vivo, by dynamic PET and biodistribution measurements in mice bearing human tumor xenografts treated with a TRAILR2 agonist or with conventional chemotherapy. 18f-c2am 11-19 TNF receptor superfamily member 10b Homo sapiens 201-208 33007289-4 2020 AMC-04 upregulated the expression of activating transcription factor-4 (ATF4)-C/EBP homologous protein (CHOP) and death receptor 5 (DR5) in cancer cells, as revealed by microarray analysis, small-interference RNA assay, and western blotting. 7-amino-4-methylcoumarin 0-3 TNF receptor superfamily member 10b Homo sapiens 114-130 33007289-4 2020 AMC-04 upregulated the expression of activating transcription factor-4 (ATF4)-C/EBP homologous protein (CHOP) and death receptor 5 (DR5) in cancer cells, as revealed by microarray analysis, small-interference RNA assay, and western blotting. 7-amino-4-methylcoumarin 0-3 TNF receptor superfamily member 10b Homo sapiens 132-135 32200503-7 2020 Knockdown of DR5 abrogated apoptosis induced by the combination of CUDC-907 and TRAIL in breast cancer cells. CUDC-907 67-75 TNF receptor superfamily member 10b Homo sapiens 13-16 32200503-9 2020 JNK inhibitor pretreatment attenuated CUDC-907-induced upregulation of DR5. CUDC-907 38-46 TNF receptor superfamily member 10b Homo sapiens 71-74 32200503-0 2020 CUDC-907 enhances TRAIL-induced apoptosis through upregulation of DR5 in breast cancer cells. CUDC-907 0-8 TNF receptor superfamily member 10b Homo sapiens 66-69 32200503-10 2020 In summary, CUDC-907 shows potent cytotoxicity against breast cancer cells and facilitates TRAIL-mediated apoptosis through DR5 upregulation. CUDC-907 12-20 TNF receptor superfamily member 10b Homo sapiens 124-127 32200503-6 2020 CUDC-907 enhanced expressions of death receptor 5 (DR5), reduced the levels of anti-apoptotic molecules XIAP, Bcl-2 and Bcl-xL. CUDC-907 0-8 TNF receptor superfamily member 10b Homo sapiens 33-49 33180729-6 2020 We show that the action of the herbal drug, Cryptotanshinone on OS cell lines induces apoptosis by increasing sensitivity to TNF-related apoptosis-inducing ligand (TRAIL) through its multi-pronged action on STAT3, DRP1 and DR5. cryptotanshinone 44-60 TNF receptor superfamily member 10b Homo sapiens 223-226 32200503-6 2020 CUDC-907 enhanced expressions of death receptor 5 (DR5), reduced the levels of anti-apoptotic molecules XIAP, Bcl-2 and Bcl-xL. CUDC-907 0-8 TNF receptor superfamily member 10b Homo sapiens 51-54 32777260-0 2020 Luteolin enhances TRAIL sensitivity in non-small cell lung cancer cells through increasing DR5 expression and Drp1-mediated mitochondrial fission. Luteolin 0-8 TNF receptor superfamily member 10b Homo sapiens 91-94 33122623-7 2020 Moreover, DR5 knockdown or knockout attenuated siRALB-induced apoptosis, confirming the role of the extrinsic apoptotic pathway as a regulator of siRALB-induced cell death. siralb 47-53 TNF receptor superfamily member 10b Homo sapiens 10-13 32433558-5 2020 In contrast, oxygen, and nutrient deprivation promote high amounts of TRAILR2 expression in TRAIL-hypersensitive cells in inner spheroid layers. Oxygen 13-19 TNF receptor superfamily member 10b Homo sapiens 70-77 32433558-6 2020 COX-II inhibitor celecoxib further enhanced TRAILR2 expression in spheroids, likely resulting from increased ER stress, and thereby re-sensitized TRAIL-resistant cell layers to treatment. Celecoxib 17-26 TNF receptor superfamily member 10b Homo sapiens 44-51 32777260-8 2020 Moreover, luteolin treatment alone or in combination with TRAIL increased the phosphorylation of c-Jun N-terminal kinase (JNK), while SP600125 (the JNK inhibitor) significantly abolished the synergistic effect on DR5 expression and Drp1 translocation, indicating that JNK signaling activation was greatly associated with the synergistic effect exerted by luteolin in NSCLC cells. Luteolin 10-18 TNF receptor superfamily member 10b Homo sapiens 213-216 32777260-8 2020 Moreover, luteolin treatment alone or in combination with TRAIL increased the phosphorylation of c-Jun N-terminal kinase (JNK), while SP600125 (the JNK inhibitor) significantly abolished the synergistic effect on DR5 expression and Drp1 translocation, indicating that JNK signaling activation was greatly associated with the synergistic effect exerted by luteolin in NSCLC cells. pyrazolanthrone 134-142 TNF receptor superfamily member 10b Homo sapiens 213-216 33050333-0 2020 Dexamethasone Inhibits TRAIL-Induced Apoptosis through c-FLIP(L) Upregulation and DR5 Downregulation by GSK3beta Activation in Cancer Cells. Dexamethasone 0-13 TNF receptor superfamily member 10b Homo sapiens 82-85 33240775-4 2020 Mechanistically, curcumol directly targets NQO2 to cause reactive oxygen species (ROS) generation, which triggers endoplasmic reticulum (ER) stress-C/EBP homologous protein (CHOP) death receptor (DR5) signaling, sensitizing NSCLC cell to TRAIL-induced apoptosis. Reactive Oxygen Species 82-85 TNF receptor superfamily member 10b Homo sapiens 196-199 33240775-4 2020 Mechanistically, curcumol directly targets NQO2 to cause reactive oxygen species (ROS) generation, which triggers endoplasmic reticulum (ER) stress-C/EBP homologous protein (CHOP) death receptor (DR5) signaling, sensitizing NSCLC cell to TRAIL-induced apoptosis. curcumol 17-25 TNF receptor superfamily member 10b Homo sapiens 196-199 33050333-3 2020 We found that upregulation of c-FLIP(L) and downregulation of death receptor 5 (DR5; receptor for TRAIL ligand) contribute to the anti-apoptotic effect of DEX on TRAIL-induced apoptosis. Dexamethasone 155-158 TNF receptor superfamily member 10b Homo sapiens 62-78 33050333-3 2020 We found that upregulation of c-FLIP(L) and downregulation of death receptor 5 (DR5; receptor for TRAIL ligand) contribute to the anti-apoptotic effect of DEX on TRAIL-induced apoptosis. Dexamethasone 155-158 TNF receptor superfamily member 10b Homo sapiens 80-83 33050333-7 2020 In addition, DEX decreased protein stability of DR5 via GSK-3beta-mediated upregulation of Cbl, an E3 ligase of DR5. Dexamethasone 13-16 TNF receptor superfamily member 10b Homo sapiens 48-51 33050333-7 2020 In addition, DEX decreased protein stability of DR5 via GSK-3beta-mediated upregulation of Cbl, an E3 ligase of DR5. Dexamethasone 13-16 TNF receptor superfamily member 10b Homo sapiens 112-115 33050333-8 2020 Knockdown of Cbl by siRNA markedly inhibited DEX-induced DR5 downregulation. Dexamethasone 45-48 TNF receptor superfamily member 10b Homo sapiens 57-60 33050333-9 2020 Taken together, these results suggest that DEX inhibits TRAIL-mediated apoptosis via GSK-3beta-mediated DR5 downregulation and c-FLIP(L) upregulation in cancer cells. Dexamethasone 43-46 TNF receptor superfamily member 10b Homo sapiens 104-107 33050112-0 2020 Magnolol Enhances the Therapeutic Effects of TRAIL through DR5 Upregulation and Downregulation of c-FLIP and Mcl-1 Proteins in Cancer Cells. magnolol 0-8 TNF receptor superfamily member 10b Homo sapiens 59-62 33050112-2 2020 We found that magnolol sensitizes TRAIL-induced apoptotic cell death via upregulation of DR5 and downregulation of cellular FLICE-inhibitory protein (c-FLIP) and Mcl-1 in cancer cells, but not in normal cells. magnolol 14-22 TNF receptor superfamily member 10b Homo sapiens 89-92 32941010-1 2020 Fatty acid-induced upregulation of death receptor 5 (DR5) and its cognate ligand, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), promotes hepatocyte lipoapoptosis, which is a key mechanism in the progression of fatty liver disease. Fatty Acids 0-10 TNF receptor superfamily member 10b Homo sapiens 35-51 33050112-3 2020 Mechanistically, magnolol increased ATF4-dependent DR5 expression at the transcription level, and knockdown of ATF4 markedly inhibited magnolol-induced DR5 upregulation. magnolol 17-25 TNF receptor superfamily member 10b Homo sapiens 51-54 32941010-1 2020 Fatty acid-induced upregulation of death receptor 5 (DR5) and its cognate ligand, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), promotes hepatocyte lipoapoptosis, which is a key mechanism in the progression of fatty liver disease. Fatty Acids 0-10 TNF receptor superfamily member 10b Homo sapiens 53-56 33050112-3 2020 Mechanistically, magnolol increased ATF4-dependent DR5 expression at the transcription level, and knockdown of ATF4 markedly inhibited magnolol-induced DR5 upregulation. magnolol 135-143 TNF receptor superfamily member 10b Homo sapiens 152-155 33050112-4 2020 Silencing DR5 with siRNA prevented combined treatment with magnolol and TRAIL-induced apoptosis and PARP cleavage. magnolol 59-67 TNF receptor superfamily member 10b Homo sapiens 10-13 33050112-6 2020 Our results revealed that magnolol enhanced TRAIL-induced apoptosis via ATF4-dependent DR5 upregulation and downregulation of c-FLIP and Mcl-1 proteins. magnolol 26-34 TNF receptor superfamily member 10b Homo sapiens 87-90 32941010-4 2020 To this end, we used yeast display and a designed combinatorial library to identify a synthetic 58-amino acid affibody ligand that specifically binds the DR5. 58-amino acid 96-109 TNF receptor superfamily member 10b Homo sapiens 154-157 33023194-0 2020 Selinexor Sensitizes TRAIL-R2-Positive TNBC Cells to the Activity of TRAIL-R2xCD3 Bispecific Antibody. selinexor 0-9 TNF receptor superfamily member 10b Homo sapiens 21-29 32692906-7 2020 These results represent the first mechanistic insight into how O-glycan structures on cell surface modulate their sensitivity to apoptotic stimuli, suggesting expression of Tn/STn may offer tumor cell survival advantages through altering DR4 and/or DR5 activity. o-glycan 63-71 TNF receptor superfamily member 10b Homo sapiens 249-252 32899699-4 2020 ATS/DHA induces the expression of DR5 in a P53 dependent manner in HCT116 and DLD-1 cells. artenimol 4-7 TNF receptor superfamily member 10b Homo sapiens 34-37 32899699-5 2020 Both ATS and DHA overcome the resistance to DHER-induced apoptosis in HCT116, mainly through upregulating death receptor 5 (DR5). artenimol 13-16 TNF receptor superfamily member 10b Homo sapiens 106-122 32899699-5 2020 Both ATS and DHA overcome the resistance to DHER-induced apoptosis in HCT116, mainly through upregulating death receptor 5 (DR5). artenimol 13-16 TNF receptor superfamily member 10b Homo sapiens 124-127 32899699-5 2020 Both ATS and DHA overcome the resistance to DHER-induced apoptosis in HCT116, mainly through upregulating death receptor 5 (DR5). dher 44-48 TNF receptor superfamily member 10b Homo sapiens 106-122 32899699-5 2020 Both ATS and DHA overcome the resistance to DHER-induced apoptosis in HCT116, mainly through upregulating death receptor 5 (DR5). dher 44-48 TNF receptor superfamily member 10b Homo sapiens 124-127 32899699-6 2020 We also demonstrate that DHA sensitizes HCT116 cells to DHER-induced apoptosis via P53 regulated DR5 expression in P53 knockdown assays. artenimol 25-28 TNF receptor superfamily member 10b Homo sapiens 97-100 32899699-6 2020 We also demonstrate that DHA sensitizes HCT116 cells to DHER-induced apoptosis via P53 regulated DR5 expression in P53 knockdown assays. dher 56-60 TNF receptor superfamily member 10b Homo sapiens 97-100 32506763-0 2020 ROS-mediated PERK-eIF2alpha-ATF4 pathway plays an important role in arsenite-induced L-02 cells apoptosis via regulating CHOP-DR5 signaling. Reactive Oxygen Species 0-3 TNF receptor superfamily member 10b Homo sapiens 126-129 32506763-0 2020 ROS-mediated PERK-eIF2alpha-ATF4 pathway plays an important role in arsenite-induced L-02 cells apoptosis via regulating CHOP-DR5 signaling. arsenite 68-76 TNF receptor superfamily member 10b Homo sapiens 126-129 32506763-5 2020 Results showed that death receptor 5 (DR5) was a mediator of NaAsO2 -induced apoptosis by enhancing construction of the death-inducing signaling complex (DISC). sodium arsenite 61-67 TNF receptor superfamily member 10b Homo sapiens 20-36 32506763-5 2020 Results showed that death receptor 5 (DR5) was a mediator of NaAsO2 -induced apoptosis by enhancing construction of the death-inducing signaling complex (DISC). sodium arsenite 61-67 TNF receptor superfamily member 10b Homo sapiens 38-41 32506763-8 2020 PERK inhibitor and ATF4 siRNA significantly attenuated NaAsO2 -induced CHOP and DR5 expressions. sodium arsenite 55-61 TNF receptor superfamily member 10b Homo sapiens 80-83 32506763-10 2020 Taken together, the results indicate that ROS-mediated PERK-eIF2alpha-ATF4 pathway activated by NaAsO2 is the critical upstream event for subsequent apoptosis induction via regulating CHOP-DR5 signaling in L-02 cells when chronic exposure to arsenic, and support that antioxidants might be potential therapeutic agents for preventing or delaying the onset and progress of arsenic-induced hepatotoxicity. Reactive Oxygen Species 42-45 TNF receptor superfamily member 10b Homo sapiens 189-192 32506763-10 2020 Taken together, the results indicate that ROS-mediated PERK-eIF2alpha-ATF4 pathway activated by NaAsO2 is the critical upstream event for subsequent apoptosis induction via regulating CHOP-DR5 signaling in L-02 cells when chronic exposure to arsenic, and support that antioxidants might be potential therapeutic agents for preventing or delaying the onset and progress of arsenic-induced hepatotoxicity. sodium arsenite 96-102 TNF receptor superfamily member 10b Homo sapiens 189-192 32974652-3 2020 In this work, we present structural insights for the recognition of DR5 and DR0 elements by RXR-RAR heterodimer using x-ray crystallography, small angle x-ray scattering, and hydrogen/deuterium exchange coupled to mass spectrometry. Hydrogen 175-183 TNF receptor superfamily member 10b Homo sapiens 68-71 32974652-3 2020 In this work, we present structural insights for the recognition of DR5 and DR0 elements by RXR-RAR heterodimer using x-ray crystallography, small angle x-ray scattering, and hydrogen/deuterium exchange coupled to mass spectrometry. Deuterium 184-193 TNF receptor superfamily member 10b Homo sapiens 68-71 32899699-0 2020 Artemisinin Derivatives Stimulate DR5-Specific TRAIL-Induced Apoptosis by Regulating Wildtype P53. artemisinin 0-11 TNF receptor superfamily member 10b Homo sapiens 34-37 32878791-0 2020 Up-regulation of Death Receptor 5/TRAIL-R2 Mediates Apoptosis Induced by N,N"-[(3,4-dimethoxyphenyl)methylene] Biscinnamide in Cancer Cells. n,n"-[(3,4-dimethoxyphenyl)methylene] biscinnamide 73-123 TNF receptor superfamily member 10b Homo sapiens 17-33 32878791-0 2020 Up-regulation of Death Receptor 5/TRAIL-R2 Mediates Apoptosis Induced by N,N"-[(3,4-dimethoxyphenyl)methylene] Biscinnamide in Cancer Cells. n,n"-[(3,4-dimethoxyphenyl)methylene] biscinnamide 73-123 TNF receptor superfamily member 10b Homo sapiens 34-42 32878791-8 2020 RESULTS: DPMBC up-regulated expression levels of DR5 protein and induced apoptosis through the extrinsic apoptotic pathway mediated by DR5 and caspases. dpmbc 9-14 TNF receptor superfamily member 10b Homo sapiens 49-52 32878791-8 2020 RESULTS: DPMBC up-regulated expression levels of DR5 protein and induced apoptosis through the extrinsic apoptotic pathway mediated by DR5 and caspases. dpmbc 9-14 TNF receptor superfamily member 10b Homo sapiens 135-138 32141025-6 2020 RESULTS: Curcumin augments TRAIL-apoptotic signaling in leukemic cells by upregulating the expression of DR4 and DR5 along with suppression of cFLIP and anti-apoptotic proteins Mcl-1, Bcl-xl, and XIAP. Curcumin 9-17 TNF receptor superfamily member 10b Homo sapiens 113-116 32825566-3 2020 In this study, we showed the sub-lethal concentrations of a lysosomotropic autophagy inhibitor, IITZ-01, sensitizes cancer cells (renal, lung, and breast carcinoma) to TRAIL-induced apoptosis through DR5 upregulation and survivin downregulation through ubiquitin-proteasome pathway. IITZ-01 96-103 TNF receptor superfamily member 10b Homo sapiens 200-203 32825566-4 2020 Knockdown of DR5 or overexpression of survivin inhibited combined treatment with IITZ-01 and TRAIL-induced apoptosis. IITZ-01 81-88 TNF receptor superfamily member 10b Homo sapiens 13-16 32825566-7 2020 Taken together, these results provide the first evidence that IITZ-01 enhances TRAIL-mediated apoptosis through DR5 stabilization by downregulation of Cbl and USP9X-dependent survivin ubiquitination and degradation in renal carcinoma cells. IITZ-01 62-69 TNF receptor superfamily member 10b Homo sapiens 112-115 32626921-0 2020 Inhibition of autophagy flux by sertraline attenuates TRAIL resistance in lung cancer via death receptor 5 upregulation. Sertraline 32-42 TNF receptor superfamily member 10b Homo sapiens 90-106 32626921-5 2020 To the best of our knowledge, this is the first study to demonstrate that sertraline inhibits autophagic flux and increases the expression of death receptor 5 (DR5) on TRAIL-resistant lung cancer cells. Sertraline 74-84 TNF receptor superfamily member 10b Homo sapiens 142-158 32626921-5 2020 To the best of our knowledge, this is the first study to demonstrate that sertraline inhibits autophagic flux and increases the expression of death receptor 5 (DR5) on TRAIL-resistant lung cancer cells. Sertraline 74-84 TNF receptor superfamily member 10b Homo sapiens 160-163 32626921-6 2020 Inhibition of autophagy using autophagy inhibitors 3-methyladenine and chloroquine upregulated the expression of DR5 and enhanced TRAIL-induced apoptosis, as confirmed by the increase of pro-apoptotic proteins caspase-8 and caspase-3. 3-methyladenine 51-66 TNF receptor superfamily member 10b Homo sapiens 113-116 32626921-6 2020 Inhibition of autophagy using autophagy inhibitors 3-methyladenine and chloroquine upregulated the expression of DR5 and enhanced TRAIL-induced apoptosis, as confirmed by the increase of pro-apoptotic proteins caspase-8 and caspase-3. Chloroquine 71-82 TNF receptor superfamily member 10b Homo sapiens 113-116 32626921-7 2020 Silencing DR5 expression using DR5 small interfering RNA prevented sertraline-induced TRAIL-mediated apoptosis, indicating the role of DR5 in TRAIL-mediated apoptosis. Sertraline 67-77 TNF receptor superfamily member 10b Homo sapiens 10-13 32626921-7 2020 Silencing DR5 expression using DR5 small interfering RNA prevented sertraline-induced TRAIL-mediated apoptosis, indicating the role of DR5 in TRAIL-mediated apoptosis. Sertraline 67-77 TNF receptor superfamily member 10b Homo sapiens 31-34 32626921-7 2020 Silencing DR5 expression using DR5 small interfering RNA prevented sertraline-induced TRAIL-mediated apoptosis, indicating the role of DR5 in TRAIL-mediated apoptosis. Sertraline 67-77 TNF receptor superfamily member 10b Homo sapiens 31-34 32626921-8 2020 Overall, sertraline enhanced TRAIL-mediated apoptosis via the downregulation of AMP-activated protein kinase phosphorylation, resulting in the inhibition of autophagic flux, upregulation of DR5 expression, and activation of the apoptotic caspase cascade. Sertraline 9-19 TNF receptor superfamily member 10b Homo sapiens 190-193 32722598-7 2020 Treatment of cancer cell lines with the antioxidant N-acetylcysteine reduces the extent of membrane dysfunction and the expression of both CHOP-DR5 and miR-425-PTEN axes, attenuating PAM/TRAIL-induced cancer cell apoptosis. Acetylcysteine 52-68 TNF receptor superfamily member 10b Homo sapiens 144-147 32416217-8 2020 Moreover, DT-13 induced the death receptor pathway-dependent apoptosis of HL-60 and Kasumi-1 cells by up-regulating Fas, FasL, DR5 and TRAIL as well as promoted the cleavage of caspase 8, caspase 3 and PARP. DT-13 10-15 TNF receptor superfamily member 10b Homo sapiens 127-130 32303681-4 2020 Here, we found that TNFRSF10B, along with a vesicular trafficking regulator protein, YIPF2, were upregulated after treatment with pemetrexed (PEM) in NSCLC cells. Pemetrexed 130-140 TNF receptor superfamily member 10b Homo sapiens 20-29 32714568-5 2020 Depletion of the anti-apoptotic protein FLIP, which we identify as a novel mediator of resistance to MLN4924, enhanced apoptosis in a p53-, TRAIL-R2/DR5-, and caspase-8-dependent manner. pevonedistat 101-108 TNF receptor superfamily member 10b Homo sapiens 140-148 32714568-5 2020 Depletion of the anti-apoptotic protein FLIP, which we identify as a novel mediator of resistance to MLN4924, enhanced apoptosis in a p53-, TRAIL-R2/DR5-, and caspase-8-dependent manner. pevonedistat 101-108 TNF receptor superfamily member 10b Homo sapiens 149-152 32714568-6 2020 Notably, TRAIL-R2 was involved in potentiating the apoptotic response to MLN4924 in the absence of FLIP, in a ligand-independent manner. pevonedistat 73-80 TNF receptor superfamily member 10b Homo sapiens 9-17 32319535-8 2020 Overall, the results of the present study revealed that cantharidin effectively sensitized cells to TRAIL-mediated apoptosis and its effects are likely to be mediated by autophagy, the downregulation of c-FLIP and the upregulation of DR-5. Cantharidin 56-67 TNF receptor superfamily member 10b Homo sapiens 234-238 32441887-0 2020 Lestaurtinib potentiates TRAIL-induced apoptosis in glioma via CHOP-dependent DR5 induction. lestaurtinib 0-12 TNF receptor superfamily member 10b Homo sapiens 78-81 32441887-5 2020 While U87 and U251 cells showed resistance to TRAIL single treatment, they were sensitized to apoptosis induced by TRAIL in the presence of lestaurtinib because of increased death receptor 5 (DR5) levels through CHOP-dependent manner. lestaurtinib 140-152 TNF receptor superfamily member 10b Homo sapiens 174-190 32441887-5 2020 While U87 and U251 cells showed resistance to TRAIL single treatment, they were sensitized to apoptosis induced by TRAIL in the presence of lestaurtinib because of increased death receptor 5 (DR5) levels through CHOP-dependent manner. lestaurtinib 140-152 TNF receptor superfamily member 10b Homo sapiens 192-195 32559583-11 2020 Moreover, periplocin increased the mRNA expression of NOXA, Bak, Bcl-2, and death receptors such as TRAIL-R1 and TRAIL-R2 and the protein expression of ERK/phospho ERK, p38/phospho p38, and JNK/phospho JNK. periplocin 10-20 TNF receptor superfamily member 10b Homo sapiens 113-121 31955141-0 2020 Curcumin-loaded nanostructured lipid carrier induced apoptosis in human HepG2 cells through activation of the DR5/caspase-mediated extrinsic apoptosis pathway. Curcumin 0-8 TNF receptor superfamily member 10b Homo sapiens 110-113 31955141-5 2020 This study showed that Cur-NLC significantly increased total expression of DR5 protein while simultaneously upregulated cell membrane expression of DR5. Cur1 compound 23-26 TNF receptor superfamily member 10b Homo sapiens 75-78 31955141-5 2020 This study showed that Cur-NLC significantly increased total expression of DR5 protein while simultaneously upregulated cell membrane expression of DR5. Cur1 compound 23-26 TNF receptor superfamily member 10b Homo sapiens 148-151 31955141-8 2020 Therefore, Cur-NLC induced activation of the extrinsic apoptosis pathway via modulating the DR5/caspase-8/-3 mediated apoptosis pathway in HepG2 cells, suggesting that Cur-NLC is a promising therapeutic agent or supplement for the treatment of hepatocellular carcinoma. Cur1 compound 11-14 TNF receptor superfamily member 10b Homo sapiens 92-95 31955141-8 2020 Therefore, Cur-NLC induced activation of the extrinsic apoptosis pathway via modulating the DR5/caspase-8/-3 mediated apoptosis pathway in HepG2 cells, suggesting that Cur-NLC is a promising therapeutic agent or supplement for the treatment of hepatocellular carcinoma. Cur1 compound 168-171 TNF receptor superfamily member 10b Homo sapiens 92-95 32156787-6 2020 Therapeutic importin beta1 knockdown, administered via the atelocollagen delivery system, as well as treatment with the importin beta inhibitor, importazole, induced regression and/or eradication of two human TRAIL-resistant tumor cells when combined with agonistic anti-hDR5 antibody treatment. importazole 145-156 TNF receptor superfamily member 10b Homo sapiens 271-275 32365976-5 2020 Bortezomib and doxorubicin greatly enhanced the surface expression of the death receptors DR5 and DR4, while panobinostat increased expression of DR5 and suppressed expression of DR4 in both cell lines. Bortezomib 0-10 TNF receptor superfamily member 10b Homo sapiens 90-93 32365976-5 2020 Bortezomib and doxorubicin greatly enhanced the surface expression of the death receptors DR5 and DR4, while panobinostat increased expression of DR5 and suppressed expression of DR4 in both cell lines. Doxorubicin 15-26 TNF receptor superfamily member 10b Homo sapiens 90-93 32319535-0 2020 Downregulation of c-FLIP and upregulation of DR-5 by cantharidin sensitizes TRAIL-mediated apoptosis in prostate cancer cells via autophagy flux. Cantharidin 53-64 TNF receptor superfamily member 10b Homo sapiens 45-49 32319535-5 2020 Notably, it was also identified that cantharidin treatment initiated the downregulation of cellular FLICE-like inhibitory protein (c-FLIP) and upregulation of death receptor 5 (DR-5), and sensitized cells to TRAIL-mediated apoptosis by initiating autophagy flux. Cantharidin 37-48 TNF receptor superfamily member 10b Homo sapiens 159-175 32319535-5 2020 Notably, it was also identified that cantharidin treatment initiated the downregulation of cellular FLICE-like inhibitory protein (c-FLIP) and upregulation of death receptor 5 (DR-5), and sensitized cells to TRAIL-mediated apoptosis by initiating autophagy flux. Cantharidin 37-48 TNF receptor superfamily member 10b Homo sapiens 177-181 32509779-8 2020 This results in a strongly reduced number of apoptotic cells upon treatment with DR5-specific TRAIL variant DHER in CM. dher 108-112 TNF receptor superfamily member 10b Homo sapiens 81-84 32375399-8 2020 Dinaciclib was shown to drastically enhance EV-T killing effects on cancer lines that express good levels of death receptor (DR) 5, which are associated with suppression of CDK1, CDK9 and anti-apoptotic proteins. dinaciclib 0-10 TNF receptor superfamily member 10b Homo sapiens 109-130 32303681-4 2020 Here, we found that TNFRSF10B, along with a vesicular trafficking regulator protein, YIPF2, were upregulated after treatment with pemetrexed (PEM) in NSCLC cells. Pemetrexed 142-145 TNF receptor superfamily member 10b Homo sapiens 20-29 31812766-5 2020 Moreover, formaldehyde significantly potentiated the induction of death receptor-5, caspase 8/10, cleaved caspase 3/7/9, pro-apoptotic proteins (Bim, Bad and Bax), depolarization of MMP (mitochondrial membrane potential) and AIF (apoptosis-inducing factor) induced by acrolein, and synergistically decreased expressions of pro-survival proteins (Bcl-2 and Bcl-XL) and poly ADP-ribose polymerase. Formaldehyde 10-22 TNF receptor superfamily member 10b Homo sapiens 66-82 31923762-0 2020 Oxidative stress-driven DR5 upregulation restores TRAIL/Apo2L sensitivity induced by iron oxide nanoparticles in colorectal cancer. ferric oxide 85-95 TNF receptor superfamily member 10b Homo sapiens 24-27 32213959-6 2020 While PGA2 induced apoptosis in HCT116 cells, phosphorylation of p53 and transcriptional induction of p53-target genes such as p21WAF1, PUMA, BAX, NOXA, and DR5 occurred. prostaglandin A2 6-10 TNF receptor superfamily member 10b Homo sapiens 157-160 32213959-9 2020 Moreover, among target genes of p53, knockdown of DR5 expression by RNA interference, suppressed PGA2-induced apoptosis. prostaglandin A2 97-101 TNF receptor superfamily member 10b Homo sapiens 50-53 32213959-13 2020 PGA2 may induce p53-dependent apoptosis in which DNA-PK activates p53, and DR5, a transcriptional target of p53, plays a pivotal role in HCT116 cells. prostaglandin A2 0-4 TNF receptor superfamily member 10b Homo sapiens 75-78 32266096-11 2020 DR5 is one of the upstream receptors of the JNK pathway, and shRNA knockdown of DR5 can partially reverse gossypol-induced apoptosis. Gossypol 106-114 TNF receptor superfamily member 10b Homo sapiens 0-3 32266096-11 2020 DR5 is one of the upstream receptors of the JNK pathway, and shRNA knockdown of DR5 can partially reverse gossypol-induced apoptosis. Gossypol 106-114 TNF receptor superfamily member 10b Homo sapiens 80-83 31923762-3 2020 Specifically, NanoTRAIL (TRAIL/Apo2L-iron oxide nanoparticles) generated ROS (reactive oxygen species)-triggered JNK (c-Jun N-terminal kinase) activation and induced subsequent autophagy-assisted DR5 upregulation, resulting in a significant enhanced antitumor efficacy of TRAIL/Apo2L, which confirmed in both TRAIL-resistant HT-29, intermediately resistant SW-480 and sensitive HCT-116 cells. ferric oxide 37-47 TNF receptor superfamily member 10b Homo sapiens 196-199 31923762-3 2020 Specifically, NanoTRAIL (TRAIL/Apo2L-iron oxide nanoparticles) generated ROS (reactive oxygen species)-triggered JNK (c-Jun N-terminal kinase) activation and induced subsequent autophagy-assisted DR5 upregulation, resulting in a significant enhanced antitumor efficacy of TRAIL/Apo2L, which confirmed in both TRAIL-resistant HT-29, intermediately resistant SW-480 and sensitive HCT-116 cells. Oxygen 87-93 TNF receptor superfamily member 10b Homo sapiens 196-199 32124960-13 2020 Overall, the present study provided evidence that ICA sensitized tumor cells to TRAIL-induced apoptosis via ROS-, ERK- and CHOP-mediated upregulation of DR5 and DR4. icariin 50-53 TNF receptor superfamily member 10b Homo sapiens 153-156 32124960-13 2020 Overall, the present study provided evidence that ICA sensitized tumor cells to TRAIL-induced apoptosis via ROS-, ERK- and CHOP-mediated upregulation of DR5 and DR4. Reactive Oxygen Species 108-111 TNF receptor superfamily member 10b Homo sapiens 153-156 31641820-8 2020 Our study concluded that P5H induced the death receptor, DR5 in HCT-116 and mitochondria-mediated apoptosis pathway in HT-29. Hydrogen 25-28 TNF receptor superfamily member 10b Homo sapiens 57-60 31839995-11 2019 Our results indicate that DDAs are unique in causing DR5 accumulation and oligomerization and inducing downstream caspase activation and cancer cell death through mechanisms involving altered DR5 disulfide bonding. Disulfides 196-205 TNF receptor superfamily member 10b Homo sapiens 192-195 31806377-5 2020 Interestingly, we observed that the addition of four different autophagy inducers, rapamycin (RAP), CCI779, ABT737 and temozolomide (TMZ), induced the differences of DR5 expression and cell apoptosis after irradiation. Sirolimus 83-92 TNF receptor superfamily member 10b Homo sapiens 166-169 31806377-5 2020 Interestingly, we observed that the addition of four different autophagy inducers, rapamycin (RAP), CCI779, ABT737 and temozolomide (TMZ), induced the differences of DR5 expression and cell apoptosis after irradiation. Sirolimus 94-97 TNF receptor superfamily member 10b Homo sapiens 166-169 31806377-5 2020 Interestingly, we observed that the addition of four different autophagy inducers, rapamycin (RAP), CCI779, ABT737 and temozolomide (TMZ), induced the differences of DR5 expression and cell apoptosis after irradiation. ABT-737 108-114 TNF receptor superfamily member 10b Homo sapiens 166-169 31806377-5 2020 Interestingly, we observed that the addition of four different autophagy inducers, rapamycin (RAP), CCI779, ABT737 and temozolomide (TMZ), induced the differences of DR5 expression and cell apoptosis after irradiation. temozolomide 119-131 TNF receptor superfamily member 10b Homo sapiens 166-169 31806377-5 2020 Interestingly, we observed that the addition of four different autophagy inducers, rapamycin (RAP), CCI779, ABT737 and temozolomide (TMZ), induced the differences of DR5 expression and cell apoptosis after irradiation. temozolomide 133-136 TNF receptor superfamily member 10b Homo sapiens 166-169 31806377-6 2020 Unlike RAP and CCI779, ABT737 and TMZ were able to increase DR5 expression and further induce cell death. ABT-737 23-29 TNF receptor superfamily member 10b Homo sapiens 60-63 31806377-6 2020 Unlike RAP and CCI779, ABT737 and TMZ were able to increase DR5 expression and further induce cell death. temozolomide 34-37 TNF receptor superfamily member 10b Homo sapiens 60-63 31698509-9 2020 Ecn induced ROS production and GRP78, CHOP, DR5 and DR4 expression as well as depolarized the mitochondria membrane potential. ecn 0-3 TNF receptor superfamily member 10b Homo sapiens 44-47 32083004-8 2020 In addition, ZBTB7A transactivates TRAIL-R2, which sensitizes cells to cisplatin-induced apoptosis. Cisplatin 71-80 TNF receptor superfamily member 10b Homo sapiens 35-43 32083004-9 2020 The ZBTB7A-TRAIL-R2 cascade is involved in both the extrinsic and intrinsic cisplatin-induced pathways of apoptosis. Cisplatin 76-85 TNF receptor superfamily member 10b Homo sapiens 11-19 32187278-8 2020 CASPASE-8 and TNFRSF10B expression levels could predict the response of both the cell lines to rhTRAIL alone or the response to a combination of rhTRAIL and cisplatin. Cisplatin 157-166 TNF receptor superfamily member 10b Homo sapiens 14-23 31294850-7 2020 Our modelization results show that TRAIL can bind to DR4 and DR5 when transported by graphene nanoflake, as a proof of concept. Graphite 85-93 TNF receptor superfamily member 10b Homo sapiens 61-64 31839995-5 2019 At the protein level, DDAs alter DR5 disulfide bonding to increase steady-state DR5 levels and oligomerization, leading to downstream caspase 8 and 3 activation. Disulfides 37-46 TNF receptor superfamily member 10b Homo sapiens 33-36 31839995-5 2019 At the protein level, DDAs alter DR5 disulfide bonding to increase steady-state DR5 levels and oligomerization, leading to downstream caspase 8 and 3 activation. Disulfides 37-46 TNF receptor superfamily member 10b Homo sapiens 80-83 31641820-0 2020 Activation of death receptor, DR5 and mitochondria-mediated apoptosis by a 3,4,5-trimethoxybenzyloxy derivative in wild-type and p53 mutant colorectal cancer cell lines. BW 1069C85 75-100 TNF receptor superfamily member 10b Homo sapiens 30-33 31987044-0 2020 Cell death induced by the ER stressor thapsigargin involves death receptor 5, a non-autophagic function of MAP1LC3B, and distinct contributions from unfolded protein response components. Thapsigargin 38-50 TNF receptor superfamily member 10b Homo sapiens 60-76 31987044-7 2020 RESULTS: In both cell lines, Tg-induced cell death depended on death receptor 5 and caspase-8. Thapsigargin 29-31 TNF receptor superfamily member 10b Homo sapiens 63-79 31987044-9 2020 PERK, ATF4 and CHOP were required for Tg-induced cell death, but surprisingly acted in parallel rather than as a linear pathway; ATF4 and CHOP were independently required for Tg-mediated upregulation of death receptor 5 and MAP1LC3B proteins, whereas PERK acted via other pathways. Thapsigargin 38-40 TNF receptor superfamily member 10b Homo sapiens 203-219 31987044-9 2020 PERK, ATF4 and CHOP were required for Tg-induced cell death, but surprisingly acted in parallel rather than as a linear pathway; ATF4 and CHOP were independently required for Tg-mediated upregulation of death receptor 5 and MAP1LC3B proteins, whereas PERK acted via other pathways. Thapsigargin 175-177 TNF receptor superfamily member 10b Homo sapiens 203-219 31306050-4 2020 In particular, AMP induced caspase-8 dependent apoptosis via upregulation of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and death receptor (DR5). ampelopsin 15-18 TNF receptor superfamily member 10b Homo sapiens 161-164 31306050-5 2020 Knockdown of DR5 by RNA interference blocked AMP-induced apoptosis. ampelopsin 45-48 TNF receptor superfamily member 10b Homo sapiens 13-16 31306050-8 2020 These results demonstrate that treatment with AMP induces the apoptosis of EBV-positive cells through upregulation of TRAIL/DR5 and activation of p38 signaling. ampelopsin 46-49 TNF receptor superfamily member 10b Homo sapiens 124-127 31206933-0 2019 TRAIL-inspired multivalent dextran conjugates efficiently induce apoptosis upon DR5 receptor clustering. Dextrans 27-34 TNF receptor superfamily member 10b Homo sapiens 80-83 31206933-5 2019 Enzyme-catalyzed ligation of a hydrophilic dextran conjugate bearing multiple DR5-targeting sites to a human Fc did not affect potency (EC50 = 2 - 7 nM), providing an option for improved in vivo half-life and prospective conjugation to an antibody of interest in view of bispecific tumor targeting. Dextrans 43-50 TNF receptor superfamily member 10b Homo sapiens 78-81 31817791-7 2019 In addition, BITC increased death receptor DR5 expression, and activated the cysteine-aspartic proteases (caspases) cascade. benzyl isothiocyanate 13-17 TNF receptor superfamily member 10b Homo sapiens 43-46 33304996-4 2019 We report here on a peptide amphiphile (PA) containing a dimeric, cyclic peptide that self-assembles into cylindrical supramolecular nanofibers and targets DR5. Peptides, Cyclic 66-80 TNF receptor superfamily member 10b Homo sapiens 156-159 31004033-7 2019 RG7388 induced a pro-apoptotic gene expression signature with upregulation of p53-target genes involved in the intrinsic (PUMA, BAX) and extrinsic (TNFRSF10B, FAS) pathways of apoptosis, as well as MDM2 Only a slight induction of CDKN1A was observed and upregulation of pro-apoptotic genes dominated, indicating that CLL cells are primed for p53-dependent apoptosis. RG7388 0-6 TNF receptor superfamily member 10b Homo sapiens 148-157 33304996-5 2019 Coassembly of the DR5-targeting PA and a pegylated PA creates a supramolecular nanoscale construct with enhanced binding affinity to DR5 relative to a monomeric targeting PA, and was found to be cytotoxic in vitro. Protactinium 32-34 TNF receptor superfamily member 10b Homo sapiens 18-21 33304996-5 2019 Coassembly of the DR5-targeting PA and a pegylated PA creates a supramolecular nanoscale construct with enhanced binding affinity to DR5 relative to a monomeric targeting PA, and was found to be cytotoxic in vitro. Protactinium 32-34 TNF receptor superfamily member 10b Homo sapiens 133-136 33304996-5 2019 Coassembly of the DR5-targeting PA and a pegylated PA creates a supramolecular nanoscale construct with enhanced binding affinity to DR5 relative to a monomeric targeting PA, and was found to be cytotoxic in vitro. Protactinium 51-53 TNF receptor superfamily member 10b Homo sapiens 133-136 33304996-5 2019 Coassembly of the DR5-targeting PA and a pegylated PA creates a supramolecular nanoscale construct with enhanced binding affinity to DR5 relative to a monomeric targeting PA, and was found to be cytotoxic in vitro. Protactinium 51-53 TNF receptor superfamily member 10b Homo sapiens 133-136 33304996-6 2019 When combined with the chemotherapy paclitaxel, DR5-targeting carriers showed potent antitumor activity in vivo, demonstrating the multifunctional capabilities of peptide-based supramolecular nanostructures. Paclitaxel 36-46 TNF receptor superfamily member 10b Homo sapiens 48-51 31623058-0 2019 Pseudolaric Acid B Induces Growth Inhibition and Caspase-Dependent Apoptosis on Head and Neck Cancer Cell lines through Death Receptor 5. pseudolaric acid B 0-18 TNF receptor superfamily member 10b Homo sapiens 120-136 31695422-4 2019 Results: Methyloleanolate increased the fraction of Annexin V/PI apoptotic cells and activated caspase-8, caspase-3, and death receptor 5 (DR5) more than oleanolate in A549 and H1299 cells pretreated with pancaspase inhibitor z-VAD-fmk and DR5 depletion. Methyloleanolate 9-25 TNF receptor superfamily member 10b Homo sapiens 121-137 31695422-4 2019 Results: Methyloleanolate increased the fraction of Annexin V/PI apoptotic cells and activated caspase-8, caspase-3, and death receptor 5 (DR5) more than oleanolate in A549 and H1299 cells pretreated with pancaspase inhibitor z-VAD-fmk and DR5 depletion. Methyloleanolate 9-25 TNF receptor superfamily member 10b Homo sapiens 139-142 31695422-4 2019 Results: Methyloleanolate increased the fraction of Annexin V/PI apoptotic cells and activated caspase-8, caspase-3, and death receptor 5 (DR5) more than oleanolate in A549 and H1299 cells pretreated with pancaspase inhibitor z-VAD-fmk and DR5 depletion. Methyloleanolate 9-25 TNF receptor superfamily member 10b Homo sapiens 240-243 31695422-4 2019 Results: Methyloleanolate increased the fraction of Annexin V/PI apoptotic cells and activated caspase-8, caspase-3, and death receptor 5 (DR5) more than oleanolate in A549 and H1299 cells pretreated with pancaspase inhibitor z-VAD-fmk and DR5 depletion. 28-O-glucopyranosyl-3-O-(O-rhamnopyranosyl-1-3-O-(O-glucopyranosyl)glucopyranosyl)oleanolate 15-25 TNF receptor superfamily member 10b Homo sapiens 121-137 31695422-4 2019 Results: Methyloleanolate increased the fraction of Annexin V/PI apoptotic cells and activated caspase-8, caspase-3, and death receptor 5 (DR5) more than oleanolate in A549 and H1299 cells pretreated with pancaspase inhibitor z-VAD-fmk and DR5 depletion. 28-O-glucopyranosyl-3-O-(O-rhamnopyranosyl-1-3-O-(O-glucopyranosyl)glucopyranosyl)oleanolate 15-25 TNF receptor superfamily member 10b Homo sapiens 139-142 31695422-4 2019 Results: Methyloleanolate increased the fraction of Annexin V/PI apoptotic cells and activated caspase-8, caspase-3, and death receptor 5 (DR5) more than oleanolate in A549 and H1299 cells pretreated with pancaspase inhibitor z-VAD-fmk and DR5 depletion. 28-O-glucopyranosyl-3-O-(O-rhamnopyranosyl-1-3-O-(O-glucopyranosyl)glucopyranosyl)oleanolate 15-25 TNF receptor superfamily member 10b Homo sapiens 240-243 31623058-3 2019 PAB-induced apoptosis is through inducing death receptor 5 (DR5) together with the increase in the expression of cleaved caspase-8. pseudolaric acid B 0-3 TNF receptor superfamily member 10b Homo sapiens 42-58 31623058-3 2019 PAB-induced apoptosis is through inducing death receptor 5 (DR5) together with the increase in the expression of cleaved caspase-8. pseudolaric acid B 0-3 TNF receptor superfamily member 10b Homo sapiens 60-63 31673231-8 2019 Bortezomib also increased DR5 expression and knockdown of DR5 expression significantly recovered cell viability reduced by the combination treatment. bortezomib 0-10 TNF receptor superfamily member 10b Homo sapiens 26-29 31281953-15 2019 Furthermore, celastrol upregulated death receptor 5 (DR5) at the mRNA and protein levels, and siRNA-mediated DR5 knockdown reduced the killing effect of the combination drug treatment on glioma cells and reduced the activation of caspase-3, caspase-8 and PARP. celastrol 13-22 TNF receptor superfamily member 10b Homo sapiens 35-51 31281953-15 2019 Furthermore, celastrol upregulated death receptor 5 (DR5) at the mRNA and protein levels, and siRNA-mediated DR5 knockdown reduced the killing effect of the combination drug treatment on glioma cells and reduced the activation of caspase-3, caspase-8 and PARP. celastrol 13-22 TNF receptor superfamily member 10b Homo sapiens 53-56 31673231-11 2019 However, upregulation of DR5 by bortezomib was knocked down only by inhibition of ERK1/2 activation significantly, but not by JNK activity inhibition. bortezomib 32-42 TNF receptor superfamily member 10b Homo sapiens 25-28 31673231-12 2019 In summary, upregulation of DR5 by bortezomib is of critical significance in the synergy of bortezomib with TRAIL in apoptosis of TRAIL-resistant SNU-216 and that activity of ERK1/2 is required in the bortezomib-induced DR5 overexpression. bortezomib 35-45 TNF receptor superfamily member 10b Homo sapiens 28-31 31673231-12 2019 In summary, upregulation of DR5 by bortezomib is of critical significance in the synergy of bortezomib with TRAIL in apoptosis of TRAIL-resistant SNU-216 and that activity of ERK1/2 is required in the bortezomib-induced DR5 overexpression. bortezomib 35-45 TNF receptor superfamily member 10b Homo sapiens 220-223 31673231-12 2019 In summary, upregulation of DR5 by bortezomib is of critical significance in the synergy of bortezomib with TRAIL in apoptosis of TRAIL-resistant SNU-216 and that activity of ERK1/2 is required in the bortezomib-induced DR5 overexpression. bortezomib 92-102 TNF receptor superfamily member 10b Homo sapiens 28-31 31673231-12 2019 In summary, upregulation of DR5 by bortezomib is of critical significance in the synergy of bortezomib with TRAIL in apoptosis of TRAIL-resistant SNU-216 and that activity of ERK1/2 is required in the bortezomib-induced DR5 overexpression. bortezomib 92-102 TNF receptor superfamily member 10b Homo sapiens 220-223 31673231-12 2019 In summary, upregulation of DR5 by bortezomib is of critical significance in the synergy of bortezomib with TRAIL in apoptosis of TRAIL-resistant SNU-216 and that activity of ERK1/2 is required in the bortezomib-induced DR5 overexpression. bortezomib 92-102 TNF receptor superfamily member 10b Homo sapiens 28-31 31673231-12 2019 In summary, upregulation of DR5 by bortezomib is of critical significance in the synergy of bortezomib with TRAIL in apoptosis of TRAIL-resistant SNU-216 and that activity of ERK1/2 is required in the bortezomib-induced DR5 overexpression. bortezomib 92-102 TNF receptor superfamily member 10b Homo sapiens 220-223 31362889-5 2019 Leptomycin B works synergistically with both TRAIL and death receptor 5 agonistic antibodies to induce apoptosis in TRAIL sensitive cell lines. leptomycin B 0-12 TNF receptor superfamily member 10b Homo sapiens 55-71 31322237-6 2019 Western blotting demonstrated that DP induced apoptosis through extrinsic and intrinsic pathways by upregulating death receptor 4 (DR4), DR5, cleaved caspase-3/-7/-9 and cleaved poly (ADP-ribose) polymerase (PARP), and by decreasing total PARP, total caspase-3/7, Bcl-2 and caspase-9/-10. dracorhodin 35-37 TNF receptor superfamily member 10b Homo sapiens 137-140 31546731-0 2019 p53-Mediated Oxidative Stress Enhances Indirubin-3"-Monoxime-Induced Apoptosis in HCT116 Colon Cancer Cells by Upregulating Death Receptor 5 and TNF-Related Apoptosis-Inducing Ligand Expression. indirubin-3'-monoxime 39-60 TNF receptor superfamily member 10b Homo sapiens 124-140 31546731-2 2019 This study revealed that I3M promotes the expression of death receptor 5 (DR5) and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) in HCT116 p53+/+ cells, resulting in caspase-mediated apoptosis. indirubin-3'-monoxime 25-28 TNF receptor superfamily member 10b Homo sapiens 56-72 31546731-2 2019 This study revealed that I3M promotes the expression of death receptor 5 (DR5) and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) in HCT116 p53+/+ cells, resulting in caspase-mediated apoptosis. indirubin-3'-monoxime 25-28 TNF receptor superfamily member 10b Homo sapiens 74-77 31546731-6 2019 Our results also showed that I3M enhanced transcription factor C/EBP homologous protein (CHOP) expression, resulted in endoplasmic reticulum (ER) stress-mediated DR5 expression, which was upregulated by ROS production in HCT116 p53+/+ cells. indirubin-3'-monoxime 29-32 TNF receptor superfamily member 10b Homo sapiens 162-165 31546731-7 2019 Moreover, co-treatment with I3M and TRAIL enhanced DR5 expression, thereby triggering TRAIL-induced apoptosis of HCT116 p53+/+ cells, which was interfered by a DR5-specific blocking chimeric antibody. indirubin-3'-monoxime 28-31 TNF receptor superfamily member 10b Homo sapiens 51-54 31546731-7 2019 Moreover, co-treatment with I3M and TRAIL enhanced DR5 expression, thereby triggering TRAIL-induced apoptosis of HCT116 p53+/+ cells, which was interfered by a DR5-specific blocking chimeric antibody. indirubin-3'-monoxime 28-31 TNF receptor superfamily member 10b Homo sapiens 160-163 31546731-8 2019 In summary, I3M potently enhances TRAIL-induced apoptosis by upregulating DR5 expression via p53-mediated ROS production in HCT116 p53+/+ cells. indirubin-3'-monoxime 12-15 TNF receptor superfamily member 10b Homo sapiens 74-77 31416165-9 2019 Mutation of a putative nuclear export sequence (NES) in TRAIL-R2 or the inhibition of CRM-1 by Leptomycin-B resulted in the nuclear accumulation of TRAIL-R2. leptomycin B 95-107 TNF receptor superfamily member 10b Homo sapiens 148-156 30556589-0 2019 Chrysanthemulide A induces apoptosis through DR5 upregulation via JNK-mediated autophagosome accumulation in human osteosarcoma cells. chrysanthemulide a 0-18 TNF receptor superfamily member 10b Homo sapiens 45-48 30556589-8 2019 Furthermore, CA activated the c-Jun N-terminal kinase (JNK) signaling pathway, and treatment with JNK siRNAs or inhibitor SP600125 significantly attenuated CA-mediated autophagosome accumulation and DR5-mediated cell apoptosis. pyrazolanthrone 122-130 TNF receptor superfamily member 10b Homo sapiens 199-202 30958996-4 2019 Transfection of DR5 siRNA into H1299 cells significantly diminished the enhancing effects of Q on TSA-induced apoptosis. Quercetin 93-94 TNF receptor superfamily member 10b Homo sapiens 16-19 31233200-8 2019 However, in the resistant cells, the oxaliplatin-triggered extrinsic apoptotic pathway appeared to be suppressed by decreased lipid raft formation, and recruitment of death receptor 5 and FADD into lipid rafts. Oxaliplatin 37-48 TNF receptor superfamily member 10b Homo sapiens 167-183 30958996-4 2019 Transfection of DR5 siRNA into H1299 cells significantly diminished the enhancing effects of Q on TSA-induced apoptosis. trichostatin A 98-101 TNF receptor superfamily member 10b Homo sapiens 16-19 30958996-8 2019 These data suggest that the up-regulation of p300 expression, which in turn increases histone acetylation and DR5 expression, plays an important role in the enhancing effect of Q on TSA/vorinostat- induced apoptosis in H1299 cells. Quercetin 177-178 TNF receptor superfamily member 10b Homo sapiens 110-113 30958996-8 2019 These data suggest that the up-regulation of p300 expression, which in turn increases histone acetylation and DR5 expression, plays an important role in the enhancing effect of Q on TSA/vorinostat- induced apoptosis in H1299 cells. trichostatin A 182-185 TNF receptor superfamily member 10b Homo sapiens 110-113 30958996-8 2019 These data suggest that the up-regulation of p300 expression, which in turn increases histone acetylation and DR5 expression, plays an important role in the enhancing effect of Q on TSA/vorinostat- induced apoptosis in H1299 cells. Vorinostat 186-196 TNF receptor superfamily member 10b Homo sapiens 110-113 30730256-8 2019 These results indicated that ERK1/2-mediated ELK1/CHOP/DR5 pathway is involved in 6-MSITC-induced apoptosis in colorectal cancer cells. 6-(Methylsulfinyl)hexyl isothiocyanate 82-89 TNF receptor superfamily member 10b Homo sapiens 55-58 30737960-5 2019 Through upregulating the expression of DR4 and DR5 at transcriptional and protein levels, Periplocin enhanced the sensitivity of gastric cancer cells to TRAIL. periplocin 90-100 TNF receptor superfamily member 10b Homo sapiens 47-50 30724400-3 2019 RESULTS: Cisplatin treatment in IMR-32 cells increased the expression of death receptor 5 (DR5; TRAIL-R2), but not other receptors, via downregulation of NF-kappaB activity. Cisplatin 9-18 TNF receptor superfamily member 10b Homo sapiens 73-89 30724400-3 2019 RESULTS: Cisplatin treatment in IMR-32 cells increased the expression of death receptor 5 (DR5; TRAIL-R2), but not other receptors, via downregulation of NF-kappaB activity. Cisplatin 9-18 TNF receptor superfamily member 10b Homo sapiens 91-94 30724400-3 2019 RESULTS: Cisplatin treatment in IMR-32 cells increased the expression of death receptor 5 (DR5; TRAIL-R2), but not other receptors, via downregulation of NF-kappaB activity. Cisplatin 9-18 TNF receptor superfamily member 10b Homo sapiens 96-104 30724400-4 2019 However, the cisplatin-mediated increase in DR5 failed to induce cell death following TRAIL treatment. Cisplatin 13-22 TNF receptor superfamily member 10b Homo sapiens 44-47 30724400-9 2019 Concurrent treatment with cisplatin enhanced TRAIL-mediated cytotoxicity, which was abrogated by an additional pretreatment with DR5:Fc chimera protein. Cisplatin 26-35 TNF receptor superfamily member 10b Homo sapiens 129-132 30730256-0 2019 Involvement of ERK1/2-mediated ELK1/CHOP/DR5 pathway in 6-(methylsulfinyl)hexyl isothiocyanate-induced apoptosis of colorectal cancer cells. 6-(Methylsulfinyl)hexyl isothiocyanate 56-94 TNF receptor superfamily member 10b Homo sapiens 41-44 30765502-8 2019 Furthermore, intracellular cholesterol regulates temozolomide-induced cell death in glioblastoma cells via accumulation and activation of death receptor 5 in plasma membrane lipid rafts. Cholesterol 27-38 TNF receptor superfamily member 10b Homo sapiens 138-154 30730256-6 2019 6-MSITC stimulated ERK1/2 phosphorylation, and then activated ERK1/2 signaling including ELK1 phosphorylation, and upregulation of C/EBP homologous protein (CHOP) and death receptor 5 (DR5). 6-(Methylsulfinyl)hexyl isothiocyanate 0-7 TNF receptor superfamily member 10b Homo sapiens 167-183 30730256-6 2019 6-MSITC stimulated ERK1/2 phosphorylation, and then activated ERK1/2 signaling including ELK1 phosphorylation, and upregulation of C/EBP homologous protein (CHOP) and death receptor 5 (DR5). 6-(Methylsulfinyl)hexyl isothiocyanate 0-7 TNF receptor superfamily member 10b Homo sapiens 185-188 30864676-0 2019 Reversine induces cell cycle arrest and apoptosis via upregulation of the Fas and DR5 signaling pathways in human colorectal cancer cells. 2-(4-morpholinoanilino)-6-cyclohexylaminopurine 0-9 TNF receptor superfamily member 10b Homo sapiens 82-85 30864676-10 2019 Reversine-induced apoptosis and cell cycle arrest were suppressed by inhibition of Fas and DR5 expression via siRNA. 2-(4-morpholinoanilino)-6-cyclohexylaminopurine 0-9 TNF receptor superfamily member 10b Homo sapiens 91-94 30642878-7 2019 We used this assay to screen compounds from three commercially available libraries, and identified 1-benzyl-3-cetyl-2-methylimidazolium iodide (NH125) as a potent inducer of DR5 expression. NH 125 99-142 TNF receptor superfamily member 10b Homo sapiens 174-177 30642878-7 2019 We used this assay to screen compounds from three commercially available libraries, and identified 1-benzyl-3-cetyl-2-methylimidazolium iodide (NH125) as a potent inducer of DR5 expression. NH 125 144-149 TNF receptor superfamily member 10b Homo sapiens 174-177 30817944-5 2019 The sensitizing effect of AC to TRAIL was well correlated with inhibition of death receptor 5 (DR5) CHOP, and p53 expression. auriculasin 26-28 TNF receptor superfamily member 10b Homo sapiens 77-93 30692634-5 2019 Overexpression of RUNX3 increased DR5 expression via induction of the reactive oxygen species (ROS)-endoplasmic reticulum (ER) stress-effector CHOP. Reactive Oxygen Species 70-93 TNF receptor superfamily member 10b Homo sapiens 34-37 30692634-5 2019 Overexpression of RUNX3 increased DR5 expression via induction of the reactive oxygen species (ROS)-endoplasmic reticulum (ER) stress-effector CHOP. Reactive Oxygen Species 95-98 TNF receptor superfamily member 10b Homo sapiens 34-37 30817944-5 2019 The sensitizing effect of AC to TRAIL was well correlated with inhibition of death receptor 5 (DR5) CHOP, and p53 expression. auriculasin 26-28 TNF receptor superfamily member 10b Homo sapiens 95-98 30817944-7 2019 These results suggest that non-toxic concentrations of AC sensitize TRAIL-resistant primary prostate cancer cells to TRAIL-mediated apoptosis via up-regulation of DR5 and downstream signaling pathways. auriculasin 55-57 TNF receptor superfamily member 10b Homo sapiens 163-166 31032087-0 2019 Artonin E sensitizes TRAIL-induced apoptosis by DR5 upregulation and cFLIP downregulation in TRAIL-refractory colorectal cancer LoVo cells. artonin E 0-9 TNF receptor superfamily member 10b Homo sapiens 48-51 31032087-3 2019 In an effort to overcome TRAIL-refractory cancer, we investigated the effect of artonin E in regulating death receptor 5 (DR5) and cellular FLICE (FADD-like IL-1beta-converting enzyme)-inhibitory protein (cFLIP), two major mediators regulate TRAIL-induced apoptosis, in LoVo cells as a model of TRAIL refractory CRC. artonin E 80-89 TNF receptor superfamily member 10b Homo sapiens 104-120 31032087-3 2019 In an effort to overcome TRAIL-refractory cancer, we investigated the effect of artonin E in regulating death receptor 5 (DR5) and cellular FLICE (FADD-like IL-1beta-converting enzyme)-inhibitory protein (cFLIP), two major mediators regulate TRAIL-induced apoptosis, in LoVo cells as a model of TRAIL refractory CRC. artonin E 80-89 TNF receptor superfamily member 10b Homo sapiens 122-125 31032087-9 2019 Artonin E enhanced both mRNA and protein expression of DR5. artonin E 0-9 TNF receptor superfamily member 10b Homo sapiens 55-58 31032087-11 2019 All together we showed that artonin E enhanced TRAIL-induced apoptosis in LoVo cells through DR5 upregulation and cFLIP downregulation. artonin E 28-37 TNF receptor superfamily member 10b Homo sapiens 93-96 31032087-12 2019 Conclusions: Artonin E was able to increase DR5 expression and decrease cFLIP expression in LoVo cells. artonin E 13-22 TNF receptor superfamily member 10b Homo sapiens 44-47 30765502-8 2019 Furthermore, intracellular cholesterol regulates temozolomide-induced cell death in glioblastoma cells via accumulation and activation of death receptor 5 in plasma membrane lipid rafts. Temozolomide 49-61 TNF receptor superfamily member 10b Homo sapiens 138-154 30689998-0 2019 Galbanic acid potentiates TRAIL induced apoptosis in resistant non-small cell lung cancer cells via inhibition of MDR1 and activation of caspases and DR5. galbanic acid 0-13 TNF receptor superfamily member 10b Homo sapiens 150-153 31019655-12 2019 The blockage of autophagy by bafilomycin A1 or Atg7 siRNA abolished SNX-2112-induced upregulation of DR5. SNX 2112 68-76 TNF receptor superfamily member 10b Homo sapiens 101-104 31019655-13 2019 Meanwhile, ROS scavenger NAC, JNK inhibitor SP600125, and p53 inhibitor PFTalpha were used to verify that autophagy-mediated upregulation of DR5 was regulated by the SNX-2112-stimulated activation of the ROS-JNK-p53 signaling pathway. Reactive Oxygen Species 11-14 TNF receptor superfamily member 10b Homo sapiens 141-144 31019655-13 2019 Meanwhile, ROS scavenger NAC, JNK inhibitor SP600125, and p53 inhibitor PFTalpha were used to verify that autophagy-mediated upregulation of DR5 was regulated by the SNX-2112-stimulated activation of the ROS-JNK-p53 signaling pathway. pyrazolanthrone 44-52 TNF receptor superfamily member 10b Homo sapiens 141-144 31019655-13 2019 Meanwhile, ROS scavenger NAC, JNK inhibitor SP600125, and p53 inhibitor PFTalpha were used to verify that autophagy-mediated upregulation of DR5 was regulated by the SNX-2112-stimulated activation of the ROS-JNK-p53 signaling pathway. pifithrin 72-80 TNF receptor superfamily member 10b Homo sapiens 141-144 31019655-13 2019 Meanwhile, ROS scavenger NAC, JNK inhibitor SP600125, and p53 inhibitor PFTalpha were used to verify that autophagy-mediated upregulation of DR5 was regulated by the SNX-2112-stimulated activation of the ROS-JNK-p53 signaling pathway. SNX 2112 166-174 TNF receptor superfamily member 10b Homo sapiens 141-144 31019655-13 2019 Meanwhile, ROS scavenger NAC, JNK inhibitor SP600125, and p53 inhibitor PFTalpha were used to verify that autophagy-mediated upregulation of DR5 was regulated by the SNX-2112-stimulated activation of the ROS-JNK-p53 signaling pathway. Reactive Oxygen Species 204-207 TNF receptor superfamily member 10b Homo sapiens 141-144 30689998-10 2019 Overall, our findings suggest that galbanic acid enhances TRAIL induced apoptosis via inhibition of MDR1 and activation of caspases and DR5 in H460/R cells as a potent TRAIL sensitizer. galbanic acid 35-48 TNF receptor superfamily member 10b Homo sapiens 136-139 31019655-0 2019 Hsp90 Inhibitor SNX-2112 Enhances TRAIL-Induced Apoptosis of Human Cervical Cancer Cells via the ROS-Mediated JNK-p53-Autophagy-DR5 Pathway. SNX 2112 16-24 TNF receptor superfamily member 10b Homo sapiens 128-131 31019655-0 2019 Hsp90 Inhibitor SNX-2112 Enhances TRAIL-Induced Apoptosis of Human Cervical Cancer Cells via the ROS-Mediated JNK-p53-Autophagy-DR5 Pathway. Reactive Oxygen Species 97-100 TNF receptor superfamily member 10b Homo sapiens 128-131 31019655-9 2019 Moreover, SNX-2112 induced the upregulation of death-receptor DR5 in HeLa cells. SNX 2112 10-18 TNF receptor superfamily member 10b Homo sapiens 62-65 31019655-10 2019 The silencing of DR5 by siRNA significantly decreased cell apoptosis by the combined effect of SNX-2112 and TRAIL. SNX 2112 95-103 TNF receptor superfamily member 10b Homo sapiens 17-20 30674532-5 2019 In this study, we report that death receptor 5 (DR5), a key component of the extrinsic apoptotic pathway, is markedly induced in response to BRD4 depletion and BETi treatment in colorectal cancer cells. beti 160-164 TNF receptor superfamily member 10b Homo sapiens 30-46 30674532-5 2019 In this study, we report that death receptor 5 (DR5), a key component of the extrinsic apoptotic pathway, is markedly induced in response to BRD4 depletion and BETi treatment in colorectal cancer cells. beti 160-164 TNF receptor superfamily member 10b Homo sapiens 48-51 30674532-7 2019 Enhanced DR5 induction was necessary for the chemosensitization and apoptotic effects of BETi and was responsible for increased BETi sensitivity in colorectal cancer cells containing a mutation in speckle-type POZ protein (SPOP), a subunit of BRD4 E3 ubiquitin ligase. beti 89-93 TNF receptor superfamily member 10b Homo sapiens 9-12 30674532-7 2019 Enhanced DR5 induction was necessary for the chemosensitization and apoptotic effects of BETi and was responsible for increased BETi sensitivity in colorectal cancer cells containing a mutation in speckle-type POZ protein (SPOP), a subunit of BRD4 E3 ubiquitin ligase. beti 128-132 TNF receptor superfamily member 10b Homo sapiens 9-12 30674532-8 2019 In a colorectal cancer xenograft model, BETi combined with chemotherapy suppressed the tumor growth in a DR5-dependent manner and potently inhibited patient-derived xenograft tumor growth with enhanced DR5 induction and apoptosis. beti 40-44 TNF receptor superfamily member 10b Homo sapiens 105-108 30674532-8 2019 In a colorectal cancer xenograft model, BETi combined with chemotherapy suppressed the tumor growth in a DR5-dependent manner and potently inhibited patient-derived xenograft tumor growth with enhanced DR5 induction and apoptosis. beti 40-44 TNF receptor superfamily member 10b Homo sapiens 202-205 30674532-9 2019 These findings suggest that BETi alone or in combination with chemotherapy is effective against colorectal cancer due to enhanced DR5 induction and apoptosis. beti 28-32 TNF receptor superfamily member 10b Homo sapiens 130-133 30870485-13 2019 This is the first study to demonstrate that XN alters the expression of DR5 as well as the synergistic effect of XN on TRAIL in NB and provides a strong rationale for further preclinical analysis. xanthohumol 44-46 TNF receptor superfamily member 10b Homo sapiens 72-75 30647121-7 2019 NEO214-induced cytotoxicity involves apoptosis triggered by endoplasmic reticulum (ER) stress, as well as activating the Death Receptor 5 (DR5)/TNF-related apoptosis-inducing ligand (TRAIL/Apo2L) pathway. neo214 0-6 TNF receptor superfamily member 10b Homo sapiens 121-137 29916141-11 2019 R-CRT overexpression in the cytoplasm of MO59K rendered the cells susceptible to BT-induced, DR5-mediated cell death. Bortezomib 81-83 TNF receptor superfamily member 10b Homo sapiens 93-96 30647121-7 2019 NEO214-induced cytotoxicity involves apoptosis triggered by endoplasmic reticulum (ER) stress, as well as activating the Death Receptor 5 (DR5)/TNF-related apoptosis-inducing ligand (TRAIL/Apo2L) pathway. neo214 0-6 TNF receptor superfamily member 10b Homo sapiens 139-142 30431076-5 2019 The attenuation of the autophagic flux by applying the specific inhibitor of autophagy, chloroquine, significantly suppressed DR5 expression. Chloroquine 88-99 TNF receptor superfamily member 10b Homo sapiens 126-129 30589515-9 2019 Interestingly, we observed that induction of the salvage pathway by external administration of l-fucose restores DR5-mediated apoptosis in both DLD-1 and HCT 116 cells. Fucose 95-103 TNF receptor superfamily member 10b Homo sapiens 113-116 30431076-7 2019 Furthermore, pre-treatment with the c-Jun N-terminal kinase (JNK) inhibitor, SP600125, significantly attenuated the luteolin-induced upregulation of DR5 expression, thereby suggesting that JNK activation promotes DR5 expression. pyrazolanthrone 77-85 TNF receptor superfamily member 10b Homo sapiens 149-152 30431076-7 2019 Furthermore, pre-treatment with the c-Jun N-terminal kinase (JNK) inhibitor, SP600125, significantly attenuated the luteolin-induced upregulation of DR5 expression, thereby suggesting that JNK activation promotes DR5 expression. pyrazolanthrone 77-85 TNF receptor superfamily member 10b Homo sapiens 213-216 30359552-0 2019 Ixazomib promotes CHOP-dependent DR5 induction and apoptosis in colorectal cancer cells. ixazomib 0-8 TNF receptor superfamily member 10b Homo sapiens 33-36 30359552-4 2019 Western blotting and real-time RT-PCR were performed to detect ixazomib-induced DR5 upregulation. ixazomib 63-71 TNF receptor superfamily member 10b Homo sapiens 80-83 30991885-6 2019 We found that MS-444 treatment of glioblastoma cells resulted in loss of viability and induction of apoptosis, with evidence implicating death receptor 5. 5,8-dihydroxy-3-methyl-4-(9H)-naphtho(2,3-c)furanone 14-20 TNF receptor superfamily member 10b Homo sapiens 137-153 30359552-8 2019 Our findings indicated that ixazomib treatment induces CHOP-dependent DR5 induction, irrespective of p53 status. ixazomib 28-36 TNF receptor superfamily member 10b Homo sapiens 70-73 30359552-9 2019 Furthermore, DR5 is necessary for ixazomib-mediated apoptosis. ixazomib 34-42 TNF receptor superfamily member 10b Homo sapiens 13-16 30359552-10 2019 Ixazomib also synergized with TRAIL to induce marked apoptosis via DR5 in CRC cells. ixazomib 0-8 TNF receptor superfamily member 10b Homo sapiens 67-70 30359552-11 2019 CONCLUSIONS: Our findings further suggested that ixazomib sensitizes TRAIL/death receptor signaling pathway-targeted CRC and suggested that DR5 induction could be a valuable indicator of ixazomib sensitivity. ixazomib 187-195 TNF receptor superfamily member 10b Homo sapiens 140-143 30423122-7 2019 Pretreatment with flutamide, as well as knockdown of androgen receptor, decreased testosterone-induced DR5 and CHOP expression, as well as apoptosis. Flutamide 18-27 TNF receptor superfamily member 10b Homo sapiens 103-106 30423122-7 2019 Pretreatment with flutamide, as well as knockdown of androgen receptor, decreased testosterone-induced DR5 and CHOP expression, as well as apoptosis. Testosterone 82-94 TNF receptor superfamily member 10b Homo sapiens 103-106 30423122-4 2019 In this study, we found that testosterone induced expression of various UPR genes, including CHOP, as well as DR5, in cultured human granulosa-lutein cells (GLCs). Testosterone 29-41 TNF receptor superfamily member 10b Homo sapiens 110-113 30423122-8 2019 Expression of DR5 and CHOP was upregulated in GLCs obtained from patients with PCOS, as well as in granulosa cells of antral follicles in ovarian sections obtained from patients with PCOS and dehydroepiandrosterone-induced PCOS mice. Dehydroepiandrosterone 192-214 TNF receptor superfamily member 10b Homo sapiens 14-17 30423122-5 2019 Pretreatment with the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) inhibited testosterone-induced apoptosis and expression of DR5 and CHOP. ursodoxicoltaurine 42-67 TNF receptor superfamily member 10b Homo sapiens 135-138 30423122-5 2019 Pretreatment with the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) inhibited testosterone-induced apoptosis and expression of DR5 and CHOP. ursodoxicoltaurine 69-74 TNF receptor superfamily member 10b Homo sapiens 135-138 30202993-3 2019 In this study, we investigated the hepatotoxicity caused by KMTR2, an anti-human TRAIL-R2 monoclonal antibody, in chimeric mice with humanized livers (PXB-mice). kmtr2 60-65 TNF receptor superfamily member 10b Homo sapiens 81-89 30423122-5 2019 Pretreatment with the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) inhibited testosterone-induced apoptosis and expression of DR5 and CHOP. Testosterone 86-98 TNF receptor superfamily member 10b Homo sapiens 135-138 30423122-6 2019 Knockdown of CHOP inhibited testosterone-induced DR5 expression and apoptosis, and knockdown of DR5 inhibited testosterone-induced apoptosis. Testosterone 28-40 TNF receptor superfamily member 10b Homo sapiens 49-52 30423122-6 2019 Knockdown of CHOP inhibited testosterone-induced DR5 expression and apoptosis, and knockdown of DR5 inhibited testosterone-induced apoptosis. Testosterone 110-122 TNF receptor superfamily member 10b Homo sapiens 96-99 30486888-9 2018 We show here for the first time that (a) olaparib significantly increased tumor cell sensitivity to NK killing and ADCC in both BRCA WT and BRCA mutant prostate carcinoma cells, independent of PD-L1 or EGFR modulation; (b) mechanistically, treatment with olaparib upregulated death receptor TRAIL-R2; and (c) olaparib significantly enhanced NK killing of additional tumor types, including breast, non-small cell lung carcinoma, and chordoma. olaparib 41-49 TNF receptor superfamily member 10b Homo sapiens 291-299 30359578-0 2018 C-27-carboxylated oleanane triterpenoids up-regulate TRAIL DISC assembly via p38 MAPK and CHOP-mediated DR5 expression in human glioblastoma cells. c-27-carboxylated oleanane 0-26 TNF receptor superfamily member 10b Homo sapiens 104-107 30359578-0 2018 C-27-carboxylated oleanane triterpenoids up-regulate TRAIL DISC assembly via p38 MAPK and CHOP-mediated DR5 expression in human glioblastoma cells. triterpenoids 27-40 TNF receptor superfamily member 10b Homo sapiens 104-107 30359578-5 2018 The upregulation of DR5 expression by C27OAs strictly depends on transactivation of C/EBP homology protein, which is regulated through the p38 MAPK pathway, rather than p53 and intracellular reactive oxygen species status. Reactive Oxygen Species 191-214 TNF receptor superfamily member 10b Homo sapiens 20-23 30713790-0 2019 Bortezomib sensitizes multiple myeloma to NK cells via ER-stress-induced suppression of HLA-E and upregulation of DR5. Bortezomib 0-10 TNF receptor superfamily member 10b Homo sapiens 114-117 30463333-5 2018 Maritoclax-induced DR5 upregulation was regulated by induction of C/EBP homologous protein (CHOP) expression. marinopyrrole A 0-10 TNF receptor superfamily member 10b Homo sapiens 19-22 29964331-5 2018 BIX-01294 markedly increased death receptor 5 (DR5) expression, while silencing of DR5 using small interfering RNAs abolished the TRAIL-sensitizing effect of BIX-01294. BIX 01294 158-167 TNF receptor superfamily member 10b Homo sapiens 83-86 30132536-0 2018 3-Bromopyruvate sensitizes human breast cancer cells to TRAIL-induced apoptosis via the phosphorylated AMPK-mediated upregulation of DR5. bromopyruvate 0-15 TNF receptor superfamily member 10b Homo sapiens 133-136 30132536-5 2018 In the present study, it was demonstrated that 3-BP synergistically sensitized breast cancer cells to TRAIL-induced apoptosis via the upregulation of death receptor 5 (DR5). bromopyruvate 47-51 TNF receptor superfamily member 10b Homo sapiens 150-166 30132536-5 2018 In the present study, it was demonstrated that 3-BP synergistically sensitized breast cancer cells to TRAIL-induced apoptosis via the upregulation of death receptor 5 (DR5). bromopyruvate 47-51 TNF receptor superfamily member 10b Homo sapiens 168-171 30132536-12 2018 In conclusion, these results indicated that 3-BP sensitized breast cancer cells to TRAIL via the AMPK-mediated upregulation of DR5. bromopyruvate 44-48 TNF receptor superfamily member 10b Homo sapiens 127-130 29964331-0 2018 Inhibition of euchromatin histone-lysine N-methyltransferase 2 sensitizes breast cancer cells to tumor necrosis factor-related apoptosis-inducing ligand through reactive oxygen species-mediated activating transcription factor 4-C/EBP homologous protein-death receptor 5 pathway activation. Reactive Oxygen Species 161-184 TNF receptor superfamily member 10b Homo sapiens 253-269 29964331-6 2018 Specifically, BIX-01294 induced C/EBP homologous protein (CHOP)-mediated DR5 gene transcriptional activation and DR5 promoter activation was induced by upregulation of the protein kinase R-like endoplasmic reticulum kinase-mediated activating transcription factor 4 (ATF4). BIX 01294 14-23 TNF receptor superfamily member 10b Homo sapiens 73-76 29964331-5 2018 BIX-01294 markedly increased death receptor 5 (DR5) expression, while silencing of DR5 using small interfering RNAs abolished the TRAIL-sensitizing effect of BIX-01294. BIX 01294 0-9 TNF receptor superfamily member 10b Homo sapiens 29-45 29964331-5 2018 BIX-01294 markedly increased death receptor 5 (DR5) expression, while silencing of DR5 using small interfering RNAs abolished the TRAIL-sensitizing effect of BIX-01294. BIX 01294 0-9 TNF receptor superfamily member 10b Homo sapiens 47-50 29964331-7 2018 Moreover, inhibition of reactive oxygen species by N-acetyl-L-cysteine efficiently blocked BIX-01294-induced DR5 upregulation by inhibiting ATF4/CHOP expression, leading to diminished sensitization to TRAIL. Reactive Oxygen Species 24-47 TNF receptor superfamily member 10b Homo sapiens 109-112 29964331-7 2018 Moreover, inhibition of reactive oxygen species by N-acetyl-L-cysteine efficiently blocked BIX-01294-induced DR5 upregulation by inhibiting ATF4/CHOP expression, leading to diminished sensitization to TRAIL. Acetylcysteine 51-70 TNF receptor superfamily member 10b Homo sapiens 109-112 29964331-7 2018 Moreover, inhibition of reactive oxygen species by N-acetyl-L-cysteine efficiently blocked BIX-01294-induced DR5 upregulation by inhibiting ATF4/CHOP expression, leading to diminished sensitization to TRAIL. BIX 01294 91-100 TNF receptor superfamily member 10b Homo sapiens 109-112 29964331-8 2018 These findings suggest that BIX-01294 sensitizes breast cancer cells to TRAIL by upregulating ATF4/CHOP-dependent DR5 expression with a reactive oxygen species-dependent manner. BIX 01294 28-37 TNF receptor superfamily member 10b Homo sapiens 114-117 29964331-8 2018 These findings suggest that BIX-01294 sensitizes breast cancer cells to TRAIL by upregulating ATF4/CHOP-dependent DR5 expression with a reactive oxygen species-dependent manner. Reactive Oxygen Species 136-159 TNF receptor superfamily member 10b Homo sapiens 114-117 29869749-0 2018 1H, 13C, 15N NMR resonance assignments and secondary structure determination of the extra-cellular domain from the human proapoptotic TRAIL-R2 death receptor 5 (DR5-ECD). Hydrogen 0-2 TNF receptor superfamily member 10b Homo sapiens 143-159 29869749-0 2018 1H, 13C, 15N NMR resonance assignments and secondary structure determination of the extra-cellular domain from the human proapoptotic TRAIL-R2 death receptor 5 (DR5-ECD). Hydrogen 0-2 TNF receptor superfamily member 10b Homo sapiens 161-164 30177492-5 2018 The results of MTT assay of Jurkat, K562 and Molt-4 cells with IPH and co-staining experiments with IPH and an anti-DR5 antibody indicate that IPH binds to DR5 and induces apoptosis in a manner parallel to the DR5 expression level. monooxyethylene trimethylolpropane tristearate 15-18 TNF receptor superfamily member 10b Homo sapiens 156-159 30177492-5 2018 The results of MTT assay of Jurkat, K562 and Molt-4 cells with IPH and co-staining experiments with IPH and an anti-DR5 antibody indicate that IPH binds to DR5 and induces apoptosis in a manner parallel to the DR5 expression level. monooxyethylene trimethylolpropane tristearate 15-18 TNF receptor superfamily member 10b Homo sapiens 156-159 30177492-5 2018 The results of MTT assay of Jurkat, K562 and Molt-4 cells with IPH and co-staining experiments with IPH and an anti-DR5 antibody indicate that IPH binds to DR5 and induces apoptosis in a manner parallel to the DR5 expression level. iph 100-103 TNF receptor superfamily member 10b Homo sapiens 156-159 30177492-5 2018 The results of MTT assay of Jurkat, K562 and Molt-4 cells with IPH and co-staining experiments with IPH and an anti-DR5 antibody indicate that IPH binds to DR5 and induces apoptosis in a manner parallel to the DR5 expression level. iph 100-103 TNF receptor superfamily member 10b Homo sapiens 156-159 30177492-5 2018 The results of MTT assay of Jurkat, K562 and Molt-4 cells with IPH and co-staining experiments with IPH and an anti-DR5 antibody indicate that IPH binds to DR5 and induces apoptosis in a manner parallel to the DR5 expression level. iph 100-103 TNF receptor superfamily member 10b Homo sapiens 156-159 30177492-5 2018 The results of MTT assay of Jurkat, K562 and Molt-4 cells with IPH and co-staining experiments with IPH and an anti-DR5 antibody indicate that IPH binds to DR5 and induces apoptosis in a manner parallel to the DR5 expression level. iph 100-103 TNF receptor superfamily member 10b Homo sapiens 156-159 30189637-5 2018 Niclosamide markedly increased DR5 protein levels, including cell-surface DR5, and decreased c-FLIP protein levels. Niclosamide 0-11 TNF receptor superfamily member 10b Homo sapiens 31-34 30189637-5 2018 Niclosamide markedly increased DR5 protein levels, including cell-surface DR5, and decreased c-FLIP protein levels. Niclosamide 0-11 TNF receptor superfamily member 10b Homo sapiens 74-77 30189637-6 2018 Down-regulation of DR5 by specific small interfering RNA (siRNA) and ectopic expression of c-FLIP markedly blocked niclosamide plus TRAIL-induced apoptosis. Niclosamide 115-126 TNF receptor superfamily member 10b Homo sapiens 19-22 30189637-7 2018 Our findings provide that niclosamide could overcome resistance to TRAIL through up-regulating DR5 on the cell surface and down-regulating c-FLIP in cancer cells. Niclosamide 26-37 TNF receptor superfamily member 10b Homo sapiens 95-98 29957841-7 2018 The cholesterol-sequestering agent nystatin partially reversed DR5 clustering and DISC formation, preventing apoptosis triggered by the combination of beta-elemene and TRAIL. Cholesterol 4-15 TNF receptor superfamily member 10b Homo sapiens 63-66 29957841-7 2018 The cholesterol-sequestering agent nystatin partially reversed DR5 clustering and DISC formation, preventing apoptosis triggered by the combination of beta-elemene and TRAIL. Nystatin 35-43 TNF receptor superfamily member 10b Homo sapiens 63-66 29957841-5 2018 Compared to beta-elemene or TRAIL alone, treatment with beta-elemene and TRAIL obviously promoted DR5 clustering as well as translocation of Caspase-8, DR5 and FADD into lipid rafts. beta-elemene 56-68 TNF receptor superfamily member 10b Homo sapiens 98-101 29929000-0 2018 Telmisartan generates ROS-dependent upregulation of death receptor 5 to sensitize TRAIL in lung cancer via inhibition of autophagy flux. Telmisartan 0-11 TNF receptor superfamily member 10b Homo sapiens 52-68 29957841-5 2018 Compared to beta-elemene or TRAIL alone, treatment with beta-elemene and TRAIL obviously promoted DR5 clustering as well as translocation of Caspase-8, DR5 and FADD into lipid rafts. beta-elemene 56-68 TNF receptor superfamily member 10b Homo sapiens 152-155 29929000-0 2018 Telmisartan generates ROS-dependent upregulation of death receptor 5 to sensitize TRAIL in lung cancer via inhibition of autophagy flux. ros 22-25 TNF receptor superfamily member 10b Homo sapiens 52-68 29929000-4 2018 In this study, we evaluated telmisartan as a novel TRAIL-DR5-targeting agent with the aim of rendering TRAIL-based cancer therapies more active. Telmisartan 28-39 TNF receptor superfamily member 10b Homo sapiens 57-60 29929000-6 2018 The molecular mechanism includes the blocking of AMPK phosphorylation causes inhibition of autophagy flux by telmisartan resulting in ROS generation leading to death receptor (DR5) upregulation and subsequent activation of the caspase cascade by TRAIL treatment. ros 134-137 TNF receptor superfamily member 10b Homo sapiens 176-179 29929000-7 2018 Furthermore, using chloroquine and siATG5 significantly enhances ROS production and application of the ROS scavenger N-acetyl-cysteine (NAC) rescues the cells undergoing apoptosis by abrogating the expression of DR5 and finally the caspase cascade. Chloroquine 19-30 TNF receptor superfamily member 10b Homo sapiens 212-215 29929000-7 2018 Furthermore, using chloroquine and siATG5 significantly enhances ROS production and application of the ROS scavenger N-acetyl-cysteine (NAC) rescues the cells undergoing apoptosis by abrogating the expression of DR5 and finally the caspase cascade. ros 103-106 TNF receptor superfamily member 10b Homo sapiens 212-215 29929000-7 2018 Furthermore, using chloroquine and siATG5 significantly enhances ROS production and application of the ROS scavenger N-acetyl-cysteine (NAC) rescues the cells undergoing apoptosis by abrogating the expression of DR5 and finally the caspase cascade. Acetylcysteine 117-134 TNF receptor superfamily member 10b Homo sapiens 212-215 29929000-7 2018 Furthermore, using chloroquine and siATG5 significantly enhances ROS production and application of the ROS scavenger N-acetyl-cysteine (NAC) rescues the cells undergoing apoptosis by abrogating the expression of DR5 and finally the caspase cascade. Acetylcysteine 136-139 TNF receptor superfamily member 10b Homo sapiens 212-215 30061216-6 2018 Nelfinavir induced ER stress and increased the expression of TRAIL death receptor (DR) 4 and DR5, sensitizing the cancer cells to TRAIL. Nelfinavir 0-10 TNF receptor superfamily member 10b Homo sapiens 93-96 30157799-4 2018 We show that due to to its dual action on DR5, a death receptor for TRAIL and on STAT3, Cryptotanshinone can be used to increase sensitivity to TRAIL. cryptotanshinone 88-104 TNF receptor superfamily member 10b Homo sapiens 42-45 30120241-0 2018 Author Correction: Bisphosphonate enhances TRAIL sensitivity to human osteosarcoma cells via death receptor 5 upregulation. Diphosphonates 19-33 TNF receptor superfamily member 10b Homo sapiens 93-109 29800641-0 2018 Isoalantolactone induces apoptosis through reactive oxygen species-dependent upregulation of death receptor 5 in human esophageal cancer cells. isoalantolactone 0-16 TNF receptor superfamily member 10b Homo sapiens 93-109 29596115-7 2018 Quercetin action is manifested by the upregulation of rhTRAIL-binding receptors DR4 and DR5 on the surface of cancer cells and by increased rate of the proteasome-mediated degradation of the antiapoptotic protein FLIP. Quercetin 0-9 TNF receptor superfamily member 10b Homo sapiens 88-91 29792947-4 2018 Combined treatment with SFN and TRAIL (SFN/TRAIL) significantly induced apoptosis concomitant with activation of caspases, loss of mitochondrial membrane potential (MMP), Bid truncation, and induction of death receptor 5. sulforaphane 24-27 TNF receptor superfamily member 10b Homo sapiens 204-220 29800641-0 2018 Isoalantolactone induces apoptosis through reactive oxygen species-dependent upregulation of death receptor 5 in human esophageal cancer cells. Reactive Oxygen Species 43-66 TNF receptor superfamily member 10b Homo sapiens 93-109 29792947-4 2018 Combined treatment with SFN and TRAIL (SFN/TRAIL) significantly induced apoptosis concomitant with activation of caspases, loss of mitochondrial membrane potential (MMP), Bid truncation, and induction of death receptor 5. sulforaphane 39-42 TNF receptor superfamily member 10b Homo sapiens 204-220 29800641-5 2018 Treatment with isoalantolactone activated caspases-3, -7, and -10, and upregulated death receptor (DR)5. isoalantolactone 15-31 TNF receptor superfamily member 10b Homo sapiens 83-103 29800641-6 2018 Furthermore, DR5 knockdown partially reversed the effect of isoalantolactone. isoalantolactone 60-76 TNF receptor superfamily member 10b Homo sapiens 13-16 29800641-12 2018 In conclusion, our results revealed that isoalantolactone induced apoptosis through the extrinsic pathway via upregulation of DR5 and elevation of ROS in human esophageal cancer cells. isoalantolactone 41-57 TNF receptor superfamily member 10b Homo sapiens 126-129 30111400-6 2018 CONCLUSION: Osthole can sensitize HL-60 cells to TRAIL-induced apoptosis, which may be related with the activation of mitochondrial pathways and up-regulation of DR5. osthol 12-19 TNF receptor superfamily member 10b Homo sapiens 162-165 30004456-0 2018 Garcinol Enhances TRAIL-Induced Apoptotic Cell Death through Up-Regulation of DR5 and Down-Regulation of c-FLIP Expression. garcinol 0-8 TNF receptor superfamily member 10b Homo sapiens 78-81 30111400-5 2018 RESULTS: The combined treatment (100micromol/L Osthole + 40 ng/ml TRAIL) of HL-60 cells for 48 h induced an apoptotic rate of (33.9+-2.7) %, which was significantly higher than that of cells treated with Osthole or TRAIL alone (P<0.05); at the same time, the combined treatment promoted the decrease of MMP and the expression rate of BCL-2/BAX, and potentiated the expression of DR5 and Caspase-3,-8,-9 activity. osthol 47-54 TNF receptor superfamily member 10b Homo sapiens 382-385 29970185-14 2018 Knockdown of DR4 and DR5 with siRNAs increased cell survival rate and decreased the expression of cleaved-PARP induced by the combination of azithromycin and TRAIL. Azithromycin 141-153 TNF receptor superfamily member 10b Homo sapiens 21-24 29970185-16 2018 CONCLUSION: The synergistic antitumor effect of azithromycin and TRAIL mainly relies on the up-regulations of DR4 and DR5, which in turn result from LC-3B-involved autophagy inhibition. Azithromycin 48-60 TNF receptor superfamily member 10b Homo sapiens 118-121 29970185-13 2018 Azithromycin increased the expressions of DR4, DR5, p62 and LC-3B proteins and potentiated induction of apoptosis by TRAIL. Azithromycin 0-12 TNF receptor superfamily member 10b Homo sapiens 47-50 30004456-6 2018 Garcinol plus TRAIL induced up-regulation of DR5 and down-regulation of c-FLIP expression at post-translational levels. garcinol 0-8 TNF receptor superfamily member 10b Homo sapiens 45-48 30004456-7 2018 Furthermore, knock-down of DR5 by siRNA and ectopic expression of c-FLIP blocked apoptotic cell death induced by garcinol plus TRAIL. garcinol 113-121 TNF receptor superfamily member 10b Homo sapiens 27-30 29694835-3 2018 In this research, we showed for the first time that candesartan treatment significantly sensitized human lung adenocarcinoma cells to Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis by targeting TRAIL-DR5. candesartan 52-63 TNF receptor superfamily member 10b Homo sapiens 236-239 29694835-6 2018 The molecular mechanisms underlying candesartan-induced TRAIL-mediated apoptosis involved the downstream of AMPK phosphorylation resulting inhibition of autophagy flux, recruitment of death receptor 5 (DR5) and activation of apoptotic caspase cascade. candesartan 36-47 TNF receptor superfamily member 10b Homo sapiens 184-200 29694835-6 2018 The molecular mechanisms underlying candesartan-induced TRAIL-mediated apoptosis involved the downstream of AMPK phosphorylation resulting inhibition of autophagy flux, recruitment of death receptor 5 (DR5) and activation of apoptotic caspase cascade. candesartan 36-47 TNF receptor superfamily member 10b Homo sapiens 202-205 29615720-0 2018 Involvement of AMP-activated protein kinase and Death Receptor 5 in TRAIL-Berberine-induced apoptosis of cancer cells. Berberine 74-83 TNF receptor superfamily member 10b Homo sapiens 48-64 29891932-4 2018 We demonstrate that in vivo accumulation or exogenous application of MEcPP alters the expression of two auxin reporters, DR5:GFP and DII-VENUS, and reduces the abundance of the auxin-efflux carrier PIN-FORMED1 (PIN1) at the plasma membrane. methyl-D-erythritol 2,4-cyclodiphosphate 69-74 TNF receptor superfamily member 10b Homo sapiens 121-124 29768226-3 2018 Three conserved domains of Death receptor (DR5) protein extracellular domain, which are fortified cysteine, were chosen and chemically synthesised. Cysteine 98-106 TNF receptor superfamily member 10b Homo sapiens 43-46 29764340-4 2018 A human apoptosis protein array kit showed that death receptor 5 may be involved in silymarin-induced apoptosis, which was also shown through western blotting, immunocytochemistry, and reverse transcription-polymerase chain reaction. Silymarin 84-93 TNF receptor superfamily member 10b Homo sapiens 48-64 29615720-7 2018 These showed induction of several genes including TNFRSF10B (expresses DR5) and Harakiri following BBR treatment, which were further validated by qPCR analysis. Berberine 99-102 TNF receptor superfamily member 10b Homo sapiens 50-59 29549167-0 2018 Tetrandrine (TET) Induces Death Receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5) and Sensitizes Prostate Cancer Cells to TRAIL-Induced Apoptosis. tetrandrine 0-11 TNF receptor superfamily member 10b Homo sapiens 69-77 29549167-0 2018 Tetrandrine (TET) Induces Death Receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5) and Sensitizes Prostate Cancer Cells to TRAIL-Induced Apoptosis. tetrandrine 0-11 TNF receptor superfamily member 10b Homo sapiens 79-82 29549167-0 2018 Tetrandrine (TET) Induces Death Receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5) and Sensitizes Prostate Cancer Cells to TRAIL-Induced Apoptosis. tetrandrine 13-16 TNF receptor superfamily member 10b Homo sapiens 69-77 29549167-0 2018 Tetrandrine (TET) Induces Death Receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5) and Sensitizes Prostate Cancer Cells to TRAIL-Induced Apoptosis. tetrandrine 13-16 TNF receptor superfamily member 10b Homo sapiens 79-82 29549167-4 2018 We demonstrate here that small-molecule tetrandrine (TET) potentially sensitizes previously resistant (LNCaP and C4-2B cells) and mildly sensitive (PC3 cells) prostate cancer cells to TRAIL-induced apoptosis, and they do so by upregulating mRNA expression and protein levels of death receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5). tetrandrine 40-51 TNF receptor superfamily member 10b Homo sapiens 321-329 29549167-4 2018 We demonstrate here that small-molecule tetrandrine (TET) potentially sensitizes previously resistant (LNCaP and C4-2B cells) and mildly sensitive (PC3 cells) prostate cancer cells to TRAIL-induced apoptosis, and they do so by upregulating mRNA expression and protein levels of death receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5). tetrandrine 40-51 TNF receptor superfamily member 10b Homo sapiens 331-334 29549167-4 2018 We demonstrate here that small-molecule tetrandrine (TET) potentially sensitizes previously resistant (LNCaP and C4-2B cells) and mildly sensitive (PC3 cells) prostate cancer cells to TRAIL-induced apoptosis, and they do so by upregulating mRNA expression and protein levels of death receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5). tetrandrine 53-56 TNF receptor superfamily member 10b Homo sapiens 321-329 29549167-4 2018 We demonstrate here that small-molecule tetrandrine (TET) potentially sensitizes previously resistant (LNCaP and C4-2B cells) and mildly sensitive (PC3 cells) prostate cancer cells to TRAIL-induced apoptosis, and they do so by upregulating mRNA expression and protein levels of death receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5). tetrandrine 53-56 TNF receptor superfamily member 10b Homo sapiens 331-334 29549167-6 2018 We show that double knockdown of DR4 and DR5 abrogated the apoptotic effects of TET and TRAIL. tetrandrine 80-83 TNF receptor superfamily member 10b Homo sapiens 41-44 29549167-7 2018 We also demonstrate that TET-induced DR4 and DR5 expression is independent of p53 status. tetrandrine 25-28 TNF receptor superfamily member 10b Homo sapiens 45-48 29615720-7 2018 These showed induction of several genes including TNFRSF10B (expresses DR5) and Harakiri following BBR treatment, which were further validated by qPCR analysis. Berberine 99-102 TNF receptor superfamily member 10b Homo sapiens 71-74 29599323-6 2018 To determine the pathway and relevant components by which gallic acid-induced apoptosis is mediated through, cells were transfected with siRNA (Fas, FasL, DR5, p53) using Lipofectamine 2000. Gallic Acid 58-69 TNF receptor superfamily member 10b Homo sapiens 155-158 29599323-8 2018 Gallic acid induced up-regulation of Fas, FasL, and DR5 expression in AGS cells. Gallic Acid 0-11 TNF receptor superfamily member 10b Homo sapiens 52-55 29599323-9 2018 Transfection of cells with Fas, FasL, or DR5 siRNA reduced gallic acid-induced cell death. Gallic Acid 59-70 TNF receptor superfamily member 10b Homo sapiens 41-44 29599323-10 2018 In addition, p53 was shown to be involved in gallic acid-mediated Fas, FasL, and DR5 expression as well as cell apoptosis in AGS cells. Gallic Acid 45-56 TNF receptor superfamily member 10b Homo sapiens 81-84 29556305-6 2018 The present data indicate that Taxol may enhance the pro-apoptotic effects of TRAIL overexpression in HeLa cells by increasing cleaved caspase-3 and DR5 expression levels and decreasing Bcl-2 expression levels. Paclitaxel 31-36 TNF receptor superfamily member 10b Homo sapiens 149-152 29300929-8 2018 Furthermore, bortezomib and TRAIL synergistically induced reactive oxygen species (ROS) generation and caused death receptor 5 upregulation through activation of the extracellular signal-regulated kinase-C/EBP homologous protein signaling cascade. Bortezomib 13-23 TNF receptor superfamily member 10b Homo sapiens 110-126 29744288-0 2018 Dasatinib promotes TRAIL-mediated apoptosis by upregulating CHOP-dependent death receptor 5 in gastric cancer. Dasatinib 0-9 TNF receptor superfamily member 10b Homo sapiens 75-91 29744288-7 2018 Moreover, increased DR5 expression facilitated dasatinib-induced apoptosis; the dasatinib-induced increase in DR5 expression was mediated by CCAAT/enhancer-binding protein homologous protein (CHOP). Dasatinib 47-56 TNF receptor superfamily member 10b Homo sapiens 20-23 29744288-5 2018 In addition, we found that dasatinib increased the expression of death receptor 5 (DR5) in GC cells. Dasatinib 27-36 TNF receptor superfamily member 10b Homo sapiens 65-81 29744288-7 2018 Moreover, increased DR5 expression facilitated dasatinib-induced apoptosis; the dasatinib-induced increase in DR5 expression was mediated by CCAAT/enhancer-binding protein homologous protein (CHOP). Dasatinib 80-89 TNF receptor superfamily member 10b Homo sapiens 110-113 29744288-5 2018 In addition, we found that dasatinib increased the expression of death receptor 5 (DR5) in GC cells. Dasatinib 27-36 TNF receptor superfamily member 10b Homo sapiens 83-86 29744288-8 2018 Furthermore, dasatinib also synergized with TRAIL to induce apoptosis via DR5 in GC cells. Dasatinib 13-22 TNF receptor superfamily member 10b Homo sapiens 74-77 29158005-9 2018 In the 1,25(OH)2D3 + HDAC2 overexpression group, the expressions of p53, Bax, DR5 and caspase 8 were significantly lower but the expression of Bcl-2 was significantly higher than those of the 1,25(OH)2D3 treatment group (P < 0.05). 25(oh) 9-15 TNF receptor superfamily member 10b Homo sapiens 78-81 29744288-9 2018 Our results show that dasatinib promoted TRAIL-mediated apoptosis via upregulation of CHOP-dependent DR5 expression in GC, suggesting that DR5 induction can be used as an indicator of dasatinib sensitivity. Dasatinib 22-31 TNF receptor superfamily member 10b Homo sapiens 101-104 29744288-9 2018 Our results show that dasatinib promoted TRAIL-mediated apoptosis via upregulation of CHOP-dependent DR5 expression in GC, suggesting that DR5 induction can be used as an indicator of dasatinib sensitivity. Dasatinib 22-31 TNF receptor superfamily member 10b Homo sapiens 139-142 29498673-4 2018 Remarkably, though, despite the fact that all membrane-bound TRAIL receptors harbor putative glycosylation sites, only pro-apoptotic signaling through DR4 and DR5 has, so far, been found to be regulated by N- and O-glycosylation, respectively. Nitrogen 206-207 TNF receptor superfamily member 10b Homo sapiens 159-162 29278689-7 2018 TRAIL-induced apoptosis, enhanced by the inhibitor of the Bcl-2 family ABT-737, or by the translation inhibitor homoharringtonine, proceeded in both cell lines predominantly via the DR5 receptor. Homoharringtonine 112-129 TNF receptor superfamily member 10b Homo sapiens 182-185 29158005-9 2018 In the 1,25(OH)2D3 + HDAC2 overexpression group, the expressions of p53, Bax, DR5 and caspase 8 were significantly lower but the expression of Bcl-2 was significantly higher than those of the 1,25(OH)2D3 treatment group (P < 0.05). Calcitriol 7-18 TNF receptor superfamily member 10b Homo sapiens 78-81 29435013-0 2018 Enhancement of cisplatin cytotoxicity by Retigeric acid B involves blocking DNA repair and activating DR5 in prostate cancer cells. Cisplatin 15-24 TNF receptor superfamily member 10b Homo sapiens 102-105 29435013-6 2018 Concurrently, the proapoptotic protein death receptor 5 (DR5) was activated by RAB, which also enhanced the chemotherapeutic response of CDDP. Cisplatin 137-141 TNF receptor superfamily member 10b Homo sapiens 39-55 29435013-8 2018 The results of the present study identified that RAB functioned synergistically with CDDP to increase the efficacy of CDDP by inhibiting DNA damage repair and activating DR5, suggesting the mechanistic basis for the antitumor effect of RAB in combination with current chemotherapeutics. retigeric acid B 49-52 TNF receptor superfamily member 10b Homo sapiens 170-173 29435013-6 2018 Concurrently, the proapoptotic protein death receptor 5 (DR5) was activated by RAB, which also enhanced the chemotherapeutic response of CDDP. Cisplatin 137-141 TNF receptor superfamily member 10b Homo sapiens 57-60 29435013-7 2018 Knockdown of DR5 partially blocked RAB-CDDP synergism, suggesting the crucial involvement of DR5 in this event. retigeric acid B 35-38 TNF receptor superfamily member 10b Homo sapiens 13-16 29435013-7 2018 Knockdown of DR5 partially blocked RAB-CDDP synergism, suggesting the crucial involvement of DR5 in this event. retigeric acid B 35-38 TNF receptor superfamily member 10b Homo sapiens 93-96 29435013-7 2018 Knockdown of DR5 partially blocked RAB-CDDP synergism, suggesting the crucial involvement of DR5 in this event. Cisplatin 39-43 TNF receptor superfamily member 10b Homo sapiens 13-16 29435013-7 2018 Knockdown of DR5 partially blocked RAB-CDDP synergism, suggesting the crucial involvement of DR5 in this event. Cisplatin 39-43 TNF receptor superfamily member 10b Homo sapiens 93-96 29435013-8 2018 The results of the present study identified that RAB functioned synergistically with CDDP to increase the efficacy of CDDP by inhibiting DNA damage repair and activating DR5, suggesting the mechanistic basis for the antitumor effect of RAB in combination with current chemotherapeutics. Cisplatin 85-89 TNF receptor superfamily member 10b Homo sapiens 170-173 29435013-8 2018 The results of the present study identified that RAB functioned synergistically with CDDP to increase the efficacy of CDDP by inhibiting DNA damage repair and activating DR5, suggesting the mechanistic basis for the antitumor effect of RAB in combination with current chemotherapeutics. Cisplatin 118-122 TNF receptor superfamily member 10b Homo sapiens 170-173 29531826-14 2018 Upregulation of DR5 is involved in BIX plus TRAIL-mediated apoptosis. 2-(3,4-dihydroxyphenyl)-2-oxoethyl thiocyanate 35-38 TNF receptor superfamily member 10b Homo sapiens 16-19 29531826-0 2018 BIX-01294 sensitizes renal cancer Caki cells to TRAIL-induced apoptosis through downregulation of survivin expression and upregulation of DR5 expression. BIX 01294 0-9 TNF receptor superfamily member 10b Homo sapiens 138-141 29272788-0 2018 Combination of novel DR5 targeting agonistic scFv antibody TR2-3 with cisplatin shows enhanced synergistic antitumor activity in vitro and in vivo. Cisplatin 70-79 TNF receptor superfamily member 10b Homo sapiens 21-24 29531826-7 2018 Furthermore, BIX induced upregulation of DR5 expression at the transcriptional levels, and knockdown of DR5 expression using small interfering RNAs (siRNAs) markedly attenuated BIX and TRAIL-induced apoptosis. 2-(3,4-dihydroxyphenyl)-2-oxoethyl thiocyanate 13-16 TNF receptor superfamily member 10b Homo sapiens 41-44 29531826-7 2018 Furthermore, BIX induced upregulation of DR5 expression at the transcriptional levels, and knockdown of DR5 expression using small interfering RNAs (siRNAs) markedly attenuated BIX and TRAIL-induced apoptosis. 2-(3,4-dihydroxyphenyl)-2-oxoethyl thiocyanate 177-180 TNF receptor superfamily member 10b Homo sapiens 104-107 29531826-11 2018 Taken together, these results suggest that BIX facilitates TRAIL-mediated apoptosis via downregulation of survivin and upregulation of DR5 expression in renal carcinoma Caki cells. 2-(3,4-dihydroxyphenyl)-2-oxoethyl thiocyanate 43-46 TNF receptor superfamily member 10b Homo sapiens 135-138 29434187-6 2018 Activation of the amino-acid-sensing kinase general control nonderepressible 2 (GCN2) upon glutamine deprivation is responsible for TRAIL-R2 upregulation through a signaling pathway involving ATF4 and CHOP transcription factors. Glutamine 91-100 TNF receptor superfamily member 10b Homo sapiens 132-140 29272788-8 2018 In addition, treatment with cisplatin alone for 24 h resulted in significantly up-regulating the mRNA and protein levels of DR5 in both COLO205 and MDA-MB-231 cell lines by q-PCR and Western blot assay. Cisplatin 28-37 TNF receptor superfamily member 10b Homo sapiens 124-127 29272788-10 2018 These results indicated that cisplatin sensitized COLO205 and MDA-MB-231 cancer cells to TR2-3-mediated apoptosis by up-regulation of DR5 expression. Cisplatin 29-38 TNF receptor superfamily member 10b Homo sapiens 134-137 29434473-6 2018 Quercetin was able to sensitize rhTRAIL-resistant breast cancers to rhTRAIL-induced apoptosis as detected by Western blotting through the proteasome-mediated degradation of c-FLIPL and through the upregulation of DR5 expression transcriptionally. Quercetin 0-9 TNF receptor superfamily member 10b Homo sapiens 213-216 29272788-12 2018 CONCLUSIONS: Our findings suggest that cisplatin enhanced the antitumor activity of TR2-3 in COLO205 and MDA-MB-231 cancer cells through up-regulation of DR5 expression. Cisplatin 39-48 TNF receptor superfamily member 10b Homo sapiens 154-157 29535810-13 2018 Our results showed that silibinin enhanced TRAIL-induced apoptosis by upregulating DR5 expression through the ROS-ER stress-CaMKII-Sp1 axis. Silybin 24-33 TNF receptor superfamily member 10b Homo sapiens 83-86 29162448-0 2018 Intracellular cholesterol level regulates sensitivity of glioblastoma cells against temozolomide-induced cell death by modulation of caspase-8 activation via death receptor 5-accumulation and activation in the plasma membrane lipid raft. Cholesterol 14-25 TNF receptor superfamily member 10b Homo sapiens 158-174 29162448-0 2018 Intracellular cholesterol level regulates sensitivity of glioblastoma cells against temozolomide-induced cell death by modulation of caspase-8 activation via death receptor 5-accumulation and activation in the plasma membrane lipid raft. Temozolomide 84-96 TNF receptor superfamily member 10b Homo sapiens 158-174 29162448-7 2018 TMZ without or with MbetaCD and/or Chol caused accumulation of DR5 into the plasma membrane lipid raft and formed a complex with caspase-8, an extrinsic caspase cascade inducer, reflected in the induction of cell death. Temozolomide 0-3 TNF receptor superfamily member 10b Homo sapiens 63-66 29162448-7 2018 TMZ without or with MbetaCD and/or Chol caused accumulation of DR5 into the plasma membrane lipid raft and formed a complex with caspase-8, an extrinsic caspase cascade inducer, reflected in the induction of cell death. methyl-beta-cyclodextrin 20-27 TNF receptor superfamily member 10b Homo sapiens 63-66 29162448-7 2018 TMZ without or with MbetaCD and/or Chol caused accumulation of DR5 into the plasma membrane lipid raft and formed a complex with caspase-8, an extrinsic caspase cascade inducer, reflected in the induction of cell death. Cholesterol 35-39 TNF receptor superfamily member 10b Homo sapiens 63-66 29162448-8 2018 In addition, treatment with caspase-8 inhibitor or knockdown of DR5 dramatically suppressed U251-Con cell death induced by combination treatment with TMZ, MbetaCD, and Chol. Temozolomide 150-153 TNF receptor superfamily member 10b Homo sapiens 64-67 29162448-8 2018 In addition, treatment with caspase-8 inhibitor or knockdown of DR5 dramatically suppressed U251-Con cell death induced by combination treatment with TMZ, MbetaCD, and Chol. methyl-beta-cyclodextrin 155-162 TNF receptor superfamily member 10b Homo sapiens 64-67 29162448-8 2018 In addition, treatment with caspase-8 inhibitor or knockdown of DR5 dramatically suppressed U251-Con cell death induced by combination treatment with TMZ, MbetaCD, and Chol. Cholesterol 168-172 TNF receptor superfamily member 10b Homo sapiens 64-67 29535810-0 2018 Silibinin sensitizes TRAIL-mediated apoptosis by upregulating DR5 through ROS-induced endoplasmic reticulum stress-Ca2+-CaMKII-Sp1 pathway. Silybin 0-9 TNF receptor superfamily member 10b Homo sapiens 62-65 29535810-0 2018 Silibinin sensitizes TRAIL-mediated apoptosis by upregulating DR5 through ROS-induced endoplasmic reticulum stress-Ca2+-CaMKII-Sp1 pathway. Reactive Oxygen Species 74-77 TNF receptor superfamily member 10b Homo sapiens 62-65 29535810-4 2018 Silibinin also triggered TRAIL-induced apoptosis in A549 cells through upregulation of death receptor 5 (DR5). Silybin 0-9 TNF receptor superfamily member 10b Homo sapiens 87-103 29535810-4 2018 Silibinin also triggered TRAIL-induced apoptosis in A549 cells through upregulation of death receptor 5 (DR5). Silybin 0-9 TNF receptor superfamily member 10b Homo sapiens 105-108 29535810-5 2018 Pretreatment with DR5/Fc chimeric protein and DR5-targeted small interfering RNA (siRNA) significantly blocked silibinin/TRAIL-mediated apoptosis in A549 cells. Silybin 111-120 TNF receptor superfamily member 10b Homo sapiens 18-21 29535810-5 2018 Pretreatment with DR5/Fc chimeric protein and DR5-targeted small interfering RNA (siRNA) significantly blocked silibinin/TRAIL-mediated apoptosis in A549 cells. Silybin 111-120 TNF receptor superfamily member 10b Homo sapiens 46-49 29535810-6 2018 Furthermore, silibinin increased the production of reactive oxygen species (ROS), which led to the induction of TRAIL-mediated apoptosis through DR5 upregulation. Silybin 13-22 TNF receptor superfamily member 10b Homo sapiens 145-148 29162448-10 2018 These findings suggest that intracellular cholesterol level affects TMZ treatment of GBM mediated via a DR5-caspase-8 mechanism. Cholesterol 42-53 TNF receptor superfamily member 10b Homo sapiens 104-107 29162448-10 2018 These findings suggest that intracellular cholesterol level affects TMZ treatment of GBM mediated via a DR5-caspase-8 mechanism. Temozolomide 68-71 TNF receptor superfamily member 10b Homo sapiens 104-107 30257253-0 2018 miR-128 Targets the SIRT1/ROS/DR5 Pathway to Sensitize Colorectal Cancer to TRAIL-Induced Apoptosis. mir-128 0-7 TNF receptor superfamily member 10b Homo sapiens 30-33 30257253-12 2018 This increase of ROS subsequently induced DR5 expression, and thus increased TRAIL-induced apoptosis in CRC cells. Reactive Oxygen Species 17-20 TNF receptor superfamily member 10b Homo sapiens 42-45 29164887-9 2017 Moreover, we found that PTER induced the expression of DR4 and DR5 through the reactive oxygen species (ROS)/ endoplasmic reticulum (ER) stress/ERK 1/2 and p38/C/EBP-homologous protein (CHOP) signaling pathways. Reactive Oxygen Species 79-102 TNF receptor superfamily member 10b Homo sapiens 63-66 29164887-9 2017 Moreover, we found that PTER induced the expression of DR4 and DR5 through the reactive oxygen species (ROS)/ endoplasmic reticulum (ER) stress/ERK 1/2 and p38/C/EBP-homologous protein (CHOP) signaling pathways. Reactive Oxygen Species 104-107 TNF receptor superfamily member 10b Homo sapiens 63-66 29164887-10 2017 Overall, our results showed that PTER potentiated TRAIL-induced apoptosis via ROS-mediated CHOP activation leading to the expression of DR4 and DR5. Reactive Oxygen Species 78-81 TNF receptor superfamily member 10b Homo sapiens 144-147 29535810-6 2018 Furthermore, silibinin increased the production of reactive oxygen species (ROS), which led to the induction of TRAIL-mediated apoptosis through DR5 upregulation. Reactive Oxygen Species 51-74 TNF receptor superfamily member 10b Homo sapiens 145-148 29535810-13 2018 Our results showed that silibinin enhanced TRAIL-induced apoptosis by upregulating DR5 expression through the ROS-ER stress-CaMKII-Sp1 axis. Reactive Oxygen Species 110-113 TNF receptor superfamily member 10b Homo sapiens 83-86 29535810-6 2018 Furthermore, silibinin increased the production of reactive oxygen species (ROS), which led to the induction of TRAIL-mediated apoptosis through DR5 upregulation. Reactive Oxygen Species 76-79 TNF receptor superfamily member 10b Homo sapiens 145-148 29072615-8 2017 Collectively, our data show that LSS-11 is a potent naphthalimide-based chemosensitizer that could enhance cell death in paclitaxel-resistant lung cancer cells through the DR5/PARP1 pathway and STAT3/MDR1/MRP1 STAT3 inhibition. LSS-11 33-39 TNF receptor superfamily member 10b Homo sapiens 172-175 29535810-11 2018 Ca2+/calmodulin-dependent protein kinase (CaMKII) inhibitor, K252a, blocked silibinin/TRAIL-induced DR5 expression along with TRAIL-mediated apoptosis. Silybin 76-85 TNF receptor superfamily member 10b Homo sapiens 100-103 29535810-12 2018 Accordingly, we showed that ROS/ER stress-induced CaMKII activated Sp1, which is an important transcription factor for DR5 expression. Reactive Oxygen Species 28-31 TNF receptor superfamily member 10b Homo sapiens 119-122 29296202-5 2017 Ciglitazone kills cervical cancer cells by activating death receptor signalling pathway, caspase cascade and BH3 interacting-domain death agonist (Bid) cleavage through the up-regulation of Death Receptor 4 (DR4)/DR5 and soluble and membrane-bound TNF related apoptosis inducing ligand (TRAIL). ciglitazone 0-11 TNF receptor superfamily member 10b Homo sapiens 213-216 29070784-10 2017 RESULTS Kaempferol enhanced apoptosis and drastically up-regulated DR4, DR5, CHOP, JNK, ERK1/2, p38 and apoptotic protein expression with decline in the expression of anti-apoptotic proteins. kaempferol 8-18 TNF receptor superfamily member 10b Homo sapiens 72-75 29070784-12 2017 CONCLUSIONS Kaempferol sensitized ovarian cancer cells to TRAIL-induced apoptosis via up-regulation of DR4 and DR5 through ERK/JNK/CHOP pathways. kaempferol 12-22 TNF receptor superfamily member 10b Homo sapiens 111-114 29297126-6 2017 The cytotoxicity of DR5-B variant is significantly higher compared to wild-type TRAIL in combination with cisplatin in 9 of 12 tumor cell lines. Cisplatin 106-115 TNF receptor superfamily member 10b Homo sapiens 20-23 29072615-8 2017 Collectively, our data show that LSS-11 is a potent naphthalimide-based chemosensitizer that could enhance cell death in paclitaxel-resistant lung cancer cells through the DR5/PARP1 pathway and STAT3/MDR1/MRP1 STAT3 inhibition. Naphthalimides 52-65 TNF receptor superfamily member 10b Homo sapiens 172-175 29072615-8 2017 Collectively, our data show that LSS-11 is a potent naphthalimide-based chemosensitizer that could enhance cell death in paclitaxel-resistant lung cancer cells through the DR5/PARP1 pathway and STAT3/MDR1/MRP1 STAT3 inhibition. Paclitaxel 121-131 TNF receptor superfamily member 10b Homo sapiens 172-175 28702823-0 2017 Etoposide and doxorubicin enhance the sensitivity of triple negative breast cancers through modulation of TRAIL-DR5 axis. Doxorubicin 14-25 TNF receptor superfamily member 10b Homo sapiens 112-115 28702823-7 2017 The binding energy for TRAIL-DR5 complexation in the ternary complexes containing ET (-111.08 kcal/mol) and DOX (-76.35 kcal/mol) were higher than reported binding energy of binary complex (-53.70 kcal/mol). Doxorubicin 108-111 TNF receptor superfamily member 10b Homo sapiens 29-32 28702823-0 2017 Etoposide and doxorubicin enhance the sensitivity of triple negative breast cancers through modulation of TRAIL-DR5 axis. Etoposide 0-9 TNF receptor superfamily member 10b Homo sapiens 112-115 28702823-12 2017 Thus, data suggests ET and DOX act as DR5 agonistic ligands and enhance the cellular apoptosis in TNBC. Doxorubicin 27-30 TNF receptor superfamily member 10b Homo sapiens 38-41 28702823-2 2017 Recently, we reported the molecular level details for the modulation of TRAIL-DR5 axis by quinacrine (QC) in breast cancer cells. Quinacrine 90-100 TNF receptor superfamily member 10b Homo sapiens 78-81 28702823-3 2017 In this work, the DR5 mediated anticancer potential of topoisomerase inhibitor etoposide (ET) and doxorubicin (DOX) against TNBC has been evaluated. Etoposide 79-88 TNF receptor superfamily member 10b Homo sapiens 18-21 28702823-3 2017 In this work, the DR5 mediated anticancer potential of topoisomerase inhibitor etoposide (ET) and doxorubicin (DOX) against TNBC has been evaluated. Etoposide 90-92 TNF receptor superfamily member 10b Homo sapiens 18-21 28702823-3 2017 In this work, the DR5 mediated anticancer potential of topoisomerase inhibitor etoposide (ET) and doxorubicin (DOX) against TNBC has been evaluated. Doxorubicin 98-109 TNF receptor superfamily member 10b Homo sapiens 18-21 28702823-3 2017 In this work, the DR5 mediated anticancer potential of topoisomerase inhibitor etoposide (ET) and doxorubicin (DOX) against TNBC has been evaluated. Doxorubicin 111-114 TNF receptor superfamily member 10b Homo sapiens 18-21 28702823-4 2017 ET and DOX enhanced the DR5 expression in TNBC cells, whereas non-topoisomerase inhibitors pifithrin-alpha (PIF) and dexamethasone (DEX) failed to do so. Etoposide 0-2 TNF receptor superfamily member 10b Homo sapiens 24-27 28702823-4 2017 ET and DOX enhanced the DR5 expression in TNBC cells, whereas non-topoisomerase inhibitors pifithrin-alpha (PIF) and dexamethasone (DEX) failed to do so. Doxorubicin 7-10 TNF receptor superfamily member 10b Homo sapiens 24-27 28467689-5 2017 Vernodalol increased the expression of death receptor (DR) 5, and silencing of DR5 with a small interfering RNA (siRNA) reduced the effect of vernodalol on TRAIL-mediated apoptosis. vernodalol 0-10 TNF receptor superfamily member 10b Homo sapiens 39-60 28631177-6 2017 The expression of TRAIL and TRAIL-R2 in SAA patients was significantly decreased compared with controls; however, there was no statistical difference in TRAIL mRNA expression between the two groups. saa 40-43 TNF receptor superfamily member 10b Homo sapiens 28-36 28467689-5 2017 Vernodalol increased the expression of death receptor (DR) 5, and silencing of DR5 with a small interfering RNA (siRNA) reduced the effect of vernodalol on TRAIL-mediated apoptosis. vernodalol 142-152 TNF receptor superfamily member 10b Homo sapiens 79-82 28467689-8 2017 In addition, a c-Jun N-terminal kinase (JNK) inhibitor blocked the vernodalol-induced up-regulation of DR5, indicating that the effect depended on JNK activation. vernodalol 67-77 TNF receptor superfamily member 10b Homo sapiens 103-106 28467689-11 2017 Based on our results, vernodalol enhanced TRAIL-induced apoptosis by down-regulating Mcl-1 and up-regulating DR5, and the effects of DR5 depended on JNK activation and CHOP induction. vernodalol 22-32 TNF receptor superfamily member 10b Homo sapiens 109-112 28467689-11 2017 Based on our results, vernodalol enhanced TRAIL-induced apoptosis by down-regulating Mcl-1 and up-regulating DR5, and the effects of DR5 depended on JNK activation and CHOP induction. vernodalol 22-32 TNF receptor superfamily member 10b Homo sapiens 133-136 28918048-0 2017 miR-133a Promotes TRAIL Resistance in Glioblastoma via Suppressing Death Receptor 5 and Activating NF-kappaB Signaling. mir-133a 0-8 TNF receptor superfamily member 10b Homo sapiens 67-83 28918048-3 2017 In this study, a negative correlation between DR5 expression and TRAIL resistance was observed, and miR-133a was predicted to be the most promising candidate to suppress DR5 expression. mir-133a 100-108 TNF receptor superfamily member 10b Homo sapiens 170-173 28918048-7 2017 In conclusion, our data demonstrate that miR-133a promotes TRAIL resistance in glioblastoma by suppressing DR5 expression and activating NF-kappaB signaling. mir-133a 41-49 TNF receptor superfamily member 10b Homo sapiens 107-110 28793767-3 2017 In our study, 20(S)-Rh2 induced the expression of orphan nuclear receptor Nur77 and death receptor proteins Fas, FasL, DR5, and TRAIL, as well as the cleavage of caspase 8 and caspase 3 in HL-60 cells. rh2 20-23 TNF receptor superfamily member 10b Homo sapiens 119-122 28092099-6 2017 Results show that upon DR5 activation, CaM was recruited into DR5-mediated DISC in a calcium dependent manner. Calcium 85-92 TNF receptor superfamily member 10b Homo sapiens 23-26 28899403-8 2017 Additionally, DR5 in patients with AOSD are associated with macrophage activation syndrome (MAS) and steroid pulse therapy. Steroids 101-108 TNF receptor superfamily member 10b Homo sapiens 14-17 28767099-0 2017 Fisetin Induces Apoptosis Through p53-Mediated Up-Regulation of DR5 Expression in Human Renal Carcinoma Caki Cells. fisetin 0-7 TNF receptor superfamily member 10b Homo sapiens 64-67 28471808-0 2017 3-Bromopyruvate enhances TRAIL-induced apoptosis in human nasopharyngeal carcinoma cells through CHOP-dependent upregulation of TRAIL-R2. bromopyruvate 0-15 TNF receptor superfamily member 10b Homo sapiens 128-136 29018571-1 2017 To investigate the ability of SAHA-induced TRAIL DR5-CTSB crosstalk to initiate the breast cancer autophagy, RTCA assay was performed to assess the effect of SAHA on breast cancer cells, and western blot and ELISA were used to verify the inductive effects on expression of CTSB. Vorinostat 30-34 TNF receptor superfamily member 10b Homo sapiens 49-52 29018571-4 2017 The distribution of LC3-II in TRAIL DR5-silenced breast cancer cells treated with SAHA was observed by immunofluorescence microscopy, the mRNA levels of autophagy-related genes were detected by RNA microarray, and the activity of autophagy-related signaling pathways was screened by MAPK antibody array. Vorinostat 82-86 TNF receptor superfamily member 10b Homo sapiens 36-39 29018571-6 2017 Western blot and ELISA results indicated that TRAIL DR5 was involved in the expression of CTSB in SAHA-induced breast cancer cells. Vorinostat 98-102 TNF receptor superfamily member 10b Homo sapiens 52-55 29018571-7 2017 Cell viability and apoptosis assays showed that the inactivation of TRAIL DR5 can significantly inhibit the effects of SAHA. Vorinostat 119-123 TNF receptor superfamily member 10b Homo sapiens 74-77 29018571-9 2017 While SAHA was added in the TRAIL-DR5 blocked cells, the distribution of LC3-II signal was dispersed, the intensity of fluorescence signal was weaker than that of SAHA alone. Vorinostat 6-10 TNF receptor superfamily member 10b Homo sapiens 34-37 29018571-12 2017 These results provide more evidence that SAHA may stimulate TRAIL DR5-CTSB crosstalk, influence the activity of downstream TOR signalling pathway mainly through the AKTs pathway, and initiate the autophagy of breast cancer cells. Vorinostat 41-45 TNF receptor superfamily member 10b Homo sapiens 66-69 28571773-0 2017 DATS sensitizes glioma cells to TRAIL-mediated apoptosis by up-regulation of death receptor 5 via ROS. ros 98-101 TNF receptor superfamily member 10b Homo sapiens 77-93 28571773-6 2017 In addition, DATS enhances TRAIL-induced apoptosis through the downregulation of anti-apoptotic protein (Mcl-1) and the upregulation of DR5 receptors through actions on the ROS- induced-p53. diallyl trisulfide 13-17 TNF receptor superfamily member 10b Homo sapiens 136-139 28571773-6 2017 In addition, DATS enhances TRAIL-induced apoptosis through the downregulation of anti-apoptotic protein (Mcl-1) and the upregulation of DR5 receptors through actions on the ROS- induced-p53. ros 173-176 TNF receptor superfamily member 10b Homo sapiens 136-139 28624529-0 2017 Cyclopamine sensitizes TRAIL-resistant gastric cancer cells to TRAIL-induced apoptosis via endoplasmic reticulum stress-mediated increase of death receptor 5 and survivin degradation. cyclopamine 0-11 TNF receptor superfamily member 10b Homo sapiens 141-157 28624529-3 2017 In this study, we show that cyclopamine sensitizes gastric cancer cells to TRAIL-induced apoptosis by elevating the expression of DR5. cyclopamine 28-39 TNF receptor superfamily member 10b Homo sapiens 130-133 28092099-6 2017 Results show that upon DR5 activation, CaM was recruited into DR5-mediated DISC in a calcium dependent manner. Calcium 85-92 TNF receptor superfamily member 10b Homo sapiens 62-65 28092099-11 2017 These results demonstrated CaM binding to DR5-mediated DISC in a calcium dependent manner and may identify CaM as a key regulator of DR5-mediated DISC formation for apoptosis in breast cancer. Calcium 65-72 TNF receptor superfamily member 10b Homo sapiens 42-45 28092099-11 2017 These results demonstrated CaM binding to DR5-mediated DISC in a calcium dependent manner and may identify CaM as a key regulator of DR5-mediated DISC formation for apoptosis in breast cancer. Calcium 65-72 TNF receptor superfamily member 10b Homo sapiens 133-136 28758908-9 2017 We discovered also that taxanes can increase the expression of death receptor TRAIL-R2 in PC3 prostate cancer cells. Taxoids 24-31 TNF receptor superfamily member 10b Homo sapiens 78-86 28511296-5 2017 In particular, the expression of the death receptors DR5 and FAS was significantly increased by MMPP treatment. 2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol 96-100 TNF receptor superfamily member 10b Homo sapiens 53-56 28067150-0 2017 Low-dose 5-fluorouracil sensitizes HepG2 cells to TRAIL through TRAIL receptor DR5 and survivin-dependent mechanisms. Fluorouracil 9-23 TNF receptor superfamily member 10b Homo sapiens 79-82 28414160-0 2017 Paraquat induces extrinsic pathway of apoptosis in A549 cells by induction of DR5 and repression of anti-apoptotic proteins, DDX3 and GSK3 expression. Paraquat 0-8 TNF receptor superfamily member 10b Homo sapiens 78-81 28746849-2 2017 Recent studies have proposed that activation of several tumor necrosis factor receptors, including Death Receptor 5, involves a scissorlike opening of the disulfide-linked transmembrane (TM) dimer. Disulfides 155-164 TNF receptor superfamily member 10b Homo sapiens 99-115 28120533-2 2017 We have recently demonstrated that nanovectorization of TRAIL with single-walled carbon nanotubes enhanced TRAIL affinity to DR5. Carbon 81-87 TNF receptor superfamily member 10b Homo sapiens 125-128 28476883-10 2017 We further demonstrated that HOTAIR regulates DR5 expression via the epigenetic regulator enhancer of zeste homolog 2 (EZH2) and that EZH2 controls histone H3 lysine 27 trimethylation on the DR5 gene. Lysine 159-165 TNF receptor superfamily member 10b Homo sapiens 191-194 28414160-4 2017 Additionally, PQ treatment caused an increase in DR5 (death receptor-5) and caspase-8 interaction, indicating formation of DISC (death-inducing signaling complex). Paraquat 14-16 TNF receptor superfamily member 10b Homo sapiens 49-52 28414160-4 2017 Additionally, PQ treatment caused an increase in DR5 (death receptor-5) and caspase-8 interaction, indicating formation of DISC (death-inducing signaling complex). Paraquat 14-16 TNF receptor superfamily member 10b Homo sapiens 54-70 28414160-6 2017 Moreover, PQ drastically increased DR5 expression and membrane localization. Paraquat 10-12 TNF receptor superfamily member 10b Homo sapiens 35-38 28414160-7 2017 Furthermore, PQ caused prominent concentration dependent reductions of DDX3 (the DEAD box protein-3) and GSK3 (glycogen synthase kinase-3) which can associate with DR5 and prevent DISC formation. Paraquat 13-15 TNF receptor superfamily member 10b Homo sapiens 164-167 28414160-8 2017 Additionally, PQ decreased DR5-DDX3 interaction, suggesting a reduction of DDX3/GSK3 anti-apoptotic complex. Paraquat 14-16 TNF receptor superfamily member 10b Homo sapiens 27-30 28414160-11 2017 Taken together, these results suggest that PQ may induce extrinsic pathway of apoptosis in A549 cells through upregulation of DR5 and repression of anti-apoptotic proteins, DDX3/GSK3 leading to reduction of anti-apoptotic complex. Paraquat 43-45 TNF receptor superfamily member 10b Homo sapiens 126-129 27965309-0 2017 Selective Targeting of Myeloid-Derived Suppressor Cells in Cancer Patients Using DS-8273a, an Agonistic TRAIL-R2 Antibody. DS-8273a 81-89 TNF receptor superfamily member 10b Homo sapiens 104-112 28067150-5 2017 The enhanced induction of cell death by the 5-FU/TRAIL combination was mediated by DR5 up-regulation and survivin down-regulation. Fluorouracil 44-48 TNF receptor superfamily member 10b Homo sapiens 83-86 28498435-6 2017 The effects of plumbagin on the expression of DR5, Bax and cFLIP could be partially abolished by the reactive oxygen species (ROS) scavenger NAC. Reactive Oxygen Species 101-124 TNF receptor superfamily member 10b Homo sapiens 46-49 28498435-6 2017 The effects of plumbagin on the expression of DR5, Bax and cFLIP could be partially abolished by the reactive oxygen species (ROS) scavenger NAC. Reactive Oxygen Species 126-129 TNF receptor superfamily member 10b Homo sapiens 46-49 28593135-5 2017 Gene expression studies revealed that cotreatment with melatonin and VPA triggered the up-regulation of certain genes related to apoptosis (TNFRSF10A and TNFRSF10B), autophagy (BECN, ATG3 and ATG5) and necrosis (MLKL, PARP-1 and RIPK1). Melatonin 55-64 TNF receptor superfamily member 10b Homo sapiens 154-163 28498480-0 2017 Miconazole induces apoptosis via the death receptor 5-dependent and mitochondrial-mediated pathways in human bladder cancer cells. Miconazole 0-10 TNF receptor superfamily member 10b Homo sapiens 37-53 28529596-6 2017 It was demonstrated that activity of the TRAIL-R2 (DR5) pathway was critical in the development of andrographolide-mediated rhTRAIL sensitization, since its inhibition significantly reduced the extent of apoptosis induced by the combination of rhTRAIL and andrographolide. andrographolide 99-114 TNF receptor superfamily member 10b Homo sapiens 41-49 28529596-6 2017 It was demonstrated that activity of the TRAIL-R2 (DR5) pathway was critical in the development of andrographolide-mediated rhTRAIL sensitization, since its inhibition significantly reduced the extent of apoptosis induced by the combination of rhTRAIL and andrographolide. andrographolide 99-114 TNF receptor superfamily member 10b Homo sapiens 51-54 28529596-6 2017 It was demonstrated that activity of the TRAIL-R2 (DR5) pathway was critical in the development of andrographolide-mediated rhTRAIL sensitization, since its inhibition significantly reduced the extent of apoptosis induced by the combination of rhTRAIL and andrographolide. andrographolide 256-271 TNF receptor superfamily member 10b Homo sapiens 41-49 28529596-6 2017 It was demonstrated that activity of the TRAIL-R2 (DR5) pathway was critical in the development of andrographolide-mediated rhTRAIL sensitization, since its inhibition significantly reduced the extent of apoptosis induced by the combination of rhTRAIL and andrographolide. andrographolide 256-271 TNF receptor superfamily member 10b Homo sapiens 51-54 28529596-8 2017 N-acetyl cysteine prevented andrographolide-mediated DR5 induction and the apoptotic effect induced by the combination of rhTRAIL and andrographolide. Acetylcysteine 0-17 TNF receptor superfamily member 10b Homo sapiens 53-56 28529596-8 2017 N-acetyl cysteine prevented andrographolide-mediated DR5 induction and the apoptotic effect induced by the combination of rhTRAIL and andrographolide. andrographolide 28-43 TNF receptor superfamily member 10b Homo sapiens 53-56 28529596-9 2017 Collectively, the present study demonstrated that andrographolide enhances TRAIL-induced apoptosis through induction of DR5 expression. andrographolide 50-65 TNF receptor superfamily member 10b Homo sapiens 120-123 28482915-0 2017 DR5 suppression induces sphingosine-1-phosphate-dependent TRAF2 polyubiquitination, leading to activation of JNK/AP-1 and promotion of cancer cell invasion. sphingosine 1-phosphate 24-47 TNF receptor superfamily member 10b Homo sapiens 0-3 28529565-8 2017 The knockdown of p53 by specific small interfering RNA resulted in the depletion of p53, and inhibited the OPA and PPA treatment-induced increases in p53, which led to a decrease in the expression of p21, DR5, Fas, PUMA and phosphatase and tensin homolog proteins. ppa 115-118 TNF receptor superfamily member 10b Homo sapiens 205-208 28459212-3 2017 Our previous article reported that small molecule YM155 induced apoptosis in pancreatic cancer cells via activation of death receptor 5. YM 155 50-55 TNF receptor superfamily member 10b Homo sapiens 119-135 28459212-7 2017 Western blotting analysis revealed that lexatumumab and PF573228 combination treatment increased death receptor 5 but decreased Bcl-xL expression. 6-(4-(3-(methylsulfonyl)benzylamino)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3,4-dihydroquinolin-2(1H)-one 56-64 TNF receptor superfamily member 10b Homo sapiens 97-113 28387244-6 2017 MT-6 treatment also induced mitochondrial membrane potential loss, JNK activation, and DR5 expression. Melatonin 0-4 TNF receptor superfamily member 10b Homo sapiens 87-90 28487767-0 2017 Inducement of apoptosis by cucurbitacin E, a tetracyclic triterpenes, through death receptor 5 in human cervical cancer cell lines. cucurbitacin E 27-41 TNF receptor superfamily member 10b Homo sapiens 78-94 28487767-0 2017 Inducement of apoptosis by cucurbitacin E, a tetracyclic triterpenes, through death receptor 5 in human cervical cancer cell lines. Triterpenes 57-68 TNF receptor superfamily member 10b Homo sapiens 78-94 28387244-7 2017 Cotreatment of cells with the JNK inhibitor SP600125 considerably attenuated MT-6-induced apoptosis, mitochondria membrane potential loss, DR5 upregulation, and suppression of cell viability. pyrazolanthrone 44-52 TNF receptor superfamily member 10b Homo sapiens 139-142 28202415-10 2017 Accordingly, TRAIL/Gln deprivation enhanced the expression of death receptor 5 (DR5) and transient knockdown of DR5 completely restored TRAIL/Gln deprivation-mediated apoptosis. Glutamine 19-22 TNF receptor superfamily member 10b Homo sapiens 62-78 28202415-10 2017 Accordingly, TRAIL/Gln deprivation enhanced the expression of death receptor 5 (DR5) and transient knockdown of DR5 completely restored TRAIL/Gln deprivation-mediated apoptosis. Glutamine 19-22 TNF receptor superfamily member 10b Homo sapiens 80-83 28202415-10 2017 Accordingly, TRAIL/Gln deprivation enhanced the expression of death receptor 5 (DR5) and transient knockdown of DR5 completely restored TRAIL/Gln deprivation-mediated apoptosis. Glutamine 142-145 TNF receptor superfamily member 10b Homo sapiens 62-78 28202415-10 2017 Accordingly, TRAIL/Gln deprivation enhanced the expression of death receptor 5 (DR5) and transient knockdown of DR5 completely restored TRAIL/Gln deprivation-mediated apoptosis. Glutamine 142-145 TNF receptor superfamily member 10b Homo sapiens 112-115 28270124-0 2017 Cyproterone acetate enhances TRAIL-induced androgen-independent prostate cancer cell apoptosis via up-regulation of death receptor 5. Cyproterone Acetate 0-19 TNF receptor superfamily member 10b Homo sapiens 116-132 28126677-4 2017 Furthermore, ATS exposure in TRAIL resistant cells resulted in significant increase of both DR4/DR5 expression and STAT3 inhibition thereby arbitrating TRAIL mediated apoptosis in HepG2 cells. Artesunate 13-16 TNF receptor superfamily member 10b Homo sapiens 96-99 28553347-6 2017 Mitomycin C upregulated the expression levels of Fas, DR4, DR5, cleaved caspase-8/9, Bax, Bim and cleaved caspase-3 proteins, and it downregulated Bcl-2 and Bcl-xL expression. Mitomycin 0-11 TNF receptor superfamily member 10b Homo sapiens 59-62 28300219-4 2017 We found that doxorubicin and related anthracycline agents (e.g., daunorubicin, idarubicin, and epirubicin) significantly upregulated the expression of death receptors (DRs) (TNFR1, Fas, DR4 and DR5) in iPS-derived cardiomyocytes at both protein and mRNA levels. Doxorubicin 14-25 TNF receptor superfamily member 10b Homo sapiens 195-198 28300219-4 2017 We found that doxorubicin and related anthracycline agents (e.g., daunorubicin, idarubicin, and epirubicin) significantly upregulated the expression of death receptors (DRs) (TNFR1, Fas, DR4 and DR5) in iPS-derived cardiomyocytes at both protein and mRNA levels. Anthracyclines 38-51 TNF receptor superfamily member 10b Homo sapiens 195-198 28300219-4 2017 We found that doxorubicin and related anthracycline agents (e.g., daunorubicin, idarubicin, and epirubicin) significantly upregulated the expression of death receptors (DRs) (TNFR1, Fas, DR4 and DR5) in iPS-derived cardiomyocytes at both protein and mRNA levels. Daunorubicin 66-78 TNF receptor superfamily member 10b Homo sapiens 195-198 28300219-4 2017 We found that doxorubicin and related anthracycline agents (e.g., daunorubicin, idarubicin, and epirubicin) significantly upregulated the expression of death receptors (DRs) (TNFR1, Fas, DR4 and DR5) in iPS-derived cardiomyocytes at both protein and mRNA levels. Idarubicin 80-90 TNF receptor superfamily member 10b Homo sapiens 195-198 28300219-4 2017 We found that doxorubicin and related anthracycline agents (e.g., daunorubicin, idarubicin, and epirubicin) significantly upregulated the expression of death receptors (DRs) (TNFR1, Fas, DR4 and DR5) in iPS-derived cardiomyocytes at both protein and mRNA levels. Epirubicin 96-106 TNF receptor superfamily member 10b Homo sapiens 195-198 28103535-3 2017 Our experiments showed that the combination of EGCG and Am80 synergistically induced both apoptosis in human lung cancer cell line PC-9 and up-regulated expressions of growth arrest and DNA damage-inducible gene 153 (GADD153), death receptor 5, and p21waf1 genes in the cells. epigallocatechin gallate 47-51 TNF receptor superfamily member 10b Homo sapiens 227-243 28103535-3 2017 Our experiments showed that the combination of EGCG and Am80 synergistically induced both apoptosis in human lung cancer cell line PC-9 and up-regulated expressions of growth arrest and DNA damage-inducible gene 153 (GADD153), death receptor 5, and p21waf1 genes in the cells. tamibarotene 56-60 TNF receptor superfamily member 10b Homo sapiens 227-243 28270124-9 2017 Further investigation of the TRAIL-induced apoptosis pathway revealed that cyproterone acetate exerted its effect by selectively increasing death receptor 5 (DR5) mRNA and protein expression. Cyproterone Acetate 75-94 TNF receptor superfamily member 10b Homo sapiens 140-156 28270124-9 2017 Further investigation of the TRAIL-induced apoptosis pathway revealed that cyproterone acetate exerted its effect by selectively increasing death receptor 5 (DR5) mRNA and protein expression. Cyproterone Acetate 75-94 TNF receptor superfamily member 10b Homo sapiens 158-161 28270124-10 2017 Cyproterone acetate treatment also increased DR5 gene promoter activity, which could be abolished by mutation of a consensus binding domain of transcription factor CCAAT-enhancer-binding protein homologous protein (CHOP) in the DR5 gene promoter. Cyproterone Acetate 0-19 TNF receptor superfamily member 10b Homo sapiens 45-48 28270124-10 2017 Cyproterone acetate treatment also increased DR5 gene promoter activity, which could be abolished by mutation of a consensus binding domain of transcription factor CCAAT-enhancer-binding protein homologous protein (CHOP) in the DR5 gene promoter. Cyproterone Acetate 0-19 TNF receptor superfamily member 10b Homo sapiens 228-231 28270124-11 2017 Cyproterone acetate increases CHOP expression in a concentration and time-dependent manner and endoplasmic reticulum stress reducer 4-phenylbutyrate could block cyproterone acetate-induced CHOP and DR5 up-regulation. Cyproterone Acetate 0-19 TNF receptor superfamily member 10b Homo sapiens 198-201 28270124-11 2017 Cyproterone acetate increases CHOP expression in a concentration and time-dependent manner and endoplasmic reticulum stress reducer 4-phenylbutyrate could block cyproterone acetate-induced CHOP and DR5 up-regulation. 4-phenylbutyric acid 132-148 TNF receptor superfamily member 10b Homo sapiens 198-201 28270124-11 2017 Cyproterone acetate increases CHOP expression in a concentration and time-dependent manner and endoplasmic reticulum stress reducer 4-phenylbutyrate could block cyproterone acetate-induced CHOP and DR5 up-regulation. Cyproterone Acetate 161-180 TNF receptor superfamily member 10b Homo sapiens 198-201 28270124-12 2017 More importantly, siRNA silencing of CHOP significantly reduced cyproterone acetate-induced DR5 up-regulation and TRAIL sensitivity in prostate cancer cells. Cyproterone Acetate 64-83 TNF receptor superfamily member 10b Homo sapiens 92-95 27993669-6 2017 Suppression of etoposide-induced cell death correlated with a downregulation of p53 expression, which, among other functions, regulates the expression of death receptor 5, one of the activators of caspase-8. Etoposide 15-24 TNF receptor superfamily member 10b Homo sapiens 154-170 27903966-3 2017 [Pemetrexed + sildenafil] activated an eIF2alpha - ATF4 - CHOP - Beclin1 pathway causing formation of toxic autophagosomes; activated a protective IRE1 - XBP-1 - chaperone induction pathway; and activated a toxic eIF2alpha - CHOP - DR4 / DR5 / CD95 induction pathway. Pemetrexed 1-11 TNF receptor superfamily member 10b Homo sapiens 238-241 28209994-4 2017 The underlying apoptotic mechanisms involved upregulation of death receptor 5 (DR5) and CCAAT/enhancer binding protein homologous protein, which is related to the endoplasmic reticulum stress response, and is further associated with reactive oxygen species generation and Ca2+ accumulation. Reactive Oxygen Species 233-256 TNF receptor superfamily member 10b Homo sapiens 61-77 28209994-4 2017 The underlying apoptotic mechanisms involved upregulation of death receptor 5 (DR5) and CCAAT/enhancer binding protein homologous protein, which is related to the endoplasmic reticulum stress response, and is further associated with reactive oxygen species generation and Ca2+ accumulation. Reactive Oxygen Species 233-256 TNF receptor superfamily member 10b Homo sapiens 79-82 28098861-8 2017 Triptolide treatment of HepG2 cells caused a significant increase in the expression of p21, Bax and DR5 genes in HepG2 cells. triptolide 0-10 TNF receptor superfamily member 10b Homo sapiens 100-103 28076753-2 2017 LSCs isolated from methotrexate resistant side population (SP) of leukemic cell lines HL60 and MOLT4 exhibited high levels of CD25 and TRAIL R2/DR5 which are potential targets. Methotrexate 19-31 TNF receptor superfamily member 10b Homo sapiens 135-143 28076753-2 2017 LSCs isolated from methotrexate resistant side population (SP) of leukemic cell lines HL60 and MOLT4 exhibited high levels of CD25 and TRAIL R2/DR5 which are potential targets. Methotrexate 19-31 TNF receptor superfamily member 10b Homo sapiens 144-147 28228218-0 2017 [Thioridazine Sensitizes Apoptotic Effect of TRAIL in Human Lung Cancer PC9 Cells Through ER Stress Mediated Up-regulation of DR5]. Thioridazine 1-13 TNF receptor superfamily member 10b Homo sapiens 126-129 28228218-2 2017 This study is to observe the proliferation inhibition effect of TRAIL sensitized by thioridazine on PC9 cells through endoplasmic reticulum (ER) stress mediated up-regulation of death receptor 5 (DR5) and investigate its mechanism. Thioridazine 84-96 TNF receptor superfamily member 10b Homo sapiens 178-194 28228218-2 2017 This study is to observe the proliferation inhibition effect of TRAIL sensitized by thioridazine on PC9 cells through endoplasmic reticulum (ER) stress mediated up-regulation of death receptor 5 (DR5) and investigate its mechanism. Thioridazine 84-96 TNF receptor superfamily member 10b Homo sapiens 196-199 28228218-7 2017 Thioridazine increased the inhibition and apoptosis of PC9 cells and up-regulated the expression of cell-surface DR5 induced by TRAIL. Thioridazine 0-12 TNF receptor superfamily member 10b Homo sapiens 113-116 28228218-9 2017 Western blotting indicated that compared with TRAIL group, expressions of Cleaved-caspase-8, Cleaved-PARP and DR5 increased significantly in combination group of TRAIL and thioridazine. Thioridazine 172-184 TNF receptor superfamily member 10b Homo sapiens 110-113 28228218-10 2017 The induction of DR5 and pro-apoptotic effect were mediated through activation of ER stress accompanying by increased synthesis of GRP78 and CHOP, which can be blocked by adding of ER stress inhibitor 4-PBA. 4-phenylbutylamine 201-206 TNF receptor superfamily member 10b Homo sapiens 17-20 28228218-11 2017 CONCLUSIONS: Thioridazine enhanced proliferation inhibition effect of TRAIL in PC9 cells may be facilitated through ER stress mediated upregulation of DR5. Thioridazine 13-25 TNF receptor superfamily member 10b Homo sapiens 151-154 28182009-10 2017 Expression of the extracellular domains of Fas or TRAILR2 with a glycolipid membrane anchor specifically reduced the intrinsic signalling pathway of CD40 in 293T cells. Glycolipids 65-75 TNF receptor superfamily member 10b Homo sapiens 50-57 28135339-8 2017 These results suggest that lovastatin sensitizes TRAIL-induced apoptosis by up-regulation of DR5 level via NF-kappaB inactivation, but also directly induces apoptosis by dysregulation of MAPK pathway. Lovastatin 27-37 TNF receptor superfamily member 10b Homo sapiens 93-96 27903966-3 2017 [Pemetrexed + sildenafil] activated an eIF2alpha - ATF4 - CHOP - Beclin1 pathway causing formation of toxic autophagosomes; activated a protective IRE1 - XBP-1 - chaperone induction pathway; and activated a toxic eIF2alpha - CHOP - DR4 / DR5 / CD95 induction pathway. Sildenafil Citrate 14-24 TNF receptor superfamily member 10b Homo sapiens 238-241 27192488-6 2016 We demonstrated that 3-HTMC treatment on both cell lines induced apoptotic process associated with overexpression of death receptor DR5, activation of caspase-8 and -3, PARP cleavage, and DNA fragmentation. 3-hydroxy-3',4,4',5'-tetramethoxychalcone 21-27 TNF receptor superfamily member 10b Homo sapiens 132-135 28662515-9 2017 CONCLUSIONS: Our results revealed that shikonin could inhibit cells viability and induce apoptosis of CCA cells, effects enhanced by TRAIL treatment via ROS mediated JNK signalling pathways, involving up-regulation of DR5 expression. shikonin 39-47 TNF receptor superfamily member 10b Homo sapiens 218-221 27978543-7 2016 Furthermore, bortezomib treatment significantly increased expression of the death receptors TRAILR-1 and TRAILR-2 in chondrosarcoma cells. Bortezomib 13-23 TNF receptor superfamily member 10b Homo sapiens 105-113 27720987-0 2016 Death Receptor 5 Networks Require Membrane Cholesterol for Proper Structure and Function. Cholesterol 43-54 TNF receptor superfamily member 10b Homo sapiens 0-16 27720987-1 2016 Death receptor 5 (DR5) is an apoptosis-inducing member of the tumor necrosis factor receptor superfamily, whose activity has been linked to membrane cholesterol content. Cholesterol 149-160 TNF receptor superfamily member 10b Homo sapiens 0-16 27720987-1 2016 Death receptor 5 (DR5) is an apoptosis-inducing member of the tumor necrosis factor receptor superfamily, whose activity has been linked to membrane cholesterol content. Cholesterol 149-160 TNF receptor superfamily member 10b Homo sapiens 18-21 27720987-2 2016 Upon ligand binding, DR5 forms large clusters within the plasma membrane that have often been assumed to be manifestations of receptor co-localization in cholesterol-rich membrane domains. Cholesterol 154-165 TNF receptor superfamily member 10b Homo sapiens 21-24 27720987-11 2016 As evidence, we show that cholesterol is necessary for the covalent dimerization of DR5 transmembrane domains. Cholesterol 26-37 TNF receptor superfamily member 10b Homo sapiens 84-87 27603596-0 2017 Tunicamycin enhances human colon cancer cells to TRAIL-induced apoptosis by JNK-CHOP-mediated DR5 upregulation and the inhibition of the EGFR pathway. Tunicamycin 0-11 TNF receptor superfamily member 10b Homo sapiens 94-97 27603596-4 2017 Knockdown of DR5 but not DR4 expression by specific shRNAs or siRNAs significantly increased tunicamycin-mediated and TRAIL-mediated cell viability. Tunicamycin 93-104 TNF receptor superfamily member 10b Homo sapiens 13-16 27603596-5 2017 DR5 induction was regulated by C/EBP homologous protein (CHOP) and JNK as CHOP siRNA or JNK inhibitor SP600125 considerably abolished the DR5 induction. pyrazolanthrone 102-110 TNF receptor superfamily member 10b Homo sapiens 0-3 27603596-5 2017 DR5 induction was regulated by C/EBP homologous protein (CHOP) and JNK as CHOP siRNA or JNK inhibitor SP600125 considerably abolished the DR5 induction. pyrazolanthrone 102-110 TNF receptor superfamily member 10b Homo sapiens 138-141 27603596-7 2017 In summary, tunicamycin effectively enhanced TRAIL-induced apoptosis might through JNK-CHOP-mediated DR5 upregulation and the inhibition of the epidermal growth factor receptor pathway. Tunicamycin 12-23 TNF receptor superfamily member 10b Homo sapiens 101-104 28028438-11 2016 Unfortunately, the initial increased reduction in tumor growth compared with hvTRA mono treatment was not sustained, and this was related to downregulation of the DR5 level by vemurafenib. Vemurafenib 176-187 TNF receptor superfamily member 10b Homo sapiens 163-166 26822174-6 2016 Antioxidant N-acetyl-L-cysteine (NAC) and c-Jun NH2-terminal kinase (JNK) inhibitor SP600125 inhibited PEITC-induced DR4 and DR5 expression. Acetylcysteine 33-36 TNF receptor superfamily member 10b Homo sapiens 125-128 26822174-6 2016 Antioxidant N-acetyl-L-cysteine (NAC) and c-Jun NH2-terminal kinase (JNK) inhibitor SP600125 inhibited PEITC-induced DR4 and DR5 expression. pyrazolanthrone 84-92 TNF receptor superfamily member 10b Homo sapiens 125-128 26822174-6 2016 Antioxidant N-acetyl-L-cysteine (NAC) and c-Jun NH2-terminal kinase (JNK) inhibitor SP600125 inhibited PEITC-induced DR4 and DR5 expression. phenethyl isothiocyanate 103-108 TNF receptor superfamily member 10b Homo sapiens 125-128 26932761-2 2016 To solve the toxicity versus efficacy and tumor-targeting challenges of PEI used as gene delivery vector, we constructed a novel non-viral vector DR5-TAT-modified Pluronic-PEI (Pluronic-PEI-DR5-TAT), which was based on the attachment of low-molecular-weight polyethylenimine (LMW-PEI) to the amphiphilic polymer Pluronic to prepare Pluronic-modified LMW-PEI (Pluronic-PEI). Polyethyleneimine 72-75 TNF receptor superfamily member 10b Homo sapiens 146-149 26932761-2 2016 To solve the toxicity versus efficacy and tumor-targeting challenges of PEI used as gene delivery vector, we constructed a novel non-viral vector DR5-TAT-modified Pluronic-PEI (Pluronic-PEI-DR5-TAT), which was based on the attachment of low-molecular-weight polyethylenimine (LMW-PEI) to the amphiphilic polymer Pluronic to prepare Pluronic-modified LMW-PEI (Pluronic-PEI). Polyethyleneimine 72-75 TNF receptor superfamily member 10b Homo sapiens 190-193 26932761-2 2016 To solve the toxicity versus efficacy and tumor-targeting challenges of PEI used as gene delivery vector, we constructed a novel non-viral vector DR5-TAT-modified Pluronic-PEI (Pluronic-PEI-DR5-TAT), which was based on the attachment of low-molecular-weight polyethylenimine (LMW-PEI) to the amphiphilic polymer Pluronic to prepare Pluronic-modified LMW-PEI (Pluronic-PEI). Polyethyleneimine 258-274 TNF receptor superfamily member 10b Homo sapiens 146-149 26932761-2 2016 To solve the toxicity versus efficacy and tumor-targeting challenges of PEI used as gene delivery vector, we constructed a novel non-viral vector DR5-TAT-modified Pluronic-PEI (Pluronic-PEI-DR5-TAT), which was based on the attachment of low-molecular-weight polyethylenimine (LMW-PEI) to the amphiphilic polymer Pluronic to prepare Pluronic-modified LMW-PEI (Pluronic-PEI). lmw-pei 276-283 TNF receptor superfamily member 10b Homo sapiens 146-149 26932761-2 2016 To solve the toxicity versus efficacy and tumor-targeting challenges of PEI used as gene delivery vector, we constructed a novel non-viral vector DR5-TAT-modified Pluronic-PEI (Pluronic-PEI-DR5-TAT), which was based on the attachment of low-molecular-weight polyethylenimine (LMW-PEI) to the amphiphilic polymer Pluronic to prepare Pluronic-modified LMW-PEI (Pluronic-PEI). lmw-pei 350-357 TNF receptor superfamily member 10b Homo sapiens 146-149 26932761-2 2016 To solve the toxicity versus efficacy and tumor-targeting challenges of PEI used as gene delivery vector, we constructed a novel non-viral vector DR5-TAT-modified Pluronic-PEI (Pluronic-PEI-DR5-TAT), which was based on the attachment of low-molecular-weight polyethylenimine (LMW-PEI) to the amphiphilic polymer Pluronic to prepare Pluronic-modified LMW-PEI (Pluronic-PEI). pluronic-pei 163-175 TNF receptor superfamily member 10b Homo sapiens 146-149 26932761-2 2016 To solve the toxicity versus efficacy and tumor-targeting challenges of PEI used as gene delivery vector, we constructed a novel non-viral vector DR5-TAT-modified Pluronic-PEI (Pluronic-PEI-DR5-TAT), which was based on the attachment of low-molecular-weight polyethylenimine (LMW-PEI) to the amphiphilic polymer Pluronic to prepare Pluronic-modified LMW-PEI (Pluronic-PEI). pluronic-pei 163-175 TNF receptor superfamily member 10b Homo sapiens 190-193 27933820-2 2016 Here, hyaluronate-gold nanorod/death receptor 5 antibody (HA-AuNR/DR5 Ab) complex was developed for transdermal theranosis of skin cancer. hyaluronate 6-17 TNF receptor superfamily member 10b Homo sapiens 31-47 27933820-2 2016 Here, hyaluronate-gold nanorod/death receptor 5 antibody (HA-AuNR/DR5 Ab) complex was developed for transdermal theranosis of skin cancer. hyaluronate 6-17 TNF receptor superfamily member 10b Homo sapiens 66-69 27629794-3 2016 Interestingly, gefitinib increases the expression of cell surface receptors DR4 and DR5, possibly explaining the synergistic effect. Gefitinib 15-24 TNF receptor superfamily member 10b Homo sapiens 84-87 27750195-2 2016 Our working group has recently demonstrated that nanovectorization of TRAIL with single wall carbon nanotubes (abbreviated NPT) enhanced TRAIL affinity to the high affinity site DR5 and increased pro apoptotic potential in different human tumor cell lines. Carbon 93-99 TNF receptor superfamily member 10b Homo sapiens 178-181 27750195-13 2016 As well, it was shown that the extra cellular acidosis led to the protonation of the TRAIL residue histidine by flipping the His switch to the on position with a concomitant decrease in affinity for DR4 and DR5 receptors. Histidine 99-108 TNF receptor superfamily member 10b Homo sapiens 207-210 27629794-4 2016 Knockdown of DR4 and DR5 by siRNA significantly decreases gefitinib- and TRAIL-mediated cell apoptosis, supporting this idea. Gefitinib 58-67 TNF receptor superfamily member 10b Homo sapiens 21-24 27629794-5 2016 Because the inhibition of gefitinib-induced autophagy by 3-MA significantly decreases DR4 and DR5 upregulation, as well as reduces gefitinib- and TRAIL-induced apoptosis, we conclude that death receptor upregulation is autophagy mediated. Gefitinib 26-35 TNF receptor superfamily member 10b Homo sapiens 94-97 27295176-0 2016 Improved anti-tumor activity of oxaliplatin by encapsulating in anti-DR5 targeted gold nanoparticles. Oxaliplatin 32-43 TNF receptor superfamily member 10b Homo sapiens 69-72 27543608-6 2016 Collectively, we identified CPI-7c as a novel small-molecule inhibitor of MDM2 with a unique two-prong mechanism of action that sensitized TRAIL-resistant cancer cells to apoptosis by modulating the MDM2-p53-DR4/DR5 pathway. cpi-7c 28-34 TNF receptor superfamily member 10b Homo sapiens 212-215 27515963-7 2016 Yet, AZD1152 treatment enhanced death receptor TRAIL-R2 levels in all tumor cell lines investigated. 2-((3-((4-((5-(2-((3-fluorophenyl)amino)-2-oxoethyl)-1H-pyrazol-3-yl)amino)quinazolin-7-yl)oxy)propyl)(ethyl)amino)ethyl dihydrogen phosphate 5-12 TNF receptor superfamily member 10b Homo sapiens 47-55 27731378-3 2016 In this study, we show that ER stress mediated by thapsigargin promoted CHOP and DR5 synthesis thus sensitizing TRAIL treatment, which induced ESCC cells apoptosis. Thapsigargin 50-62 TNF receptor superfamily member 10b Homo sapiens 81-84 27594528-0 2016 A new brominated chalcone derivative suppresses the growth of gastric cancer cells in vitro and in vivo involving ROS mediated up-regulation of DR5 and 4 expression and apoptosis. Chalcone 17-25 TNF receptor superfamily member 10b Homo sapiens 144-153 27594528-0 2016 A new brominated chalcone derivative suppresses the growth of gastric cancer cells in vitro and in vivo involving ROS mediated up-regulation of DR5 and 4 expression and apoptosis. Reactive Oxygen Species 114-117 TNF receptor superfamily member 10b Homo sapiens 144-153 27387452-0 2016 A new oridonin analog suppresses triple-negative breast cancer cells and tumor growth via the induction of death receptor 5. oridonin 6-14 TNF receptor superfamily member 10b Homo sapiens 107-123 27698877-5 2016 The present study demonstrated, using immunofluorescence and western blotting, that blocking gamma-secretase activity in T-ALL cells with N-[(3,5-difluorophenyl) acetyl]-L-alanyl-2-phenyl] glycine-1,1-dimethylethyl ester (DAPT) downregulated NCID and upregulated the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor 5 (DR5). n-[(3,5-difluorophenyl) acetyl]-l-alanyl-2-phenyl] glycine-1,1-dimethylethyl ester 138-220 TNF receptor superfamily member 10b Homo sapiens 332-348 27698877-5 2016 The present study demonstrated, using immunofluorescence and western blotting, that blocking gamma-secretase activity in T-ALL cells with N-[(3,5-difluorophenyl) acetyl]-L-alanyl-2-phenyl] glycine-1,1-dimethylethyl ester (DAPT) downregulated NCID and upregulated the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor 5 (DR5). n-[(3,5-difluorophenyl) acetyl]-l-alanyl-2-phenyl] glycine-1,1-dimethylethyl ester 138-220 TNF receptor superfamily member 10b Homo sapiens 350-353 27565731-5 2016 Meanwhile, ONC201-induced TRAIL/death receptor-5 (DR-5) expression, caspase-8 activation and CRC cell apoptosis were also potentiated with AZD-8055 co-treatment. (5-(2,4-bis((3S)-3-methylmorpholin-4-yl)pyrido(2,3-d)pyrimidin-7-yl)-2-methoxyphenyl)methanol 139-147 TNF receptor superfamily member 10b Homo sapiens 26-48 27648301-3 2016 We demonstrate TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) resistance in the majority of human colon cancers tested and utilize the glucose analog 2-deoxy-d-glucose to sensitize TRAIL-resistant gastrointestinal adenocarcinoma cells, and not normal gastrointestinal epithelial cells, to TRAIL-induced apoptosis through enhanced death receptor 5 expression, downstream modulation of MAPK signaling and subsequent miRNA expression modulation by increasing the expression of miR-494 via MEK activation. Glucose 152-159 TNF receptor superfamily member 10b Homo sapiens 347-363 27924159-0 2016 In Vitro and In Vivo Evaluation of 89Zr-DS-8273a as a Theranostic for Anti-Death Receptor 5 Therapy. Deuterium 40-42 TNF receptor superfamily member 10b Homo sapiens 75-91 27924159-1 2016 Background: DS-8273a, an anti-human death receptor 5 (DR5) agonistic antibody, has cytotoxic activity against human cancer cells and induces apoptosis after specific binding to DR5. DS-8273a 12-20 TNF receptor superfamily member 10b Homo sapiens 36-52 27924159-1 2016 Background: DS-8273a, an anti-human death receptor 5 (DR5) agonistic antibody, has cytotoxic activity against human cancer cells and induces apoptosis after specific binding to DR5. DS-8273a 12-20 TNF receptor superfamily member 10b Homo sapiens 54-57 27924159-1 2016 Background: DS-8273a, an anti-human death receptor 5 (DR5) agonistic antibody, has cytotoxic activity against human cancer cells and induces apoptosis after specific binding to DR5. DS-8273a 12-20 TNF receptor superfamily member 10b Homo sapiens 177-180 27569287-4 2016 Unexpectedly, DR5 is upregulated in the cells treated with DCA/metformin, and sustained under hypoxia. Dichloroacetic Acid 59-62 TNF receptor superfamily member 10b Homo sapiens 14-17 27569287-4 2016 Unexpectedly, DR5 is upregulated in the cells treated with DCA/metformin, and sustained under hypoxia. Metformin 63-72 TNF receptor superfamily member 10b Homo sapiens 14-17 27569287-5 2016 Blocking DR5 by siRNA inhibited DCA/metformin/TRAIL-induced cell death, indicating that DR5 upregulation plays an important role in sensitizing cancer cells to TRAIL-induced cell death. Dichloroacetic Acid 32-35 TNF receptor superfamily member 10b Homo sapiens 9-12 27569287-5 2016 Blocking DR5 by siRNA inhibited DCA/metformin/TRAIL-induced cell death, indicating that DR5 upregulation plays an important role in sensitizing cancer cells to TRAIL-induced cell death. Dichloroacetic Acid 32-35 TNF receptor superfamily member 10b Homo sapiens 88-91 27569287-5 2016 Blocking DR5 by siRNA inhibited DCA/metformin/TRAIL-induced cell death, indicating that DR5 upregulation plays an important role in sensitizing cancer cells to TRAIL-induced cell death. Metformin 36-45 TNF receptor superfamily member 10b Homo sapiens 9-12 27569287-5 2016 Blocking DR5 by siRNA inhibited DCA/metformin/TRAIL-induced cell death, indicating that DR5 upregulation plays an important role in sensitizing cancer cells to TRAIL-induced cell death. Metformin 36-45 TNF receptor superfamily member 10b Homo sapiens 88-91 27569287-6 2016 Furthermore, we found that activation of JNK and c-Jun is responsible for upregulation of DR5 induced by DCA/metformin. Dichloroacetic Acid 105-108 TNF receptor superfamily member 10b Homo sapiens 90-93 27569287-6 2016 Furthermore, we found that activation of JNK and c-Jun is responsible for upregulation of DR5 induced by DCA/metformin. Metformin 109-118 TNF receptor superfamily member 10b Homo sapiens 90-93 27648301-3 2016 We demonstrate TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) resistance in the majority of human colon cancers tested and utilize the glucose analog 2-deoxy-d-glucose to sensitize TRAIL-resistant gastrointestinal adenocarcinoma cells, and not normal gastrointestinal epithelial cells, to TRAIL-induced apoptosis through enhanced death receptor 5 expression, downstream modulation of MAPK signaling and subsequent miRNA expression modulation by increasing the expression of miR-494 via MEK activation. Deoxyglucose 167-184 TNF receptor superfamily member 10b Homo sapiens 347-363 26952193-11 2016 Moreover, because of negative values of H and S * quantities, we demonstrated that van der Waals and hydrogen bonds governed the strong NPT-DR5 association for pH > 7.4 (as for TRAIL alone). Sulfur 49-50 TNF receptor superfamily member 10b Homo sapiens 143-146 27506940-4 2016 DR5, which is essential for 5-FU-induced apoptosis, accumulated in lysosomes and autophagosomes upon chloroquine treatment. Fluorouracil 28-32 TNF receptor superfamily member 10b Homo sapiens 0-3 27506940-4 2016 DR5, which is essential for 5-FU-induced apoptosis, accumulated in lysosomes and autophagosomes upon chloroquine treatment. Chloroquine 101-112 TNF receptor superfamily member 10b Homo sapiens 0-3 27506940-8 2016 We conclude that targeting of lysosomes by chloroquine deregulates DR5 trafficking and abrogates 5-FU- but not TRAIL-stimulated cell elimination, hence suggesting a novel mechanism for receptor activation. Chloroquine 43-54 TNF receptor superfamily member 10b Homo sapiens 67-70 27406983-0 2016 A Histone Deacetylase Inhibitor, OBP-801, and Celecoxib Synergistically Inhibit the Cell Growth with Apoptosis via a DR5-Dependent Pathway in Bladder Cancer Cells. Celecoxib 46-55 TNF receptor superfamily member 10b Homo sapiens 117-120 27406983-7 2016 Therefore, we conjectured that the apoptosis induced by OBP-801 and celecoxib is at least partially dependent on DR5. Celecoxib 68-77 TNF receptor superfamily member 10b Homo sapiens 113-116 26952193-11 2016 Moreover, because of negative values of H and S * quantities, we demonstrated that van der Waals and hydrogen bonds governed the strong NPT-DR5 association for pH > 7.4 (as for TRAIL alone). Hydrogen 104-112 TNF receptor superfamily member 10b Homo sapiens 143-146 27461934-4 2016 In addition, by using caspase-3, caspase-8 and caspase-9 activity assays and western blot analysis, the anticancer effects of triptolide against osteosarcoma growth were found to involve activation of the DR-5/p53/Bax/caspase-9/ caspase-3 signaling pathway and the DR-5/FADD/caspase-8/lysosomal/cathepsin B/caspase-3 signaling pathway in the MG-63 cells. triptolide 126-136 TNF receptor superfamily member 10b Homo sapiens 205-209 27461934-4 2016 In addition, by using caspase-3, caspase-8 and caspase-9 activity assays and western blot analysis, the anticancer effects of triptolide against osteosarcoma growth were found to involve activation of the DR-5/p53/Bax/caspase-9/ caspase-3 signaling pathway and the DR-5/FADD/caspase-8/lysosomal/cathepsin B/caspase-3 signaling pathway in the MG-63 cells. triptolide 126-136 TNF receptor superfamily member 10b Homo sapiens 265-269 27461934-5 2016 An important factor in the anticancer effects of triptolide against osteosarcoma was TRAIL-DR-5. triptolide 49-59 TNF receptor superfamily member 10b Homo sapiens 91-95 27461934-6 2016 The data suggest that triptolide may be a potential novel chemotherapeutic agent for osteosarcoma and acts through the TRAIL-DR-5 signaling pathway. triptolide 22-32 TNF receptor superfamily member 10b Homo sapiens 125-129 27581364-4 2016 Physapubescin decreases the expression of HIF-2alpha and increases the expression of CCAAT/enhancer-binding protein homologus protein (CHOP), which leads to up-regulation of death receptor 5 (DR5), activation of caspase-8 and -3, cleavage of poly (ADP-Ribose) polymerase (PARP) and apoptosis. physapubescin 0-13 TNF receptor superfamily member 10b Homo sapiens 174-190 27268146-6 2016 In A549 and H1299 cells, magnolol and PM remarkably induced cell apoptosis by arresting the cell cycle in the G0/G1 phase while simultaneously activating various pro-apoptotic signals, including TRAIL-R2 (DR5), Bax, caspase 3, cleaved caspase 3, and cleaved PARP. magnolol 25-33 TNF receptor superfamily member 10b Homo sapiens 195-203 27268146-6 2016 In A549 and H1299 cells, magnolol and PM remarkably induced cell apoptosis by arresting the cell cycle in the G0/G1 phase while simultaneously activating various pro-apoptotic signals, including TRAIL-R2 (DR5), Bax, caspase 3, cleaved caspase 3, and cleaved PARP. magnolol 25-33 TNF receptor superfamily member 10b Homo sapiens 205-208 27268146-8 2016 ChIP assays also demonstrated that magnolol and PM significantly enriched the histone acetyl mark (H3K27ac) in the promoter region of DR5. magnolol 35-43 TNF receptor superfamily member 10b Homo sapiens 134-137 27268146-10 2016 Therefore, magnolol and PM, as potential inhibitors of class I HDACs, induced tumor cell apoptosis and suppressed tumor growth partially by epigenetically activating DR5, which is a key protein in death receptor signaling pathway. magnolol 11-19 TNF receptor superfamily member 10b Homo sapiens 166-169 27268146-10 2016 Therefore, magnolol and PM, as potential inhibitors of class I HDACs, induced tumor cell apoptosis and suppressed tumor growth partially by epigenetically activating DR5, which is a key protein in death receptor signaling pathway. pipermethystine 24-26 TNF receptor superfamily member 10b Homo sapiens 166-169 27581364-4 2016 Physapubescin decreases the expression of HIF-2alpha and increases the expression of CCAAT/enhancer-binding protein homologus protein (CHOP), which leads to up-regulation of death receptor 5 (DR5), activation of caspase-8 and -3, cleavage of poly (ADP-Ribose) polymerase (PARP) and apoptosis. physapubescin 0-13 TNF receptor superfamily member 10b Homo sapiens 192-195 27277541-0 2016 Proteasome inhibitor MG132 potentiates TRAIL-induced apoptosis in gallbladder carcinoma GBC-SD cells via DR5-dependent pathway. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 21-26 TNF receptor superfamily member 10b Homo sapiens 105-108 27512955-7 2016 Importantly, our results indicate, for the first time, that DR5 upregulation is mediated by autophagy, as blockade of CK-induced autophagy by 3-MA, LY294002 or Atg7 siRNAs substantially decreases DR5 upregulation and reduces the synergistic effect. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 148-156 TNF receptor superfamily member 10b Homo sapiens 60-63 27363951-3 2016 Sanguinarine treatment increased DR5/TRAILR2 (death receptor 5/TRAIL receptor 2) expression and enhanced the activation of caspase-8 and cleavage of its substrate, Bid. sanguinarine 0-12 TNF receptor superfamily member 10b Homo sapiens 33-36 27363951-3 2016 Sanguinarine treatment increased DR5/TRAILR2 (death receptor 5/TRAIL receptor 2) expression and enhanced the activation of caspase-8 and cleavage of its substrate, Bid. sanguinarine 0-12 TNF receptor superfamily member 10b Homo sapiens 37-44 27363951-3 2016 Sanguinarine treatment increased DR5/TRAILR2 (death receptor 5/TRAIL receptor 2) expression and enhanced the activation of caspase-8 and cleavage of its substrate, Bid. sanguinarine 0-12 TNF receptor superfamily member 10b Homo sapiens 46-62 27277541-8 2016 Taken together, these findings indicate that MG132 possesses anti-gallbladder cancer potential that correlate with regulation of DR5-dependent pathway, and suggest that MG132 may be a promising agent for sensitizing GBC-SD cells to TRAIL-induced apoptosis. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 45-50 TNF receptor superfamily member 10b Homo sapiens 129-132 27277541-8 2016 Taken together, these findings indicate that MG132 possesses anti-gallbladder cancer potential that correlate with regulation of DR5-dependent pathway, and suggest that MG132 may be a promising agent for sensitizing GBC-SD cells to TRAIL-induced apoptosis. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 169-174 TNF receptor superfamily member 10b Homo sapiens 129-132 27453990-7 2016 Tumor cell selective apoptotic action of UA was mainly attributed due to the activation of extrinsic apoptosis pathway via up regulation of DR4, DR5 and PARP cleavage in MCF-7 cells over non-tumorigenic MCF-10A cells. ursolic acid 41-43 TNF receptor superfamily member 10b Homo sapiens 145-148 27176604-9 2016 Manu A caused depolarization of the mitochondrial membrane with induction of CHOP, DR4 and DR5. manumycin 0-6 TNF receptor superfamily member 10b Homo sapiens 91-94 27053682-8 2016 Pre-treatment of PDA cells with MS-444 (Novartis), an established small molecule inhibitor of HuR, substantially increased DR4 and DR5 cell surface levels and enhanced TRAIL sensitivity, further validating HuR"s role in affecting TRAIL apoptotic resistance. 5,8-dihydroxy-3-methyl-4-(9H)-naphtho(2,3-c)furanone 32-38 TNF receptor superfamily member 10b Homo sapiens 131-134 27260301-7 2016 Additionally, we found that CAPE stimulated the expression of death receptor 5 (DR5) and treatment with DR5/Fc chimera protein significantly blocked CAPE/TRAIL-induced apoptosis, which indicates that CAPE/TRAIL stimulated apoptosis through the binding of TRAIL to DR5. caffeic acid phenethyl ester 149-153 TNF receptor superfamily member 10b Homo sapiens 104-107 27260301-7 2016 Additionally, we found that CAPE stimulated the expression of death receptor 5 (DR5) and treatment with DR5/Fc chimera protein significantly blocked CAPE/TRAIL-induced apoptosis, which indicates that CAPE/TRAIL stimulated apoptosis through the binding of TRAIL to DR5. caffeic acid phenethyl ester 149-153 TNF receptor superfamily member 10b Homo sapiens 104-107 27176505-0 2016 The pyrrolo-1,5-benzoxazepine, PBOX-15, enhances TRAIL-induced apoptosis by upregulation of DR5 and downregulation of core cell survival proteins in acute lymphoblastic leukaemia cells. pyrrolo-1,5-benzoxazepine 4-29 TNF receptor superfamily member 10b Homo sapiens 92-95 27176505-0 2016 The pyrrolo-1,5-benzoxazepine, PBOX-15, enhances TRAIL-induced apoptosis by upregulation of DR5 and downregulation of core cell survival proteins in acute lymphoblastic leukaemia cells. PBOX-15 31-38 TNF receptor superfamily member 10b Homo sapiens 92-95 27176505-7 2016 PBOX-15 synergistically enhanced apoptosis induced by TRAIL and a DR5-selective TRAIL variant in ALL-derived cells. PBOX-15 0-7 TNF receptor superfamily member 10b Homo sapiens 66-69 27176505-10 2016 We demonstrate that PBOX-15 can enhance TRAIL-induced apoptosis by upregulation of DR5, reduction of cellular mitochondrial potential, activation of the caspase cascade and downregulation of PI3K/Akt, c-FLIP, Mcl-1 and IAP survival pathways. PBOX-15 20-27 TNF receptor superfamily member 10b Homo sapiens 83-86 27260301-0 2016 Caffeic acid phenethyl ester enhances TRAIL-mediated apoptosis via CHOP-induced death receptor 5 upregulation in hepatocarcinoma Hep3B cells. caffeic acid phenethyl ester 0-28 TNF receptor superfamily member 10b Homo sapiens 80-96 27260301-7 2016 Additionally, we found that CAPE stimulated the expression of death receptor 5 (DR5) and treatment with DR5/Fc chimera protein significantly blocked CAPE/TRAIL-induced apoptosis, which indicates that CAPE/TRAIL stimulated apoptosis through the binding of TRAIL to DR5. caffeic acid phenethyl ester 28-32 TNF receptor superfamily member 10b Homo sapiens 62-78 27260301-7 2016 Additionally, we found that CAPE stimulated the expression of death receptor 5 (DR5) and treatment with DR5/Fc chimera protein significantly blocked CAPE/TRAIL-induced apoptosis, which indicates that CAPE/TRAIL stimulated apoptosis through the binding of TRAIL to DR5. caffeic acid phenethyl ester 28-32 TNF receptor superfamily member 10b Homo sapiens 80-83 26984266-10 2016 We further investigated the upstream extrinsic pathway and showed that kaempferol stimulated death receptor signals [Fas/CD95, death receptor 4 (DR4) and DR5] through increasing the levels of phosphorylated p53 and phosphorylated ATM pathways in HUVECs, which can be individually confirmed by N-acetylcysteine (NAC), ATM specific inhibitor (caffeine) and p53 siRNA. kaempferol 71-81 TNF receptor superfamily member 10b Homo sapiens 154-157 26669750-7 2016 Aotaphenazine (1) enhanced the levels of apoptosis inducing proteins DR4, DR5, p53 and also decreased the levels of cell survival protein Bcl-2 in TRAIL-resistant human gastric adenocarcinoma (AGS) cells in a dose-dependent manner. Aotaphenazine 0-13 TNF receptor superfamily member 10b Homo sapiens 74-77 27029345-9 2016 We investigated the downstream changes in apoptosis pathway components upon Mitoxantrone treatment, and observed that Death Receptors (DR4 and DR5) expression was upregulated, and pro-apoptotic and anti-apoptotic gene expression patterns were altered in favor of apoptosis. Mitoxantrone 76-88 TNF receptor superfamily member 10b Homo sapiens 143-146 26983803-6 2016 Furthermore, DR5 knockdown attenuated the combination treatment of tanshinone IIA with TRAIL-mediated cell death in human NSCLC cells. tanshinone 67-81 TNF receptor superfamily member 10b Homo sapiens 13-16 27129269-5 2016 Results show that CaM directly binds to DR5 in a calcium dependent manner in breast cancer cells. Calcium 49-56 TNF receptor superfamily member 10b Homo sapiens 40-43 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Tryptophan 16-19 TNF receptor superfamily member 10b Homo sapiens 67-70 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Tryptophan 16-19 TNF receptor superfamily member 10b Homo sapiens 107-110 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Tryptophan 16-19 TNF receptor superfamily member 10b Homo sapiens 107-110 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Arginine 25-28 TNF receptor superfamily member 10b Homo sapiens 67-70 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Arginine 25-28 TNF receptor superfamily member 10b Homo sapiens 107-110 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Arginine 25-28 TNF receptor superfamily member 10b Homo sapiens 107-110 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Glutamic Acid 34-37 TNF receptor superfamily member 10b Homo sapiens 67-70 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Glutamic Acid 34-37 TNF receptor superfamily member 10b Homo sapiens 107-110 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Glutamic Acid 34-37 TNF receptor superfamily member 10b Homo sapiens 107-110 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Leucine 43-46 TNF receptor superfamily member 10b Homo sapiens 67-70 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Leucine 43-46 TNF receptor superfamily member 10b Homo sapiens 107-110 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Leucine 43-46 TNF receptor superfamily member 10b Homo sapiens 107-110 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Glutamic Acid 56-59 TNF receptor superfamily member 10b Homo sapiens 67-70 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Glutamic Acid 56-59 TNF receptor superfamily member 10b Homo sapiens 107-110 27129269-8 2016 The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. Glutamic Acid 56-59 TNF receptor superfamily member 10b Homo sapiens 107-110 27222248-5 2016 Using chemical and genetic approaches, we have demonstrated that the B-Raf inhibitor PLX4032 induces DR5 upregulation exclusively in Ras-mutant cancer cells; this effect is dependent on Ras/c-Raf/MEK/ERK signaling activation. Vemurafenib 85-92 TNF receptor superfamily member 10b Homo sapiens 101-104 27222248-6 2016 PLX4032 induces DR5 expression at transcriptional levels, largely due to enhancing CHOP/Elk1-mediated DR5 transcription. Vemurafenib 0-7 TNF receptor superfamily member 10b Homo sapiens 16-19 27222248-6 2016 PLX4032 induces DR5 expression at transcriptional levels, largely due to enhancing CHOP/Elk1-mediated DR5 transcription. Vemurafenib 0-7 TNF receptor superfamily member 10b Homo sapiens 102-105 26983803-7 2016 Tanshinone IIA also increased CHOP and activated the PERK-ATF4 pathway suggesting that tanshinone IIA increased DR5 and CHOP by activating the PERK-ATF4 pathway. tanshinone 87-97 TNF receptor superfamily member 10b Homo sapiens 112-115 26983803-9 2016 Taken together, these results indicate that tanshinone IIA increases TRAIL-induced cell death via upregulating DR5 and downregulating survivin mediated by, respectively, selective activation of PERK/ATF4 and inhibition of STAT3, suggesting combinatorial intervention of tanshinone IIA and TRAIL as a new therapeutic strategy for human NSCLC. tanshinone 44-58 TNF receptor superfamily member 10b Homo sapiens 111-114 26662956-0 2016 Synergistic effect of fisetin combined with sorafenib in human cervical cancer HeLa cells through activation of death receptor-5 mediated caspase-8/caspase-3 and the mitochondria-dependent apoptotic pathway. fisetin 22-29 TNF receptor superfamily member 10b Homo sapiens 112-128 26615420-0 2016 The alkyllysophospholipid edelfosine enhances TRAIL-mediated apoptosis in gastric cancer cells through death receptor 5 and the mitochondrial pathway. alkyllysophospholipid edelfosine 4-36 TNF receptor superfamily member 10b Homo sapiens 103-119 27037412-9 2016 In combination with irinotecan or doxorubicin, FAP-DR5 treatment resulted in substantial tumor regression in patient-derived xenograft models. Irinotecan 20-30 TNF receptor superfamily member 10b Homo sapiens 51-54 27037412-9 2016 In combination with irinotecan or doxorubicin, FAP-DR5 treatment resulted in substantial tumor regression in patient-derived xenograft models. Doxorubicin 34-45 TNF receptor superfamily member 10b Homo sapiens 51-54 26662956-0 2016 Synergistic effect of fisetin combined with sorafenib in human cervical cancer HeLa cells through activation of death receptor-5 mediated caspase-8/caspase-3 and the mitochondria-dependent apoptotic pathway. Sorafenib 44-53 TNF receptor superfamily member 10b Homo sapiens 112-128 26662956-7 2016 Our study showed that the combination of fisetin and sorafenib exerted better synergistic effects in vitro and in vivo than either agent used alone against human cervical cancer, and this synergism was based on apoptotic potential through a mitochondrial- and DR5-dependent caspase-8/caspase-3 signaling pathway. fisetin 41-48 TNF receptor superfamily member 10b Homo sapiens 260-263 26615420-5 2016 Edelfosine upregulated the protein level of death receptor 5 (DR5/TNFRSF10B) and/or increased DR5 upregulation in lipid rafts. edelfosine 0-10 TNF receptor superfamily member 10b Homo sapiens 44-60 26615420-5 2016 Edelfosine upregulated the protein level of death receptor 5 (DR5/TNFRSF10B) and/or increased DR5 upregulation in lipid rafts. edelfosine 0-10 TNF receptor superfamily member 10b Homo sapiens 62-65 26662956-7 2016 Our study showed that the combination of fisetin and sorafenib exerted better synergistic effects in vitro and in vivo than either agent used alone against human cervical cancer, and this synergism was based on apoptotic potential through a mitochondrial- and DR5-dependent caspase-8/caspase-3 signaling pathway. Sorafenib 53-62 TNF receptor superfamily member 10b Homo sapiens 260-263 26615420-5 2016 Edelfosine upregulated the protein level of death receptor 5 (DR5/TNFRSF10B) and/or increased DR5 upregulation in lipid rafts. edelfosine 0-10 TNF receptor superfamily member 10b Homo sapiens 66-75 26942880-9 2016 The induction of DR5 also sensitized NPC cells to radiotherapy, and the SER was 1.195. Serine 72-75 TNF receptor superfamily member 10b Homo sapiens 17-20 26615420-5 2016 Edelfosine upregulated the protein level of death receptor 5 (DR5/TNFRSF10B) and/or increased DR5 upregulation in lipid rafts. edelfosine 0-10 TNF receptor superfamily member 10b Homo sapiens 94-97 26615420-6 2016 In addition, edelfosine-mediated rhTRAIL sensitization was regulated by the DR5 pathway. edelfosine 13-23 TNF receptor superfamily member 10b Homo sapiens 76-79 27033605-6 2016 Depletion of DR4 or DR5 by small interfering RNA significantly reversed the cell growth inhibitory effects of bakuchiol in HCT116 and HT-29 cells. bakuchiol 110-119 TNF receptor superfamily member 10b Homo sapiens 20-23 27033605-8 2016 The collective results suggest that bakuchiol facilitates TRAIL-induced apoptosis in colon cancer cells through up-regulation of the TRAIL receptors; DR4 and DR5 via ROS/JNK pathway signals. bakuchiol 36-45 TNF receptor superfamily member 10b Homo sapiens 158-161 26971531-3 2016 Baicalein stimulated the expression of DR5, FasL, and FADD, and activated caspase-8 by reducing the levels of FLIPs (FLICE-inhibitory proteins). baicalein 0-9 TNF receptor superfamily member 10b Homo sapiens 39-42 26991930-2 2016 Herein, we present a modular approach to generate death receptor 5 (DR5) binding constructs comprising multiple copies of DR5 targeting peptide (DR5TP) covalently bound to biomolecular scaffolds of peptidic nature. dr5tp 145-150 TNF receptor superfamily member 10b Homo sapiens 50-66 26991930-2 2016 Herein, we present a modular approach to generate death receptor 5 (DR5) binding constructs comprising multiple copies of DR5 targeting peptide (DR5TP) covalently bound to biomolecular scaffolds of peptidic nature. dr5tp 145-150 TNF receptor superfamily member 10b Homo sapiens 68-71 26991930-2 2016 Herein, we present a modular approach to generate death receptor 5 (DR5) binding constructs comprising multiple copies of DR5 targeting peptide (DR5TP) covalently bound to biomolecular scaffolds of peptidic nature. dr5tp 145-150 TNF receptor superfamily member 10b Homo sapiens 122-125 26769704-6 2016 Moreover, the expression of tBid, DR5, and Fas proteins was enhanced by FBRA extract, and the pretreatment with caspase-8 inhibitor, but not caspase-9 inhibitor, restored the reduction of the cell viability induced by FBRA extract. fbra 72-76 TNF receptor superfamily member 10b Homo sapiens 34-37 26995783-4 2016 This study sought to provide structural and molecular insight for the roles of four selected DR5 DD mutations (E355K, E367K, K415N, and L363F) in the oligomerization of DR5 DD-FADD complex during the DISC formation. dd-fadd 173-180 TNF receptor superfamily member 10b Homo sapiens 93-96 26995783-4 2016 This study sought to provide structural and molecular insight for the roles of four selected DR5 DD mutations (E355K, E367K, K415N, and L363F) in the oligomerization of DR5 DD-FADD complex during the DISC formation. dd-fadd 173-180 TNF receptor superfamily member 10b Homo sapiens 169-172 26956619-0 2016 ERK mediated upregulation of death receptor 5 overcomes the lack of p53 functionality in the diaminothiazole DAT1 induced apoptosis in colon cancer models: efficiency of DAT1 in Ras-Raf mutated cells. Thiazole-2,4-diamine 93-108 TNF receptor superfamily member 10b Homo sapiens 29-45 26679052-5 2016 This study showed that treatment with cytotoxic concentration of antrocin induced both intrinsic and extrinsic apoptotic pathways in human bladder cancer 5637 cells, evidenced by increase of Fas, DR5, Bax expression and caspase-3, -8 and -9 activation. antrocin 65-73 TNF receptor superfamily member 10b Homo sapiens 196-199 26679052-13 2016 Both antrocin-induced intrinsic and extrinsic apoptosis is through upregulation of pro-apoptotic proteins, including Bax, Fas, and DR5. antrocin 5-13 TNF receptor superfamily member 10b Homo sapiens 131-134 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. dd-fadd 157-164 TNF receptor superfamily member 10b Homo sapiens 153-156 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. dd-fadd 157-164 TNF receptor superfamily member 10b Homo sapiens 340-343 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. dd-fadd 157-164 TNF receptor superfamily member 10b Homo sapiens 340-343 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. fadd 160-164 TNF receptor superfamily member 10b Homo sapiens 153-156 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. fadd 160-164 TNF receptor superfamily member 10b Homo sapiens 340-343 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. fadd 160-164 TNF receptor superfamily member 10b Homo sapiens 340-343 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. dd-fadd 344-351 TNF receptor superfamily member 10b Homo sapiens 153-156 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. dd-fadd 344-351 TNF receptor superfamily member 10b Homo sapiens 340-343 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. dd-fadd 344-351 TNF receptor superfamily member 10b Homo sapiens 340-343 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. fadd 255-259 TNF receptor superfamily member 10b Homo sapiens 153-156 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. fadd 255-259 TNF receptor superfamily member 10b Homo sapiens 340-343 26995783-5 2016 Results from the molecular dynamics simulations show that the simulated mutants induce conformational, dynamical motions and interactions changes in the DR5 DD-FADD tetramer complex, including changes in a protein"s backbone flexibility, less exposure of FADD DED"s caspase-8 binding site, reduced H-bonding and hydrophobic contacts at the DR5 DD-FADD DD binding, altered distribution of the electrostatic potentials and correlated motions of residues, and reduced binding affinity of DR5 DD binding to FADD. fadd 255-259 TNF receptor superfamily member 10b Homo sapiens 340-343 26964637-0 2016 Chloroquine enhances TRAIL-mediated apoptosis through up-regulation of DR5 by stabilization of mRNA and protein in cancer cells. Chloroquine 0-11 TNF receptor superfamily member 10b Homo sapiens 71-74 26964637-4 2016 CQ up-regulates DR5 mRNA and protein expression in a dose- and time- dependent manner. Chloroquine 0-2 TNF receptor superfamily member 10b Homo sapiens 16-19 26964637-6 2016 Moreover, we found that CQ decreased the expression of Cbl, an E3 ligase of DR5, and knock-down of Cbl markedly enhanced DR5 up-regulation. Chloroquine 24-26 TNF receptor superfamily member 10b Homo sapiens 76-79 26964637-7 2016 Other lysosomal inhibitors, including monensin and nigericin, also up-regulated DR5 and sensitized TRAIL-mediated apoptosis. Nigericin 51-60 TNF receptor superfamily member 10b Homo sapiens 80-83 26964637-8 2016 Therefore, this study demonstrates that lysosomal inhibition by CQ may sensitize TRAIL-mediated apoptosis in human renal cancer Caki cells via DR5 up-regulation. Chloroquine 64-66 TNF receptor superfamily member 10b Homo sapiens 143-146 26661339-12 2016 CONCLUSIONS AND IMPLICATIONS: ROS generation by decursin selectively activated the PERK/ATF4 axis of the endoplasmic reticulum stress signalling pathway, leading to enhanced TRAIL sensitivity in TRAIL-resistant NSCLC cell lines, partly via up-regulation of DR5. Reactive Oxygen Species 30-33 TNF receptor superfamily member 10b Homo sapiens 257-260 26771233-8 2016 Whole transcriptomic analysis demonstrated that propionibacterial supernatant or propionibacterial metabolites (propionate and acetate), in combination with TRAIL, increased pro-apoptotic gene expression (TRAIL-R2/DR5) and decreased anti-apoptotic gene expression (FLIP, XIAP) in HT29 human colon cancer cells. Propionates 112-122 TNF receptor superfamily member 10b Homo sapiens 205-213 26332363-7 2016 MEAG also caused an increase in truncated Bid (t-Bid), cleaved caspase-8, and death receptor 5 (DR5). meag 0-4 TNF receptor superfamily member 10b Homo sapiens 78-94 26332363-7 2016 MEAG also caused an increase in truncated Bid (t-Bid), cleaved caspase-8, and death receptor 5 (DR5). meag 0-4 TNF receptor superfamily member 10b Homo sapiens 96-99 26332363-8 2016 Interestingly, withaferin A (WA), a bioactive component of MEAG, clearly induced apoptosis accompanied by upregulation of Bim, t-Bid, caspase-8, and DR5 similar to the effects of MEAG. withaferin A 15-27 TNF receptor superfamily member 10b Homo sapiens 149-152 26843620-3 2016 Importantly, treatment with AD5-10, a DR5-specific agonistic monoclonal antibody, was able to mimic TRAIL-induced apoptosis in CABYR-a/b-silenced cells. ad5-10 28-34 TNF receptor superfamily member 10b Homo sapiens 38-41 26771233-8 2016 Whole transcriptomic analysis demonstrated that propionibacterial supernatant or propionibacterial metabolites (propionate and acetate), in combination with TRAIL, increased pro-apoptotic gene expression (TRAIL-R2/DR5) and decreased anti-apoptotic gene expression (FLIP, XIAP) in HT29 human colon cancer cells. Propionates 112-122 TNF receptor superfamily member 10b Homo sapiens 214-217 26771233-8 2016 Whole transcriptomic analysis demonstrated that propionibacterial supernatant or propionibacterial metabolites (propionate and acetate), in combination with TRAIL, increased pro-apoptotic gene expression (TRAIL-R2/DR5) and decreased anti-apoptotic gene expression (FLIP, XIAP) in HT29 human colon cancer cells. Acetates 127-134 TNF receptor superfamily member 10b Homo sapiens 205-213 26771233-8 2016 Whole transcriptomic analysis demonstrated that propionibacterial supernatant or propionibacterial metabolites (propionate and acetate), in combination with TRAIL, increased pro-apoptotic gene expression (TRAIL-R2/DR5) and decreased anti-apoptotic gene expression (FLIP, XIAP) in HT29 human colon cancer cells. Acetates 127-134 TNF receptor superfamily member 10b Homo sapiens 214-217 26859847-6 2016 Our results also revealed that FKC induced intrinsic and extrinsic apoptosis via upregulation of the levels of pro-apoptotic proteins (Bak) and death receptors (DR5), while downregulation of the levels of anti-apoptotic proteins (XIAP, cIAP-1, c-FlipL, Bcl-xL and survivin), resulting in the activation of caspase-3, -8 and -9 and cleavage of poly(ADP-ribose) polymerase (PARP). flavokawain C 31-34 TNF receptor superfamily member 10b Homo sapiens 161-164 26959625-11 2016 CONCLUSION: Ellagic acid potentially up-regulated DR4, DR5 and MAP kinases (JNK, ERK1/2 and p38). Ellagic Acid 12-24 TNF receptor superfamily member 10b Homo sapiens 55-58 25417048-7 2016 Exploration of NF-kappaB inhibitor (Phenylarsine oxide, 0.1 muM) synergistically further increased SVT-induced DR3 and DR5 expressions accompanied with further inhibition of cancer cells growth. oxophenylarsine 36-54 TNF receptor superfamily member 10b Homo sapiens 119-122 27268643-0 2016 Combination of Nimbolide and TNF-alpha-Increases Human Colon Adenocarcinoma Cell Death through JNK-mediated DR5 Up- regulation. nimbolide 15-24 TNF receptor superfamily member 10b Homo sapiens 108-111 25867072-6 2016 ATP-competitive mTOR/PI3K inhibitors also promote DR5 induction and FADD-dependent apoptosis in colon cancer cells. Adenosine Triphosphate 0-3 TNF receptor superfamily member 10b Homo sapiens 50-53 25867065-7 2016 We found that both B-Raf (for example, PLX4032) and MEK inhibitors (for example, AZD6244 and PD0325901) effectively inhibited ERK1/2 phosphorylation and reduced DR5 levels in both human thyroid cancer and melanoma cells. Vemurafenib 39-46 TNF receptor superfamily member 10b Homo sapiens 161-164 25867065-7 2016 We found that both B-Raf (for example, PLX4032) and MEK inhibitors (for example, AZD6244 and PD0325901) effectively inhibited ERK1/2 phosphorylation and reduced DR5 levels in both human thyroid cancer and melanoma cells. AZD 6244 81-88 TNF receptor superfamily member 10b Homo sapiens 161-164 25867065-7 2016 We found that both B-Raf (for example, PLX4032) and MEK inhibitors (for example, AZD6244 and PD0325901) effectively inhibited ERK1/2 phosphorylation and reduced DR5 levels in both human thyroid cancer and melanoma cells. mirdametinib 93-102 TNF receptor superfamily member 10b Homo sapiens 161-164 26711147-4 2016 Furthermore, ASA could up-regulate TNF-R1 and DR5 via activation of p38 MAPK, thereby activating caspase 8, revealing the death receptor pathway was also involved in this process. Aspirin 13-16 TNF receptor superfamily member 10b Homo sapiens 46-49 27268643-8 2016 In apoptotic pathway, nimbolide activated c-Jun N-terminal kinase (JNK) phosphorylation, BH3 interacting-domain death agonist (Bid) and up-regulated the death receptor 5 (DR5) level. nimbolide 22-31 TNF receptor superfamily member 10b Homo sapiens 153-169 27268643-8 2016 In apoptotic pathway, nimbolide activated c-Jun N-terminal kinase (JNK) phosphorylation, BH3 interacting-domain death agonist (Bid) and up-regulated the death receptor 5 (DR5) level. nimbolide 22-31 TNF receptor superfamily member 10b Homo sapiens 171-174 27268643-9 2016 In the combination group, nimbolide markedly sensitized TNF-alpha-induced JNK, Bid, caspase-3 activation and the up-regulation of DR5. nimbolide 26-35 TNF receptor superfamily member 10b Homo sapiens 130-133 27268643-10 2016 Our findings overall indicate that nimbolide may enhance TNF-alpha-mediated cellular proliferation inhibition through increasing cell apoptosis of HT-29 cells by up-reglation of DR5 expression via the JNK pathway. nimbolide 35-44 TNF receptor superfamily member 10b Homo sapiens 178-181 26499766-7 2015 In addition, goniothalamin enhanced TRAIL-induced apoptosis through increased death receptor DR5 expression and decreased anti-apoptotic regulator cFLIP. goniothalamin 13-26 TNF receptor superfamily member 10b Homo sapiens 93-96 26798390-0 2016 Isoproterenol Enhances Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand-Induced Apoptosis in Human Embryonic Kidney Cells through Death Receptor 5 up-Regulation. Isoproterenol 0-13 TNF receptor superfamily member 10b Homo sapiens 137-153 27822364-6 2016 Molecular docking analysis revealed that compound 1 could bind stably to the TRAIL/DR5 complex through hydrogen bonds. Hydrogen 103-111 TNF receptor superfamily member 10b Homo sapiens 83-86 26381901-6 2015 We discovered that doxorubicin (DOX) sensitizes TRAIL-resistant HT-29 colon cancer cells to TRAIL by upregulating mRNA expression of DR5 by 60% in vitro. Doxorubicin 19-30 TNF receptor superfamily member 10b Homo sapiens 133-136 26381901-6 2015 We discovered that doxorubicin (DOX) sensitizes TRAIL-resistant HT-29 colon cancer cells to TRAIL by upregulating mRNA expression of DR5 by 60% in vitro. Doxorubicin 32-35 TNF receptor superfamily member 10b Homo sapiens 133-136 26381901-8 2015 Alternatively, when DOX was encapsulated in hyaluronic acid-based nanoparticles (HAC/DOX) and intravenously administered with TRAILPEG, DR-mediated apoptosis was potentiated in HT-29 tumors by upregulating DR5 protein expression by 70% and initiating both extrinsic and intrinsic apoptotic pathways with reduced systemic toxicity compared to HAC/DOX or free DOX combined with TRAILPEG (80% vs. 40% survival rate; 75% vs. 34% tumor growth inhibition). Doxorubicin 20-23 TNF receptor superfamily member 10b Homo sapiens 206-209 26381901-8 2015 Alternatively, when DOX was encapsulated in hyaluronic acid-based nanoparticles (HAC/DOX) and intravenously administered with TRAILPEG, DR-mediated apoptosis was potentiated in HT-29 tumors by upregulating DR5 protein expression by 70% and initiating both extrinsic and intrinsic apoptotic pathways with reduced systemic toxicity compared to HAC/DOX or free DOX combined with TRAILPEG (80% vs. 40% survival rate; 75% vs. 34% tumor growth inhibition). Hyaluronic Acid 44-59 TNF receptor superfamily member 10b Homo sapiens 206-209 26381901-8 2015 Alternatively, when DOX was encapsulated in hyaluronic acid-based nanoparticles (HAC/DOX) and intravenously administered with TRAILPEG, DR-mediated apoptosis was potentiated in HT-29 tumors by upregulating DR5 protein expression by 70% and initiating both extrinsic and intrinsic apoptotic pathways with reduced systemic toxicity compared to HAC/DOX or free DOX combined with TRAILPEG (80% vs. 40% survival rate; 75% vs. 34% tumor growth inhibition). hac 81-84 TNF receptor superfamily member 10b Homo sapiens 206-209 26506422-9 2015 In Panc-1 cells, ABT-263 increased the surface expression of death receptor (DR) 5; the NF-kappaB pathway, but not endoplasmic reticulum stress, participated in the increase. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 17-20 TNF receptor superfamily member 10b Homo sapiens 61-82 26426449-7 2015 The triptolide-induced sensitization was accompanied by increased TRAIL-R2 (DR5) and decreased heat shock protein 70 expression. triptolide 4-14 TNF receptor superfamily member 10b Homo sapiens 66-74 26426449-7 2015 The triptolide-induced sensitization was accompanied by increased TRAIL-R2 (DR5) and decreased heat shock protein 70 expression. triptolide 4-14 TNF receptor superfamily member 10b Homo sapiens 76-79 26499766-0 2015 Goniothalamin enhances TRAIL-induced apoptosis in colorectal cancer cells through DR5 upregulation and cFLIP downregulation. goniothalamin 0-13 TNF receptor superfamily member 10b Homo sapiens 82-85 26692822-13 2015 In addition, As2O3 in combination with 5-FU treatment caused up-regulation of DR5, caspase 3, caspase 8, and caspase 9, and down-regulation of BCL-2, but had no effect of DR4. Arsenic Trioxide 13-18 TNF receptor superfamily member 10b Homo sapiens 78-81 26692822-13 2015 In addition, As2O3 in combination with 5-FU treatment caused up-regulation of DR5, caspase 3, caspase 8, and caspase 9, and down-regulation of BCL-2, but had no effect of DR4. Fluorouracil 39-43 TNF receptor superfamily member 10b Homo sapiens 78-81 26499766-8 2015 Interestingly, goniothalamin increased translocation of DR5 to cell surface and consequently contributed to the enhancement of TRAIL-induced apoptosis. goniothalamin 15-28 TNF receptor superfamily member 10b Homo sapiens 56-59 26349783-5 2015 We demonstrated here that chaetocin effectively suppressed the growth of multiple lung cancer cells through inducing apoptosis in a death receptor 5 (DR5)-dependent manner. chaetocin 26-35 TNF receptor superfamily member 10b Homo sapiens 132-148 25640641-6 2015 Phenylacetylamide triggered the expression of C/EBP homologous protein at the early treatment stage, followed by death receptor-5 upregulation, caspase activation, and beta-actin/tubulin degradation. phenylacetylamide 0-17 TNF receptor superfamily member 10b Homo sapiens 113-129 26349783-0 2015 Chaetocin induces endoplasmic reticulum stress response and leads to death receptor 5-dependent apoptosis in human non-small cell lung cancer cells. chaetocin 0-9 TNF receptor superfamily member 10b Homo sapiens 69-85 26349783-5 2015 We demonstrated here that chaetocin effectively suppressed the growth of multiple lung cancer cells through inducing apoptosis in a death receptor 5 (DR5)-dependent manner. chaetocin 26-35 TNF receptor superfamily member 10b Homo sapiens 150-153 26349783-10 2015 In summary, chaetocin pharmacologically inhibits the activity of SUV39H1 which provokes ER stress and results in up-regulation of ATF3 and CHOP, leading to DR5-dependent apoptosis eventually. chaetocin 12-21 TNF receptor superfamily member 10b Homo sapiens 156-159 26193760-4 2015 Compound 4b and the thiazole derivatives, 5b and 6b, increased the expression of tumor necrosis factor receptors TRAIL-R2 and TRAIL-R1, accompanied by down-modulation of pro-caspase 3 levels, and the augmentation of cleaved caspase 3. Thiazoles 20-28 TNF receptor superfamily member 10b Homo sapiens 113-121 26837170-12 2015 In conclusion, GA sensitizes HT-29 cells to TRAIL-induced apoptosis by promoting ROS-activated ERS pathways, up-regulating of DR4 and DR5, and inhibiting c-FLIP expression. Gallium 15-17 TNF receptor superfamily member 10b Homo sapiens 134-137 26491348-10 2015 The protein expressions of caspase-8, -3, and -9 were decreased in human anti-DR5 mAb-treated FLS in a dose-dependent manner through exposure to a caspase inhibitor, indicating that anti-DR5 mAb induction of apoptosis is through the caspase pathway. CHEMBL1232769 94-97 TNF receptor superfamily member 10b Homo sapiens 78-81 26491348-10 2015 The protein expressions of caspase-8, -3, and -9 were decreased in human anti-DR5 mAb-treated FLS in a dose-dependent manner through exposure to a caspase inhibitor, indicating that anti-DR5 mAb induction of apoptosis is through the caspase pathway. CHEMBL1232769 94-97 TNF receptor superfamily member 10b Homo sapiens 187-190 26304927-9 2015 Interestingly, alpha-mangostin, which is a xanthone derivative, canceled the resistance by increasing the expression level of DR5 through down-regulation of miR-133b and effectively induced the translocation of DR5 to the cancer cell surface membrane in TRAIL-resistant DLD-1 cells. mangostin 15-30 TNF receptor superfamily member 10b Homo sapiens 126-129 26304927-9 2015 Interestingly, alpha-mangostin, which is a xanthone derivative, canceled the resistance by increasing the expression level of DR5 through down-regulation of miR-133b and effectively induced the translocation of DR5 to the cancer cell surface membrane in TRAIL-resistant DLD-1 cells. mangostin 15-30 TNF receptor superfamily member 10b Homo sapiens 211-214 26304927-9 2015 Interestingly, alpha-mangostin, which is a xanthone derivative, canceled the resistance by increasing the expression level of DR5 through down-regulation of miR-133b and effectively induced the translocation of DR5 to the cancer cell surface membrane in TRAIL-resistant DLD-1 cells. xanthone 43-51 TNF receptor superfamily member 10b Homo sapiens 126-129 26547077-7 2015 Chemotherapeutic drugs such as doxorubicin, paclitaxel, and bortezomib augmented the effect of both TRAIL variants, and the enhancing effect was more pronounced for DR5-B. Doxorubicin 31-42 TNF receptor superfamily member 10b Homo sapiens 165-168 26295571-8 2015 Doxorubicin-resistant breast cancer cells (MCF-7/ADR) presented a higher expression of DR5 and were more sensitive to NK cells compared with doxorubicin-sensitive breast cancer cells (MCF-7). Doxorubicin 0-11 TNF receptor superfamily member 10b Homo sapiens 87-90 26136424-9 2015 Verticillin A also increased DR5 expression and overcame colon carcinoma resistance to DR5 agonist drozitumab-induced apoptosis. verticillins 0-13 TNF receptor superfamily member 10b Homo sapiens 29-32 26136424-9 2015 Verticillin A also increased DR5 expression and overcame colon carcinoma resistance to DR5 agonist drozitumab-induced apoptosis. verticillins 0-13 TNF receptor superfamily member 10b Homo sapiens 87-90 26124477-14 2015 CONCLUSION: Death-receptor-5 imaging with (111)In-CS-1008 reveals interpatient and intrapatient heterogeneity of uptake in tumor, is not dose dependent, and is predictive of clinical benefit in the treatment of patients who have mCRC. Cesium 50-52 TNF receptor superfamily member 10b Homo sapiens 12-28 26203587-16 2015 Taken together, these data suggest that TIIA enhances TRAIL-induced apoptosis by upregulating DR5 receptors through the ROS-JNK-CHOP signaling axis in human ovarian carcinoma cells. ros 120-123 TNF receptor superfamily member 10b Homo sapiens 94-97 26547077-7 2015 Chemotherapeutic drugs such as doxorubicin, paclitaxel, and bortezomib augmented the effect of both TRAIL variants, and the enhancing effect was more pronounced for DR5-B. Paclitaxel 44-54 TNF receptor superfamily member 10b Homo sapiens 165-168 26547077-7 2015 Chemotherapeutic drugs such as doxorubicin, paclitaxel, and bortezomib augmented the effect of both TRAIL variants, and the enhancing effect was more pronounced for DR5-B. Bortezomib 60-70 TNF receptor superfamily member 10b Homo sapiens 165-168 26044191-0 2015 NVP-BKM120 potentiates apoptosis in tumor necrosis factor-related apoptosis-inducing ligand-resistant glioma cell lines via upregulation of Noxa and death receptor 5. NVP-BKM120 4-10 TNF receptor superfamily member 10b Homo sapiens 149-165 26044191-8 2015 BKM120 upregulated cell surface expression of death receptor 5 (DR5), but did not increase levels of the other major TRAIL receptor, death receptor 4 (DR4). NVP-BKM120 0-6 TNF receptor superfamily member 10b Homo sapiens 46-62 26044191-8 2015 BKM120 upregulated cell surface expression of death receptor 5 (DR5), but did not increase levels of the other major TRAIL receptor, death receptor 4 (DR4). NVP-BKM120 0-6 TNF receptor superfamily member 10b Homo sapiens 64-67 25917567-0 2015 FLLL12 induces apoptosis in lung cancer cells through a p53/p73-independent but death receptor 5-dependent pathway. flll12 0-6 TNF receptor superfamily member 10b Homo sapiens 80-96 25930665-10 2015 DR4 or DR5 neutralizing antibodies abolished the effect of alpha-H on TRAIL-induced apoptosis, suggesting that sensitization was mediated through the death receptor pathway. alpha-h 59-66 TNF receptor superfamily member 10b Homo sapiens 7-10 26059173-6 2015 In an apoptosis array, betulin activated the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors TRAIL R1/DR4 and R2/DR5, and tumour necrosis factor receptor 1 (TNFR1), suggesting that betulin treatment leads to induction of apoptosis through both intrinsic and extrinsic apoptosis pathways in RCC4 cells. betulin 23-30 TNF receptor superfamily member 10b Homo sapiens 140-143 25917567-6 2015 Moreover, FLLL12 induced the expression of death receptor-5 (DR5). flll12 10-16 TNF receptor superfamily member 10b Homo sapiens 43-59 25917567-6 2015 Moreover, FLLL12 induced the expression of death receptor-5 (DR5). flll12 10-16 TNF receptor superfamily member 10b Homo sapiens 61-64 25917567-11 2015 Taken together, our results strongly suggest that FLLL12 induces apoptosis of lung cancer cell lines by posttranscriptional regulation of DR5 through activation of protein tyrosine phosphatase(s). flll12 50-56 TNF receptor superfamily member 10b Homo sapiens 138-141 25724522-2 2015 Because gene profiling studies have revealed that methionine depletion increases TNF-related apoptosis-inducing ligand receptor-2 (TRAIL-R2) mRNA, we postulated that methionine stress sensitizes breast cancer cells to proapoptotic TRAIL-R2 agonists. Methionine 50-60 TNF receptor superfamily member 10b Homo sapiens 81-129 26009898-0 2015 The novel proteasome inhibitor carfilzomib activates and enhances extrinsic apoptosis involving stabilization of death receptor 5. carfilzomib 31-42 TNF receptor superfamily member 10b Homo sapiens 113-129 26170696-0 2015 Gefitinib upregulates death receptor 5 expression to mediate rmhTRAIL-induced apoptosis in Gefitinib-sensitive NSCLC cell line. Gefitinib 0-9 TNF receptor superfamily member 10b Homo sapiens 22-38 26170696-0 2015 Gefitinib upregulates death receptor 5 expression to mediate rmhTRAIL-induced apoptosis in Gefitinib-sensitive NSCLC cell line. Gefitinib 91-100 TNF receptor superfamily member 10b Homo sapiens 22-38 26170696-4 2015 Therefore, we investigated the effects of the combination of drugs and the expression of the DR5 to analyze the growth of a gefitinib-responsive non-small cell lung cancer cell line PC9, which was treated with rmhTRAIL and gefitinib individually or in combination. Gefitinib 124-133 TNF receptor superfamily member 10b Homo sapiens 93-96 25724522-2 2015 Because gene profiling studies have revealed that methionine depletion increases TNF-related apoptosis-inducing ligand receptor-2 (TRAIL-R2) mRNA, we postulated that methionine stress sensitizes breast cancer cells to proapoptotic TRAIL-R2 agonists. Methionine 50-60 TNF receptor superfamily member 10b Homo sapiens 131-139 25724522-2 2015 Because gene profiling studies have revealed that methionine depletion increases TNF-related apoptosis-inducing ligand receptor-2 (TRAIL-R2) mRNA, we postulated that methionine stress sensitizes breast cancer cells to proapoptotic TRAIL-R2 agonists. Methionine 50-60 TNF receptor superfamily member 10b Homo sapiens 231-239 25724522-2 2015 Because gene profiling studies have revealed that methionine depletion increases TNF-related apoptosis-inducing ligand receptor-2 (TRAIL-R2) mRNA, we postulated that methionine stress sensitizes breast cancer cells to proapoptotic TRAIL-R2 agonists. Methionine 166-176 TNF receptor superfamily member 10b Homo sapiens 81-129 25724522-2 2015 Because gene profiling studies have revealed that methionine depletion increases TNF-related apoptosis-inducing ligand receptor-2 (TRAIL-R2) mRNA, we postulated that methionine stress sensitizes breast cancer cells to proapoptotic TRAIL-R2 agonists. Methionine 166-176 TNF receptor superfamily member 10b Homo sapiens 131-139 25724522-2 2015 Because gene profiling studies have revealed that methionine depletion increases TNF-related apoptosis-inducing ligand receptor-2 (TRAIL-R2) mRNA, we postulated that methionine stress sensitizes breast cancer cells to proapoptotic TRAIL-R2 agonists. Methionine 166-176 TNF receptor superfamily member 10b Homo sapiens 231-239 25724522-7 2015 RESULTS: Methionine depletion sensitized TNBC cells to lexatumumab-induced caspase activation and apoptosis by increasing TRAIL-R2 mRNA and cell surface expression. Methionine 9-19 TNF receptor superfamily member 10b Homo sapiens 122-130 25724522-9 2015 Proteomics analyses revealed that MAGED2, which has been reported to reduce TRAIL-R2 expression, was suppressed by methionine stress. Methionine 115-125 TNF receptor superfamily member 10b Homo sapiens 76-84 25724522-12 2015 CONCLUSIONS: Methionine depletion exposes a targetable defect in TNBC cells by increasing TRAIL-R2 expression. Methionine 13-23 TNF receptor superfamily member 10b Homo sapiens 90-98 25955843-0 2015 Metformin Causes G1-Phase Arrest via Down-Regulation of MiR-221 and Enhances TRAIL Sensitivity through DR5 Up-Regulation in Pancreatic Cancer Cells. Metformin 0-9 TNF receptor superfamily member 10b Homo sapiens 103-106 25770930-4 2015 Combination of chrysin and cisplatin increased the phosphorylation and accumulation of p53 through activating ERK1/2 in Hep G2 cells, which led to the overexpression of the pro-apoptotic proteins Bax and DR5 and the inhibition of the anti-apoptotic protein Bcl-2. chrysin 15-22 TNF receptor superfamily member 10b Homo sapiens 204-207 25770930-4 2015 Combination of chrysin and cisplatin increased the phosphorylation and accumulation of p53 through activating ERK1/2 in Hep G2 cells, which led to the overexpression of the pro-apoptotic proteins Bax and DR5 and the inhibition of the anti-apoptotic protein Bcl-2. Cisplatin 27-36 TNF receptor superfamily member 10b Homo sapiens 204-207 25955843-9 2015 Metformin induced the expressions of death receptor 5 (DR5), a receptor for TRAIL, and Bim with a pro-apoptotic function in the downstream of TRAIL-DR5 pathway. Metformin 0-9 TNF receptor superfamily member 10b Homo sapiens 37-53 25955843-9 2015 Metformin induced the expressions of death receptor 5 (DR5), a receptor for TRAIL, and Bim with a pro-apoptotic function in the downstream of TRAIL-DR5 pathway. Metformin 0-9 TNF receptor superfamily member 10b Homo sapiens 55-58 25955843-9 2015 Metformin induced the expressions of death receptor 5 (DR5), a receptor for TRAIL, and Bim with a pro-apoptotic function in the downstream of TRAIL-DR5 pathway. Metformin 0-9 TNF receptor superfamily member 10b Homo sapiens 148-151 25991668-0 2015 Delphinidin sensitizes prostate cancer cells to TRAIL-induced apoptosis, by inducing DR5 and causing caspase-mediated HDAC3 cleavage. delphinidin 0-11 TNF receptor superfamily member 10b Homo sapiens 85-88 25721064-0 2015 Unexpected hepatotoxicity in a phase I study of TAS266, a novel tetravalent agonistic Nanobody targeting the DR5 receptor. tas266 48-54 TNF receptor superfamily member 10b Homo sapiens 110-113 25721064-1 2015 PURPOSE: TAS266 is a novel agonistic tetravalent Nanobody( ) targeting the DR5 receptor. tas266 9-15 TNF receptor superfamily member 10b Homo sapiens 75-78 25991668-6 2015 TRAIL-induced apoptosis in prostate cancer cells pretreated with delphinidin was dependent on death receptor 5 (DR5) and downstream cleavage of histone deacetylase 3 (HDAC3). delphinidin 65-76 TNF receptor superfamily member 10b Homo sapiens 94-110 25991668-6 2015 TRAIL-induced apoptosis in prostate cancer cells pretreated with delphinidin was dependent on death receptor 5 (DR5) and downstream cleavage of histone deacetylase 3 (HDAC3). delphinidin 65-76 TNF receptor superfamily member 10b Homo sapiens 112-115 25991668-7 2015 In conclusion, delphinidin sensitizes prostate cancer cells to TRAIL-induced apoptosis by inducing DR5, thus causing caspase-mediated HDAC3 cleavage. delphinidin 15-26 TNF receptor superfamily member 10b Homo sapiens 99-102 25766325-0 2015 A benzimidazole derivative exhibiting antitumor activity blocks EGFR and HER2 activity and upregulates DR5 in breast cancer cells. benzimidazole 2-15 TNF receptor superfamily member 10b Homo sapiens 103-106 25932013-4 2015 We here show that selective activation of DR5 specifically induces calcium influx only in the primary cilia, whereas non-selective activation of dopamine receptor induces calcium fluxes in both cilioplasm and cytoplasm. Calcium 67-74 TNF receptor superfamily member 10b Homo sapiens 42-45 25932013-6 2015 Furthermore, calcium activation in the cilioplasm by DR5 increases length and mechanosensory function of primary cilia, leading to a greater response to fluid-shear stress. Calcium 13-20 TNF receptor superfamily member 10b Homo sapiens 53-56 25796504-8 2015 Together, we conclude that in TRAIL-treated MDR gastric carcinoma cells, cisplatin induces the death receptors DR4 and DR5 through the up-regulation of c-myc and strengthens the activation of caspases via promoting the release of cyt c. Cisplatin 73-82 TNF receptor superfamily member 10b Homo sapiens 119-122 25796504-0 2015 Cisplatin-mediated c-myc overexpression and cytochrome c (cyt c) release result in the up-regulation of the death receptors DR4 and DR5 and the activation of caspase 3 and caspase 9, likely responsible for the TRAIL-sensitizing effect of cisplatin. Cisplatin 0-9 TNF receptor superfamily member 10b Homo sapiens 132-135 25796504-0 2015 Cisplatin-mediated c-myc overexpression and cytochrome c (cyt c) release result in the up-regulation of the death receptors DR4 and DR5 and the activation of caspase 3 and caspase 9, likely responsible for the TRAIL-sensitizing effect of cisplatin. Cisplatin 238-247 TNF receptor superfamily member 10b Homo sapiens 132-135 25502339-0 2015 Parthenolide enhances sensitivity of colorectal cancer cells to TRAIL by inducing death receptor 5 and promotes TRAIL-induced apoptosis. parthenolide 0-12 TNF receptor superfamily member 10b Homo sapiens 82-98 25541375-7 2015 TaqMan low density array analysis of A549 cells also confirmed that the induction of apoptosis by the polysaccharide occurred through the TRAIL-DR4/DR5 pathways. Polysaccharides 102-116 TNF receptor superfamily member 10b Homo sapiens 148-151 25208926-7 2015 Evidence that such genes may be targets of regulation via RA signalling initiated by ALDH1A activity was provided by the TNFRSF10B gene, encoding the apoptotic death receptor TNFRSF10B (also termed TRAIL-R2), which negatively correlated with ALDH1A2 and showed elevated transcription following treatment of T-ALL cell lines with the ALDH1A inhibitor citral (3,7-dimethyl-2,6-octadienal). Tretinoin 58-60 TNF receptor superfamily member 10b Homo sapiens 121-130 25487639-0 2015 Thioether analogues of disulfide-bridged cyclic peptides targeting death receptor 5: conformational analysis, dimerisation and consequences for receptor activation. Sulfides 0-9 TNF receptor superfamily member 10b Homo sapiens 67-83 25487639-0 2015 Thioether analogues of disulfide-bridged cyclic peptides targeting death receptor 5: conformational analysis, dimerisation and consequences for receptor activation. Disulfides 23-32 TNF receptor superfamily member 10b Homo sapiens 67-83 25487639-2 2015 We report the effect of replacing the disulfide bridge with a lanthionine linkage in a 16-mer cyclic peptide that binds to death receptor 5 (DR5, TRAIL-R2). Disulfides 38-47 TNF receptor superfamily member 10b Homo sapiens 123-139 25487639-2 2015 We report the effect of replacing the disulfide bridge with a lanthionine linkage in a 16-mer cyclic peptide that binds to death receptor 5 (DR5, TRAIL-R2). Disulfides 38-47 TNF receptor superfamily member 10b Homo sapiens 141-144 25487639-2 2015 We report the effect of replacing the disulfide bridge with a lanthionine linkage in a 16-mer cyclic peptide that binds to death receptor 5 (DR5, TRAIL-R2). Disulfides 38-47 TNF receptor superfamily member 10b Homo sapiens 146-154 25487639-2 2015 We report the effect of replacing the disulfide bridge with a lanthionine linkage in a 16-mer cyclic peptide that binds to death receptor 5 (DR5, TRAIL-R2). lanthionine 62-73 TNF receptor superfamily member 10b Homo sapiens 123-139 25487639-2 2015 We report the effect of replacing the disulfide bridge with a lanthionine linkage in a 16-mer cyclic peptide that binds to death receptor 5 (DR5, TRAIL-R2). lanthionine 62-73 TNF receptor superfamily member 10b Homo sapiens 141-144 25487639-2 2015 We report the effect of replacing the disulfide bridge with a lanthionine linkage in a 16-mer cyclic peptide that binds to death receptor 5 (DR5, TRAIL-R2). lanthionine 62-73 TNF receptor superfamily member 10b Homo sapiens 146-154 25487639-3 2015 Upon covalent oligomerisation, the disulfide-bridged peptide has previously shown similar behaviour to that of TNF-related apoptosis inducing ligand (TRAIL), by selectively triggering the DR5 cell death pathway. Disulfides 35-44 TNF receptor superfamily member 10b Homo sapiens 188-191 25487639-6 2015 The same holds true for dimeric versions of these peptides: the thioether is able to induce DR5-mediated apoptosis of BJAB lymphoma and tumorigenic BJELR cells, albeit to a slightly lower extent compared to its disulfide homologue. Sulfides 64-73 TNF receptor superfamily member 10b Homo sapiens 92-95 25487639-6 2015 The same holds true for dimeric versions of these peptides: the thioether is able to induce DR5-mediated apoptosis of BJAB lymphoma and tumorigenic BJELR cells, albeit to a slightly lower extent compared to its disulfide homologue. Disulfides 211-220 TNF receptor superfamily member 10b Homo sapiens 92-95 25218028-2 2015 Celecoxib and its non-coxib analog, 2,5-dimethyl celecoxib, induced ULBP-1 and DR5 in both COX-2 negative HCT-15 cells and COX-2 positive HT-29 cells. Celecoxib 0-9 TNF receptor superfamily member 10b Homo sapiens 79-82 25218028-2 2015 Celecoxib and its non-coxib analog, 2,5-dimethyl celecoxib, induced ULBP-1 and DR5 in both COX-2 negative HCT-15 cells and COX-2 positive HT-29 cells. 2,5-dimethylcelecoxib 36-58 TNF receptor superfamily member 10b Homo sapiens 79-82 25218028-4 2015 The inducibility of ULBP-1 and DR5 by celecoxib was not different between CD44- and CD44+ HCT-15 cells, and CD133- and CD133+ HT-29 cells. Celecoxib 38-47 TNF receptor superfamily member 10b Homo sapiens 31-34 25218028-6 2015 In addition, celecoxib induced CHOP, and thapsigargin, an inducer of ER (endoplasmic reticulum) stress, induced DR5 but not ULBP1 in HCT-15. Thapsigargin 41-53 TNF receptor superfamily member 10b Homo sapiens 112-115 25218028-7 2015 Taken together, these findings suggest that celecoxib induces the expression of ULBP-1 as well as DR5 in clonogenic colon cancer cells via COX-2 and ER stress-independent pathways, and increases their susceptibility to NK cells. Celecoxib 44-53 TNF receptor superfamily member 10b Homo sapiens 98-101 25593641-5 2015 Combined treatment with TSA and TRAIL significantly reduced the levels of the cellular Fas-associated death domain (FADD)-like interleukin-1beta-converting enzyme (FLICE) inhibitory protein (c-FLIP), whereas those of death receptor (DR) 4, DR5, and FADD remained unchanged. trichostatin A 24-27 TNF receptor superfamily member 10b Homo sapiens 240-243 25208926-7 2015 Evidence that such genes may be targets of regulation via RA signalling initiated by ALDH1A activity was provided by the TNFRSF10B gene, encoding the apoptotic death receptor TNFRSF10B (also termed TRAIL-R2), which negatively correlated with ALDH1A2 and showed elevated transcription following treatment of T-ALL cell lines with the ALDH1A inhibitor citral (3,7-dimethyl-2,6-octadienal). Tretinoin 58-60 TNF receptor superfamily member 10b Homo sapiens 175-184 25208926-7 2015 Evidence that such genes may be targets of regulation via RA signalling initiated by ALDH1A activity was provided by the TNFRSF10B gene, encoding the apoptotic death receptor TNFRSF10B (also termed TRAIL-R2), which negatively correlated with ALDH1A2 and showed elevated transcription following treatment of T-ALL cell lines with the ALDH1A inhibitor citral (3,7-dimethyl-2,6-octadienal). Tretinoin 58-60 TNF receptor superfamily member 10b Homo sapiens 198-206 25208926-7 2015 Evidence that such genes may be targets of regulation via RA signalling initiated by ALDH1A activity was provided by the TNFRSF10B gene, encoding the apoptotic death receptor TNFRSF10B (also termed TRAIL-R2), which negatively correlated with ALDH1A2 and showed elevated transcription following treatment of T-ALL cell lines with the ALDH1A inhibitor citral (3,7-dimethyl-2,6-octadienal). citral 350-356 TNF receptor superfamily member 10b Homo sapiens 121-130 25208926-7 2015 Evidence that such genes may be targets of regulation via RA signalling initiated by ALDH1A activity was provided by the TNFRSF10B gene, encoding the apoptotic death receptor TNFRSF10B (also termed TRAIL-R2), which negatively correlated with ALDH1A2 and showed elevated transcription following treatment of T-ALL cell lines with the ALDH1A inhibitor citral (3,7-dimethyl-2,6-octadienal). citral 350-356 TNF receptor superfamily member 10b Homo sapiens 175-184 25208926-7 2015 Evidence that such genes may be targets of regulation via RA signalling initiated by ALDH1A activity was provided by the TNFRSF10B gene, encoding the apoptotic death receptor TNFRSF10B (also termed TRAIL-R2), which negatively correlated with ALDH1A2 and showed elevated transcription following treatment of T-ALL cell lines with the ALDH1A inhibitor citral (3,7-dimethyl-2,6-octadienal). citral 350-356 TNF receptor superfamily member 10b Homo sapiens 198-206 25208926-7 2015 Evidence that such genes may be targets of regulation via RA signalling initiated by ALDH1A activity was provided by the TNFRSF10B gene, encoding the apoptotic death receptor TNFRSF10B (also termed TRAIL-R2), which negatively correlated with ALDH1A2 and showed elevated transcription following treatment of T-ALL cell lines with the ALDH1A inhibitor citral (3,7-dimethyl-2,6-octadienal). citral 358-385 TNF receptor superfamily member 10b Homo sapiens 121-130 25208926-7 2015 Evidence that such genes may be targets of regulation via RA signalling initiated by ALDH1A activity was provided by the TNFRSF10B gene, encoding the apoptotic death receptor TNFRSF10B (also termed TRAIL-R2), which negatively correlated with ALDH1A2 and showed elevated transcription following treatment of T-ALL cell lines with the ALDH1A inhibitor citral (3,7-dimethyl-2,6-octadienal). citral 358-385 TNF receptor superfamily member 10b Homo sapiens 175-184 25208926-7 2015 Evidence that such genes may be targets of regulation via RA signalling initiated by ALDH1A activity was provided by the TNFRSF10B gene, encoding the apoptotic death receptor TNFRSF10B (also termed TRAIL-R2), which negatively correlated with ALDH1A2 and showed elevated transcription following treatment of T-ALL cell lines with the ALDH1A inhibitor citral (3,7-dimethyl-2,6-octadienal). citral 358-385 TNF receptor superfamily member 10b Homo sapiens 198-206 25344582-0 2015 Small molecule inhibitor YM155-mediated activation of death receptor 5 is crucial for chemotherapy-induced apoptosis in pancreatic carcinoma. YM 155 25-30 TNF receptor superfamily member 10b Homo sapiens 54-70 25333675-9 2015 Further investigation of the mechanism underlying these effects revealed that the evodiamine + TRAIL effect is associated with the increased expression of death receptor (DR)4, DR5, caspase-8 and cleaved caspase-3. evodiamine 82-92 TNF receptor superfamily member 10b Homo sapiens 177-180 26111475-10 2015 The ROS inhibitor NAC and silencing of DR5 by siRNA transfection inhibited the ability of combination to induce PARP cleavage and generate ROS. Reactive Oxygen Species 139-142 TNF receptor superfamily member 10b Homo sapiens 39-42 25344582-8 2015 We found that YM155 induced apoptosis by broad-spectrum inhibition of IAP family member proteins (e.g., CIAP1/2 and FLIP) and induced proapoptotic Bak protein upregulation and activation; the antitumor effect of YM155 treatment with either the DR5 agonist lexatumumab or gemcitabine on pancreatic cancer cells was synergistic. YM 155 14-19 TNF receptor superfamily member 10b Homo sapiens 244-247 25320841-3 2014 In this study, isoegomaketone (1) acted as a synergistic TRAIL sensitizer by mediating up-regulation of DR5 expression in primary prostate cancer RC-58T/h/SA#4 cells. isoegomaketone 15-29 TNF receptor superfamily member 10b Homo sapiens 104-107 25200008-4 2014 We describe the development of an optimized polymer-based nanotherapeutic incorporating both a functionalized polyethylene glycol (PEG) layer and targeting antibodies to limit premature phagocytic clearance whilst enabling targeting of DR5-expressing tumor cells. Polymers 44-51 TNF receptor superfamily member 10b Homo sapiens 236-239 25200008-5 2014 Using the HCT116 colorectal cancer model, we show that following binding to DR5, the nanoparticles activate caspase 8, enhancing the anti-tumor activity of the camptothecin payload both in vitro and in vivo. Camptothecin 160-172 TNF receptor superfamily member 10b Homo sapiens 76-79 25200008-6 2014 Importantly, the combination of nanoparticle-induced DR5 clustering with camptothecin delivery overcomes resistance to DR5-induced apoptosis caused by loss of BAX or overexpression of anti-apoptotic FLIP. Camptothecin 73-85 TNF receptor superfamily member 10b Homo sapiens 119-122 25506265-0 2014 Upregulation of death receptor 5 and activation of caspase 8/3 play a critical role in ergosterol peroxide induced apoptosis in DU 145 prostate cancer cells. ergosterol-5,8-peroxide 87-106 TNF receptor superfamily member 10b Homo sapiens 16-32 25506265-11 2014 CONCLUSION: Overall, our findings suggest that ergosterol peroxide induces apoptosis via activation of death receptor 5 and caspase 8/3 in DU 145 prostate cancer cells as a cancer chemopreventive agent or dietary factor. ergosterol-5,8-peroxide 47-66 TNF receptor superfamily member 10b Homo sapiens 103-119 25320841-7 2014 Furthermore, DR5 expression induced by 1 was mediated via a ROS-independent pathway that required CHOP and p53. ros 60-63 TNF receptor superfamily member 10b Homo sapiens 13-16 25320841-8 2014 Overall, these findings provide evidence that 1 potentiates TRAIL-mediated apoptosis through up-regulation of DR5 via a ROS-independent pathway. ros 120-123 TNF receptor superfamily member 10b Homo sapiens 110-113 25461556-0 2014 Camptothecin sensitizes human hepatoma Hep3B cells to TRAIL-mediated apoptosis via ROS-dependent death receptor 5 upregulation with the involvement of MAPKs. Camptothecin 0-12 TNF receptor superfamily member 10b Homo sapiens 97-113 25461556-6 2014 CPT-induced generation of reactive oxygen species (ROS) also preceded the upregulation of DR5 in response to TRAIL. Reactive Oxygen Species 26-49 TNF receptor superfamily member 10b Homo sapiens 90-93 25461556-6 2014 CPT-induced generation of reactive oxygen species (ROS) also preceded the upregulation of DR5 in response to TRAIL. Reactive Oxygen Species 51-54 TNF receptor superfamily member 10b Homo sapiens 90-93 25461556-7 2014 The involvement of ROS in DR5 upregulation confirmed that pretreatment with antioxidants, including N-acetyl-L-cysteine and glutathione, significantly inhibits CPT-TRAIL-induced cell death by suppressing DR5 expression. Reactive Oxygen Species 19-22 TNF receptor superfamily member 10b Homo sapiens 26-29 25461556-7 2014 The involvement of ROS in DR5 upregulation confirmed that pretreatment with antioxidants, including N-acetyl-L-cysteine and glutathione, significantly inhibits CPT-TRAIL-induced cell death by suppressing DR5 expression. Acetylcysteine 100-119 TNF receptor superfamily member 10b Homo sapiens 26-29 25461556-7 2014 The involvement of ROS in DR5 upregulation confirmed that pretreatment with antioxidants, including N-acetyl-L-cysteine and glutathione, significantly inhibits CPT-TRAIL-induced cell death by suppressing DR5 expression. Acetylcysteine 100-119 TNF receptor superfamily member 10b Homo sapiens 204-207 25461556-7 2014 The involvement of ROS in DR5 upregulation confirmed that pretreatment with antioxidants, including N-acetyl-L-cysteine and glutathione, significantly inhibits CPT-TRAIL-induced cell death by suppressing DR5 expression. Glutathione 124-135 TNF receptor superfamily member 10b Homo sapiens 26-29 25461556-7 2014 The involvement of ROS in DR5 upregulation confirmed that pretreatment with antioxidants, including N-acetyl-L-cysteine and glutathione, significantly inhibits CPT-TRAIL-induced cell death by suppressing DR5 expression. Glutathione 124-135 TNF receptor superfamily member 10b Homo sapiens 204-207 24905183-4 2014 Moreover, honokiol elicited the extrinsic death receptor pathway of DR5 and caspase 8 and the intrinsic pathway of caspase 9. honokiol 10-18 TNF receptor superfamily member 10b Homo sapiens 68-71 25289891-7 2014 We tested synergistic apoptosis-inducing cotreatment strategies and determined that only the proteasome inhibitor bortezomib potently enhanced expression of the TRAIL receptors TRAIL-R1 and/or TRAIL-R2, the formation of the TRAIL death-inducing signaling complex, and activation of caspases; this treatment resulted in sensitization of all TRAIL-resistant meningioma samples to TRAIL-induced apoptosis. Bortezomib 114-124 TNF receptor superfamily member 10b Homo sapiens 193-201 25017974-4 2014 We found that CCT327 enhanced TRAIL-induced apoptosis through upregulation of death receptors DR4 and DR5. CCT327 14-20 TNF receptor superfamily member 10b Homo sapiens 102-105 25341043-6 2014 Bortezomib-mediated sensitisation to TRAIL was associated with enhanced activation of caspase-8, -9 and -3, elevated membrane expression levels of TRAIL-R2, cytochrome c release and G2/M arrest. Bortezomib 0-10 TNF receptor superfamily member 10b Homo sapiens 147-155 24212133-5 2014 Immunohistochemical analysis for DR4 and DR5 was carried out on formalin-fixed paraffin-embedded sections of skin tissues using avidin-biotin peroxidase method. Formaldehyde 64-72 TNF receptor superfamily member 10b Homo sapiens 41-44 24970682-11 2014 Furthermore, upregulation of death receptor 5 by metformin-mediated Sirt1 downregulation enhanced the sensitivity of wild-type p53 cancer cells to TRAIL-induced apoptosis. Metformin 49-58 TNF receptor superfamily member 10b Homo sapiens 29-45 25321472-9 2014 In conclusion, our results suggest that Med sensitizes myeloid leukemia cells to TRAIL-induced apoptosis through the upregulation of DR5 through activation of the ROS-JNK-CHOP pathway. ros 163-166 TNF receptor superfamily member 10b Homo sapiens 133-136 25310712-0 2014 Combination of TRAIL with bortezomib shifted apoptotic signaling from DR4 to DR5 death receptor by selective internalization and degradation of DR4. Bortezomib 26-36 TNF receptor superfamily member 10b Homo sapiens 77-80 25310712-5 2014 The expression of death (DR4 and DR5) as well as decoy (DcR1 and DcR2) receptors was upregulated in the both cell lines either by TRAIL or by bortezomib. Bortezomib 142-152 TNF receptor superfamily member 10b Homo sapiens 33-36 25310712-7 2014 Interestingly DR5-B variant of TRAIL which do not bind with DR4 receptor also induced elimination of DR4 from cell surface in combination with bortezomib indicating the ligand-independent mechanism of the receptor internalization. Bortezomib 143-153 TNF receptor superfamily member 10b Homo sapiens 14-17 24930757-6 2014 Ilimaquinone also reduced the cell survival proteins Bcl2 and Bcl-xL, while strongly up-regulating death receptor (DR) 4 and DR5 expression. ilimaquinone 0-12 TNF receptor superfamily member 10b Homo sapiens 125-128 24930757-7 2014 Induction of DR4 and DR5 by ilimaquinone was mediated through up-regulation of CCAAT/enhancer-binding protein homologous protein (CHOP). ilimaquinone 28-40 TNF receptor superfamily member 10b Homo sapiens 21-24 24930757-9 2014 Finally, the generation of ROS was required for CHOP and DR5 up-regulation by ilimaquinone. ros 27-30 TNF receptor superfamily member 10b Homo sapiens 57-60 24930757-9 2014 Finally, the generation of ROS was required for CHOP and DR5 up-regulation by ilimaquinone. ilimaquinone 78-90 TNF receptor superfamily member 10b Homo sapiens 57-60 24930757-10 2014 These results demonstrate that ilimaquinone enhanced the sensitivity of human colon cancer cells to TRAIL-induced apoptosis through ROS-ERK/p38 MAPK-CHOP-mediated up-regulation of DR4 and DR5 expression, suggesting that ilimaquinone could be developed into an adjuvant chemotherapeutic drug. ilimaquinone 31-43 TNF receptor superfamily member 10b Homo sapiens 188-191 24930757-10 2014 These results demonstrate that ilimaquinone enhanced the sensitivity of human colon cancer cells to TRAIL-induced apoptosis through ROS-ERK/p38 MAPK-CHOP-mediated up-regulation of DR4 and DR5 expression, suggesting that ilimaquinone could be developed into an adjuvant chemotherapeutic drug. ros 132-135 TNF receptor superfamily member 10b Homo sapiens 188-191 24188478-0 2014 Cariporide sensitizes leukemic cells to tumor necrosis factor related apoptosis-inducing ligand by up-regulation of death receptor 5 via endoplasmic reticulum stress-CCAAT/enhancer binding protein homologous protein dependent mechanism. cariporide 0-10 TNF receptor superfamily member 10b Homo sapiens 116-132 24188478-4 2014 The present study showed that treatment with the NHE1 inhibitor cariporide led to ER stress-induced up-regulation of the death receptor 5 (DR5) which is mediated by CHOP at the transcriptional level. cariporide 64-74 TNF receptor superfamily member 10b Homo sapiens 121-137 24188478-4 2014 The present study showed that treatment with the NHE1 inhibitor cariporide led to ER stress-induced up-regulation of the death receptor 5 (DR5) which is mediated by CHOP at the transcriptional level. cariporide 64-74 TNF receptor superfamily member 10b Homo sapiens 139-142 25017974-8 2014 Moreover, CCT327-induced induction of DR5 and DR4 was mediated by reactive oxygen species (ROS), and N-acetylcysteine (NAC) blocked the induction of DRs by CCT327. CCT327 10-16 TNF receptor superfamily member 10b Homo sapiens 38-41 25017974-8 2014 Moreover, CCT327-induced induction of DR5 and DR4 was mediated by reactive oxygen species (ROS), and N-acetylcysteine (NAC) blocked the induction of DRs by CCT327. Reactive Oxygen Species 66-89 TNF receptor superfamily member 10b Homo sapiens 38-41 25017974-8 2014 Moreover, CCT327-induced induction of DR5 and DR4 was mediated by reactive oxygen species (ROS), and N-acetylcysteine (NAC) blocked the induction of DRs by CCT327. Reactive Oxygen Species 91-94 TNF receptor superfamily member 10b Homo sapiens 38-41 24789319-0 2014 Tumoricidal activity of combining the agonistic DR5 antibody D-6 with cisplatin in C30 cisplatin-resistant ovarian cancer in vitro and in vivo. Cisplatin 87-96 TNF receptor superfamily member 10b Homo sapiens 48-51 24939851-2 2014 In this report, we demonstrate that the stress response gene ATF3 is required for endoplasmic reticulum stress-mediated DR5 induction upon zerumbone (ZER) and celecoxib (CCB) in human p53-deficient colorectal cancer cells. zerumbone 139-148 TNF receptor superfamily member 10b Homo sapiens 120-123 24939851-2 2014 In this report, we demonstrate that the stress response gene ATF3 is required for endoplasmic reticulum stress-mediated DR5 induction upon zerumbone (ZER) and celecoxib (CCB) in human p53-deficient colorectal cancer cells. Celecoxib 159-168 TNF receptor superfamily member 10b Homo sapiens 120-123 24939851-2 2014 In this report, we demonstrate that the stress response gene ATF3 is required for endoplasmic reticulum stress-mediated DR5 induction upon zerumbone (ZER) and celecoxib (CCB) in human p53-deficient colorectal cancer cells. Celecoxib 170-173 TNF receptor superfamily member 10b Homo sapiens 120-123 24939851-6 2014 A reporter assay demonstrated that at least two ATF/cAMP response element motifs as well as C/EBP homologous protein motif at the proximal region of the human DR5 gene promoter were required for ZER-induced DR5 gene transcription. Cyclic AMP 52-56 TNF receptor superfamily member 10b Homo sapiens 207-210 24919794-0 2014 Quinazoline analog HMJ-30 inhibits angiogenesis: involvement of endothelial cell apoptosis through ROS-JNK-mediated death receptor 5 signaling. Quinazolines 0-11 TNF receptor superfamily member 10b Homo sapiens 116-132 24919794-0 2014 Quinazoline analog HMJ-30 inhibits angiogenesis: involvement of endothelial cell apoptosis through ROS-JNK-mediated death receptor 5 signaling. 6-fluoro-2-(3-fluorophenyl)-4-(cyanoanilino)quinazoline 19-25 TNF receptor superfamily member 10b Homo sapiens 116-132 24919794-8 2014 We investigated the upstream of the death receptor pathway and further observed that the levels of death receptor 5 (DR5) and phosphorylated c-Jun N-terminal kinase (JNK) signals were upregulated in HUVECs following HMJ-30 challenge, which was confirmed by a JNK-specific inhibitor (SP600125). 6-fluoro-2-(3-fluorophenyl)-4-(cyanoanilino)quinazoline 216-222 TNF receptor superfamily member 10b Homo sapiens 99-115 24919794-8 2014 We investigated the upstream of the death receptor pathway and further observed that the levels of death receptor 5 (DR5) and phosphorylated c-Jun N-terminal kinase (JNK) signals were upregulated in HUVECs following HMJ-30 challenge, which was confirmed by a JNK-specific inhibitor (SP600125). 6-fluoro-2-(3-fluorophenyl)-4-(cyanoanilino)quinazoline 216-222 TNF receptor superfamily member 10b Homo sapiens 117-120 24919794-8 2014 We investigated the upstream of the death receptor pathway and further observed that the levels of death receptor 5 (DR5) and phosphorylated c-Jun N-terminal kinase (JNK) signals were upregulated in HUVECs following HMJ-30 challenge, which was confirmed by a JNK-specific inhibitor (SP600125). pyrazolanthrone 283-291 TNF receptor superfamily member 10b Homo sapiens 99-115 24919794-8 2014 We investigated the upstream of the death receptor pathway and further observed that the levels of death receptor 5 (DR5) and phosphorylated c-Jun N-terminal kinase (JNK) signals were upregulated in HUVECs following HMJ-30 challenge, which was confirmed by a JNK-specific inhibitor (SP600125). pyrazolanthrone 283-291 TNF receptor superfamily member 10b Homo sapiens 117-120 24919794-9 2014 Hence, HMJ-30-induced endothelial cell apoptosis involved the ROS/JNK-regulated DR5 pathway. 6-fluoro-2-(3-fluorophenyl)-4-(cyanoanilino)quinazoline 7-13 TNF receptor superfamily member 10b Homo sapiens 80-83 24919794-9 2014 Hence, HMJ-30-induced endothelial cell apoptosis involved the ROS/JNK-regulated DR5 pathway. Reactive Oxygen Species 62-65 TNF receptor superfamily member 10b Homo sapiens 80-83 25015549-0 2014 Upregulation of microRNA135a-3p and death receptor 5 plays a critical role in Tanshinone I sensitized prostate cancer cells to TRAIL induced apoptosis. tanshinone 78-88 TNF receptor superfamily member 10b Homo sapiens 36-52 25015549-5 2014 RT-PCR and RT-qPCR analyses confirmed that co-treatment of Tanshinone I and TRAIL up-regulated DR5 and microRNA 135a-3p at mRNA level or activity of DR5 promoter and attenuated phosphorylation of extracellular signal regulated kinases in PC-3. tanshinone 59-71 TNF receptor superfamily member 10b Homo sapiens 95-98 25015549-5 2014 RT-PCR and RT-qPCR analyses confirmed that co-treatment of Tanshinone I and TRAIL up-regulated DR5 and microRNA 135a-3p at mRNA level or activity of DR5 promoter and attenuated phosphorylation of extracellular signal regulated kinases in PC-3. tanshinone 59-71 TNF receptor superfamily member 10b Homo sapiens 149-152 25015549-6 2014 Conversely, the silencing of DR5 blocked the increased cytotoxicity, sub G1 population and PARP cleavages induced by co-treatment of Tanshinone I and TRAIL. tanshinone 133-145 TNF receptor superfamily member 10b Homo sapiens 29-32 25015549-8 2014 Overall, our findings suggest that Tanshinone I enhances TRAIL mediated apoptosis via upregulation of miR135a-3p mediated DR5 in prostate cancer cells as a potent TRAIL sensitizer. tanshinone 35-47 TNF receptor superfamily member 10b Homo sapiens 122-125 25033286-6 2014 In the KatoIII cell line (TP53-null), DR5 silencing markedly attenuated tenovin-6-induced apoptosis, suggesting that the pivotal mechanism behind its antitumor effects is based on activation of the death receptor signal pathway. tenovin-6 72-81 TNF receptor superfamily member 10b Homo sapiens 38-41 24973666-6 2014 Cordycepin-induced cell death was also associated with induction of Fas and death receptor 5, activation of caspase-8, and truncation of Bid (tBid), suggesting that tBid might serve to connect activation of both the mitochondrial-mediated intrinsic and death receptor-mediated extrinsic apoptotic pathways. cordycepin 0-10 TNF receptor superfamily member 10b Homo sapiens 76-92 25102912-0 2014 Myeloid zinc finger 1 mediates sulindac sulfide-induced upregulation of death receptor 5 of human colon cancer cells. sulindac sulfide 31-47 TNF receptor superfamily member 10b Homo sapiens 72-88 25102912-2 2014 Sulindac sulfide had been shown to induce the expression of death receptor 5 (DR5), a receptor for TRAIL, and sensitize cancer cells to TRAIL-induced apoptosis; however, the molecular mechanism underlying the upregulation of DR5 has not yet been elucidated. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 60-76 25102912-2 2014 Sulindac sulfide had been shown to induce the expression of death receptor 5 (DR5), a receptor for TRAIL, and sensitize cancer cells to TRAIL-induced apoptosis; however, the molecular mechanism underlying the upregulation of DR5 has not yet been elucidated. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 78-81 25102912-2 2014 Sulindac sulfide had been shown to induce the expression of death receptor 5 (DR5), a receptor for TRAIL, and sensitize cancer cells to TRAIL-induced apoptosis; however, the molecular mechanism underlying the upregulation of DR5 has not yet been elucidated. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 225-228 25102912-3 2014 We demonstrate here that myeloid zinc finger 1 (MZF1) mediates the induction of DR5 by sulindac sulfide. sulindac sulfide 87-103 TNF receptor superfamily member 10b Homo sapiens 80-83 25102912-4 2014 Sulindac sulfide induced the expression of DR5 at the protein and mRNA levels in colon cancer SW480 cells. sulindac sulfide 0-16 TNF receptor superfamily member 10b Homo sapiens 43-46 25102912-5 2014 Furthermore, sulindac sulfide increased DR5 promoter activity. sulindac sulfide 13-29 TNF receptor superfamily member 10b Homo sapiens 40-43 25102912-6 2014 We showed that sulindac sulfide stimulated DR5 promoter activity via the -301 to -253 region. sulindac sulfide 15-31 TNF receptor superfamily member 10b Homo sapiens 43-46 25102912-8 2014 Site-directed mutations in the site abrogated the enhancement in DR5 promoter activity by sulindac sulfide. sulindac sulfide 90-106 TNF receptor superfamily member 10b Homo sapiens 65-68 25102912-9 2014 MZF1 directly bound to the putative MZF1-binding site of the DR5 promoter and the binding was increased by sulindac sulfide. sulindac sulfide 107-123 TNF receptor superfamily member 10b Homo sapiens 61-64 25102912-10 2014 The expression of MZF1 was also increased by sulindac sulfide, and MZF1 siRNA attenuated the upregulation of DR5 by sulindac sulfide. sulindac sulfide 116-132 TNF receptor superfamily member 10b Homo sapiens 109-112 25102912-11 2014 These results indicate that sulindac sulfide induces the expression of DR5 by up-regulating MZF1. sulindac sulfide 28-44 TNF receptor superfamily member 10b Homo sapiens 71-74 24710190-5 2014 Bufalin also promoted the clustering of death receptor 4 (DR4) and DR5 in aggregated lipid rafts. bufalin 0-7 TNF receptor superfamily member 10b Homo sapiens 67-70 24710190-6 2014 The cholesterol-sequestering agent methyl-beta-cyclodextrin reversed the DR4 and DR5 clustering and reduced bufalin+TRAIL-induced apoptosis. Cholesterol 4-15 TNF receptor superfamily member 10b Homo sapiens 81-84 24710190-6 2014 The cholesterol-sequestering agent methyl-beta-cyclodextrin reversed the DR4 and DR5 clustering and reduced bufalin+TRAIL-induced apoptosis. methyl-beta-cyclodextrin 35-59 TNF receptor superfamily member 10b Homo sapiens 81-84 24789319-1 2014 A previous study by our group reported that the agonistic DR5 antibody D-6 was capable of triggering apoptosis in A2780 cisplatin-sensitive ovarian cancer cells and that this marked effect was enhanced by cisplatin in vitro. Cisplatin 120-129 TNF receptor superfamily member 10b Homo sapiens 58-61 24789319-1 2014 A previous study by our group reported that the agonistic DR5 antibody D-6 was capable of triggering apoptosis in A2780 cisplatin-sensitive ovarian cancer cells and that this marked effect was enhanced by cisplatin in vitro. Cisplatin 205-214 TNF receptor superfamily member 10b Homo sapiens 58-61 25233628-5 2014 MTT assay showed that, rClone30- hDR5 in combination with TRAIL more efficiently inhibited the tumor growth of HepG2 cells compared to rClone30-hDR5 or TRAIL in vitro. monooxyethylene trimethylolpropane tristearate 0-3 TNF receptor superfamily member 10b Homo sapiens 33-37 25052611-8 2014 The expression of DR5 on the surface of KG-1a treated with resveratrol was (9.05 +- 3.57)%, significantly higher than (3.11 +- 0.54)% of untreated KG-1a (P<0.05). Resveratrol 59-70 TNF receptor superfamily member 10b Homo sapiens 18-21 24662747-3 2014 Triptolide increases levels of death receptor DR5 and decreases the pro-survival FLICE-like inhibitory protein (c-FLIP), which contribute to the activation of caspase-8. triptolide 0-10 TNF receptor superfamily member 10b Homo sapiens 46-49 24882208-6 2014 E-cadherin bound specifically to ligated DR4/DR5, requiring extracellular cadherin domain 1 and calcium. Calcium 96-103 TNF receptor superfamily member 10b Homo sapiens 45-48 24104765-3 2014 Fluid-shear stress and dopamine receptor type-5 (DR5) agonist are among the few stimuli that require cilia for intracellular calcium signal transduction. Calcium 125-132 TNF receptor superfamily member 10b Homo sapiens 23-47 24104765-3 2014 Fluid-shear stress and dopamine receptor type-5 (DR5) agonist are among the few stimuli that require cilia for intracellular calcium signal transduction. Calcium 125-132 TNF receptor superfamily member 10b Homo sapiens 49-52 24104765-7 2014 While DR5-specific agonist induces calcium signaling mainly in the cilioplasm via ciliary CaV1.2, thrombin specifically induces cytosolic calcium signaling through the IP3 receptor. Calcium 35-42 TNF receptor superfamily member 10b Homo sapiens 6-9 24740347-5 2014 Treatment with low doses of salinomycin in combination with TRAIL augmented the activation of caspase-3 and increased TRAIL-R2 cell surface expression. salinomycin 28-39 TNF receptor superfamily member 10b Homo sapiens 118-126 24401154-7 2014 Moreover, OSU-A9 increased DR5 expression through a reactive oxygen species (ROS)-dependent mechanism. Reactive Oxygen Species 52-75 TNF receptor superfamily member 10b Homo sapiens 27-30 24401154-7 2014 Moreover, OSU-A9 increased DR5 expression through a reactive oxygen species (ROS)-dependent mechanism. Reactive Oxygen Species 77-80 TNF receptor superfamily member 10b Homo sapiens 27-30 24732433-4 2014 Through this route, WFA acted as an effective DR5 activator capable of potentiating the biologic effects of celecoxib, etoposide, and TRAIL. withaferin A 20-23 TNF receptor superfamily member 10b Homo sapiens 46-49 24732433-4 2014 Through this route, WFA acted as an effective DR5 activator capable of potentiating the biologic effects of celecoxib, etoposide, and TRAIL. Celecoxib 108-117 TNF receptor superfamily member 10b Homo sapiens 46-49 24732433-4 2014 Through this route, WFA acted as an effective DR5 activator capable of potentiating the biologic effects of celecoxib, etoposide, and TRAIL. Etoposide 119-128 TNF receptor superfamily member 10b Homo sapiens 46-49 24213373-0 2014 Indomethacin sensitizes TRAIL-resistant melanoma cells to TRAIL-induced apoptosis through ROS-mediated upregulation of death receptor 5 and downregulation of survivin. Indomethacin 0-12 TNF receptor superfamily member 10b Homo sapiens 119-135 24213373-0 2014 Indomethacin sensitizes TRAIL-resistant melanoma cells to TRAIL-induced apoptosis through ROS-mediated upregulation of death receptor 5 and downregulation of survivin. Reactive Oxygen Species 90-93 TNF receptor superfamily member 10b Homo sapiens 119-135 24213373-5 2014 Mechanistically, indomethacin induced cell surface expression of death receptor 5 (DR5) in melanoma cells and also in various types of cancer cells. Indomethacin 17-29 TNF receptor superfamily member 10b Homo sapiens 65-81 24213373-5 2014 Mechanistically, indomethacin induced cell surface expression of death receptor 5 (DR5) in melanoma cells and also in various types of cancer cells. Indomethacin 17-29 TNF receptor superfamily member 10b Homo sapiens 83-86 24213373-6 2014 DR5 knockdown abolished the enhancing effect of indomethacin on TRAIL responses. Indomethacin 48-60 TNF receptor superfamily member 10b Homo sapiens 0-3 24213373-7 2014 Induction of the DR5 by indomethacin was found to be p53 independent but dependent on the induction of CCAAT/enhancer-binding protein homologous protein (CHOP). Indomethacin 24-36 TNF receptor superfamily member 10b Homo sapiens 17-20 24213373-8 2014 Knockdown of CHOP abolished indomethacin-induced DR5 expression and the associated potentiation of TRAIL-mediated cell death. Indomethacin 28-40 TNF receptor superfamily member 10b Homo sapiens 49-52 24213373-9 2014 In addition, indomethacin-induced reactive oxygen species (ROS) production preceded upregulation of CHOP and DR5, and consequent sensitization of cells to TRAIL. Indomethacin 13-25 TNF receptor superfamily member 10b Homo sapiens 109-112 24213373-9 2014 In addition, indomethacin-induced reactive oxygen species (ROS) production preceded upregulation of CHOP and DR5, and consequent sensitization of cells to TRAIL. Reactive Oxygen Species 34-57 TNF receptor superfamily member 10b Homo sapiens 109-112 24213373-9 2014 In addition, indomethacin-induced reactive oxygen species (ROS) production preceded upregulation of CHOP and DR5, and consequent sensitization of cells to TRAIL. Reactive Oxygen Species 59-62 TNF receptor superfamily member 10b Homo sapiens 109-112 24491546-6 2014 PEITC enhances TRAIL-induced apoptosis through the downregulation of cell survival proteins and the upregulation of DR5 receptors through actions on the ROS-induced-p53. ros 153-156 TNF receptor superfamily member 10b Homo sapiens 116-119 24612139-4 2014 The quercetin induced expression of death receptor DR5 but did not affect expression of DR4 in cancer cells. Quercetin 4-13 TNF receptor superfamily member 10b Homo sapiens 51-54 24612139-5 2014 The induction of DR5 was mediated through activation of JNK and through upregulation of a transcription factor CCAAT enhancer-binding protein homologous protein (CHOP); as silencing of these signaling molecules abrogated the effect of quercetin. Quercetin 235-244 TNF receptor superfamily member 10b Homo sapiens 17-20 24612139-6 2014 Upregulation of DR5 was mediated through the generation of reactive oxygen species (ROS), as ROS scavengers reduced the effect of quercetin on JNK activation, CHOP upregulation, DR induction, TRAIL sensitization, downregulated the expression of cell survival proteins and upregulated the proapoptotic proteins. Reactive Oxygen Species 59-82 TNF receptor superfamily member 10b Homo sapiens 16-19 24612139-6 2014 Upregulation of DR5 was mediated through the generation of reactive oxygen species (ROS), as ROS scavengers reduced the effect of quercetin on JNK activation, CHOP upregulation, DR induction, TRAIL sensitization, downregulated the expression of cell survival proteins and upregulated the proapoptotic proteins. Reactive Oxygen Species 84-87 TNF receptor superfamily member 10b Homo sapiens 16-19 24612139-6 2014 Upregulation of DR5 was mediated through the generation of reactive oxygen species (ROS), as ROS scavengers reduced the effect of quercetin on JNK activation, CHOP upregulation, DR induction, TRAIL sensitization, downregulated the expression of cell survival proteins and upregulated the proapoptotic proteins. Reactive Oxygen Species 93-96 TNF receptor superfamily member 10b Homo sapiens 16-19 24612139-6 2014 Upregulation of DR5 was mediated through the generation of reactive oxygen species (ROS), as ROS scavengers reduced the effect of quercetin on JNK activation, CHOP upregulation, DR induction, TRAIL sensitization, downregulated the expression of cell survival proteins and upregulated the proapoptotic proteins. Quercetin 130-139 TNF receptor superfamily member 10b Homo sapiens 16-19 24612139-7 2014 Furthermore, quercetin enhances TRAIL mediated inhibition of tumor growth of human SKOV-3 xenograft was associated with induction of apoptosis, activation of caspase-3, CHOP and DR5. Quercetin 13-22 TNF receptor superfamily member 10b Homo sapiens 178-181 24612139-8 2014 Overall, our data suggest that quercetin enhances apoptotic death of ovarian cancer cells to TRAIL through upregulation of CHOP-induced DR5 expression following ROS mediated endoplasmic reticulum-stress. Quercetin 31-40 TNF receptor superfamily member 10b Homo sapiens 136-139 24740347-6 2014 TRAIL-R2 upmodulation was required for mediating the stimulatory effect of salinomycin on TRAIL-mediated apoptosis, since it was abrogated by siRNA-mediated TRAIL-R2 knockdown. salinomycin 75-86 TNF receptor superfamily member 10b Homo sapiens 0-8 24740347-6 2014 TRAIL-R2 upmodulation was required for mediating the stimulatory effect of salinomycin on TRAIL-mediated apoptosis, since it was abrogated by siRNA-mediated TRAIL-R2 knockdown. salinomycin 75-86 TNF receptor superfamily member 10b Homo sapiens 157-165 24535016-10 2014 Combined treatment of ZD55-TRAIL and MG132 (a proteasome inhibitor) robustly increased the expression of death receptor 5 (DR5), which enhanced the apoptosis response without significant toxicity to normal liver cells. zd55 22-26 TNF receptor superfamily member 10b Homo sapiens 105-121 24639349-8 2014 CTL resistance was due to DR5 downregulation and an inverted ratio of pro- to antiapoptotic molecules, both of which were reversed by the histone deacetylase inhibitor suberoylanilide hydroxanic acid. suberoylanilide hydroxanic acid 168-199 TNF receptor superfamily member 10b Homo sapiens 26-29 24535016-10 2014 Combined treatment of ZD55-TRAIL and MG132 (a proteasome inhibitor) robustly increased the expression of death receptor 5 (DR5), which enhanced the apoptosis response without significant toxicity to normal liver cells. zd55 22-26 TNF receptor superfamily member 10b Homo sapiens 123-126 24535016-10 2014 Combined treatment of ZD55-TRAIL and MG132 (a proteasome inhibitor) robustly increased the expression of death receptor 5 (DR5), which enhanced the apoptosis response without significant toxicity to normal liver cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 37-42 TNF receptor superfamily member 10b Homo sapiens 105-121 24535016-10 2014 Combined treatment of ZD55-TRAIL and MG132 (a proteasome inhibitor) robustly increased the expression of death receptor 5 (DR5), which enhanced the apoptosis response without significant toxicity to normal liver cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 37-42 TNF receptor superfamily member 10b Homo sapiens 123-126 24616705-5 2014 To determine the roles of DR5 and sensory cilia, we used dopamine to non-selectively and fenoldopam to selectively activate ciliary DR5. Fenoldopam 89-99 TNF receptor superfamily member 10b Homo sapiens 132-135 24286513-7 2014 The acetyl-L-carnitine-cisplatin combination caused reduced expression of genes Casp8, Fas, Casp1, Tnfrsf11b, Tnfrsf10b induced by cisplatin. Acetylcarnitine 4-22 TNF receptor superfamily member 10b Homo sapiens 110-119 24286513-7 2014 The acetyl-L-carnitine-cisplatin combination caused reduced expression of genes Casp8, Fas, Casp1, Tnfrsf11b, Tnfrsf10b induced by cisplatin. Cisplatin 23-32 TNF receptor superfamily member 10b Homo sapiens 110-119 24286513-7 2014 The acetyl-L-carnitine-cisplatin combination caused reduced expression of genes Casp8, Fas, Casp1, Tnfrsf11b, Tnfrsf10b induced by cisplatin. Cisplatin 131-140 TNF receptor superfamily member 10b Homo sapiens 110-119 24276615-2 2014 In the present study, we showed that co-treatment with troglitazone (TGZ), a synthetic ligand of peroxisome proliferator-activated receptor gamma (PPARgamma), and TRAIL synergistically induced apoptosis through DR5 upregulation in human colon cancer DLD-1 cells. Troglitazone 55-67 TNF receptor superfamily member 10b Homo sapiens 211-214 24276615-2 2014 In the present study, we showed that co-treatment with troglitazone (TGZ), a synthetic ligand of peroxisome proliferator-activated receptor gamma (PPARgamma), and TRAIL synergistically induced apoptosis through DR5 upregulation in human colon cancer DLD-1 cells. Troglitazone 69-72 TNF receptor superfamily member 10b Homo sapiens 211-214 24045365-0 2014 Digitoxin sensitizes glioma cells to TRAIL-mediated apoptosis by upregulation of death receptor 5 and downregulation of survivin. Digitoxin 0-9 TNF receptor superfamily member 10b Homo sapiens 81-97 24270523-7 2014 Co-treatment with MESC and an ERK inhibitor (PD98059) significantly increased the expression of DR5 to induce apoptosis in MC-3 cells. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 45-52 TNF receptor superfamily member 10b Homo sapiens 96-99 24270523-7 2014 Co-treatment with MESC and an ERK inhibitor (PD98059) significantly increased the expression of DR5 to induce apoptosis in MC-3 cells. mc-3 123-127 TNF receptor superfamily member 10b Homo sapiens 96-99 24387758-0 2014 Parthenolide induces apoptosis via TNFRSF10B and PMAIP1 pathways in human lung cancer cells. parthenolide 0-12 TNF receptor superfamily member 10b Homo sapiens 35-44 24387758-13 2014 CONCLUSION: We showed that parthenolide not only triggered extrinsic apoptosis by up-regulating TNFRSF10B and down-regulating CFLAR, but also induced intrinsic apoptosis through increasing the expression of PMAIP1 and decreasing the level of MCL1 in NSCLC cells. parthenolide 27-39 TNF receptor superfamily member 10b Homo sapiens 96-105 24270523-0 2014 Extracellular signal-regulated kinase inhibition is required for methanol extract of Smilax china L.-induced apoptosis through death receptor 5 in human oral mucoepidermoid carcinoma cells. Methanol 65-73 TNF receptor superfamily member 10b Homo sapiens 127-143 24436439-8 2014 We demonstrated that TMZ effectively increased the sensitivity to TRAIL-induced apoptosis via extracellular signal-regulated kinase-mediated upregulation of the death receptor 5 and downregulation of antiapoptotic proteins, such as X-linked inhibitor of apoptosis protein and cellular FLICE-inhibitory protein. Temozolomide 21-24 TNF receptor superfamily member 10b Homo sapiens 161-177 24045365-7 2014 We demonstrate that inactivation of survivin and death receptor 5 expression by DT is sufficient to restore TRAIL sensitivity in resistant glioma cells. Digitoxin 80-82 TNF receptor superfamily member 10b Homo sapiens 49-65 24734951-5 2014 Increasing expressions of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and death receptor 5 (DR5) were found in 2"-hydroxyflavanone-treated cells. 2'-hydroxyflavanone 131-150 TNF receptor superfamily member 10b Homo sapiens 94-110 25292102-6 2014 In addition, inhibition of IDO activity by the IDO inhibitor 1-MT or tryptophan significantly reduced IFN-gamma-induced apoptosis and death receptor 5 (DR5) expression, which suggests that IDO enzymatic activity plays an important role in the anti-NSCLC cancer effect of IFN-gamma. Tryptophan 69-79 TNF receptor superfamily member 10b Homo sapiens 134-150 25292102-6 2014 In addition, inhibition of IDO activity by the IDO inhibitor 1-MT or tryptophan significantly reduced IFN-gamma-induced apoptosis and death receptor 5 (DR5) expression, which suggests that IDO enzymatic activity plays an important role in the anti-NSCLC cancer effect of IFN-gamma. Tryptophan 69-79 TNF receptor superfamily member 10b Homo sapiens 152-155 24734951-5 2014 Increasing expressions of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and death receptor 5 (DR5) were found in 2"-hydroxyflavanone-treated cells. 2'-hydroxyflavanone 131-150 TNF receptor superfamily member 10b Homo sapiens 112-115 24287697-0 2013 Identification of DR5 as a critical, NF-kappaB-regulated mediator of Smac-induced apoptosis. smac 69-73 TNF receptor superfamily member 10b Homo sapiens 18-21 24008361-2 2013 We have previously observed that the saturated free fatty acids (FFAs) induce hepatocyte apoptosis in part via a death receptor 5 (DR5)-mediated signaling pathway. saturated free fatty acids 37-63 TNF receptor superfamily member 10b Homo sapiens 131-134 24136147-9 2013 Addition of the chemotherapeutic drug cisplatin to the combination further increased p53 and DR5 levels and rhTRAIL- and D269H/E195R-induced apoptosis. Cisplatin 38-47 TNF receptor superfamily member 10b Homo sapiens 93-96 24136147-12 2013 Addition of DNA-damaging agents such as cisplatin further enhances DR5-mediated apoptosis. Cisplatin 40-49 TNF receptor superfamily member 10b Homo sapiens 67-70 24008361-2 2013 We have previously observed that the saturated free fatty acids (FFAs) induce hepatocyte apoptosis in part via a death receptor 5 (DR5)-mediated signaling pathway. saturated free fatty acids 37-63 TNF receptor superfamily member 10b Homo sapiens 113-129 24002210-5 2013 As detected by western blot analysis and real-time RT-PCR, ibuprofen upregulated the expression of death receptor 5 (DR5), a TRAIL receptor. Ibuprofen 59-68 TNF receptor superfamily member 10b Homo sapiens 99-115 24045184-7 2013 (111)In-labeled CS-1008 was specifically taken up in mice bearing COLO 205 and WiDr tumors with prolonged tumor retention (26.25 +- 2.85%ID/g vs. 12.20 +- 2.24 at 168 hours post injection; n = 5, SD), and uptake correlated both with DR5 expression on tumor cells and antitumor activity. Cesium 16-18 TNF receptor superfamily member 10b Homo sapiens 233-236 23846485-6 2013 Curcumin-induced production of reactive oxygen species did not affect total expression of DR5 but it enhanced mobilization of DR5 to the plasma membrane. Curcumin 0-8 TNF receptor superfamily member 10b Homo sapiens 126-129 23846485-6 2013 Curcumin-induced production of reactive oxygen species did not affect total expression of DR5 but it enhanced mobilization of DR5 to the plasma membrane. Reactive Oxygen Species 31-54 TNF receptor superfamily member 10b Homo sapiens 126-129 24078627-4 2013 The limonoid induced expression of death receptor (DR) 5 and DR4 but did not affect expression of decoy receptors in cancer cells. Limonins 4-12 TNF receptor superfamily member 10b Homo sapiens 35-56 24078627-7 2013 The CHOP binding site on the DR5 gene was required for induction of DR5 by azadirone. azadirone 75-84 TNF receptor superfamily member 10b Homo sapiens 29-32 24078627-7 2013 The CHOP binding site on the DR5 gene was required for induction of DR5 by azadirone. azadirone 75-84 TNF receptor superfamily member 10b Homo sapiens 68-71 24078627-12 2013 Overall, this study indicates that azadirone can sensitize cancer cells to TRAIL through ROS-ERK-CHOP-mediated up-regulation of DR5 and DR4 signaling, down-regulation of cell survival proteins, and up-regulation of proapoptotic proteins. azadirone 35-44 TNF receptor superfamily member 10b Homo sapiens 128-131 24078627-12 2013 Overall, this study indicates that azadirone can sensitize cancer cells to TRAIL through ROS-ERK-CHOP-mediated up-regulation of DR5 and DR4 signaling, down-regulation of cell survival proteins, and up-regulation of proapoptotic proteins. Reactive Oxygen Species 89-92 TNF receptor superfamily member 10b Homo sapiens 128-131 24002210-5 2013 As detected by western blot analysis and real-time RT-PCR, ibuprofen upregulated the expression of death receptor 5 (DR5), a TRAIL receptor. Ibuprofen 59-68 TNF receptor superfamily member 10b Homo sapiens 117-120 24002210-6 2013 TRAIL-induced apoptosis enhanced by ibuprofen was effectively decreased by a caspase inhibitor and dominant-negative DR5. Ibuprofen 36-45 TNF receptor superfamily member 10b Homo sapiens 117-120 23732517-5 2013 Knockdown of DR5 and DR4 significantly (>85%) reduced the sensitizing effect of the AR inhibitor fidarestat on TRAIL-induced apoptosis. fidarestat 100-110 TNF receptor superfamily member 10b Homo sapiens 13-16 23700228-8 2013 Emodin-induced induction of DR5 was mediated through the generation of reactive oxygen species (ROS), as N-acetylcysteine blocked the induction of DR5 and the induction of apoptosis. Reactive Oxygen Species 71-94 TNF receptor superfamily member 10b Homo sapiens 28-31 24002210-0 2013 Ibuprofen enhances TRAIL-induced apoptosis through DR5 upregulation. Ibuprofen 0-9 TNF receptor superfamily member 10b Homo sapiens 51-54 23700228-8 2013 Emodin-induced induction of DR5 was mediated through the generation of reactive oxygen species (ROS), as N-acetylcysteine blocked the induction of DR5 and the induction of apoptosis. Reactive Oxygen Species 96-99 TNF receptor superfamily member 10b Homo sapiens 28-31 23700228-8 2013 Emodin-induced induction of DR5 was mediated through the generation of reactive oxygen species (ROS), as N-acetylcysteine blocked the induction of DR5 and the induction of apoptosis. Acetylcysteine 105-121 TNF receptor superfamily member 10b Homo sapiens 28-31 23700228-8 2013 Emodin-induced induction of DR5 was mediated through the generation of reactive oxygen species (ROS), as N-acetylcysteine blocked the induction of DR5 and the induction of apoptosis. Acetylcysteine 105-121 TNF receptor superfamily member 10b Homo sapiens 147-150 23857473-4 2013 Moreover, TRAIL receptors, such as DR4 and DR5 were significantly upregulated by CAPE treatment, and both DR4/Fc and DR5/Fc chimera markedly abrogated apoptosis induced by CAPE and TRAIL, demonstrating the critical role of these death receptors in combination-induced apoptosis. caffeic acid phenethyl ester 81-85 TNF receptor superfamily member 10b Homo sapiens 43-46 23857473-8 2013 In contrast, JNK-specific inhibition, by SP600125, triggered upregulation of DR4 and DR5, and sensitized SK-Hep1 cells to TRAIL, indicating that the CAPE-induced suppression of JNK may contribute to the sensitizing effect of CAPE through the upregulation of death receptors. caffeic acid phenethyl ester 149-153 TNF receptor superfamily member 10b Homo sapiens 85-88 23504627-5 2013 The central role of the TRAIL signaling pathway was further supported by an increased expression of TRAIL-R2 on DOX-treated tumor cells and by downregulation of cellular FLICE inhibitory protein, the inhibitors of death receptor-mediated apoptosis. Doxorubicin 112-115 TNF receptor superfamily member 10b Homo sapiens 100-108 23857473-4 2013 Moreover, TRAIL receptors, such as DR4 and DR5 were significantly upregulated by CAPE treatment, and both DR4/Fc and DR5/Fc chimera markedly abrogated apoptosis induced by CAPE and TRAIL, demonstrating the critical role of these death receptors in combination-induced apoptosis. caffeic acid phenethyl ester 172-176 TNF receptor superfamily member 10b Homo sapiens 43-46 23857473-4 2013 Moreover, TRAIL receptors, such as DR4 and DR5 were significantly upregulated by CAPE treatment, and both DR4/Fc and DR5/Fc chimera markedly abrogated apoptosis induced by CAPE and TRAIL, demonstrating the critical role of these death receptors in combination-induced apoptosis. caffeic acid phenethyl ester 172-176 TNF receptor superfamily member 10b Homo sapiens 117-120 23912708-8 2013 Pretreatment with cisplatin significantly enhanced the expression of DR5. Cisplatin 18-27 TNF receptor superfamily member 10b Homo sapiens 69-72 23754197-4 2013 We demonstrated that Asp induced cell death through the activation of TRAIL DR4/DR5 death receptors leading to the activation of caspase-8 and caspase-3 and to cell apoptosis. Aspartic Acid 21-24 TNF receptor superfamily member 10b Homo sapiens 80-83 23615398-5 2013 Interestingly, other polyether antibiotics, such as salinomycin, nigericin, narasin and lasalocid A, also stimulated TRAIL-mediated apoptosis in glioma cells via ER stress, CHOP-mediated DR5 upregulation and c-FLIP downregulation. polyether antibiotics 21-42 TNF receptor superfamily member 10b Homo sapiens 187-190 23615398-5 2013 Interestingly, other polyether antibiotics, such as salinomycin, nigericin, narasin and lasalocid A, also stimulated TRAIL-mediated apoptosis in glioma cells via ER stress, CHOP-mediated DR5 upregulation and c-FLIP downregulation. salinomycin 52-63 TNF receptor superfamily member 10b Homo sapiens 187-190 23615398-5 2013 Interestingly, other polyether antibiotics, such as salinomycin, nigericin, narasin and lasalocid A, also stimulated TRAIL-mediated apoptosis in glioma cells via ER stress, CHOP-mediated DR5 upregulation and c-FLIP downregulation. Nigericin 65-74 TNF receptor superfamily member 10b Homo sapiens 187-190 23615398-5 2013 Interestingly, other polyether antibiotics, such as salinomycin, nigericin, narasin and lasalocid A, also stimulated TRAIL-mediated apoptosis in glioma cells via ER stress, CHOP-mediated DR5 upregulation and c-FLIP downregulation. narasin 76-83 TNF receptor superfamily member 10b Homo sapiens 187-190 23615398-5 2013 Interestingly, other polyether antibiotics, such as salinomycin, nigericin, narasin and lasalocid A, also stimulated TRAIL-mediated apoptosis in glioma cells via ER stress, CHOP-mediated DR5 upregulation and c-FLIP downregulation. Lasalocid 88-99 TNF receptor superfamily member 10b Homo sapiens 187-190 23994633-6 2013 The results indicated that combination of MG132 and TRAIL had the effect of up-regulating expression of DR5, caspase-3, caspase-8 and p27(kip1), down-regulating expression of MMP-9 and inducing apoptosis as well as suppressing the ability of invasion of OS732 cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 42-47 TNF receptor superfamily member 10b Homo sapiens 104-107 23554101-0 2013 The chalcone 2"-hydroxy-4",5"-dimethoxychalcone activates death receptor 5 pathway and leads to apoptosis in human nonsmall cell lung cancer cells. Chalcone 4-12 TNF receptor superfamily member 10b Homo sapiens 58-74 23605004-0 2013 Induction of death receptor 5 expression in tumor vasculature by perifosine restores the vascular disruption activity of TRAIL-expressing CD34(+) cells. perifosine 65-75 TNF receptor superfamily member 10b Homo sapiens 13-29 23428290-0 2013 Carbenoxolone enhances TRAIL-induced apoptosis through the upregulation of death receptor 5 and inhibition of gap junction intercellular communication in human glioma. Carbenoxolone 0-13 TNF receptor superfamily member 10b Homo sapiens 75-91 23428290-7 2013 CBX enhanced TRAIL-induced apoptosis through upregulation of death receptor 5, blockade of GJ intercellular communication, and downregulation of connexin 43. Carbenoxolone 0-3 TNF receptor superfamily member 10b Homo sapiens 61-77 23435998-0 2013 Upregulation of DR5 receptor by the diaminothiazole DAT1 [4-amino-5-benzoyl-2-(4-methoxy phenyl amino) thiazole] triggers an independent extrinsic pathway of apoptosis in colon cancer cells with compromised pro and antiapoptotic proteins. Thiazole-2,4-diamine 36-51 TNF receptor superfamily member 10b Homo sapiens 16-19 23435998-0 2013 Upregulation of DR5 receptor by the diaminothiazole DAT1 [4-amino-5-benzoyl-2-(4-methoxy phenyl amino) thiazole] triggers an independent extrinsic pathway of apoptosis in colon cancer cells with compromised pro and antiapoptotic proteins. 4-amino-5-benzoyl-2-(4-methoxyphenylamino)thiazole 58-111 TNF receptor superfamily member 10b Homo sapiens 16-19 23620163-6 2013 Notably, while SAHA induced upregulation of death receptor 4 (DR4) and DR5, HHT upregulated tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) expression in a dose-dependent manner. Vorinostat 15-19 TNF receptor superfamily member 10b Homo sapiens 71-74 23554101-0 2013 The chalcone 2"-hydroxy-4",5"-dimethoxychalcone activates death receptor 5 pathway and leads to apoptosis in human nonsmall cell lung cancer cells. 2'-hydroxy-4',5'-dimethoxychalcone 13-47 TNF receptor superfamily member 10b Homo sapiens 58-74 23554101-6 2013 In conclusion, HDMC induces apoptosis in human nonsmall cell lung cancer cells via activation of DR5 signaling pathway, and ROS-mediated ATF4-CHOP axis is involved in the process. 2'-hydroxy-4',5'-dimethoxychalcone 15-19 TNF receptor superfamily member 10b Homo sapiens 97-100 23499682-6 2013 Indeed, treatment with 2-DG resulted in upregulation of TRAIL receptor 2 (TRAIL-R2), downregulation of cIAP1 and XIAP, and reduction in RIP1. Deoxyglucose 23-27 TNF receptor superfamily member 10b Homo sapiens 56-72 23523585-7 2013 HMJ-38-influenced HUVECs were performed by determining the oxidative stress (ROS production) and ATM/p53-modulated Fas and DR4/DR5 signals that were examined by flow cytometry, Western blotting, siRNA and real-time RT-PCR analyses, respectively. 2-(3'-methoxyphenyl)-6-pyrrolidinyl-4-quinazolinone 0-6 TNF receptor superfamily member 10b Homo sapiens 127-130 23696862-6 2013 Additionally, DTCD stimulated extracellular signal-regulated kinase (ERK) activation, while the ERK inhibitor PD98059 blocked DTCD-induced DR5 expression and suppressed binding of Sp1 to the DR5 promoter. dtcd 126-130 TNF receptor superfamily member 10b Homo sapiens 139-142 23696862-6 2013 Additionally, DTCD stimulated extracellular signal-regulated kinase (ERK) activation, while the ERK inhibitor PD98059 blocked DTCD-induced DR5 expression and suppressed binding of Sp1 to the DR5 promoter. dtcd 126-130 TNF receptor superfamily member 10b Homo sapiens 191-194 23608376-0 2013 Casticin induces caspase-mediated apoptosis via activation of mitochondrial pathway and upregulation of DR5 in human lung cancer cells. casticin 0-8 TNF receptor superfamily member 10b Homo sapiens 104-107 23608376-1 2013 OBJECTIVE: To assess if casticin induces caspase-mediated apoptosis via activation of mitochondrial pathway and upregulation of DR5 in human lung cancer cells. casticin 24-32 TNF receptor superfamily member 10b Homo sapiens 128-131 23608376-12 2013 In H157 cell line, casticin increased expression of DR5 at protein levels but not affect the expression of DR4. casticin 19-27 TNF receptor superfamily member 10b Homo sapiens 52-55 23608376-13 2013 The pretreatment with DR5/Fc chimera protein effectively attenuated casticin-induced apoptosis in H157 cells. casticin 68-76 TNF receptor superfamily member 10b Homo sapiens 22-25 23608376-15 2013 CONCLUSIONS: Our results demonstrate that casticin induces caspase-mediated apoptosis via activation of mitochondrial pathway and upregulation of DR5 in human lung cancer cells. casticin 42-50 TNF receptor superfamily member 10b Homo sapiens 146-149 23696862-4 2013 In vitro, DTCD enhanced TRAIL-induced cytotoxicity in human ovarian cancer cells through up-regulation of DR5. dtcd 10-14 TNF receptor superfamily member 10b Homo sapiens 106-109 23696862-5 2013 Luciferase analysis and CHIP assays showed that DTCD increased DR5 promoter activity via Sp1 activation. dtcd 48-52 TNF receptor superfamily member 10b Homo sapiens 63-66 23696862-6 2013 Additionally, DTCD stimulated extracellular signal-regulated kinase (ERK) activation, while the ERK inhibitor PD98059 blocked DTCD-induced DR5 expression and suppressed binding of Sp1 to the DR5 promoter. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 110-117 TNF receptor superfamily member 10b Homo sapiens 139-142 23696862-6 2013 Additionally, DTCD stimulated extracellular signal-regulated kinase (ERK) activation, while the ERK inhibitor PD98059 blocked DTCD-induced DR5 expression and suppressed binding of Sp1 to the DR5 promoter. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 110-117 TNF receptor superfamily member 10b Homo sapiens 191-194 23396090-10 2013 In contrast, liposomal CDDP induced expression of genes from DNA damage pathways and several genes of the extrinsic pathway of apoptosis (TNFRSF10B-DR5, CD70-TNFSF7). Cisplatin 23-27 TNF receptor superfamily member 10b Homo sapiens 138-151 23499682-6 2013 Indeed, treatment with 2-DG resulted in upregulation of TRAIL receptor 2 (TRAIL-R2), downregulation of cIAP1 and XIAP, and reduction in RIP1. Deoxyglucose 23-27 TNF receptor superfamily member 10b Homo sapiens 74-82 23001792-5 2013 This increased apoptosis in human colon cancer cells after RA or DZNep treatment was associated with a ~2.5-fold increase in TNFRSF10B (DR5) transcript levels and a 42% reduction in the H3K27me3 epigenetic mark at the TNFRSF10B promoter after DZNep addition. 3-deazaneplanocin 65-70 TNF receptor superfamily member 10b Homo sapiens 125-134 23564786-0 2013 Salinomycin induces apoptosis via death receptor-5 up-regulation in cisplatin-resistant ovarian cancer cells. salinomycin 0-11 TNF receptor superfamily member 10b Homo sapiens 34-50 23564786-0 2013 Salinomycin induces apoptosis via death receptor-5 up-regulation in cisplatin-resistant ovarian cancer cells. Cisplatin 68-77 TNF receptor superfamily member 10b Homo sapiens 34-50 23564786-9 2013 Salinomycin increased the expression of death receptor-5 (DR5), caspase-8 and Fas-associated protein with death domain (FADD). salinomycin 0-11 TNF receptor superfamily member 10b Homo sapiens 40-56 23564786-9 2013 Salinomycin increased the expression of death receptor-5 (DR5), caspase-8 and Fas-associated protein with death domain (FADD). salinomycin 0-11 TNF receptor superfamily member 10b Homo sapiens 58-61 23564786-12 2013 CONCLUSION: These findings provide important insights regarding the activation of caspase-8 and DR5, to our knowledge, for the first time in salinomycin-treated cisplatin-resistant ovarian cancer and demonstrate that salinomycin could be a prominent anticancer agent. salinomycin 141-152 TNF receptor superfamily member 10b Homo sapiens 96-99 23564786-12 2013 CONCLUSION: These findings provide important insights regarding the activation of caspase-8 and DR5, to our knowledge, for the first time in salinomycin-treated cisplatin-resistant ovarian cancer and demonstrate that salinomycin could be a prominent anticancer agent. Cisplatin 161-170 TNF receptor superfamily member 10b Homo sapiens 96-99 23564786-12 2013 CONCLUSION: These findings provide important insights regarding the activation of caspase-8 and DR5, to our knowledge, for the first time in salinomycin-treated cisplatin-resistant ovarian cancer and demonstrate that salinomycin could be a prominent anticancer agent. salinomycin 217-228 TNF receptor superfamily member 10b Homo sapiens 96-99 23378009-4 2013 We observed that LiCl significantly enhanced A549 cells apoptosis through up-regulation of death receptors DR4 and DR5 and activation of caspase cascades. Lithium Chloride 17-21 TNF receptor superfamily member 10b Homo sapiens 115-118 23001792-5 2013 This increased apoptosis in human colon cancer cells after RA or DZNep treatment was associated with a ~2.5-fold increase in TNFRSF10B (DR5) transcript levels and a 42% reduction in the H3K27me3 epigenetic mark at the TNFRSF10B promoter after DZNep addition. 3-deazaneplanocin 65-70 TNF receptor superfamily member 10b Homo sapiens 136-139 23001792-5 2013 This increased apoptosis in human colon cancer cells after RA or DZNep treatment was associated with a ~2.5-fold increase in TNFRSF10B (DR5) transcript levels and a 42% reduction in the H3K27me3 epigenetic mark at the TNFRSF10B promoter after DZNep addition. 3-deazaneplanocin 65-70 TNF receptor superfamily member 10b Homo sapiens 218-227 23644112-4 2013 KB cells treated with 5 mmol/L 2-DG with or without TRAIL for 0, 6, 16, or 24 h were examined with Western blotting for protein expressions of death receptor 5 (DR5) and caspase-3. Deoxyglucose 31-35 TNF receptor superfamily member 10b Homo sapiens 143-159 23644112-4 2013 KB cells treated with 5 mmol/L 2-DG with or without TRAIL for 0, 6, 16, or 24 h were examined with Western blotting for protein expressions of death receptor 5 (DR5) and caspase-3. Deoxyglucose 31-35 TNF receptor superfamily member 10b Homo sapiens 161-164 23644112-7 2013 2-DG treatment markedly up-regulated DR5 and caspase-3 expression and enhanced the inhibitory effect of TRAIL on cell colony formation. Deoxyglucose 0-4 TNF receptor superfamily member 10b Homo sapiens 37-40 23644112-8 2013 CONCLUSION: 2-DG sensitizes oral cancer cells to TRAIL- induced apoptosis by up-regulating DR5 and caspase-3 expressions. Deoxyglucose 12-16 TNF receptor superfamily member 10b Homo sapiens 91-94 22665066-4 2013 Second-generation tyrosine kinase inhibitors (dasatinib and nilotinib) and short hairpin RNA against bcr-abl also downregulated DR4 and DR5 expression in Ph(+) leukemia cells, and transfection of bcr-abl into a Ph(-) leukemia cell line induced DR4 and DR5 expression, which was abrogated by imatinib treatment. Dasatinib 46-55 TNF receptor superfamily member 10b Homo sapiens 136-139 21803611-10 2013 The isoflavone sensitized the TRAIL-resistant LNCaP cells through the inhibition of transcription factor NF-kappaB(p65) activity, increased the expression of the death receptor TRAIL-R2 (DR5), and disrupted mitochondrial membrane potential (DeltaPsim). Isoflavones 4-14 TNF receptor superfamily member 10b Homo sapiens 177-185 21803611-10 2013 The isoflavone sensitized the TRAIL-resistant LNCaP cells through the inhibition of transcription factor NF-kappaB(p65) activity, increased the expression of the death receptor TRAIL-R2 (DR5), and disrupted mitochondrial membrane potential (DeltaPsim). Isoflavones 4-14 TNF receptor superfamily member 10b Homo sapiens 187-190 22665066-3 2013 Here we demonstrate that imatinib specifically downregulated DR4 and DR5 expression in cell lines and clinical samples of Ph(+) leukemia. Imatinib Mesylate 25-33 TNF receptor superfamily member 10b Homo sapiens 69-72 22665066-4 2013 Second-generation tyrosine kinase inhibitors (dasatinib and nilotinib) and short hairpin RNA against bcr-abl also downregulated DR4 and DR5 expression in Ph(+) leukemia cells, and transfection of bcr-abl into a Ph(-) leukemia cell line induced DR4 and DR5 expression, which was abrogated by imatinib treatment. nilotinib 60-69 TNF receptor superfamily member 10b Homo sapiens 136-139 23082969-0 2013 (E)-2,4-bis(p-hydroxyphenyl)-2-butenal has an antiproliferative effect on NSCLC cells induced by p38 MAPK-mediated suppression of NF-kappaB and up-regulation of TNFRSF10B (DR5). SCHEMBL6784264 0-38 TNF receptor superfamily member 10b Homo sapiens 161-170 23352978-7 2013 Both I3C and EGCG significantly increased caspase-3 activity, DNA fragmentation percentage, DR4 and DR5 protein expression as well as decreased Bcl-2 protein expression when compared to control groups. epigallocatechin gallate 13-17 TNF receptor superfamily member 10b Homo sapiens 100-103 23082969-0 2013 (E)-2,4-bis(p-hydroxyphenyl)-2-butenal has an antiproliferative effect on NSCLC cells induced by p38 MAPK-mediated suppression of NF-kappaB and up-regulation of TNFRSF10B (DR5). SCHEMBL6784264 0-38 TNF receptor superfamily member 10b Homo sapiens 172-175 23082969-9 2013 Even though all the MAPKs were activated, only pretreatment with a p38 MAPK inhibitor reversed (E)-2,4-bis(p-hydroxyphenyl)-2-butenal-induced cell growth inhibition, increase in cleaved caspase-3, -9 and TNFRSF10B expression, and NF-kappaB inactivation. SCHEMBL6784264 95-133 TNF receptor superfamily member 10b Homo sapiens 204-213 23082969-8 2013 Of the death receptors activated, only TNFRSF10B knock down with siRNA reversed the effect of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal. SCHEMBL6784264 94-132 TNF receptor superfamily member 10b Homo sapiens 39-48 23082969-10 2013 CONCLUSIONS AND IMPLICATIONS: (E)-2,4-bis(p-hydroxyphenyl)-2-butenal induces apoptosis in NSCLC cells by p38 MAPK-mediated suppression of NF-kappaB and activation of TNFRSF10B, which then activates the caspase-3 and caspase-9 pathways. SCHEMBL6784264 30-68 TNF receptor superfamily member 10b Homo sapiens 166-175 22978563-0 2013 Sorafenib and its derivative SC-49 sensitize hepatocellular carcinoma cells to CS-1008, a humanized anti-TNFRSF10B (DR5) antibody. Sorafenib 0-9 TNF receptor superfamily member 10b Homo sapiens 105-114 23291741-10 2013 The expression levels of DR5 and p-AKT proteins were increased and decreased, respectively, as a result of the effects of sphingoid bases from sea cucumber. sphingoid bases 134-149 TNF receptor superfamily member 10b Homo sapiens 25-28 23291741-11 2013 The results indicate that sphingoid bases from sea cucumber induce apoptosis in HepG2 cells through upregulation of DR5, Bax, GADD45 and PPARgamma and downregulation of p-AKT. sphingoid bases 26-41 TNF receptor superfamily member 10b Homo sapiens 128-131 23261452-0 2013 Lipoxygenase inhibitor MK886 potentiates TRAIL-induced apoptosis through CHOP- and p38 MAPK-mediated up-regulation of death receptor 5 in malignant glioma. MK-886 23-28 TNF receptor superfamily member 10b Homo sapiens 118-134 23261452-5 2013 We found that MK886 sensitized glioma cells to TRAIL-induced apoptosis by upregulating the death receptor 5 (DR5) and that specific knockdown of DR5 attenuated cell death. MK-886 14-19 TNF receptor superfamily member 10b Homo sapiens 91-107 23261452-5 2013 We found that MK886 sensitized glioma cells to TRAIL-induced apoptosis by upregulating the death receptor 5 (DR5) and that specific knockdown of DR5 attenuated cell death. MK-886 14-19 TNF receptor superfamily member 10b Homo sapiens 109-112 23261452-6 2013 The mechanisms underlying this sensitization involved activation of the MK886-induced p38 mitogen-activated protein kinase (MAPK) pathway and subsequent DR5 overexpression. MK-886 72-77 TNF receptor superfamily member 10b Homo sapiens 153-156 23261452-8 2013 Taken together, our findings indicate that the increased expression of DR5 in a p38 MAPK-dependent manner plays an important role in the sensitization of MK886 to TRAIL-induced apoptosis. MK-886 154-159 TNF receptor superfamily member 10b Homo sapiens 71-74 23291741-0 2013 Sphingoid bases from sea cucumber induce apoptosis in human hepatoma HepG2 cells through p-AKT and DR5. Sphingosine 0-9 TNF receptor superfamily member 10b Homo sapiens 111-114 23429289-6 2013 Consequently, when we generated MSCs that secreted TRAIL-R2-specific variants of soluble TRAIL (sTRAIL), we found that such engineered MSCs, labeled MSC.sTRAIL(DR5), had enhanced antitumor activity in combination with 5-FU when compared with MSC.sTRAIL. Fluorouracil 218-222 TNF receptor superfamily member 10b Homo sapiens 51-59 23429289-6 2013 Consequently, when we generated MSCs that secreted TRAIL-R2-specific variants of soluble TRAIL (sTRAIL), we found that such engineered MSCs, labeled MSC.sTRAIL(DR5), had enhanced antitumor activity in combination with 5-FU when compared with MSC.sTRAIL. Fluorouracil 218-222 TNF receptor superfamily member 10b Homo sapiens 160-163 22978563-0 2013 Sorafenib and its derivative SC-49 sensitize hepatocellular carcinoma cells to CS-1008, a humanized anti-TNFRSF10B (DR5) antibody. Sorafenib 0-9 TNF receptor superfamily member 10b Homo sapiens 116-119 22978563-2 2013 Here, we report that sorafenib and SC-49 sensitize HCC cells to CS-1008, a novel anti-human death receptor 5 (TNFRSF10B) antibody. Sorafenib 21-30 TNF receptor superfamily member 10b Homo sapiens 92-108 22978563-2 2013 Here, we report that sorafenib and SC-49 sensitize HCC cells to CS-1008, a novel anti-human death receptor 5 (TNFRSF10B) antibody. Sorafenib 21-30 TNF receptor superfamily member 10b Homo sapiens 110-119 23258590-9 2013 Further studies showed that nystatin partially prevented lipid raft aggregation and DR4 and DR5 clustering and reduced apoptosis in H460 cells again. Nystatin 28-36 TNF receptor superfamily member 10b Homo sapiens 92-95 22960596-3 2013 Exogenous addition of either mevalonate or geranylgeranyl pyrophosphate (GGPP) inhibited SVA activation of JNK/CHOP/DR5 pro-apoptotic pathway, indicating that activation of JNK/CHOP/DR5 pro-apoptotic pathway is dependent on SVA inhibition of 3-hydroxy-3-methylglutaryl Coenzyme A (HMG-CoA) reductase and its intermediate GGPP. geranylgeranyl pyrophosphate 43-71 TNF receptor superfamily member 10b Homo sapiens 116-119 22960596-3 2013 Exogenous addition of either mevalonate or geranylgeranyl pyrophosphate (GGPP) inhibited SVA activation of JNK/CHOP/DR5 pro-apoptotic pathway, indicating that activation of JNK/CHOP/DR5 pro-apoptotic pathway is dependent on SVA inhibition of 3-hydroxy-3-methylglutaryl Coenzyme A (HMG-CoA) reductase and its intermediate GGPP. geranylgeranyl pyrophosphate 43-71 TNF receptor superfamily member 10b Homo sapiens 182-185 22960596-3 2013 Exogenous addition of either mevalonate or geranylgeranyl pyrophosphate (GGPP) inhibited SVA activation of JNK/CHOP/DR5 pro-apoptotic pathway, indicating that activation of JNK/CHOP/DR5 pro-apoptotic pathway is dependent on SVA inhibition of 3-hydroxy-3-methylglutaryl Coenzyme A (HMG-CoA) reductase and its intermediate GGPP. geranylgeranyl pyrophosphate 73-77 TNF receptor superfamily member 10b Homo sapiens 116-119 22960596-3 2013 Exogenous addition of either mevalonate or geranylgeranyl pyrophosphate (GGPP) inhibited SVA activation of JNK/CHOP/DR5 pro-apoptotic pathway, indicating that activation of JNK/CHOP/DR5 pro-apoptotic pathway is dependent on SVA inhibition of 3-hydroxy-3-methylglutaryl Coenzyme A (HMG-CoA) reductase and its intermediate GGPP. geranylgeranyl pyrophosphate 73-77 TNF receptor superfamily member 10b Homo sapiens 182-185 22960596-0 2013 Simvastatin inhibition of mevalonate pathway induces apoptosis in human breast cancer cells via activation of JNK/CHOP/DR5 signaling pathway. Simvastatin 0-11 TNF receptor superfamily member 10b Homo sapiens 119-122 22960596-0 2013 Simvastatin inhibition of mevalonate pathway induces apoptosis in human breast cancer cells via activation of JNK/CHOP/DR5 signaling pathway. Mevalonic Acid 26-36 TNF receptor superfamily member 10b Homo sapiens 119-122 22960596-1 2013 Simvastatin (SVA) was shown to up-regulate expression of death receptor-5 (DR5), CCAAT/enhancer binding protein homologous protein (CHOP) and phosphorylated c-Jun N-terminal kinase (pJNK) in human breast cancer cell lines. Simvastatin 0-11 TNF receptor superfamily member 10b Homo sapiens 57-73 22960596-1 2013 Simvastatin (SVA) was shown to up-regulate expression of death receptor-5 (DR5), CCAAT/enhancer binding protein homologous protein (CHOP) and phosphorylated c-Jun N-terminal kinase (pJNK) in human breast cancer cell lines. Simvastatin 0-11 TNF receptor superfamily member 10b Homo sapiens 75-78 22960596-3 2013 Exogenous addition of either mevalonate or geranylgeranyl pyrophosphate (GGPP) inhibited SVA activation of JNK/CHOP/DR5 pro-apoptotic pathway, indicating that activation of JNK/CHOP/DR5 pro-apoptotic pathway is dependent on SVA inhibition of 3-hydroxy-3-methylglutaryl Coenzyme A (HMG-CoA) reductase and its intermediate GGPP. Mevalonic Acid 29-39 TNF receptor superfamily member 10b Homo sapiens 116-119 22960596-3 2013 Exogenous addition of either mevalonate or geranylgeranyl pyrophosphate (GGPP) inhibited SVA activation of JNK/CHOP/DR5 pro-apoptotic pathway, indicating that activation of JNK/CHOP/DR5 pro-apoptotic pathway is dependent on SVA inhibition of 3-hydroxy-3-methylglutaryl Coenzyme A (HMG-CoA) reductase and its intermediate GGPP. Mevalonic Acid 29-39 TNF receptor superfamily member 10b Homo sapiens 182-185 23348585-0 2013 Salirasib sensitizes hepatocarcinoma cells to TRAIL-induced apoptosis through DR5 and survivin-dependent mechanisms. farnesylthiosalicylic acid 0-9 TNF receptor superfamily member 10b Homo sapiens 78-81 23135489-0 2013 Casticin, a flavonoid, potentiates TRAIL-induced apoptosis through modulation of anti-apoptotic proteins and death receptor 5 in colon cancer cells. casticin 0-8 TNF receptor superfamily member 10b Homo sapiens 121-137 23135489-3 2013 Casticin downregulated cell survival proteins including Bcl-xL, Bcl-2, survivin, XIAP and cFLIP, and induced death receptor 5 (DR5), but had no effect on DR4 and decoy receptors (DcR1 or DcR2). casticin 0-8 TNF receptor superfamily member 10b Homo sapiens 133-149 23135489-3 2013 Casticin downregulated cell survival proteins including Bcl-xL, Bcl-2, survivin, XIAP and cFLIP, and induced death receptor 5 (DR5), but had no effect on DR4 and decoy receptors (DcR1 or DcR2). casticin 0-8 TNF receptor superfamily member 10b Homo sapiens 151-154 23135489-4 2013 Deletion of DR5 by siRNA significantly reduced the apoptosis induced by TRAIL and casticin. casticin 94-102 TNF receptor superfamily member 10b Homo sapiens 12-15 23135489-6 2013 Collectively, the present study showed that casticin potentiates TRAIL-induced apoptosis through downregulation of cell survival proteins and induction of DR5 mediated by ROS. casticin 56-64 TNF receptor superfamily member 10b Homo sapiens 179-182 23135489-6 2013 Collectively, the present study showed that casticin potentiates TRAIL-induced apoptosis through downregulation of cell survival proteins and induction of DR5 mediated by ROS. Reactive Oxygen Species 195-198 TNF receptor superfamily member 10b Homo sapiens 179-182 23335397-0 2013 Oligomycin A enhances apoptotic effect of TRAIL through CHOP-mediated death receptor 5 expression. oligomycin A 0-12 TNF receptor superfamily member 10b Homo sapiens 70-86 23335397-6 2013 Collectively, our data suggest that OMA enhances apoptotic death of cervical cancer cells to TRAIL through upregulation of CHOP-mediated DR5 expression following ER-stress. oligomycin A 36-39 TNF receptor superfamily member 10b Homo sapiens 137-140 22982206-0 2013 Verrucarin A sensitizes TRAIL-induced apoptosis via the upregulation of DR5 in an eIF2alpha/CHOP-dependent manner. muconomycin A 0-12 TNF receptor superfamily member 10b Homo sapiens 72-75 22982206-5 2013 We first found that VA induces a major molecule of ER stress, CCAAT/enhancer binding protein homologous protein (CHOP)-dependent DR5 induction and subsequently increases TRAIL-induced cleavage of caspases and PARP in TRAIL-resistant Hep3B cells. muconomycin A 20-22 TNF receptor superfamily member 10b Homo sapiens 129-132 22982206-10 2013 Therefore, VA-induced eIF2alpha phosphorylation seemed to be important for CHOP and DR5 upregulation and TRAIL-induced apoptosis. muconomycin A 11-13 TNF receptor superfamily member 10b Homo sapiens 84-87 22982206-12 2013 We concluded that VA triggers TRAIL-induced apoptosis by eIF2alpha/CHOP-dependent DR5 induction via ROS generation. Reactive Oxygen Species 100-103 TNF receptor superfamily member 10b Homo sapiens 82-85 23348585-9 2013 Furthermore, TRAIL-induced apoptosis in HCC cells pretreated with salirasib was dependent on activation of death receptor (DR) 5. farnesylthiosalicylic acid 66-75 TNF receptor superfamily member 10b Homo sapiens 107-128 23348585-10 2013 In conclusion, salirasib sensitizes hepatocarcinoma cells to TRAIL-induced apoptosis by a mechanism involving the DR5 receptor and survivin inhibition. farnesylthiosalicylic acid 15-24 TNF receptor superfamily member 10b Homo sapiens 114-117 24289635-7 2013 The general caspase inhibitor Z-VAD- FMK could inhibit the embelin-enhanced sensitivity of A549 cells to TRAILR2 mAb (75.97+-3.17%)(P<0.05). benzyloxycarbonylvalyl-alanyl-aspartyl fluoromethyl ketone 30-40 TNF receptor superfamily member 10b Homo sapiens 105-112 24324958-6 2013 Our results showed that there were significant synergistic effects of low dose 5-Fu and TRAIL on TRAIL-resistant AGS cells, and this effect was supposed to be mediated by decreasing DcR2 expression and increasing DR5 expression. Fluorouracil 79-83 TNF receptor superfamily member 10b Homo sapiens 213-216 23065795-3 2012 siRNA knockdown of JNK, CHOP, or DR5 shows that gammaT-induced apoptosis is JNK/CHOP/DR5 signaling dependent, which is similar to gammaT3-mediated apoptotic signaling. gamma-Tocopherol 48-54 TNF receptor superfamily member 10b Homo sapiens 33-36 24377552-7 2013 Expression of DR4 and DR5 on the cell surfaces, and the apoptosis rate were increased after treatment by ATO alone or combined with bortezomib. Arsenic Trioxide 105-108 TNF receptor superfamily member 10b Homo sapiens 22-25 24377552-7 2013 Expression of DR4 and DR5 on the cell surfaces, and the apoptosis rate were increased after treatment by ATO alone or combined with bortezomib. Bortezomib 132-142 TNF receptor superfamily member 10b Homo sapiens 22-25 23255955-9 2013 In addition, PA increased the expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis signaling-related death receptor 5 (DR5), mediating caspase-8-involved extrinsic pathway. pomolic acid 13-15 TNF receptor superfamily member 10b Homo sapiens 145-161 23255955-9 2013 In addition, PA increased the expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis signaling-related death receptor 5 (DR5), mediating caspase-8-involved extrinsic pathway. pomolic acid 13-15 TNF receptor superfamily member 10b Homo sapiens 163-166 23431365-3 2013 We and others have shown that several anticancer agents and flavonoids overcome resistance to Apo2L/TRAIL by upregulating death receptor 5 (DR5) in malignant tumor cells. Flavonoids 60-70 TNF receptor superfamily member 10b Homo sapiens 122-138 23431365-3 2013 We and others have shown that several anticancer agents and flavonoids overcome resistance to Apo2L/TRAIL by upregulating death receptor 5 (DR5) in malignant tumor cells. Flavonoids 60-70 TNF receptor superfamily member 10b Homo sapiens 140-143 23431365-5 2013 Here we show that the dietary flavonoid apigenin binds and inhibits adenine nucleotide translocase-2 (ANT2), resulting in enhancement of Apo2L/TRAIL-induced apoptosis by upregulation of DR5. Flavonoids 30-39 TNF receptor superfamily member 10b Homo sapiens 186-189 22923501-8 2012 The free radical scavenger Mn(III)tetrakis(4-benzoic acid)porphyrin chloride blocked ROS generation, DR5 up-regulation, caspase-8 activation, DNA damage, and apoptosis, which indicates a primary role for oxidative injury in lethality. mn(iii)tetrakis(4-benzoic acid)porphyrin chloride 27-76 TNF receptor superfamily member 10b Homo sapiens 101-104 23224239-9 2013 Inhibition of ERK1/2 by U0126 significantly decreased the apigenin/TRAIL-induced DR5 expression and apoptosis. U 0126 24-29 TNF receptor superfamily member 10b Homo sapiens 81-84 23536831-7 2013 Casticin dramatically upregulated DR5 receptor expression but had no effects on DR4 or decoy receptors. casticin 0-8 TNF receptor superfamily member 10b Homo sapiens 34-37 23536831-8 2013 Deletion of DR5 by siRNA significantly reduced the apoptosis induced by the co-application of TRAIL and casticin. casticin 104-112 TNF receptor superfamily member 10b Homo sapiens 12-15 23536831-9 2013 Gene silencing of the CCAAT/enhancer binding protein homologous protein (CHOP) and pretreatment with salubrinal, an endoplasmic reticulum (ER) stress inhibitor, attenuated casticin-induced DR5 receptor expression, and apoptosis and ROS production. casticin 172-180 TNF receptor superfamily member 10b Homo sapiens 189-192 23536831-9 2013 Gene silencing of the CCAAT/enhancer binding protein homologous protein (CHOP) and pretreatment with salubrinal, an endoplasmic reticulum (ER) stress inhibitor, attenuated casticin-induced DR5 receptor expression, and apoptosis and ROS production. Reactive Oxygen Species 232-235 TNF receptor superfamily member 10b Homo sapiens 189-192 23536831-11 2013 In addition, casticin also induced the expressions of DR5 protein in other gastric cancer cells (SGC-7901 and MGC-803). casticin 13-21 TNF receptor superfamily member 10b Homo sapiens 54-57 23536831-12 2013 CONCLUSION/SIGNIFICANCE: Casticin enhances TRAIL-induced apoptosis through the downregulation of cell survival proteins and the upregulation of DR5 receptors through actions on the ROS-ER stress-CHOP pathway. casticin 25-33 TNF receptor superfamily member 10b Homo sapiens 144-147 23536831-12 2013 CONCLUSION/SIGNIFICANCE: Casticin enhances TRAIL-induced apoptosis through the downregulation of cell survival proteins and the upregulation of DR5 receptors through actions on the ROS-ER stress-CHOP pathway. Reactive Oxygen Species 181-184 TNF receptor superfamily member 10b Homo sapiens 144-147 23007278-8 2012 Pretreatment with the c-Jun N-terminal kinase (JNK) inhibitor SP600125 and the reactive oxygen species (ROS) scavenger N-acetylcysteine reduced the SVT and TRAIL-induced upregulation of DR4 and DR5 expression, expression of the apoptosis related protein such as caspase-3 and-9, as well as cell growth inhibitory effects. pyrazolanthrone 62-70 TNF receptor superfamily member 10b Homo sapiens 194-197 23007278-8 2012 Pretreatment with the c-Jun N-terminal kinase (JNK) inhibitor SP600125 and the reactive oxygen species (ROS) scavenger N-acetylcysteine reduced the SVT and TRAIL-induced upregulation of DR4 and DR5 expression, expression of the apoptosis related protein such as caspase-3 and-9, as well as cell growth inhibitory effects. Reactive Oxygen Species 79-102 TNF receptor superfamily member 10b Homo sapiens 194-197 23007278-8 2012 Pretreatment with the c-Jun N-terminal kinase (JNK) inhibitor SP600125 and the reactive oxygen species (ROS) scavenger N-acetylcysteine reduced the SVT and TRAIL-induced upregulation of DR4 and DR5 expression, expression of the apoptosis related protein such as caspase-3 and-9, as well as cell growth inhibitory effects. Reactive Oxygen Species 104-107 TNF receptor superfamily member 10b Homo sapiens 194-197 23007278-8 2012 Pretreatment with the c-Jun N-terminal kinase (JNK) inhibitor SP600125 and the reactive oxygen species (ROS) scavenger N-acetylcysteine reduced the SVT and TRAIL-induced upregulation of DR4 and DR5 expression, expression of the apoptosis related protein such as caspase-3 and-9, as well as cell growth inhibitory effects. Acetylcysteine 119-135 TNF receptor superfamily member 10b Homo sapiens 194-197 23065795-3 2012 siRNA knockdown of JNK, CHOP, or DR5 shows that gammaT-induced apoptosis is JNK/CHOP/DR5 signaling dependent, which is similar to gammaT3-mediated apoptotic signaling. gamma-Tocopherol 48-54 TNF receptor superfamily member 10b Homo sapiens 85-88 23065795-5 2012 Inhibition of de novo ceramide synthesis using chemical inhibitors blocked the ability of gammaT and gammaT3 to induce apoptosis as detected by Annexin V-FITC/PI assay and to activate JNK/CHOP/DR5 pro-apoptotic signaling thereby demonstrating the involvement of de novo ceramide synthesis in gammaT- and gammaT3-induced apoptosis. Ceramides 22-30 TNF receptor superfamily member 10b Homo sapiens 193-196 23065795-6 2012 CONCLUSION: Taken together, data show that both gammaT and gammaT3 induce apoptosis via de novo ceramide synthesis dependent activation of JNK/CHOP/DR5 pro-apoptotic signaling. Ceramides 96-104 TNF receptor superfamily member 10b Homo sapiens 148-151 23203129-11 2012 Chalcones enhance TRAIL-induced apoptosis in HeLa cells through increased expression of TRAIL-R2. Chalcones 0-9 TNF receptor superfamily member 10b Homo sapiens 89-97 23203129-0 2012 Targeting death receptor TRAIL-R2 by chalcones for TRAIL-induced apoptosis in cancer cells. Chalcones 37-46 TNF receptor superfamily member 10b Homo sapiens 25-33 22922338-5 2012 Capsazepine induced death receptors (DRs) DR5 and DR4, but not decoy receptors, at the transcriptional level and in a non-cell-type-specific manner. capsazepine 0-11 TNF receptor superfamily member 10b Homo sapiens 42-45 22922338-7 2012 CHOP induction was also reactive oxygen species (ROS)-dependent, as shown by capsazepine"s ability to induce ROS and by the quenching of ROS by N-acetylcysteine or glutathione, which prevented induction of CHOP and DR5 and consequent sensitization to TRAIL. Reactive Oxygen Species 24-47 TNF receptor superfamily member 10b Homo sapiens 215-218 22922338-8 2012 Capsazepine"s effects appeared to be mediated via JNK, as shown by capsazepine"s ability to induce JNK and by the suppression of both CHOP and DR5 activation by inhibition of JNK. capsazepine 0-11 TNF receptor superfamily member 10b Homo sapiens 143-146 22922338-7 2012 CHOP induction was also reactive oxygen species (ROS)-dependent, as shown by capsazepine"s ability to induce ROS and by the quenching of ROS by N-acetylcysteine or glutathione, which prevented induction of CHOP and DR5 and consequent sensitization to TRAIL. Reactive Oxygen Species 49-52 TNF receptor superfamily member 10b Homo sapiens 215-218 22922338-7 2012 CHOP induction was also reactive oxygen species (ROS)-dependent, as shown by capsazepine"s ability to induce ROS and by the quenching of ROS by N-acetylcysteine or glutathione, which prevented induction of CHOP and DR5 and consequent sensitization to TRAIL. capsazepine 77-88 TNF receptor superfamily member 10b Homo sapiens 215-218 22922338-7 2012 CHOP induction was also reactive oxygen species (ROS)-dependent, as shown by capsazepine"s ability to induce ROS and by the quenching of ROS by N-acetylcysteine or glutathione, which prevented induction of CHOP and DR5 and consequent sensitization to TRAIL. Acetylcysteine 144-160 TNF receptor superfamily member 10b Homo sapiens 215-218 22922338-7 2012 CHOP induction was also reactive oxygen species (ROS)-dependent, as shown by capsazepine"s ability to induce ROS and by the quenching of ROS by N-acetylcysteine or glutathione, which prevented induction of CHOP and DR5 and consequent sensitization to TRAIL. Glutathione 164-175 TNF receptor superfamily member 10b Homo sapiens 215-218 22354145-0 2012 AW00179 potentiates TRAIL-mediated death of human lung cancer H1299 cells through ROS-JNK-c-Jun-mediated up-regulation of DR5 and down-regulation of anti-apoptotic molecules. Reactive Oxygen Species 82-85 TNF receptor superfamily member 10b Homo sapiens 122-125 22922573-9 2012 The synergistic apoptotic effect of CPT-11 and TRAIL was proposed to be mediated by upregulating DR5 mRNA expression and increasing expression of Bax and caspase-9 proteins. Irinotecan 36-42 TNF receptor superfamily member 10b Homo sapiens 97-100 23063980-3 2012 Ayanin diacetate-induced cell death was found to be associated with: (i) loss of inner mitochondrial membrane potential, (ii) the release of cytochrome c, (iii) the activation of multiple caspases, (iv) cleavage of poly(ADP-ribose) polymerase and (v) the up-regulation of death receptors for TRAIL, DR4 and DR5. ayanin diacetate 0-16 TNF receptor superfamily member 10b Homo sapiens 307-310 22354145-7 2012 In conclusion, AW00179 has the potential to sensitize H1299 cells to TRAIL-mediated apoptosis through two distinct mechanisms: ROS-JNK-c-Jun-mediated up-regulation of DR5, and down-regulation of anti-apoptotic molecules. Reactive Oxygen Species 127-130 TNF receptor superfamily member 10b Homo sapiens 167-170 22784708-5 2012 Methylglyoxal suppressed the expression of antiapoptotic factors, X-linked inhibitor of apoptosis protein (XIAP), survivin, cIAP1, Bcl-2, and Bcl-xL, without affecting TRAIL receptors, DR4 and DR5. Pyruvaldehyde 0-13 TNF receptor superfamily member 10b Homo sapiens 193-196 22842544-0 2012 Antimycin A sensitizes cells to TRAIL-induced apoptosis through upregulation of DR5 and downregulation of c-FLIP and Bcl-2. Antimycin A 0-11 TNF receptor superfamily member 10b Homo sapiens 80-83 22735465-14 2012 Artepillin C increased the expression of TRAIL-R2 and decreased the activity of NF-kappaB. artepillin C 0-12 TNF receptor superfamily member 10b Homo sapiens 41-49 22848091-2 2012 Our previous work has shown that PS-341 induces death receptor 5 (DR5)-dependent apoptosis and enhances the TNF-related apoptosis-inducing ligand-induced apoptosis in human non-small cell lung cancer cells. Bortezomib 33-39 TNF receptor superfamily member 10b Homo sapiens 48-64 22848091-2 2012 Our previous work has shown that PS-341 induces death receptor 5 (DR5)-dependent apoptosis and enhances the TNF-related apoptosis-inducing ligand-induced apoptosis in human non-small cell lung cancer cells. Bortezomib 33-39 TNF receptor superfamily member 10b Homo sapiens 66-69 22848091-4 2012 In the present study, we reveal that PKCdelta and RSK2 mediate PS-341-induced DR5 upregulation, involving coactivation of endoplasmic reticulum (ER) stress. Bortezomib 63-69 TNF receptor superfamily member 10b Homo sapiens 78-81 22848091-10 2012 In addition, we discovered that the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 and RSK2 were elevated after PS-341 treatment and inhibition of their phosphorylation using MAP-ERK kinase 1/2 inhibitor decreased the DR5 level, indicating that ERK/RSK2 signaling is involved in DR5 upregulation. Bortezomib 132-138 TNF receptor superfamily member 10b Homo sapiens 238-241 22848091-10 2012 In addition, we discovered that the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 and RSK2 were elevated after PS-341 treatment and inhibition of their phosphorylation using MAP-ERK kinase 1/2 inhibitor decreased the DR5 level, indicating that ERK/RSK2 signaling is involved in DR5 upregulation. Bortezomib 132-138 TNF receptor superfamily member 10b Homo sapiens 299-302 22848091-13 2012 Collectively, we show that PS-341 induces PKCdelta-dependent DR5 expression through activation of ERK/RSK2 and ER stress signaling pathway. Bortezomib 27-33 TNF receptor superfamily member 10b Homo sapiens 61-64 22359244-6 2012 Expression of TRAIL-mediated apoptosis signaling-related death receptor DR5 was increased in 3-O-acetyloleanolic acid-treated HCT-116 cells. oleanolic acid 3-acetate 93-117 TNF receptor superfamily member 10b Homo sapiens 72-75 22359244-8 2012 The results indicate that 3-O-acetyloleanolic acid induces apoptosis in HCT-116 cells mediated by an extrinsic apoptosis signaling cascade via up-regulation of DR5. oleanolic acid 3-acetate 26-50 TNF receptor superfamily member 10b Homo sapiens 160-163 22962275-4 2012 MK886 effectively increased the sensitivity to TRAIL-induced apoptosis via upregulation of the death receptor 5 and downregulation of the antiapoptotic protein survivin in human glioma cell lines and in primary glioma cells. MK-886 0-5 TNF receptor superfamily member 10b Homo sapiens 95-111 22824956-7 2012 In addition, U0126 reduced UDCA-triggered TNF-related apoptosis-inducing ligand receptor 2 (TRAIL-R2/DR5) expression. U 0126 13-18 TNF receptor superfamily member 10b Homo sapiens 54-102 22824956-7 2012 In addition, U0126 reduced UDCA-triggered TNF-related apoptosis-inducing ligand receptor 2 (TRAIL-R2/DR5) expression. U 0126 13-18 TNF receptor superfamily member 10b Homo sapiens 104-112 22824956-7 2012 In addition, U0126 reduced UDCA-triggered TNF-related apoptosis-inducing ligand receptor 2 (TRAIL-R2/DR5) expression. U 0126 13-18 TNF receptor superfamily member 10b Homo sapiens 113-116 22936321-0 2012 Induction of apoptosis in human Hep3B hepatoma cells by norcantharidin through a p53 independent pathway via TRAIL/DR5 signal transduction. norcantharidin 56-70 TNF receptor superfamily member 10b Homo sapiens 115-118 22815059-5 2012 The anthracycline anticancer drug epirubicin (EPB) synergizes the cytotoxicity of HMCAZ5 in cancer cells by upregulating DR5 expression on the cell surfaces, enhancing p53 expression, Bid cleavage, and JNK phosphorylation and downregulating c-FLIP expression and Akt phosphorylation. Anthracyclines 4-17 TNF receptor superfamily member 10b Homo sapiens 121-124 22555452-6 2012 Up-regulation of death receptor 4 (DR4) and DR5 by silibinin was shown by RT-PCR and by flow cytometry. Silybin 51-60 TNF receptor superfamily member 10b Homo sapiens 44-47 22815059-5 2012 The anthracycline anticancer drug epirubicin (EPB) synergizes the cytotoxicity of HMCAZ5 in cancer cells by upregulating DR5 expression on the cell surfaces, enhancing p53 expression, Bid cleavage, and JNK phosphorylation and downregulating c-FLIP expression and Akt phosphorylation. Epirubicin 34-44 TNF receptor superfamily member 10b Homo sapiens 121-124 22707197-0 2012 Triptolide sensitizes TRAIL-induced apoptosis in prostate cancer cells via p53-mediated DR5 up-regulation. triptolide 0-10 TNF receptor superfamily member 10b Homo sapiens 88-91 22707197-3 2012 In this study, we found that Triptolide, a Chinese medicine, significantly sensitizes prostate cancer cells to TRAIL-mediated cellular apoptosis by up-regulating DR5 expression. triptolide 29-39 TNF receptor superfamily member 10b Homo sapiens 162-165 22707197-4 2012 Triptolide treatment can suppress Akt/Hdm2 signaling pathway, and lead to p53 accumulation, thereby up-regulating DR5 expression. triptolide 0-10 TNF receptor superfamily member 10b Homo sapiens 114-117 22483777-5 2012 Knockdown of SIRT1 or treatment with amurensin G, a potent new SIRT1 inhibitor, up-regulated the levels of DR5 and c-Myc and down-regulated the level of c-FLIP(L/S). amurensin G 37-48 TNF receptor superfamily member 10b Homo sapiens 107-110 22555452-7 2012 Human recombinant DR5/Fc chimera protein that has a dominant-negative effect by competing with the endogenous receptors abrogated cell death induced by silibinin and TRAIL, demonstrating the activation of the death receptor pathway. Silybin 152-161 TNF receptor superfamily member 10b Homo sapiens 18-21 22562097-8 2012 Our results showed that DR5 monoclonal antibody (D-6) was able to induce the apoptosis and increase the cell growth inhibition rates of ovarian cancer cells in a dose-dependent manner, and the effect was enhanced by cisplatin. Cisplatin 216-225 TNF receptor superfamily member 10b Homo sapiens 24-27 22562097-10 2012 The combination treatment of the DR5 monoclonal antibody (D-6) with cisplatin may be a promising treatment for ovarian cancer. Cisplatin 68-77 TNF receptor superfamily member 10b Homo sapiens 33-36 21801305-0 2012 Salermide up-regulates death receptor 5 expression through the ATF4-ATF3-CHOP axis and leads to apoptosis in human cancer cells. N-(3-((2-hydroxynaphthalen-1-ylmethylene)amino)phenyl)-2-phenylpropionamide 0-9 TNF receptor superfamily member 10b Homo sapiens 23-39 22447040-7 2012 Transfection studies were used to determine the involvement of DR4, DR5 and Cbl-b in the synergistic effect of bufalin and TRAIL. bufalin 111-118 TNF receptor superfamily member 10b Homo sapiens 68-71 22447040-11 2012 Blocking the up-regulation of DR4 and DR5 by siRNA rendered cells less sensitive to apoptosis induced by the combination of bufalin and TRAIL. bufalin 124-131 TNF receptor superfamily member 10b Homo sapiens 38-41 22447040-15 2012 Bufalin-induced down-regulation of Cbl-b contributed to the up-regulation of DR4 and DR5, which might be partially mediated by the activation of ERK, JNK and p38 MAPK. bufalin 0-7 TNF receptor superfamily member 10b Homo sapiens 85-88 22788777-6 2012 Based on distance geometry method and intermolecular hydrogen bonding analysis, the key functional domain in DR5 was predicted and the DR5 mutants were designed. Hydrogen 53-61 TNF receptor superfamily member 10b Homo sapiens 109-112 22788777-6 2012 Based on distance geometry method and intermolecular hydrogen bonding analysis, the key functional domain in DR5 was predicted and the DR5 mutants were designed. Hydrogen 53-61 TNF receptor superfamily member 10b Homo sapiens 135-138 21801305-11 2012 This suggests that DR5 is modulated by the ATF4-ATF3-CHOP axis in NSCLC after Sirt1/2 inhibition or salermide treatment. N-(3-((2-hydroxynaphthalen-1-ylmethylene)amino)phenyl)-2-phenylpropionamide 100-109 TNF receptor superfamily member 10b Homo sapiens 19-22 21801305-4 2012 In this study, we show that salermide up-regulates the expression of death receptor 5 (DR5) in human non-small cell lung cancer (NSCLC) cells. N-(3-((2-hydroxynaphthalen-1-ylmethylene)amino)phenyl)-2-phenylpropionamide 28-37 TNF receptor superfamily member 10b Homo sapiens 69-85 21801305-4 2012 In this study, we show that salermide up-regulates the expression of death receptor 5 (DR5) in human non-small cell lung cancer (NSCLC) cells. N-(3-((2-hydroxynaphthalen-1-ylmethylene)amino)phenyl)-2-phenylpropionamide 28-37 TNF receptor superfamily member 10b Homo sapiens 87-90 21801305-6 2012 Increasing DR5 protein expression correlates with salermide-induced apoptosis in human NSCLC cells. N-(3-((2-hydroxynaphthalen-1-ylmethylene)amino)phenyl)-2-phenylpropionamide 50-59 TNF receptor superfamily member 10b Homo sapiens 11-14 21801305-7 2012 We discovered that IRE-1alpha, Bip, activating transcription factor 3 (ATF4), activating transcription factor 3 (ATF3) and C/EBP homologous protein (CHOP) are induced by salermide, which suggests that DR5-dependent apoptosis is induced by endoplasmic reticulum stress. N-(3-((2-hydroxynaphthalen-1-ylmethylene)amino)phenyl)-2-phenylpropionamide 170-179 TNF receptor superfamily member 10b Homo sapiens 201-204 23124518-2 2012 We provide evidence that pretreatment with etoposide significantly enhanced TRAIL-mediated apoptosis via up-regulation of DR5 (death receptor 5 or TRAIL-R2) expression in the caspase 8 expressing neuroblastoma cell line, SK-N-MC. Etoposide 43-52 TNF receptor superfamily member 10b Homo sapiens 122-125 22309674-0 2012 beta-Phenethyl isothiocyanate induces death receptor 5 to induce apoptosis in human oral cancer cells via p38. phenethyl isothiocyanate 0-29 TNF receptor superfamily member 10b Homo sapiens 38-54 22309674-8 2012 This result was confirmed by blockage of PEITC-induced cleavages of Poly(ADP-ribose) Polymerase, caspase-3, caspase-8, and DR5 by p38 MAPK inhibitor, SB203580. phenethyl isothiocyanate 41-46 TNF receptor superfamily member 10b Homo sapiens 123-126 22309674-8 2012 This result was confirmed by blockage of PEITC-induced cleavages of Poly(ADP-ribose) Polymerase, caspase-3, caspase-8, and DR5 by p38 MAPK inhibitor, SB203580. SB 203580 150-158 TNF receptor superfamily member 10b Homo sapiens 123-126 23124518-4 2012 Although TRAIL-R2 expression increased in IMR-32 cells in response to etoposide treatment, cell death was not increased by concurrent treatment with TRAIL compared with etoposide alone, because the cells lacked caspase 8 expression. Etoposide 70-79 TNF receptor superfamily member 10b Homo sapiens 9-17 23124518-2 2012 We provide evidence that pretreatment with etoposide significantly enhanced TRAIL-mediated apoptosis via up-regulation of DR5 (death receptor 5 or TRAIL-R2) expression in the caspase 8 expressing neuroblastoma cell line, SK-N-MC. Etoposide 43-52 TNF receptor superfamily member 10b Homo sapiens 127-143 23124518-7 2012 These results indicate that the etoposide-mediated sensitization of neuroblastoma cells to TRAIL is associated with an increase in TRAIL-R2 expression and requires caspase 8 expression. Etoposide 32-41 TNF receptor superfamily member 10b Homo sapiens 131-139 23124518-2 2012 We provide evidence that pretreatment with etoposide significantly enhanced TRAIL-mediated apoptosis via up-regulation of DR5 (death receptor 5 or TRAIL-R2) expression in the caspase 8 expressing neuroblastoma cell line, SK-N-MC. Etoposide 43-52 TNF receptor superfamily member 10b Homo sapiens 147-155 22367066-3 2012 Exposure to MEMC was found to result in growth inhibition by the induction of caspase-dependent apoptosis in NCI-H460 cells, which correlated with upregulated expression of death receptor (DR)4, DR5 and FasL, downregulation of anti-apoptotic Bcl-2 and Bcl-xL expression, cleavage of Bid, and loss of mitochondrial membrane potential. Mercury, chloro(2-methoxyethyl)- 12-16 TNF receptor superfamily member 10b Homo sapiens 219-222 22681760-9 2012 Snake venom toxin also induced JNK phosphorylation and ROS generation, however, pretreatment of JNK inhibitor and ROS scavenger reversed the inhibitory effect of snake venom toxin on cancer cell proliferation, and reduced the snake venom toxin-induced upregulation of DR4 and DR5 expression. Reactive Oxygen Species 114-117 TNF receptor superfamily member 10b Homo sapiens 276-279 22681760-10 2012 CONCLUSIONS: Our results indicated that snake venom toxin could inhibit human colon cancer cell growth, and these effects may be related to ROS and JNK mediated activation of death receptor (DR4 and DR5) signals. Reactive Oxygen Species 140-143 TNF receptor superfamily member 10b Homo sapiens 199-202 22522053-6 2012 Curcumin induced upregulation of Fas, FasL, and DR5 expression in chondrosarcoma cells. Curcumin 0-8 TNF receptor superfamily member 10b Homo sapiens 48-51 22522053-7 2012 Transfection of cells with Fas, FasL, or DR5 siRNA reduced curcumin-induced cell death. Curcumin 59-67 TNF receptor superfamily member 10b Homo sapiens 41-44 22522053-8 2012 In addition, p53 involved in curcumin-mediated Fas, FasL, and DR5 expression and cell apoptosis in chondrosarcoma cells. Curcumin 29-37 TNF receptor superfamily member 10b Homo sapiens 62-65 22395735-8 2012 In view of the finding that DR5 silencing abrogated the combination-stimulated apoptosis, we propose that apoptotic synergy induced by sulindac sulfide plus DHA is mediated via DR5. Docosahexaenoic Acids 181-184 TNF receptor superfamily member 10b Homo sapiens 201-204 22472572-0 2012 Inostamycin enhanced TRAIL-induced apoptosis through DR5 upregulation on the cell surface. inostamycin 0-11 TNF receptor superfamily member 10b Homo sapiens 53-56 22472572-6 2012 Inostamycin upregulated DR5, and a knockdown of DR5 suppressed the apoptosis that was synergistically induced by co-treatment with inostamycin and TRAIL. inostamycin 0-11 TNF receptor superfamily member 10b Homo sapiens 24-27 22472572-6 2012 Inostamycin upregulated DR5, and a knockdown of DR5 suppressed the apoptosis that was synergistically induced by co-treatment with inostamycin and TRAIL. inostamycin 131-142 TNF receptor superfamily member 10b Homo sapiens 48-51 22472572-7 2012 Moreover, inostamycin increased the expression of DR5 on the cell surface. inostamycin 10-21 TNF receptor superfamily member 10b Homo sapiens 50-53 22472572-8 2012 Therefore, inostamycin-increased cell surface expression of DR5 may have contributed to the enhancement of TRAIL-induced apoptosis. inostamycin 11-22 TNF receptor superfamily member 10b Homo sapiens 60-63 22395735-8 2012 In view of the finding that DR5 silencing abrogated the combination-stimulated apoptosis, we propose that apoptotic synergy induced by sulindac sulfide plus DHA is mediated via DR5. sulindac sulfide 159-175 TNF receptor superfamily member 10b Homo sapiens 40-43 22131069-8 2012 Epirubicin treatment up-regulated death receptor 4 (DR-4) and DR-5 expression and down-regulated FLIP expression, thereby enhancing the activation of procaspase 3, procaspase 8, and procaspase 9. Epirubicin 0-10 TNF receptor superfamily member 10b Homo sapiens 62-66 22395735-8 2012 In view of the finding that DR5 silencing abrogated the combination-stimulated apoptosis, we propose that apoptotic synergy induced by sulindac sulfide plus DHA is mediated via DR5. sulindac sulfide 159-175 TNF receptor superfamily member 10b Homo sapiens 201-204 22395735-8 2012 In view of the finding that DR5 silencing abrogated the combination-stimulated apoptosis, we propose that apoptotic synergy induced by sulindac sulfide plus DHA is mediated via DR5. Docosahexaenoic Acids 181-184 TNF receptor superfamily member 10b Homo sapiens 40-43 22707267-3 2012 METHODS AND RESULTS: DHA induces apoptosis as detected by Annexin V-FITC/PI assay as well as induces cleavage of caspase-8 and -9, endoplasmic reticulum stress (ERS), and elevated levels of death receptor-5 (DR5) protein expression as detected by western blot assays. Docosahexaenoic Acids 21-24 TNF receptor superfamily member 10b Homo sapiens 190-206 22707267-3 2012 METHODS AND RESULTS: DHA induces apoptosis as detected by Annexin V-FITC/PI assay as well as induces cleavage of caspase-8 and -9, endoplasmic reticulum stress (ERS), and elevated levels of death receptor-5 (DR5) protein expression as detected by western blot assays. Docosahexaenoic Acids 21-24 TNF receptor superfamily member 10b Homo sapiens 208-211 22707267-4 2012 Chemical inhibitors of caspase-8 and -9 and small interfering RNAs (siRNAs) show DHA to induce ERS/CHOP/DR5-mediated caspase-8 and -9 dependent apoptosis. Docosahexaenoic Acids 81-84 TNF receptor superfamily member 10b Homo sapiens 104-107 22707267-7 2012 CONCLUSION: Data, for the first time, demonstrate that DHA induces apoptosis in TNBC cells via activation of ERS/CHOP/DR5-mediated caspase-8 and -9 dependent pro-apoptotic events, and that different forms of vitamin E exhibit distinct effects on DHA-induced apoptosis; namely, inhibition by alphaT and enhancement by gammaT3. Docosahexaenoic Acids 55-58 TNF receptor superfamily member 10b Homo sapiens 118-121 22400995-0 2012 Isoobtusilactone A sensitizes human hepatoma Hep G2 cells to TRAIL-induced apoptosis via ROS and CHOP-mediated up-regulation of DR5. isoobtusilactone A 0-18 TNF receptor superfamily member 10b Homo sapiens 128-131 21927023-4 2012 In this report, we demonstrate that ATF3 is required for efficient DR5 induction upon DNA damage by camptothecin (CPT) in colorectal cancer cells. Camptothecin 100-112 TNF receptor superfamily member 10b Homo sapiens 67-70 21927023-4 2012 In this report, we demonstrate that ATF3 is required for efficient DR5 induction upon DNA damage by camptothecin (CPT) in colorectal cancer cells. Camptothecin 114-117 TNF receptor superfamily member 10b Homo sapiens 67-70 22328338-8 2012 Moreover, orlistat induced reactive oxygen species (ROS), and a ROS scavenger diminished the sensitivity to TRAIL through the suppression of CHOP and DR5 expression in both cell lines. Reactive Oxygen Species 76-79 TNF receptor superfamily member 10b Homo sapiens 174-177 22328338-9 2012 These results suggest that there are two pathways of upregulation of DR5 by orlistat, which are the ROS-CHOP pathway and the ROS-direct pathway. Reactive Oxygen Species 112-115 TNF receptor superfamily member 10b Homo sapiens 81-84 22328338-9 2012 These results suggest that there are two pathways of upregulation of DR5 by orlistat, which are the ROS-CHOP pathway and the ROS-direct pathway. Reactive Oxygen Species 137-140 TNF receptor superfamily member 10b Homo sapiens 81-84 22400995-8 2012 DR5 expression after IOA treatment was accompanied by provoking intracellular reactive oxygen species (ROS) generation. Reactive Oxygen Species 78-101 TNF receptor superfamily member 10b Homo sapiens 0-3 22400995-8 2012 DR5 expression after IOA treatment was accompanied by provoking intracellular reactive oxygen species (ROS) generation. Reactive Oxygen Species 103-106 TNF receptor superfamily member 10b Homo sapiens 0-3 22200406-0 2012 Capsaicin sensitizes TRAIL-induced apoptosis through Sp1-mediated DR5 up-regulation: involvement of Ca(2+) influx. Capsaicin 0-9 TNF receptor superfamily member 10b Homo sapiens 66-69 22244504-6 2012 Furthermore, epirubicin therapy significantly improved recurrence-free rate for the patients with DR4-high (P = .006) or DR5-high (P = .042) tumor. Epirubicin 13-23 TNF receptor superfamily member 10b Homo sapiens 121-124 22244504-8 2012 In addition, patients with high expression of both DR4 and DR5 might benefit from epirubicin therapy. Epirubicin 82-92 TNF receptor superfamily member 10b Homo sapiens 59-62 22008998-11 2012 Taken together, RU486 enhances TRAIL-mediated apoptosis through down-regulation of Bcl-2 and c-FLIP(L) as well as CHOP-mediated DR5 up-regulation. Mifepristone 16-21 TNF receptor superfamily member 10b Homo sapiens 128-131 22205156-6 2012 Epirubicin and pirarubicin significantly increased the TRAIL-R2 expression at both the mRNA and the protein levels. Epirubicin 0-10 TNF receptor superfamily member 10b Homo sapiens 55-63 22205156-6 2012 Epirubicin and pirarubicin significantly increased the TRAIL-R2 expression at both the mRNA and the protein levels. pirarubicin 15-26 TNF receptor superfamily member 10b Homo sapiens 55-63 22205156-11 2012 The present study demonstrates that anthracylines sensitize RCC cells to lexatumumab-mediated apoptosis by inducing TRAIL-R2 expression, and the utility of PCR array to elucidate the mechanism of synergistic apoptosis. anthracylines 36-49 TNF receptor superfamily member 10b Homo sapiens 116-124 22230479-0 2012 Dovitinib sensitizes hepatocellular carcinoma cells to TRAIL and tigatuzumab, a novel anti-DR5 antibody, through SHP-1-dependent inhibition of STAT3. 4-amino-5-fluoro-3-(5-(4-methylpiperazin-1-yl)-1H-benzimidazol-2-yl)quinolin-2(1H)-one 0-9 TNF receptor superfamily member 10b Homo sapiens 91-94 22008998-5 2012 We found that up-regulation of death receptor 5 (DR5; receptor for TRAIL ligand), and down-regulation of Bcl-2 and c-FLIP (caspase regulator) contributes to RU-486 induced TRAIL sensitization. Mifepristone 157-163 TNF receptor superfamily member 10b Homo sapiens 31-47 22008998-5 2012 We found that up-regulation of death receptor 5 (DR5; receptor for TRAIL ligand), and down-regulation of Bcl-2 and c-FLIP (caspase regulator) contributes to RU-486 induced TRAIL sensitization. Mifepristone 157-163 TNF receptor superfamily member 10b Homo sapiens 49-52 22008998-6 2012 Down-regulation of DR5 by siRNA also blocked RU486 induced TRAIL sensitization. Mifepristone 45-50 TNF receptor superfamily member 10b Homo sapiens 19-22 22200406-4 2012 In addition, we found that capsaicin induces the cell surface expression of TRAIL receptor DR5, but not DR4 through the activation Sp1 on its promoter region. Capsaicin 27-36 TNF receptor superfamily member 10b Homo sapiens 91-94 22200406-5 2012 Furthermore, we investigated that capsaicin-induced DR5 expression and apoptosis are inhibited by calcium chelator or inhibitors for calmodulin-dependent protein kinase. Capsaicin 34-43 TNF receptor superfamily member 10b Homo sapiens 52-55 22200406-6 2012 Taken together, our data suggest that capsaicin sensitizes TRAIL-mediated HCC cell apoptosis by DR5 up-regulation via calcium influx-dependent Sp1 activation. Capsaicin 38-47 TNF receptor superfamily member 10b Homo sapiens 96-99 22077238-0 2012 Aclarubicin enhances tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis through death receptor 5 upregulation. Aclarubicin 0-11 TNF receptor superfamily member 10b Homo sapiens 103-119 22077238-6 2012 In contrast, another anthracycline, doxorubicin (DOX), only slightly sensitized Jurkat cells and A549 cells to TRAIL-induced apoptosis, with weaker enhancement of death receptor 5 (DR5) expression than ACR. Doxorubicin 36-47 TNF receptor superfamily member 10b Homo sapiens 163-179 22077238-6 2012 In contrast, another anthracycline, doxorubicin (DOX), only slightly sensitized Jurkat cells and A549 cells to TRAIL-induced apoptosis, with weaker enhancement of death receptor 5 (DR5) expression than ACR. Doxorubicin 36-47 TNF receptor superfamily member 10b Homo sapiens 181-184 22077238-6 2012 In contrast, another anthracycline, doxorubicin (DOX), only slightly sensitized Jurkat cells and A549 cells to TRAIL-induced apoptosis, with weaker enhancement of death receptor 5 (DR5) expression than ACR. Doxorubicin 49-52 TNF receptor superfamily member 10b Homo sapiens 163-179 21660448-0 2012 5, 7-Dimethoxyflavone sensitizes TRAIL-induced apoptosis through DR5 upregulation in hepatocellular carcinoma cells. 5,7-dimethoxyflavone 0-21 TNF receptor superfamily member 10b Homo sapiens 65-68 22158615-2 2012 Here we report that stimulation of DR4 and/or DR5 by the agonistic protein KD548-Fc, an Fc-fused DR4/DR5 dual-specific Kringle domain variant, activates plasma membrane-associated Nox1 NADPH oxidase to generate superoxide anion and subsequently accumulates intracellular reactive oxygen species (ROS), leading to sustained c-Jun N-terminal kinase activation and eventual apoptotic cell death in human HeLa and Jurkat tumor cells. Superoxides 211-227 TNF receptor superfamily member 10b Homo sapiens 46-49 22158615-2 2012 Here we report that stimulation of DR4 and/or DR5 by the agonistic protein KD548-Fc, an Fc-fused DR4/DR5 dual-specific Kringle domain variant, activates plasma membrane-associated Nox1 NADPH oxidase to generate superoxide anion and subsequently accumulates intracellular reactive oxygen species (ROS), leading to sustained c-Jun N-terminal kinase activation and eventual apoptotic cell death in human HeLa and Jurkat tumor cells. Superoxides 211-227 TNF receptor superfamily member 10b Homo sapiens 101-104 22158615-2 2012 Here we report that stimulation of DR4 and/or DR5 by the agonistic protein KD548-Fc, an Fc-fused DR4/DR5 dual-specific Kringle domain variant, activates plasma membrane-associated Nox1 NADPH oxidase to generate superoxide anion and subsequently accumulates intracellular reactive oxygen species (ROS), leading to sustained c-Jun N-terminal kinase activation and eventual apoptotic cell death in human HeLa and Jurkat tumor cells. Reactive Oxygen Species 271-294 TNF receptor superfamily member 10b Homo sapiens 46-49 22158615-2 2012 Here we report that stimulation of DR4 and/or DR5 by the agonistic protein KD548-Fc, an Fc-fused DR4/DR5 dual-specific Kringle domain variant, activates plasma membrane-associated Nox1 NADPH oxidase to generate superoxide anion and subsequently accumulates intracellular reactive oxygen species (ROS), leading to sustained c-Jun N-terminal kinase activation and eventual apoptotic cell death in human HeLa and Jurkat tumor cells. Reactive Oxygen Species 271-294 TNF receptor superfamily member 10b Homo sapiens 101-104 22158615-2 2012 Here we report that stimulation of DR4 and/or DR5 by the agonistic protein KD548-Fc, an Fc-fused DR4/DR5 dual-specific Kringle domain variant, activates plasma membrane-associated Nox1 NADPH oxidase to generate superoxide anion and subsequently accumulates intracellular reactive oxygen species (ROS), leading to sustained c-Jun N-terminal kinase activation and eventual apoptotic cell death in human HeLa and Jurkat tumor cells. Reactive Oxygen Species 296-299 TNF receptor superfamily member 10b Homo sapiens 46-49 22158615-2 2012 Here we report that stimulation of DR4 and/or DR5 by the agonistic protein KD548-Fc, an Fc-fused DR4/DR5 dual-specific Kringle domain variant, activates plasma membrane-associated Nox1 NADPH oxidase to generate superoxide anion and subsequently accumulates intracellular reactive oxygen species (ROS), leading to sustained c-Jun N-terminal kinase activation and eventual apoptotic cell death in human HeLa and Jurkat tumor cells. Reactive Oxygen Species 296-299 TNF receptor superfamily member 10b Homo sapiens 101-104 22158615-7 2012 Our results suggest that DR4 and DR5 have a capability to activate Nox1 by recruiting RFK, resulting in ROS-mediated apoptotic cell death in tumor cells. Reactive Oxygen Species 104-107 TNF receptor superfamily member 10b Homo sapiens 33-36 21660448-9 2012 Pretreatment with N-acetylcysteine (NAC) inhibited DMF-induced upregulation of DR5, CHOP, GPR78, and ATF4 protein expression and blocked the cotreatment-induced apoptosis. Acetylcysteine 18-34 TNF receptor superfamily member 10b Homo sapiens 79-82 21660448-9 2012 Pretreatment with N-acetylcysteine (NAC) inhibited DMF-induced upregulation of DR5, CHOP, GPR78, and ATF4 protein expression and blocked the cotreatment-induced apoptosis. Acetylcysteine 36-39 TNF receptor superfamily member 10b Homo sapiens 79-82 21660448-9 2012 Pretreatment with N-acetylcysteine (NAC) inhibited DMF-induced upregulation of DR5, CHOP, GPR78, and ATF4 protein expression and blocked the cotreatment-induced apoptosis. Dimethylformamide 51-54 TNF receptor superfamily member 10b Homo sapiens 79-82 21660448-8 2012 RESULTS: Our results demonstrated subtoxic concentrations of DMF sensitize HCC cells to TRAIL-induced apoptosis and induce the death receptor 5 (DR5) expression level, accompanying the generation of reactive oxygen species (ROS) and the upregulation of CHOP, GPR78, and ATF4 protein expression. Dimethylformamide 61-64 TNF receptor superfamily member 10b Homo sapiens 127-143 21660448-10 2012 Furthermore, DMF-mediated sensitization of HCC cells to TRAIL was reduced by administration of a blocking antibody or small interfering RNAs for DR5, salubrinal, an inhibitor of ER stress, and the small interfering RNAs for CHOP. Dimethylformamide 13-16 TNF receptor superfamily member 10b Homo sapiens 145-148 21660448-12 2012 CONCLUSION: The present study demonstrates that DMF selectively enhances TRAIL-induced apoptosis by ROS-stimulated ER-stress triggering CHOP-mediated DR5 upregulation in HCC. Dimethylformamide 48-51 TNF receptor superfamily member 10b Homo sapiens 150-153 21660448-8 2012 RESULTS: Our results demonstrated subtoxic concentrations of DMF sensitize HCC cells to TRAIL-induced apoptosis and induce the death receptor 5 (DR5) expression level, accompanying the generation of reactive oxygen species (ROS) and the upregulation of CHOP, GPR78, and ATF4 protein expression. Dimethylformamide 61-64 TNF receptor superfamily member 10b Homo sapiens 145-148 21660448-12 2012 CONCLUSION: The present study demonstrates that DMF selectively enhances TRAIL-induced apoptosis by ROS-stimulated ER-stress triggering CHOP-mediated DR5 upregulation in HCC. Reactive Oxygen Species 100-103 TNF receptor superfamily member 10b Homo sapiens 150-153 21887462-5 2012 Cotreatment of bufotalin with TRAIL resulted in the downregulation of anti-apoptotic proteins, including Bcl-XL, Mcl-1, survivin and XIAP, and the up-regulation of MAPKs and TRAIL receptor DR5. bufotalin 15-24 TNF receptor superfamily member 10b Homo sapiens 213-216 22666346-9 2012 CONCLUSIONS/SIGNIFICANCE: These results suggest that DHA enhances Apo2L/TRAIL-mediated apoptosis in human pancreatic cancer cells through reactive oxygen species-mediated up-regulation of DR5. Reactive Oxygen Species 138-161 TNF receptor superfamily member 10b Homo sapiens 188-191 21978492-6 2011 PHA665752 treatment also caused increased expression of DR5 via generation of reactive oxygen species, and combination treatment with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and PHA665752 induced significant apoptosis. Reactive Oxygen Species 78-101 TNF receptor superfamily member 10b Homo sapiens 56-59 22666346-0 2012 Dihydroartemisinin enhances Apo2L/TRAIL-mediated apoptosis in pancreatic cancer cells via ROS-mediated up-regulation of death receptor 5. artenimol 0-18 TNF receptor superfamily member 10b Homo sapiens 120-136 22666346-0 2012 Dihydroartemisinin enhances Apo2L/TRAIL-mediated apoptosis in pancreatic cancer cells via ROS-mediated up-regulation of death receptor 5. Reactive Oxygen Species 90-93 TNF receptor superfamily member 10b Homo sapiens 120-136 22666346-6 2012 The effect of DHA was mediated through the generation of reactive oxygen species, the induction of death receptor 5 (DR5) and the modulation of apoptosis-related proteins. artenimol 14-17 TNF receptor superfamily member 10b Homo sapiens 99-115 22666346-6 2012 The effect of DHA was mediated through the generation of reactive oxygen species, the induction of death receptor 5 (DR5) and the modulation of apoptosis-related proteins. artenimol 14-17 TNF receptor superfamily member 10b Homo sapiens 117-120 22666346-8 2012 In addition, knockdown of DR5 by small interfering RNA also significantly reduced the amount of apoptosis induced by DHA and Apo2L/TRAIL. artenimol 117-120 TNF receptor superfamily member 10b Homo sapiens 26-29 22666346-9 2012 CONCLUSIONS/SIGNIFICANCE: These results suggest that DHA enhances Apo2L/TRAIL-mediated apoptosis in human pancreatic cancer cells through reactive oxygen species-mediated up-regulation of DR5. artenimol 53-56 TNF receptor superfamily member 10b Homo sapiens 188-191 22120628-10 2011 Both Velcade and MG132 increased the protein levels of DR5, a TRAIL receptor known to be up-regulated by p53, in U373MG cells where p53 is mutated. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 17-22 TNF receptor superfamily member 10b Homo sapiens 55-58 21903093-0 2011 Guggulsterone sensitizes hepatoma cells to TRAIL-induced apoptosis through the induction of CHOP-dependent DR5: involvement of ROS-dependent ER-stress. pregna-4,17-diene-3,16-dione 0-13 TNF receptor superfamily member 10b Homo sapiens 107-110 21903093-11 2011 Pretreatment with N-acetyl-L-cysteine and glutathione inhibited GGS-induced ER-stress, and CHOP and DR5 upregulation and almost completely blocked GGS/TRAIL-induced apoptosis. Acetylcysteine 18-37 TNF receptor superfamily member 10b Homo sapiens 100-103 21903093-11 2011 Pretreatment with N-acetyl-L-cysteine and glutathione inhibited GGS-induced ER-stress, and CHOP and DR5 upregulation and almost completely blocked GGS/TRAIL-induced apoptosis. Glutathione 42-53 TNF receptor superfamily member 10b Homo sapiens 100-103 22019170-12 2011 CONCLUSIONS: Data suggest that paclitaxel induces nuclear translocation and activation of PKC-delta, which in turn causes Golgi-Cdk1 activation, leading to Golgi associated DR5 up-regulation, and caspase-8 and 3 activation. Paclitaxel 31-41 TNF receptor superfamily member 10b Homo sapiens 173-176 21769428-7 2011 It suggested that the TRAIL-induced redistribution of DR4 and DR5 in lipid rafts contributed to the sensitivity to TRAIL in TRAIL-sensitive NSCLC H460 cell line, which was also confirmed by intervention tests of the cholesterol-sequestering agent nystatin. Cholesterol 216-227 TNF receptor superfamily member 10b Homo sapiens 62-65 21769428-7 2011 It suggested that the TRAIL-induced redistribution of DR4 and DR5 in lipid rafts contributed to the sensitivity to TRAIL in TRAIL-sensitive NSCLC H460 cell line, which was also confirmed by intervention tests of the cholesterol-sequestering agent nystatin. Nystatin 247-255 TNF receptor superfamily member 10b Homo sapiens 62-65 22019170-7 2011 Data revealed that paclitaxel induced an increase of Cdk1 activity and DR5 expression on the Golgi complex that was associated with increased cleavage of caspase-8, a DR5 downstream factor, and caspase-3 into catalytically active fragments. Paclitaxel 19-29 TNF receptor superfamily member 10b Homo sapiens 71-74 22019170-7 2011 Data revealed that paclitaxel induced an increase of Cdk1 activity and DR5 expression on the Golgi complex that was associated with increased cleavage of caspase-8, a DR5 downstream factor, and caspase-3 into catalytically active fragments. Paclitaxel 19-29 TNF receptor superfamily member 10b Homo sapiens 167-170 21941003-5 2011 The toxic saturated free fatty acid palmitate induces an increase in DR5 mRNA and protein expression in Huh-7 human hepatoma cells leading to DR5 localization into lipid rafts, cell surface receptor clustering with subsequent recruitment of the initiator caspase-8, and ultimately cellular demise. saturated 10-19 TNF receptor superfamily member 10b Homo sapiens 69-72 21941003-5 2011 The toxic saturated free fatty acid palmitate induces an increase in DR5 mRNA and protein expression in Huh-7 human hepatoma cells leading to DR5 localization into lipid rafts, cell surface receptor clustering with subsequent recruitment of the initiator caspase-8, and ultimately cellular demise. saturated 10-19 TNF receptor superfamily member 10b Homo sapiens 142-145 21941003-5 2011 The toxic saturated free fatty acid palmitate induces an increase in DR5 mRNA and protein expression in Huh-7 human hepatoma cells leading to DR5 localization into lipid rafts, cell surface receptor clustering with subsequent recruitment of the initiator caspase-8, and ultimately cellular demise. free fatty acid palmitate 20-45 TNF receptor superfamily member 10b Homo sapiens 69-72 21875750-6 2011 We also demonstrate that DR5-targeted nanoparticles encapsulating the cytotoxic drug camptothecin are effectively targeted to the tumour cells, thereby producing enhanced cytotoxic effects through simultaneous drug delivery and apoptosis induction. Camptothecin 85-97 TNF receptor superfamily member 10b Homo sapiens 25-28 21941003-5 2011 The toxic saturated free fatty acid palmitate induces an increase in DR5 mRNA and protein expression in Huh-7 human hepatoma cells leading to DR5 localization into lipid rafts, cell surface receptor clustering with subsequent recruitment of the initiator caspase-8, and ultimately cellular demise. free fatty acid palmitate 20-45 TNF receptor superfamily member 10b Homo sapiens 142-145 21941003-8 2011 Likewise, knockdown of DR5 or caspase-8 expression by shRNA technology attenuated palmitate-induced Bax activation and apoptosis in Huh-7 cells, without altering induction of ER stress markers. Palmitates 82-91 TNF receptor superfamily member 10b Homo sapiens 23-26 21726997-0 2011 Death receptor 5 and cellular FLICE-inhibitory protein regulate pemetrexed-induced apoptosis in human lung cancer cells. Pemetrexed 64-74 TNF receptor superfamily member 10b Homo sapiens 0-16 21726997-5 2011 We showed that pemetrexed induced apoptosis via up-regulation of Death Receptor 5 (DR5), an important death receptor for tumour necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL). Pemetrexed 15-25 TNF receptor superfamily member 10b Homo sapiens 65-81 21726997-5 2011 We showed that pemetrexed induced apoptosis via up-regulation of Death Receptor 5 (DR5), an important death receptor for tumour necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL). Pemetrexed 15-25 TNF receptor superfamily member 10b Homo sapiens 83-86 21726997-7 2011 Modulating DR5 induction by small interfering RNA abrogated the ability of pemetrexed to induce apoptosis. Pemetrexed 75-85 TNF receptor superfamily member 10b Homo sapiens 11-14 21726997-8 2011 In addition, silencing of C/EBP homologous protein (CHOP) expression reduced DR5 expression, demonstrating that the transcriptional factor CHOP has a pivotal role on DR5 up-regulation following pemetrexed treatment. Pemetrexed 194-204 TNF receptor superfamily member 10b Homo sapiens 77-80 21726997-8 2011 In addition, silencing of C/EBP homologous protein (CHOP) expression reduced DR5 expression, demonstrating that the transcriptional factor CHOP has a pivotal role on DR5 up-regulation following pemetrexed treatment. Pemetrexed 194-204 TNF receptor superfamily member 10b Homo sapiens 166-169 21757445-2 2011 We have identified lanatoside C as a sensitizer of GBM cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced cell death partly by upregulation of the death receptor 5. lanatoside C 19-31 TNF receptor superfamily member 10b Homo sapiens 177-193 21854768-0 2011 Acrolein sensitizes human renal cancer Caki cells to TRAIL-induced apoptosis via ROS-mediated up-regulation of death receptor-5 (DR5) and down-regulation of Bcl-2. Acrolein 0-8 TNF receptor superfamily member 10b Homo sapiens 111-127 21854768-0 2011 Acrolein sensitizes human renal cancer Caki cells to TRAIL-induced apoptosis via ROS-mediated up-regulation of death receptor-5 (DR5) and down-regulation of Bcl-2. Acrolein 0-8 TNF receptor superfamily member 10b Homo sapiens 129-132 21854768-0 2011 Acrolein sensitizes human renal cancer Caki cells to TRAIL-induced apoptosis via ROS-mediated up-regulation of death receptor-5 (DR5) and down-regulation of Bcl-2. ros 81-84 TNF receptor superfamily member 10b Homo sapiens 111-127 21854768-0 2011 Acrolein sensitizes human renal cancer Caki cells to TRAIL-induced apoptosis via ROS-mediated up-regulation of death receptor-5 (DR5) and down-regulation of Bcl-2. ros 81-84 TNF receptor superfamily member 10b Homo sapiens 129-132 21854768-8 2011 Interestingly, pretreatment with an antioxidant, N-acetylcysteine (NAC), inhibited not only CHOP and DR5 up-regulation but also the cell death induced by acrolein plus TRAIL. Acetylcysteine 49-65 TNF receptor superfamily member 10b Homo sapiens 101-104 21854768-8 2011 Interestingly, pretreatment with an antioxidant, N-acetylcysteine (NAC), inhibited not only CHOP and DR5 up-regulation but also the cell death induced by acrolein plus TRAIL. Acetylcysteine 67-70 TNF receptor superfamily member 10b Homo sapiens 101-104 21854768-9 2011 Taken together, our results demonstrated that acrolein enhances TRAIL-induced apoptosis in Caki cells through down-regulation of Bcl-2 and ROS dependent up-regulation of DR5. Acrolein 46-54 TNF receptor superfamily member 10b Homo sapiens 170-173 21965726-7 2011 However, PA also increases surface expression of death receptors 4 and 5 (DR4 and DR5, respectively). Paclitaxel 9-11 TNF receptor superfamily member 10b Homo sapiens 82-85 21779837-5 2011 Silibinin enhanced the expression (protein and mRNA) of TNF-related apoptosis-inducing ligand (TRAIL) death receptors (DR4/DR5) at the cell surface in SW480 cells, and induced their expression in TRAIL-resistant SW620 cells normally not expressing DR4/DR5. Silybin 0-9 TNF receptor superfamily member 10b Homo sapiens 123-126 22090786-0 2011 Casticin-induced apoptosis involves death receptor 5 upregulation in hepatocellular carcinoma cells. casticin 0-8 TNF receptor superfamily member 10b Homo sapiens 36-52 22090786-15 2011 In the HCC cells treated with casticin for 24 h, DR5 protein level was increased. casticin 30-38 TNF receptor superfamily member 10b Homo sapiens 49-52 22090786-16 2011 The expression of DR5 protein induced by casticin was inhibited by NAC. casticin 41-49 TNF receptor superfamily member 10b Homo sapiens 18-21 22090786-17 2011 Pretreatment with DR5/Fc chimera protein, a blocking antibody, effectively attenuated the induction of apoptosis by casticin. casticin 116-124 TNF receptor superfamily member 10b Homo sapiens 18-21 22090786-18 2011 CONCLUSION: Casticin-induced apoptosis of HCC cells is involved in GSH depletion and DR5 upregulation. casticin 12-20 TNF receptor superfamily member 10b Homo sapiens 85-88 21751122-8 2011 Etoposide and doxorubicin induced DR5 but not DR4 in NB cell lines. Etoposide 0-9 TNF receptor superfamily member 10b Homo sapiens 34-37 21751122-8 2011 Etoposide and doxorubicin induced DR5 but not DR4 in NB cell lines. Doxorubicin 14-25 TNF receptor superfamily member 10b Homo sapiens 34-37 21722984-3 2011 This study focused on the interaction of cisplatin/radiation with the anti-human DR5 monoclonal antibody TRA-8 in SCLC cells. Cisplatin 41-50 TNF receptor superfamily member 10b Homo sapiens 81-84 21877938-0 2011 Cisplatin restores TRAIL apoptotic pathway in glioblastoma-derived stem cells through up-regulation of DR5 and down-regulation of c-FLIP. Cisplatin 0-9 TNF receptor superfamily member 10b Homo sapiens 103-106 21877938-3 2011 However, treatment with cisplatin leads to the upregulation of DR5 and downregulation of c-FLIP and restores TRAIL apoptotic pathway in the neurospheres. Cisplatin 24-33 TNF receptor superfamily member 10b Homo sapiens 63-66 21325634-9 2011 Fourth, curcumin upregulated death receptors, DR4 and DR5. Curcumin 8-16 TNF receptor superfamily member 10b Homo sapiens 54-57 21597379-7 2011 In addition, C-DIMs inhibited cell proliferation and induced PARP cleavage through death receptor 5 and CHOP in HEp-2 and HN22 cells. c-dims 13-19 TNF receptor superfamily member 10b Homo sapiens 83-99 21697087-0 2011 Modulation of CCAAT/enhancer binding protein homologous protein (CHOP)-dependent DR5 expression by nelfinavir sensitizes glioblastoma multiforme cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Nelfinavir 99-109 TNF receptor superfamily member 10b Homo sapiens 81-84 21697087-5 2011 We found that Nelfinavir treatment led to ER stress-induced up-regulation of the DR5 receptor. Nelfinavir 14-24 TNF receptor superfamily member 10b Homo sapiens 81-84 21697087-10 2011 We provide evidence that Nelfinavir-induced ER stress modulates DR5 expression in human glioblastoma multiforme cells and can enhance TRAIL efficacy. Nelfinavir 25-35 TNF receptor superfamily member 10b Homo sapiens 64-67 21697087-9 2011 Combining Nelfinavir with TRAIL led to a significantly enhanced level of apoptosis that was abrogated by siRNA silencing of DR5. Nelfinavir 10-20 TNF receptor superfamily member 10b Homo sapiens 124-127 21354251-9 2011 Bupropion also induced the mRNA expression of the death receptors DR4 (TRAILR1) and DR5 (TRAILR2). Bupropion 0-9 TNF receptor superfamily member 10b Homo sapiens 84-87 21702500-9 2011 PD98059 increased the expression of DR4 and DR5, activated caspase-3, and cleaved PARP. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 0-7 TNF receptor superfamily member 10b Homo sapiens 44-47 21601561-7 2011 We further found that Redd1-induced mTOR down-regulation and ATF4/CHOP-induced expression of the TRAIL receptor DR5 associated with sorafenib treatment helped sensitize cells to TRAIL-induced apoptosis. Sorafenib 132-141 TNF receptor superfamily member 10b Homo sapiens 112-115 21822052-0 2011 Mcl-1 and YY1 inhibition and induction of DR5 by the BH3-mimetic Obatoclax (GX15-070) contribute in the sensitization of B-NHL cells to TRAIL apoptosis. BH 3 53-56 TNF receptor superfamily member 10b Homo sapiens 42-45 21725212-5 2011 Co-treatment of these cells with quinacrine and TRAIL induces overwhelming cell death within 3-4 h. Levels of DR5, a pro-apoptotic death receptor of TRAIL, are increased upon treatment with quinacrine, while levels of Mcl-1, an anti-apoptotic member of the Bcl-2 family, are decreased. Quinacrine 33-43 TNF receptor superfamily member 10b Homo sapiens 110-113 21725212-5 2011 Co-treatment of these cells with quinacrine and TRAIL induces overwhelming cell death within 3-4 h. Levels of DR5, a pro-apoptotic death receptor of TRAIL, are increased upon treatment with quinacrine, while levels of Mcl-1, an anti-apoptotic member of the Bcl-2 family, are decreased. Quinacrine 190-200 TNF receptor superfamily member 10b Homo sapiens 110-113 21565489-0 2011 Seleno-cyclodextrin sensitises human breast cancer cells to TRAIL-induced apoptosis through DR5 induction and NF-kappaB suppression. seleno-cyclodextrin 0-19 TNF receptor superfamily member 10b Homo sapiens 92-95 21801359-9 2011 In addition, we found that treatment of A549 cells with AFMC significantly induced the expression of death receptor 5 (DR5). afmc 56-60 TNF receptor superfamily member 10b Homo sapiens 101-117 21801359-9 2011 In addition, we found that treatment of A549 cells with AFMC significantly induced the expression of death receptor 5 (DR5). afmc 56-60 TNF receptor superfamily member 10b Homo sapiens 119-122 21801359-10 2011 AFMC-mediated sensitization of A549 cells to TRAIL was efficiently reduced by administration of a blocking antibody or small interfering RNAs against DR5. afmc 0-4 TNF receptor superfamily member 10b Homo sapiens 150-153 21801359-11 2011 AFMC also caused increase of the Sub-G1 cells by TRAIL treatment and increased the expression levels of DR5 in other NSCLC H460 and H157 cell lines. afmc 0-4 TNF receptor superfamily member 10b Homo sapiens 104-107 21801359-12 2011 In contrast, AFMC-mediated induction of DR5 expression was not observed in human embryo lung WI-38 cells, and AFMC did not sensitize WI-38 cells to TRAIL-induced apoptosis. afmc 13-17 TNF receptor superfamily member 10b Homo sapiens 40-43 21801359-13 2011 CONCLUSIONS: AFMC synergistically enhances TRAIL-mediated apoptosis in NSCLC cells through up-regulating DR5 expression. afmc 13-17 TNF receptor superfamily member 10b Homo sapiens 105-108 21354251-9 2011 Bupropion also induced the mRNA expression of the death receptors DR4 (TRAILR1) and DR5 (TRAILR2). Bupropion 0-9 TNF receptor superfamily member 10b Homo sapiens 89-96 21653830-6 2011 Lapatinib up-regulated the proapoptotic TRAIL death receptors DR4 and DR5, leading to more efficient induction of apoptosis in the presence of TRAIL receptor agonists. Lapatinib 0-9 TNF receptor superfamily member 10b Homo sapiens 70-73 21519792-1 2011 We previously reported that the chemopreventive agent lupulone induces apoptosis through activation of the extrinsic pathway via TRAIL DR4/DR5 death receptors overcoming SW620 cell resistance to TRAIL. lupulon 54-62 TNF receptor superfamily member 10b Homo sapiens 139-142 21519792-6 2011 In contrast to JNK and ERK inhibition, the specific inactivation of p38 inhibited the lupulone-triggered up-regulation of p53 and TRAIL-death receptor DR4/DR5 expression, and prevented DNA fragmentation. lupulon 86-94 TNF receptor superfamily member 10b Homo sapiens 155-158 21653830-8 2011 The off-target induction of DR5 by lapatinib resulted from activation of the c-Jun amino-terminal kinase (JNK)/c-Jun signaling axis. Lapatinib 35-44 TNF receptor superfamily member 10b Homo sapiens 28-31 21811429-0 2011 Up-regulation of the DR5 expression by proteasome inhibitor MG132 augments TRAIL-induced apoptosis in soft tissue sarcoma cell lines. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 60-65 TNF receptor superfamily member 10b Homo sapiens 21-24 21362627-0 2011 Lipid raft-dependent death receptor 5 (DR5) expression and activation are critical for ursodeoxycholic acid-induced apoptosis in gastric cancer cells. Ursodeoxycholic Acid 87-107 TNF receptor superfamily member 10b Homo sapiens 21-37 21571804-2 2011 The purpose of this study was to characterize (64)Cu-labeled conatumumab as a PET tracer for imaging DR5 in tumors. Copper 50-52 TNF receptor superfamily member 10b Homo sapiens 101-104 21362627-0 2011 Lipid raft-dependent death receptor 5 (DR5) expression and activation are critical for ursodeoxycholic acid-induced apoptosis in gastric cancer cells. Ursodeoxycholic Acid 87-107 TNF receptor superfamily member 10b Homo sapiens 39-42 21362627-7 2011 In addition, reactive oxygen species (ROS) and protein kinase C (PKC) delta appeared to be implicated in UDCA-induced raft-dependent DR5 expression. Reactive Oxygen Species 13-36 TNF receptor superfamily member 10b Homo sapiens 133-136 21362627-7 2011 In addition, reactive oxygen species (ROS) and protein kinase C (PKC) delta appeared to be implicated in UDCA-induced raft-dependent DR5 expression. Reactive Oxygen Species 38-41 TNF receptor superfamily member 10b Homo sapiens 133-136 21362627-8 2011 Our results indicate that UDCA-induced apoptosis is mediated by DR5 expression, which is regulated by the raft formation/ROS production/PKCdelta activation pathway and DR5 localization into lipid rafts in gastric cancer cells. Reactive Oxygen Species 121-124 TNF receptor superfamily member 10b Homo sapiens 64-67 21393428-2 2011 Here, we report that bortezomib sensitizes these TRAIL-resistant cells, including Huh-7, Hep3B, and Sk-Hep1, to CS-1008, a humanized agonistic antihuman death receptor 5 antibody. Bortezomib 21-31 TNF receptor superfamily member 10b Homo sapiens 153-169 21539448-4 2011 The apoptotic effects of mDRA-6 on Jurkat cells, which express DR5 on the cell surface, were detected by flow cytometry and western blot after exposure to different doses of mDRA-6 and at fixed doses of mDRA-6 at different times. mdra-6 25-31 TNF receptor superfamily member 10b Homo sapiens 63-66 21468584-6 2011 Epirubicin significantly promoted lipid raft DR4 and DR5 aggregation, as well as the localization of DR4 and DR5 in the lipid rafts. Epirubicin 0-10 TNF receptor superfamily member 10b Homo sapiens 53-56 21468584-6 2011 Epirubicin significantly promoted lipid raft DR4 and DR5 aggregation, as well as the localization of DR4 and DR5 in the lipid rafts. Epirubicin 0-10 TNF receptor superfamily member 10b Homo sapiens 109-112 21468584-8 2011 Pretreatment with 50 microg/ml nystatin, a cholesterol-sequestering agent, partially prevented epirubicin-induced lipid raft aggregation and DR4 and DR5 clustering. Nystatin 31-39 TNF receptor superfamily member 10b Homo sapiens 149-152 21468584-8 2011 Pretreatment with 50 microg/ml nystatin, a cholesterol-sequestering agent, partially prevented epirubicin-induced lipid raft aggregation and DR4 and DR5 clustering. Epirubicin 95-105 TNF receptor superfamily member 10b Homo sapiens 149-152 21483669-3 2011 Furthermore, recombinant soluble TRAIL and an agonistic anti-DR5 antibody, AD5-10, treatment stimulates reactive oxygen species (ROS) generation and JNK phosphorylation. Reactive Oxygen Species 104-127 TNF receptor superfamily member 10b Homo sapiens 61-64 21483669-3 2011 Furthermore, recombinant soluble TRAIL and an agonistic anti-DR5 antibody, AD5-10, treatment stimulates reactive oxygen species (ROS) generation and JNK phosphorylation. Reactive Oxygen Species 129-132 TNF receptor superfamily member 10b Homo sapiens 61-64 21168266-6 2011 Celastrol"s ability to modulate the expression of pro-inflammatory cytokines, MHC II, HO-1, iNOS, NF-kappaB, Notch-1, AKT/mTOR, CXCR4, TRAIL receptors DR4 and DR5, CHOP, JNK, VEGF, adhesion molecules, proteasome activity, topoisomerase II, potassium channels, and heat shock response has been reported. celastrol 0-9 TNF receptor superfamily member 10b Homo sapiens 159-162 21393428-0 2011 Bortezomib sensitizes HCC cells to CS-1008, an antihuman death receptor 5 antibody, through the inhibition of CIP2A. Bortezomib 0-10 TNF receptor superfamily member 10b Homo sapiens 57-73 20619923-6 2011 Bortezomib treatment activated endoplasmic reticulum (ER) stress-mediated apoptosis, as demonstrated by the induction of GADD153, an ER stress-inducible transcription factor, and of the death receptor DR5, in EGFR inhibitor-resistant cells, but not in parental cells. Bortezomib 0-10 TNF receptor superfamily member 10b Homo sapiens 201-204 21167276-0 2011 Piceatannol enhances TRAIL-induced apoptosis in human leukemia THP-1 cells through Sp1- and ERK-dependent DR5 up-regulation. 3,3',4,5'-tetrahydroxystilbene 0-11 TNF receptor superfamily member 10b Homo sapiens 106-109 21167276-8 2011 In conclusion, our results suggest that PIC sensitizes TRAIL-induced-apoptosis via Sp1- and ERK-dependent DR5 up-regulation. 3,3',4,5'-tetrahydroxystilbene 40-43 TNF receptor superfamily member 10b Homo sapiens 106-109 21297379-0 2011 Bisphosphonate enhances TRAIL sensitivity to human osteosarcoma cells via death receptor 5 upregulation. Diphosphonates 0-14 TNF receptor superfamily member 10b Homo sapiens 74-90 21297379-6 2011 Bisphosphonates significantly induced mRNA and protein expression of the TRAIL receptor, DR5. Diphosphonates 0-15 TNF receptor superfamily member 10b Homo sapiens 89-92 21206980-7 2011 MG132 enhanced the expression of the TRAIL receptors DR4 and DR5, and neutralization of DR5 receptors showed a marked reduction of TRAIL-mediated apoptosis, whereas that of DR4 was a partial reduction. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-5 TNF receptor superfamily member 10b Homo sapiens 61-64 21292685-0 2011 Amiodarone sensitizes human glioma cells but not astrocytes to TRAIL-induced apoptosis via CHOP-mediated DR5 upregulation. Amiodarone 0-10 TNF receptor superfamily member 10b Homo sapiens 105-108 21292685-5 2011 Suppression of DR5 expression by small interfering (si) RNAs almost completely blocked amiodarone/TRAIL-induced apoptosis in U251MG glioma cells, demonstrating that DR5 is critical to this cell death. Amiodarone 87-97 TNF receptor superfamily member 10b Homo sapiens 15-18 21292685-5 2011 Suppression of DR5 expression by small interfering (si) RNAs almost completely blocked amiodarone/TRAIL-induced apoptosis in U251MG glioma cells, demonstrating that DR5 is critical to this cell death. Amiodarone 87-97 TNF receptor superfamily member 10b Homo sapiens 165-168 21292685-6 2011 siRNA-mediated CHOP suppression reduced amiodarone-induced DR5 upregulation and attenuated the cell death induced by amiodarone plus TRAIL. Amiodarone 40-50 TNF receptor superfamily member 10b Homo sapiens 59-62 21292685-7 2011 In addition, omitting Ca(2+) from the external medium using ethylene glycol tetraacetic acid markedly inhibited this cell death, reducing the protein levels of CHOP and DR5. Egtazic Acid 60-92 TNF receptor superfamily member 10b Homo sapiens 169-172 21292685-8 2011 These results suggest that amiodarone-induced influx of Ca(2+) plays an important role in sensitizing U251MG cells to TRAIL-mediated apoptosis through CHOP-mediated DR5 upregulation. Amiodarone 27-37 TNF receptor superfamily member 10b Homo sapiens 165-168 21292685-9 2011 Furthermore, subtoxic doses of bepridil and cibenzoline, two other anti-arrhythmic drugs with NCX-inhibitor activity, also sensitized glioma cells to TRAIL-mediated apoptosis, via the upregulation of both CHOP and DR5. Bepridil 31-39 TNF receptor superfamily member 10b Homo sapiens 214-217 21292685-9 2011 Furthermore, subtoxic doses of bepridil and cibenzoline, two other anti-arrhythmic drugs with NCX-inhibitor activity, also sensitized glioma cells to TRAIL-mediated apoptosis, via the upregulation of both CHOP and DR5. cifenline 44-55 TNF receptor superfamily member 10b Homo sapiens 214-217 21282356-5 2011 Curcumin-mediated ER stress via inhibiting the activity of SERCA2 caused increasing expressions of CHOP and its transcription target death receptor 5 (TRAIL-R2), leading to a caspase-3 and caspase-8 cascade-dependent apoptosis in SW872 cells in vitro and in vivo. Curcumin 0-8 TNF receptor superfamily member 10b Homo sapiens 133-149 21282356-5 2011 Curcumin-mediated ER stress via inhibiting the activity of SERCA2 caused increasing expressions of CHOP and its transcription target death receptor 5 (TRAIL-R2), leading to a caspase-3 and caspase-8 cascade-dependent apoptosis in SW872 cells in vitro and in vivo. Curcumin 0-8 TNF receptor superfamily member 10b Homo sapiens 151-159 21206980-9 2011 Therefore, MG132 provides partial regulation of TRAIL-mediated apoptosis in OSCC cells via modulation of DR5, c-FLIP, cIAP-1, XIAP and survivin. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 11-16 TNF receptor superfamily member 10b Homo sapiens 105-108 21156789-9 2011 Induction of DRs was independent of p53 because UA induced DR4 and DR5 in HCT116 p53(-/-) cells. ursolic acid 48-50 TNF receptor superfamily member 10b Homo sapiens 67-70 21270522-6 2011 In addition, differently from saquinavir, Saq-NO restored TRAIL sensitivity that was correlated with increased expression of DR5 independent from ROS/RNS production and YY1 repression. saquinavir-NO 42-48 TNF receptor superfamily member 10b Homo sapiens 125-128 21195543-5 2011 Mechanisms underlying this sensitization mainly involved diosgenin-induced p38 MAPK pathway activation and subsequent DR5 overexpression. Diosgenin 57-66 TNF receptor superfamily member 10b Homo sapiens 118-121 21081474-0 2011 2-Tellurium-bridged beta-cyclodextrin, a thioredoxin reductase inhibitor, sensitizes human breast cancer cells to TRAIL-induced apoptosis through DR5 induction and NF-kappaB suppression. 2-tellurium 0-11 TNF receptor superfamily member 10b Homo sapiens 146-149 21081474-0 2011 2-Tellurium-bridged beta-cyclodextrin, a thioredoxin reductase inhibitor, sensitizes human breast cancer cells to TRAIL-induced apoptosis through DR5 induction and NF-kappaB suppression. betadex 20-37 TNF receptor superfamily member 10b Homo sapiens 146-149 21081474-6 2011 From a mechanistic standpoint, we showed that 2-TeCD treatment of breast cancer cells significantly upregulated the messenger RNA and protein levels of TRAIL receptor, death receptor (DR) 5, in a transcription factor Sp1-dependent manner. 2-tecd 46-52 TNF receptor superfamily member 10b Homo sapiens 168-189 21270522-7 2011 NF-kappaB activation may be responsible of the Saq-NO induced DR5 expression. saquinavir-NO 47-53 TNF receptor superfamily member 10b Homo sapiens 62-65 21152876-0 2011 Cisplatin sensitizes human hepatocellular carcinoma cells, but not hepatocytes and mesenchymal stem cells, to TRAIL within a therapeutic window partially depending on the upregulation of DR5. Cisplatin 0-9 TNF receptor superfamily member 10b Homo sapiens 187-190 20669244-0 2011 Naringenin up-regulates the expression of death receptor 5 and enhances TRAIL-induced apoptosis in human lung cancer A549 cells. naringenin 0-10 TNF receptor superfamily member 10b Homo sapiens 42-58 20669244-9 2011 We could show that following exposure to naringenin, DR5 proteins were up-regulated and knockdown of DR5 expression by siRNA attenuated naringenin plus TRAIL-induced apoptosis. naringenin 41-51 TNF receptor superfamily member 10b Homo sapiens 53-56 20669244-9 2011 We could show that following exposure to naringenin, DR5 proteins were up-regulated and knockdown of DR5 expression by siRNA attenuated naringenin plus TRAIL-induced apoptosis. naringenin 41-51 TNF receptor superfamily member 10b Homo sapiens 101-104 20669244-9 2011 We could show that following exposure to naringenin, DR5 proteins were up-regulated and knockdown of DR5 expression by siRNA attenuated naringenin plus TRAIL-induced apoptosis. naringenin 136-146 TNF receptor superfamily member 10b Homo sapiens 101-104 21152878-0 2011 Death receptor 5-mediated TNFR family signaling pathways modulate gamma-humulene-induced apoptosis in human colorectal cancer HT29 cells. gamma-Humulene 66-80 TNF receptor superfamily member 10b Homo sapiens 0-16 21152878-6 2011 gamma-Humulene stimulated the death receptor 5 (DR5), DR4, Fas-associated protein with death domain (FADD), the cleavage of caspase-8 and cleavage caspase-3, but reduced the protein levels of cellular Fas-associated death-domain-like IL-1ss-converting enzyme inhibitory protein (c-FLIP) by Western blot analysis. gamma-Humulene 0-14 TNF receptor superfamily member 10b Homo sapiens 30-46 21152878-6 2011 gamma-Humulene stimulated the death receptor 5 (DR5), DR4, Fas-associated protein with death domain (FADD), the cleavage of caspase-8 and cleavage caspase-3, but reduced the protein levels of cellular Fas-associated death-domain-like IL-1ss-converting enzyme inhibitory protein (c-FLIP) by Western blot analysis. gamma-Humulene 0-14 TNF receptor superfamily member 10b Homo sapiens 48-51 21152878-7 2011 Consequently, gamma-humulene-triggered cell death was significantly attenuated by DR5 siRNA and the caspase-8 inhibitor. gamma-Humulene 14-28 TNF receptor superfamily member 10b Homo sapiens 82-85 21152878-8 2011 Based on our results, we suggest that gamma-humulene induced apoptotic cell death in HT29 cells through a DR5-mediated caspase-8 and -3-dependent signaling pathway. gamma-Humulene 38-52 TNF receptor superfamily member 10b Homo sapiens 106-109 21935477-10 2011 Microarray analysis of islets exposed to Imatinib for 4 hours revealed increased expression of the inflammatory genes IL-4R, TCF5, DR5, I-TRAF, I-CAM, HSP27 and IL-8. Imatinib Mesylate 41-49 TNF receptor superfamily member 10b Homo sapiens 131-134 21272366-13 2011 Surprisingly, MG-132 but not IFN-gamma could also increase DR5-mediated apoptosis in SW948-TR. Magnesium 14-16 TNF receptor superfamily member 10b Homo sapiens 59-62 21078664-4 2011 This limonoid also enhanced expression of death receptors (DRs) DR5 and DR4 in cancer cells. Limonins 5-13 TNF receptor superfamily member 10b Homo sapiens 64-67 20951126-0 2011 Activation of p38 MAPK by damnacanthal mediates apoptosis in SKHep 1 cells through the DR5/TRAIL and TNFR1/TNF-alpha and p53 pathways. damnacanthal 26-38 TNF receptor superfamily member 10b Homo sapiens 87-90 20951126-4 2011 This is first demonstration that damnacanthal-mediated apoptosis involves the sustained activation of the p38 MAPK pathway, leading to the transcription of the death receptor family genes encoding DR5/TRAIL and TNF-R1/TNF-alpha genes as well as the p53-regulated Bax gene. damnacanthal 33-45 TNF receptor superfamily member 10b Homo sapiens 197-200 21214929-8 2011 RESULTS: Combination treatments of alpha-TEA plus DOXO or CDDP act cooperatively to induce apoptosis, caspase-8 and caspase-9 cleavage, p73, phospho-c-Ab1 and phospho-JNK protein expression, and increase expression of p53 downstream mediators; namely, death receptor-5, CD95/APO-1 (Fas), Bax and Noxa, as well as Yap nuclear translocation - plus reduce expression of Bcl-2. 2,5,7,8-tetramethyl-2R-(4R,8R,12-trimethyltridecyl)chroman-6-yloxy acetic acid 35-44 TNF receptor superfamily member 10b Homo sapiens 252-268 21214929-8 2011 RESULTS: Combination treatments of alpha-TEA plus DOXO or CDDP act cooperatively to induce apoptosis, caspase-8 and caspase-9 cleavage, p73, phospho-c-Ab1 and phospho-JNK protein expression, and increase expression of p53 downstream mediators; namely, death receptor-5, CD95/APO-1 (Fas), Bax and Noxa, as well as Yap nuclear translocation - plus reduce expression of Bcl-2. Doxorubicin 50-54 TNF receptor superfamily member 10b Homo sapiens 252-268 21214929-8 2011 RESULTS: Combination treatments of alpha-TEA plus DOXO or CDDP act cooperatively to induce apoptosis, caspase-8 and caspase-9 cleavage, p73, phospho-c-Ab1 and phospho-JNK protein expression, and increase expression of p53 downstream mediators; namely, death receptor-5, CD95/APO-1 (Fas), Bax and Noxa, as well as Yap nuclear translocation - plus reduce expression of Bcl-2. Cisplatin 58-62 TNF receptor superfamily member 10b Homo sapiens 252-268 21037225-5 2011 Both platinum complexes increased DR5 surface expression in colon cancer cells. Platinum 5-13 TNF receptor superfamily member 10b Homo sapiens 34-37 21037225-6 2011 Small interfering RNA-mediated DR5 silencing rescued cells from sensitizing effects of platinum drugs on TRAIL-induced caspase-8 activation and apoptosis, showing the functional importance of DR5 in the effects observed. Platinum 87-95 TNF receptor superfamily member 10b Homo sapiens 31-34 21037225-6 2011 Small interfering RNA-mediated DR5 silencing rescued cells from sensitizing effects of platinum drugs on TRAIL-induced caspase-8 activation and apoptosis, showing the functional importance of DR5 in the effects observed. Platinum 87-95 TNF receptor superfamily member 10b Homo sapiens 192-195 21037225-7 2011 In addition, both cisplatin and LA-12 triggered the relocalization of DR4 and DR5 receptors to lipid rafts and accelerated internalization of TRAIL, which may also affect TRAIL signaling. Cisplatin 18-27 TNF receptor superfamily member 10b Homo sapiens 78-81 21037225-7 2011 In addition, both cisplatin and LA-12 triggered the relocalization of DR4 and DR5 receptors to lipid rafts and accelerated internalization of TRAIL, which may also affect TRAIL signaling. UNII-37WM0V5E17 32-37 TNF receptor superfamily member 10b Homo sapiens 78-81 21037225-8 2011 Collectively, modulations of the initial steps of the extrinsic apoptotic pathway at the level of DR5 and plasma membrane are important for sensitization of colon and prostate cancer cells to TRAIL-induced apoptosis mediated by LA-12 and cisplatin. UNII-37WM0V5E17 228-233 TNF receptor superfamily member 10b Homo sapiens 98-101 21037225-8 2011 Collectively, modulations of the initial steps of the extrinsic apoptotic pathway at the level of DR5 and plasma membrane are important for sensitization of colon and prostate cancer cells to TRAIL-induced apoptosis mediated by LA-12 and cisplatin. Cisplatin 238-247 TNF receptor superfamily member 10b Homo sapiens 98-101 21976975-0 2011 Anti-DR5 monoclonal antibody-mediated DTIC-loaded nanoparticles combining chemotherapy and immunotherapy for malignant melanoma: target formulation development and in vitro anticancer activity. Dacarbazine 38-42 TNF receptor superfamily member 10b Homo sapiens 5-8 21976975-4 2011 METHODS: The chemotherapeutic drug, dacarbazine (DTIC), that induces apoptosis through the intrinsic pathway which typically responds to severe DNA damage, was used as a model drug to prepare DTIC-loaded polylactic acid (PLA) nanoparticles (DTIC-NPs), which were covalently conjugated to a highly specific targeting functional TRAIL-receptor 2 (DR5) monoclonal antibody (mAb) that can contribute directly to cancer cell apoptosis or growth inhibition through the extrinsic pathway. Dacarbazine 36-47 TNF receptor superfamily member 10b Homo sapiens 345-348 21976975-4 2011 METHODS: The chemotherapeutic drug, dacarbazine (DTIC), that induces apoptosis through the intrinsic pathway which typically responds to severe DNA damage, was used as a model drug to prepare DTIC-loaded polylactic acid (PLA) nanoparticles (DTIC-NPs), which were covalently conjugated to a highly specific targeting functional TRAIL-receptor 2 (DR5) monoclonal antibody (mAb) that can contribute directly to cancer cell apoptosis or growth inhibition through the extrinsic pathway. Dacarbazine 49-53 TNF receptor superfamily member 10b Homo sapiens 345-348 21976975-4 2011 METHODS: The chemotherapeutic drug, dacarbazine (DTIC), that induces apoptosis through the intrinsic pathway which typically responds to severe DNA damage, was used as a model drug to prepare DTIC-loaded polylactic acid (PLA) nanoparticles (DTIC-NPs), which were covalently conjugated to a highly specific targeting functional TRAIL-receptor 2 (DR5) monoclonal antibody (mAb) that can contribute directly to cancer cell apoptosis or growth inhibition through the extrinsic pathway. Dacarbazine 192-196 TNF receptor superfamily member 10b Homo sapiens 345-348 21976975-4 2011 METHODS: The chemotherapeutic drug, dacarbazine (DTIC), that induces apoptosis through the intrinsic pathway which typically responds to severe DNA damage, was used as a model drug to prepare DTIC-loaded polylactic acid (PLA) nanoparticles (DTIC-NPs), which were covalently conjugated to a highly specific targeting functional TRAIL-receptor 2 (DR5) monoclonal antibody (mAb) that can contribute directly to cancer cell apoptosis or growth inhibition through the extrinsic pathway. poly(lactide) 204-219 TNF receptor superfamily member 10b Homo sapiens 345-348 21976975-4 2011 METHODS: The chemotherapeutic drug, dacarbazine (DTIC), that induces apoptosis through the intrinsic pathway which typically responds to severe DNA damage, was used as a model drug to prepare DTIC-loaded polylactic acid (PLA) nanoparticles (DTIC-NPs), which were covalently conjugated to a highly specific targeting functional TRAIL-receptor 2 (DR5) monoclonal antibody (mAb) that can contribute directly to cancer cell apoptosis or growth inhibition through the extrinsic pathway. Dacarbazine 192-196 TNF receptor superfamily member 10b Homo sapiens 345-348 22087285-8 2011 Furthermore, Bcl-2 was down-regulated whereas TRAIL-R1/DR4 and TRAIL-R2/DR5 expression was increased following the treatment of CSCs with GDC-0449. HhAntag691 138-146 TNF receptor superfamily member 10b Homo sapiens 63-71 22087285-8 2011 Furthermore, Bcl-2 was down-regulated whereas TRAIL-R1/DR4 and TRAIL-R2/DR5 expression was increased following the treatment of CSCs with GDC-0449. HhAntag691 138-146 TNF receptor superfamily member 10b Homo sapiens 72-75 21931821-4 2011 Simultaneously, Resveratrol treatment of DLBCL cell lines also caused ROS dependent upregulation of DR5; and interestingly, co-treatment of DLBCL with sub-toxic doses of TRAIL and Resveratrol synergistically induced apoptosis via utilizing DR5, on the other hand, gene silencing of DR5 abolished this effect. Resveratrol 180-191 TNF receptor superfamily member 10b Homo sapiens 240-243 21931821-5 2011 CONCLUSION/SIGNIFICANCE: Altogether, these data suggest that Resveratrol acts as a suppressor of AKT/PKB pathway leading to apoptosis via generation of ROS and at the same time primes DLBCL cells via up-regulation of DR5 to TRAIL-mediated apoptosis. Resveratrol 61-72 TNF receptor superfamily member 10b Homo sapiens 217-220 21127198-7 2011 DBA also induced both death receptor (DR)-5 and DR4. dibenzylidene acetone 0-3 TNF receptor superfamily member 10b Homo sapiens 22-43 21127198-8 2011 Knockdown of DR5 and DR4 by small interfering RNA (SiRNA) reduced the sensitizing effect of DBA on TRAIL-induced apoptosis. dibenzylidene acetone 92-95 TNF receptor superfamily member 10b Homo sapiens 13-16 21127198-10 2011 Knockdown of CHOP by siRNA decreased the induction of DBA-induced DR5 expression and apoptosis. dibenzylidene acetone 54-57 TNF receptor superfamily member 10b Homo sapiens 66-69 21127198-12 2011 We observed that DBA-induced induction of DR5 and DR4 was mediated through generation of reactive oxygen species (ROS), as N-acetylcysteine blocked the induction of death receptors and suppression of cell survival proteins by DBA. dibenzylidene acetone 17-20 TNF receptor superfamily member 10b Homo sapiens 42-45 21127198-12 2011 We observed that DBA-induced induction of DR5 and DR4 was mediated through generation of reactive oxygen species (ROS), as N-acetylcysteine blocked the induction of death receptors and suppression of cell survival proteins by DBA. Reactive Oxygen Species 89-112 TNF receptor superfamily member 10b Homo sapiens 42-45 21127198-12 2011 We observed that DBA-induced induction of DR5 and DR4 was mediated through generation of reactive oxygen species (ROS), as N-acetylcysteine blocked the induction of death receptors and suppression of cell survival proteins by DBA. Reactive Oxygen Species 114-117 TNF receptor superfamily member 10b Homo sapiens 42-45 21127198-12 2011 We observed that DBA-induced induction of DR5 and DR4 was mediated through generation of reactive oxygen species (ROS), as N-acetylcysteine blocked the induction of death receptors and suppression of cell survival proteins by DBA. Acetylcysteine 123-139 TNF receptor superfamily member 10b Homo sapiens 42-45 21127198-12 2011 We observed that DBA-induced induction of DR5 and DR4 was mediated through generation of reactive oxygen species (ROS), as N-acetylcysteine blocked the induction of death receptors and suppression of cell survival proteins by DBA. dibenzylidene acetone 226-229 TNF receptor superfamily member 10b Homo sapiens 42-45 21078664-11 2011 Overall, our results indicate that nimbolide can sensitize colon cancer cells to TRAIL-induced apoptosis through three distinct mechanisms: reactive oxygen species- and ERK-mediated up-regulation of DR5 and DR4, down-regulation of cell survival proteins, and up-regulation of p53 and Bax. nimbolide 35-44 TNF receptor superfamily member 10b Homo sapiens 199-202 21078664-11 2011 Overall, our results indicate that nimbolide can sensitize colon cancer cells to TRAIL-induced apoptosis through three distinct mechanisms: reactive oxygen species- and ERK-mediated up-regulation of DR5 and DR4, down-regulation of cell survival proteins, and up-regulation of p53 and Bax. Reactive Oxygen Species 140-163 TNF receptor superfamily member 10b Homo sapiens 199-202 20951126-8 2011 TRAIL- and TNF-alpha-mediated damnacanthal-induced apoptosis could be suppressed by treatment with caspase inhibitors as well as soluble death receptors Fc:DR5 and Fc:TNF-R1 chimera. damnacanthal 30-42 TNF receptor superfamily member 10b Homo sapiens 156-159 22115051-8 2011 alpha-TEA disrupted cholesterol-rich microdomains, acted cooperatively with TAM to reduce prosurvival mediators, and induced DR5-mediated mitochondria-dependent apoptosis via an endoplasmic reticulum stress-triggered pro-death pJNK/CHOP/DR5 amplification loop. alpha-TEA 0-9 TNF receptor superfamily member 10b Homo sapiens 125-128 22115051-8 2011 alpha-TEA disrupted cholesterol-rich microdomains, acted cooperatively with TAM to reduce prosurvival mediators, and induced DR5-mediated mitochondria-dependent apoptosis via an endoplasmic reticulum stress-triggered pro-death pJNK/CHOP/DR5 amplification loop. alpha-TEA 0-9 TNF receptor superfamily member 10b Homo sapiens 237-240 21931821-4 2011 Simultaneously, Resveratrol treatment of DLBCL cell lines also caused ROS dependent upregulation of DR5; and interestingly, co-treatment of DLBCL with sub-toxic doses of TRAIL and Resveratrol synergistically induced apoptosis via utilizing DR5, on the other hand, gene silencing of DR5 abolished this effect. Resveratrol 16-27 TNF receptor superfamily member 10b Homo sapiens 100-103 21931821-4 2011 Simultaneously, Resveratrol treatment of DLBCL cell lines also caused ROS dependent upregulation of DR5; and interestingly, co-treatment of DLBCL with sub-toxic doses of TRAIL and Resveratrol synergistically induced apoptosis via utilizing DR5, on the other hand, gene silencing of DR5 abolished this effect. Resveratrol 16-27 TNF receptor superfamily member 10b Homo sapiens 240-243 21931821-4 2011 Simultaneously, Resveratrol treatment of DLBCL cell lines also caused ROS dependent upregulation of DR5; and interestingly, co-treatment of DLBCL with sub-toxic doses of TRAIL and Resveratrol synergistically induced apoptosis via utilizing DR5, on the other hand, gene silencing of DR5 abolished this effect. Resveratrol 16-27 TNF receptor superfamily member 10b Homo sapiens 240-243 21931821-4 2011 Simultaneously, Resveratrol treatment of DLBCL cell lines also caused ROS dependent upregulation of DR5; and interestingly, co-treatment of DLBCL with sub-toxic doses of TRAIL and Resveratrol synergistically induced apoptosis via utilizing DR5, on the other hand, gene silencing of DR5 abolished this effect. Reactive Oxygen Species 70-73 TNF receptor superfamily member 10b Homo sapiens 100-103 21931821-4 2011 Simultaneously, Resveratrol treatment of DLBCL cell lines also caused ROS dependent upregulation of DR5; and interestingly, co-treatment of DLBCL with sub-toxic doses of TRAIL and Resveratrol synergistically induced apoptosis via utilizing DR5, on the other hand, gene silencing of DR5 abolished this effect. Resveratrol 180-191 TNF receptor superfamily member 10b Homo sapiens 100-103 21931821-4 2011 Simultaneously, Resveratrol treatment of DLBCL cell lines also caused ROS dependent upregulation of DR5; and interestingly, co-treatment of DLBCL with sub-toxic doses of TRAIL and Resveratrol synergistically induced apoptosis via utilizing DR5, on the other hand, gene silencing of DR5 abolished this effect. Resveratrol 180-191 TNF receptor superfamily member 10b Homo sapiens 240-243 21172010-6 2010 Perifosine increased phosphorylated c-Jun NH2-terminal kinase (JNK) and c-Jun levels, which were paralleled with DR4 and DR5 induction. perifosine 0-10 TNF receptor superfamily member 10b Homo sapiens 121-124 21044953-3 2010 The current study has revealed a novel ERK/ribosomal S6 kinase (RSK)-dependent mechanism that regulates DR5 expression primarily using celecoxib as a DR5 inducer. Celecoxib 135-144 TNF receptor superfamily member 10b Homo sapiens 104-107 21044953-3 2010 The current study has revealed a novel ERK/ribosomal S6 kinase (RSK)-dependent mechanism that regulates DR5 expression primarily using celecoxib as a DR5 inducer. Celecoxib 135-144 TNF receptor superfamily member 10b Homo sapiens 150-153 21044953-4 2010 Both C/EBP homologous protein (CHOP) and Elk1 are required for celecoxib-induced DR5 expression based on promoter deletion and mutation analysis and siRNA-mediated gene silencing results. Celecoxib 63-72 TNF receptor superfamily member 10b Homo sapiens 81-84 21044953-7 2010 Inhibition of either ERK signaling with a MEK inhibitor or ERK1/2 siRNA, or RSK2 signaling with an RSK2 inhibitor or RSK2 siRNA abrogated DR5 up-regulation by celecoxib as well as other agents. Celecoxib 159-168 TNF receptor superfamily member 10b Homo sapiens 138-141 21044953-10 2010 Additionally, celecoxib increased CHOP promoter activity in an ATF4-dependent manner, and siRNA-mediated blockade of ATF4 abrogated both CHOP induction and DR5 up-regulation, indicating that ATF4 is involved in celecoxib-induced CHOP and DR5 expression. Celecoxib 211-220 TNF receptor superfamily member 10b Homo sapiens 156-159 21044953-10 2010 Additionally, celecoxib increased CHOP promoter activity in an ATF4-dependent manner, and siRNA-mediated blockade of ATF4 abrogated both CHOP induction and DR5 up-regulation, indicating that ATF4 is involved in celecoxib-induced CHOP and DR5 expression. Celecoxib 211-220 TNF receptor superfamily member 10b Homo sapiens 238-241 21044953-11 2010 Collectively, we conclude that small molecules such as celecoxib induce DR5 expression through activating ERK/RSK signaling and subsequent Elk1 activation and ATF4-dependent CHOP induction. Celecoxib 55-64 TNF receptor superfamily member 10b Homo sapiens 72-75 21172010-0 2010 c-Jun NH2-terminal kinase-dependent upregulation of DR5 mediates cooperative induction of apoptosis by perifosine and TRAIL. perifosine 103-113 TNF receptor superfamily member 10b Homo sapiens 52-55 21172010-4 2010 Interestingly, perifosine primarily increased cell surface levels of DR5 although it elevated the expression of both DR4 and DR5. perifosine 15-25 TNF receptor superfamily member 10b Homo sapiens 69-72 21172010-4 2010 Interestingly, perifosine primarily increased cell surface levels of DR5 although it elevated the expression of both DR4 and DR5. perifosine 15-25 TNF receptor superfamily member 10b Homo sapiens 125-128 21172010-7 2010 However, only DR5 upregulaiton induced by perifosine could be abrogated by both the JNK inhibitor SP600125 and JNK siRNA. perifosine 42-52 TNF receptor superfamily member 10b Homo sapiens 14-17 21172010-5 2010 Blockade of DR5, but not DR4 upregulation, via small interfering RNA (siRNA) inhibited perifosine/TRAIL-induced apoptosis. perifosine 87-97 TNF receptor superfamily member 10b Homo sapiens 12-15 21172010-7 2010 However, only DR5 upregulaiton induced by perifosine could be abrogated by both the JNK inhibitor SP600125 and JNK siRNA. pyrazolanthrone 98-106 TNF receptor superfamily member 10b Homo sapiens 14-17 21172010-8 2010 The antioxidants, N-acetylcysteine and glutathione, but not vitamin C or tiron, inhibited perifosine-induced elevation of p-c-Jun, DR4 and DR5. Acetylcysteine 18-34 TNF receptor superfamily member 10b Homo sapiens 139-142 21172010-8 2010 The antioxidants, N-acetylcysteine and glutathione, but not vitamin C or tiron, inhibited perifosine-induced elevation of p-c-Jun, DR4 and DR5. Glutathione 39-50 TNF receptor superfamily member 10b Homo sapiens 139-142 21172010-8 2010 The antioxidants, N-acetylcysteine and glutathione, but not vitamin C or tiron, inhibited perifosine-induced elevation of p-c-Jun, DR4 and DR5. perifosine 90-100 TNF receptor superfamily member 10b Homo sapiens 139-142 21172010-10 2010 CONCLUSIONS: DR5 induction plays a critical role in mediating perifosine/TRAIL-induced apoptosis. perifosine 62-72 TNF receptor superfamily member 10b Homo sapiens 13-16 21172010-11 2010 Perifosine induces DR5 expression through a JNK-dependent mechanism independent of reactive oxygen species. perifosine 0-10 TNF receptor superfamily member 10b Homo sapiens 19-22 21067180-2 2010 TCN induced cancer cell death treatment by triggering mitochondrial and death receptor 5 (DR5) apoptotic pathways. tricetin 0-3 TNF receptor superfamily member 10b Homo sapiens 72-88 21179458-15 2010 Inhibition of FOXO transcription factors by shRNA blocked resveratrol-induced upregulation of Bim, TRAIL, DR4, DR5, p27/KIP1 and apoptosis, and inhibition of cyclin D1 by resveratrol. Resveratrol 58-69 TNF receptor superfamily member 10b Homo sapiens 111-114 21067180-2 2010 TCN induced cancer cell death treatment by triggering mitochondrial and death receptor 5 (DR5) apoptotic pathways. tricetin 0-3 TNF receptor superfamily member 10b Homo sapiens 90-93 21067180-4 2010 Both of the antioxidants vitamin C and catalase significantly decreased apoptosis by inhibiting the phosphorylation of JNK and subsequently triggering DR5 cell death pathways. Ascorbic Acid 25-34 TNF receptor superfamily member 10b Homo sapiens 151-154 20804743-7 2010 Moreover, combined silencing of DR4 and DR5 by small interfering RNA abrogates the ability of PTX to induce TRAIL-mediated apoptosis. Pentoxifylline 94-97 TNF receptor superfamily member 10b Homo sapiens 40-43 21159614-0 2010 Contrasting effects of nutlin-3 on TRAIL- and docetaxel-induced apoptosis due to upregulation of TRAIL-R2 and Mcl-1 in human melanoma cells. Docetaxel 46-55 TNF receptor superfamily member 10b Homo sapiens 97-105 20804743-6 2010 PTX induces the expression of death receptors DR4 and DR5 on cell surface of both the cell types where c-Jun NH2-terminal kinase (JNK) pathway plays an important role. Pentoxifylline 0-3 TNF receptor superfamily member 10b Homo sapiens 54-57 20832390-6 2010 Knocking down DR5 using siRNA completely attenuated Chl-induced caspase-8 cleavage but partially inhibited apoptosis. Chlorogenic Acid 52-55 TNF receptor superfamily member 10b Homo sapiens 14-17 20832390-7 2010 Antioxidant NAC attenuated Chl-induced oxidative stress-mediated inhibition of Bcr-Abl phosphorylation, DR5 upregulation, caspase activation and CML cell death. Chlorogenic Acid 27-30 TNF receptor superfamily member 10b Homo sapiens 104-107 20623264-8 2010 The enhancement of Ad-TRAIL by cisplatin was due to the up-regulation of DR5 but not DR4 expression, and followed by the down-regulation of survivin and activation of Caspase 3. Cisplatin 31-40 TNF receptor superfamily member 10b Homo sapiens 73-76 20605676-2 2010 Here in this study, we show that celastrol up-regulates death receptor 4 (DR4) and 5 (DR5) expression at mRNA, total protein and cell surface levels, and the specific knockdown using DR4- or DR5-targeting siRNA transfection attenuates the PARP cleavage caused by the combination of celastrol and TRAIL/Apo-2L, denoting the critical roles of DR induction in this sensitization. celastrol 33-42 TNF receptor superfamily member 10b Homo sapiens 86-89 20605676-2 2010 Here in this study, we show that celastrol up-regulates death receptor 4 (DR4) and 5 (DR5) expression at mRNA, total protein and cell surface levels, and the specific knockdown using DR4- or DR5-targeting siRNA transfection attenuates the PARP cleavage caused by the combination of celastrol and TRAIL/Apo-2L, denoting the critical roles of DR induction in this sensitization. celastrol 33-42 TNF receptor superfamily member 10b Homo sapiens 191-194 20605676-5 2010 Taken together, the present study demonstrates that the enhanced mRNA and protein expression of DR4 and DR5 play prominent roles in the sensitization of celastrol to TRAIL/Apo-2L-induced apoptosis, in a p38 MAPK-independent manner. celastrol 153-162 TNF receptor superfamily member 10b Homo sapiens 104-107 20605676-2 2010 Here in this study, we show that celastrol up-regulates death receptor 4 (DR4) and 5 (DR5) expression at mRNA, total protein and cell surface levels, and the specific knockdown using DR4- or DR5-targeting siRNA transfection attenuates the PARP cleavage caused by the combination of celastrol and TRAIL/Apo-2L, denoting the critical roles of DR induction in this sensitization. celastrol 282-291 TNF receptor superfamily member 10b Homo sapiens 86-89 20837473-11 2010 Overall, our results show that gossypol enhances TRAIL-induced apoptosis through the down-regulation of cell survival proteins and the up-regulation of TRAIL death receptors through the ROS-ERK-CHOP-DR5 pathway. Reactive Oxygen Species 186-189 TNF receptor superfamily member 10b Homo sapiens 199-202 20580868-4 2010 Sensitizing doses of vorinostat, valproic acid, sodium butyrate and MS-275 regulated the expression of TRAIL-R2, c-FLIP and Apaf-1 in leukemic cells while TSA modulated only the expression of Apaf-1. Vorinostat 21-31 TNF receptor superfamily member 10b Homo sapiens 103-111 20837473-6 2010 Gossypol-induced receptor induction was not cell type-specific, as DR5 induction was observed in other cell types. Gossypol 0-8 TNF receptor superfamily member 10b Homo sapiens 67-70 20837473-7 2010 Deletion of DR5 by siRNA significantly reduced the apoptosis induced by TRAIL and gossypol. Gossypol 82-90 TNF receptor superfamily member 10b Homo sapiens 12-15 20837473-8 2010 Gossypol induction of the death receptor required the induction of CHOP, and thus, gene silencing of CHOP abolished gossypol-induced DR5 expression and associated potentiation of apoptosis. Gossypol 116-124 TNF receptor superfamily member 10b Homo sapiens 133-136 20837473-9 2010 ERK1/2 (but not p38 MAPK or JNK) activation was also required for gossypol-induced TRAIL receptor induction; gene silencing of ERK abolished both DR5 induction and potentiation of apoptosis by TRAIL. Gossypol 66-74 TNF receptor superfamily member 10b Homo sapiens 146-149 20837473-11 2010 Overall, our results show that gossypol enhances TRAIL-induced apoptosis through the down-regulation of cell survival proteins and the up-regulation of TRAIL death receptors through the ROS-ERK-CHOP-DR5 pathway. Gossypol 31-39 TNF receptor superfamily member 10b Homo sapiens 199-202 20580868-4 2010 Sensitizing doses of vorinostat, valproic acid, sodium butyrate and MS-275 regulated the expression of TRAIL-R2, c-FLIP and Apaf-1 in leukemic cells while TSA modulated only the expression of Apaf-1. Valproic Acid 33-46 TNF receptor superfamily member 10b Homo sapiens 103-111 20580868-4 2010 Sensitizing doses of vorinostat, valproic acid, sodium butyrate and MS-275 regulated the expression of TRAIL-R2, c-FLIP and Apaf-1 in leukemic cells while TSA modulated only the expression of Apaf-1. Butyric Acid 48-63 TNF receptor superfamily member 10b Homo sapiens 103-111 20851102-6 2010 Mechanistically, 2DG and GD enhanced surface levels for both death receptors (DR4 and DR5); which was accompanied by reductions in levels of Mcl-1, Bcl-2 and survivin. Deoxyglucose 17-20 TNF receptor superfamily member 10b Homo sapiens 86-89 20886583-0 2010 alpha-TEA induces apoptosis of human breast cancer cells via activation of TRAIL/DR5 death receptor pathway. alpha-TEA 0-9 TNF receptor superfamily member 10b Homo sapiens 81-84 20886583-2 2010 alpha-TEA induces increased levels of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and death receptor-5 (DR5) and decreased levels of antiapoptotic factor, cellular FLICE-like inhibitory protein (c-FLIP L). alpha-TEA 0-9 TNF receptor superfamily member 10b Homo sapiens 106-122 20886583-2 2010 alpha-TEA induces increased levels of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and death receptor-5 (DR5) and decreased levels of antiapoptotic factor, cellular FLICE-like inhibitory protein (c-FLIP L). alpha-TEA 0-9 TNF receptor superfamily member 10b Homo sapiens 124-127 20886583-3 2010 DR5/TRAIL induced apoptosis involves downregulation of c-FLIP (L), caspase-8 activation, activated proapoptotic mediators tBid and Bax, mitochondrial permeability transition, and activation of caspase-9. tBID 122-126 TNF receptor superfamily member 10b Homo sapiens 0-3 20886583-4 2010 siRNA knockdown of either DR5 or TRAIL blocks the ability of alpha-TEA to enhance DR5 protein levels, downregulate c-FLIP(L) protein levels and induce apoptosis. alpha-TEA 61-70 TNF receptor superfamily member 10b Homo sapiens 26-29 20886583-4 2010 siRNA knockdown of either DR5 or TRAIL blocks the ability of alpha-TEA to enhance DR5 protein levels, downregulate c-FLIP(L) protein levels and induce apoptosis. alpha-TEA 61-70 TNF receptor superfamily member 10b Homo sapiens 82-85 20886583-5 2010 Combination of alpha-TEA + TRAIL acts cooperatively to induce apoptosis, and increase DR5 and decrease c-FLIP (L) protein levels. alpha-tea + 15-26 TNF receptor superfamily member 10b Homo sapiens 86-89 20886583-7 2010 Taken together, these studies show that alpha-TEA induces TRAIL/DR5 mitochondria-dependent apoptosis in human breast cancer cells, and that TRAIL/DR5-dependent increases in DR5 and decreases in c-FLIP expression are triggered by TRAIL or alpha-TEA treatments. alpha-TEA 40-49 TNF receptor superfamily member 10b Homo sapiens 64-67 20886583-7 2010 Taken together, these studies show that alpha-TEA induces TRAIL/DR5 mitochondria-dependent apoptosis in human breast cancer cells, and that TRAIL/DR5-dependent increases in DR5 and decreases in c-FLIP expression are triggered by TRAIL or alpha-TEA treatments. alpha-TEA 40-49 TNF receptor superfamily member 10b Homo sapiens 146-149 20886583-7 2010 Taken together, these studies show that alpha-TEA induces TRAIL/DR5 mitochondria-dependent apoptosis in human breast cancer cells, and that TRAIL/DR5-dependent increases in DR5 and decreases in c-FLIP expression are triggered by TRAIL or alpha-TEA treatments. alpha-TEA 40-49 TNF receptor superfamily member 10b Homo sapiens 146-149 20851102-7 2010 Mannose pre-treatment reduced enhanced killing by combination treatments, accompanied by reduced DR5 levels. Mannose 0-7 TNF receptor superfamily member 10b Homo sapiens 97-100 20674558-6 2010 DP increased the levels of reactive oxygen species (ROS) in HepG2 cells, and antioxidant N-acetylcysteine (NAC) completely blocked DP-induced ROS accumulation and the disruption of the balance between Bax and Bcl-2 proteins, and effectively blocked the decreased MMP and apoptosis, but had no effect on the activation of caspase-8 and the up-regulations of DR4 and DR5 induced by DP. 1-(3',4',5'-trimethoxyphenyl)-3-(3'',4''-dimethoxy-2'-hydroxyphenyl)propane 0-2 TNF receptor superfamily member 10b Homo sapiens 365-368 20974006-2 2010 alpha-TEA induces apoptosis via activation of extrinsic death receptors Fas (CD95) and DR5, JNK/p73/Noxa pathways, and suppression of anti-apoptotic mediators Akt, ERK, c-FLIP and survivin in breast, ovarian and prostate cancer cells. alpha-TEA 0-9 TNF receptor superfamily member 10b Homo sapiens 87-90 20974006-3 2010 RESULTS: In this study, we demonstrate that alpha-TEA induces the accumulation of cell surface membrane ceramide, leading to co-localization with Fas, DR5, and FADD, followed by activation of caspases-8 and -9 and apoptosis in human MDA-MB-231 breast cancer cells. alpha-TEA 44-53 TNF receptor superfamily member 10b Homo sapiens 151-154 20974006-3 2010 RESULTS: In this study, we demonstrate that alpha-TEA induces the accumulation of cell surface membrane ceramide, leading to co-localization with Fas, DR5, and FADD, followed by activation of caspases-8 and -9 and apoptosis in human MDA-MB-231 breast cancer cells. Ceramides 104-112 TNF receptor superfamily member 10b Homo sapiens 151-154 20974006-5 2010 Functional knockdown of ASMase with either the chemical inhibitor, desipramine, or siRNA markedly reduces alpha-TEA-induced cell surface membrane accumulation of ceramide and its co-localization with Fas, DR5, and FADD, cleavage of caspases-8 and -9 and apoptosis, suggesting an early and critical role for ASMase in alpha-TEA-induced apoptosis. alpha-TEA 106-115 TNF receptor superfamily member 10b Homo sapiens 205-208 20674558-6 2010 DP increased the levels of reactive oxygen species (ROS) in HepG2 cells, and antioxidant N-acetylcysteine (NAC) completely blocked DP-induced ROS accumulation and the disruption of the balance between Bax and Bcl-2 proteins, and effectively blocked the decreased MMP and apoptosis, but had no effect on the activation of caspase-8 and the up-regulations of DR4 and DR5 induced by DP. 1-(3',4',5'-trimethoxyphenyl)-3-(3'',4''-dimethoxy-2'-hydroxyphenyl)propane 131-133 TNF receptor superfamily member 10b Homo sapiens 365-368 20674558-6 2010 DP increased the levels of reactive oxygen species (ROS) in HepG2 cells, and antioxidant N-acetylcysteine (NAC) completely blocked DP-induced ROS accumulation and the disruption of the balance between Bax and Bcl-2 proteins, and effectively blocked the decreased MMP and apoptosis, but had no effect on the activation of caspase-8 and the up-regulations of DR4 and DR5 induced by DP. Acetylcysteine 89-105 TNF receptor superfamily member 10b Homo sapiens 365-368 20674558-6 2010 DP increased the levels of reactive oxygen species (ROS) in HepG2 cells, and antioxidant N-acetylcysteine (NAC) completely blocked DP-induced ROS accumulation and the disruption of the balance between Bax and Bcl-2 proteins, and effectively blocked the decreased MMP and apoptosis, but had no effect on the activation of caspase-8 and the up-regulations of DR4 and DR5 induced by DP. Acetylcysteine 107-110 TNF receptor superfamily member 10b Homo sapiens 365-368 20674558-6 2010 DP increased the levels of reactive oxygen species (ROS) in HepG2 cells, and antioxidant N-acetylcysteine (NAC) completely blocked DP-induced ROS accumulation and the disruption of the balance between Bax and Bcl-2 proteins, and effectively blocked the decreased MMP and apoptosis, but had no effect on the activation of caspase-8 and the up-regulations of DR4 and DR5 induced by DP. 1-(3',4',5'-trimethoxyphenyl)-3-(3'',4''-dimethoxy-2'-hydroxyphenyl)propane 131-133 TNF receptor superfamily member 10b Homo sapiens 365-368 21713362-2 2010 Previously, apple procyanidins (Pcy) have been shown to upregulate the expression of TRAIL-DR4/-DR5 and thereby overcoming the resistance of human colon cancer-derived metastatic SW620 cells to TRAIL. Proanthocyanidins 32-35 TNF receptor superfamily member 10b Homo sapiens 96-99 20410100-7 2010 We genotyped single nucleotide polymorphisms (SNPs) in candidate genes in 8p12-p21 and found a significant association between fatty acids and SNPs in apolipoprotein J (APOJ), lipoprotein lipase (LPL), macrophage scavenger receptor 1 (MSR1), and tumor necrosis factor receptor superfamily member 10b (TNFRSF10B). Fatty Acids 127-138 TNF receptor superfamily member 10b Homo sapiens 246-299 20354842-7 2010 Interestingly, inhibition of PKC by Goe6983 enabled DR5 to trigger apoptotic signaling in response to TRAIL and also strongly enhanced lexatumumab-mediated cell death. goe6983 36-43 TNF receptor superfamily member 10b Homo sapiens 52-55 21713362-2 2010 Previously, apple procyanidins (Pcy) have been shown to upregulate the expression of TRAIL-DR4/-DR5 and thereby overcoming the resistance of human colon cancer-derived metastatic SW620 cells to TRAIL. Proanthocyanidins 18-30 TNF receptor superfamily member 10b Homo sapiens 96-99 20660600-0 2010 A combination of DR5 agonistic monoclonal antibody with gemcitabine targets pancreatic cancer stem cells and results in long-term disease control in human pancreatic cancer model. gemcitabine 56-67 TNF receptor superfamily member 10b Homo sapiens 17-20 20686688-0 2010 Role of endoplasmic reticulum stress in alpha-TEA mediated TRAIL/DR5 death receptor dependent apoptosis. 2,5,7,8-tetramethyl-2R-(4R,8R,12-trimethyltridecyl)chroman-6-yloxy acetic acid 40-49 TNF receptor superfamily member 10b Homo sapiens 65-68 20686688-5 2010 CONCLUSION: Taken together, ER stress plays an important role in alpha-TEA induced apoptosis by enhancing DR5/caspase-8 pro-apoptotic signaling and suppressing anti-apoptotic factors c-FLIP and Bcl-2 via ER stress mediated JNK/CHOP/DR5/caspase-8 signaling. 2,5,7,8-tetramethyl-2R-(4R,8R,12-trimethyltridecyl)chroman-6-yloxy acetic acid 65-74 TNF receptor superfamily member 10b Homo sapiens 106-109 20686688-5 2010 CONCLUSION: Taken together, ER stress plays an important role in alpha-TEA induced apoptosis by enhancing DR5/caspase-8 pro-apoptotic signaling and suppressing anti-apoptotic factors c-FLIP and Bcl-2 via ER stress mediated JNK/CHOP/DR5/caspase-8 signaling. 2,5,7,8-tetramethyl-2R-(4R,8R,12-trimethyltridecyl)chroman-6-yloxy acetic acid 65-74 TNF receptor superfamily member 10b Homo sapiens 232-235 20410100-0 2010 Genetic variation in APOJ, LPL, and TNFRSF10B affects plasma fatty acid distribution in Alaskan Eskimos. Fatty Acids 61-71 TNF receptor superfamily member 10b Homo sapiens 36-45 20410100-7 2010 We genotyped single nucleotide polymorphisms (SNPs) in candidate genes in 8p12-p21 and found a significant association between fatty acids and SNPs in apolipoprotein J (APOJ), lipoprotein lipase (LPL), macrophage scavenger receptor 1 (MSR1), and tumor necrosis factor receptor superfamily member 10b (TNFRSF10B). Fatty Acids 127-138 TNF receptor superfamily member 10b Homo sapiens 301-310 20410100-8 2010 A Bayesian quantitative trait nucleotide analysis based on a measured genotype model showed that SNPs in LPL, TNFRSF10B, and APOJ had strong statistical evidence of a functional effect (posterior probability > or =75%) on plasma fatty acid distribution. Fatty Acids 232-242 TNF receptor superfamily member 10b Homo sapiens 110-119 19538462-3 2010 While nickel treatment increased surface expression of the apoptosis-inducing TRAIL receptors TRAIL-R1 and TRAIL-R2, it also up-regulated the apoptosis-deficient TRAIL-R4, suggesting that modulation of TRAIL receptor expression alone is unlikely to fully account for the dramatic sensitization effect of nickel. Nickel 6-12 TNF receptor superfamily member 10b Homo sapiens 107-115 19538480-6 2010 Gene reporter assays confirmed that the RA-induced t-PA gene expression occurred through interactions of retinoid receptors (RARs and RXRs) with a DR5 response element located at -7 kb from the transcription site. Tretinoin 40-42 TNF receptor superfamily member 10b Homo sapiens 147-150 19538480-8 2010 Chromatin immunoprecipitation demonstrated that interactions between RARs, RXRs and t-PA promoter were time dependent: RAR-alpha/RXR-alpha bound DR5 motif before and up to 12 hrs of RA exposure, and RAR-beta/RXR-alpha bound DR5 response element after 12 hrs of RA treatment. Tretinoin 69-71 TNF receptor superfamily member 10b Homo sapiens 145-148 19538480-8 2010 Chromatin immunoprecipitation demonstrated that interactions between RARs, RXRs and t-PA promoter were time dependent: RAR-alpha/RXR-alpha bound DR5 motif before and up to 12 hrs of RA exposure, and RAR-beta/RXR-alpha bound DR5 response element after 12 hrs of RA treatment. Tretinoin 69-71 TNF receptor superfamily member 10b Homo sapiens 224-227 19538480-8 2010 Chromatin immunoprecipitation demonstrated that interactions between RARs, RXRs and t-PA promoter were time dependent: RAR-alpha/RXR-alpha bound DR5 motif before and up to 12 hrs of RA exposure, and RAR-beta/RXR-alpha bound DR5 response element after 12 hrs of RA treatment. Tretinoin 119-121 TNF receptor superfamily member 10b Homo sapiens 145-148 19538480-8 2010 Chromatin immunoprecipitation demonstrated that interactions between RARs, RXRs and t-PA promoter were time dependent: RAR-alpha/RXR-alpha bound DR5 motif before and up to 12 hrs of RA exposure, and RAR-beta/RXR-alpha bound DR5 response element after 12 hrs of RA treatment. Tretinoin 119-121 TNF receptor superfamily member 10b Homo sapiens 224-227 20515942-0 2010 Butein sensitizes human hepatoma cells to TRAIL-induced apoptosis via extracellular signal-regulated kinase/Sp1-dependent DR5 upregulation and NF-kappaB inactivation. butein 0-6 TNF receptor superfamily member 10b Homo sapiens 122-125 20515942-4 2010 In this study, we determined that butein enhances TRAIL-induced apoptosis in hepatoma cells through upregulation of DR5. butein 34-40 TNF receptor superfamily member 10b Homo sapiens 116-119 20515942-6 2010 Electrophoretic mobility shift assays and chromatin immunoprecipitation studies were used to analyze the elevation of Sp1 binding to DR5 promoter sites by butein. butein 155-161 TNF receptor superfamily member 10b Homo sapiens 133-136 20515942-8 2010 Furthermore, pretreatment of the blocking chimeric antibody and small interfering RNA for DR5 significantly suppressed TRAIL-mediated apoptosis by butein in Hep3B cells. butein 147-153 TNF receptor superfamily member 10b Homo sapiens 90-93 20515942-9 2010 Butein also stimulated extracellular signal-regulated kinase (ERK) activation, and the ERK inhibitor PD98059 blocked butein-induced DR5 expression and suppressed binding of Sp1 to the DR5 promoter. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 101-108 TNF receptor superfamily member 10b Homo sapiens 132-135 20515942-9 2010 Butein also stimulated extracellular signal-regulated kinase (ERK) activation, and the ERK inhibitor PD98059 blocked butein-induced DR5 expression and suppressed binding of Sp1 to the DR5 promoter. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 101-108 TNF receptor superfamily member 10b Homo sapiens 184-187 20515942-9 2010 Butein also stimulated extracellular signal-regulated kinase (ERK) activation, and the ERK inhibitor PD98059 blocked butein-induced DR5 expression and suppressed binding of Sp1 to the DR5 promoter. butein 117-123 TNF receptor superfamily member 10b Homo sapiens 132-135 20224297-3 2010 Our data indicate that DHA suppresses the PI3-K/Akt and ERK cell survival pathways and triggers the induction of death receptor DR5 and activation of extrinsic and intrinsic cell death signaling. artenimol 23-26 TNF receptor superfamily member 10b Homo sapiens 128-131 20224297-4 2010 DHA-mediated DR5 induction appears to occur via increased transcriptional activity of DR5 promoter. artenimol 0-3 TNF receptor superfamily member 10b Homo sapiens 13-16 20224297-4 2010 DHA-mediated DR5 induction appears to occur via increased transcriptional activity of DR5 promoter. artenimol 0-3 TNF receptor superfamily member 10b Homo sapiens 86-89 20356045-0 2010 6-dehydrogingerdione sensitizes human hepatoblastoma Hep G2 cells to TRAIL-induced apoptosis via reactive oxygen species-mediated increase of DR5. 6-dehydrogingerdione 0-20 TNF receptor superfamily member 10b Homo sapiens 142-145 19900759-2 2010 In the gene expression assay using a DNA microalley, adenosine upregulated mRNAs for tumor necrosis factor (TNF), TNF receptor 1-associated death domain protein (TRADD), TNF related apoptosis-inducing ligand receptor 2 (TRAIL-R2), TRADD/receptor-interacting protein kinase 1 (RIPK1), Fas-associated death domain protein (FADD), and caspase-9, involving activation of caspase-8 and -9 followed by the effector caspase-3. Adenosine 53-62 TNF receptor superfamily member 10b Homo sapiens 170-218 19900759-2 2010 In the gene expression assay using a DNA microalley, adenosine upregulated mRNAs for tumor necrosis factor (TNF), TNF receptor 1-associated death domain protein (TRADD), TNF related apoptosis-inducing ligand receptor 2 (TRAIL-R2), TRADD/receptor-interacting protein kinase 1 (RIPK1), Fas-associated death domain protein (FADD), and caspase-9, involving activation of caspase-8 and -9 followed by the effector caspase-3. Adenosine 53-62 TNF receptor superfamily member 10b Homo sapiens 220-228 20356045-9 2010 Taken together, these data suggested that in addition to the mitochondrial- and Fas receptor-mediated apoptotic pathways involved, ROS-dependent and p53-regulated DR5 expression was also demonstrated to play a pivotal role in the synergistic enhancement of TRAIL-induced apoptosis instigated by 6-DG in Hep G2 cells. Reactive Oxygen Species 131-134 TNF receptor superfamily member 10b Homo sapiens 163-166 20356045-0 2010 6-dehydrogingerdione sensitizes human hepatoblastoma Hep G2 cells to TRAIL-induced apoptosis via reactive oxygen species-mediated increase of DR5. Reactive Oxygen Species 97-120 TNF receptor superfamily member 10b Homo sapiens 142-145 20356045-9 2010 Taken together, these data suggested that in addition to the mitochondrial- and Fas receptor-mediated apoptotic pathways involved, ROS-dependent and p53-regulated DR5 expression was also demonstrated to play a pivotal role in the synergistic enhancement of TRAIL-induced apoptosis instigated by 6-DG in Hep G2 cells. 6-dehydrogingerdione 295-299 TNF receptor superfamily member 10b Homo sapiens 163-166 20356045-5 2010 Abrogation of p53 expression by p53 small interfering RNA significantly attenuated 6-DG-induced DR5 expression, thus rendering these cells resistant to TRAIL-induced apoptosis. 6-dehydrogingerdione 83-87 TNF receptor superfamily member 10b Homo sapiens 96-99 20356045-6 2010 DR5 expression after 6-DG treatment was accompanied by provoking intracellular reactive oxygen species (ROS) generation. 6-dehydrogingerdione 21-25 TNF receptor superfamily member 10b Homo sapiens 0-3 20356045-6 2010 DR5 expression after 6-DG treatment was accompanied by provoking intracellular reactive oxygen species (ROS) generation. Reactive Oxygen Species 79-102 TNF receptor superfamily member 10b Homo sapiens 0-3 20356045-6 2010 DR5 expression after 6-DG treatment was accompanied by provoking intracellular reactive oxygen species (ROS) generation. Reactive Oxygen Species 104-107 TNF receptor superfamily member 10b Homo sapiens 0-3 20356045-7 2010 Pretreatment with N-acetyl-l-cysteine (NAC) attenuated 6-DG-induced DR5 expression and inhibited TRAIL-induced apoptosis. Acetylcysteine 18-37 TNF receptor superfamily member 10b Homo sapiens 68-71 20356045-7 2010 Pretreatment with N-acetyl-l-cysteine (NAC) attenuated 6-DG-induced DR5 expression and inhibited TRAIL-induced apoptosis. Acetylcysteine 39-42 TNF receptor superfamily member 10b Homo sapiens 68-71 20356045-7 2010 Pretreatment with N-acetyl-l-cysteine (NAC) attenuated 6-DG-induced DR5 expression and inhibited TRAIL-induced apoptosis. 6-dehydrogingerdione 55-59 TNF receptor superfamily member 10b Homo sapiens 68-71 20356045-8 2010 In contrast to Hep G2 cells, DR5 up-regulation and sensitization to TRAIL-induced apoptosis instigated by 6-DG were not observed in normal MDCK cells. 6-dehydrogingerdione 106-110 TNF receptor superfamily member 10b Homo sapiens 29-32 19965642-7 2010 In CLL patients receiving lenalidomide, similar evidence of CD154 activation is observed including BID, DR5, and p73 induction and also development of anti-ROR1 tumor-directed antibodies. Lenalidomide 26-38 TNF receptor superfamily member 10b Homo sapiens 104-107 20154087-8 2010 We found that celastrol also induced reactive oxygen species (ROS) generation, and ROS sequestration inhibited celastrol-induced expression of CHOP and DR5, and consequent sensitization to TRAIL. Reactive Oxygen Species 83-86 TNF receptor superfamily member 10b Homo sapiens 152-155 20154087-8 2010 We found that celastrol also induced reactive oxygen species (ROS) generation, and ROS sequestration inhibited celastrol-induced expression of CHOP and DR5, and consequent sensitization to TRAIL. celastrol 111-120 TNF receptor superfamily member 10b Homo sapiens 152-155 20154087-4 2010 In addition, we found that celastrol induced the cell surface expression of both the TRAIL receptors DR4 and DR5. celastrol 27-36 TNF receptor superfamily member 10b Homo sapiens 109-112 20154087-7 2010 Induction of the death receptor by the triterpenoid was found to be p53-independent but required the induction of CAAT/enhancer-binding protein homologous protein (CHOP), inasmuch as gene silencing of CHOP abolished the induction of DR5 expression by celastrol and associated enhancement of TRAIL-induced apoptosis. triterpenoid TP-222 39-51 TNF receptor superfamily member 10b Homo sapiens 233-236 20371719-5 2010 The combination of AD5-10 with carboplatin exerts a more than additive effect in vitro, which may at least partially be explained by the fact that carboplatin triggers DR5 expression on ovarian cancer cells. ad5-10 19-25 TNF receptor superfamily member 10b Homo sapiens 168-171 20156289-4 2010 To identify peptides that specifically interact with DR5, a disulfide-constrained phage display peptide library was screened for binders towards this receptor. Disulfides 60-69 TNF receptor superfamily member 10b Homo sapiens 53-56 20354117-0 2010 beta-Ionone enhances TRAIL-induced apoptosis in hepatocellular carcinoma cells through Sp1-dependent upregulation of DR5 and downregulation of NF-kappaB activity. beta-ionone 0-11 TNF receptor superfamily member 10b Homo sapiens 117-120 20371719-5 2010 The combination of AD5-10 with carboplatin exerts a more than additive effect in vitro, which may at least partially be explained by the fact that carboplatin triggers DR5 expression on ovarian cancer cells. Carboplatin 31-42 TNF receptor superfamily member 10b Homo sapiens 168-171 20371719-5 2010 The combination of AD5-10 with carboplatin exerts a more than additive effect in vitro, which may at least partially be explained by the fact that carboplatin triggers DR5 expression on ovarian cancer cells. Carboplatin 147-158 TNF receptor superfamily member 10b Homo sapiens 168-171 20096292-0 2010 Quercetin enhances TRAIL-induced apoptosis in prostate cancer cells via increased protein stability of death receptor 5. Quercetin 0-9 TNF receptor superfamily member 10b Homo sapiens 103-119 19922463-10 2010 Bortezomib also increased DR4 and DR5 expression in the presence of stroma. Bortezomib 0-10 TNF receptor superfamily member 10b Homo sapiens 34-37 20106985-4 2010 The cysteine-rich domains of DR5 have a conservative role in tumor necrosis factor-related apoptosis-inducing ligand-DR5-mediated apoptosis, and the pre-ligand assembly domain within the N1-cap contributes to the ligand-independent formation of receptor complexes. Cysteine 4-12 TNF receptor superfamily member 10b Homo sapiens 29-32 20106985-4 2010 The cysteine-rich domains of DR5 have a conservative role in tumor necrosis factor-related apoptosis-inducing ligand-DR5-mediated apoptosis, and the pre-ligand assembly domain within the N1-cap contributes to the ligand-independent formation of receptor complexes. Cysteine 4-12 TNF receptor superfamily member 10b Homo sapiens 117-120 20106985-9 2010 Moreover, permutation analysis showed that Leu(6) was pivotal for the interaction of DR5 and the agonistic antibody. Leucine 43-46 TNF receptor superfamily member 10b Homo sapiens 85-88 20096292-1 2010 AIMS: Quercetin has been shown to enhance tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis of prostate cancer cells via mechanisms that include upregulation of death receptor (DR) 5, a protein reported to play an important role in sensitizing cancer cells to apoptosis. Quercetin 6-15 TNF receptor superfamily member 10b Homo sapiens 193-214 20096292-2 2010 We aimed to determine the specific mechanisms underlying quercetin-induced DR5 expression. Quercetin 57-66 TNF receptor superfamily member 10b Homo sapiens 75-78 20096292-8 2010 Quercetin dose-dependently increased DR5 levels in prostate cancer cells, which was mediated by increased transcription and protein stability, but not mRNA stability. Quercetin 0-9 TNF receptor superfamily member 10b Homo sapiens 37-40