PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 2794752-7 1989 Additionally, qinghaosu resulted in a dose-dependent increase in red cell lysis and methemoglobin generation while decreasing reduced glutathione concentration. artemisinin 14-23 hemoglobin subunit gamma 2 Homo sapiens 84-97 33894419-0 2021 Artemisinin derivatives inhibit adipogenic differentiation of 3T3-L1 preadipocytes through upregulation of CHOP. artemisinin 0-11 DNA-damage inducible transcript 3 Mus musculus 107-111 33976269-5 2021 Artemisinin-resistance was assessed through in-vivo parasite clearance half-life (PC1/2), ex-vivo ring-stage survivability (RSA), and genome analysis of kelch13 and other candidate gene (pfcrt, pfmdr1, pfatpase 6, pfdhfr and pfdhps). artemisinin 0-11 proprotein convertase subtilisin/kexin type 1 Homo sapiens 82-87 33757384-0 2021 Prolongation of allograft survival by artemisinin treatment is associated with blockade of OX40-OX40L. artemisinin 38-49 tumor necrosis factor receptor superfamily, member 4 Mus musculus 91-95 33757384-0 2021 Prolongation of allograft survival by artemisinin treatment is associated with blockade of OX40-OX40L. artemisinin 38-49 tumor necrosis factor (ligand) superfamily, member 4 Mus musculus 96-101 33977847-0 2021 Computational and experimental insights on the interaction of artemisinin, dihydroartemisinin and chloroquine with SARS-CoV-2 spike protein receptor-binding domain (RBD). artemisinin 62-73 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 126-131 33556535-0 2021 Artemisinin protects endothelial function and vasodilation from oxidative damage via activation of PI3K/Akt/eNOS pathway. artemisinin 0-11 AKT serine/threonine kinase 1 Homo sapiens 104-107 33556535-0 2021 Artemisinin protects endothelial function and vasodilation from oxidative damage via activation of PI3K/Akt/eNOS pathway. artemisinin 0-11 nitric oxide synthase 3 Homo sapiens 108-112 33453127-7 2021 Artemisinin bound MD2 showed much less collapse during the molecular dynamic simulations, which supports the increased stability of MD2 upon artemisinin binding. artemisinin 0-11 lymphocyte antigen 96 Mus musculus 132-135 33453127-7 2021 Artemisinin bound MD2 showed much less collapse during the molecular dynamic simulations, which supports the increased stability of MD2 upon artemisinin binding. artemisinin 141-152 lymphocyte antigen 96 Mus musculus 18-21 33453127-7 2021 Artemisinin bound MD2 showed much less collapse during the molecular dynamic simulations, which supports the increased stability of MD2 upon artemisinin binding. artemisinin 141-152 lymphocyte antigen 96 Mus musculus 132-135 33453127-8 2021 Flow cytometry analysis showed artemisinin inhibited LPS-induced TLR4 dimerization and endocytosis in microglial BV-2 cells. artemisinin 31-42 toll-like receptor 4 Mus musculus 65-69 33453127-9 2021 Therefore, artemisinin was found to inhibit the TLR4-JNK signaling axis and block LPS-induced pro-inflammatory factors nitric oxide, IL-1beta and TNF-alpha in BV-2 cells. artemisinin 11-22 toll-like receptor 4 Mus musculus 48-52 33453127-0 2021 Artemisinin inhibits TLR4 signaling by targeting co-receptor MD2 in microglial BV-2 cells and prevents lipopolysaccharideinduced blood brain barrier leakage in mice. artemisinin 0-11 toll-like receptor 4 Mus musculus 21-25 33453127-9 2021 Therefore, artemisinin was found to inhibit the TLR4-JNK signaling axis and block LPS-induced pro-inflammatory factors nitric oxide, IL-1beta and TNF-alpha in BV-2 cells. artemisinin 11-22 mitogen-activated protein kinase 8 Mus musculus 53-56 33453127-0 2021 Artemisinin inhibits TLR4 signaling by targeting co-receptor MD2 in microglial BV-2 cells and prevents lipopolysaccharideinduced blood brain barrier leakage in mice. artemisinin 0-11 lymphocyte antigen 96 Mus musculus 61-64 33453127-5 2021 In vitro protein intrinsic fluorescence titrations and saturation transfer difference (STD)-NMR showed the direct binding of artemisinin to TLR4 co-receptor MD2. artemisinin 125-136 toll-like receptor 4 Mus musculus 140-144 33453127-9 2021 Therefore, artemisinin was found to inhibit the TLR4-JNK signaling axis and block LPS-induced pro-inflammatory factors nitric oxide, IL-1beta and TNF-alpha in BV-2 cells. artemisinin 11-22 interleukin 1 alpha Mus musculus 133-141 33453127-5 2021 In vitro protein intrinsic fluorescence titrations and saturation transfer difference (STD)-NMR showed the direct binding of artemisinin to TLR4 co-receptor MD2. artemisinin 125-136 lymphocyte antigen 96 Mus musculus 157-160 33453127-6 2021 Cellular thermal shift assay (CETSA) showed that artemisinin binding increased MD2 stability, which implies that artemisinin directly binds to MD2 in the cellular context. artemisinin 49-60 lymphocyte antigen 96 Mus musculus 79-82 33453127-6 2021 Cellular thermal shift assay (CETSA) showed that artemisinin binding increased MD2 stability, which implies that artemisinin directly binds to MD2 in the cellular context. artemisinin 49-60 lymphocyte antigen 96 Mus musculus 143-146 33453127-9 2021 Therefore, artemisinin was found to inhibit the TLR4-JNK signaling axis and block LPS-induced pro-inflammatory factors nitric oxide, IL-1beta and TNF-alpha in BV-2 cells. artemisinin 11-22 tumor necrosis factor Mus musculus 146-155 33453127-6 2021 Cellular thermal shift assay (CETSA) showed that artemisinin binding increased MD2 stability, which implies that artemisinin directly binds to MD2 in the cellular context. artemisinin 113-124 lymphocyte antigen 96 Mus musculus 79-82 33453127-6 2021 Cellular thermal shift assay (CETSA) showed that artemisinin binding increased MD2 stability, which implies that artemisinin directly binds to MD2 in the cellular context. artemisinin 113-124 lymphocyte antigen 96 Mus musculus 143-146 33453127-10 2021 Furthermore, artemisinin restored LPS-induced decrease of junction proteins ZO-1, Occludin and Claudin-5 in primary brain microvessel endothelial cells, and attenuated LPS-induced blood brain barrier disruption in mice as assessed by Evans blue. artemisinin 13-24 tight junction protein 1 Mus musculus 76-80 33453127-7 2021 Artemisinin bound MD2 showed much less collapse during the molecular dynamic simulations, which supports the increased stability of MD2 upon artemisinin binding. artemisinin 0-11 lymphocyte antigen 96 Mus musculus 18-21 33453127-10 2021 Furthermore, artemisinin restored LPS-induced decrease of junction proteins ZO-1, Occludin and Claudin-5 in primary brain microvessel endothelial cells, and attenuated LPS-induced blood brain barrier disruption in mice as assessed by Evans blue. artemisinin 13-24 occludin Mus musculus 82-90 33453127-10 2021 Furthermore, artemisinin restored LPS-induced decrease of junction proteins ZO-1, Occludin and Claudin-5 in primary brain microvessel endothelial cells, and attenuated LPS-induced blood brain barrier disruption in mice as assessed by Evans blue. artemisinin 13-24 claudin 5 Mus musculus 95-104 33453127-11 2021 In all, this study unambiguously adds MD2 as a direct binding target of artemisinin in its anti-neuroinflammatory function. artemisinin 72-83 lymphocyte antigen 96 Mus musculus 38-41 33933588-7 2021 Immunofluorescence microscopy demonstrated the rescue of gephyrin and gamma2-GABAA-receptor protein levels in the brain of treated mice with both, artemisinin and artesunate, most efficiently with a low dose of artesunate. artemisinin 147-158 gephyrin Mus musculus 57-65 33875422-10 2021 Between one in three and one in six individuals harbored reduced activity alleles of CYP2A6, CYP2B6, CYP2D6 and CYP2C8 with important implications for artemisinin, chloroquine and amodiaquine therapy. artemisinin 151-162 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 85-91 33903109-2 2021 Single nucleotide polymorphisms (SNPs) in other molecular markers, such as ap2mu and ubp1, were associated with artemisinin resistance in rodent malaria and clinical failure in African malaria patients. artemisinin 112-123 upstream binding protein 1 Homo sapiens 85-89 33878252-0 2022 Artemisinin-treatment in pre-symptomatic APP-PS1 mice increases gephyrin phosphorylation at Ser270: a modification regulating postsynaptic GABAAR density. artemisinin 0-11 presenilin 1 Mus musculus 45-48 33878252-0 2022 Artemisinin-treatment in pre-symptomatic APP-PS1 mice increases gephyrin phosphorylation at Ser270: a modification regulating postsynaptic GABAAR density. artemisinin 0-11 gephyrin Mus musculus 64-72 33878252-5 2022 Here, we studied the effects of artemisinin on gephyrin in the brain of young APP-PS1 mice. artemisinin 32-43 gephyrin Mus musculus 47-55 33878252-10 2022 Thus, our results reveal that artemisinin modulates expression as well as phosphorylation of gephyrin at sites that might have important impact on GABAergic synapses in AD. artemisinin 30-41 gephyrin Mus musculus 93-101 33875422-10 2021 Between one in three and one in six individuals harbored reduced activity alleles of CYP2A6, CYP2B6, CYP2D6 and CYP2C8 with important implications for artemisinin, chloroquine and amodiaquine therapy. artemisinin 151-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 33875422-10 2021 Between one in three and one in six individuals harbored reduced activity alleles of CYP2A6, CYP2B6, CYP2D6 and CYP2C8 with important implications for artemisinin, chloroquine and amodiaquine therapy. artemisinin 151-162 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 101-107 33875422-10 2021 Between one in three and one in six individuals harbored reduced activity alleles of CYP2A6, CYP2B6, CYP2D6 and CYP2C8 with important implications for artemisinin, chloroquine and amodiaquine therapy. artemisinin 151-162 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 112-118 32747720-0 2021 The artemisinin analog SM934 alleviates dry eye disease in rodent models by regulating TLR4/NF-kappaB/NLRP3 signaling. artemisinin 4-15 toll-like receptor 4 Rattus norvegicus 87-91 33827592-0 2021 Molecular surveillance of drug resistance: Plasmodium falciparum artemisinin resistance single nucleotide polymorphisms in Kelch protein propeller (K13) domain from Southern Pakistan. artemisinin 65-76 kelch like family member 2 Homo sapiens 123-128 33827592-0 2021 Molecular surveillance of drug resistance: Plasmodium falciparum artemisinin resistance single nucleotide polymorphisms in Kelch protein propeller (K13) domain from Southern Pakistan. artemisinin 65-76 keratin 13 Homo sapiens 148-151 33827592-1 2021 BACKGROUND: K13 propeller (k13) polymorphism are useful molecular markers for tracking the emergence and spread of artemisinin resistance in Plasmodium falciparum. artemisinin 115-126 keratin 13 Homo sapiens 12-15 33827592-1 2021 BACKGROUND: K13 propeller (k13) polymorphism are useful molecular markers for tracking the emergence and spread of artemisinin resistance in Plasmodium falciparum. artemisinin 115-126 keratin 13 Homo sapiens 27-30 33827592-3 2021 The study describes single nucleotide polymorphisms in Kelch protein propeller domain of P. falciparum associated with artemisinin resistance from Southern Pakistan. artemisinin 119-130 kelch like family member 2 Homo sapiens 55-60 32747720-0 2021 The artemisinin analog SM934 alleviates dry eye disease in rodent models by regulating TLR4/NF-kappaB/NLRP3 signaling. artemisinin 4-15 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 92-101 32747720-0 2021 The artemisinin analog SM934 alleviates dry eye disease in rodent models by regulating TLR4/NF-kappaB/NLRP3 signaling. artemisinin 4-15 NLR family, pyrin domain containing 3 Rattus norvegicus 102-107 33460658-0 2021 Interaction of artemisinin protects the activity of antioxidant enzyme catalase: A biophysical study. artemisinin 15-26 catalase Bos taurus 71-79 33758353-7 2021 In BV2 microglial cells treated with LPS (100 ng/mL), pre-application of artemisinin (40 muMu) significantly reduced the production of proinflammatory cytokines (i.e., TNF-alpha, IL-6) and suppressed microglial migration. artemisinin 73-84 tumor necrosis factor Mus musculus 168-177 33758353-7 2021 In BV2 microglial cells treated with LPS (100 ng/mL), pre-application of artemisinin (40 muMu) significantly reduced the production of proinflammatory cytokines (i.e., TNF-alpha, IL-6) and suppressed microglial migration. artemisinin 73-84 interleukin 6 Mus musculus 179-183 33758353-8 2021 Furthermore, we revealed that artemisinin significantly suppressed the nuclear translocation of NF-kappaB and the expression of proinflammatory cytokines by activating the AMPKalpha1 pathway; knockdown of AMPKalpha1 markedly abolished the anti-inflammatory effects of artemisinin in BV2 microglial cells. artemisinin 30-41 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 96-105 33758353-8 2021 Furthermore, we revealed that artemisinin significantly suppressed the nuclear translocation of NF-kappaB and the expression of proinflammatory cytokines by activating the AMPKalpha1 pathway; knockdown of AMPKalpha1 markedly abolished the anti-inflammatory effects of artemisinin in BV2 microglial cells. artemisinin 30-41 protein kinase, AMP-activated, alpha 1 catalytic subunit Mus musculus 172-182 33758353-8 2021 Furthermore, we revealed that artemisinin significantly suppressed the nuclear translocation of NF-kappaB and the expression of proinflammatory cytokines by activating the AMPKalpha1 pathway; knockdown of AMPKalpha1 markedly abolished the anti-inflammatory effects of artemisinin in BV2 microglial cells. artemisinin 30-41 protein kinase, AMP-activated, alpha 1 catalytic subunit Mus musculus 205-215 33758353-8 2021 Furthermore, we revealed that artemisinin significantly suppressed the nuclear translocation of NF-kappaB and the expression of proinflammatory cytokines by activating the AMPKalpha1 pathway; knockdown of AMPKalpha1 markedly abolished the anti-inflammatory effects of artemisinin in BV2 microglial cells. artemisinin 268-279 protein kinase, AMP-activated, alpha 1 catalytic subunit Mus musculus 205-215 33508479-4 2021 In this review, antimalarial activity of the most active artemisinin derivatives modified at C-10/C-11/C-16/C-6 positions and synthetic peroxides (endoperoxides, 1,2,4-trioxolanes, 1,2,4-trioxanes, and 1,2,4,5-tetraoxanes) are systematically summarized. artemisinin 57-68 homeobox C10 Homo sapiens 93-97 33508479-4 2021 In this review, antimalarial activity of the most active artemisinin derivatives modified at C-10/C-11/C-16/C-6 positions and synthetic peroxides (endoperoxides, 1,2,4-trioxolanes, 1,2,4-trioxanes, and 1,2,4,5-tetraoxanes) are systematically summarized. artemisinin 57-68 RNA polymerase III subunit K Homo sapiens 98-102 33632304-0 2021 Artemisinin attenuated oxidative stress and apoptosis by inhibiting autophagy in MPP+-treated SH-SY5Y cells. artemisinin 0-11 M-phase phosphoprotein 6 Homo sapiens 81-84 33359205-0 2021 Polymorphisms in Plasmodium falciparum chloroquine resistance transporter (Pfcrt) and multidrug-resistant gene 1 (Pfmdr-1) in Nigerian children 10 years post-adoption of artemisinin-based combination treatments. artemisinin 170-181 plasmodium falciparum chloroquine resistance transporter (pfcrt) and multidrug-resistant gene 1 None 17-112 33599403-2 2021 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. artemisinin 43-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 33632304-4 2021 The purpose of this study is to explore the neuroprotective effect of ART on 1-methyl-4-phenyliodine iodide (MPP +)-treated SH-SY5Y cells and underlying mechanism. artemisinin 70-73 M-phase phosphoprotein 6 Homo sapiens 109-112 33632304-12 2021 ART could significantly increase the viability of SH-SY5Y cells treated with MPP+ and reduce oxidative stress damage and apoptosis. artemisinin 0-3 M-phase phosphoprotein 6 Homo sapiens 77-80 33620584-2 2021 Recently, artemisinin (ART) and its derivatives have been investigated as potential anticancer agents for the treatment of highly aggressive cancers via the induction of ferroptosis by iron-mediated cleavage of the endoperoxide bridge. artemisinin 10-21 artemin Homo sapiens 23-26 33483501-0 2021 Artemisinin-resistant K13 mutations rewire Plasmodium falciparum"s intra-erythrocytic metabolic program to enhance survival. artemisinin 0-11 keratin 13 Homo sapiens 22-25 33596950-5 2021 RESULTS: Among all the compounds screened, 1-aryltetrahydro-beta-carbolines 2 and 3 displayed significant anti-plasmodial activity against both the artemisinin-sensitive and artemisinin-resistant strain of Plasmodium falciparum. artemisinin 148-159 carbolines 2 and 3 None 65-83 33596950-5 2021 RESULTS: Among all the compounds screened, 1-aryltetrahydro-beta-carbolines 2 and 3 displayed significant anti-plasmodial activity against both the artemisinin-sensitive and artemisinin-resistant strain of Plasmodium falciparum. artemisinin 174-185 carbolines 2 and 3 None 65-83 33560385-3 2021 Screening of a targeted antineoplastic drug library revealed that B-cell lymphoma 2 (BCL2) inhibitors synergize with artemisinins, and validation assays confirmed that the selective BCL2 inhibitor, venetoclax (VEN), synergized with artemisinin analogs to inhibit growth and induce apoptotic cell death of multiple acute leukemia cell lines in vitro. artemisinin 117-128 BCL2 apoptosis regulator Homo sapiens 66-83 33560385-3 2021 Screening of a targeted antineoplastic drug library revealed that B-cell lymphoma 2 (BCL2) inhibitors synergize with artemisinins, and validation assays confirmed that the selective BCL2 inhibitor, venetoclax (VEN), synergized with artemisinin analogs to inhibit growth and induce apoptotic cell death of multiple acute leukemia cell lines in vitro. artemisinin 117-128 BCL2 apoptosis regulator Homo sapiens 85-89 33483501-1 2021 The emergence and spread of artemisinin resistance, driven by mutations in Plasmodium falciparum K13, has compromised antimalarial efficacy and threatens the global malaria elimination campaign. artemisinin 28-39 keratin 13 Homo sapiens 97-100 33483501-4 2021 K13-mediated artemisinin resistance in the Cambodian Cam3.II line was reversed by atovaquone, a mitochondrial electron transport chain inhibitor. artemisinin 13-24 keratin 13 Homo sapiens 0-3 33483501-5 2021 These results suggest that mitochondrial processes including damage sensing and anti-oxidant properties might augment the ability of mutant K13 to protect P. falciparum against artemisinin action by helping these parasites undergo temporary quiescence and accelerated growth recovery post drug elimination. artemisinin 177-188 keratin 13 Homo sapiens 140-143 31965935-7 2021 RESULTS: Artemisinin derivatives, artemisinin-derived dimers, hybrids and artemisinin-transferrin conjugates, could significantly improve anticancer activity, and their IC50 values are lower than those of reference molecules such as doxorubicin and paclitaxel. artemisinin 74-85 transferrin Homo sapiens 86-97 33481164-0 2021 Artemisinin analogue SM934 protects against lupus-associated antiphospholipid syndrome via activation of Nrf2 and its targets. artemisinin 0-11 nuclear factor, erythroid derived 2, like 2 Mus musculus 105-109 33441725-4 2021 Furthermore, we show that upon artemisinin exposure, genome-wide binding sites for PfGCN5 are increased and it is directly associated with the genes implicated in artemisinin resistance generation like BiP and TRiC chaperone. artemisinin 31-42 heat shock protein family A (Hsp70) member 5 Homo sapiens 202-205 33441725-4 2021 Furthermore, we show that upon artemisinin exposure, genome-wide binding sites for PfGCN5 are increased and it is directly associated with the genes implicated in artemisinin resistance generation like BiP and TRiC chaperone. artemisinin 31-42 MARVEL domain containing 2 Homo sapiens 210-214 33441725-4 2021 Furthermore, we show that upon artemisinin exposure, genome-wide binding sites for PfGCN5 are increased and it is directly associated with the genes implicated in artemisinin resistance generation like BiP and TRiC chaperone. artemisinin 163-174 heat shock protein family A (Hsp70) member 5 Homo sapiens 202-205 33441725-4 2021 Furthermore, we show that upon artemisinin exposure, genome-wide binding sites for PfGCN5 are increased and it is directly associated with the genes implicated in artemisinin resistance generation like BiP and TRiC chaperone. artemisinin 163-174 MARVEL domain containing 2 Homo sapiens 210-214 32939611-0 2020 Antinociception induced by artemisinin nanocapsule in a model of postoperative pain via spinal TLR4 inhibition. artemisinin 27-38 toll-like receptor 4 Mus musculus 95-99 32386317-10 2020 Finally, Artemisinin treatment in PC3 cells repressed E2F5 along with MMP-2/MMP-9 while triggering TFPI2 expression which alleviated PC3 aggressiveness possibly through inhibition of MMP activities. artemisinin 9-20 microseminoprotein, prostate associated S homeolog Xenopus laevis 34-37 32386317-10 2020 Finally, Artemisinin treatment in PC3 cells repressed E2F5 along with MMP-2/MMP-9 while triggering TFPI2 expression which alleviated PC3 aggressiveness possibly through inhibition of MMP activities. artemisinin 9-20 E2F transcription factor 5 Homo sapiens 54-58 32386317-10 2020 Finally, Artemisinin treatment in PC3 cells repressed E2F5 along with MMP-2/MMP-9 while triggering TFPI2 expression which alleviated PC3 aggressiveness possibly through inhibition of MMP activities. artemisinin 9-20 matrix metallopeptidase 2 Homo sapiens 70-75 32386317-10 2020 Finally, Artemisinin treatment in PC3 cells repressed E2F5 along with MMP-2/MMP-9 while triggering TFPI2 expression which alleviated PC3 aggressiveness possibly through inhibition of MMP activities. artemisinin 9-20 matrix metallopeptidase 9 Homo sapiens 76-81 32386317-10 2020 Finally, Artemisinin treatment in PC3 cells repressed E2F5 along with MMP-2/MMP-9 while triggering TFPI2 expression which alleviated PC3 aggressiveness possibly through inhibition of MMP activities. artemisinin 9-20 tissue factor pathway inhibitor 2 Homo sapiens 99-104 32386317-10 2020 Finally, Artemisinin treatment in PC3 cells repressed E2F5 along with MMP-2/MMP-9 while triggering TFPI2 expression which alleviated PC3 aggressiveness possibly through inhibition of MMP activities. artemisinin 9-20 microseminoprotein, prostate associated S homeolog Xenopus laevis 133-136 32386317-12 2020 This study also indicates a therapeutic potential of artemisinin, a natural compound which acts by correcting dysfunctional E2F5/TFPI2/MMP axis in PCa. artemisinin 53-64 E2F transcription factor 5 Homo sapiens 124-128 32386317-12 2020 This study also indicates a therapeutic potential of artemisinin, a natural compound which acts by correcting dysfunctional E2F5/TFPI2/MMP axis in PCa. artemisinin 53-64 tissue factor pathway inhibitor 2 Homo sapiens 129-134 33078701-11 2020 Purported artemisinin and lumefantrine drug resistance SNPs in atp6, 3D7_1451200, and mdr1 were detected but did not correlate with parasite recurrence, nor did day 7 lumefantrine concentrations. artemisinin 10-21 mitochondrially encoded ATP synthase 6 Homo sapiens 63-67 33078701-11 2020 Purported artemisinin and lumefantrine drug resistance SNPs in atp6, 3D7_1451200, and mdr1 were detected but did not correlate with parasite recurrence, nor did day 7 lumefantrine concentrations. artemisinin 10-21 ATP binding cassette subfamily B member 1 Homo sapiens 86-90 32861782-1 2020 The present study aims to evaluate the inhibitory effects of artesunate (a semi-synthetic derivative of artemisinin) on HSP70 and Bcl-2 expression in two breast cancer cell lines, 4T1 and MCF-7. artemisinin 104-115 heat shock protein family A (Hsp70) member 4 Homo sapiens 120-125 32729005-8 2020 The artemisinin-treated myocardial I/R rats demonstrated less severe myocardial I/R injury (smaller infarct size and lower CK-MB, LDH), significant inhibition of cardiac autophagy (decreased LC3II/I and increased p62), improved mitochondrial electron transport chain activity, concomitant with decreased activation of NLRP3 inflammasome (decreased NLRP3, ASC, cleaved caspase-1, IL-1beta). artemisinin 4-15 microtubule-associated protein 1 light chain 3 alpha Rattus norvegicus 191-198 32861782-1 2020 The present study aims to evaluate the inhibitory effects of artesunate (a semi-synthetic derivative of artemisinin) on HSP70 and Bcl-2 expression in two breast cancer cell lines, 4T1 and MCF-7. artemisinin 104-115 BCL2 apoptosis regulator Homo sapiens 130-135 33381036-5 2020 Artemisinin treatments not only attenuate insulin resistance and restore islet ss-cell function in T2DM but also have potential therapeutic effects on diabetic complications, including diabetic kidney disease, cognitive impairment, diabetic retinopathy, and diabetic cardiovascular disease. artemisinin 0-11 insulin Homo sapiens 42-49 33173001-5 2020 Mutant P. berghei K13 parasites also displayed delayed clearance in vivo upon treatment with artesunate and achieved faster recrudescence upon treatment with artemisinin. artemisinin 158-169 keratin 13 Homo sapiens 18-21 33173001-7 2020 Furthermore, a Plasmodium-selective proteasome inhibitor strongly synergized dihydroartemisinin action in these P. berghei K13 mutant lines, providing further evidence that the proteasome can be targeted to overcome artemisinin resistance. artemisinin 84-95 keratin 13 Homo sapiens 123-126 33173001-8 2020 Taken together, our findings provide clear experimental evidence for the involvement of K13 polymorphisms in mediating susceptibility to artemisinins in vitro and, most importantly, under in vivo conditions.IMPORTANCE Recent successes in malaria control have been seriously threatened by the emergence of Plasmodium falciparum parasite resistance to the frontline artemisinin drugs in Southeast Asia. artemisinin 137-148 keratin 13 Homo sapiens 88-91 33173001-9 2020 P. falciparum artemisinin resistance is associated with mutations in the parasite K13 protein, which associates with a delay in the time required to clear the parasites upon drug treatment. artemisinin 14-25 keratin 13 Homo sapiens 82-85 33173001-10 2020 Gene editing technologies have been used to validate the role of several candidate K13 mutations in mediating P. falciparum artemisinin resistance in vitro under laboratory conditions. artemisinin 124-135 keratin 13 Homo sapiens 83-86 33173001-12 2020 Here, we have used CRISPR/Cas9 gene editing to introduce K13 mutations associated with artemisinin resistance into the related rodent-infecting parasite, Plasmodium berghei Phenotyping of these P. berghei K13 mutant parasites provides evidence of their role in mediating artemisinin resistance in vivo, which supports in vitro artemisinin resistance observations. artemisinin 87-98 keratin 13 Homo sapiens 57-60 32868072-7 2020 We demonstrate that TGF-beta mediated Smad2 linker region phosphorylation and GAG chain elongation was attenuated by artemisinin; however, we observed no effect on VSMC proliferation. artemisinin 117-128 transforming growth factor alpha Homo sapiens 20-28 32868072-7 2020 We demonstrate that TGF-beta mediated Smad2 linker region phosphorylation and GAG chain elongation was attenuated by artemisinin; however, we observed no effect on VSMC proliferation. artemisinin 117-128 SMAD family member 2 Homo sapiens 38-43 33142414-8 2020 All the results proved the effectiveness of the hybridization approach to develop novel artemisinin-sulfonamide compounds targeting CA IX for cancer treatment. artemisinin 88-99 carbonic anhydrase 9 Homo sapiens 132-137 33239368-0 2020 Elucidation of DNA Repair Function of PfBlm and Potentiation of Artemisinin Action by a Small-Molecule Inhibitor of RecQ Helicase. artemisinin 64-75 recq helicase None 116-129 33150340-2 2020 Dihydroartemisinin (DHA), a main active metabolite of anti-malarial drug artemisinin (ART), inhibits cancer cell invasion and migration by decreasing the translationally controlled tumor protein (TCTP), as reported in a few literature studies. artemisinin 7-18 tumor protein, translationally-controlled 1 Homo sapiens 154-194 33150340-2 2020 Dihydroartemisinin (DHA), a main active metabolite of anti-malarial drug artemisinin (ART), inhibits cancer cell invasion and migration by decreasing the translationally controlled tumor protein (TCTP), as reported in a few literature studies. artemisinin 7-18 tumor protein, translationally-controlled 1 Homo sapiens 196-200 33150340-2 2020 Dihydroartemisinin (DHA), a main active metabolite of anti-malarial drug artemisinin (ART), inhibits cancer cell invasion and migration by decreasing the translationally controlled tumor protein (TCTP), as reported in a few literature studies. artemisinin 86-89 tumor protein, translationally-controlled 1 Homo sapiens 154-194 33150340-2 2020 Dihydroartemisinin (DHA), a main active metabolite of anti-malarial drug artemisinin (ART), inhibits cancer cell invasion and migration by decreasing the translationally controlled tumor protein (TCTP), as reported in a few literature studies. artemisinin 86-89 tumor protein, translationally-controlled 1 Homo sapiens 196-200 33173001-0 2020 Plasmodium berghei K13 Mutations Mediate In Vivo Artemisinin Resistance That Is Reversed by Proteasome Inhibition. artemisinin 49-60 keratin 13 Homo sapiens 19-22 33173001-2 2020 Artemisinin resistance is primarily driven by mutations in the P. falciparum K13 protein, which enhance survival of early ring-stage parasites treated with the artemisinin active metabolite dihydroartemisinin in vitro and associate with delayed parasite clearance in vivo However, association of K13 mutations with in vivo artemisinin resistance has been problematic due to the absence of a tractable model. artemisinin 0-11 keratin 13 Homo sapiens 77-80 33173001-2 2020 Artemisinin resistance is primarily driven by mutations in the P. falciparum K13 protein, which enhance survival of early ring-stage parasites treated with the artemisinin active metabolite dihydroartemisinin in vitro and associate with delayed parasite clearance in vivo However, association of K13 mutations with in vivo artemisinin resistance has been problematic due to the absence of a tractable model. artemisinin 0-11 keratin 13 Homo sapiens 296-299 33173001-2 2020 Artemisinin resistance is primarily driven by mutations in the P. falciparum K13 protein, which enhance survival of early ring-stage parasites treated with the artemisinin active metabolite dihydroartemisinin in vitro and associate with delayed parasite clearance in vivo However, association of K13 mutations with in vivo artemisinin resistance has been problematic due to the absence of a tractable model. artemisinin 160-171 keratin 13 Homo sapiens 77-80 33173001-2 2020 Artemisinin resistance is primarily driven by mutations in the P. falciparum K13 protein, which enhance survival of early ring-stage parasites treated with the artemisinin active metabolite dihydroartemisinin in vitro and associate with delayed parasite clearance in vivo However, association of K13 mutations with in vivo artemisinin resistance has been problematic due to the absence of a tractable model. artemisinin 197-208 keratin 13 Homo sapiens 77-80 33173001-3 2020 Herein, we have employed CRISPR/Cas9 genome editing to engineer selected orthologous P. falciparum K13 mutations into the K13 gene of an artemisinin-sensitive Plasmodium berghei rodent model of malaria. artemisinin 137-148 keratin 13 Homo sapiens 99-102 33173001-3 2020 Herein, we have employed CRISPR/Cas9 genome editing to engineer selected orthologous P. falciparum K13 mutations into the K13 gene of an artemisinin-sensitive Plasmodium berghei rodent model of malaria. artemisinin 137-148 keratin 13 Homo sapiens 122-125 32729005-8 2020 The artemisinin-treated myocardial I/R rats demonstrated less severe myocardial I/R injury (smaller infarct size and lower CK-MB, LDH), significant inhibition of cardiac autophagy (decreased LC3II/I and increased p62), improved mitochondrial electron transport chain activity, concomitant with decreased activation of NLRP3 inflammasome (decreased NLRP3, ASC, cleaved caspase-1, IL-1beta). artemisinin 4-15 KH RNA binding domain containing, signal transduction associated 1 Rattus norvegicus 213-216 32729005-8 2020 The artemisinin-treated myocardial I/R rats demonstrated less severe myocardial I/R injury (smaller infarct size and lower CK-MB, LDH), significant inhibition of cardiac autophagy (decreased LC3II/I and increased p62), improved mitochondrial electron transport chain activity, concomitant with decreased activation of NLRP3 inflammasome (decreased NLRP3, ASC, cleaved caspase-1, IL-1beta). artemisinin 4-15 NLR family, pyrin domain containing 3 Rattus norvegicus 318-323 32729005-8 2020 The artemisinin-treated myocardial I/R rats demonstrated less severe myocardial I/R injury (smaller infarct size and lower CK-MB, LDH), significant inhibition of cardiac autophagy (decreased LC3II/I and increased p62), improved mitochondrial electron transport chain activity, concomitant with decreased activation of NLRP3 inflammasome (decreased NLRP3, ASC, cleaved caspase-1, IL-1beta). artemisinin 4-15 NLR family, pyrin domain containing 3 Rattus norvegicus 348-353 32729005-8 2020 The artemisinin-treated myocardial I/R rats demonstrated less severe myocardial I/R injury (smaller infarct size and lower CK-MB, LDH), significant inhibition of cardiac autophagy (decreased LC3II/I and increased p62), improved mitochondrial electron transport chain activity, concomitant with decreased activation of NLRP3 inflammasome (decreased NLRP3, ASC, cleaved caspase-1, IL-1beta). artemisinin 4-15 PYD and CARD domain containing Rattus norvegicus 355-358 32729005-8 2020 The artemisinin-treated myocardial I/R rats demonstrated less severe myocardial I/R injury (smaller infarct size and lower CK-MB, LDH), significant inhibition of cardiac autophagy (decreased LC3II/I and increased p62), improved mitochondrial electron transport chain activity, concomitant with decreased activation of NLRP3 inflammasome (decreased NLRP3, ASC, cleaved caspase-1, IL-1beta). artemisinin 4-15 caspase 1 Rattus norvegicus 368-377 32729005-8 2020 The artemisinin-treated myocardial I/R rats demonstrated less severe myocardial I/R injury (smaller infarct size and lower CK-MB, LDH), significant inhibition of cardiac autophagy (decreased LC3II/I and increased p62), improved mitochondrial electron transport chain activity, concomitant with decreased activation of NLRP3 inflammasome (decreased NLRP3, ASC, cleaved caspase-1, IL-1beta). artemisinin 4-15 interleukin 1 alpha Rattus norvegicus 379-387 32729005-9 2020 In conclusion, our findings further confirmed that activation of the NLRP3 inflammasome pathway is involved in myocardial I/R injury, whereas artemisinin preconditioning could effectively protect against myocardial I/R injury through suppression of NLRP3 inflammasome activation. artemisinin 142-153 NLR family, pyrin domain containing 3 Rattus norvegicus 249-254 32729005-10 2020 Therefore, the NLRP3 inflammasome might serve as a promising therapeutic target providing new mechanisms for understanding the effect of artemisinin during the evolution of myocardial infarction. artemisinin 137-148 NLR family, pyrin domain containing 3 Rattus norvegicus 15-20 32747827-2 2020 Here we genotyped the P. falciparum K13 (Pfkelch13) propeller domain, mutations in which can mediate artemisinin resistance5,6, in pretreatment samples collected from recent dihydroarteminisin-piperaquine and artemether-lumefantrine efficacy trials in Rwanda7. artemisinin 101-112 keratin 13 Homo sapiens 36-39 32855235-2 2020 Using a biotin-labeled artemisinin we identified the intermediate filament protein vimentin as an artemisinin target, validated by detailed biochemical and biological assays. artemisinin 23-34 vimentin Homo sapiens 83-91 32855235-2 2020 Using a biotin-labeled artemisinin we identified the intermediate filament protein vimentin as an artemisinin target, validated by detailed biochemical and biological assays. artemisinin 98-109 vimentin Homo sapiens 83-91 32855235-8 2020 Binding of artesunate, an artemisinin monomer, to vimentin prevents virus-induced vimentin degradation, decreasing vimentin phosphorylation at Ser55 and Ser83 and resisting calpain digestion. artemisinin 26-37 vimentin Homo sapiens 50-58 32855235-8 2020 Binding of artesunate, an artemisinin monomer, to vimentin prevents virus-induced vimentin degradation, decreasing vimentin phosphorylation at Ser55 and Ser83 and resisting calpain digestion. artemisinin 26-37 vimentin Homo sapiens 82-90 32855235-8 2020 Binding of artesunate, an artemisinin monomer, to vimentin prevents virus-induced vimentin degradation, decreasing vimentin phosphorylation at Ser55 and Ser83 and resisting calpain digestion. artemisinin 26-37 vimentin Homo sapiens 82-90 32535079-0 2020 Artemisinin attenuates early renal damage on diabetic nephropathy rats through suppressing TGF-beta1 regulator and activating the Nrf2 signaling pathway. artemisinin 0-11 transforming growth factor, beta 1 Rattus norvegicus 91-100 32899699-2 2020 It has been reported that artemisinin derivatives can overcome resistance to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in liver and cervical cancer cells. artemisinin 26-37 TNF superfamily member 10 Homo sapiens 77-132 32899699-2 2020 It has been reported that artemisinin derivatives can overcome resistance to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in liver and cervical cancer cells. artemisinin 26-37 TNF superfamily member 10 Homo sapiens 134-139 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 32899699-0 2020 Artemisinin Derivatives Stimulate DR5-Specific TRAIL-Induced Apoptosis by Regulating Wildtype P53. artemisinin 0-11 TNF superfamily member 10 Homo sapiens 47-52 32535079-0 2020 Artemisinin attenuates early renal damage on diabetic nephropathy rats through suppressing TGF-beta1 regulator and activating the Nrf2 signaling pathway. artemisinin 0-11 NFE2 like bZIP transcription factor 2 Rattus norvegicus 130-134 32118504-3 2020 The results showed that bisdemethoxycurcumin, glycyrrhetnic acid, rotenone, artemisinin, dihydroartemisinin, ligustilide and matrine strongly induced the mRNA levels of PXR. artemisinin 76-87 nuclear receptor subfamily 1 group I member 2 Homo sapiens 169-172 32598944-0 2020 Artemisinin protects motoneurons against axotomy-induced apoptosis through activation of the PKA-Akt signaling pathway and promotes Neural stem/progenitor cells differentiation into NeuN+ neurons. artemisinin 0-11 AKT serine/threonine kinase 1 Homo sapiens 97-100 32598944-0 2020 Artemisinin protects motoneurons against axotomy-induced apoptosis through activation of the PKA-Akt signaling pathway and promotes Neural stem/progenitor cells differentiation into NeuN+ neurons. artemisinin 0-11 RNA binding fox-1 homolog 3 Homo sapiens 182-186 32598944-6 2020 Additionally, artemisinin inhibited the molecular signals of apoptosis, activated signaling pathways related to cell survival and induced NSCPs differentiation into NeuN-positive neurons. artemisinin 14-25 RNA binding fox-1 homolog 3 Homo sapiens 165-169 32598944-7 2020 Further validation of the involved key signaling molecules, using an in vitro model of hydrogen peroxide-induced neurotoxicity, revealed that both the inhibition of PKA signaling pathway or the silencing of Akt reversed the neuroprotective action of artemisinin on motoneurons. artemisinin 250-261 AKT serine/threonine kinase 1 Homo sapiens 207-210 32598944-8 2020 Our results indicate that artemisinin provides neuroprotection against axotomy and hydrogen peroxide-induced neurotoxicity, an effect that might be mediated by the PKA-Akt signaling pathway. artemisinin 26-37 AKT serine/threonine kinase 1 Homo sapiens 168-171 32118504-4 2020 Additionally, the up-regulation of CYP3A30 gene ran parallel with PXR gene after the treatment of demethoxycurcumin, glycyrrhetnic acid, artemisinin, matrine, baicalein, schisantherin A, ligustilide, and dihydroartemisinin. artemisinin 137-148 nuclear receptor subfamily 1 group I member 2 Homo sapiens 66-69 32696720-0 2021 Antimalarial-agent artemisinin and derivatives portray more potent binding to Lys353 and Lys31-binding hotspots of SARS-CoV-2 spike protein than hydroxychloroquine: potential repurposing of artenimol for COVID-19. artemisinin 19-30 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 126-131 32657048-0 2020 Combination treatment with artemisinin and oxaliplatin inhibits tumorigenesis in esophageal cancer EC109 cell through Wnt/beta-catenin signaling pathway. artemisinin 27-38 catenin beta 1 Homo sapiens 122-134 32657048-18 2020 WHAT THIS STUDY ADDS: Combination treatment with artemisinin and oxaliplatin inhibits tumorigenesis in esophageal cancer EC109 cells through the Wnt/beta-catenin signaling pathway. artemisinin 49-60 catenin beta 1 Homo sapiens 149-161 32765947-0 2020 Artemisinin Improved Neuronal Functions in Alzheimer"s Disease Animal Model 3xtg Mice and Neuronal Cells via Stimulating the ERK/CREB Signaling Pathway. artemisinin 0-11 mitogen-activated protein kinase 1 Mus musculus 125-128 32765947-0 2020 Artemisinin Improved Neuronal Functions in Alzheimer"s Disease Animal Model 3xtg Mice and Neuronal Cells via Stimulating the ERK/CREB Signaling Pathway. artemisinin 0-11 cAMP responsive element binding protein 1 Mus musculus 129-133 32765947-6 2020 Western blot assay showed that artemisinin stimulated the activation of ERK/CREB signaling pathway. artemisinin 31-42 mitogen-activated protein kinase 1 Mus musculus 72-75 32765947-6 2020 Western blot assay showed that artemisinin stimulated the activation of ERK/CREB signaling pathway. artemisinin 31-42 cAMP responsive element binding protein 1 Mus musculus 76-80 32765947-8 2020 Artemisinin also stimulated the phosphorylation of ERK1/2 and CREB in SH-SY5Y cells in time and concentration-dependent manner. artemisinin 0-11 mitogen-activated protein kinase 3 Homo sapiens 51-57 32765947-8 2020 Artemisinin also stimulated the phosphorylation of ERK1/2 and CREB in SH-SY5Y cells in time and concentration-dependent manner. artemisinin 0-11 cAMP responsive element binding protein 1 Homo sapiens 62-66 32765947-9 2020 Inhibition of ERK/CREB pathway attenuated the protective effect of artemisinin. artemisinin 67-78 mitogen-activated protein kinase 1 Homo sapiens 14-17 32765947-9 2020 Inhibition of ERK/CREB pathway attenuated the protective effect of artemisinin. artemisinin 67-78 cAMP responsive element binding protein 1 Homo sapiens 18-22 32765947-10 2020 These data put together suggested that artemisinin has the potential to protect neuronal cells in vitro as well as in vivo animal model 3xTg mice via, at least in part, the activation of the ERK/CREB pathway. artemisinin 39-50 mitogen-activated protein kinase 1 Mus musculus 191-194 32765947-10 2020 These data put together suggested that artemisinin has the potential to protect neuronal cells in vitro as well as in vivo animal model 3xTg mice via, at least in part, the activation of the ERK/CREB pathway. artemisinin 39-50 cAMP responsive element binding protein 1 Mus musculus 195-199 32696720-5 2021 Artesunate, artemisinin and artenimol, showed two mode of interactions with Lys353 and Lys31 binding hotspots of the Spike protein. artemisinin 12-23 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 117-122 32612358-1 2020 Objective: Artemisinin (ART) is a natural anti-malarial sesquiterpene lactone which has the ability to treat and activate the CLRN1 pathway to play a pivotal role in hearing loss and hair cell function. artemisinin 11-22 clarin 1 Homo sapiens 126-131 32653033-13 2020 Also, high prevalence of dhfr and dhps mutant alleles occurred in the study areas in Lagos, Nigeria five to eight years after the introduction of artemisinin combination therapy underscores the need for continuous monitoring. artemisinin 146-157 dihydrofolate reductase Homo sapiens 25-29 32653033-13 2020 Also, high prevalence of dhfr and dhps mutant alleles occurred in the study areas in Lagos, Nigeria five to eight years after the introduction of artemisinin combination therapy underscores the need for continuous monitoring. artemisinin 146-157 deoxyhypusine synthase Homo sapiens 34-38 32799228-0 2020 [In vitro and in silico Determination of the Interaction of Artemisinin with Human Serum Albumin]. artemisinin 60-71 albumin Mus musculus 89-96 32799228-3 2020 The interaction of artemisinin with human serum albumin was studied both in vitro and in silico, and compared with dexamethasone. artemisinin 19-30 albumin Mus musculus 42-55 32799228-4 2020 The quenching of the fluorescence emission of human serum albumin with artemisinin at different temperatures proceeded according to a single mechanism and indicated the static nature, which is similar to the effect of dexamethasone. artemisinin 71-82 albumin Mus musculus 52-65 32799228-5 2020 Artemisinin and dexamethasone interact with Drug site I on human serum albumin. artemisinin 0-11 albumin Mus musculus 65-78 32799228-10 2020 Therefore, we assume that one of the main transporters of artemisinin is human serum albumin. artemisinin 58-69 albumin Mus musculus 79-92 32799228-11 2020 Moreover, the interaction parameters of artemisinin with human serum albumin coincide with those of dexamethasone. artemisinin 40-51 albumin Mus musculus 63-76 32360483-3 2020 An increasing number of studies report that artemisinin can impact the fibrotic process through various ways, such as TGF-beta, MAPK, Wnt/beta-catenin, PI3K/AKT/mTRO, FRX and Notch signaling pathways, as well as regulation of BMP-7 and cell autophagy. artemisinin 44-55 transforming growth factor alpha Homo sapiens 118-126 32360483-3 2020 An increasing number of studies report that artemisinin can impact the fibrotic process through various ways, such as TGF-beta, MAPK, Wnt/beta-catenin, PI3K/AKT/mTRO, FRX and Notch signaling pathways, as well as regulation of BMP-7 and cell autophagy. artemisinin 44-55 bone morphogenetic protein 7 Homo sapiens 226-231 32304346-0 2020 Artemisinin-Derivative-NHC-gold(I)-Hybrid with enhanced cytotoxic activity through inhibiting NRF2 transcriptional activity. artemisinin 0-11 NFE2 like bZIP transcription factor 2 Homo sapiens 94-98 32612358-1 2020 Objective: Artemisinin (ART) is a natural anti-malarial sesquiterpene lactone which has the ability to treat and activate the CLRN1 pathway to play a pivotal role in hearing loss and hair cell function. artemisinin 24-27 clarin 1 Homo sapiens 126-131 31987792-3 2020 We found that artemisinin and clinical artemisinin derivatives are able to compensate for a mutation in the yeast Bcs1 protein, a key chaperon involved in biogenesis of the mitochondrial respiratory complex III. artemisinin 14-25 bifunctional AAA family ATPase chaperone/translocase BCS1 Saccharomyces cerevisiae S288C 114-118 32532194-10 2021 Treatment with Artemisinin (50 mg/kg) reduced the levels of PAP, LDH, prostate weight and prostatic index to a significant extent and restored the histoarchitectural features of the cells. artemisinin 15-26 acid phosphatase 3 Rattus norvegicus 60-63 31987792-3 2020 We found that artemisinin and clinical artemisinin derivatives are able to compensate for a mutation in the yeast Bcs1 protein, a key chaperon involved in biogenesis of the mitochondrial respiratory complex III. artemisinin 39-50 bifunctional AAA family ATPase chaperone/translocase BCS1 Saccharomyces cerevisiae S288C 114-118 32103124-0 2020 Detection of mutations associated with artemisinin resistance at k13-propeller gene and a near complete return of chloroquine susceptible falciparum malaria in Southeast of Tanzania. artemisinin 39-50 keratin 13 Homo sapiens 65-68 32862573-4 2020 Apoptotic effects of artemisinin were finally confirmed by western blot assay by analysing its effects on Bcl-2 and Bax protein expressions. artemisinin 21-32 bcl-2 None 106-111 32862573-4 2020 Apoptotic effects of artemisinin were finally confirmed by western blot assay by analysing its effects on Bcl-2 and Bax protein expressions. artemisinin 21-32 bax None 116-119 32862573-11 2020 The molecule also targeted G2/M phase cell cycle along with targeting some key cell cycle related proteins including cyclin-B1, cyclin D1 and cyclin E. CONCLUSION: The results show that artemisinin showed strong anticancer effects in MDA-MB-231 cisplatin-resistant cancer cells by triggering apoptosis and autophagy and G2/M phase arrest. artemisinin 186-197 cyclin-b1 None 117-126 32862573-11 2020 The molecule also targeted G2/M phase cell cycle along with targeting some key cell cycle related proteins including cyclin-B1, cyclin D1 and cyclin E. CONCLUSION: The results show that artemisinin showed strong anticancer effects in MDA-MB-231 cisplatin-resistant cancer cells by triggering apoptosis and autophagy and G2/M phase arrest. artemisinin 186-197 cyclin d1 None 128-137 32105722-1 2020 OBJECTIVE: The dimeric artesunate phospholipid conjugate (Di-ART-GPC) is a novel amphipathic artemisinin derivative, which can be assembled into liposomes. artemisinin 93-104 glycophorin C Rattus norvegicus 65-68 32435422-0 2020 Artemisinin Derivatives with Antimelanoma Activity Show Inhibitory Effect against Human DNA Topoisomerase 1. artemisinin 0-11 DNA topoisomerase I Homo sapiens 88-107 32152546-5 2020 This included dependencies involving the transporters SLC11A2/SLC16A1 for artemisinin derivatives and SLC35A2/SLC38A5 for cisplatin. artemisinin 74-85 solute carrier family 11 member 2 Homo sapiens 54-61 32152546-5 2020 This included dependencies involving the transporters SLC11A2/SLC16A1 for artemisinin derivatives and SLC35A2/SLC38A5 for cisplatin. artemisinin 74-85 solute carrier family 16 member 1 Homo sapiens 62-69 31945670-10 2020 The result of fluorescence quantitative section showed that artemisinin could down-regulate the expression of notch signaling related factors notch1, Dll4 and Jagged1, and 200 mg/kg dose group had the most significant effect. artemisinin 60-71 notch 1 Mus musculus 142-148 31945670-10 2020 The result of fluorescence quantitative section showed that artemisinin could down-regulate the expression of notch signaling related factors notch1, Dll4 and Jagged1, and 200 mg/kg dose group had the most significant effect. artemisinin 60-71 delta like canonical Notch ligand 4 Mus musculus 150-154 31945670-10 2020 The result of fluorescence quantitative section showed that artemisinin could down-regulate the expression of notch signaling related factors notch1, Dll4 and Jagged1, and 200 mg/kg dose group had the most significant effect. artemisinin 60-71 jagged 1 Mus musculus 159-166 32258150-0 2020 Sequence Analysis of the K13-Propeller Gene in Artemisinin Challenging Plasmodium falciparum Isolates from Malaria Endemic Areas of Odisha, India: A Molecular Surveillance Study. artemisinin 47-58 keratin 13 Homo sapiens 25-28 32143118-8 2020 ARS compounds selectively inhibit osteoclast differentiation by downregulation of pathways involved in receptor activator of nuclear factor kappa-B ligand (RANKL) -induced osteoclastogenesis, and have no effect on osteogenic differentiation of osteoblasts. artemisinin 0-3 TNF superfamily member 11 Homo sapiens 103-154 32143118-8 2020 ARS compounds selectively inhibit osteoclast differentiation by downregulation of pathways involved in receptor activator of nuclear factor kappa-B ligand (RANKL) -induced osteoclastogenesis, and have no effect on osteogenic differentiation of osteoblasts. artemisinin 0-3 TNF superfamily member 11 Homo sapiens 156-161 32310999-0 2020 Insights into the intracellular localization, protein associations and artemisinin resistance properties of Plasmodium falciparum K13. artemisinin 71-82 keratin 13 Homo sapiens 130-133 32310999-9 2020 These data help define the biological properties of K13 and its role in mediating P. falciparum resistance to ART treatment. artemisinin 110-113 keratin 13 Homo sapiens 52-55 32103124-6 2020 About 422 samples were successful sequenced for K13 gene (marker for artemisinin resistance), the wild type (WT) was found in 391 samples (92.7%) whereby 31 samples (7.3%) had mutations in K13 gene. artemisinin 69-80 keratin 13 Homo sapiens 48-51 31284865-0 2020 In Silico Screening Reveals Histone Deacetylase 7 and ERK1/2 as Potential Targets for Artemisinin Dimer and Artemisinin Dimer Hemisuccinate. artemisinin 86-97 histone deacetylase 7 Homo sapiens 28-49 32098816-1 2020 The efficacy of current antimalarial drugs is threatened by reduced susceptibility of Plasmodium falciparum to artemisinin, associated with mutations in pfkelch13 Another gene with variants known to modulate the response to artemisinin encodes the mu subunit of the AP-2 adaptin trafficking complex. artemisinin 111-122 transcription factor AP-2 alpha Homo sapiens 266-270 32098816-1 2020 The efficacy of current antimalarial drugs is threatened by reduced susceptibility of Plasmodium falciparum to artemisinin, associated with mutations in pfkelch13 Another gene with variants known to modulate the response to artemisinin encodes the mu subunit of the AP-2 adaptin trafficking complex. artemisinin 224-235 transcription factor AP-2 alpha Homo sapiens 266-270 31791643-4 2020 Given the high antitumor activity of artemisinin dimers in comparison to their monomers, we report here the synthesis of simple 1,2,3-triazole conjugated 1,2,4-trioxanes and their potential antitumor activity by studying their inhibitory effect on osteopontin (OPN) expression in MDA-MB-435 breast cancer cells. artemisinin 37-48 secreted phosphoprotein 1 Homo sapiens 248-259 31791643-4 2020 Given the high antitumor activity of artemisinin dimers in comparison to their monomers, we report here the synthesis of simple 1,2,3-triazole conjugated 1,2,4-trioxanes and their potential antitumor activity by studying their inhibitory effect on osteopontin (OPN) expression in MDA-MB-435 breast cancer cells. artemisinin 37-48 secreted phosphoprotein 1 Homo sapiens 261-264 31284865-0 2020 In Silico Screening Reveals Histone Deacetylase 7 and ERK1/2 as Potential Targets for Artemisinin Dimer and Artemisinin Dimer Hemisuccinate. artemisinin 86-97 mitogen-activated protein kinase 3 Homo sapiens 54-60 31284865-0 2020 In Silico Screening Reveals Histone Deacetylase 7 and ERK1/2 as Potential Targets for Artemisinin Dimer and Artemisinin Dimer Hemisuccinate. artemisinin 108-119 histone deacetylase 7 Homo sapiens 28-49 31284865-0 2020 In Silico Screening Reveals Histone Deacetylase 7 and ERK1/2 as Potential Targets for Artemisinin Dimer and Artemisinin Dimer Hemisuccinate. artemisinin 108-119 mitogen-activated protein kinase 3 Homo sapiens 54-60 31358588-3 2019 Artemisinin resistance has been associated in Southeast Asia with multiple nonsynonymous single nucleotide polymorphisms (NS-SNPs) in the propeller domain of the gene encoding the Plasmodium falciparum K13 protein (K13PD). artemisinin 0-11 keratin 13 Homo sapiens 202-205 31636063-2 2019 Mutations in two other genes, ubp1 and ap2mu, are associated with artemisinin resistance in rodent malaria and with clinical failure of combination therapy in African malaria patients. artemisinin 66-77 upstream binding protein 1 Homo sapiens 30-34 31636063-8 2019 Therefore, variants of the AP2 adaptor complex mu-subunit and of the ubiquitin hydrolase UBP1 reduce in vitro artemisinin susceptibility at the early ring stage in P. falciparum These findings confirm the existence of multiple pathways to perturbation of either the mode of action of artemisinin, the parasite"s adaptive mechanisms of resistance, or both. artemisinin 110-121 upstream binding protein 1 Homo sapiens 89-93 31636063-8 2019 Therefore, variants of the AP2 adaptor complex mu-subunit and of the ubiquitin hydrolase UBP1 reduce in vitro artemisinin susceptibility at the early ring stage in P. falciparum These findings confirm the existence of multiple pathways to perturbation of either the mode of action of artemisinin, the parasite"s adaptive mechanisms of resistance, or both. artemisinin 284-295 upstream binding protein 1 Homo sapiens 89-93 31604109-2 2019 MAIN METHODS: Eight-week-old male ApoE-/- mice were treated with ART for eight weeks. artemisinin 65-68 apolipoprotein E Mus musculus 34-38 31604109-5 2019 KEY FINDINGS: Artemisinin treatment significantly reduced plaque area in the ApoE-/- mice and increased the expression of contractile phenotypic markers. artemisinin 14-25 apolipoprotein E Mus musculus 77-81 31619243-3 2019 Whole-genome sequencing of the artemisinin-resistant parasite line revealed mutations on the k13 gene associated with drug resistance in P. falciparum. artemisinin 31-42 keratin 13 Homo sapiens 93-96 31619243-4 2019 To understand the artemisinin resistance of the imported P. falciparum cases from Africa, the mutations in the k13 gene in parasites from imported malaria cases in Guangxi Province were detected and the treatment efficiency of artesunate monotherapy was observed. artemisinin 18-29 keratin 13 Homo sapiens 111-114 31094238-0 2020 Artemisinin alleviates atherosclerotic lesion by reducing macrophage inflammation via regulation of AMPK/NF-kappaB/NLRP3 inflammasomes pathway. artemisinin 0-11 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 105-114 31094238-0 2020 Artemisinin alleviates atherosclerotic lesion by reducing macrophage inflammation via regulation of AMPK/NF-kappaB/NLRP3 inflammasomes pathway. artemisinin 0-11 NLR family, pyrin domain containing 3 Mus musculus 115-120 31094238-3 2020 The primary aim of this study was to investigate whether artemisinin could be conferred an anti-atherosclerotic effect in high-fat diet (HFD)-fed ApoE-/- mice and explore the possible mechanism. artemisinin 57-68 apolipoprotein E Mus musculus 146-150 31094238-5 2020 Atherosclerotic mice treated with artemisinin showed reduced inflammation by up-regulating adenosine 5"-monophosphate (AMP) activated protein kinase (AMPK) activation and by down-regulating nuclear factor-kappaB (NF-kappaB) phosphorylation and nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome expression in the aortas. artemisinin 34-45 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 190-211 31094238-5 2020 Atherosclerotic mice treated with artemisinin showed reduced inflammation by up-regulating adenosine 5"-monophosphate (AMP) activated protein kinase (AMPK) activation and by down-regulating nuclear factor-kappaB (NF-kappaB) phosphorylation and nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome expression in the aortas. artemisinin 34-45 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 213-222 31094238-5 2020 Atherosclerotic mice treated with artemisinin showed reduced inflammation by up-regulating adenosine 5"-monophosphate (AMP) activated protein kinase (AMPK) activation and by down-regulating nuclear factor-kappaB (NF-kappaB) phosphorylation and nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome expression in the aortas. artemisinin 34-45 NLR family, pyrin domain containing 3 Mus musculus 296-301 31094238-7 2020 In conclusion, we demonstrate that artemisinin may protect the aortas from atherosclerotic lesions by suppression of inflammatory reaction via AMPK/NF-kappaB/NLRP3 inflammasomes signalling in macrophages. artemisinin 35-46 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 148-157 31094238-7 2020 In conclusion, we demonstrate that artemisinin may protect the aortas from atherosclerotic lesions by suppression of inflammatory reaction via AMPK/NF-kappaB/NLRP3 inflammasomes signalling in macrophages. artemisinin 35-46 NLR family, pyrin domain containing 3 Mus musculus 158-163 31577904-0 2019 Regulation of Artemisinin and Its Derivatives on the Assembly Behavior and Cytotoxicity of Amyloid Polypeptides hIAPP and Abeta. artemisinin 14-25 islet amyloid polypeptide Homo sapiens 112-117 31577904-0 2019 Regulation of Artemisinin and Its Derivatives on the Assembly Behavior and Cytotoxicity of Amyloid Polypeptides hIAPP and Abeta. artemisinin 14-25 amyloid beta precursor protein Homo sapiens 122-127 31577904-5 2019 In this work, four compounds, namely, artemisinin (1), dihydroartemisinin (2), artesunate (3), and artemether (4), were exploited to inhibit the assembly behavior of hIAPP and compared with that of Abeta. artemisinin 38-49 islet amyloid polypeptide Homo sapiens 166-171 31465776-0 2019 Renoprotective effects of artemisinin and hydroxychloroquine combination therapy on IgA nephropathy via suppressing NF-kappaB signaling and NLRP3 inflammasome activation by exosomes in rats. artemisinin 26-37 nuclear factor kappa B subunit 1 Homo sapiens 116-125 31465776-0 2019 Renoprotective effects of artemisinin and hydroxychloroquine combination therapy on IgA nephropathy via suppressing NF-kappaB signaling and NLRP3 inflammasome activation by exosomes in rats. artemisinin 26-37 NLR family, pyrin domain containing 3 Rattus norvegicus 140-145 31358323-0 2019 Anti-inflammatory effect of artemisinin on uric acid-induced NLRP3 inflammasome activation through blocking interaction between NLRP3 and NEK7. artemisinin 28-39 NLR family pyrin domain containing 3 Homo sapiens 61-66 31358323-0 2019 Anti-inflammatory effect of artemisinin on uric acid-induced NLRP3 inflammasome activation through blocking interaction between NLRP3 and NEK7. artemisinin 28-39 NLR family pyrin domain containing 3 Homo sapiens 128-133 31358323-9 2019 Enhanced expression of NLRP3, caspase-1, and IL-1beta was noted in macrophages treated with LPS (10 ng/ml) and MSU crystals (0.1 mg/ml), which was markedly suppressed by treatment with artemisinin (1, 10, and 100 muM). artemisinin 185-196 interleukin 1 beta Homo sapiens 45-53 31358323-0 2019 Anti-inflammatory effect of artemisinin on uric acid-induced NLRP3 inflammasome activation through blocking interaction between NLRP3 and NEK7. artemisinin 28-39 NIMA related kinase 7 Homo sapiens 138-142 31358323-9 2019 Enhanced expression of NLRP3, caspase-1, and IL-1beta was noted in macrophages treated with LPS (10 ng/ml) and MSU crystals (0.1 mg/ml), which was markedly suppressed by treatment with artemisinin (1, 10, and 100 muM). artemisinin 185-196 latexin Homo sapiens 213-216 31358323-10 2019 Artemisinin significantly inhibited interaction between NLRP3 and NEK7 in NLRP3 inflammasome activation. artemisinin 0-11 NLR family pyrin domain containing 3 Homo sapiens 56-61 31358323-3 2019 The aim of this study was to clarify the anti-inflammatory effect of artemisinin on activation of uric acid-induced NLRP3 inflammasome through regulation of NEK7. artemisinin 69-80 NLR family pyrin domain containing 3 Homo sapiens 116-121 31358323-3 2019 The aim of this study was to clarify the anti-inflammatory effect of artemisinin on activation of uric acid-induced NLRP3 inflammasome through regulation of NEK7. artemisinin 69-80 NIMA related kinase 7 Homo sapiens 157-161 31358323-10 2019 Artemisinin significantly inhibited interaction between NLRP3 and NEK7 in NLRP3 inflammasome activation. artemisinin 0-11 NIMA related kinase 7 Homo sapiens 66-70 31358323-10 2019 Artemisinin significantly inhibited interaction between NLRP3 and NEK7 in NLRP3 inflammasome activation. artemisinin 0-11 NLR family pyrin domain containing 3 Homo sapiens 74-79 31358323-9 2019 Enhanced expression of NLRP3, caspase-1, and IL-1beta was noted in macrophages treated with LPS (10 ng/ml) and MSU crystals (0.1 mg/ml), which was markedly suppressed by treatment with artemisinin (1, 10, and 100 muM). artemisinin 185-196 NLR family pyrin domain containing 3 Homo sapiens 23-28 31358323-11 2019 Artemisinin (10 and 100 muM) attenuated intracellular K+ efflux in macrophages stimulated with LPS and MSU crystals. artemisinin 0-11 latexin Homo sapiens 24-27 31112710-0 2019 A novel derivative of artemisinin inhibits cell proliferation and metastasis via down-regulation of cathepsin K in breast cancer. artemisinin 22-33 cathepsin K Homo sapiens 100-111 31358323-13 2019 CONCLUSION: This study revealed that artemisinin inhibited activation of NLRP3 inflammasome by suppressing interaction between NEK7 and NLRP3 in uric acid-induced inflammation. artemisinin 37-48 NLR family pyrin domain containing 3 Homo sapiens 73-78 31358323-13 2019 CONCLUSION: This study revealed that artemisinin inhibited activation of NLRP3 inflammasome by suppressing interaction between NEK7 and NLRP3 in uric acid-induced inflammation. artemisinin 37-48 NIMA related kinase 7 Homo sapiens 127-131 31358323-13 2019 CONCLUSION: This study revealed that artemisinin inhibited activation of NLRP3 inflammasome by suppressing interaction between NEK7 and NLRP3 in uric acid-induced inflammation. artemisinin 37-48 NLR family pyrin domain containing 3 Homo sapiens 136-141 31440140-9 2019 Finally, we will also summarize how anti-malarial artemisinin drugs modulate gephyrin-mediated inhibitory neurotransmission. artemisinin 50-61 gephyrin Homo sapiens 77-85 31387196-8 2019 RESULT: The skin erythema and histopathological alteration, as well as the elevated pro-inflammatory factors (IL-1beta, IL6, TNFalpha) and TLR2 were significantly ameliorated by ART treatment in LL37-induced rosacea-like mice. artemisinin 178-181 interleukin 1 alpha Mus musculus 110-118 31387196-8 2019 RESULT: The skin erythema and histopathological alteration, as well as the elevated pro-inflammatory factors (IL-1beta, IL6, TNFalpha) and TLR2 were significantly ameliorated by ART treatment in LL37-induced rosacea-like mice. artemisinin 178-181 interleukin 6 Mus musculus 120-123 31387196-8 2019 RESULT: The skin erythema and histopathological alteration, as well as the elevated pro-inflammatory factors (IL-1beta, IL6, TNFalpha) and TLR2 were significantly ameliorated by ART treatment in LL37-induced rosacea-like mice. artemisinin 178-181 tumor necrosis factor Mus musculus 125-133 31387196-8 2019 RESULT: The skin erythema and histopathological alteration, as well as the elevated pro-inflammatory factors (IL-1beta, IL6, TNFalpha) and TLR2 were significantly ameliorated by ART treatment in LL37-induced rosacea-like mice. artemisinin 178-181 toll-like receptor 2 Mus musculus 139-143 31387196-8 2019 RESULT: The skin erythema and histopathological alteration, as well as the elevated pro-inflammatory factors (IL-1beta, IL6, TNFalpha) and TLR2 were significantly ameliorated by ART treatment in LL37-induced rosacea-like mice. artemisinin 178-181 cathelicidin antimicrobial peptide Homo sapiens 195-199 31452821-7 2019 By contrast, artemisinin decreased proliferative capacity, decreased migration, decreased vimentin expression and increased E-cadherin expression of EMT model cells, indicating that MET was induced. artemisinin 13-24 vimentin Homo sapiens 90-98 31452821-7 2019 By contrast, artemisinin decreased proliferative capacity, decreased migration, decreased vimentin expression and increased E-cadherin expression of EMT model cells, indicating that MET was induced. artemisinin 13-24 cadherin 1 Homo sapiens 124-134 31523201-2 2019 In the current study, we investigated the role of AMPK in the protective effect of artemisinin, an FDA approved anti-malarial Chinese herbal drug, on RPE cell line D407, against H2O2 induced oxidative stress. artemisinin 83-94 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 50-54 31009824-1 2019 Mutations in the Kelch domain of the K13 gene (PF3D7_1343700) were previously associated with artemisinin resistance in Plasmodium falciparum. artemisinin 94-105 kelch like family member 2 Homo sapiens 17-22 30536376-9 2019 Based on our data, we believe that Art can inhibit proliferation of breast cancer cells by activating apoptosis pathways, and inhibit osteoclast formation and differentiation by inhibiting activation of cathepsin K, ATPase H+ transporting V0 subunit D2, nuclear factor of activated T cells 1, calcitonin receptor, and tartrate-resistant acid phosphatase and by inhibiting nuclear factor-kappaB activation. artemisinin 35-38 cathepsin K Homo sapiens 203-214 31085516-0 2019 Evolution and Genetic Diversity of the k13 Gene Associated with Artemisinin Delayed Parasite Clearance in Plasmodium falciparum. artemisinin 64-75 keratin 13 Homo sapiens 39-42 31523201-5 2019 Western blotting showed that artemisinin concentration- and time-dependently stimulated the phosphorylation of AMP-activated protein kinase (AMPK) in D407 cells while AMPK inhibitor Compound C or knock-down of AMPK by si-RNA, inhibited the survival protective effect of artemisinin. artemisinin 29-40 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 111-139 31523201-5 2019 Western blotting showed that artemisinin concentration- and time-dependently stimulated the phosphorylation of AMP-activated protein kinase (AMPK) in D407 cells while AMPK inhibitor Compound C or knock-down of AMPK by si-RNA, inhibited the survival protective effect of artemisinin. artemisinin 29-40 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 141-145 31523201-5 2019 Western blotting showed that artemisinin concentration- and time-dependently stimulated the phosphorylation of AMP-activated protein kinase (AMPK) in D407 cells while AMPK inhibitor Compound C or knock-down of AMPK by si-RNA, inhibited the survival protective effect of artemisinin. artemisinin 29-40 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 167-171 31523201-5 2019 Western blotting showed that artemisinin concentration- and time-dependently stimulated the phosphorylation of AMP-activated protein kinase (AMPK) in D407 cells while AMPK inhibitor Compound C or knock-down of AMPK by si-RNA, inhibited the survival protective effect of artemisinin. artemisinin 29-40 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 167-171 31523201-5 2019 Western blotting showed that artemisinin concentration- and time-dependently stimulated the phosphorylation of AMP-activated protein kinase (AMPK) in D407 cells while AMPK inhibitor Compound C or knock-down of AMPK by si-RNA, inhibited the survival protective effect of artemisinin. artemisinin 270-281 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 111-139 31523201-5 2019 Western blotting showed that artemisinin concentration- and time-dependently stimulated the phosphorylation of AMP-activated protein kinase (AMPK) in D407 cells while AMPK inhibitor Compound C or knock-down of AMPK by si-RNA, inhibited the survival protective effect of artemisinin. artemisinin 270-281 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 141-145 31523201-6 2019 More importantly, artemisinin produced a similar protective effect in primary cultured retinal pigment cells which was also blocked by inhibitors of AMPK. artemisinin 18-29 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 149-153 31523201-7 2019 Taken together, these results suggested that artemisinin promotes survival of human retinal pigment cells against H2O2-induced cell death at least in part through enhancing the activation of AMPK. artemisinin 45-56 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 191-195 31097578-6 2019 On expression of GRASP55 mRNA, or on exposure to the drug artemisinin (which activates GCUSP), the localization of CLRN1N48K to the hair bundles was enhanced. artemisinin 58-69 golgi reassembly stacking protein 2 Homo sapiens 17-24 31263819-0 2019 Identification of HSP90 as a direct target of artemisinin for its anti-inflammatory activity via quantitative chemical proteomics. artemisinin 46-57 heat shock protein 90 alpha family class A member 1 Homo sapiens 18-23 30869127-0 2019 Plasmodium falciparum K13 expression associated with parasite clearance during artemisinin-based combination therapy. artemisinin 79-90 keratin 13 Homo sapiens 22-25 30869127-1 2019 BACKGROUND: Delayed parasite clearance and, consequently, reduced efficacy of artemisinin-based combination therapies have been linked with Plasmodium falciparum K13 gene SNPs in Southeast Asia. artemisinin 78-89 keratin 13 Homo sapiens 162-165 31301971-0 2019 Interactions between artemisinin derivatives and P-glycoprotein. artemisinin 21-32 ATP binding cassette subfamily B member 1 Homo sapiens 49-63 31301971-4 2019 Interestingly, many artemisinin derivatives exhibit excellent ability to overcome P-gp mediated MDR and even show collateral sensitivity against MDR cancer cells. artemisinin 20-31 ATP binding cassette subfamily B member 1 Homo sapiens 82-86 31301971-5 2019 Furthermore, some artemisinin derivatives show P-gp-mediated MDR reversal activity. artemisinin 18-29 ATP binding cassette subfamily B member 1 Homo sapiens 47-51 31301971-6 2019 Therefore, the interaction between P-gp and artemisinin derivatives is important to develop novel combination treatment protocols with artemisinin derivatives and established anticancer drugs that are P-gp substrates. artemisinin 44-55 ATP binding cassette subfamily B member 1 Homo sapiens 201-205 31301971-6 2019 Therefore, the interaction between P-gp and artemisinin derivatives is important to develop novel combination treatment protocols with artemisinin derivatives and established anticancer drugs that are P-gp substrates. artemisinin 135-146 ATP binding cassette subfamily B member 1 Homo sapiens 35-39 31301971-7 2019 PURPOSE: This systematic review provides an updated overview on the interaction between artemisinin derivatives and P-gp and the effect of artemisinin derivatives on the P-gp expression level. artemisinin 88-99 ATP binding cassette subfamily B member 1 Homo sapiens 116-120 31301971-7 2019 PURPOSE: This systematic review provides an updated overview on the interaction between artemisinin derivatives and P-gp and the effect of artemisinin derivatives on the P-gp expression level. artemisinin 139-150 ATP binding cassette subfamily B member 1 Homo sapiens 170-174 31301971-8 2019 RESULTS: Artemisinin derivatives exhibit multi-specific interactions with P-gp. artemisinin 9-20 ATP binding cassette subfamily B member 1 Homo sapiens 74-78 31301971-10 2019 However, some of novel synthetized artemisinin derivatives exhibit P-gp substrate properties. artemisinin 35-46 ATP binding cassette subfamily B member 1 Homo sapiens 67-71 31301971-11 2019 Furthermore, many artemisinin derivatives act as P-gp inhibitors, which exhibit the potential to reverse MDR towards clinically used anticancer drugs. artemisinin 18-29 ATP binding cassette subfamily B member 1 Homo sapiens 49-53 31301971-12 2019 CONCLUSION: Therefore, studies on the interaction between artemisinin derivatives and P-gp provide important information for the development of novel anti-cancer artemisinin derivatives to reverse P-gp mediated MDR and for the design of rational artemisinin-based combination therapies against cancer. artemisinin 58-69 ATP binding cassette subfamily B member 1 Homo sapiens 86-90 31301971-12 2019 CONCLUSION: Therefore, studies on the interaction between artemisinin derivatives and P-gp provide important information for the development of novel anti-cancer artemisinin derivatives to reverse P-gp mediated MDR and for the design of rational artemisinin-based combination therapies against cancer. artemisinin 58-69 ATP binding cassette subfamily B member 1 Homo sapiens 197-201 31301971-12 2019 CONCLUSION: Therefore, studies on the interaction between artemisinin derivatives and P-gp provide important information for the development of novel anti-cancer artemisinin derivatives to reverse P-gp mediated MDR and for the design of rational artemisinin-based combination therapies against cancer. artemisinin 162-173 ATP binding cassette subfamily B member 1 Homo sapiens 86-90 31301971-12 2019 CONCLUSION: Therefore, studies on the interaction between artemisinin derivatives and P-gp provide important information for the development of novel anti-cancer artemisinin derivatives to reverse P-gp mediated MDR and for the design of rational artemisinin-based combination therapies against cancer. artemisinin 162-173 ATP binding cassette subfamily B member 1 Homo sapiens 197-201 31301971-12 2019 CONCLUSION: Therefore, studies on the interaction between artemisinin derivatives and P-gp provide important information for the development of novel anti-cancer artemisinin derivatives to reverse P-gp mediated MDR and for the design of rational artemisinin-based combination therapies against cancer. artemisinin 162-173 ATP binding cassette subfamily B member 1 Homo sapiens 86-90 31301971-12 2019 CONCLUSION: Therefore, studies on the interaction between artemisinin derivatives and P-gp provide important information for the development of novel anti-cancer artemisinin derivatives to reverse P-gp mediated MDR and for the design of rational artemisinin-based combination therapies against cancer. artemisinin 162-173 ATP binding cassette subfamily B member 1 Homo sapiens 197-201 31269339-5 2019 Each concentration of artemisinin increased the total serum proteins, gamma-globulins and the serum activity of CK and decreased the serum ALP level. artemisinin 22-33 cytidine/uridine monophosphate kinase 1 Gallus gallus 112-114 31269339-5 2019 Each concentration of artemisinin increased the total serum proteins, gamma-globulins and the serum activity of CK and decreased the serum ALP level. artemisinin 22-33 PDZ and LIM domain 3 Gallus gallus 139-142 31151322-0 2019 Artemisinin Attenuated Hydrogen Peroxide (H2O2)-Induced Oxidative Injury in SH-SY5Y and Hippocampal Neurons via the Activation of AMPK Pathway. artemisinin 0-11 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 130-134 31151322-6 2019 Further studies showed that artemisinin significantly restored the nuclear morphology, improved the abnormal changes in intracellular reactive oxygen species (ROS), reduced the mitochondrial membrane potential, and caspase-3 activation, thereby attenuating apoptosis. artemisinin 28-39 caspase 3 Homo sapiens 215-224 31151322-7 2019 Artemisinin also stimulated the phosphorylation of the adenosine monophosphate -activated protein kinase (AMPK) pathway in SH-SY5Y cells in a time- and concentration-dependent manner. artemisinin 0-11 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 55-104 31151322-7 2019 Artemisinin also stimulated the phosphorylation of the adenosine monophosphate -activated protein kinase (AMPK) pathway in SH-SY5Y cells in a time- and concentration-dependent manner. artemisinin 0-11 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 106-110 31151322-8 2019 Inhibition of the AMPK pathway attenuated the protective effect of artemisinin. artemisinin 67-78 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 18-22 31151322-11 2019 Cumulatively, these results indicated that artemisinin protected neuronal cells from oxidative damage, at least in part through the activation of AMPK. artemisinin 43-54 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 146-150 31097578-7 2019 Artemisinin treatment also effectively restored hair cell mechanotransduction and attenuated progressive hair cell dysfunction in clrn1 KO/KO larvae that express CLRN1 c.144T>G , highlighting the potential of artemisinin to prevent sensory loss in CLRN1 c.144T>G patients. artemisinin 0-11 clarin 1 Homo sapiens 130-135 31097578-7 2019 Artemisinin treatment also effectively restored hair cell mechanotransduction and attenuated progressive hair cell dysfunction in clrn1 KO/KO larvae that express CLRN1 c.144T>G , highlighting the potential of artemisinin to prevent sensory loss in CLRN1 c.144T>G patients. artemisinin 0-11 clarin 1 Homo sapiens 162-167 31097578-7 2019 Artemisinin treatment also effectively restored hair cell mechanotransduction and attenuated progressive hair cell dysfunction in clrn1 KO/KO larvae that express CLRN1 c.144T>G , highlighting the potential of artemisinin to prevent sensory loss in CLRN1 c.144T>G patients. artemisinin 0-11 clarin 1 Homo sapiens 251-256 30707255-4 2019 In addition to its well-established antimalarial properties, emerging evidence hints that artemisinin family drugs also possess preferential immunoregulatory and anti-inflammation properties, such as modifying T lymphocytes" activation and cytokines release, modulating Th1/Th2 balance, activating regulatory T cells (Tregs), modulating inflammatory signaling pathways, as well as acting on non-specific mechanisms of OLP. artemisinin 90-101 negative elongation factor complex member C/D Homo sapiens 270-273 31156427-10 2019 It is also important to highlight that several plant-derived products (compounds, essential oils) from Artemisia plants have shown high inhibitory potential against Leishmania spp., such as artemisinin and its derivatives. artemisinin 190-201 histocompatibility minor 13 Homo sapiens 176-179 30746754-0 2019 Artemisinin inhibits angiogenesis by regulating p38 MAPK/CREB/TSP-1 signaling pathway in osteosarcoma. artemisinin 0-11 cAMP responsive element binding protein 1 Homo sapiens 57-61 30934859-0 2019 Possible Role of the Ca2+/Mn2+ P-Type ATPase Pmr1p on Artemisinin Toxicity through an Induction of Intracellular Oxidative Stress. artemisinin 54-65 Ca(2+)/Mn(2+)-transporting P-type ATPase PMR1 Saccharomyces cerevisiae S288C 45-50 30934859-3 2019 We utilized Saccharomyces cerevisiae as a model to examine the involvement of Pmr1p, a functional homolog of PfATP6, on the toxicity of artemisinin. artemisinin 136-147 Ca(2+)/Mn(2+)-transporting P-type ATPase PMR1 Saccharomyces cerevisiae S288C 78-83 30934859-4 2019 Our analysis demonstrated that cells lacking Pmr1p are less susceptible to growth inhibition from artemisinin and its derivatives. artemisinin 98-109 Ca(2+)/Mn(2+)-transporting P-type ATPase PMR1 Saccharomyces cerevisiae S288C 45-50 30934859-9 2019 We propose that loss of function mutations in Pmr1p in yeast cells and PfATP6 in P. falciparum are protective against artemisinin toxicity due to reduced intracellular oxidative damage. artemisinin 118-129 Ca(2+)/Mn(2+)-transporting P-type ATPase PMR1 Saccharomyces cerevisiae S288C 46-51 30260039-0 2019 Artemisinin attenuates tubulointerstitial inflammation and fibrosis via the NF-kappaB/NLRP3 pathway in rats with 5/6 subtotal nephrectomy. artemisinin 0-11 NLR family, pyrin domain containing 3 Rattus norvegicus 86-91 30260039-10 2019 Furthermore, Art inhibited the activation of NLRP3 inflammasome and NF-kappaB in the kidneys. artemisinin 13-16 NLR family, pyrin domain containing 3 Rattus norvegicus 45-50 30790532-3 2019 (2019) demonstrate that artemisinin antimalarial drugs bind to gephyrin at the same site where the receptor interaction occurs. artemisinin 24-35 gephyrin Homo sapiens 63-71 30783079-2 2019 Here, we evaluated a library of natural compounds for inhibitors of Axl and identified dihydroartemisinin, the active principle of the anti-malarial drug artemisinin, as an Axl-inhibitor in prostate cancer. artemisinin 94-105 AXL receptor tyrosine kinase Homo sapiens 173-176 30746754-0 2019 Artemisinin inhibits angiogenesis by regulating p38 MAPK/CREB/TSP-1 signaling pathway in osteosarcoma. artemisinin 0-11 thrombospondin 1 Homo sapiens 62-67 30746754-6 2019 In this study, we found that artemisinin could induce both the expression and secretion of thrombospondin-1 (TSP-1) in a dose-dependent way in osteosarcoma cells. artemisinin 29-40 thrombospondin 1 Homo sapiens 91-107 30746754-6 2019 In this study, we found that artemisinin could induce both the expression and secretion of thrombospondin-1 (TSP-1) in a dose-dependent way in osteosarcoma cells. artemisinin 29-40 thrombospondin 1 Homo sapiens 109-114 30746754-7 2019 In addition, TSP-1 could effectively restore the artemisinin-induced suppression of angiogenesis in human umbilical vein endothelial cells (HUVECs). artemisinin 49-60 thrombospondin 1 Homo sapiens 13-18 30746754-9 2019 Moreover, our results showed that artemisinin could induce the phosphorylation of CREB via the activation of p38 mitogen-activated protein kinase (MAPK) signaling pathway in osteosarcoma cells. artemisinin 34-45 cAMP responsive element binding protein 1 Homo sapiens 82-86 30746754-9 2019 Moreover, our results showed that artemisinin could induce the phosphorylation of CREB via the activation of p38 mitogen-activated protein kinase (MAPK) signaling pathway in osteosarcoma cells. artemisinin 34-45 mitogen-activated protein kinase 14 Homo sapiens 109-145 30746754-10 2019 In vivo, we also found that artemisinin could inhibit osteosarcoma proliferation and angiogenesis by regulating the p38 MAPK/CREB/TSP-1 signaling pathway. artemisinin 28-39 cAMP responsive element binding protein 1 Homo sapiens 125-129 30746754-10 2019 In vivo, we also found that artemisinin could inhibit osteosarcoma proliferation and angiogenesis by regulating the p38 MAPK/CREB/TSP-1 signaling pathway. artemisinin 28-39 thrombospondin 1 Homo sapiens 130-135 30746754-11 2019 Taken together, our findings indicated that artemisinin could inhibit angiogenesis by regulating the p38 MAPK/CREB/TSP-1 signaling pathway in osteosarcoma. artemisinin 44-55 cAMP responsive element binding protein 1 Homo sapiens 110-114 30746754-11 2019 Taken together, our findings indicated that artemisinin could inhibit angiogenesis by regulating the p38 MAPK/CREB/TSP-1 signaling pathway in osteosarcoma. artemisinin 44-55 thrombospondin 1 Homo sapiens 115-120 30607137-0 2018 Patients" adherence to artemisinin-based combination therapy and healthcare workers" perception and practice in Savannakhet province, Lao PDR. artemisinin 23-34 interleukin 4 induced 1 Homo sapiens 134-137 30337251-8 2018 An enhancement of SP1 and CD14 expression was observed in artemisinin treated human monocyte cell line. artemisinin 58-69 CD14 molecule Homo sapiens 26-30 30278333-0 2018 Development of high potent and selective Bcl-2 inhibitors bearing the structural elements of natural product artemisinin. artemisinin 109-120 BCL2 apoptosis regulator Homo sapiens 41-46 30278333-1 2018 By taking advantage of the apoptosis-inducing capacity of artemisinin derivatives, we developed several series of compounds by merging the basic structural elements of the natural product artemisinin into the P2 interaction pocket of the clinically prescribed Bcl-2 inhibitor venetoclax. artemisinin 58-69 BCL2 apoptosis regulator Homo sapiens 260-265 30278333-1 2018 By taking advantage of the apoptosis-inducing capacity of artemisinin derivatives, we developed several series of compounds by merging the basic structural elements of the natural product artemisinin into the P2 interaction pocket of the clinically prescribed Bcl-2 inhibitor venetoclax. artemisinin 188-199 BCL2 apoptosis regulator Homo sapiens 260-265 30505200-4 2018 We found that artemisinin inhibited the proliferation in C6 cells and induced cell cycle arrest and a caspase-3-dependent cell apoptosis. artemisinin 14-25 caspase 3 Rattus norvegicus 102-111 30271498-2 2018 Dihydroartemisinin (DHA), a the effective anti-malarial derivative of artemisinin, demonstrated potent anti-angiogenic activities that closely related to the regulation of vascular endothelial growth factor (VEGF) signaling cascade. artemisinin 7-18 vascular endothelial growth factor A Homo sapiens 172-206 30367653-0 2018 A single nucleotide polymorphism in the Plasmodium falciparum atg18 gene associates with artemisinin resistance and confers enhanced parasite survival under nutrient deprivation. artemisinin 89-100 phosphoinositide binding protein ATG18 Saccharomyces cerevisiae S288C 62-67 30367653-4 2018 RESULTS: A SNP in autophagy-related gene 18 (atg18) was associated with long parasite clearance half-life in patients following artemisinin-based combination therapy. artemisinin 128-139 phosphoinositide binding protein ATG18 Saccharomyces cerevisiae S288C 45-50 30367653-10 2018 CONCLUSIONS: These results suggest that the atg18 T38I polymorphism may provide additional resistance against artemisinin derivatives, but not partner drugs, even in the absence of kelch13 mutations, and may also be important in parasite survival during nutrient deprivation. artemisinin 110-121 phosphoinositide binding protein ATG18 Saccharomyces cerevisiae S288C 44-49 30443164-0 2018 Plasmodium falciparum Treated with Artemisinin-based Combined Therapy Exhibits Enhanced Mutation, Heightened Cortisol and TNF-alpha Induction. artemisinin 35-46 tumor necrosis factor Homo sapiens 122-131 30271498-2 2018 Dihydroartemisinin (DHA), a the effective anti-malarial derivative of artemisinin, demonstrated potent anti-angiogenic activities that closely related to the regulation of vascular endothelial growth factor (VEGF) signaling cascade. artemisinin 7-18 vascular endothelial growth factor A Homo sapiens 208-212 29731711-6 2018 Furthermore, our study demonstrated that artemisinin activated Akt/Bcl-2 signaling and that neuroprotective effect of artemisinin was blocked by Akt-specific inhibitor, MK2206. artemisinin 41-52 thymoma viral proto-oncogene 1 Mus musculus 63-66 28737446-10 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.46, 0.17, 0.15, 0.60, 0.51, 0.66 and 0.77, respectively, in HLM determined by chemical inhibition method in the presence of BSA (0.5% w/v). artemisinin 26-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 29991950-2 2018 This study aimed to explore the effects of artemisinin alone or combined with neurectomy of pterygoid canal in ovalbumin-induced AR mouse model and illustrate the underlying mechanisms. artemisinin 43-54 serine (or cysteine) peptidase inhibitor, clade B, member 1, pseudogene Mus musculus 111-120 29991950-10 2018 An increased Treg cell proportion and inhibited ERK phosphorylation were observed in artemisinin-treated groups as compared to those in the AR group. artemisinin 85-96 mitogen-activated protein kinase 1 Mus musculus 48-51 29991950-12 2018 Conclusions: These results indicated that artemisinin exhibited anti-allergic effect by inhibiting ERK activation and increasing Treg cell proportion, which subsequently decreased the expressions of allergic mediators. artemisinin 42-53 mitogen-activated protein kinase 1 Mus musculus 99-102 29690890-0 2018 A novel field-based molecular assay to detect validated artemisinin-resistant k13 mutants. artemisinin 56-67 keratin 13 Homo sapiens 78-81 30159225-1 2018 Objectives: The CYP2B6 is one of the most polymorphic CYP genes in humans that has the potential to modify the pharmacological and toxicological responses to clinically important drugs such as antimalarial artemisinin and its derivatives. artemisinin 206-217 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 29967165-3 2018 The beta5 inhibitors synergize with a beta2 inhibitor in vitro and in mice and with artemisinin. artemisinin 84-95 integrin beta 5 Mus musculus 4-9 30011856-1 2018 According to the precepts that C-10 amino-artemisinins display optimum biological activities for the artemisinin drug class, and that attachment of a sugar enhances specificity of drug delivery, polarity and solubility so as to attenuate toxicity, we assessed the effects of attaching sugars to N-4 of the dihydroartemisinin (DHA)-piperazine derivative prepared in one step from DHA and piperazine. artemisinin 42-53 homeobox C10 Homo sapiens 31-35 28737446-9 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.24, 0.28, 0.15, 0.45, 0.46, 0.42 and 0.54, respectively, in HLM determined by chemical inhibition method. artemisinin 26-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 29731711-7 2018 Taken together, our study indicated that artemisinin prevented neuronal HT-22 cell from glutamate-induced oxidative injury by activation of Akt signaling pathway. artemisinin 41-52 thymoma viral proto-oncogene 1 Mus musculus 140-143 29731711-6 2018 Furthermore, our study demonstrated that artemisinin activated Akt/Bcl-2 signaling and that neuroprotective effect of artemisinin was blocked by Akt-specific inhibitor, MK2206. artemisinin 41-52 B cell leukemia/lymphoma 2 Mus musculus 67-72 29731711-6 2018 Furthermore, our study demonstrated that artemisinin activated Akt/Bcl-2 signaling and that neuroprotective effect of artemisinin was blocked by Akt-specific inhibitor, MK2206. artemisinin 41-52 thymoma viral proto-oncogene 1 Mus musculus 145-148 29731711-6 2018 Furthermore, our study demonstrated that artemisinin activated Akt/Bcl-2 signaling and that neuroprotective effect of artemisinin was blocked by Akt-specific inhibitor, MK2206. artemisinin 118-129 thymoma viral proto-oncogene 1 Mus musculus 145-148 30266137-1 2018 In the present study, a transferrin-conjugated nanostructured lipid carrier (TF-NLCs) for brain delivery of artemisinin (ART) was developed. artemisinin 108-119 transferrin Homo sapiens 24-35 29456648-0 2018 Immunosuppressive effects of hydroxychloroquine and artemisinin combination therapy via the nuclear factor-kappaB signaling pathway in lupus nephritis mice. artemisinin 52-63 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 92-113 30562742-0 2018 Artemisinin Ameliorates Osteoarthritis by Inhibiting the Wnt/beta-Catenin Signaling Pathway. artemisinin 0-11 catenin beta 1 Homo sapiens 61-73 29246124-12 2017 Along with orphan nuclear receptors (ERRalpha, ERRbeta and ERRgamma), artemisinin altered the ERalpha/ERbeta/PR/Her expression status of MCF-7 cells. artemisinin 70-81 estrogen receptor 1 Homo sapiens 94-101 33525848-8 2017 Alternative candidate markers of artemisinin susceptibility are also described, as K13-independent treatment failure has been observed in African P. falciparum and in the rodent malaria parasite Plasmodium chabaudi. artemisinin 33-44 keratin 13 Homo sapiens 83-86 29246124-12 2017 Along with orphan nuclear receptors (ERRalpha, ERRbeta and ERRgamma), artemisinin altered the ERalpha/ERbeta/PR/Her expression status of MCF-7 cells. artemisinin 70-81 estrogen receptor 2 Homo sapiens 102-108 28952817-2 2017 Artemisinin, as an anti-malaria as its agent, has been used widely in the treatment of malaria, shifts the lymphocyte responses from Th1 to Th2. artemisinin 0-11 heart and neural crest derivatives expressed 2 Mus musculus 140-143 29241036-2 2017 (2017) show that translational repression through eIF2alpha phosphorylation mediated by PK4 kinase activity plays a key role in artemisinin resistance in recrudescent malaria infections. artemisinin 128-139 eukaryotic translation initiation factor 2A Homo sapiens 50-59 29241041-0 2017 Inhibiting the Plasmodium eIF2alpha Kinase PK4 Prevents Artemisinin-Induced Latency. artemisinin 56-67 eukaryotic translation initiation factor 2A Mus musculus 26-35 28952817-9 2017 The concentration of IFN-gamma in the low dose of artemisinin treated group showed significantly (p value < .05) lower in comparison to the untreated group. artemisinin 50-61 interferon gamma Mus musculus 21-30 28952817-11 2017 CONCLUSIONS: Since, artemisinin can shift the immune responses from Th1 to Th2, therefore, it can be helpful in the treatment of MS after more investigation. artemisinin 20-31 heart and neural crest derivatives expressed 2 Mus musculus 75-78 27931149-0 2017 Chrysosplenetin inhibits artemisinin efflux in P-gp-over-expressing Caco-2 cells and reverses P-gp/MDR1 mRNA up-regulated expression induced by artemisinin in mouse small intestine. artemisinin 25-36 phosphoglycolate phosphatase Homo sapiens 47-51 27931149-0 2017 Chrysosplenetin inhibits artemisinin efflux in P-gp-over-expressing Caco-2 cells and reverses P-gp/MDR1 mRNA up-regulated expression induced by artemisinin in mouse small intestine. artemisinin 144-155 phosphoglycolate phosphatase Homo sapiens 94-98 27931149-0 2017 Chrysosplenetin inhibits artemisinin efflux in P-gp-over-expressing Caco-2 cells and reverses P-gp/MDR1 mRNA up-regulated expression induced by artemisinin in mouse small intestine. artemisinin 144-155 ATP binding cassette subfamily B member 1 Homo sapiens 99-103 29387451-3 2017 Herein, we report that dihydroartemisinin, one of the most active derivatives of Artemisinin, directly targets platelet-derived growth factor receptor-alpha (PDGFRalpha) to inhibit ovarian cancer cell growth and metastasis. artemisinin 81-92 platelet derived growth factor receptor alpha Homo sapiens 111-156 29178921-1 2017 BACKGROUND: Artemisinin resistance, linked to polymorphisms in the Kelch gene on chromosome 13 of Plasmodium falciparum (k13), has outpaced containment efforts in South East Asia. artemisinin 12-23 keratin 13 Homo sapiens 121-124 29178921-9 2017 The prevalence of k13 mutations as well as allele diversity varies considerably across short distances, presumably because of historical patterns of artemisinin use and population movements. artemisinin 149-160 keratin 13 Homo sapiens 18-21 29387451-3 2017 Herein, we report that dihydroartemisinin, one of the most active derivatives of Artemisinin, directly targets platelet-derived growth factor receptor-alpha (PDGFRalpha) to inhibit ovarian cancer cell growth and metastasis. artemisinin 81-92 platelet derived growth factor receptor alpha Homo sapiens 158-168 29387451-7 2017 Considering that Artemisinin compounds are currently clinically used drugs with favorable safety profiles, the results from this study will potentiate their use in combination with clinically used PDGFRalpha inhibitors, leading to maximal therapeutic efficacy with minimal adverse effects in PDGFRalpha-positive cancer patients. artemisinin 17-28 platelet derived growth factor receptor alpha Homo sapiens 197-207 29387451-7 2017 Considering that Artemisinin compounds are currently clinically used drugs with favorable safety profiles, the results from this study will potentiate their use in combination with clinically used PDGFRalpha inhibitors, leading to maximal therapeutic efficacy with minimal adverse effects in PDGFRalpha-positive cancer patients. artemisinin 17-28 platelet derived growth factor receptor alpha Homo sapiens 292-302 28447781-7 2017 Taken together, these results demonstrated that artemisinin promoted the survival of RGC-5 cells from H2O2 toxicity via the activation of the p38 and ERK1/2 pathways. artemisinin 48-59 mitogen-activated protein kinase 14 Mus musculus 142-145 28341405-0 2017 Synthesis of novel C-9 carbon substituted derivatives of artemisinin. artemisinin 57-68 complement C9 Homo sapiens 19-22 29078767-0 2017 Correction to: Prevalence of K13 mutation and Day-3 positive parasitaemia in artemisinin-resistant malaria endemic area of Cambodia: a cross-sectional study. artemisinin 77-88 keratin 13 Homo sapiens 29-32 28708672-0 2017 Artemisinin disrupts androgen responsiveness of human prostate cancer cells by stimulating the 26S proteasome-mediated degradation of the androgen receptor protein. artemisinin 0-11 androgen receptor Homo sapiens 138-155 28708672-3 2017 Artemisinin treatment induced the 26S proteasome-mediated degradation of the receptor protein, without altering AR transcript levels, in androgen-responsive LNCaP prostate cancer cells or PC-3 prostate cancer cells expressing exogenous wild-type AR. artemisinin 0-11 androgen receptor Homo sapiens 246-248 28708672-4 2017 Furthermore, artemisinin stimulated AR ubiquitination and AR receptor interactions with the E3 ubiquitin ligase MDM2 in LNCaP cells. artemisinin 13-24 androgen receptor Homo sapiens 36-38 28708672-4 2017 Furthermore, artemisinin stimulated AR ubiquitination and AR receptor interactions with the E3 ubiquitin ligase MDM2 in LNCaP cells. artemisinin 13-24 MDM2 proto-oncogene Homo sapiens 112-116 28708672-5 2017 The artemisinin-induced loss of AR protein prevented androgen-responsive cell proliferation and ablated total AR transcriptional activity. artemisinin 4-15 androgen receptor Homo sapiens 32-34 28708672-5 2017 The artemisinin-induced loss of AR protein prevented androgen-responsive cell proliferation and ablated total AR transcriptional activity. artemisinin 4-15 androgen receptor Homo sapiens 110-112 28708672-6 2017 The serine/threonine protein kinase AKT-1 was shown to be highly associated with artemisinin-induced proteasome-mediated degradation of AR protein. artemisinin 81-92 AKT serine/threonine kinase 1 Homo sapiens 36-41 28708672-6 2017 The serine/threonine protein kinase AKT-1 was shown to be highly associated with artemisinin-induced proteasome-mediated degradation of AR protein. artemisinin 81-92 androgen receptor Homo sapiens 136-138 28708672-7 2017 Artemisinin treatment activated AKT-1 enzymatic activity, enhanced receptor association with AKT-1, and induced AR serine phosphorylation. artemisinin 0-11 AKT serine/threonine kinase 1 Homo sapiens 32-37 28708672-7 2017 Artemisinin treatment activated AKT-1 enzymatic activity, enhanced receptor association with AKT-1, and induced AR serine phosphorylation. artemisinin 0-11 AKT serine/threonine kinase 1 Homo sapiens 93-98 28708672-7 2017 Artemisinin treatment activated AKT-1 enzymatic activity, enhanced receptor association with AKT-1, and induced AR serine phosphorylation. artemisinin 0-11 androgen receptor Homo sapiens 112-114 28708672-8 2017 Treatment of LNCaP cells with the PI3-kinase inhibitor LY294002, which inhibits the PI3-kinase-dependent activation of AKT-1, prevented the artemisinin-induced AR degradation. artemisinin 140-151 AKT serine/threonine kinase 1 Homo sapiens 119-124 28708672-8 2017 Treatment of LNCaP cells with the PI3-kinase inhibitor LY294002, which inhibits the PI3-kinase-dependent activation of AKT-1, prevented the artemisinin-induced AR degradation. artemisinin 140-151 androgen receptor Homo sapiens 160-162 28708672-10 2017 Taken together, our results indicate that artemisinin induces the degradation of AR protein and disrupts androgen responsiveness of human prostate cancer cells, suggesting that this natural compound represents a new potential therapeutic molecule that selectively targets AR levels. artemisinin 42-53 androgen receptor Homo sapiens 81-83 28708672-10 2017 Taken together, our results indicate that artemisinin induces the degradation of AR protein and disrupts androgen responsiveness of human prostate cancer cells, suggesting that this natural compound represents a new potential therapeutic molecule that selectively targets AR levels. artemisinin 42-53 androgen receptor Homo sapiens 272-274 28903755-0 2017 Prevalence of K13 mutation and Day-3 positive parasitaemia in artemisinin-resistant malaria endemic area of Cambodia: a cross-sectional study. artemisinin 62-73 keratin 13 Homo sapiens 14-17 28903755-2 2017 In 2013, mutations in the propeller domain of the kelch protein K13 was found to be associated with artemisinin resistance. artemisinin 100-111 keratin 13 Homo sapiens 64-67 28903755-12 2017 Further investigation should be made to determine if k13 marker remains useful as a tool for tracking artemisinin resistance and predicting the trend of the efficacy of artemisinin combination therapy once the mutant alleles have been well established in the population. artemisinin 102-113 keratin 13 Homo sapiens 53-56 28447781-4 2017 Western blot analysis showed that artemisinin upregulated the phosphorylation of p38 and extracellular signal-regulated kinases1/2 (ERK1/2) and reversed the inhibitory effect of H2O2 on the phosphorylation of these two kinases. artemisinin 34-45 mitogen activated protein kinase 14 Rattus norvegicus 81-84 28447781-4 2017 Western blot analysis showed that artemisinin upregulated the phosphorylation of p38 and extracellular signal-regulated kinases1/2 (ERK1/2) and reversed the inhibitory effect of H2O2 on the phosphorylation of these two kinases. artemisinin 34-45 mitogen activated protein kinase 3 Rattus norvegicus 89-130 28447781-4 2017 Western blot analysis showed that artemisinin upregulated the phosphorylation of p38 and extracellular signal-regulated kinases1/2 (ERK1/2) and reversed the inhibitory effect of H2O2 on the phosphorylation of these two kinases. artemisinin 34-45 mitogen activated protein kinase 3 Rattus norvegicus 132-138 28447781-5 2017 Moreover, protective effect of artemisinin was blocked by the p38 kinase inhibitor PD169316 or ERK1/2 kinase pathway inhibitor PD98059, respectively. artemisinin 31-42 mitogen-activated protein kinase 14 Mus musculus 62-65 28447781-5 2017 Moreover, protective effect of artemisinin was blocked by the p38 kinase inhibitor PD169316 or ERK1/2 kinase pathway inhibitor PD98059, respectively. artemisinin 31-42 mitogen activated protein kinase 3 Rattus norvegicus 95-101 29132370-2 2017 In September 2008, a public-private mix (PPM) strategy was launched initially in four northern and southern provinces in Lao PDR to increase access to rapid diagnostic tests (RDTs) and artemisinin-based combination therapy (ACT), improve quality of care, and collect routine malaria data from the private sector. artemisinin 185-196 interleukin 4 induced 1 Homo sapiens 121-124 28192150-0 2017 Effect of artemisinin on neuropathic pain mediated by P2X4 receptor in dorsal root ganglia. artemisinin 10-21 purinergic receptor P2X 4 Homo sapiens 54-58 28192150-7 2017 The results demonstrated that artemisinin relieved pain behaviors in the CCI rats, inhibited the expression of P2X4 receptor in the DRG, and decreased the ATP-activated currents in HEK293 cells transfected with P2X4 plasmid. artemisinin 30-41 purinergic receptor P2X 4 Homo sapiens 111-115 28192150-7 2017 The results demonstrated that artemisinin relieved pain behaviors in the CCI rats, inhibited the expression of P2X4 receptor in the DRG, and decreased the ATP-activated currents in HEK293 cells transfected with P2X4 plasmid. artemisinin 30-41 purinergic receptor P2X 4 Homo sapiens 211-215 28192150-9 2017 After CCI rats were treated with artemisinin, the coexpression of P2X4 receptor and GFAP in the DRG was significantly decreased compared to the CCI group. artemisinin 33-44 purinergic receptor P2X 4 Homo sapiens 66-70 28192150-10 2017 This finding suggested that artemisinin could inhibit the nociceptive transmission mediated by P2X4 receptor in the DRG SGCs and thus relieve pain behaviors in the CCI rats. artemisinin 28-39 purinergic receptor P2X 4 Homo sapiens 95-99 29225511-6 2017 Trapping of vanillin and (+)-artemisinin elicited single-crystal-to-single-crystal transformations where space group symmetry reduced from C2/c to P1 and C2, respectively, and the absolute configuration of (+)-artemisinin was determined through anomalous dispersion. artemisinin 25-40 complement C2 Homo sapiens 139-156 28447781-7 2017 Taken together, these results demonstrated that artemisinin promoted the survival of RGC-5 cells from H2O2 toxicity via the activation of the p38 and ERK1/2 pathways. artemisinin 48-59 mitogen-activated protein kinase 3 Mus musculus 150-156 28476599-5 2017 This up-regulation is not caused by haem but rather by Hz-generated lipoperoxidation products (15-HETE) and fibrinogen associated to Hz, and is, at least in part, triggered by the activation of NF-kappaB, as it was significantly inhibited by artemisinin and other NF-kappaB pathway inhibitors. artemisinin 242-253 nuclear factor kappa B subunit 1 Homo sapiens 194-203 28806957-2 2017 Although K13 kelch propeller has been assessed for artemisinin resistance molecular marker, most of the mutations need to be validated. artemisinin 51-62 keratin 13 Homo sapiens 9-12 28806957-3 2017 In this study, artemisinin resistance was assessed by clinical and molecular analysis, including k13 and recently reported markers, pfarps10, pffd and pfmdr2. artemisinin 15-26 keratin 13 Homo sapiens 97-100 28806957-10 2017 CONCLUSIONS: Apart from k13, pfarps10, pffd and pfmdr2 are also useful for molecular surveillance of artemisinin resistance especially where k13 mutation has not been reported. artemisinin 101-112 keratin 13 Homo sapiens 141-144 28476599-5 2017 This up-regulation is not caused by haem but rather by Hz-generated lipoperoxidation products (15-HETE) and fibrinogen associated to Hz, and is, at least in part, triggered by the activation of NF-kappaB, as it was significantly inhibited by artemisinin and other NF-kappaB pathway inhibitors. artemisinin 242-253 nuclear factor kappa B subunit 1 Homo sapiens 264-273 28391183-0 2017 Artemisinin protects PC12 cells against beta-amyloid-induced apoptosis through activation of the ERK1/2 signaling pathway. artemisinin 0-11 mitogen activated protein kinase 3 Rattus norvegicus 97-103 28391183-8 2017 Western blotting analysis demonstrated that artemisinin activated extracellular regulated kinase ERK1/2 but not Akt survival signaling. artemisinin 44-55 mitogen activated protein kinase 3 Rattus norvegicus 97-103 28391183-9 2017 Consistent with the role of ERK1/2, preincubation of cells with ERK1/2 pathway inhibitor PD98059 blocked the effect of artemisinin while PI3K inhibitor LY294002 has no effect. artemisinin 119-130 mitogen activated protein kinase 3 Rattus norvegicus 28-34 28391183-9 2017 Consistent with the role of ERK1/2, preincubation of cells with ERK1/2 pathway inhibitor PD98059 blocked the effect of artemisinin while PI3K inhibitor LY294002 has no effect. artemisinin 119-130 mitogen activated protein kinase 3 Rattus norvegicus 64-70 28391183-11 2017 Taken together, these results, at the first time, suggest that artemisinin is a potential protectant against beta amyloid insult through activation of the ERK1/2 pathway. artemisinin 63-74 mitogen activated protein kinase 3 Rattus norvegicus 155-161 28737711-6 2017 Additionally, the artemisinin-enhanced cytotoxic effect of NK-92MI cells on tumor cells was accompanied by the stimulation of granule exocytosis, as evidenced by the detection of CD107a expression in NK cells. artemisinin 18-29 lysosomal associated membrane protein 1 Homo sapiens 179-185 27825878-6 2017 By virtue of confocal fluorescence imaging, the artemisinin location in lysosome, ROS-triggered LMP and ultimate cell apoptosis can be visualized with the cathepsin B and caspase-3 activatable nanoprobe. artemisinin 48-59 cathepsin B Mus musculus 155-166 28454894-0 2017 The antimalarial drug artemisinin induces an additional, Sod1-supressible anti-mitochondrial action in yeast. artemisinin 22-33 superoxide dismutase SOD1 Saccharomyces cerevisiae S288C 57-61 28289248-5 2017 Single point mutations in the gene coding for the Kelch propeller domain of the K13 protein strongly correlate with artemisinin resistance. artemisinin 116-127 keratin 13 Homo sapiens 80-83 27825878-6 2017 By virtue of confocal fluorescence imaging, the artemisinin location in lysosome, ROS-triggered LMP and ultimate cell apoptosis can be visualized with the cathepsin B and caspase-3 activatable nanoprobe. artemisinin 48-59 caspase 3 Mus musculus 171-180 28450175-4 2017 Mutations in the propeller domain of the Kelch 13 gene (K13-propeller, PF3D71343700), such as Y493H, R539T, I543T and C580Y, were recently associated with in vivo and in vitro resistance to artemisinin in Southeast Asia. artemisinin 190-201 keratin 13 Homo sapiens 56-59 28448816-8 2017 We successfully demonstrated that treatment with high dose of artemisinin significantly decreased the elevated levels of AST (p<0.05) and ALT (p<0.01) in plasma, as well as the liver weight index (p<0.01). artemisinin 62-73 transmembrane protease, serine 11d Mus musculus 121-124 28448816-8 2017 We successfully demonstrated that treatment with high dose of artemisinin significantly decreased the elevated levels of AST (p<0.05) and ALT (p<0.01) in plasma, as well as the liver weight index (p<0.01). artemisinin 62-73 glutamic pyruvic transaminase, soluble Mus musculus 141-144 28294340-0 2017 Neuroprotective effects of artemisinin against isoflurane-induced cognitive impairments and neuronal cell death involve JNK/ERK1/2 signalling and improved hippocampal histone acetylation in neonatal rats. artemisinin 27-38 mitogen-activated protein kinase 8 Rattus norvegicus 120-123 28294340-10 2017 CONCLUSIONS: Artemisinin effectively inhibited neuronal apoptosis and improved cognition and memory via regulating histone acetylation and JNK/ERK1/2 signalling. artemisinin 13-24 mitogen-activated protein kinase 8 Rattus norvegicus 139-142 28294340-10 2017 CONCLUSIONS: Artemisinin effectively inhibited neuronal apoptosis and improved cognition and memory via regulating histone acetylation and JNK/ERK1/2 signalling. artemisinin 13-24 mitogen activated protein kinase 3 Rattus norvegicus 143-149 28294340-0 2017 Neuroprotective effects of artemisinin against isoflurane-induced cognitive impairments and neuronal cell death involve JNK/ERK1/2 signalling and improved hippocampal histone acetylation in neonatal rats. artemisinin 27-38 mitogen activated protein kinase 3 Rattus norvegicus 124-130 28294340-5 2017 Western blotting analysis revealed that treatment with artemisinin significantly enhanced the expression of anti-apoptotic proteins (Bcl-2, Bcl-xL, c-IAP-1, c-IAP-2, xIAP and survivin). artemisinin 55-66 BCL2, apoptosis regulator Rattus norvegicus 133-138 28294340-5 2017 Western blotting analysis revealed that treatment with artemisinin significantly enhanced the expression of anti-apoptotic proteins (Bcl-2, Bcl-xL, c-IAP-1, c-IAP-2, xIAP and survivin). artemisinin 55-66 Bcl2-like 1 Rattus norvegicus 140-146 28294340-5 2017 Western blotting analysis revealed that treatment with artemisinin significantly enhanced the expression of anti-apoptotic proteins (Bcl-2, Bcl-xL, c-IAP-1, c-IAP-2, xIAP and survivin). artemisinin 55-66 X-linked inhibitor of apoptosis Rattus norvegicus 166-170 28294340-6 2017 Artemisinin increased the acetylation of H3K9 and H4K12 while reducing the expression of histone deacetlyases (HDACs) - HDAC-2 and HDAC-3. artemisinin 0-11 histone deacetylase 2 Rattus norvegicus 120-126 28294340-6 2017 Artemisinin increased the acetylation of H3K9 and H4K12 while reducing the expression of histone deacetlyases (HDACs) - HDAC-2 and HDAC-3. artemisinin 0-11 histone deacetylase 3 Rattus norvegicus 131-137 28294340-7 2017 Isoflurane-induced activation of JNK signalling and downregulated ERK1/2 expression was effectively modulated by artemisinin. artemisinin 113-124 mitogen-activated protein kinase 8 Rattus norvegicus 33-36 28294340-7 2017 Isoflurane-induced activation of JNK signalling and downregulated ERK1/2 expression was effectively modulated by artemisinin. artemisinin 113-124 mitogen activated protein kinase 3 Rattus norvegicus 66-72 28438155-1 2017 BACKGROUND: In the context of national and regional goals to eliminate malaria by 2030, the Center for Malaria Parasitology and Entomology in the Lao PDR is implementing strategies to ensure all malaria cases are detected and appropriately treated with first-line artemisinin combination therapy, artemether-lumefantrine (AL). artemisinin 264-275 interleukin 4 induced 1 Homo sapiens 146-149 28249583-2 2017 After discovery of artemisinin resistance marker (K13), molecular surveillance on artemisinin resistance in endemic regions have been conducted. artemisinin 19-30 keratin 13 Homo sapiens 50-53 28361857-10 2017 The most enhanced increase of miRNA expression under artemisinin influence were found for miRNA-200b in MCF-7/DDP cells (7.1 +- 0.98 fold change), miRNA-320a in MCF-7/Dox cells (2.9 +- 0.45 fold change) and miRNA-34a (1.7 +- 0.15 fold change) in MCF-7/S cells. artemisinin 53-64 microRNA 34a Homo sapiens 207-216 28249583-6 2017 Amplification and sequence analysis of artemisinin resistance molecular markers, such as k13, pfarps10, pffd, pfmdr2, pfmrp1, pfrad5, and pfcnbp, were carried out and pfmdr1 copy number analysis was conducted by real-time PCR. artemisinin 39-50 keratin 13 Homo sapiens 89-92 28000518-0 2017 Artemisinin inhibits inflammatory response via regulating NF-kappaB and MAPK signaling pathways. artemisinin 0-11 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 58-67 28000518-0 2017 Artemisinin inhibits inflammatory response via regulating NF-kappaB and MAPK signaling pathways. artemisinin 0-11 mitogen-activated protein kinase 1 Mus musculus 72-76 28000518-4 2017 Then the artemisinin significantly inhibited the expression of NF-kappaB reporter gene induced by TNF-alpha in a dose-dependent manner. artemisinin 9-20 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 63-72 28000518-4 2017 Then the artemisinin significantly inhibited the expression of NF-kappaB reporter gene induced by TNF-alpha in a dose-dependent manner. artemisinin 9-20 tumor necrosis factor Mus musculus 98-107 28000518-5 2017 Artemisinin also inhibited TNF-alpha induced phosphorylation and degradation of IkappaBalpha, p65 nuclear translocation. artemisinin 0-11 tumor necrosis factor Mus musculus 27-36 28000518-5 2017 Artemisinin also inhibited TNF-alpha induced phosphorylation and degradation of IkappaBalpha, p65 nuclear translocation. artemisinin 0-11 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 80-92 28000518-5 2017 Artemisinin also inhibited TNF-alpha induced phosphorylation and degradation of IkappaBalpha, p65 nuclear translocation. artemisinin 0-11 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 94-97 28000518-6 2017 Artemisinin also has an impact on upstream signaling of IKK through the inhibition of expression of adaptor proteins, TNF receptor-associated factor 2 (TRAF2) and receptor interacting protein 1 (RIP1). artemisinin 0-11 TNF receptor-associated factor 2 Mus musculus 118-150 28000518-6 2017 Artemisinin also has an impact on upstream signaling of IKK through the inhibition of expression of adaptor proteins, TNF receptor-associated factor 2 (TRAF2) and receptor interacting protein 1 (RIP1). artemisinin 0-11 TNF receptor-associated factor 2 Mus musculus 152-157 28000518-6 2017 Artemisinin also has an impact on upstream signaling of IKK through the inhibition of expression of adaptor proteins, TNF receptor-associated factor 2 (TRAF2) and receptor interacting protein 1 (RIP1). artemisinin 0-11 receptor (TNFRSF)-interacting serine-threonine kinase 1 Mus musculus 163-193 28000518-6 2017 Artemisinin also has an impact on upstream signaling of IKK through the inhibition of expression of adaptor proteins, TNF receptor-associated factor 2 (TRAF2) and receptor interacting protein 1 (RIP1). artemisinin 0-11 receptor (TNFRSF)-interacting serine-threonine kinase 1 Mus musculus 195-199 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 tumor necrosis factor Mus musculus 66-75 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 98-107 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 baculoviral IAP repeat-containing 3 Mus musculus 146-152 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 B cell leukemia/lymphoma 2 Mus musculus 154-159 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 cytochrome c oxidase II, mitochondrial Mus musculus 187-192 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 cyclin D1 Mus musculus 194-202 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 matrix metallopeptidase 9 Mus musculus 215-220 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 vascular endothelial growth factor A Mus musculus 237-241 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 tumor necrosis factor Mus musculus 278-287 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 nitric oxide synthase 2, inducible Mus musculus 289-293 28000518-7 2017 Furthermore, pretreatment of cells with artemisinin prevented the TNF-alpha-induced expression of NF-kappaB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-alpha, iNOS, and MCP1). artemisinin 40-51 mast cell protease 1 Mus musculus 299-303 28000518-8 2017 We also proved that artemisinin potentiated TNF-alpha-induced apoptosis. artemisinin 20-31 tumor necrosis factor Mus musculus 44-53 28000518-9 2017 Moreover, artemisinin significantly impaired the ROS production and phosphorylation of p38 and ERK, but did not affect the phosphorylation of JNK. artemisinin 10-21 mitogen-activated protein kinase 14 Mus musculus 87-90 28000518-9 2017 Moreover, artemisinin significantly impaired the ROS production and phosphorylation of p38 and ERK, but did not affect the phosphorylation of JNK. artemisinin 10-21 mitogen-activated protein kinase 1 Mus musculus 95-98 27515812-0 2016 Efficacy of artemisinin-based combination therapies for the treatment of falciparum malaria in Pakistan (2007-2015): In vivo response and dhfr and dhps mutations. artemisinin 12-23 deoxyhypusine synthase Homo sapiens 147-151 27736063-1 2016 A series of novel Smo antagonists were developed either by directly incorporating the basic skeleton of the natural product artemisinin or by first breaking artemisinin into structurally simpler and stable intermediates and then reconstructing into diversified heterocyclic derivatives, equipped with a Smo-targeting bullet. artemisinin 124-135 smoothened, frizzled class receptor Homo sapiens 18-21 27736063-1 2016 A series of novel Smo antagonists were developed either by directly incorporating the basic skeleton of the natural product artemisinin or by first breaking artemisinin into structurally simpler and stable intermediates and then reconstructing into diversified heterocyclic derivatives, equipped with a Smo-targeting bullet. artemisinin 157-168 smoothened, frizzled class receptor Homo sapiens 18-21 27660233-0 2016 Role of K13 Mutations in Artemisinin-Based Combination Therapy. artemisinin 25-36 keratin 13 Homo sapiens 8-11 27515812-0 2016 Efficacy of artemisinin-based combination therapies for the treatment of falciparum malaria in Pakistan (2007-2015): In vivo response and dhfr and dhps mutations. artemisinin 12-23 dihydrofolate reductase Homo sapiens 138-142 27732639-10 2016 For canine CAR3, 50 muM artemisinin proved to be the best activator (7.3 +- 1.8 and 10.5 +- 2.2 fold) while clotrimazole (10 muM) was the primary activator of the rat variant (13.7 +- 0.8 and 26.9 +- 1.3 fold). artemisinin 24-35 carbonic anhydrase 3 Rattus norvegicus 11-15 27372058-0 2016 Artemisinin protects human retinal pigment epithelial cells from hydrogen peroxide-induced oxidative damage through activation of ERK/CREB signaling. artemisinin 0-11 mitogen-activated protein kinase 1 Homo sapiens 130-133 27012321-1 2016 In this study, we used a self-contrast method, which excluded the individual difference, to evaluate the inhibitory effect of chrysosplentin (CHR) in the presence or absence of artemisinin (ART) on the P-glycoprotein (P-gp) transport activity. artemisinin 190-193 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 202-216 27372058-0 2016 Artemisinin protects human retinal pigment epithelial cells from hydrogen peroxide-induced oxidative damage through activation of ERK/CREB signaling. artemisinin 0-11 cAMP responsive element binding protein 1 Homo sapiens 134-138 27372058-5 2016 Western blotting analysis showed that Artemisinin was able to activate extracellular regulated ERK/CREB survival signaling. artemisinin 38-49 mitogen-activated protein kinase 1 Homo sapiens 95-98 27372058-5 2016 Western blotting analysis showed that Artemisinin was able to activate extracellular regulated ERK/CREB survival signaling. artemisinin 38-49 cAMP responsive element binding protein 1 Homo sapiens 99-103 27372058-7 2016 Taken together, these results suggest that Artemisinin is a potential protectant with the pro-survival effects against H2O2 insult through activation of the ERK/CREB pathway. artemisinin 43-54 mitogen-activated protein kinase 1 Homo sapiens 157-160 27372058-7 2016 Taken together, these results suggest that Artemisinin is a potential protectant with the pro-survival effects against H2O2 insult through activation of the ERK/CREB pathway. artemisinin 43-54 cAMP responsive element binding protein 1 Homo sapiens 161-165 27039397-0 2016 25-methoxyl-dammarane-3beta, 12beta, 20-triol and artemisinin synergistically inhibit MDA-MB-231 cell proliferation through downregulation of testes-specific protease 50 (TSP50) expression. artemisinin 50-61 serine protease 50 Homo sapiens 142-169 27590543-5 2016 The relative standard deviation is 4.83% for the determination of 10 muM artemisinin. artemisinin 73-84 latexin Homo sapiens 69-72 27713827-7 2016 Induction of CYP3A4 by PB, artemisinin, and phenytoin was also much reduced in PXR-KO cells, while the response to CITCO was maintained. artemisinin 27-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 27713827-7 2016 Induction of CYP3A4 by PB, artemisinin, and phenytoin was also much reduced in PXR-KO cells, while the response to CITCO was maintained. artemisinin 27-38 nuclear receptor subfamily 1 group I member 2 Homo sapiens 79-82 27313266-2 2016 The role of Plasmodium falciparum K13 mutations (a marker of artemisinin resistance) in reducing treatment efficacy remains controversial. artemisinin 61-72 keratin 13 Homo sapiens 34-37 27313266-5 2016 K13 mutations conferring moderate artemisinin resistance (notably E252Q) predominated initially but were later overtaken by propeller mutations associated with slower parasite clearance (notably C580Y). artemisinin 34-45 keratin 13 Homo sapiens 0-3 27313266-10 2016 CONCLUSIONS: The increasing prevalence of K13 mutations was the decisive factor for the recent and rapid decline in efficacy of artemisinin-based combination (MAS3) on the Thailand-Myanmar border. artemisinin 128-139 keratin 13 Homo sapiens 42-45 27039397-0 2016 25-methoxyl-dammarane-3beta, 12beta, 20-triol and artemisinin synergistically inhibit MDA-MB-231 cell proliferation through downregulation of testes-specific protease 50 (TSP50) expression. artemisinin 50-61 serine protease 50 Homo sapiens 171-176 27039397-5 2016 We identified the compound 7P3A, which consists of 70 % 25-methoxyl-dammarane-3beta, 12beta, 20-triol and 30 % artemisinin, as being capable of inhibiting the TSP50-3"-UTR reporter activity, as well as the expression of TSP50. artemisinin 111-122 serine protease 50 Homo sapiens 159-164 27480521-4 2016 We found that artesunate (ART), a small molecular derivative of artemisinin, had a significant inhibitory effect on ocular NV by downregulating the expression of VEGFR2, PKCalpha, and PDGFR. artemisinin 64-75 kinase insert domain receptor Homo sapiens 162-168 26999297-0 2016 Enhanced Efficacy of Artemisinin Loaded in Transferrin-Conjugated Liposomes versus Stealth Liposomes against HCT-8 Colon Cancer Cells. artemisinin 21-32 transferrin Homo sapiens 43-54 27385212-0 2016 Halofuginone and artemisinin synergistically arrest cancer cells at the G1/G0 phase by upregulating p21Cip1 and p27Kip1. artemisinin 17-28 cyclin dependent kinase inhibitor 1A Homo sapiens 100-107 27385212-0 2016 Halofuginone and artemisinin synergistically arrest cancer cells at the G1/G0 phase by upregulating p21Cip1 and p27Kip1. artemisinin 17-28 cyclin dependent kinase inhibitor 1B Homo sapiens 112-119 27480521-4 2016 We found that artesunate (ART), a small molecular derivative of artemisinin, had a significant inhibitory effect on ocular NV by downregulating the expression of VEGFR2, PKCalpha, and PDGFR. artemisinin 64-75 protein kinase C alpha Homo sapiens 170-178 27480521-4 2016 We found that artesunate (ART), a small molecular derivative of artemisinin, had a significant inhibitory effect on ocular NV by downregulating the expression of VEGFR2, PKCalpha, and PDGFR. artemisinin 64-75 platelet derived growth factor receptor beta Homo sapiens 184-189 25789847-4 2016 Real-time PCR and western blot analysis of extracted RNA and total protein revealed artemsinin and artesunate increased miR-34a expression in a dose-dependent manner correlating with down-regulation of the miR-34a target gene, CDK4. artemisinin 84-94 microRNA 34a Homo sapiens 120-127 27242266-0 2016 Artemisinin conferred ERK mediated neuroprotection to PC12 cells and cortical neurons exposed to sodium nitroprusside-induced oxidative insult. artemisinin 0-11 Eph receptor B1 Rattus norvegicus 22-25 27242266-3 2016 Pretreatment of PC12 cells with artemisinin significantly suppressed SNP-induced cell death by decreasing the extent of oxidation, preventing the decline of mitochondrial membrane potential, restoring abnormal changes in nuclear morphology and reducing lactate dehydrogenase release and inhibiting caspase 3/7 activities. artemisinin 32-43 caspase 3 Rattus norvegicus 298-307 27242266-4 2016 Western blotting analysis revealed that artemisinin was able to activate extracellular regulated protein kinases (ERK) pathway. artemisinin 40-51 Eph receptor B1 Rattus norvegicus 73-112 27242266-4 2016 Western blotting analysis revealed that artemisinin was able to activate extracellular regulated protein kinases (ERK) pathway. artemisinin 40-51 Eph receptor B1 Rattus norvegicus 114-117 27242266-5 2016 Furthermore, the ERK inhibitor PD98059 blocked the neuroprotective effect of artemisinin whereas the PI3K inhibitor LY294002 had no effect. artemisinin 77-88 Eph receptor B1 Rattus norvegicus 17-20 27242266-6 2016 Cumulatively these findings support the notion that artemisinin confers neuroprotection from SNP-induce neuronal cell death insult, a phenomenon coincidentally related to activation of ERK phosphorylation. artemisinin 52-63 Eph receptor B1 Rattus norvegicus 185-188 27337450-6 2016 Specifically, these studies have established that artemisinin resistance manifests as slow parasite clearance in patients and increased survival of early-ring-stage parasites in vitro; is caused by single nucleotide polymorphisms in the parasite"s K13 gene, is associated with an upregulated "unfolded protein response" pathway that may antagonize the pro-oxidant activity of artemisinins, and selects for partner drug resistance that rapidly leads to ACT failures. artemisinin 50-61 keratin 13 Homo sapiens 248-251 26830216-0 2016 Artemisinin protects mice against burn sepsis through inhibiting NLRP3 inflammasome activation. artemisinin 0-11 NLR family, pyrin domain containing 3 Mus musculus 65-70 26830216-13 2016 Artemisinin down-regulated protein levels of NLRP3 and caspase 1 and inhibited the increases of IL-1beta and IL-18 messenger RNA expression from Raw 264.7 cells that were stimulated with burn sepsis serum. artemisinin 0-11 NLR family, pyrin domain containing 3 Mus musculus 45-50 26830216-13 2016 Artemisinin down-regulated protein levels of NLRP3 and caspase 1 and inhibited the increases of IL-1beta and IL-18 messenger RNA expression from Raw 264.7 cells that were stimulated with burn sepsis serum. artemisinin 0-11 caspase 1 Mus musculus 55-64 26830216-13 2016 Artemisinin down-regulated protein levels of NLRP3 and caspase 1 and inhibited the increases of IL-1beta and IL-18 messenger RNA expression from Raw 264.7 cells that were stimulated with burn sepsis serum. artemisinin 0-11 interleukin 1 beta Mus musculus 96-104 26830216-13 2016 Artemisinin down-regulated protein levels of NLRP3 and caspase 1 and inhibited the increases of IL-1beta and IL-18 messenger RNA expression from Raw 264.7 cells that were stimulated with burn sepsis serum. artemisinin 0-11 interleukin 18 Mus musculus 109-114 25789847-5 2016 I3C stimulation of miR-34a expression required functional p53, whereas, both artemisinin and artesunate up-regulated miR-34a expression regardless of p53 mutational status or in the presence of dominant negative p53. artemisinin 77-88 microRNA 34a Homo sapiens 117-124 25789847-7 2016 Our results suggest that miR-34a is an essential component of the anti-proliferative activities of I3C, artemisinin, and artesunate and demonstrate that both wild-type p53 dependent and independent pathways are responsible for miR-34a induction. artemisinin 104-115 microRNA 34a Homo sapiens 25-32 27434054-3 2016 Eight of 20 patients with uncomplicated malaria who were given oral artemisinin-based combination therapy met the definition of posttreatment hemolysis (low haptoglobin level and increased lactate dehydrogenase level on day 14). artemisinin 68-79 haptoglobin Homo sapiens 157-168 27122190-0 2016 Artemisinin inhibits monocyte adhesion to HUVECs through the NF-kappaB and MAPK pathways in vitro. artemisinin 0-11 nuclear factor kappa B subunit 1 Homo sapiens 61-70 27122190-4 2016 In the present study, we found that artemisinin significantly decreased the adhesion of monocytes to tumor necrosis factor-alpha (TNF-alpha)-stimulated HUVECs in a dose-dependent manner and suppressed the mRNA and protein level of ICAM-1 and VCAM-1 in the TNF-alpha-stimulated HUVECs. artemisinin 36-47 tumor necrosis factor Homo sapiens 101-128 27122190-4 2016 In the present study, we found that artemisinin significantly decreased the adhesion of monocytes to tumor necrosis factor-alpha (TNF-alpha)-stimulated HUVECs in a dose-dependent manner and suppressed the mRNA and protein level of ICAM-1 and VCAM-1 in the TNF-alpha-stimulated HUVECs. artemisinin 36-47 tumor necrosis factor Homo sapiens 130-139 27122190-4 2016 In the present study, we found that artemisinin significantly decreased the adhesion of monocytes to tumor necrosis factor-alpha (TNF-alpha)-stimulated HUVECs in a dose-dependent manner and suppressed the mRNA and protein level of ICAM-1 and VCAM-1 in the TNF-alpha-stimulated HUVECs. artemisinin 36-47 intercellular adhesion molecule 1 Homo sapiens 231-237 27122190-4 2016 In the present study, we found that artemisinin significantly decreased the adhesion of monocytes to tumor necrosis factor-alpha (TNF-alpha)-stimulated HUVECs in a dose-dependent manner and suppressed the mRNA and protein level of ICAM-1 and VCAM-1 in the TNF-alpha-stimulated HUVECs. artemisinin 36-47 vascular cell adhesion molecule 1 Homo sapiens 242-248 27122190-4 2016 In the present study, we found that artemisinin significantly decreased the adhesion of monocytes to tumor necrosis factor-alpha (TNF-alpha)-stimulated HUVECs in a dose-dependent manner and suppressed the mRNA and protein level of ICAM-1 and VCAM-1 in the TNF-alpha-stimulated HUVECs. artemisinin 36-47 tumor necrosis factor Homo sapiens 256-265 27122190-6 2016 Moreover, artemisinin impeded the activation of the NF-kappaB and MAPK signaling pathways. artemisinin 10-21 nuclear factor kappa B subunit 1 Homo sapiens 52-61 27122190-8 2016 Taken together, the findings of our study indicated that artemisinin blocked monocyte adhesion to TNF-alpha-stimulated to HUVECs by downregulating ICAM-1 and VCAM-1 expression in the TNF-alpha-stimulated HUVECs. artemisinin 57-68 tumor necrosis factor Homo sapiens 98-107 27122190-8 2016 Taken together, the findings of our study indicated that artemisinin blocked monocyte adhesion to TNF-alpha-stimulated to HUVECs by downregulating ICAM-1 and VCAM-1 expression in the TNF-alpha-stimulated HUVECs. artemisinin 57-68 intercellular adhesion molecule 1 Homo sapiens 147-153 27122190-8 2016 Taken together, the findings of our study indicated that artemisinin blocked monocyte adhesion to TNF-alpha-stimulated to HUVECs by downregulating ICAM-1 and VCAM-1 expression in the TNF-alpha-stimulated HUVECs. artemisinin 57-68 vascular cell adhesion molecule 1 Homo sapiens 158-164 27122190-8 2016 Taken together, the findings of our study indicated that artemisinin blocked monocyte adhesion to TNF-alpha-stimulated to HUVECs by downregulating ICAM-1 and VCAM-1 expression in the TNF-alpha-stimulated HUVECs. artemisinin 57-68 tumor necrosis factor Homo sapiens 183-192 27119499-0 2016 Artemisinin and its derivatives can significantly inhibit lung tumorigenesis and tumor metastasis through Wnt/beta-catenin signaling. artemisinin 0-11 catenin beta 1 Homo sapiens 110-122 26830216-15 2016 CONCLUSION: Artemisinin protects mice from burn sepsis by attenuating the inflammatory response and alleviating inflammatory infiltration in vital organs, likely through inhibiting the activation of NLRP3 inflammasome. artemisinin 12-23 NLR family, pyrin domain containing 3 Mus musculus 199-204 25789847-4 2016 Real-time PCR and western blot analysis of extracted RNA and total protein revealed artemsinin and artesunate increased miR-34a expression in a dose-dependent manner correlating with down-regulation of the miR-34a target gene, CDK4. artemisinin 84-94 cyclin dependent kinase 4 Homo sapiens 227-231 25789847-4 2016 Real-time PCR and western blot analysis of extracted RNA and total protein revealed artemsinin and artesunate increased miR-34a expression in a dose-dependent manner correlating with down-regulation of the miR-34a target gene, CDK4. artemisinin 84-94 microRNA 34a Homo sapiens 206-213 27035431-7 2016 Western blot analysis indicated that artemisinin induced the expression of p16, while down-regulating phosphorylated extracellular signal-regulated kinase (ERK)1/2, CDK4 and cyclin D1 expression, leading to inhibition of the ERK1/2 pathway. artemisinin 37-48 mitogen-activated protein kinase 3 Homo sapiens 225-231 27035431-8 2016 Furthermore, flow cytometry and western blot analysis showed that artemisinin caused G1-phase arrest of the cell cycle, promoted the generation of reactive oxygen species (ROS), led to a collapse of the mitochondrial membrane potential and to triggered cytochrome c release from the mitochondria into the cytoplasm, which finally activated caspase-3-mediated apoptosis. artemisinin 66-77 cytochrome c, somatic Homo sapiens 253-265 27035431-7 2016 Western blot analysis indicated that artemisinin induced the expression of p16, while down-regulating phosphorylated extracellular signal-regulated kinase (ERK)1/2, CDK4 and cyclin D1 expression, leading to inhibition of the ERK1/2 pathway. artemisinin 37-48 cyclin dependent kinase inhibitor 2A Homo sapiens 75-78 27035431-8 2016 Furthermore, flow cytometry and western blot analysis showed that artemisinin caused G1-phase arrest of the cell cycle, promoted the generation of reactive oxygen species (ROS), led to a collapse of the mitochondrial membrane potential and to triggered cytochrome c release from the mitochondria into the cytoplasm, which finally activated caspase-3-mediated apoptosis. artemisinin 66-77 caspase 3 Homo sapiens 340-349 27035431-7 2016 Western blot analysis indicated that artemisinin induced the expression of p16, while down-regulating phosphorylated extracellular signal-regulated kinase (ERK)1/2, CDK4 and cyclin D1 expression, leading to inhibition of the ERK1/2 pathway. artemisinin 37-48 mitogen-activated protein kinase 1 Homo sapiens 117-163 26518641-5 2015 The dosage of 5 ppm of artemisinin improved the WG and FCR for the chickens infected with E. acervulina. artemisinin 23-34 FCR Gallus gallus 55-58 27035431-7 2016 Western blot analysis indicated that artemisinin induced the expression of p16, while down-regulating phosphorylated extracellular signal-regulated kinase (ERK)1/2, CDK4 and cyclin D1 expression, leading to inhibition of the ERK1/2 pathway. artemisinin 37-48 cyclin dependent kinase 4 Homo sapiens 165-169 27035431-7 2016 Western blot analysis indicated that artemisinin induced the expression of p16, while down-regulating phosphorylated extracellular signal-regulated kinase (ERK)1/2, CDK4 and cyclin D1 expression, leading to inhibition of the ERK1/2 pathway. artemisinin 37-48 cyclin D1 Homo sapiens 174-183 26932893-10 2016 Cytochrome C was released from mitochondria to cytoplasm leading to the activation of caspase-3 and PARP after dealt with artemisinin(P<0.05). artemisinin 122-133 cytochrome c, somatic Homo sapiens 0-12 26932893-10 2016 Cytochrome C was released from mitochondria to cytoplasm leading to the activation of caspase-3 and PARP after dealt with artemisinin(P<0.05). artemisinin 122-133 caspase 3 Homo sapiens 86-95 26932893-10 2016 Cytochrome C was released from mitochondria to cytoplasm leading to the activation of caspase-3 and PARP after dealt with artemisinin(P<0.05). artemisinin 122-133 collagen type XI alpha 2 chain Homo sapiens 100-104 26932893-11 2016 CONCLUSION: The inhibition effect of artemisinin on the proliferation gallbladder cancer cells is accompanied by down-regulation of ERK1/2 signaling pathway, G1 phase arrest and triggering caspase-3-mediate apoptosis. artemisinin 37-48 mitogen-activated protein kinase 3 Homo sapiens 132-138 26932893-11 2016 CONCLUSION: The inhibition effect of artemisinin on the proliferation gallbladder cancer cells is accompanied by down-regulation of ERK1/2 signaling pathway, G1 phase arrest and triggering caspase-3-mediate apoptosis. artemisinin 37-48 caspase 3 Homo sapiens 189-198 26932893-9 2016 In addition, expression of p16 was increased, and expressions of p-ERK1/2, CDK4 and cyclin D1 were down-regulated by artemisinin(all P<0.05). artemisinin 117-128 cyclin dependent kinase inhibitor 2A Homo sapiens 27-30 26932893-9 2016 In addition, expression of p16 was increased, and expressions of p-ERK1/2, CDK4 and cyclin D1 were down-regulated by artemisinin(all P<0.05). artemisinin 117-128 mitogen-activated protein kinase 3 Homo sapiens 67-73 26932893-9 2016 In addition, expression of p16 was increased, and expressions of p-ERK1/2, CDK4 and cyclin D1 were down-regulated by artemisinin(all P<0.05). artemisinin 117-128 cyclin dependent kinase 4 Homo sapiens 75-79 26932893-9 2016 In addition, expression of p16 was increased, and expressions of p-ERK1/2, CDK4 and cyclin D1 were down-regulated by artemisinin(all P<0.05). artemisinin 117-128 cyclin D1 Homo sapiens 84-93 26655404-13 2015 The high expression of MYC, TFR, and VEGFC in the patient biopsy corresponded with high expression of these markers in the artemisinin-sensitive PC-3 cells compared to artemisinin-resistant DU-145 cells. artemisinin 123-134 MYC proto-oncogene, bHLH transcription factor Homo sapiens 23-26 26655404-13 2015 The high expression of MYC, TFR, and VEGFC in the patient biopsy corresponded with high expression of these markers in the artemisinin-sensitive PC-3 cells compared to artemisinin-resistant DU-145 cells. artemisinin 123-134 transferrin receptor Homo sapiens 28-31 26655404-13 2015 The high expression of MYC, TFR, and VEGFC in the patient biopsy corresponded with high expression of these markers in the artemisinin-sensitive PC-3 cells compared to artemisinin-resistant DU-145 cells. artemisinin 123-134 vascular endothelial growth factor C Homo sapiens 37-42 26655404-13 2015 The high expression of MYC, TFR, and VEGFC in the patient biopsy corresponded with high expression of these markers in the artemisinin-sensitive PC-3 cells compared to artemisinin-resistant DU-145 cells. artemisinin 168-179 MYC proto-oncogene, bHLH transcription factor Homo sapiens 23-26 26655404-13 2015 The high expression of MYC, TFR, and VEGFC in the patient biopsy corresponded with high expression of these markers in the artemisinin-sensitive PC-3 cells compared to artemisinin-resistant DU-145 cells. artemisinin 168-179 transferrin receptor Homo sapiens 28-31 26655404-13 2015 The high expression of MYC, TFR, and VEGFC in the patient biopsy corresponded with high expression of these markers in the artemisinin-sensitive PC-3 cells compared to artemisinin-resistant DU-145 cells. artemisinin 168-179 vascular endothelial growth factor C Homo sapiens 37-42 26084957-0 2015 Stereoselective chlorothiolation of artemisinin-derived C-10 oxa terminal alkynes. artemisinin 36-47 homeobox C10 Homo sapiens 56-60 26695060-9 2015 Artemisinin-resistant K13 mutants with ex vivo PSA survival rates >=10% were associated with 32-fold higher risk of recrudescence (95% CI, 4.5-224; P = 0.0005). artemisinin 0-11 keratin 13 Homo sapiens 22-25 26695060-11 2015 Combined ex vivo PSA and K13 genotyping provides a convenient monitor for both artemisinin and piperaquine resistance where dihydroartemisinin-piperaquine is used. artemisinin 79-90 keratin 13 Homo sapiens 25-28 31157126-1 2015 Objective: To determine if oral artemisinin is safe and has a short-term effect on prostate specific antigen (PSA) kinetics in patients with prostate cancer (CaP). artemisinin 32-43 kallikrein related peptidase 3 Homo sapiens 83-108 26084957-1 2015 A mild and efficient strategy is explored on the highly sensitive artemisinin-derived C-10 oxa terminal alkynes. artemisinin 66-77 homeobox C10 Homo sapiens 86-90 25862844-0 2015 Quantitative structure-activity relationship and molecular docking of artemisinin derivatives to vascular endothelial growth factor receptor 1. artemisinin 70-81 fms related receptor tyrosine kinase 1 Homo sapiens 97-142 25936561-8 2015 The protection was associated with a Th1-biased immune response as evident from a positive delayed-type hypersensitivity reaction, escalated IgG2a levels, augmented lymphoproliferation and enhancement in proinflammatory cytokines (IFN-gamma and IL-2) with significant suppression of Th2 cytokines (IL-10 and IL-4) after in vitro recall, compared to infected control and free artemisinin treatment. artemisinin 375-386 negative elongation factor complex member C/D, Th1l Mus musculus 37-40 26237549-3 2015 RECENT FINDINGS: Multiple international collaborations have established that artemisinin resistance is associated with slow parasite clearance in patients, increased survival of early-ring-stage parasites in vitro, single-nucleotide polymorphisms (SNPs) in the parasite"s kelch protein gene (K13), parasite "founder" populations sharing a genetic background of four additional SNPs, parasite transcriptional profiles reflecting an "unfolded protein response" and decelerated parasite development, and elevated parasite phosphatidylinositol-3-kinase activity. artemisinin 77-88 keratin 13 Homo sapiens 292-295 26390866-7 2015 In areas with artemisinin-sensitive parasites, the median PC1/2 following three-day artesunate treatment (4 mg/kg/day) ranged from 1.8 to 3.0 h and the proportion of patients with PC1/2 >5 h from 0 to 10 %. artemisinin 14-25 polycystin 1, transient receptor potential channel interacting Homo sapiens 58-63 26390866-7 2015 In areas with artemisinin-sensitive parasites, the median PC1/2 following three-day artesunate treatment (4 mg/kg/day) ranged from 1.8 to 3.0 h and the proportion of patients with PC1/2 >5 h from 0 to 10 %. artemisinin 14-25 polycystin 1, transient receptor potential channel interacting Homo sapiens 180-185 26390866-13 2015 As substantial heterogeneity in parasite clearance exists between locations, early detection of artemisinin resistance requires reference PC1/2 data. artemisinin 96-107 polycystin 1, transient receptor potential channel interacting Homo sapiens 138-143 25755006-2 2015 In the present study, we have investigated the anticancer effects of artemisinin in human cervical cancer cells, with special emphasis on its role in inducing apoptosis and repressing cell proliferation by inhibiting the telomerase subunits, ERalpha which is essential for maintenance of the cervix, and downstream components like VEGF, which is known to activate angiogenesis. artemisinin 69-80 estrogen receptor 1 Homo sapiens 242-249 25755006-2 2015 In the present study, we have investigated the anticancer effects of artemisinin in human cervical cancer cells, with special emphasis on its role in inducing apoptosis and repressing cell proliferation by inhibiting the telomerase subunits, ERalpha which is essential for maintenance of the cervix, and downstream components like VEGF, which is known to activate angiogenesis. artemisinin 69-80 vascular endothelial growth factor A Homo sapiens 331-335 25755006-5 2015 Our findings demonstrated that artemisinin significantly downregulated the expression of ERalpha and its downstream component, VEGF. artemisinin 31-42 estrogen receptor 1 Homo sapiens 89-96 25755006-5 2015 Our findings demonstrated that artemisinin significantly downregulated the expression of ERalpha and its downstream component, VEGF. artemisinin 31-42 vascular endothelial growth factor A Homo sapiens 127-131 25755006-8 2015 Artemisinin-induced apoptosis was confirmed by FACS, nuclear chromatin condensation, annexin V staining. artemisinin 0-11 annexin A5 Homo sapiens 85-94 25755006-9 2015 Increased expression of p53 with concomitant decrease in expression of the p53 inhibitor Mdm2 further supported that artemisinin-induced apoptosis was p53-dependent. artemisinin 117-128 tumor protein p53 Homo sapiens 24-27 25755006-9 2015 Increased expression of p53 with concomitant decrease in expression of the p53 inhibitor Mdm2 further supported that artemisinin-induced apoptosis was p53-dependent. artemisinin 117-128 tumor protein p53 Homo sapiens 75-78 25755006-9 2015 Increased expression of p53 with concomitant decrease in expression of the p53 inhibitor Mdm2 further supported that artemisinin-induced apoptosis was p53-dependent. artemisinin 117-128 MDM2 proto-oncogene Homo sapiens 89-93 25755006-9 2015 Increased expression of p53 with concomitant decrease in expression of the p53 inhibitor Mdm2 further supported that artemisinin-induced apoptosis was p53-dependent. artemisinin 117-128 tumor protein p53 Homo sapiens 75-78 25897065-4 2015 All 1,737 ACAs contained the labeled artemisinin derivative, with 4.1% being outside the 85-115% artemisinin API range defined as acceptable quality. artemisinin 37-48 ALL1 Homo sapiens 0-5 25707499-1 2015 The aim of this study was to evaluate the effect of artemisinin (ART) on rat vascular smooth muscle cell (VSMC) proliferation induced by tumour necrosis factor (TNF)-alpha, cell cycle arrest, and apoptosis, and its effect on neointima formation after balloon injury of rat carotid artery. artemisinin 52-63 tumor necrosis factor Rattus norvegicus 137-171 25707499-8 2015 The results suggest that ART can effectively inhibit the proliferation of VSMC induced by TNF-alpha through the apoptotic induction pathway and cell cycle arrest. artemisinin 25-28 tumor necrosis factor Rattus norvegicus 90-99 26109756-8 2015 At 2 h after treatment, the intravenous administration of artemisinin and transferrin-loaded magnetic nanoliposomes followed using the magnetic field approximately produced 10- and 5.5-fold higher levels of artemisinin and transferrin in the tumors, respectively, compared with free artemisinin and transferrin. artemisinin 58-69 transferrin Mus musculus 223-234 26109756-8 2015 At 2 h after treatment, the intravenous administration of artemisinin and transferrin-loaded magnetic nanoliposomes followed using the magnetic field approximately produced 10- and 5.5-fold higher levels of artemisinin and transferrin in the tumors, respectively, compared with free artemisinin and transferrin. artemisinin 58-69 transferrin Mus musculus 223-234 26109756-8 2015 At 2 h after treatment, the intravenous administration of artemisinin and transferrin-loaded magnetic nanoliposomes followed using the magnetic field approximately produced 10- and 5.5-fold higher levels of artemisinin and transferrin in the tumors, respectively, compared with free artemisinin and transferrin. artemisinin 207-218 transferrin Mus musculus 74-85 26109756-8 2015 At 2 h after treatment, the intravenous administration of artemisinin and transferrin-loaded magnetic nanoliposomes followed using the magnetic field approximately produced 10- and 5.5-fold higher levels of artemisinin and transferrin in the tumors, respectively, compared with free artemisinin and transferrin. artemisinin 207-218 transferrin Mus musculus 74-85 25836766-3 2015 A total of 417 patient samples from the year 2007, collected during malaria surveillance studies across ten provinces in Thailand, were genotyped for the candidate Plasmodium falciparum molecular marker of artemisinin resistance K13. artemisinin 206-217 keratin 13 Homo sapiens 229-232 25836766-7 2015 The most prevalent artemisinin resistance-associated K13 mutation, C580Y, carried two distinct haplotype profiles that were separated based on geography, along the Thai-Cambodia and Thai-Myanmar borders. artemisinin 19-30 keratin 13 Homo sapiens 53-56 25836766-9 2015 In summary, parasites with K13 propeller mutations associated with artemisinin resistance were widely present along the Thai-Cambodia and Thai-Myanmar borders prior to the implementation of the artemisinin resistance containment project in the region. artemisinin 67-78 keratin 13 Homo sapiens 27-30 25836766-9 2015 In summary, parasites with K13 propeller mutations associated with artemisinin resistance were widely present along the Thai-Cambodia and Thai-Myanmar borders prior to the implementation of the artemisinin resistance containment project in the region. artemisinin 194-205 keratin 13 Homo sapiens 27-30 25862844-3 2015 MATERIAL AND METHODS: We performed molecular docking of 52 artemisinin derivatives to vascular endothelial growth factor receptors (VEGFR1, VEGFR2), and VEGFA ligand using Autodock4 and AutodockTools-1.5.7.rc1 using the Lamarckian genetic algorithm. artemisinin 59-70 fms related receptor tyrosine kinase 1 Homo sapiens 132-138 25862844-3 2015 MATERIAL AND METHODS: We performed molecular docking of 52 artemisinin derivatives to vascular endothelial growth factor receptors (VEGFR1, VEGFR2), and VEGFA ligand using Autodock4 and AutodockTools-1.5.7.rc1 using the Lamarckian genetic algorithm. artemisinin 59-70 kinase insert domain receptor Homo sapiens 140-146 25862844-5 2015 RESULTS: A statistically significant inverse relationship was obtained between in silico binding energies to VEGFR1 and anti-angiogenic activity in vivo of a test-set of artemisinin derivatives (R=-0.843; p=0.035). artemisinin 170-181 fms related receptor tyrosine kinase 1 Homo sapiens 109-115 25862844-7 2015 Furthermore, 52 artemisinin derivatives were docked to VEGFR1 and in selected examples also to VEGFR2 and VEGFA. artemisinin 16-27 fms related receptor tyrosine kinase 1 Homo sapiens 55-61 25862844-7 2015 Furthermore, 52 artemisinin derivatives were docked to VEGFR1 and in selected examples also to VEGFR2 and VEGFA. artemisinin 16-27 kinase insert domain receptor Homo sapiens 95-101 25862844-7 2015 Furthermore, 52 artemisinin derivatives were docked to VEGFR1 and in selected examples also to VEGFR2 and VEGFA. artemisinin 16-27 vascular endothelial growth factor A Homo sapiens 106-111 25862844-9 2015 The best binding affinities to VEGFR1 were found for an artemisinin dimer, 10-dihydroartemisinyl-2-propylpentanoate, and dihydroartemisinin alpha-hemisuccinate sodium salt. artemisinin 56-67 fms related receptor tyrosine kinase 1 Homo sapiens 31-37 25584867-5 2015 Furthermore, artemisinin induced apoptosis in erythrocytes in zebrafish embryos, as assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling assay, and preferentially acted on differentiated erythrocytes by elevating caspase 8 and caspase 9 activity in differentiated human K562 cells. artemisinin 13-24 caspase 8, apoptosis-related cysteine peptidase Danio rerio 233-242 25584867-5 2015 Furthermore, artemisinin induced apoptosis in erythrocytes in zebrafish embryos, as assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling assay, and preferentially acted on differentiated erythrocytes by elevating caspase 8 and caspase 9 activity in differentiated human K562 cells. artemisinin 13-24 caspase 9, apoptosis-related cysteine peptidase Danio rerio 247-256 25584867-4 2015 RNA-Seq revealed that artemisinin suppressed a cluster of genes in the heme biosynthesis and globin synthesis pathways. artemisinin 22-33 hemoglobin alpha embryonic-3 Danio rerio 93-99 25502316-3 2015 Polymorphisms in the kelch domain-carrying protein K13 are associated with artemisinin resistance, but the underlying molecular mechanisms are unknown. artemisinin 75-86 keratin 13 Homo sapiens 51-54 25575733-0 2015 Artemisinin inhibits the proliferation, migration, and inflammatory reaction induced by tumor necrosis factor-alpha in vascular smooth muscle cells through nuclear factor kappa B pathway. artemisinin 0-11 tumor necrosis factor Rattus norvegicus 88-115 25575733-2 2015 The aim of this study was to evaluate the effect of artemisinin (ART) on the proliferation, migration, and inflammation induced by tumor necrosis factor-alpha (TNF-alpha) of rat vascular smooth muscle cells (VSMCs). artemisinin 52-63 tumor necrosis factor Rattus norvegicus 131-158 25575733-2 2015 The aim of this study was to evaluate the effect of artemisinin (ART) on the proliferation, migration, and inflammation induced by tumor necrosis factor-alpha (TNF-alpha) of rat vascular smooth muscle cells (VSMCs). artemisinin 52-63 tumor necrosis factor Rattus norvegicus 160-169 25575733-2 2015 The aim of this study was to evaluate the effect of artemisinin (ART) on the proliferation, migration, and inflammation induced by tumor necrosis factor-alpha (TNF-alpha) of rat vascular smooth muscle cells (VSMCs). artemisinin 65-68 tumor necrosis factor Rattus norvegicus 131-158 25575733-2 2015 The aim of this study was to evaluate the effect of artemisinin (ART) on the proliferation, migration, and inflammation induced by tumor necrosis factor-alpha (TNF-alpha) of rat vascular smooth muscle cells (VSMCs). artemisinin 65-68 tumor necrosis factor Rattus norvegicus 160-169 25575733-9 2015 RESULTS: The proliferation, migration, and inflammation of VSMCs induced by TNF-alpha were significantly inhibited by ART treatment in a dose-dependent manner. artemisinin 118-121 tumor necrosis factor Rattus norvegicus 76-85 25354548-5 2015 Western blot analysis revealed that QH induced apoptosis in human prostate cancer cells via activation of caspase-3 and cleavage of poly(adenosine diphosphate ribose) polymerase. artemisinin 36-38 caspase 3 Homo sapiens 106-115 25760540-0 2015 Effect of CAR polymorphism on the pharmacokinetics of artemisinin in healthy Chinese subjects. artemisinin 54-65 nuclear receptor subfamily 1 group I member 3 Homo sapiens 10-13 25760540-1 2015 Repeated pretreatment with the antimalarial drug artemisinin (QHS) could lead to reduced exposure to the parent drug, which is mainly mediated by auto-induction of CYP2B6 activity. artemisinin 49-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-170 25502316-4 2015 We analyzed the in vivo transcriptomes of 1043 P. falciparum isolates from patients with acute malaria and found that artemisinin resistance is associated with increased expression of unfolded protein response (UPR) pathways involving the major PROSC and TRiC chaperone complexes. artemisinin 118-129 pyridoxal phosphate binding protein Homo sapiens 245-250 25313206-3 2015 While several artemisinin-type antimalarial drugs have been shown to activate PXR, data on nonartemisinin-type antimalarials are still missing. artemisinin 14-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 78-81 24876631-6 2015 Artemisinin significantly increased hydrogen peroxide (H2O2) and malondialdehyde (MDA) levels and decreased catalase, glutathione peroxidase and superoxide dismutase activities in erythrocytes and uterus of rats compared with control group (p < 0.05). artemisinin 0-11 catalase Rattus norvegicus 108-116 25754587-0 2015 Enhancement of cytotoxicity of artemisinin toward cancer cells by transferrin-mediated carbon nanotubes nanoparticles. artemisinin 31-42 transferrin Homo sapiens 66-77 25263705-0 2014 Improper protein trafficking contributes to artemisinin sensitivity in cells lacking the KDAC Rpd3p. artemisinin 44-55 histone deacetylase RPD3 Saccharomyces cerevisiae S288C 94-99 24804646-1 2014 The interactions of dihydroartemisinin (DHA) and artemisinin (ART) with bovine serum albumin (BSA) have been investigated using fluorescence, UV/vis absorption and Fourier transform infrared (FTIR) spectra under simulated physiological conditions. artemisinin 27-38 albumin Homo sapiens 79-98 25263705-3 2014 Deletion of RPD3, but not other KDAC genes, resulted in strong sensitivity to artemisinin, which was also observed in sit4Delta mutants with impaired endoplasmic reticulum (ER) to Golgi protein trafficking. artemisinin 78-89 histone deacetylase RPD3 Saccharomyces cerevisiae S288C 12-16 25363545-0 2014 Cytochrome P450 single nucleotide polymorphisms in an indigenous Tanzanian population: a concern about the metabolism of artemisinin-based combinations. artemisinin 121-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 25110145-0 2014 Artemisinin prevents electric remodeling following myocardial infarction possibly by upregulating the expression of connexin 43. artemisinin 0-11 gap junction protein, alpha 1 Rattus norvegicus 116-127 24079412-4 2014 However, increased hydrogen peroxide and lipid peroxidation levels were accompanied by a concomitant decrease in glutathione peroxidase and glutathione-S-transferase activities as well as glutathione level in spermatozoon of rats administered with overdose of artemisinin. artemisinin 260-271 hematopoietic prostaglandin D synthase Rattus norvegicus 140-165 25110145-5 2014 The electrophysiological improvement of the VFT was accompanied by increased immunofluorescence-stained connexin 43 (Cx43), myocardial Cx43 protein and mRNA levels in artemisinin-treated rats. artemisinin 167-178 gap junction protein, alpha 1 Rattus norvegicus 135-139 25110145-6 2014 The present study also demonstrated that artemisinin significantly decreased tissue tumor necrosis factor (TNF)-alpha levels at the infarcted border zone. artemisinin 41-52 tumor necrosis factor Rattus norvegicus 84-117 25110145-8 2014 Although the precise mechanism by which artemisinin modulates the dephosphorylation of Cx43 is unknown, it is likely that artemisinin increased the expression of Cx43 via the inhibition of TNF-alpha. artemisinin 40-51 gap junction protein, alpha 1 Rattus norvegicus 87-91 25110145-8 2014 Although the precise mechanism by which artemisinin modulates the dephosphorylation of Cx43 is unknown, it is likely that artemisinin increased the expression of Cx43 via the inhibition of TNF-alpha. artemisinin 122-133 gap junction protein, alpha 1 Rattus norvegicus 162-166 25110145-8 2014 Although the precise mechanism by which artemisinin modulates the dephosphorylation of Cx43 is unknown, it is likely that artemisinin increased the expression of Cx43 via the inhibition of TNF-alpha. artemisinin 122-133 tumor necrosis factor Rattus norvegicus 189-198 24400699-0 2014 Effects of artemisinin antimalarials on Cytochrome P450 enzymes in vitro using recombinant enzymes and human liver microsomes: potential implications for combination therapies. artemisinin 11-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 25074847-0 2014 A semi-synthetic derivative of artemisinin, artesunate inhibits prostaglandin E2 production in LPS/IFNgamma-activated BV2 microglia. artemisinin 31-42 interferon gamma Homo sapiens 99-107 24886881-0 2014 Artemisinin protects against dextran sulfate-sodium-induced inflammatory bowel disease, which is associated with activation of the pregnane X receptor. artemisinin 0-11 nuclear receptor subfamily 1 group I member 2 Homo sapiens 131-150 24886881-2 2014 In the present study, the effects of artemisinin on pregnane X receptor (PXR)-mediated CYP3A expression and its therapeutic role in inflammatory bowel disease were investigated. artemisinin 37-48 nuclear receptor subfamily 1 group I member 2 Homo sapiens 52-71 24886881-2 2014 In the present study, the effects of artemisinin on pregnane X receptor (PXR)-mediated CYP3A expression and its therapeutic role in inflammatory bowel disease were investigated. artemisinin 37-48 nuclear receptor subfamily 1 group I member 2 Homo sapiens 73-76 24886881-2 2014 In the present study, the effects of artemisinin on pregnane X receptor (PXR)-mediated CYP3A expression and its therapeutic role in inflammatory bowel disease were investigated. artemisinin 37-48 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 87-92 24886881-3 2014 LS174T cells exposed to artemisinin at various concentrations and for different periods of time were examined with respect to the specific induction of CYP3A4 and PXR mRNA expression. artemisinin 24-35 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 152-158 24886881-3 2014 LS174T cells exposed to artemisinin at various concentrations and for different periods of time were examined with respect to the specific induction of CYP3A4 and PXR mRNA expression. artemisinin 24-35 nuclear receptor subfamily 1 group I member 2 Homo sapiens 163-166 24886881-4 2014 Transient transfection experiments showed transcriptional activation of the CYP3A4 gene through artemisinin to be PXR-dependent. artemisinin 96-107 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-82 24886881-4 2014 Transient transfection experiments showed transcriptional activation of the CYP3A4 gene through artemisinin to be PXR-dependent. artemisinin 96-107 nuclear receptor subfamily 1 group I member 2 Homo sapiens 114-117 24886881-5 2014 An electrophoretic-mobility shift assay (EMSA) showed that artemisinin activates the DNA-binding capacity of the PXR for the CYP3A4 element. artemisinin 59-70 nuclear receptor subfamily 1 group I member 2 Homo sapiens 113-116 24886881-5 2014 An electrophoretic-mobility shift assay (EMSA) showed that artemisinin activates the DNA-binding capacity of the PXR for the CYP3A4 element. artemisinin 59-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 125-131 24886881-6 2014 These results indicate that the induction of CYP3A4 by artemisinin is mediated through the activation of PXR. artemisinin 55-66 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-51 24886881-6 2014 These results indicate that the induction of CYP3A4 by artemisinin is mediated through the activation of PXR. artemisinin 55-66 nuclear receptor subfamily 1 group I member 2 Homo sapiens 105-108 24886881-9 2014 Artemisinin was found to prevent or reduce the severity of colonic inflammation by inducing CYP3A expression by activation of PXR. artemisinin 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-97 24886881-9 2014 Artemisinin was found to prevent or reduce the severity of colonic inflammation by inducing CYP3A expression by activation of PXR. artemisinin 0-11 nuclear receptor subfamily 1 group I member 2 Homo sapiens 126-129 25017372-5 2014 Annexin V-FITC/PI staining assay revealed that artemisinin markedly induced apoptosis. artemisinin 47-58 annexin A5 Homo sapiens 0-9 25116436-9 2014 These results identify GC B cells as a target of artesunate and provide a new rationale for using artemisinin analogues to treat autoimmune diseases mediated by autoantibodies. artemisinin 98-109 natriuretic peptide receptor 2 Mus musculus 23-27 24400699-5 2014 Effects of the antimalarial agent artemisinin and its structural analogues, artemether, artesunate and dihydroartemisinin, on seven of the major human liver CYP isoforms (CYP1A2, 2A6, 2B6, 2C9, 2C19, 2D6 and 3A4) were evaluated using recombinant enzymes (fluorometric assay) and human liver microsomes (LC-MS/MS analysis). artemisinin 34-45 peptidylprolyl isomerase G Homo sapiens 157-160 24400699-5 2014 Effects of the antimalarial agent artemisinin and its structural analogues, artemether, artesunate and dihydroartemisinin, on seven of the major human liver CYP isoforms (CYP1A2, 2A6, 2B6, 2C9, 2C19, 2D6 and 3A4) were evaluated using recombinant enzymes (fluorometric assay) and human liver microsomes (LC-MS/MS analysis). artemisinin 34-45 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 171-177 24400699-10 2014 A high risk of interaction in vivo was predicted if artemisinin is coadministrated with CYP1A2 or 2C19 substrates. artemisinin 52-63 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 88-94 24974584-4 2014 Treatment with artemisinin was also associated with activation of AMP kinase and inhibition of mTOR/p70S6K/pS6 signaling in SHSY5Y cells. artemisinin 15-26 mechanistic target of rapamycin kinase Homo sapiens 95-99 24661944-0 2014 Enhanced delivery of artemisinin and its analogues to cancer cells by their adducts with human serum transferrin. artemisinin 21-32 transferrin Homo sapiens 101-112 24974584-4 2014 Treatment with artemisinin was also associated with activation of AMP kinase and inhibition of mTOR/p70S6K/pS6 signaling in SHSY5Y cells. artemisinin 15-26 ribosomal protein S6 kinase B1 Homo sapiens 100-106 24721719-0 2014 Fetal bovine serum and human constitutive androstane receptor: evidence for activation of the SV23 splice variant by artemisinin, artemether, and arteether in a serum-free cell culture system. artemisinin 117-128 nuclear receptor subfamily 1 group I member 3 Homo sapiens 29-61 24661969-6 2014 MATERIALS AND METHODS: Healthy and Plasmodium chabaudi-infected mice were oral gavaged with pACT to deliver a 100 mg kg(-1) body weight dose of artemisinin. artemisinin 144-155 retinoblastoma binding protein 6, ubiquitin ligase Mus musculus 92-96 24887240-1 2014 BACKGROUND: Artemisinin is the major sesquiterpene lactones in sweet wormwood (Artemisia annua L.), and its combination with transferrin exhibits versatile anti-cancer activities. artemisinin 12-23 transferrin Homo sapiens 125-136 24721719-7 2014 Under this condition, artemisinin, artemether, and arteether increased hCAR-SV23 activity, whereas they decreased it in cells cultured in medium supplemented with 10% regular FBS. artemisinin 22-33 CXADR Ig-like cell adhesion molecule Homo sapiens 71-75 24661969-8 2014 RESULTS: The first order elimination rate constant for artemisinin in pACT-treated healthy mice was estimated to be 0.80 h(-1) with an elimination half-life (T1/2) of 51.6 min. artemisinin 55-66 retinoblastoma binding protein 6, ubiquitin ligase Mus musculus 70-74 24661969-19 2014 The results have implications for possible therapeutic use of pACT in treating malaria and other artemisinin-susceptible diseases. artemisinin 97-108 retinoblastoma binding protein 6, ubiquitin ligase Mus musculus 62-66 24333126-3 2014 The single compound and the combined doping experiments at 0, 25 and 50 mug mL-1 doping levels showed that artemetin (50 mug mL-1) caused a reduction in the amount of artemisinin crystallized by ca. artemisinin 167-178 L1 cell adhesion molecule Mus musculus 125-129 24296733-6 2014 Artemisinin treatment stimulated the cellular levels of IkappaB-alpha protein without altering the level of IkappaB-alpha transcripts. artemisinin 0-11 NFKB inhibitor alpha Homo sapiens 56-69 24296733-7 2014 Finally, expression of exogenous p65 resulted in the accumulation of this NF-kappaB subunit in the nucleus of artemisinin-treated and artemisinin-untreated cells, reversed the artemisinin downregulation of CDK4 protein expression and promoter activity, and prevented the artemisinin-induced G1 cell cycle arrest. artemisinin 110-121 RELA proto-oncogene, NF-kB subunit Homo sapiens 33-36 24296733-7 2014 Finally, expression of exogenous p65 resulted in the accumulation of this NF-kappaB subunit in the nucleus of artemisinin-treated and artemisinin-untreated cells, reversed the artemisinin downregulation of CDK4 protein expression and promoter activity, and prevented the artemisinin-induced G1 cell cycle arrest. artemisinin 110-121 cyclin dependent kinase 4 Homo sapiens 206-210 24296733-7 2014 Finally, expression of exogenous p65 resulted in the accumulation of this NF-kappaB subunit in the nucleus of artemisinin-treated and artemisinin-untreated cells, reversed the artemisinin downregulation of CDK4 protein expression and promoter activity, and prevented the artemisinin-induced G1 cell cycle arrest. artemisinin 134-145 RELA proto-oncogene, NF-kB subunit Homo sapiens 33-36 24296733-7 2014 Finally, expression of exogenous p65 resulted in the accumulation of this NF-kappaB subunit in the nucleus of artemisinin-treated and artemisinin-untreated cells, reversed the artemisinin downregulation of CDK4 protein expression and promoter activity, and prevented the artemisinin-induced G1 cell cycle arrest. artemisinin 134-145 cyclin dependent kinase 4 Homo sapiens 206-210 24296733-7 2014 Finally, expression of exogenous p65 resulted in the accumulation of this NF-kappaB subunit in the nucleus of artemisinin-treated and artemisinin-untreated cells, reversed the artemisinin downregulation of CDK4 protein expression and promoter activity, and prevented the artemisinin-induced G1 cell cycle arrest. artemisinin 134-145 RELA proto-oncogene, NF-kB subunit Homo sapiens 33-36 24296733-7 2014 Finally, expression of exogenous p65 resulted in the accumulation of this NF-kappaB subunit in the nucleus of artemisinin-treated and artemisinin-untreated cells, reversed the artemisinin downregulation of CDK4 protein expression and promoter activity, and prevented the artemisinin-induced G1 cell cycle arrest. artemisinin 134-145 cyclin dependent kinase 4 Homo sapiens 206-210 24296733-7 2014 Finally, expression of exogenous p65 resulted in the accumulation of this NF-kappaB subunit in the nucleus of artemisinin-treated and artemisinin-untreated cells, reversed the artemisinin downregulation of CDK4 protein expression and promoter activity, and prevented the artemisinin-induced G1 cell cycle arrest. artemisinin 134-145 RELA proto-oncogene, NF-kB subunit Homo sapiens 33-36 24296733-7 2014 Finally, expression of exogenous p65 resulted in the accumulation of this NF-kappaB subunit in the nucleus of artemisinin-treated and artemisinin-untreated cells, reversed the artemisinin downregulation of CDK4 protein expression and promoter activity, and prevented the artemisinin-induced G1 cell cycle arrest. artemisinin 134-145 cyclin dependent kinase 4 Homo sapiens 206-210 24296733-8 2014 Taken together, our results demonstrate that a key event in the artemisinin antiproliferative effects in endometrial cancer cells is the transcriptional downregulation of CDK4 expression by disruption of NF-kappaB interactions with the CDK4 promoter. artemisinin 64-75 cyclin dependent kinase 4 Homo sapiens 171-175 24296733-8 2014 Taken together, our results demonstrate that a key event in the artemisinin antiproliferative effects in endometrial cancer cells is the transcriptional downregulation of CDK4 expression by disruption of NF-kappaB interactions with the CDK4 promoter. artemisinin 64-75 cyclin dependent kinase 4 Homo sapiens 236-240 24296733-0 2014 Artemisinin triggers a G1 cell cycle arrest of human Ishikawa endometrial cancer cells and inhibits cyclin-dependent kinase-4 promoter activity and expression by disrupting nuclear factor-kappaB transcriptional signaling. artemisinin 0-11 cyclin dependent kinase 4 Homo sapiens 100-125 24296733-2 2014 Artemisinin induced a G1 cell cycle arrest in cultured human Ishikawa endometrial cancer cells and downregulated cyclin-dependent kinase-2 (CDK2) and CDK4 transcript and protein levels. artemisinin 0-11 cyclin dependent kinase 2 Homo sapiens 113-138 24296733-2 2014 Artemisinin induced a G1 cell cycle arrest in cultured human Ishikawa endometrial cancer cells and downregulated cyclin-dependent kinase-2 (CDK2) and CDK4 transcript and protein levels. artemisinin 0-11 cyclin dependent kinase 2 Homo sapiens 140-144 24296733-2 2014 Artemisinin induced a G1 cell cycle arrest in cultured human Ishikawa endometrial cancer cells and downregulated cyclin-dependent kinase-2 (CDK2) and CDK4 transcript and protein levels. artemisinin 0-11 cyclin dependent kinase 4 Homo sapiens 150-154 24296733-3 2014 Analysis of CDK4 promoter-luciferase reporter constructs showed that the artemisinin ablation of CDK4 gene expression was accounted for by the loss of CDK4 promoter activity. artemisinin 73-84 cyclin dependent kinase 4 Homo sapiens 12-16 24296733-3 2014 Analysis of CDK4 promoter-luciferase reporter constructs showed that the artemisinin ablation of CDK4 gene expression was accounted for by the loss of CDK4 promoter activity. artemisinin 73-84 cyclin dependent kinase 4 Homo sapiens 97-101 24296733-3 2014 Analysis of CDK4 promoter-luciferase reporter constructs showed that the artemisinin ablation of CDK4 gene expression was accounted for by the loss of CDK4 promoter activity. artemisinin 73-84 cyclin dependent kinase 4 Homo sapiens 97-101 24296733-4 2014 Chromatin immunoprecipitation demonstrated that artemisinin inhibited nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappaB) subunit p65 and p50 interactions with the endogenous Ishikawa cell CDK4 promoter. artemisinin 48-59 RELA proto-oncogene, NF-kB subunit Homo sapiens 153-156 24296733-4 2014 Chromatin immunoprecipitation demonstrated that artemisinin inhibited nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappaB) subunit p65 and p50 interactions with the endogenous Ishikawa cell CDK4 promoter. artemisinin 48-59 nuclear factor kappa B subunit 1 Homo sapiens 161-164 24296733-4 2014 Chromatin immunoprecipitation demonstrated that artemisinin inhibited nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappaB) subunit p65 and p50 interactions with the endogenous Ishikawa cell CDK4 promoter. artemisinin 48-59 cyclin dependent kinase 4 Homo sapiens 212-216 24296733-5 2014 Coimmunoprecipitation revealed that artemisinin disrupts endogenous p65 and p50 nuclear translocation through increased protein-protein interactions with IkappaB-alpha, an NF-kappaB inhibitor, and disrupts its interaction with the CDK4 promoter, leading to a loss of CDK4 gene expression. artemisinin 36-47 RELA proto-oncogene, NF-kB subunit Homo sapiens 68-71 24296733-5 2014 Coimmunoprecipitation revealed that artemisinin disrupts endogenous p65 and p50 nuclear translocation through increased protein-protein interactions with IkappaB-alpha, an NF-kappaB inhibitor, and disrupts its interaction with the CDK4 promoter, leading to a loss of CDK4 gene expression. artemisinin 36-47 nuclear factor kappa B subunit 1 Homo sapiens 76-79 24296733-5 2014 Coimmunoprecipitation revealed that artemisinin disrupts endogenous p65 and p50 nuclear translocation through increased protein-protein interactions with IkappaB-alpha, an NF-kappaB inhibitor, and disrupts its interaction with the CDK4 promoter, leading to a loss of CDK4 gene expression. artemisinin 36-47 NFKB inhibitor alpha Homo sapiens 154-167 24296733-5 2014 Coimmunoprecipitation revealed that artemisinin disrupts endogenous p65 and p50 nuclear translocation through increased protein-protein interactions with IkappaB-alpha, an NF-kappaB inhibitor, and disrupts its interaction with the CDK4 promoter, leading to a loss of CDK4 gene expression. artemisinin 36-47 cyclin dependent kinase 4 Homo sapiens 231-235 24296733-5 2014 Coimmunoprecipitation revealed that artemisinin disrupts endogenous p65 and p50 nuclear translocation through increased protein-protein interactions with IkappaB-alpha, an NF-kappaB inhibitor, and disrupts its interaction with the CDK4 promoter, leading to a loss of CDK4 gene expression. artemisinin 36-47 cyclin dependent kinase 4 Homo sapiens 267-271 25102693-0 2014 Artemisinin ameliorated proteinuria in rats with adriamycin-induced nephropathy through regulating nephrin and podocin expressions. artemisinin 0-11 NPHS1 adhesion molecule, nephrin Rattus norvegicus 99-106 25102693-9 2014 CONCLUSION: Artemisinin might have an effect on the nephropathy in rats caused by adriamycin, which may be at least partly correlated with attenu- ation of the severity of foot process effacement and fusion, up-regulation of the expressions of Nephrin and Podocin in the glomeruli in the rats. artemisinin 12-23 NPHS1 adhesion molecule, nephrin Rattus norvegicus 244-251 25102693-0 2014 Artemisinin ameliorated proteinuria in rats with adriamycin-induced nephropathy through regulating nephrin and podocin expressions. artemisinin 0-11 NPHS2 stomatin family member, podocin Rattus norvegicus 111-118 25102693-7 2014 Compared with adriamycin group, artemisinin could reduce uric protein excretion, decrease the serum TC, TG elevation, increase the serum Alb level, up-regulate the expressions of Nephrin and Podocin (P < 0.05 or P < 0.01), but no statistical significance effects on the levels of BUN, Scr in artemisinin group (P > 0.05). artemisinin 32-43 albumin Rattus norvegicus 137-140 25102693-9 2014 CONCLUSION: Artemisinin might have an effect on the nephropathy in rats caused by adriamycin, which may be at least partly correlated with attenu- ation of the severity of foot process effacement and fusion, up-regulation of the expressions of Nephrin and Podocin in the glomeruli in the rats. artemisinin 12-23 NPHS2 stomatin family member, podocin Rattus norvegicus 256-263 25102693-7 2014 Compared with adriamycin group, artemisinin could reduce uric protein excretion, decrease the serum TC, TG elevation, increase the serum Alb level, up-regulate the expressions of Nephrin and Podocin (P < 0.05 or P < 0.01), but no statistical significance effects on the levels of BUN, Scr in artemisinin group (P > 0.05). artemisinin 32-43 NPHS1 adhesion molecule, nephrin Rattus norvegicus 179-186 25102693-7 2014 Compared with adriamycin group, artemisinin could reduce uric protein excretion, decrease the serum TC, TG elevation, increase the serum Alb level, up-regulate the expressions of Nephrin and Podocin (P < 0.05 or P < 0.01), but no statistical significance effects on the levels of BUN, Scr in artemisinin group (P > 0.05). artemisinin 32-43 NPHS2 stomatin family member, podocin Rattus norvegicus 191-198 24381053-0 2013 A SAR and QSAR study of new artemisinin compounds with antimalarial activity. artemisinin 28-39 sarcosine dehydrogenase Homo sapiens 2-5 24078446-0 2014 Artemisinin inhibits gastric cancer cell proliferation through upregulation of p53. artemisinin 0-11 tumor protein p53 Homo sapiens 79-82 24078446-4 2014 Treatment with artemisinin was also associated with induction of p27 kip1 and p21 kip1, two negative cell-cycle regulators. artemisinin 15-26 cyclin dependent kinase inhibitor 1B Homo sapiens 65-73 24078446-5 2014 Furthermore, we revealed that artemisinin treatment led to an increased expression of p53. artemisinin 30-41 tumor protein p53 Homo sapiens 86-89 23661289-0 2013 Artemisinin induces A549 cell apoptosis dominantly via a reactive oxygen species-mediated amplification activation loop among caspase-9, -8 and -3. artemisinin 0-11 caspase 9 Homo sapiens 126-146 24337849-7 2013 The induction of apoptosis and inhibition of colony formation in SMMC-7721 cells treated with artemisinin were determined by TdT-mediated dUTP nick end labeling (TUNEL) and colony formation assay, respectively. artemisinin 94-105 DNA nucleotidylexotransferase Homo sapiens 125-128 23661289-1 2013 This report is designed to explore the roles of caspase-8, -9 and -3 in artemisinin (ARTE)-induced apoptosis in non-small cell lung cancer cells (A549 cells). artemisinin 72-83 caspase 8 Homo sapiens 48-68 23661289-1 2013 This report is designed to explore the roles of caspase-8, -9 and -3 in artemisinin (ARTE)-induced apoptosis in non-small cell lung cancer cells (A549 cells). artemisinin 85-89 caspase 8 Homo sapiens 48-68 23406388-0 2013 Antimalarial drug artemisinin extenuates amyloidogenesis and neuroinflammation in APPswe/PS1dE9 transgenic mice via inhibition of nuclear factor-kappaB and NLRP3 inflammasome activation. artemisinin 18-29 NLR family, pyrin domain containing 3 Mus musculus 156-161 23782869-1 2013 BACKGROUND: This case study describes how a public-private partnership between Medicines for Malaria Venture (MMV) and Sigma-Tau Industrie Farmaceutiche Riunite SpA achieved international regulatory approval for use of the fixed-dose artemisinin-based combination therapy dihydroartemisinin-piperaquine (Eurartesim ) for the treatment of malaria, enabling more widespread access to the medicine in malaria-endemic countries. artemisinin 234-245 surfactant protein A2 Homo sapiens 161-164 23684544-0 2013 Differential interactions of the broad spectrum drugs artemisinin, dihydroartemisinin and artesunate with serum albumin. artemisinin 54-65 albumin Homo sapiens 106-119 23684544-2 2013 As the binding affinity of artemisinin and its derivatives dihydroartemisinin and artesunate to blood serum proteins might influence the effectiveness of the drug, binding of artemisinin and derivatives to serum albumin was studied under near physiological conditions. artemisinin 27-38 albumin Homo sapiens 206-219 23979886-2 2013 Considering the lack of data regarding the genotoxic effects of these compounds in human cells, the objective of this study was to evaluate the cytotoxicity and genotoxicity, and expressions of the CASP3 and SOD1 genes in a cultured human hepatocellular liver carcinoma cell line (HepG2 cells) treated with artemisinin and artesunate. artemisinin 307-318 caspase 3 Homo sapiens 198-203 23560676-3 2013 To elucidate the potential pathways to Ft recovery, repair of oxidative damage to horse spleen apoferritin (apoFt) and Ft by quercetin (QH) or rutin (RH) was studied in the presence and absence of oxygen. artemisinin 136-138 ferritin heavy chain Equus caballus 95-106 23406388-5 2013 RESULTS: We found that artemisinin treatment (1) decreased neuritic plaque burden; (2) did not alter Abeta transport across the blood-brain barrier; (3) regulated APP processing via inhibiting beta-secretase activity; (4) inhibited NF-kappaB activity and NALP3 inflammasome activation in APPswe/PS1dE9 double transgenic mice. artemisinin 23-34 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 232-241 23406388-5 2013 RESULTS: We found that artemisinin treatment (1) decreased neuritic plaque burden; (2) did not alter Abeta transport across the blood-brain barrier; (3) regulated APP processing via inhibiting beta-secretase activity; (4) inhibited NF-kappaB activity and NALP3 inflammasome activation in APPswe/PS1dE9 double transgenic mice. artemisinin 23-34 NLR family, pyrin domain containing 3 Mus musculus 255-260 23406388-6 2013 CONCLUSIONS: The in vivo study clearly demonstrates that artemisinin has protective effects on AD pathology due to its effects on suppressing NF-kappaB activity and NALP3 inflammasome activation. artemisinin 57-68 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 142-151 23406388-6 2013 CONCLUSIONS: The in vivo study clearly demonstrates that artemisinin has protective effects on AD pathology due to its effects on suppressing NF-kappaB activity and NALP3 inflammasome activation. artemisinin 57-68 NLR family, pyrin domain containing 3 Mus musculus 165-170 23497071-7 2013 RESULTS: Over 80% of health workers interviewed were able to correctly identify the malaria M&E forms; 25.4% knew the basis for categorizing Artemisinin-based combination therapy (ACT) into ACT1 - ACT4; 97.6% of the respondents felt there was need to keep proper records to have information available and 7.5% of them kept records because they were asked to do so. artemisinin 145-156 TRAF3 interacting protein 2 Homo sapiens 194-198 23229480-0 2013 Effect of single nucleotide polymorphisms in cytochrome P450 isoenzyme and N-acetyltransferase 2 genes on the metabolism of artemisinin-based combination therapies in malaria patients from Cambodia and Tanzania. artemisinin 124-135 N-acetyltransferase 2 Homo sapiens 75-96 23467454-3 2013 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. artemisinin 43-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 23201011-4 2013 In present research, we demonstrated that dihydroartemisinin (DHA), a safe and effective antimalarial analog of artemisinin, could significantly inhibit the Bcr/Abl fusion gene at the mRNA level in CML cells sensitive or resistant to imatinib (including the primary CML cells with T315I mutation) and induce cell death. artemisinin 49-60 ABL proto-oncogene 1, non-receptor tyrosine kinase Homo sapiens 157-164 23287027-0 2013 Mutation analysis in pfmdr1 and pfmrp1 as potential candidate genes for artemisinin resistance in Plasmodium falciparum clinical isolates 4years after implementation of artemisinin combination therapy in Iran. artemisinin 72-83 multidrug resistance protein 1 Plasmodium falciparum 3D7 21-27 23287027-3 2013 Also, the copy number of pfmdr1 gene was screened for its association with pfmdr1 mutations to incriminate artemisinin resistance. artemisinin 107-118 multidrug resistance protein 1 Plasmodium falciparum 3D7 25-31 23287027-8 2013 In addition, the copy number of pfmdr1 gene (N = 1) was similar as a sensitive isolate, 3D7, to artemisinin. artemisinin 96-107 multidrug resistance protein 1 Plasmodium falciparum 3D7 32-38 23123209-0 2013 Induction of neurite outgrowth in PC12 cells by artemisinin through activation of ERK and p38 MAPK signaling pathways. artemisinin 48-59 Eph receptor B1 Rattus norvegicus 82-85 23123209-0 2013 Induction of neurite outgrowth in PC12 cells by artemisinin through activation of ERK and p38 MAPK signaling pathways. artemisinin 48-59 mitogen activated protein kinase 14 Rattus norvegicus 90-93 23123209-8 2013 Artemisinin upregulated phosphorylation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK), critical signaling molecules in neuronal differentiation. artemisinin 0-11 Eph receptor B1 Rattus norvegicus 43-80 23123209-8 2013 Artemisinin upregulated phosphorylation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK), critical signaling molecules in neuronal differentiation. artemisinin 0-11 Eph receptor B1 Rattus norvegicus 82-85 23123209-8 2013 Artemisinin upregulated phosphorylation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK), critical signaling molecules in neuronal differentiation. artemisinin 0-11 mitogen activated protein kinase 14 Rattus norvegicus 91-127 23123209-9 2013 Consistent with activation of the two MAPKs, neurite outgrowth induced by artemisinin was inhibited by the MAPK/ERK kinase inhibitor PD98059 and the p38 MAPK inhibitor SB203580. artemisinin 74-85 Eph receptor B1 Rattus norvegicus 112-115 23123209-9 2013 Consistent with activation of the two MAPKs, neurite outgrowth induced by artemisinin was inhibited by the MAPK/ERK kinase inhibitor PD98059 and the p38 MAPK inhibitor SB203580. artemisinin 74-85 mitogen activated protein kinase 14 Rattus norvegicus 149-152 23123209-10 2013 Artemisinin also induced phosphorylation of cyclic AMP response element-binding protein (CREB) that was almost completely attenuated by PD98059 but not by SB203580. artemisinin 0-11 cAMP responsive element binding protein 1 Rattus norvegicus 44-87 23123209-10 2013 Artemisinin also induced phosphorylation of cyclic AMP response element-binding protein (CREB) that was almost completely attenuated by PD98059 but not by SB203580. artemisinin 0-11 cAMP responsive element binding protein 1 Rattus norvegicus 89-93 23123209-11 2013 Taken together, our results indicate that artemisinin and its derivatives containing the endoperoxide bridge induced differentiation of PC12 cells toward a neuronal phenotype and suggest that both activation of ERK signaling pathway, which leads to CREB phosphorylation, and activation of p38 MAPK signaling pathway are involved in this process. artemisinin 42-53 Eph receptor B1 Rattus norvegicus 211-214 23123209-11 2013 Taken together, our results indicate that artemisinin and its derivatives containing the endoperoxide bridge induced differentiation of PC12 cells toward a neuronal phenotype and suggest that both activation of ERK signaling pathway, which leads to CREB phosphorylation, and activation of p38 MAPK signaling pathway are involved in this process. artemisinin 42-53 cAMP responsive element binding protein 1 Rattus norvegicus 249-253 23123209-11 2013 Taken together, our results indicate that artemisinin and its derivatives containing the endoperoxide bridge induced differentiation of PC12 cells toward a neuronal phenotype and suggest that both activation of ERK signaling pathway, which leads to CREB phosphorylation, and activation of p38 MAPK signaling pathway are involved in this process. artemisinin 42-53 mitogen activated protein kinase 14 Rattus norvegicus 289-292 23267137-0 2013 Effects of transferrin conjugates of artemisinin and artemisinin dimer on breast cancer cell lines. artemisinin 37-48 transferrin Homo sapiens 11-22 23267137-0 2013 Effects of transferrin conjugates of artemisinin and artemisinin dimer on breast cancer cell lines. artemisinin 53-64 transferrin Homo sapiens 11-22 23267137-1 2013 Transferrin (Tf) conjugates of monomeric artemisinin (ART) and artemisinin dimer were synthesized. artemisinin 41-52 transferrin Homo sapiens 0-11 23267137-1 2013 Transferrin (Tf) conjugates of monomeric artemisinin (ART) and artemisinin dimer were synthesized. artemisinin 63-74 transferrin Homo sapiens 0-11 23267137-5 2013 To our knowledge, this is the first demonstration that an ART derivative can cause a decline of ERBB2 in a human cancer cell line. artemisinin 58-61 erb-b2 receptor tyrosine kinase 2 Homo sapiens 96-101 23041519-0 2012 Artemisinin inhibits lipopolysaccharide-induced interferon-beta production in RAW 264.7 cells: implications on signal transducer and activator of transcription-1 signaling and nitric oxide production. artemisinin 0-11 signal transducer and activator of transcription 1 Mus musculus 111-161 23516601-6 2013 We demonstrate for the first time, for any ART derivative, that ADP NPs can down regulate the oncogenic protein HER2, and its counterpart, HER3 in a HER2+ cell line. artemisinin 43-46 erb-b2 receptor tyrosine kinase 2 Mus musculus 112-116 23516601-6 2013 We demonstrate for the first time, for any ART derivative, that ADP NPs can down regulate the oncogenic protein HER2, and its counterpart, HER3 in a HER2+ cell line. artemisinin 43-46 erb-b2 receptor tyrosine kinase 3 Mus musculus 139-143 23041519-2 2012 In this study, we investigated the effect of artemisinin on lipopolysaccharide (LPS)-induced production of IFN-beta and characterized the potential relationship between artemisinin-mediated inhibition of IFN-beta and NO production. artemisinin 169-180 interferon beta 1, fibroblast Mus musculus 204-212 23041519-3 2012 Artemisinin suppressed IFN-beta production and mRNA expression in a dose-dependent manner in LPS-stimulated RAW 264.7 cells. artemisinin 0-11 interferon beta 1, fibroblast Mus musculus 23-31 23041519-4 2012 LPS-induced phosphorylation of signal transducer and activator of transcription-1 (STAT-1) was also inhibited by artemisinin treatment in RAW 264.7 cells. artemisinin 113-124 signal transducer and activator of transcription 1 Mus musculus 31-81 23041519-4 2012 LPS-induced phosphorylation of signal transducer and activator of transcription-1 (STAT-1) was also inhibited by artemisinin treatment in RAW 264.7 cells. artemisinin 113-124 signal transducer and activator of transcription 1 Mus musculus 83-89 23041519-6 2012 Further study demonstrated that artemisinin-mediated inhibition of NO production and STAT-1 phosphorylation was reversed by addition of exogenous IFN-beta. artemisinin 32-43 signal transducer and activator of transcription 1 Mus musculus 85-91 23041519-6 2012 Further study demonstrated that artemisinin-mediated inhibition of NO production and STAT-1 phosphorylation was reversed by addition of exogenous IFN-beta. artemisinin 32-43 interferon beta 1, fibroblast Mus musculus 146-154 23041519-8 2012 Collectively, these results suggest that the inhibition of IFN-beta production by artemisinin and concomitant attenuation of STAT-1 activation might be involved in artemisinin-mediated inhibition of NO production in macrophages. artemisinin 82-93 interferon beta 1, fibroblast Mus musculus 59-67 23041519-8 2012 Collectively, these results suggest that the inhibition of IFN-beta production by artemisinin and concomitant attenuation of STAT-1 activation might be involved in artemisinin-mediated inhibition of NO production in macrophages. artemisinin 164-175 interferon beta 1, fibroblast Mus musculus 59-67 23041519-8 2012 Collectively, these results suggest that the inhibition of IFN-beta production by artemisinin and concomitant attenuation of STAT-1 activation might be involved in artemisinin-mediated inhibition of NO production in macrophages. artemisinin 164-175 signal transducer and activator of transcription 1 Mus musculus 125-131 22922993-4 2012 Our results show that artemisinin significantly inhibited IL-1beta and TNF-alpha protein expression and the infiltration of macrophages. artemisinin 22-33 interleukin 1 beta Homo sapiens 58-66 22922993-4 2012 Our results show that artemisinin significantly inhibited IL-1beta and TNF-alpha protein expression and the infiltration of macrophages. artemisinin 22-33 tumor necrosis factor Homo sapiens 71-80 22922993-5 2012 Artemisinin significantly decreased the protein expression of NGF, GAP43, and TH compared with the control group, which was related to sympathetic nerve remodeling. artemisinin 0-11 nerve growth factor Homo sapiens 62-65 22922993-5 2012 Artemisinin significantly decreased the protein expression of NGF, GAP43, and TH compared with the control group, which was related to sympathetic nerve remodeling. artemisinin 0-11 growth associated protein 43 Homo sapiens 67-72 22922993-7 2012 In addition, the densities of both GAP-43- and TH-immunoreactive nerves in the peri-infarct zone were significantly attenuated by artemisinin treatment. artemisinin 130-141 growth associated protein 43 Homo sapiens 35-41 23212638-0 2012 Enhanced IL-12p40 production in LPS-stimulated macrophages by inhibiting JNK activation by artemisinin. artemisinin 91-102 interleukin 12b Mus musculus 9-17 23212638-0 2012 Enhanced IL-12p40 production in LPS-stimulated macrophages by inhibiting JNK activation by artemisinin. artemisinin 91-102 mitogen-activated protein kinase 8 Mus musculus 73-76 23212638-2 2012 In this study, we investigated the effect of artemisinin on the production of IL-12p40, which is important in the generation of T helper 1 responses. artemisinin 45-56 interleukin 12b Mus musculus 78-86 23212638-3 2012 Artemisinin significantly induced IL-12p40 production in LPS-stimulated RAW264.7 macrophage cells. artemisinin 0-11 interleukin 12b Mus musculus 34-42 23212638-4 2012 To elucidate the signaling molecules regulated by artemisinin in induced IL-12p40 production, the DNA-binding activity of several transcription factors and activation of mitogen-activated protein kinase (MAPK)s were investigated. artemisinin 50-61 interleukin 12b Mus musculus 73-81 23212638-6 2012 However, the induced phosphorylation of JNK was significantly decreased by artemisinin, and inhibition of the JNK signaling pathway further increased IL-12p40 production in LPS-stimulated RAW264.7 macrophage cells. artemisinin 75-86 mitogen-activated protein kinase 8 Mus musculus 40-43 23212638-7 2012 Taken together, these data suggest that artemisinin induces the production of IL-12p40 in LPS-stimulated macrophage cells by inhibiting JNK activity. artemisinin 40-51 interleukin 12b Mus musculus 78-86 23212638-7 2012 Taken together, these data suggest that artemisinin induces the production of IL-12p40 in LPS-stimulated macrophage cells by inhibiting JNK activity. artemisinin 40-51 mitogen-activated protein kinase 8 Mus musculus 136-139 22679214-4 2012 To better understand the autoinduction and metabolic drug-drug interactions (DDIs), we evaluated the P450s (particularly CYP2B6 and CYP3A4) inhibited or induced by two artemisinin drugs, Qing-hao-su (QHS) and dihydroartemisinin (DHA) using human liver microsome, recombinant P450 enzymes, and primary human hepatocytes. artemisinin 168-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-127 22853186-0 2012 LyP-1 modification to enhance delivery of artemisinin or fluorescent probe loaded polymeric micelles to highly metastatic tumor and its lymphatics. artemisinin 42-53 protein tyrosine phosphatase non-receptor type 22 Homo sapiens 0-5 22577882-5 2012 KEY RESULTS: Only two artemisinin derivatives arteether and artemether, the metabolite deoxyartemisinin and artemisinin itself demonstrated agonist binding to the major isoforms CAR1 and CAR3, while arteether and artemether were also inverse agonists of CAR2. artemisinin 22-33 nuclear receptor subfamily 1 group I member 3 Homo sapiens 178-182 22577882-5 2012 KEY RESULTS: Only two artemisinin derivatives arteether and artemether, the metabolite deoxyartemisinin and artemisinin itself demonstrated agonist binding to the major isoforms CAR1 and CAR3, while arteether and artemether were also inverse agonists of CAR2. artemisinin 22-33 carbonic anhydrase 3 Homo sapiens 187-191 22577882-5 2012 KEY RESULTS: Only two artemisinin derivatives arteether and artemether, the metabolite deoxyartemisinin and artemisinin itself demonstrated agonist binding to the major isoforms CAR1 and CAR3, while arteether and artemether were also inverse agonists of CAR2. artemisinin 22-33 nuclear receptor subfamily 1 group I member 4 Homo sapiens 254-258 22577882-5 2012 KEY RESULTS: Only two artemisinin derivatives arteether and artemether, the metabolite deoxyartemisinin and artemisinin itself demonstrated agonist binding to the major isoforms CAR1 and CAR3, while arteether and artemether were also inverse agonists of CAR2. artemisinin 92-103 nuclear receptor subfamily 1 group I member 3 Homo sapiens 178-182 22577882-7 2012 While arteether showed the highest activities in vitro, it was less active than artemisinin in inducing hepatic CYP3A4 gene expression in hepatocytes. artemisinin 80-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 112-118 22577882-8 2012 CONCLUSIONS AND IMPLICATIONS: Artemisinin derivatives and metabolites differentially affect the activities of CAR isoforms and of the pregnane X receptor (PXR). artemisinin 30-41 nuclear receptor subfamily 1 group I member 3 Homo sapiens 110-113 22577882-8 2012 CONCLUSIONS AND IMPLICATIONS: Artemisinin derivatives and metabolites differentially affect the activities of CAR isoforms and of the pregnane X receptor (PXR). artemisinin 30-41 nuclear receptor subfamily 1 group I member 2 Homo sapiens 134-153 22577882-8 2012 CONCLUSIONS AND IMPLICATIONS: Artemisinin derivatives and metabolites differentially affect the activities of CAR isoforms and of the pregnane X receptor (PXR). artemisinin 30-41 nuclear receptor subfamily 1 group I member 2 Homo sapiens 155-158 22577882-9 2012 This negates a common effect of these drugs on CAR/PXR-dependent induction of drug metabolism and further provides an explanation for artemisinin consistently inducing cytochrome P450 genes in vivo, whereas arteether and artemether do not. artemisinin 134-145 nuclear receptor subfamily 1 group I member 3 Homo sapiens 47-50 22577882-9 2012 This negates a common effect of these drugs on CAR/PXR-dependent induction of drug metabolism and further provides an explanation for artemisinin consistently inducing cytochrome P450 genes in vivo, whereas arteether and artemether do not. artemisinin 134-145 nuclear receptor subfamily 1 group I member 2 Homo sapiens 51-54 22679214-4 2012 To better understand the autoinduction and metabolic drug-drug interactions (DDIs), we evaluated the P450s (particularly CYP2B6 and CYP3A4) inhibited or induced by two artemisinin drugs, Qing-hao-su (QHS) and dihydroartemisinin (DHA) using human liver microsome, recombinant P450 enzymes, and primary human hepatocytes. artemisinin 168-179 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 132-138 22679214-4 2012 To better understand the autoinduction and metabolic drug-drug interactions (DDIs), we evaluated the P450s (particularly CYP2B6 and CYP3A4) inhibited or induced by two artemisinin drugs, Qing-hao-su (QHS) and dihydroartemisinin (DHA) using human liver microsome, recombinant P450 enzymes, and primary human hepatocytes. artemisinin 168-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 22552829-9 2012 Analyses of G2-checkpoint-related proteins, the activation of Wee 1 and depression of cyclin B1 expression induced by radiation, could be restored to the control level by artemisinin. artemisinin 171-182 WEE1 G2 checkpoint kinase Homo sapiens 62-67 23252279-6 2012 The activity and expression level of Caspase-3 were significantly increased when treated with galactosylated artemisinin for 36 h and 72 h. The expression of BAX was also increased markedly, while BCL-2 was reduced after treatment with the drug. artemisinin 109-120 caspase 3 Homo sapiens 37-46 23252279-6 2012 The activity and expression level of Caspase-3 were significantly increased when treated with galactosylated artemisinin for 36 h and 72 h. The expression of BAX was also increased markedly, while BCL-2 was reduced after treatment with the drug. artemisinin 109-120 BCL2 associated X, apoptosis regulator Homo sapiens 158-161 23252279-6 2012 The activity and expression level of Caspase-3 were significantly increased when treated with galactosylated artemisinin for 36 h and 72 h. The expression of BAX was also increased markedly, while BCL-2 was reduced after treatment with the drug. artemisinin 109-120 BCL2 apoptosis regulator Homo sapiens 197-202 23252279-8 2012 CONCLUSION: Galactosylated artemisinin exhibits great antitumor activities, which may be ralated to triggering cytochrome C apoptotic pathway which mediated by BCL-2 family. artemisinin 27-38 BCL2 apoptosis regulator Homo sapiens 160-165 23236763-7 2012 Artemisinin only inhibited the IL-6 release to a certain extent. artemisinin 0-11 interleukin 6 Mus musculus 31-35 21547369-9 2012 Taken together, these data demonstrate that metastatic melanoma cells display a specific vulnerability to DHA-induced NOXA-dependent apoptosis and suggest feasibility of future anti-melanoma intervention using artemisinin-derived clinical redox antimalarials. artemisinin 210-221 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 118-122 22434375-0 2012 Amplification activation loop between caspase-8 and -9 dominates artemisinin-induced apoptosis of ASTC-a-1 cells. artemisinin 65-76 caspase 8 Homo sapiens 38-54 22434375-1 2012 Although caspases have been demonstrated to be involved in artemisinin (ARTE)-induced apoptosis, their exact functions are not well understood. artemisinin 59-70 caspase 8 Homo sapiens 9-17 22434375-1 2012 Although caspases have been demonstrated to be involved in artemisinin (ARTE)-induced apoptosis, their exact functions are not well understood. artemisinin 72-76 caspase 8 Homo sapiens 9-17 22434375-3 2012 ARTE treatment induces a rapid generation of reactive oxygen species (ROS), and ROS-dependent apoptosis as well as the activation of caspase-8, -9 and -3 via time- and dose-dependent fashion. artemisinin 0-4 caspase 8 Homo sapiens 133-153 22434375-8 2012 Our findings imply a potential to develop new derivatives from artemisinin to effectively initiate the amplification activation loop of caspases. artemisinin 63-74 caspase 8 Homo sapiens 136-144 22552829-9 2012 Analyses of G2-checkpoint-related proteins, the activation of Wee 1 and depression of cyclin B1 expression induced by radiation, could be restored to the control level by artemisinin. artemisinin 171-182 cyclin B1 Homo sapiens 86-95 22552829-10 2012 CONCLUSIONS: Given the unique cytotoxic profile of artemisinin on cancer cells and normal cells, artemisinin may be a potentially promising radiosensitizer through the regulation of the expression of G2 checkpoint-related proteins like Wee 1 and cyclin B1, and improve therapeutic ratios for the combination of artemisinin and ionizing irradiation in the treatment of patients with cervical cancer. artemisinin 97-108 WEE1 G2 checkpoint kinase Homo sapiens 236-241 22552829-10 2012 CONCLUSIONS: Given the unique cytotoxic profile of artemisinin on cancer cells and normal cells, artemisinin may be a potentially promising radiosensitizer through the regulation of the expression of G2 checkpoint-related proteins like Wee 1 and cyclin B1, and improve therapeutic ratios for the combination of artemisinin and ionizing irradiation in the treatment of patients with cervical cancer. artemisinin 97-108 cyclin B1 Homo sapiens 246-255 22552829-10 2012 CONCLUSIONS: Given the unique cytotoxic profile of artemisinin on cancer cells and normal cells, artemisinin may be a potentially promising radiosensitizer through the regulation of the expression of G2 checkpoint-related proteins like Wee 1 and cyclin B1, and improve therapeutic ratios for the combination of artemisinin and ionizing irradiation in the treatment of patients with cervical cancer. artemisinin 97-108 WEE1 G2 checkpoint kinase Homo sapiens 236-241 22552829-10 2012 CONCLUSIONS: Given the unique cytotoxic profile of artemisinin on cancer cells and normal cells, artemisinin may be a potentially promising radiosensitizer through the regulation of the expression of G2 checkpoint-related proteins like Wee 1 and cyclin B1, and improve therapeutic ratios for the combination of artemisinin and ionizing irradiation in the treatment of patients with cervical cancer. artemisinin 97-108 cyclin B1 Homo sapiens 246-255 22252826-0 2012 PXR variants and artemisinin use in Vietnamese subjects: frequency distribution and impact on the interindividual variability of CYP3A induction by artemisinin. artemisinin 148-159 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-3 22185819-0 2012 Antiproliferative effects of artemisinin on human breast cancer cells requires the downregulated expression of the E2F1 transcription factor and loss of E2F1-target cell cycle genes. artemisinin 29-40 E2F transcription factor 1 Homo sapiens 153-157 22185819-0 2012 Antiproliferative effects of artemisinin on human breast cancer cells requires the downregulated expression of the E2F1 transcription factor and loss of E2F1-target cell cycle genes. artemisinin 29-40 E2F transcription factor 1 Homo sapiens 115-119 22252826-0 2012 PXR variants and artemisinin use in Vietnamese subjects: frequency distribution and impact on the interindividual variability of CYP3A induction by artemisinin. artemisinin 148-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-134 22185819-4 2012 Concurrent with the cell cycle arrest of MCF7 cells, artemisinin selectively downregulated the transcript and protein levels of the CDK2 and CDK4 cyclin-dependent kinases, cyclin E, cyclin D1, and the E2F1 transcription factor. artemisinin 53-64 cyclin dependent kinase 2 Homo sapiens 132-136 22252826-2 2012 Here, we report the resequencing and genotyping of PXR variants in 75 Vietnamese individuals previously characterized for CYP3A enzyme activity after artemisinin exposure. artemisinin 150-161 nuclear receptor subfamily 1 group I member 2 Homo sapiens 51-54 22252826-2 2012 Here, we report the resequencing and genotyping of PXR variants in 75 Vietnamese individuals previously characterized for CYP3A enzyme activity after artemisinin exposure. artemisinin 150-161 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 122-127 22185819-4 2012 Concurrent with the cell cycle arrest of MCF7 cells, artemisinin selectively downregulated the transcript and protein levels of the CDK2 and CDK4 cyclin-dependent kinases, cyclin E, cyclin D1, and the E2F1 transcription factor. artemisinin 53-64 cyclin E1 L homeolog Xenopus laevis 172-180 22252826-4 2012 A trend of significance was observed between the level of CYP3A4 induction by artemisinin and two PXR variants, the 8118C T (Y328Y) and 10719A G variants. artemisinin 78-89 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 58-64 22185819-4 2012 Concurrent with the cell cycle arrest of MCF7 cells, artemisinin selectively downregulated the transcript and protein levels of the CDK2 and CDK4 cyclin-dependent kinases, cyclin E, cyclin D1, and the E2F1 transcription factor. artemisinin 53-64 cyclin D1 Homo sapiens 182-191 22252826-4 2012 A trend of significance was observed between the level of CYP3A4 induction by artemisinin and two PXR variants, the 8118C T (Y328Y) and 10719A G variants. artemisinin 78-89 nuclear receptor subfamily 1 group I member 2 Homo sapiens 98-101 22185819-4 2012 Concurrent with the cell cycle arrest of MCF7 cells, artemisinin selectively downregulated the transcript and protein levels of the CDK2 and CDK4 cyclin-dependent kinases, cyclin E, cyclin D1, and the E2F1 transcription factor. artemisinin 53-64 E2F transcription factor 1 Homo sapiens 201-205 22185819-5 2012 Analysis of CDK2 promoter-luciferase reporter constructs showed that the artemisinin ablation of CDK2 gene expression was accounted for by the loss of CDK2 promoter activity. artemisinin 73-84 cyclin dependent kinase 2 Homo sapiens 12-16 23506555-0 2012 The evaluation of CYP2B6 inhibition by artemisinin antimalarials in recombinant enzymes and human liver microsomes. artemisinin 39-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 22185819-5 2012 Analysis of CDK2 promoter-luciferase reporter constructs showed that the artemisinin ablation of CDK2 gene expression was accounted for by the loss of CDK2 promoter activity. artemisinin 73-84 cyclin dependent kinase 2 Homo sapiens 97-101 22185819-5 2012 Analysis of CDK2 promoter-luciferase reporter constructs showed that the artemisinin ablation of CDK2 gene expression was accounted for by the loss of CDK2 promoter activity. artemisinin 73-84 cyclin dependent kinase 2 Homo sapiens 97-101 22185819-6 2012 Chromatin immunoprecipitation revealed that artemisinin inhibited E2F1 interactions with the endogenous MCF7 cell CDK2 and cyclin E promoters. artemisinin 44-55 E2F transcription factor 1 Homo sapiens 66-70 22185819-6 2012 Chromatin immunoprecipitation revealed that artemisinin inhibited E2F1 interactions with the endogenous MCF7 cell CDK2 and cyclin E promoters. artemisinin 44-55 cyclin dependent kinase 2 Homo sapiens 114-118 22185819-6 2012 Chromatin immunoprecipitation revealed that artemisinin inhibited E2F1 interactions with the endogenous MCF7 cell CDK2 and cyclin E promoters. artemisinin 44-55 cyclin E1 L homeolog Xenopus laevis 123-131 22185819-7 2012 Moreover, constitutive expression of exogenous E2F1 prevented the artemisinin-induced cell cycle arrest and downregulation of CDK2 and cyclin E gene expression. artemisinin 66-77 E2F transcription factor 1 L homeolog Xenopus laevis 47-51 22185819-7 2012 Moreover, constitutive expression of exogenous E2F1 prevented the artemisinin-induced cell cycle arrest and downregulation of CDK2 and cyclin E gene expression. artemisinin 66-77 cyclin-dependent kinase 2 S homeolog Xenopus laevis 126-130 22185819-7 2012 Moreover, constitutive expression of exogenous E2F1 prevented the artemisinin-induced cell cycle arrest and downregulation of CDK2 and cyclin E gene expression. artemisinin 66-77 cyclin E1 L homeolog Xenopus laevis 135-143 22185819-8 2012 Taken together, our results demonstrate that the artemisinin disruption of E2F1 transcription factor expression mediates the cell cycle arrest of human breast cancer cells and represents a critical transcriptional pathway by which artemisinin controls human reproductive cancer cell growth. artemisinin 49-60 E2F transcription factor 1 Homo sapiens 75-79 22185819-8 2012 Taken together, our results demonstrate that the artemisinin disruption of E2F1 transcription factor expression mediates the cell cycle arrest of human breast cancer cells and represents a critical transcriptional pathway by which artemisinin controls human reproductive cancer cell growth. artemisinin 231-242 E2F transcription factor 1 Homo sapiens 75-79 23019910-0 2012 [Effect of artemisinin on the expressions of GRalpha mRNA, GRbeta mRNA and P300/CBP protein in lupus nephritis mice]. artemisinin 11-22 CREB binding protein Mus musculus 75-83 23019910-1 2012 OBJECTIVE: To investigate the effects of Artemisinin on the expressions of GRalpha mRNA, GRbeta mRNA and P300/CBP protein in lupus nephritis mice. artemisinin 41-52 CREB binding protein Mus musculus 105-113 22359336-1 2012 Artemisinin (QHS) is one of the first-line antimalarials, and autoinduction of CYP-mediated metabolism can result in its reduced exposure. artemisinin 0-11 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 79-82 23506555-2 2012 Some artemisinin compounds and anti-retroviral drugs have been shown to be metabolized by CYP2B6. artemisinin 5-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 23506555-4 2012 This study aimed to investigate whether artemisinin compounds inhibit CYP2B6 activity in vitro using recombinant CYP2B6 (rCYP2B6) and human liver microsomes (HLM). artemisinin 40-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 23506555-4 2012 This study aimed to investigate whether artemisinin compounds inhibit CYP2B6 activity in vitro using recombinant CYP2B6 (rCYP2B6) and human liver microsomes (HLM). artemisinin 40-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 23506555-4 2012 This study aimed to investigate whether artemisinin compounds inhibit CYP2B6 activity in vitro using recombinant CYP2B6 (rCYP2B6) and human liver microsomes (HLM). artemisinin 40-51 cytochrome P450, family 2, subfamily b, polypeptide 3 Rattus norvegicus 121-128 23506555-7 2012 Artemisinin and artemether were shown to inhibit CYP2B6 in vitro through a partial mixed type of inhibition, while dihydroartemisinin did not inhibit the enzymatic activity. artemisinin 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 23506555-8 2012 IC50 values for artemisinin were 9.5 and 9.1 microM for rCYP2B6 and HLM, respectively, after 30 min of incubation. artemisinin 16-27 cytochrome P450, family 2, subfamily b, polypeptide 3 Rattus norvegicus 56-63 23506555-11 2012 Based on the [I]/Ki approach using rCYP2B6, the risk of DDIs for artemisinin was indicated to be medium to high, while artemether had a low risk. artemisinin 65-76 cytochrome P450, family 2, subfamily b, polypeptide 3 Rattus norvegicus 35-42 21411308-1 2011 Protein engineering of cytochrome P450 monooxygenases (P450s) has been very successful in generating valuable non-natural activities and properties, allowing these powerful catalysts to be used for the synthesis of drug metabolites and in biosynthetic pathways for the production of precursors of artemisinin and paclitaxel. artemisinin 297-308 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 22724024-8 2012 Inhibition of both routes through selected molecules (SB203580, quercetin, artemisinin, parthenolide) prevented HZ-dependent lysozyme release. artemisinin 75-86 lysozyme Homo sapiens 125-133 22615874-5 2012 In contrast, RNA interference depletion of intrinsic Sp1 or treatment with artemisinin, a Sp1 inhibitor, reduced expression of LEDGF/p75, suggesting Sp1-mediated regulation of LEDGF/p75. artemisinin 75-86 Chromatin assembly factor 1, p105 subunit Drosophila melanogaster 127-136 22615874-5 2012 In contrast, RNA interference depletion of intrinsic Sp1 or treatment with artemisinin, a Sp1 inhibitor, reduced expression of LEDGF/p75, suggesting Sp1-mediated regulation of LEDGF/p75. artemisinin 75-86 Chromatin assembly factor 1, p105 subunit Drosophila melanogaster 176-185 22514713-4 2012 Our results show that artemisinin significantly inhibited LPS-induced production of tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1) and nitric oxide (NO). artemisinin 22-33 tumor necrosis factor Homo sapiens 84-111 22514713-4 2012 Our results show that artemisinin significantly inhibited LPS-induced production of tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1) and nitric oxide (NO). artemisinin 22-33 tumor necrosis factor Homo sapiens 113-122 22514713-4 2012 Our results show that artemisinin significantly inhibited LPS-induced production of tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1) and nitric oxide (NO). artemisinin 22-33 interleukin 6 Homo sapiens 125-138 22514713-4 2012 Our results show that artemisinin significantly inhibited LPS-induced production of tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1) and nitric oxide (NO). artemisinin 22-33 interleukin 6 Homo sapiens 140-144 22514713-4 2012 Our results show that artemisinin significantly inhibited LPS-induced production of tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1) and nitric oxide (NO). artemisinin 22-33 C-C motif chemokine ligand 2 Homo sapiens 147-177 22514713-4 2012 Our results show that artemisinin significantly inhibited LPS-induced production of tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1) and nitric oxide (NO). artemisinin 22-33 C-C motif chemokine ligand 2 Homo sapiens 179-184 22514713-5 2012 Artemisinin significantly decreased both the mRNA and the protein levels of these pro-inflammatory cytokines and inducible nitric oxide synthase (iNOS) and increased the protein levels of IkappaB-alpha, which forms a cytoplasmic inactive complex with the p65-p50 heterodimeric complex. artemisinin 0-11 nitric oxide synthase 2 Homo sapiens 113-144 22514713-5 2012 Artemisinin significantly decreased both the mRNA and the protein levels of these pro-inflammatory cytokines and inducible nitric oxide synthase (iNOS) and increased the protein levels of IkappaB-alpha, which forms a cytoplasmic inactive complex with the p65-p50 heterodimeric complex. artemisinin 0-11 nitric oxide synthase 2 Homo sapiens 146-150 22514713-5 2012 Artemisinin significantly decreased both the mRNA and the protein levels of these pro-inflammatory cytokines and inducible nitric oxide synthase (iNOS) and increased the protein levels of IkappaB-alpha, which forms a cytoplasmic inactive complex with the p65-p50 heterodimeric complex. artemisinin 0-11 NFKB inhibitor alpha Homo sapiens 188-201 22514713-8 2012 The inhibitory effects of artemisinin on LPS-stimulated microglia were blocked after IkappaB-alpha was silenced with IkappaB-alpha siRNA. artemisinin 26-37 NFKB inhibitor alpha Homo sapiens 85-98 22514713-8 2012 The inhibitory effects of artemisinin on LPS-stimulated microglia were blocked after IkappaB-alpha was silenced with IkappaB-alpha siRNA. artemisinin 26-37 NFKB inhibitor alpha Homo sapiens 117-130 22389703-10 2012 CONCLUSIONS/SIGNIFICANCE: The results of this study demonstrated that artemisinin analogue SM934 had therapeutic effects on lupus-prone female NZB/W F(1) mice by inhibiting the pathogenic helper T cell development and enhancing anti-inflammatory cytokine IL-10 production. artemisinin 70-81 interleukin 10 Mus musculus 255-260 21719297-1 2011 The indole alkaloid cyclopiazonic acid (CPA) is one of the few known nanomolar inhibitors of sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) besides the anticancer drug thapsigargin and the antiplasmoidal terpenoid artemisinin. artemisinin 216-227 ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 3 Homo sapiens 93-133 21854775-6 2011 Artemisinin, primaquine, and quinine down-regulated GTP-cyclohydrolase 1 gene expression 1.26-, 1.29-, and 1.63-fold, respectively. artemisinin 0-11 GTP cyclohydrolase 1 Homo sapiens 52-72 21484768-0 2011 Oral administration of artemisinin analog SM934 ameliorates lupus syndromes in MRL/lpr mice by inhibiting Th1 and Th17 cell responses. artemisinin 23-34 negative elongation factor complex member C/D, Th1l Mus musculus 106-109 21484768-9 2011 CONCLUSION: Taken together, the results of this study demonstrated that the artemisinin analog SM934 had therapeutic effects in lupus-prone female MRL/lpr mice by inhibiting both Th1 cell and Th17 cell responses. artemisinin 76-87 negative elongation factor complex member C/D, Th1l Mus musculus 179-182 21309636-1 2011 The cytotoxicity of dihydroartemisinin (DHART; an active metabolite of artemisinin or ART) was investigated using murine GT1-7 hypothalamic neurons. artemisinin 27-38 retinoic acid induced 1 Mus musculus 121-124 21445878-0 2011 Down-regulation of BMI-1 cooperates with artemisinin on growth inhibition of nasopharyngeal carcinoma cells. artemisinin 41-52 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 19-24 21445878-7 2011 Artemisinin inhibited BMI-1 both in protein and transcript levels. artemisinin 0-11 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 22-27 21445878-8 2011 BMI-1 knockdown made the cells more sensitive to artemisinin with an increase in G1 phase, but over-expression of BMI-1 partially reversed the artemisinin-induced G1 cell cycle arrest. artemisinin 49-60 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 0-5 21445878-8 2011 BMI-1 knockdown made the cells more sensitive to artemisinin with an increase in G1 phase, but over-expression of BMI-1 partially reversed the artemisinin-induced G1 cell cycle arrest. artemisinin 143-154 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 114-119 21445878-9 2011 Depletion of BMI-1 was able to intensifying the increment of p16 and the reduction of CDK4 induced by artemisinin. artemisinin 102-113 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 13-18 21445878-9 2011 Depletion of BMI-1 was able to intensifying the increment of p16 and the reduction of CDK4 induced by artemisinin. artemisinin 102-113 cyclin dependent kinase 4 Homo sapiens 86-90 21445878-10 2011 In addition, over-expression of BMI-1 was capable of attenuating the increasing p16 and decreasing CDK4 in cells treated with artemisinin. artemisinin 126-137 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 32-37 21445878-10 2011 In addition, over-expression of BMI-1 was capable of attenuating the increasing p16 and decreasing CDK4 in cells treated with artemisinin. artemisinin 126-137 cyclin dependent kinase inhibitor 2A Homo sapiens 80-83 21445878-10 2011 In addition, over-expression of BMI-1 was capable of attenuating the increasing p16 and decreasing CDK4 in cells treated with artemisinin. artemisinin 126-137 cyclin dependent kinase 4 Homo sapiens 99-103 21445878-11 2011 Taking together, the BMI1-p16/CDK4 axis was involved in the artemisinin-driven G1 arrest in nasopharyngeal carcinoma cells, and these results indicated that a potential treatment that the combination of artemisinin and BMI-1 downregulation could enhance the growth inhibitory affects on nasopharyngeal carcinoma cells. artemisinin 60-71 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 21-25 21445878-11 2011 Taking together, the BMI1-p16/CDK4 axis was involved in the artemisinin-driven G1 arrest in nasopharyngeal carcinoma cells, and these results indicated that a potential treatment that the combination of artemisinin and BMI-1 downregulation could enhance the growth inhibitory affects on nasopharyngeal carcinoma cells. artemisinin 60-71 cyclin dependent kinase inhibitor 2A Homo sapiens 26-29 21445878-11 2011 Taking together, the BMI1-p16/CDK4 axis was involved in the artemisinin-driven G1 arrest in nasopharyngeal carcinoma cells, and these results indicated that a potential treatment that the combination of artemisinin and BMI-1 downregulation could enhance the growth inhibitory affects on nasopharyngeal carcinoma cells. artemisinin 60-71 cyclin dependent kinase 4 Homo sapiens 30-34 21445878-11 2011 Taking together, the BMI1-p16/CDK4 axis was involved in the artemisinin-driven G1 arrest in nasopharyngeal carcinoma cells, and these results indicated that a potential treatment that the combination of artemisinin and BMI-1 downregulation could enhance the growth inhibitory affects on nasopharyngeal carcinoma cells. artemisinin 60-71 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 219-224 21445878-11 2011 Taking together, the BMI1-p16/CDK4 axis was involved in the artemisinin-driven G1 arrest in nasopharyngeal carcinoma cells, and these results indicated that a potential treatment that the combination of artemisinin and BMI-1 downregulation could enhance the growth inhibitory affects on nasopharyngeal carcinoma cells. artemisinin 203-214 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 21-25 21445878-11 2011 Taking together, the BMI1-p16/CDK4 axis was involved in the artemisinin-driven G1 arrest in nasopharyngeal carcinoma cells, and these results indicated that a potential treatment that the combination of artemisinin and BMI-1 downregulation could enhance the growth inhibitory affects on nasopharyngeal carcinoma cells. artemisinin 203-214 cyclin dependent kinase inhibitor 2A Homo sapiens 26-29 21445878-11 2011 Taking together, the BMI1-p16/CDK4 axis was involved in the artemisinin-driven G1 arrest in nasopharyngeal carcinoma cells, and these results indicated that a potential treatment that the combination of artemisinin and BMI-1 downregulation could enhance the growth inhibitory affects on nasopharyngeal carcinoma cells. artemisinin 203-214 cyclin dependent kinase 4 Homo sapiens 30-34 21320288-4 2011 Here it is shown that, in the absence of reducing agents, artemisinin and artesunate targeted the S. cerevisiae calcium channels Pmr1p and Pmc1p. artemisinin 58-69 Ca(2+)/Mn(2+)-transporting P-type ATPase PMR1 Saccharomyces cerevisiae S288C 129-134 21509671-0 2011 Effect of artemisinin combined with glucocorticoid on the expressions of glucocorticoid receptor alpha mRNA, glucocorticoid receptor beta mRNA and P300/CBP protein in lupus nephritis mice. artemisinin 10-21 CREB binding protein Mus musculus 147-155 21320288-4 2011 Here it is shown that, in the absence of reducing agents, artemisinin and artesunate targeted the S. cerevisiae calcium channels Pmr1p and Pmc1p. artemisinin 58-69 calcium-transporting ATPase PMC1 Saccharomyces cerevisiae S288C 139-144 28299053-4 2011 During its work in Chad, Sierra Leone and Mali in the period 2004 to 2008, Medecins Sans Frontieres showed that it was possible to significantly improve access to effective malaria treatment through: i) the removal of health centre level user fees for essential healthcare for vulnerable population groups, ii) the introduction of free community based treatment for children using malaria village workers to diagnose and treat simple malaria in communities where geographical and financial barriers limited access to effective malaria care, iii) the improved diagnosis and treatment of malaria using rapid diagnosis tests and artemisinin based combination therapy, at both health facilities and in the community. artemisinin 626-637 chondroadherin Homo sapiens 19-23 21039753-8 2011 Artemisinin (20-80 mug/mL) significantly inhibited the induction of EMMPRIN and MMP-9 at both the transcriptional and translational levels in a dose-dependent manner in PMA-induced macrophages. artemisinin 0-11 basigin (Ok blood group) Homo sapiens 68-75 21165548-3 2011 In the present study, we demonstrate that artemisinin inhibits the secretion and the mRNA levels of tumor necrosis factor (TNF)-alpha, interleukin (IL)-1ss, and IL-6 in a dose-dependent manner in phorbol 12-myristate 13-acetate (PMA)-induced THP-1 human monocytes. artemisinin 42-53 tumor necrosis factor Homo sapiens 100-133 21165548-3 2011 In the present study, we demonstrate that artemisinin inhibits the secretion and the mRNA levels of tumor necrosis factor (TNF)-alpha, interleukin (IL)-1ss, and IL-6 in a dose-dependent manner in phorbol 12-myristate 13-acetate (PMA)-induced THP-1 human monocytes. artemisinin 42-53 interleukin 6 Homo sapiens 161-165 21039753-0 2011 Artemisinin inhibits extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinase-9 expression via a protein kinase Cdelta/p38/extracellular signal-regulated kinase pathway in phorbol myristate acetate-induced THP-1 macrophages. artemisinin 0-11 basigin (Ok blood group) Homo sapiens 21-67 21039753-8 2011 Artemisinin (20-80 mug/mL) significantly inhibited the induction of EMMPRIN and MMP-9 at both the transcriptional and translational levels in a dose-dependent manner in PMA-induced macrophages. artemisinin 0-11 matrix metallopeptidase 9 Homo sapiens 80-85 21039753-0 2011 Artemisinin inhibits extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinase-9 expression via a protein kinase Cdelta/p38/extracellular signal-regulated kinase pathway in phorbol myristate acetate-induced THP-1 macrophages. artemisinin 0-11 basigin (Ok blood group) Homo sapiens 69-76 21039753-0 2011 Artemisinin inhibits extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinase-9 expression via a protein kinase Cdelta/p38/extracellular signal-regulated kinase pathway in phorbol myristate acetate-induced THP-1 macrophages. artemisinin 0-11 matrix metallopeptidase 9 Homo sapiens 82-108 21039753-9 2011 In addition, artemisinin (20-80 mug/mL) strongly blocked PKCdelta/JNK/p38/ERK MAPK phosphorylation. artemisinin 13-24 protein kinase C delta Homo sapiens 57-65 21039753-0 2011 Artemisinin inhibits extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinase-9 expression via a protein kinase Cdelta/p38/extracellular signal-regulated kinase pathway in phorbol myristate acetate-induced THP-1 macrophages. artemisinin 0-11 mitogen-activated protein kinase 1 Homo sapiens 148-151 21039753-0 2011 Artemisinin inhibits extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinase-9 expression via a protein kinase Cdelta/p38/extracellular signal-regulated kinase pathway in phorbol myristate acetate-induced THP-1 macrophages. artemisinin 0-11 GLI family zinc finger 2 Homo sapiens 235-240 21039753-9 2011 In addition, artemisinin (20-80 mug/mL) strongly blocked PKCdelta/JNK/p38/ERK MAPK phosphorylation. artemisinin 13-24 mitogen-activated protein kinase 8 Homo sapiens 66-69 21039753-3 2011 The aim of the present study was to explore whether artemisinin, a natural extract from Artemisia annua, could decrease EMMPRIN and MMP-9 expression in phorbol myristate acetate (PMA)-induced macrophages by regulating the protein kinase (PK) Cdelta/c-Jun N-terminal kinase (JNK)/p38/extracellular signal-regulated kinase (ERK) pathway. artemisinin 52-63 basigin (Ok blood group) Homo sapiens 120-127 21039753-3 2011 The aim of the present study was to explore whether artemisinin, a natural extract from Artemisia annua, could decrease EMMPRIN and MMP-9 expression in phorbol myristate acetate (PMA)-induced macrophages by regulating the protein kinase (PK) Cdelta/c-Jun N-terminal kinase (JNK)/p38/extracellular signal-regulated kinase (ERK) pathway. artemisinin 52-63 matrix metallopeptidase 9 Homo sapiens 132-137 21039753-9 2011 In addition, artemisinin (20-80 mug/mL) strongly blocked PKCdelta/JNK/p38/ERK MAPK phosphorylation. artemisinin 13-24 mitogen-activated protein kinase 1 Homo sapiens 70-73 21039753-3 2011 The aim of the present study was to explore whether artemisinin, a natural extract from Artemisia annua, could decrease EMMPRIN and MMP-9 expression in phorbol myristate acetate (PMA)-induced macrophages by regulating the protein kinase (PK) Cdelta/c-Jun N-terminal kinase (JNK)/p38/extracellular signal-regulated kinase (ERK) pathway. artemisinin 52-63 mitogen-activated protein kinase 8 Homo sapiens 274-277 21039753-3 2011 The aim of the present study was to explore whether artemisinin, a natural extract from Artemisia annua, could decrease EMMPRIN and MMP-9 expression in phorbol myristate acetate (PMA)-induced macrophages by regulating the protein kinase (PK) Cdelta/c-Jun N-terminal kinase (JNK)/p38/extracellular signal-regulated kinase (ERK) pathway. artemisinin 52-63 mitogen-activated protein kinase 1 Homo sapiens 279-282 21039753-3 2011 The aim of the present study was to explore whether artemisinin, a natural extract from Artemisia annua, could decrease EMMPRIN and MMP-9 expression in phorbol myristate acetate (PMA)-induced macrophages by regulating the protein kinase (PK) Cdelta/c-Jun N-terminal kinase (JNK)/p38/extracellular signal-regulated kinase (ERK) pathway. artemisinin 52-63 mitogen-activated protein kinase 1 Homo sapiens 322-325 21039753-9 2011 In addition, artemisinin (20-80 mug/mL) strongly blocked PKCdelta/JNK/p38/ERK MAPK phosphorylation. artemisinin 13-24 mitogen-activated protein kinase 1 Homo sapiens 74-77 21039753-9 2011 In addition, artemisinin (20-80 mug/mL) strongly blocked PKCdelta/JNK/p38/ERK MAPK phosphorylation. artemisinin 13-24 mitogen-activated protein kinase 1 Homo sapiens 78-82 21039753-12 2011 The results of the present study suggest that artemisinin inhibits EMMPRIN and MMP-9 expression and activity by suppressing the PKCdelta/ERK/p38 cascade in PMA-induced macrophages. artemisinin 46-57 basigin (Ok blood group) Homo sapiens 67-74 21039753-12 2011 The results of the present study suggest that artemisinin inhibits EMMPRIN and MMP-9 expression and activity by suppressing the PKCdelta/ERK/p38 cascade in PMA-induced macrophages. artemisinin 46-57 matrix metallopeptidase 9 Homo sapiens 79-84 21039753-12 2011 The results of the present study suggest that artemisinin inhibits EMMPRIN and MMP-9 expression and activity by suppressing the PKCdelta/ERK/p38 cascade in PMA-induced macrophages. artemisinin 46-57 protein kinase C delta Homo sapiens 128-136 21039753-12 2011 The results of the present study suggest that artemisinin inhibits EMMPRIN and MMP-9 expression and activity by suppressing the PKCdelta/ERK/p38 cascade in PMA-induced macrophages. artemisinin 46-57 mitogen-activated protein kinase 1 Homo sapiens 137-140 21039753-12 2011 The results of the present study suggest that artemisinin inhibits EMMPRIN and MMP-9 expression and activity by suppressing the PKCdelta/ERK/p38 cascade in PMA-induced macrophages. artemisinin 46-57 mitogen-activated protein kinase 1 Homo sapiens 141-144 21106771-4 2010 These results proved that it is reliable and effective to use Art to treat LN mice, and its therapeutic mechanisms should closely be related to the fact that Art can obviously decrease the serum levels of TNF-alpha and IL-6 and down-regulate the expression of the NF-kappaBp65 protein and NF-kappaB and TGF-beta1 mRNA in renal tissues. artemisinin 62-65 tumor necrosis factor Mus musculus 205-214 21695271-2 2011 Anti-malarial drug artesunate is a semi-synthetic derivative of artemisinin, the principal active component of a medicinal plant Artemisia annua, and has been shown to inhibit PI3K/Akt activity. artemisinin 64-75 thymoma viral proto-oncogene 1 Mus musculus 181-184 20863781-5 2010 Artemisinin blocked angiotensin II-induced cardiac hypertrophy in vitro in a concentration-dependent manner. artemisinin 0-11 angiotensinogen Rattus norvegicus 20-34 20863781-8 2010 Banded rats treated with oral artemisinin, compared with untreated rats, showed significantly decreased the levels of IL-6, TNF-alpha and MCP-1 mRNA expression and increased protein levels of IkappaB-alpha, which forms a cytoplasmic inactive complex with the p65-p50 heterodimeric complex. artemisinin 30-41 interleukin 6 Rattus norvegicus 118-122 20863781-8 2010 Banded rats treated with oral artemisinin, compared with untreated rats, showed significantly decreased the levels of IL-6, TNF-alpha and MCP-1 mRNA expression and increased protein levels of IkappaB-alpha, which forms a cytoplasmic inactive complex with the p65-p50 heterodimeric complex. artemisinin 30-41 tumor necrosis factor Rattus norvegicus 124-133 20863781-8 2010 Banded rats treated with oral artemisinin, compared with untreated rats, showed significantly decreased the levels of IL-6, TNF-alpha and MCP-1 mRNA expression and increased protein levels of IkappaB-alpha, which forms a cytoplasmic inactive complex with the p65-p50 heterodimeric complex. artemisinin 30-41 mast cell protease 1-like 1 Rattus norvegicus 138-143 20863781-8 2010 Banded rats treated with oral artemisinin, compared with untreated rats, showed significantly decreased the levels of IL-6, TNF-alpha and MCP-1 mRNA expression and increased protein levels of IkappaB-alpha, which forms a cytoplasmic inactive complex with the p65-p50 heterodimeric complex. artemisinin 30-41 NFKB inhibitor alpha Rattus norvegicus 192-205 20863781-9 2010 The effect of artemisinin on cardiac hypertrophy was blocked after IkappaB-alpha was silenced by transfection of cardiomyocytes with IkappaB-alpha siRNA. artemisinin 14-25 NFKB inhibitor alpha Rattus norvegicus 67-80 20863781-9 2010 The effect of artemisinin on cardiac hypertrophy was blocked after IkappaB-alpha was silenced by transfection of cardiomyocytes with IkappaB-alpha siRNA. artemisinin 14-25 NFKB inhibitor alpha Rattus norvegicus 133-146 25214386-0 2011 Artemisinin induces caspase-8/9-mediated and Bax/Bak-independent apoptosis in human lung adenocarcinoma (ASTC-a-1) cells. artemisinin 0-11 caspase 8 Homo sapiens 20-29 25214386-0 2011 Artemisinin induces caspase-8/9-mediated and Bax/Bak-independent apoptosis in human lung adenocarcinoma (ASTC-a-1) cells. artemisinin 0-11 BCL2 associated X, apoptosis regulator Homo sapiens 45-48 25214386-0 2011 Artemisinin induces caspase-8/9-mediated and Bax/Bak-independent apoptosis in human lung adenocarcinoma (ASTC-a-1) cells. artemisinin 0-11 BCL2 antagonist/killer 1 Homo sapiens 49-52 21106771-4 2010 These results proved that it is reliable and effective to use Art to treat LN mice, and its therapeutic mechanisms should closely be related to the fact that Art can obviously decrease the serum levels of TNF-alpha and IL-6 and down-regulate the expression of the NF-kappaBp65 protein and NF-kappaB and TGF-beta1 mRNA in renal tissues. artemisinin 62-65 interleukin 6 Mus musculus 219-223 21106771-4 2010 These results proved that it is reliable and effective to use Art to treat LN mice, and its therapeutic mechanisms should closely be related to the fact that Art can obviously decrease the serum levels of TNF-alpha and IL-6 and down-regulate the expression of the NF-kappaBp65 protein and NF-kappaB and TGF-beta1 mRNA in renal tissues. artemisinin 62-65 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 264-273 21106771-4 2010 These results proved that it is reliable and effective to use Art to treat LN mice, and its therapeutic mechanisms should closely be related to the fact that Art can obviously decrease the serum levels of TNF-alpha and IL-6 and down-regulate the expression of the NF-kappaBp65 protein and NF-kappaB and TGF-beta1 mRNA in renal tissues. artemisinin 62-65 transforming growth factor, beta 1 Mus musculus 303-312 18463201-7 2008 Parthenolide and artemisinin prevented all of the statin effects by inducing RhoA/Rho kinase activation. artemisinin 17-28 ras homolog family member A Homo sapiens 77-81 20678173-5 2010 All HZ/trophozoite/15-HETE effects on MMP-9 activity and TNF/IL-1beta production were abrogated by quercetin, artemisinin and parthenolide, inhibitors of IkappaBalpha phosphorylation and subsequent degradation, NF-kappaB nuclear translocation, and NF-kappaB-p65 binding to DNA respectively. artemisinin 110-121 matrix metallopeptidase 9 Homo sapiens 38-43 20678173-5 2010 All HZ/trophozoite/15-HETE effects on MMP-9 activity and TNF/IL-1beta production were abrogated by quercetin, artemisinin and parthenolide, inhibitors of IkappaBalpha phosphorylation and subsequent degradation, NF-kappaB nuclear translocation, and NF-kappaB-p65 binding to DNA respectively. artemisinin 110-121 tumor necrosis factor Homo sapiens 57-60 20678173-5 2010 All HZ/trophozoite/15-HETE effects on MMP-9 activity and TNF/IL-1beta production were abrogated by quercetin, artemisinin and parthenolide, inhibitors of IkappaBalpha phosphorylation and subsequent degradation, NF-kappaB nuclear translocation, and NF-kappaB-p65 binding to DNA respectively. artemisinin 110-121 interleukin 1 beta Homo sapiens 61-69 20678173-5 2010 All HZ/trophozoite/15-HETE effects on MMP-9 activity and TNF/IL-1beta production were abrogated by quercetin, artemisinin and parthenolide, inhibitors of IkappaBalpha phosphorylation and subsequent degradation, NF-kappaB nuclear translocation, and NF-kappaB-p65 binding to DNA respectively. artemisinin 110-121 NFKB inhibitor alpha Homo sapiens 154-166 20335983-2 2010 Reduction of artemisinin by NaBH4 produced dihydroartemisinin (DHA), and yielded a new stereochemically labile centre at C-10, which, in turn, provided two interconverting lactol hemiacetal epimers (namely alpha and beta), whose rate of interconversion depends on buffer, pH, and solvent polarity. artemisinin 13-24 homeobox C10 Homo sapiens 121-125 20681308-0 2010 [Effects of artemisinin on proliferation, apoptosis and Caspase-3 expression of rat mesangial cell]. artemisinin 12-23 caspase 3 Rattus norvegicus 56-65 20681308-1 2010 OBJECTIVE: To study the effects of artemisinin on proliferation, apoptosis and Caspase-3 active of rat mesangial cell. artemisinin 35-46 caspase 3 Rattus norvegicus 79-88 20681308-6 2010 CONCLUSION: Artemisinin could inhibit the proliferation of mesangial cell, enhance the expression of Caspase-3 and promote the apoptosis in a dose-dependent manner. artemisinin 12-23 caspase 3 Rattus norvegicus 101-110 18956140-6 2009 In addition, artemisinin affected the migratory ability of A375M cells by reducing metalloproteinase 2 (MMP-2) production and down-regulating alpha v beta 3 integrin expression. artemisinin 13-24 matrix metallopeptidase 2 Homo sapiens 104-109 19017637-0 2009 Artemisinin blocks prostate cancer growth and cell cycle progression by disrupting Sp1 interactions with the cyclin-dependent kinase-4 (CDK4) promoter and inhibiting CDK4 gene expression. artemisinin 0-11 cyclin dependent kinase 4 Homo sapiens 109-134 19017637-0 2009 Artemisinin blocks prostate cancer growth and cell cycle progression by disrupting Sp1 interactions with the cyclin-dependent kinase-4 (CDK4) promoter and inhibiting CDK4 gene expression. artemisinin 0-11 cyclin dependent kinase 4 Homo sapiens 136-140 19017637-0 2009 Artemisinin blocks prostate cancer growth and cell cycle progression by disrupting Sp1 interactions with the cyclin-dependent kinase-4 (CDK4) promoter and inhibiting CDK4 gene expression. artemisinin 0-11 cyclin dependent kinase 4 Homo sapiens 166-170 19017637-2 2009 We have observed that artemisinin treatment triggers a stringent G1 cell cycle arrest of LNCaP (lymph node carcinoma of the prostate) human prostate cancer cells that is accompanied by a rapid down-regulation of CDK2 and CDK4 protein and transcript levels. artemisinin 22-33 cyclin dependent kinase 2 Homo sapiens 212-216 19017637-2 2009 We have observed that artemisinin treatment triggers a stringent G1 cell cycle arrest of LNCaP (lymph node carcinoma of the prostate) human prostate cancer cells that is accompanied by a rapid down-regulation of CDK2 and CDK4 protein and transcript levels. artemisinin 22-33 cyclin dependent kinase 4 Homo sapiens 221-225 19017637-3 2009 Transient transfection with promoter-linked luciferase reporter plasmids revealed that artemisinin strongly inhibits CDK2 and CDK4 promoter activity. artemisinin 87-98 cyclin dependent kinase 2 Homo sapiens 117-121 19017637-3 2009 Transient transfection with promoter-linked luciferase reporter plasmids revealed that artemisinin strongly inhibits CDK2 and CDK4 promoter activity. artemisinin 87-98 cyclin dependent kinase 4 Homo sapiens 126-130 19017637-4 2009 Deletion analysis of the CDK4 promoter revealed a 231-bp artemisinin-responsive region between -1737 and -1506. artemisinin 57-68 cyclin dependent kinase 4 Homo sapiens 25-29 19017637-5 2009 Site-specific mutations revealed that the Sp1 site at -1531 was necessary for artemisinin responsiveness in the context of the CDK4 promoter. artemisinin 78-89 cyclin dependent kinase 4 Homo sapiens 127-131 19017637-6 2009 DNA binding assays as well as chromatin immunoprecipitation assays demonstrated that this Sp1-binding site in the CDK4 promoter forms a specific artemisinin-responsive DNA-protein complex that contains the Sp1 transcription factor. artemisinin 145-156 cyclin dependent kinase 4 Homo sapiens 114-118 19017637-7 2009 Artemisinin reduced phosphorylation of Sp1, and when dephosphorylation of Sp1 was inhibited by treatment of cells with the phosphatase inhibitor okadaic acid, the ability of artemisinin to down-regulate Sp1 interactions with the CDK4 promoter was ablated, rendering the CDK4 promoter unresponsive to artemisinin. artemisinin 174-185 cyclin dependent kinase 4 Homo sapiens 229-233 19017637-7 2009 Artemisinin reduced phosphorylation of Sp1, and when dephosphorylation of Sp1 was inhibited by treatment of cells with the phosphatase inhibitor okadaic acid, the ability of artemisinin to down-regulate Sp1 interactions with the CDK4 promoter was ablated, rendering the CDK4 promoter unresponsive to artemisinin. artemisinin 174-185 cyclin dependent kinase 4 Homo sapiens 270-274 19017637-8 2009 Finally, overexpression of Sp1 mostly reversed the artemisinin down-regulation of CDK4 promoter activity and partially reversed the cell cycle arrest. artemisinin 51-62 cyclin dependent kinase 4 Homo sapiens 82-86 19017637-9 2009 Taken together, our results demonstrate that a key event in the artemisinin anti-proliferative effects in prostate cancer cells is the transcriptional down-regulation of CDK4 expression by disruption of Sp1 interactions with the CDK4 promoter. artemisinin 64-75 cyclin dependent kinase 4 Homo sapiens 170-174 19017637-9 2009 Taken together, our results demonstrate that a key event in the artemisinin anti-proliferative effects in prostate cancer cells is the transcriptional down-regulation of CDK4 expression by disruption of Sp1 interactions with the CDK4 promoter. artemisinin 64-75 cyclin dependent kinase 4 Homo sapiens 229-233 19255645-9 2009 Curcumin is reported to act synergistically with artemisinin which forms covalent adducts with the transmembrane proteins (SERCA enzyme) and inactivates them, thus inhibiting the activity of Plasmodium parasite. artemisinin 49-60 ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 3 Homo sapiens 123-128 18784357-0 2008 Artemisinin selectively decreases functional levels of estrogen receptor-alpha and ablates estrogen-induced proliferation in human breast cancer cells. artemisinin 0-11 estrogen receptor 1 Homo sapiens 55-78 18784357-2 2008 Treatment of MCF7 cells with artemisinin, an antimalarial phytochemical from the sweet wormwood plant, effectively blocked estrogen-stimulated cell cycle progression induced by either 17beta-estradiol (E(2)), an agonist for both ERs, or by propyl pyrazole triol (PPT), a selective ERalpha agonist. artemisinin 29-40 estrogen receptor 1 Homo sapiens 281-288 18784357-3 2008 Artemisinin strongly downregulated ERalpha protein and transcripts without altering expression or activity of ERbeta. artemisinin 0-11 estrogen receptor 1 Homo sapiens 35-42 18784357-4 2008 Transfection of MCF7 cells with ERalpha promoter-linked luciferase reporter plasmids revealed that the artemisinin downregulation of ERalpha promoter activity accounted for the loss of ERalpha expression. artemisinin 103-114 estrogen receptor 1 Homo sapiens 32-39 18784357-4 2008 Transfection of MCF7 cells with ERalpha promoter-linked luciferase reporter plasmids revealed that the artemisinin downregulation of ERalpha promoter activity accounted for the loss of ERalpha expression. artemisinin 103-114 estrogen receptor 1 Homo sapiens 133-140 18784357-4 2008 Transfection of MCF7 cells with ERalpha promoter-linked luciferase reporter plasmids revealed that the artemisinin downregulation of ERalpha promoter activity accounted for the loss of ERalpha expression. artemisinin 103-114 estrogen receptor 1 Homo sapiens 133-140 18784357-5 2008 Artemisinin treatment ablated the estrogenic induction of endogenous progesterone receptor (PR) transcripts by either E(2) or PPT and inhibited the estrogenic stimulation of a luciferase reporter plasmid driven by consensus estrogen response elements (EREs). artemisinin 0-11 progesterone receptor Homo sapiens 69-90 18784357-5 2008 Artemisinin treatment ablated the estrogenic induction of endogenous progesterone receptor (PR) transcripts by either E(2) or PPT and inhibited the estrogenic stimulation of a luciferase reporter plasmid driven by consensus estrogen response elements (EREs). artemisinin 0-11 progesterone receptor Homo sapiens 92-94 18784357-6 2008 Chromatin immunoprecipitation assays revealed that artemisinin significantly downregulated the level of endogeneous ERalpha bound to the PR promoter, whereas the level of bound endogeneous ERbeta was not altered. artemisinin 51-62 estrogen receptor 1 Homo sapiens 116-123 18784357-6 2008 Chromatin immunoprecipitation assays revealed that artemisinin significantly downregulated the level of endogeneous ERalpha bound to the PR promoter, whereas the level of bound endogeneous ERbeta was not altered. artemisinin 51-62 progesterone receptor Homo sapiens 137-139 18784357-7 2008 Treatment of MCF7 cells with artemisinin and the pure antiestrogen fulvestrant resulted in a cooperative reduction of ERalpha protein levels and enhanced G(1) cell cycle arrest compared with the effects of either compound alone. artemisinin 29-40 estrogen receptor 1 Homo sapiens 118-125 18784357-8 2008 Our results show that artemisinin switches proliferative human breast cancer cells from expressing a high ERalpha:ERbeta ratio to a condition in which ERbeta predominates, which parallels the physiological state linked to antiproliferative events in normal mammary epithelium. artemisinin 22-33 estrogen receptor 1 Homo sapiens 106-113 18784357-8 2008 Our results show that artemisinin switches proliferative human breast cancer cells from expressing a high ERalpha:ERbeta ratio to a condition in which ERbeta predominates, which parallels the physiological state linked to antiproliferative events in normal mammary epithelium. artemisinin 22-33 estrogen receptor 2 Homo sapiens 114-120 18784357-8 2008 Our results show that artemisinin switches proliferative human breast cancer cells from expressing a high ERalpha:ERbeta ratio to a condition in which ERbeta predominates, which parallels the physiological state linked to antiproliferative events in normal mammary epithelium. artemisinin 22-33 estrogen receptor 2 Homo sapiens 151-157 20530490-2 2010 Mutation of an amino acid residue in mammalian SERCA1 (Glu(255)) to the equivalent one predicted in PfATP6 (Leu) was reported to induce sensitivity to artemisinin in the oocyte system. artemisinin 151-162 ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 1 Homo sapiens 47-53 20130467-3 2010 Intracellular iron uptake is regulated by the transferrin receptor (TfR), and the activity of artemisinin depends on the availability of iron. artemisinin 94-105 transferrin receptor Homo sapiens 68-71 20137162-4 2010 In this article, recent progress of study on transferrin receptor used in treating hematological malignant tumor are reviewed from aspects of transferrin receptor combined with drugs including artemisinin, doxorubicin, gambogic acid and so on, genes, antibodies, polyethylene glycol and nanoparticles. artemisinin 193-204 transferrin receptor Homo sapiens 45-65 19957999-0 2010 Modular synthesis and in vitro and in vivo antimalarial assessment of C-10 pyrrole mannich base derivatives of artemisinin. artemisinin 111-122 homeobox C10 Homo sapiens 70-74 19913412-0 2010 Bio-transformation of artemisinin using soil microbe: Direct C-acetoxylation of artemisinin at C-9 by Penicillium simplissimum. artemisinin 22-33 complement C9 Homo sapiens 95-98 19913412-0 2010 Bio-transformation of artemisinin using soil microbe: Direct C-acetoxylation of artemisinin at C-9 by Penicillium simplissimum. artemisinin 80-91 complement C9 Homo sapiens 95-98 19913412-1 2010 Potent antimalarial compound artemisinin, 1 was bio-transformed to C-9 acetoxy artemisinin, 2 using soil microbe Penicillium simplissimum along with C-9 hydroxy derivative 3. artemisinin 29-40 complement C9 Homo sapiens 67-70 19913412-1 2010 Potent antimalarial compound artemisinin, 1 was bio-transformed to C-9 acetoxy artemisinin, 2 using soil microbe Penicillium simplissimum along with C-9 hydroxy derivative 3. artemisinin 29-40 complement C9 Homo sapiens 149-152 19846912-0 2009 Artemisinin-transferrin conjugate retards growth of breast tumors in the rat. artemisinin 0-11 transferrin Rattus norvegicus 12-23 19846912-3 2009 Previously, we found that covalently tagging artemisinin to transferrin enhanced the selectivity and toxicity of artemisinin toward cancer cells in vitro. artemisinin 45-56 transferrin Rattus norvegicus 60-71 19846912-3 2009 Previously, we found that covalently tagging artemisinin to transferrin enhanced the selectivity and toxicity of artemisinin toward cancer cells in vitro. artemisinin 113-124 transferrin Rattus norvegicus 60-71 19846912-4 2009 In the present research, artemisinin-transferrin conjugate was tested in a rat breast cancer model. artemisinin 25-36 transferrin Rattus norvegicus 37-48 19846912-6 2009 Once tumors were formed, daily intravenous injections of artemisinin-transferrin conjugate were administered. artemisinin 57-68 transferrin Rattus norvegicus 69-80 19846912-9 2009 CONCLUSION: Artemisinin-transferrin conjugate could be developed into a potent therapeutic agent for cancer in humans. artemisinin 12-23 transferrin Rattus norvegicus 24-35 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. artemisinin 138-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19562679-0 2009 Artemisinin--a possible CYP2B6 probe substrate? artemisinin 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 19562679-3 2009 Correlations between the metabolic rate constant for artemisinin and the other CYP2B6 substrates were examined. artemisinin 53-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 19562679-8 2009 CONCLUSIONS: The rate of in vitro metabolism of artemisinin was correlated significantly to that of bupropion, propofol and efavirenz, suggesting artemisinin to be a potential alternative marker to assess CYP2B6 activity. artemisinin 48-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 205-211 19562679-9 2009 Further studies characterizing the metabolic fate of artemisinin are needed in order to evaluate its utility as an in vitro and in vivo CYP2B6 probe substrate, since CYP2B6 might not be the only CYP isoform involved in the depletion of artemisinin. artemisinin 53-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-142 19562679-9 2009 Further studies characterizing the metabolic fate of artemisinin are needed in order to evaluate its utility as an in vitro and in vivo CYP2B6 probe substrate, since CYP2B6 might not be the only CYP isoform involved in the depletion of artemisinin. artemisinin 53-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-172 19562679-9 2009 Further studies characterizing the metabolic fate of artemisinin are needed in order to evaluate its utility as an in vitro and in vivo CYP2B6 probe substrate, since CYP2B6 might not be the only CYP isoform involved in the depletion of artemisinin. artemisinin 53-64 peptidylprolyl isomerase G Homo sapiens 136-139 19298255-0 2009 Artemisinin induces doxorubicin resistance in human colon cancer cells via calcium-dependent activation of HIF-1alpha and P-glycoprotein overexpression. artemisinin 0-11 hypoxia inducible factor 1 subunit alpha Homo sapiens 107-117 19298255-0 2009 Artemisinin induces doxorubicin resistance in human colon cancer cells via calcium-dependent activation of HIF-1alpha and P-glycoprotein overexpression. artemisinin 0-11 ATP binding cassette subfamily B member 1 Homo sapiens 122-136 19298255-2 2009 As the sesquiterpene lactone thapsigargin, a known inhibitor of mammalian SERCA, enhances the expression of P-glycoprotein (Pgp) by increasing the intracellular Ca(++) ([Ca(++)](i)) level, we investigated whether artemisinin and its structural homologue parthenolide could inhibit SERCA in human colon carcinoma HT29 cells and induce a resistance to doxorubicin. artemisinin 213-224 ATP binding cassette subfamily B member 1 Homo sapiens 108-122 19298255-2 2009 As the sesquiterpene lactone thapsigargin, a known inhibitor of mammalian SERCA, enhances the expression of P-glycoprotein (Pgp) by increasing the intracellular Ca(++) ([Ca(++)](i)) level, we investigated whether artemisinin and its structural homologue parthenolide could inhibit SERCA in human colon carcinoma HT29 cells and induce a resistance to doxorubicin. artemisinin 213-224 ATP binding cassette subfamily B member 1 Homo sapiens 124-127 19298255-8 2009 In HT29 cells, artemisinin and parthenolide induced the phosphorylation of HIF-1alpha, which was inhibited by KN93. artemisinin 15-26 hypoxia inducible factor 1 subunit alpha Homo sapiens 75-85 19298255-9 2009 CONCLUSIONS AND IMPLICATIONS: Our results suggest that artemisinin and parthenolide may act as SERCA inhibitors and, like other SERCA inhibitors, induce resistance to doxorubicin in human colon cancer cells, via the CaMKII-dependent activation of HIF-1alpha and the induction of Pgp. artemisinin 55-66 calcium/calmodulin dependent protein kinase II gamma Homo sapiens 216-222 19298255-9 2009 CONCLUSIONS AND IMPLICATIONS: Our results suggest that artemisinin and parthenolide may act as SERCA inhibitors and, like other SERCA inhibitors, induce resistance to doxorubicin in human colon cancer cells, via the CaMKII-dependent activation of HIF-1alpha and the induction of Pgp. artemisinin 55-66 hypoxia inducible factor 1 subunit alpha Homo sapiens 247-257 19298255-9 2009 CONCLUSIONS AND IMPLICATIONS: Our results suggest that artemisinin and parthenolide may act as SERCA inhibitors and, like other SERCA inhibitors, induce resistance to doxorubicin in human colon cancer cells, via the CaMKII-dependent activation of HIF-1alpha and the induction of Pgp. artemisinin 55-66 ATP binding cassette subfamily B member 1 Homo sapiens 279-282 18838215-2 2009 Artemisinin was covalently conjugated to a transferrin-receptor targeting peptide, HAIYPRH that binds to a cavity on the surface of transferrin receptor. artemisinin 0-11 transferrin Homo sapiens 43-54 18838215-2 2009 Artemisinin was covalently conjugated to a transferrin-receptor targeting peptide, HAIYPRH that binds to a cavity on the surface of transferrin receptor. artemisinin 0-11 transferrin Homo sapiens 132-143 18838215-3 2009 This enables artemisinin to be co-internalized with receptor-bound transferrin. artemisinin 13-24 transferrin Homo sapiens 67-78 18838215-4 2009 The iron released from transferrin can activate artemisinin to generate toxic radical species to kill cells. artemisinin 48-59 transferrin Homo sapiens 23-34 19408682-0 2009 [Antitumor mechanism of Qinghaosu derivatives--molecular docking studies of Qinghaosu derivatives with transferrin]. artemisinin 24-33 transferrin Homo sapiens 103-114 19408682-0 2009 [Antitumor mechanism of Qinghaosu derivatives--molecular docking studies of Qinghaosu derivatives with transferrin]. artemisinin 76-85 transferrin Homo sapiens 103-114 19408682-5 2009 Docking results unveil that Iron(II)-transferrin increased the cytotoxicity of Qinghaosu derivatives and provide a rational basis for further design and synthesis of novel Qinghaosu derivatives. artemisinin 79-88 transferrin Homo sapiens 37-48 19408682-5 2009 Docking results unveil that Iron(II)-transferrin increased the cytotoxicity of Qinghaosu derivatives and provide a rational basis for further design and synthesis of novel Qinghaosu derivatives. artemisinin 172-181 transferrin Homo sapiens 37-48 18793755-0 2008 Artemisinin can inhibit the calmodulin-mediated activation of phosphodiesterase in comparison with Cyclosporin A. artemisinin 0-11 calmodulin 1 Homo sapiens 28-38 18793755-1 2008 Artemisinin and Cyclosporin A were examined for their ability to inhibit the calmodulin-mediated activation of phosphodiesterase, which is based on the hydrolysis of cAMP to AMP by phosphodiesterase in the presence or absence of inhibitors, followed by quantitative analysis using spectrophotometer method. artemisinin 0-11 calmodulin 1 Homo sapiens 77-87 18793755-3 2008 Our results indicates that Artemisinin and Cyclosporin A induced some conformational changes on calmodulin and increased the fluorescence emission, but Artemisinin increased fluorescence emission of calmodulin in higher amounts compared with the Cyclosporin A. artemisinin 27-38 calmodulin 1 Homo sapiens 96-106 18793755-3 2008 Our results indicates that Artemisinin and Cyclosporin A induced some conformational changes on calmodulin and increased the fluorescence emission, but Artemisinin increased fluorescence emission of calmodulin in higher amounts compared with the Cyclosporin A. artemisinin 152-163 calmodulin 1 Homo sapiens 199-209 18793755-4 2008 Kinetic analysis of the Artemisinin-calmodulin and Cyclosporine A-calmodulin interaction showed that these agents competitively inhibited the activation of phosphodiesterase without affecting Vmax. artemisinin 24-35 calmodulin 1 Homo sapiens 36-46 18793755-8 2008 However, the degree of decrease in DeltaG (H2O) value was as follows: Artemisinin>Cyclosporin A, which means Artemisinin induced more instability in the calmodulin structure.In conclusion, our findings showed a good correlation between the ability of both Artemisinin and Cyclosporin A to block the activation of phosphodiesterase and their ability to bind to the activator and that Artemisinin is a more potent inhibitor of phosphodiesterase compared with Cyclosporin A. artemisinin 70-81 calmodulin 1 Homo sapiens 156-166 18793755-8 2008 However, the degree of decrease in DeltaG (H2O) value was as follows: Artemisinin>Cyclosporin A, which means Artemisinin induced more instability in the calmodulin structure.In conclusion, our findings showed a good correlation between the ability of both Artemisinin and Cyclosporin A to block the activation of phosphodiesterase and their ability to bind to the activator and that Artemisinin is a more potent inhibitor of phosphodiesterase compared with Cyclosporin A. artemisinin 112-123 calmodulin 1 Homo sapiens 156-166 18793755-8 2008 However, the degree of decrease in DeltaG (H2O) value was as follows: Artemisinin>Cyclosporin A, which means Artemisinin induced more instability in the calmodulin structure.In conclusion, our findings showed a good correlation between the ability of both Artemisinin and Cyclosporin A to block the activation of phosphodiesterase and their ability to bind to the activator and that Artemisinin is a more potent inhibitor of phosphodiesterase compared with Cyclosporin A. artemisinin 112-123 calmodulin 1 Homo sapiens 156-166 18793755-8 2008 However, the degree of decrease in DeltaG (H2O) value was as follows: Artemisinin>Cyclosporin A, which means Artemisinin induced more instability in the calmodulin structure.In conclusion, our findings showed a good correlation between the ability of both Artemisinin and Cyclosporin A to block the activation of phosphodiesterase and their ability to bind to the activator and that Artemisinin is a more potent inhibitor of phosphodiesterase compared with Cyclosporin A. artemisinin 112-123 calmodulin 1 Homo sapiens 156-166 18353653-1 2008 From 9-substituted DHA, several new artemisinin-derived C-10 acetal ethers and esters were prepared with either a 9-fluoro or a 9-sulfonyl substituent. artemisinin 36-47 homeobox C10 Homo sapiens 56-60 18456255-0 2008 Interferon-alpha enhances artemisinin-induced differentiation of HL-60 leukemia cells via a PKC alpha/ERK pathway. artemisinin 26-37 protein kinase C alpha Homo sapiens 92-101 18456255-0 2008 Interferon-alpha enhances artemisinin-induced differentiation of HL-60 leukemia cells via a PKC alpha/ERK pathway. artemisinin 26-37 mitogen-activated protein kinase 1 Homo sapiens 102-105 18456255-6 2008 The increased cell differentiation by IFN-alpha and artemisinin was significantly suppressed by the inhibitors for protein kinase C (PKC), extracellular signal-regulated kinase (ERK) and jun N-terminal kinase (JNK), but not by the inhibitors for phosphatidylinositol 3-kinase (PI3-K) and p38 mitogen-activated protein kinase (MAPK). artemisinin 52-63 protein kinase C alpha Homo sapiens 133-136 18456255-6 2008 The increased cell differentiation by IFN-alpha and artemisinin was significantly suppressed by the inhibitors for protein kinase C (PKC), extracellular signal-regulated kinase (ERK) and jun N-terminal kinase (JNK), but not by the inhibitors for phosphatidylinositol 3-kinase (PI3-K) and p38 mitogen-activated protein kinase (MAPK). artemisinin 52-63 mitogen-activated protein kinase 1 Homo sapiens 139-176 18456255-6 2008 The increased cell differentiation by IFN-alpha and artemisinin was significantly suppressed by the inhibitors for protein kinase C (PKC), extracellular signal-regulated kinase (ERK) and jun N-terminal kinase (JNK), but not by the inhibitors for phosphatidylinositol 3-kinase (PI3-K) and p38 mitogen-activated protein kinase (MAPK). artemisinin 52-63 mitogen-activated protein kinase 1 Homo sapiens 178-181 18456255-6 2008 The increased cell differentiation by IFN-alpha and artemisinin was significantly suppressed by the inhibitors for protein kinase C (PKC), extracellular signal-regulated kinase (ERK) and jun N-terminal kinase (JNK), but not by the inhibitors for phosphatidylinositol 3-kinase (PI3-K) and p38 mitogen-activated protein kinase (MAPK). artemisinin 52-63 mitogen-activated protein kinase 8 Homo sapiens 187-208 18456255-6 2008 The increased cell differentiation by IFN-alpha and artemisinin was significantly suppressed by the inhibitors for protein kinase C (PKC), extracellular signal-regulated kinase (ERK) and jun N-terminal kinase (JNK), but not by the inhibitors for phosphatidylinositol 3-kinase (PI3-K) and p38 mitogen-activated protein kinase (MAPK). artemisinin 52-63 mitogen-activated protein kinase 8 Homo sapiens 210-213 18456255-6 2008 The increased cell differentiation by IFN-alpha and artemisinin was significantly suppressed by the inhibitors for protein kinase C (PKC), extracellular signal-regulated kinase (ERK) and jun N-terminal kinase (JNK), but not by the inhibitors for phosphatidylinositol 3-kinase (PI3-K) and p38 mitogen-activated protein kinase (MAPK). artemisinin 52-63 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha Homo sapiens 246-275 18456255-6 2008 The increased cell differentiation by IFN-alpha and artemisinin was significantly suppressed by the inhibitors for protein kinase C (PKC), extracellular signal-regulated kinase (ERK) and jun N-terminal kinase (JNK), but not by the inhibitors for phosphatidylinositol 3-kinase (PI3-K) and p38 mitogen-activated protein kinase (MAPK). artemisinin 52-63 mitogen-activated protein kinase 14 Homo sapiens 288-324 18456255-6 2008 The increased cell differentiation by IFN-alpha and artemisinin was significantly suppressed by the inhibitors for protein kinase C (PKC), extracellular signal-regulated kinase (ERK) and jun N-terminal kinase (JNK), but not by the inhibitors for phosphatidylinositol 3-kinase (PI3-K) and p38 mitogen-activated protein kinase (MAPK). artemisinin 52-63 mitogen-activated protein kinase 1 Homo sapiens 326-330 18456255-8 2008 Taken together, these results indicate the enhancement of artemisinin-induced HL-60 cell differentiation by IFN-alpha through the activation of a PKC alpha/ERK signaling pathway, and suggest a possible use of IFN-alpha and artemisinin in the treatment of leukemic diseases. artemisinin 58-69 interferon alpha 1 Homo sapiens 108-117 18456255-8 2008 Taken together, these results indicate the enhancement of artemisinin-induced HL-60 cell differentiation by IFN-alpha through the activation of a PKC alpha/ERK signaling pathway, and suggest a possible use of IFN-alpha and artemisinin in the treatment of leukemic diseases. artemisinin 58-69 protein kinase C alpha Homo sapiens 146-155 18456255-8 2008 Taken together, these results indicate the enhancement of artemisinin-induced HL-60 cell differentiation by IFN-alpha through the activation of a PKC alpha/ERK signaling pathway, and suggest a possible use of IFN-alpha and artemisinin in the treatment of leukemic diseases. artemisinin 58-69 mitogen-activated protein kinase 1 Homo sapiens 156-159 18456255-8 2008 Taken together, these results indicate the enhancement of artemisinin-induced HL-60 cell differentiation by IFN-alpha through the activation of a PKC alpha/ERK signaling pathway, and suggest a possible use of IFN-alpha and artemisinin in the treatment of leukemic diseases. artemisinin 58-69 interferon alpha 1 Homo sapiens 209-218 18350255-0 2008 A model based assessment of the CYP2B6 and CYP2C19 inductive properties by artemisinin antimalarials: implications for combination regimens. artemisinin 75-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 18350255-0 2008 A model based assessment of the CYP2B6 and CYP2C19 inductive properties by artemisinin antimalarials: implications for combination regimens. artemisinin 75-86 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 43-50 18350255-6 2008 The production rate of CYP2C19 increased 51.2% by artemisinin, 14.8% by arteether and 24.9% by artemether. artemisinin 50-61 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 23-30 18350255-7 2008 In conclusion, all studied artemisinin derivatives induced CYP2B6. artemisinin 27-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 18350255-8 2008 CYP2C19 induction by arteether and artemether as well as CYP2B6 and CYP2C19 induction by artemisinin was confirmed. artemisinin 89-100 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 68-75 18350255-1 2008 The study aim was to assess the inductive properties of artemisinin antimalarials using mephenytoin as a probe for CYP2B6 and CYP2C19 enzymatic activity. artemisinin 56-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 18350255-1 2008 The study aim was to assess the inductive properties of artemisinin antimalarials using mephenytoin as a probe for CYP2B6 and CYP2C19 enzymatic activity. artemisinin 56-67 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 126-133 18350255-5 2008 The production rate of CYP2B6 was increased 79.7% by artemisinin, 61.5% by arteether, 76.1% by artemether, 19.9% by dihydroartemisinin and 16.9% by artesunate. artemisinin 53-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 18044948-8 2008 Along the artemisinin decomposition routes, especially B2 and B3, larger structural changes including formation of branched structures and CO2 release are related to increased exothermicity of the conversions, weakened attractive oxygen-oxygen interactions, and increased entropy of the formed species. artemisinin 10-21 immunoglobulin kappa variable 5-2 Homo sapiens 55-64 18223457-6 2008 PCR product sequencing was applied to regulatory regions of MDR1, the CYP3A4 proximal promoter, and to exons 2 and 5 of PXR, a gene coding for a nuclear factor activated by artemisinin antimalarials and associated with the transcription induction of most of the studied genes. artemisinin 173-184 nuclear receptor subfamily 1 group I member 2 Homo sapiens 120-123 18667841-0 2008 Artemisinin inhibits tumor lymphangiogenesis by suppression of vascular endothelial growth factor C. artemisinin 0-11 vascular endothelial growth factor C Mus musculus 63-99 18667841-6 2008 These data indicate that artemisinin may be useful for the prevention of lymph node metastasis by downregulating VEGF-C and reducing tumor lymphangiogenesis. artemisinin 25-36 vascular endothelial growth factor C Mus musculus 113-119 18667841-4 2008 Consistent with the decrease in lymph node metastasis, tumor lymphangiogenesis and expression of vascular endothelial growth factor C (VEGF-C) was significantly decreased in artemisinin-treated mice, as compared to control mice. artemisinin 174-185 vascular endothelial growth factor C Mus musculus 97-133 18667841-4 2008 Consistent with the decrease in lymph node metastasis, tumor lymphangiogenesis and expression of vascular endothelial growth factor C (VEGF-C) was significantly decreased in artemisinin-treated mice, as compared to control mice. artemisinin 174-185 vascular endothelial growth factor C Mus musculus 135-141 18667841-5 2008 Furthermore, IL-1beta-induced p38 mitogen-activated protein kinase (MAPK) activation and upregulation of VEGF-C mRNA and protein in LLC cells was also suppressed by artemisinin or by the p38 MAPK inhibitor SB-203580, suggesting that p38 MAPK could serve as a mediator of proinflammatory cytokine-induced VEGF-C expression. artemisinin 165-176 interleukin 1 beta Mus musculus 13-21 18667841-5 2008 Furthermore, IL-1beta-induced p38 mitogen-activated protein kinase (MAPK) activation and upregulation of VEGF-C mRNA and protein in LLC cells was also suppressed by artemisinin or by the p38 MAPK inhibitor SB-203580, suggesting that p38 MAPK could serve as a mediator of proinflammatory cytokine-induced VEGF-C expression. artemisinin 165-176 mitogen-activated protein kinase 14 Mus musculus 30-33 18667841-5 2008 Furthermore, IL-1beta-induced p38 mitogen-activated protein kinase (MAPK) activation and upregulation of VEGF-C mRNA and protein in LLC cells was also suppressed by artemisinin or by the p38 MAPK inhibitor SB-203580, suggesting that p38 MAPK could serve as a mediator of proinflammatory cytokine-induced VEGF-C expression. artemisinin 165-176 vascular endothelial growth factor C Mus musculus 105-111 18667841-5 2008 Furthermore, IL-1beta-induced p38 mitogen-activated protein kinase (MAPK) activation and upregulation of VEGF-C mRNA and protein in LLC cells was also suppressed by artemisinin or by the p38 MAPK inhibitor SB-203580, suggesting that p38 MAPK could serve as a mediator of proinflammatory cytokine-induced VEGF-C expression. artemisinin 165-176 mitogen-activated protein kinase 14 Mus musculus 233-241 18667841-5 2008 Furthermore, IL-1beta-induced p38 mitogen-activated protein kinase (MAPK) activation and upregulation of VEGF-C mRNA and protein in LLC cells was also suppressed by artemisinin or by the p38 MAPK inhibitor SB-203580, suggesting that p38 MAPK could serve as a mediator of proinflammatory cytokine-induced VEGF-C expression. artemisinin 165-176 vascular endothelial growth factor C Mus musculus 304-310 17521300-6 2007 The S-4"-hydroxymephenytoin/S-mephenytoin ratio (CYP2C19) was increased on day 5 by artemisinin [1.69 (1.47-1.94)] and arteether [1.33 (1.15-1.55)] compared with day -6. artemisinin 84-95 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 49-56 17521300-0 2007 Artemisinin antimalarials moderately affect cytochrome P450 enzyme activity in healthy subjects. artemisinin 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 17521300-7 2007 The paraxanthine/caffeine ratio (CYP1A2) was decreased on day 1 after administration of artemisinin [0.27 (0.18-0.39)], arteether [0.70 (0.55-0.89)] and dihydroartemisinin [0.73 (0.59-0.90)] compared with day -6. artemisinin 88-99 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 33-39 17521300-1 2007 The aim of this study was to investigate which principal human cytochrome P450 (CYP450) enzymes are affected by artemisinin and to what degree the artemisinin derivatives differ with respect to their respective induction and inhibition capacity. artemisinin 112-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 17521300-1 2007 The aim of this study was to investigate which principal human cytochrome P450 (CYP450) enzymes are affected by artemisinin and to what degree the artemisinin derivatives differ with respect to their respective induction and inhibition capacity. artemisinin 112-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-86 17521300-8 2007 The alpha-hydroxymetoprolol/metoprolol ratio (CYP2D6) was lower on day 1 compared with day -6 in the artemisinin [0.82 (0.70-0.96)] and dihydroartemisinin [0.83 (0.71-0.96)] groups, respectively. artemisinin 101-112 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 46-52 17521300-5 2007 The 1-hydroxymidazolam/midazolam 4-h plasma concentration ratio (CYP3A) was increased on day 5 by artemisinin [2.66-fold (98.75% CI: 2.10-3.36)], artemether [1.54 (1.14-2.09)] and dihydroartemisinin [1.25 (1.06-1.47)] compared with day -6. artemisinin 98-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-70 17521300-10 2007 These results show that intake of artemisinin antimalarials affect the activities of several principal human drug metabolizing CYP450 enzymes. artemisinin 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-133 17115150-11 2007 It is important to determine if these differences, along with previously reported differences in cytochrome P450 2B6 allele frequencies, are associated with altered metabolism/effectiveness of artemisinin drugs. artemisinin 193-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-116 17314215-2 2007 In this study, we explored the effect of artesunate, an artemisinin derivative, on tumour necrosis factor (TNF)-alpha-induced production of interleukins, IL-1beta, IL-6 and IL-8, in human rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLS), and further investigated the signal mechanism by which this compound modulates those cytokines" production. artemisinin 56-67 tumor necrosis factor Homo sapiens 83-117 17279482-6 2007 Among them, tanshinone IIA and cryptotanshinone are found to be potent inhibitors to CYP1A2, while artemisinin is a marginal inhibitor to CYP1A2 and glycyrrhetic acid is a weak inhibitor to CYP2C9. artemisinin 99-110 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 138-144 16412643-7 2006 Some reactivity of artemisinin with methemoglobin was indeed detected after addition of 50% v/v acetonitrile, most likely as a consequence of extensive protein unfolding. artemisinin 19-30 hemoglobin subunit gamma 2 Homo sapiens 36-49 16919048-3 2006 The objective of this study was to characterize the inductive properties of artemisinin on drug-metabolizing cytochrome P450 (CYP450) enzymes. artemisinin 76-87 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 109-124 16919048-3 2006 The objective of this study was to characterize the inductive properties of artemisinin on drug-metabolizing cytochrome P450 (CYP450) enzymes. artemisinin 76-87 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 126-132 16919048-4 2006 The possibility of artemisinin to induce CYP450 was studied in artemisinin-treated (orally for four days) and vehicle-treated rats using reverse transcriptase polymerase chain reaction (RT-PCR). artemisinin 19-30 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 41-47 16919048-6 2006 Increased CYP2B1 mRNA levels in rats could be seen after artemisinin treatment as well as a weak but reproducible increase in the intensity of CYP1A2. artemisinin 57-68 cytochrome P450, family 2, subfamily b, polypeptide 1 Rattus norvegicus 10-16 16919048-7 2006 Administration of artemisinin to mice up-regulated hepatic CYP2B10-dependent, and to a lesser extent, CYP2A5-dependent enzyme activities. artemisinin 18-29 cytochrome P450, family 2, subfamily b, polypeptide 10 Mus musculus 59-66 16919048-7 2006 Administration of artemisinin to mice up-regulated hepatic CYP2B10-dependent, and to a lesser extent, CYP2A5-dependent enzyme activities. artemisinin 18-29 cytochrome P450, family 2, subfamily a, polypeptide 5 Mus musculus 102-108 16919048-8 2006 In primary hepatocyte culture, artemisinin significantly increased the CYP2B10 mRNA levels whereas the CYP2A5 mRNA levels were increased to a lesser extent. artemisinin 31-42 cytochrome P450, family 2, subfamily b, polypeptide 10 Mus musculus 71-78 16919048-10 2006 Artemisinin was an activator of constitutive androstane receptor (CAR) but not pregnane X receptor (PXR) in HEK293 cells. artemisinin 0-11 nuclear receptor subfamily 1 group I member 3 Homo sapiens 32-64 16919048-10 2006 Artemisinin was an activator of constitutive androstane receptor (CAR) but not pregnane X receptor (PXR) in HEK293 cells. artemisinin 0-11 nuclear receptor subfamily 1 group I member 3 Homo sapiens 66-69 16919048-12 2006 The weaker up-regulation of CYP2A5 might also be CAR-dependent or alternatively, a consequence of artemisinin toxicity. artemisinin 98-109 cytochrome P450, family 2, subfamily a, polypeptide 5 Mus musculus 28-34 16919048-12 2006 The weaker up-regulation of CYP2A5 might also be CAR-dependent or alternatively, a consequence of artemisinin toxicity. artemisinin 98-109 nuclear receptor subfamily 1, group I, member 3 Mus musculus 49-52 16730793-0 2006 Study of the interaction of artemisinin with bovine serum albumin. artemisinin 28-39 albumin Homo sapiens 52-65 16730793-1 2006 The study on the interaction of artemisinin with bovine serum albumin (BSA) has been undertaken at three temperatures, 289, 296 and 303 K and investigated the effect of common ions and UV C (253.7 nm) irradiation on the binding of artemisinin with BSA. artemisinin 32-43 albumin Homo sapiens 56-69 16919048-0 2006 In vivo and mechanistic evidence of nuclear receptor CAR induction by artemisinin. artemisinin 70-81 nuclear receptor subfamily 1, group I, member 3 Mus musculus 53-56 16879742-2 2006 The change to artemisinin-based combination therapy (ACT) was initiated in 2004 using the co-blister of artesunate + sulfadoxine/pyrimethamine (AS+SP) and artemether + lumefantrine (ART+LUM), as first- and second-line, respectively. artemisinin 14-25 lumican Homo sapiens 186-189 16506047-1 2006 OBJECTIVE: Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of artemisinin drugs, a novel series of antimalarials. artemisinin 73-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-30 16506047-1 2006 OBJECTIVE: Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of artemisinin drugs, a novel series of antimalarials. artemisinin 73-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 15761118-0 2005 Antimalarial artemisinin drugs induce cytochrome P450 and MDR1 expression by activation of xenosensors pregnane X receptor and constitutive androstane receptor. artemisinin 13-24 ATP binding cassette subfamily B member 1 Homo sapiens 58-62 16512009-0 2006 [The OMS demands an immediate end to the commercialization of antimalarials only consisting of artemisinine]. artemisinin 95-107 chronic/recurrent otitis media Homo sapiens 5-8 16491843-4 2006 In regard to many possible functions, the TCTP of Plasmodium falciparum (Pf) is known to bind with an antimalarial agent, artemisinin, which is activated by heme. artemisinin 122-133 tumor protein, translationally-controlled 1 Homo sapiens 42-46 16185154-5 2005 By covalently tagging artemisinin to transferrin, artemisinin could be selectively picked up and concentrated by cancer cells. artemisinin 22-33 transferrin Homo sapiens 37-48 16185154-5 2005 By covalently tagging artemisinin to transferrin, artemisinin could be selectively picked up and concentrated by cancer cells. artemisinin 50-61 transferrin Homo sapiens 37-48 16185154-7 2005 Once an artemisinin-tagged transferrin molecule is endocytosed, iron is released and reacts with artemisinin moieties tagged to transferrin. artemisinin 8-19 transferrin Homo sapiens 27-38 16185154-7 2005 Once an artemisinin-tagged transferrin molecule is endocytosed, iron is released and reacts with artemisinin moieties tagged to transferrin. artemisinin 8-19 transferrin Homo sapiens 128-139 16185154-7 2005 Once an artemisinin-tagged transferrin molecule is endocytosed, iron is released and reacts with artemisinin moieties tagged to transferrin. artemisinin 97-108 transferrin Homo sapiens 27-38 16185154-7 2005 Once an artemisinin-tagged transferrin molecule is endocytosed, iron is released and reacts with artemisinin moieties tagged to transferrin. artemisinin 97-108 transferrin Homo sapiens 128-139 16185154-9 2005 The authors have found that artemisinin-tagged transferrin is highly selective and potent in killing cancer cells. artemisinin 28-39 transferrin Homo sapiens 47-58 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 97-108 NADH-ubiquinone reductase (H(+)-translocating) NDE1 Saccharomyces cerevisiae S288C 131-135 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 97-108 NADH-ubiquinone reductase (H(+)-translocating) NDI1 Saccharomyces cerevisiae S288C 139-143 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 97-108 NADH-ubiquinone reductase (H(+)-translocating) NDE1 Saccharomyces cerevisiae S288C 254-258 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 97-108 NADH-ubiquinone reductase (H(+)-translocating) NDI1 Saccharomyces cerevisiae S288C 262-266 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 215-226 NADH-ubiquinone reductase (H(+)-translocating) NDE1 Saccharomyces cerevisiae S288C 131-135 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 215-226 NADH-ubiquinone reductase (H(+)-translocating) NDI1 Saccharomyces cerevisiae S288C 139-143 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 215-226 NADH-ubiquinone reductase (H(+)-translocating) NDE1 Saccharomyces cerevisiae S288C 254-258 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 215-226 NADH-ubiquinone reductase (H(+)-translocating) NDI1 Saccharomyces cerevisiae S288C 262-266 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 215-226 NADH-ubiquinone reductase (H(+)-translocating) NDE1 Saccharomyces cerevisiae S288C 131-135 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 215-226 NADH-ubiquinone reductase (H(+)-translocating) NDI1 Saccharomyces cerevisiae S288C 139-143 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 215-226 NADH-ubiquinone reductase (H(+)-translocating) NDE1 Saccharomyces cerevisiae S288C 254-258 16170412-5 2005 Moreover, in a genetic study, we identify the electron transport chain as an important player in artemisinin"s action: Deletion of NDE1 or NDI1, which encode mitochondrial NADH dehydrogenases, confers resistance to artemisinin, whereas overexpression of NDE1 or NDI1 dramatically increases sensitivity to artemisinin. artemisinin 215-226 NADH-ubiquinone reductase (H(+)-translocating) NDI1 Saccharomyces cerevisiae S288C 262-266 16261361-0 2005 Artemisinin and thiabendazole are potent inhibitors of cytochrome P450 1A2 (CYP1A2) activity in humans. artemisinin 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 55-74 16261361-0 2005 Artemisinin and thiabendazole are potent inhibitors of cytochrome P450 1A2 (CYP1A2) activity in humans. artemisinin 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 76-82 16261361-1 2005 OBJECTIVE: To investigate the likelihood of artemisinin and thiabendazole causing pharmacokinetic interactions involving cytochrome P450 (CYP1A2) in humans given their potent inhibitory effects on the isoform in vitro. artemisinin 44-55 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 138-144 16261361-6 2005 RESULTS: Using the ratio of paraxanthine to caffeine after 4 h as an indicator of CYP1A2 activity, thiabendazole and artemisinin inhibited 92 and 66%, respectively, of the enzyme activity in vivo. artemisinin 117-128 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 82-88 16261361-10 2005 CONCLUSIONS: Co-administration of thiabendazole or artemisinin with CYP1A2 substrates could result in clinically significant effects. artemisinin 51-62 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 68-74 15761118-0 2005 Antimalarial artemisinin drugs induce cytochrome P450 and MDR1 expression by activation of xenosensors pregnane X receptor and constitutive androstane receptor. artemisinin 13-24 nuclear receptor subfamily 1 group I member 2 Homo sapiens 103-122 15761118-0 2005 Antimalarial artemisinin drugs induce cytochrome P450 and MDR1 expression by activation of xenosensors pregnane X receptor and constitutive androstane receptor. artemisinin 13-24 nuclear receptor subfamily 1 group I member 3 Homo sapiens 127-159 15761118-5 2005 Because the xenosensors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) have been shown to mediate induction of drug-metabolizing enzymes and drug transporters, we investigated the hypothesis that artemisinin induces cytochrome P450 expression by activating PXR and/or CAR. artemisinin 218-229 nuclear receptor subfamily 1 group I member 2 Homo sapiens 24-43 15761118-5 2005 Because the xenosensors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) have been shown to mediate induction of drug-metabolizing enzymes and drug transporters, we investigated the hypothesis that artemisinin induces cytochrome P450 expression by activating PXR and/or CAR. artemisinin 218-229 nuclear receptor subfamily 1 group I member 2 Homo sapiens 45-48 15761118-5 2005 Because the xenosensors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) have been shown to mediate induction of drug-metabolizing enzymes and drug transporters, we investigated the hypothesis that artemisinin induces cytochrome P450 expression by activating PXR and/or CAR. artemisinin 218-229 nuclear receptor subfamily 1 group I member 3 Homo sapiens 54-86 15761118-5 2005 Because the xenosensors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) have been shown to mediate induction of drug-metabolizing enzymes and drug transporters, we investigated the hypothesis that artemisinin induces cytochrome P450 expression by activating PXR and/or CAR. artemisinin 218-229 nuclear receptor subfamily 1 group I member 3 Homo sapiens 88-91 15761118-5 2005 Because the xenosensors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) have been shown to mediate induction of drug-metabolizing enzymes and drug transporters, we investigated the hypothesis that artemisinin induces cytochrome P450 expression by activating PXR and/or CAR. artemisinin 218-229 nuclear receptor subfamily 1 group I member 2 Homo sapiens 279-282 15761118-5 2005 Because the xenosensors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) have been shown to mediate induction of drug-metabolizing enzymes and drug transporters, we investigated the hypothesis that artemisinin induces cytochrome P450 expression by activating PXR and/or CAR. artemisinin 218-229 nuclear receptor subfamily 1 group I member 3 Homo sapiens 290-293 15761118-6 2005 By combining in vitro transfection methods and quantitative analyses of gene expression in cell lines and primary human hepatocytes, we here show that artemisinin drugs activate human PXR as well as human and mouse CAR and induce the expression of CYP2B6, CYP3A4, and MDR1 in primary human hepatocytes and in the human intestinal cell line LS174T. artemisinin 151-162 nuclear receptor subfamily 1 group I member 2 Homo sapiens 184-187 15761118-6 2005 By combining in vitro transfection methods and quantitative analyses of gene expression in cell lines and primary human hepatocytes, we here show that artemisinin drugs activate human PXR as well as human and mouse CAR and induce the expression of CYP2B6, CYP3A4, and MDR1 in primary human hepatocytes and in the human intestinal cell line LS174T. artemisinin 151-162 nuclear receptor subfamily 1, group I, member 3 Mus musculus 215-218 15761118-6 2005 By combining in vitro transfection methods and quantitative analyses of gene expression in cell lines and primary human hepatocytes, we here show that artemisinin drugs activate human PXR as well as human and mouse CAR and induce the expression of CYP2B6, CYP3A4, and MDR1 in primary human hepatocytes and in the human intestinal cell line LS174T. artemisinin 151-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 248-254 15761118-6 2005 By combining in vitro transfection methods and quantitative analyses of gene expression in cell lines and primary human hepatocytes, we here show that artemisinin drugs activate human PXR as well as human and mouse CAR and induce the expression of CYP2B6, CYP3A4, and MDR1 in primary human hepatocytes and in the human intestinal cell line LS174T. artemisinin 151-162 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 256-262 15761118-6 2005 By combining in vitro transfection methods and quantitative analyses of gene expression in cell lines and primary human hepatocytes, we here show that artemisinin drugs activate human PXR as well as human and mouse CAR and induce the expression of CYP2B6, CYP3A4, and MDR1 in primary human hepatocytes and in the human intestinal cell line LS174T. artemisinin 151-162 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 268-272 15761118-8 2005 In conclusion, activation of PXR and CAR and especially the resulting induction of CYP3A4 and MDR1 demonstrate that artemisinin has a higher risk of potential drug interactions than anticipated previously. artemisinin 116-127 nuclear receptor subfamily 1 group I member 2 Homo sapiens 29-32 15761118-8 2005 In conclusion, activation of PXR and CAR and especially the resulting induction of CYP3A4 and MDR1 demonstrate that artemisinin has a higher risk of potential drug interactions than anticipated previously. artemisinin 116-127 nuclear receptor subfamily 1 group I member 3 Homo sapiens 37-40 15761118-8 2005 In conclusion, activation of PXR and CAR and especially the resulting induction of CYP3A4 and MDR1 demonstrate that artemisinin has a higher risk of potential drug interactions than anticipated previously. artemisinin 116-127 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 15761118-8 2005 In conclusion, activation of PXR and CAR and especially the resulting induction of CYP3A4 and MDR1 demonstrate that artemisinin has a higher risk of potential drug interactions than anticipated previously. artemisinin 116-127 ATP binding cassette subfamily B member 1 Homo sapiens 94-98 15878303-8 2005 The Plasmodium translationally controlled tumor protein (TCTP) represents a known target protein of artemisinin and its derivatives in the malaria parasite. artemisinin 100-111 tumor protein, translationally-controlled 1 Homo sapiens 57-61 16019941-1 2005 The aim was to obtain information on the one-electron reduction of the antimalarial natural drug artemisinin (ART). artemisinin 97-108 artemin Homo sapiens 110-113 15782383-0 2005 C10-modified artemisinin derivatives: efficient heme-alkylating agents. artemisinin 13-24 homeobox C10 Homo sapiens 0-3 15642597-4 2005 In the present research, we covalently attached artemisinin to the iron-carrying plasma glycoprotein transferrin. artemisinin 48-59 transferrin Homo sapiens 101-112 15642597-6 2005 Thus, we hypothesize that by tagging artemisinin to transferrin, both iron and artemisinin would be transported into cancer cells in one package. artemisinin 37-48 transferrin Homo sapiens 52-63 15642597-6 2005 Thus, we hypothesize that by tagging artemisinin to transferrin, both iron and artemisinin would be transported into cancer cells in one package. artemisinin 79-90 transferrin Homo sapiens 52-63 15642597-7 2005 Once inside a cell, iron is released and can readily react with artemisinin close by tagged to the transferrin. artemisinin 64-75 transferrin Homo sapiens 99-110 15115411-1 2004 The synthesis of a series of C-10 trifluoromethyl ethers of artemisinin has been achieved from key bromide 8, itself carried out in two steps from artemisinin. artemisinin 60-71 gene rich cluster, C10 gene Mus musculus 29-33 15336316-3 2004 Iron(II)-glycine sulfate (Ferrosanol) and transferrin increased the cytotoxicity of free artesunate, artesunate microencapsulated in maltosyl-beta-cyclodextrin, and artemisinin toward CCRF-CEM leukemia and U373 astrocytoma cells 1.5- to 10.3-fold compared with that of artemisinins applied without iron. artemisinin 165-176 transferrin Homo sapiens 42-53 15115411-1 2004 The synthesis of a series of C-10 trifluoromethyl ethers of artemisinin has been achieved from key bromide 8, itself carried out in two steps from artemisinin. artemisinin 147-158 gene rich cluster, C10 gene Mus musculus 29-33 15115411-3 2004 The presence of the CF(3) group at C-10 of artemisinin clearly increased the chemical stability under simulated stomach acid conditions. artemisinin 43-54 gene rich cluster, C10 gene Mus musculus 35-39 12643911-1 2003 The alkylating properties of two artemisinin derivatives bearing a trifluoromethyl substituent at C10 were evaluated toward manganese(II) tetraphenylporphyrin, considered as a heme model. artemisinin 33-44 homeobox C10 Homo sapiens 98-101 14698358-0 2003 Postsynaptic dopamine (D(2))-mediated behavioural effects of high acute doses of artemisinin in rodents. artemisinin 81-92 dopamine receptor D2 Mus musculus 0-22 14660006-8 2003 Artemisinin enhanced PKC activity and protein level of PKC beta I isoform in only 1,25-(OH)(2)D(3)-treated HL-60 cells. artemisinin 0-11 proline rich transmembrane protein 2 Homo sapiens 55-58 14660006-9 2003 Taken together, these results indicate that artemisinin strongly enhanced 1,25-(OH)(2)D(3)- and all-trans RA-induced cell differentiation in which PKC is differentially involved in arteminisin-mediated enhancement of leukemia cell differentiation. artemisinin 44-55 proline rich transmembrane protein 2 Homo sapiens 147-150 13679021-4 2003 The natural antimalarial drug artemisinin is selectively toxic to iron-loaded cells (such as malarial parasites), and it has recently been suggested that, inasmuch as many cancers overexpress transferrin receptors, such cancers might be treatable with a regimen comprised of iron supplementation and high-dose artemisinin. artemisinin 30-41 transferrin Homo sapiens 192-203 13679021-4 2003 The natural antimalarial drug artemisinin is selectively toxic to iron-loaded cells (such as malarial parasites), and it has recently been suggested that, inasmuch as many cancers overexpress transferrin receptors, such cancers might be treatable with a regimen comprised of iron supplementation and high-dose artemisinin. artemisinin 310-321 transferrin Homo sapiens 192-203 12844133-0 2003 Artemisinin autoinduction is caused by involvement of cytochrome P450 2B6 but not 2C9. artemisinin 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-73 12844133-1 2003 AIM: Our goal was to investigate whether artemisinin autoinduction is caused by an increase in cytochrome P450 (CYP) 2B6 or CYP2C9 activities, we evaluated the effects of multiple-dose artemisinin administration on S-mephenytoin N-demethylation in healthy subjects. artemisinin 41-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-120 12844133-1 2003 AIM: Our goal was to investigate whether artemisinin autoinduction is caused by an increase in cytochrome P450 (CYP) 2B6 or CYP2C9 activities, we evaluated the effects of multiple-dose artemisinin administration on S-mephenytoin N-demethylation in healthy subjects. artemisinin 41-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 12844133-8 2003 The area under the concentration-time curve ratio of S-nirvanol/S-mephenytoin, an index of CYP2B6 activity, increased 1.9-fold (P <.05) in CYP2C19 poor metabolizers during artemisinin multiple-dose administration, whereas the urinary excretion ratio of hydroxytolbutamide plus carboxytolbutamide/tolbutamide remained constant during the study period. artemisinin 175-186 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 142-149 12844133-9 2003 CONCLUSIONS: These results indicate that artemisinin induces the N-demethylation of S-mephenytoin probably by an increased capacity of CYP2B6. artemisinin 41-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 12844133-10 2003 The autoinduction phenomenon of artemisinin may, therefore, be attributed, at least in part, to induction of CYP2B6, because this is the isozyme primarily involved in its metabolism. artemisinin 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 14698358-7 2003 Bromocriptine, a D(2) receptor agonist, induced locomotor activity in mice pretreated with reserpine which was attenuated by artemisinin. artemisinin 125-136 dopamine receptor D2 Mus musculus 17-30 14698358-8 2003 The results suggest that artemisinin possesses sedative property, which may be mediated via postsynaptic dopamine (D(2)) receptor in the CNS. artemisinin 25-36 dopamine receptor D2 Mus musculus 92-129 14660006-0 2003 Differential involvement of protein kinase C in human promyelocytic leukemia cell differentiation enhanced by artemisinin. artemisinin 110-121 proline rich transmembrane protein 2 Homo sapiens 28-44 14660006-7 2003 Particularly, protein kinase C (PKC) inhibitors inhibited HL-60 cell differentiation induced by artemisinin in combination with 1,25-(OH)(2)D(3) but not with all-trans RA. artemisinin 96-107 proline rich transmembrane protein 2 Homo sapiens 14-30 14660006-7 2003 Particularly, protein kinase C (PKC) inhibitors inhibited HL-60 cell differentiation induced by artemisinin in combination with 1,25-(OH)(2)D(3) but not with all-trans RA. artemisinin 96-107 proline rich transmembrane protein 2 Homo sapiens 32-35 14660006-8 2003 Artemisinin enhanced PKC activity and protein level of PKC beta I isoform in only 1,25-(OH)(2)D(3)-treated HL-60 cells. artemisinin 0-11 proline rich transmembrane protein 2 Homo sapiens 21-24 14615418-6 2003 Artemisinin down-regulated hypoxia-inducible factor-1alpha and vascular endothelial growth factor (VEGF) expression, which control endothelial cell growth. artemisinin 0-11 hypoxia inducible factor 1, alpha subunit Mus musculus 27-58 14615418-6 2003 Artemisinin down-regulated hypoxia-inducible factor-1alpha and vascular endothelial growth factor (VEGF) expression, which control endothelial cell growth. artemisinin 0-11 vascular endothelial growth factor A Mus musculus 63-97 14615418-6 2003 Artemisinin down-regulated hypoxia-inducible factor-1alpha and vascular endothelial growth factor (VEGF) expression, which control endothelial cell growth. artemisinin 0-11 vascular endothelial growth factor A Mus musculus 99-103 14615418-7 2003 The antiangiogenic effects and the inhibition of hypoxia-inducible factor-1alpha and VEGF were reversed upon cotreatment with the free radical scavengers mannitol and vitamin E, indicating that artemisinin may act via reactive oxygen species generation. artemisinin 194-205 hypoxia inducible factor 1, alpha subunit Mus musculus 49-80 14615418-7 2003 The antiangiogenic effects and the inhibition of hypoxia-inducible factor-1alpha and VEGF were reversed upon cotreatment with the free radical scavengers mannitol and vitamin E, indicating that artemisinin may act via reactive oxygen species generation. artemisinin 194-205 vascular endothelial growth factor A Mus musculus 85-89 11855985-10 2002 On the basis of the products obtained from biomimetic Fe(II) degradation of the C-10 carba analogue (23), we propose that these analogues may have a mode of action subtly different from that of the parent drug artemisinin (series 1 (7-16)) and other C-10 ether derivatives such as artemether. artemisinin 210-221 homeobox C10 Homo sapiens 80-84 12619052-3 2003 The metabolic fate of (14)C-artemisinin in microsomes from human B-lymphoblastoid cell lines transformed with CYP2A6, CYP2B6 and CYP3A4 was also investigated. artemisinin 28-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 12619052-3 2003 The metabolic fate of (14)C-artemisinin in microsomes from human B-lymphoblastoid cell lines transformed with CYP2A6, CYP2B6 and CYP3A4 was also investigated. artemisinin 28-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-135 11888669-0 2002 Transferrin overcomes drug resistance to artemisinin in human small-cell lung carcinoma cells. artemisinin 41-52 transferrin Homo sapiens 0-11 11697139-6 2001 Artemisinin was incubated with glutathione, NADPH and glutathione reductase and GSTs in a coupled assay system analogous to the standard assay scheme with cumene hydroperoxide as a substrate of GSTs. artemisinin 0-11 glutathione-disulfide reductase Homo sapiens 54-75 12536531-12 2001 Artemisinin has similar effectiveness to dexamethasone to inhibit the production of PLA2 in vitro. artemisinin 0-11 phospholipase A2 group IB Homo sapiens 84-88 11697139-6 2001 Artemisinin was incubated with glutathione, NADPH and glutathione reductase and GSTs in a coupled assay system analogous to the standard assay scheme with cumene hydroperoxide as a substrate of GSTs. artemisinin 0-11 glutathione S-transferase kappa 1 Homo sapiens 80-84 11697139-9 2001 Using recombinant GSTs the activity of GSTs with artemisinin was at least two fold higher than the reaction with CDNB. artemisinin 49-60 glutathione S-transferase kappa 1 Homo sapiens 18-22 11697139-9 2001 Using recombinant GSTs the activity of GSTs with artemisinin was at least two fold higher than the reaction with CDNB. artemisinin 49-60 glutathione S-transferase kappa 1 Homo sapiens 39-43 11697139-6 2001 Artemisinin was incubated with glutathione, NADPH and glutathione reductase and GSTs in a coupled assay system analogous to the standard assay scheme with cumene hydroperoxide as a substrate of GSTs. artemisinin 0-11 glutathione S-transferase kappa 1 Homo sapiens 194-198 11697139-10 2001 Considering these results, it is possible that GSTs may contribute to the metabolism of artemisinin in the presence of NADPH and GSSG-reductase. artemisinin 88-99 glutathione S-transferase kappa 1 Homo sapiens 47-51 11697139-11 2001 We propose a model, based on the known reactions of GSTs and sesquiterpenes, in which (1) artemisinin reacts with GSH resulting in oxidised glutathione; (2) the oxidised glutathione is then converted to reduced glutathione via glutathione reductase; and (3) the latter reaction may then result in the depletion of NADPH via GSSG-reductase. artemisinin 90-101 glutathione S-transferase kappa 1 Homo sapiens 52-56 11697139-7 2001 Artemisinin was shown to stimulate NADPH oxidation in cytosols from rat liver, kidney, intestines and in affinity purified preparations of GSTs from rat liver. artemisinin 0-11 glutathione S-transferase kappa 1 Homo sapiens 139-143 11697139-11 2001 We propose a model, based on the known reactions of GSTs and sesquiterpenes, in which (1) artemisinin reacts with GSH resulting in oxidised glutathione; (2) the oxidised glutathione is then converted to reduced glutathione via glutathione reductase; and (3) the latter reaction may then result in the depletion of NADPH via GSSG-reductase. artemisinin 90-101 glutathione-disulfide reductase Homo sapiens 227-248 11697139-8 2001 Using human recombinant GSTs hetelorogously expressed in Escherichia coli, artemisinin was similarly shown to stimulate NADPH oxidation with the highest activity observed with GST M1-1. artemisinin 75-86 glutathione S-transferase kappa 1 Homo sapiens 24-28 11697139-12 2001 The ability of artemisinin to react with GSH in the presence of GST may be responsible for the NADPH utilisation observed in vitro and suggests that cytosolic GSTs are likely to be contributing to metabolism of artemisinin and related drugs in vivo. artemisinin 15-26 glutathione S-transferase kappa 1 Homo sapiens 159-163 11697139-12 2001 The ability of artemisinin to react with GSH in the presence of GST may be responsible for the NADPH utilisation observed in vitro and suggests that cytosolic GSTs are likely to be contributing to metabolism of artemisinin and related drugs in vivo. artemisinin 211-222 glutathione S-transferase kappa 1 Homo sapiens 159-163 11575956-0 2001 Synthesis of new artemisinin analogues from artemisinic acid modified at C-3 and C-13 and their antimalarial activity. artemisinin 17-28 complement C3 Homo sapiens 73-76 11575956-0 2001 Synthesis of new artemisinin analogues from artemisinic acid modified at C-3 and C-13 and their antimalarial activity. artemisinin 17-28 homeobox C13 Homo sapiens 81-85 11124226-6 2001 Potent inhibitors of CYP1A2 were artemisinin, dihydroartemisinin, thiabendazole, primaquine, and niclosamide (K(i) = 0.43, 3.67, 1.54, 0.22, and 2.70 microM, respectively). artemisinin 33-44 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 21-27 11432537-10 2001 CYP2B6 and CYP3A4 are involved in the metabolism of artemisinin and derivatives, but further studies may reveal involvement of more enzymes. artemisinin 52-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 11432537-10 2001 CYP2B6 and CYP3A4 are involved in the metabolism of artemisinin and derivatives, but further studies may reveal involvement of more enzymes. artemisinin 52-63 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-17 11432537-11 2001 Artemisinin may induce CYP2C19. artemisinin 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 23-30 10543871-1 1999 Nine C-10 non-acetal derivatives of the natural trioxane artemisinin (1) were prepared as dimers using some novel chemistry. artemisinin 57-68 homeobox C10 Homo sapiens 5-9 10626755-0 1999 Stereospecific analysis of omeprazole supports artemisinin as a potent inducer of CYP2C19. artemisinin 47-58 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 82-89 10583023-6 1999 Recombinant CYP3A4 was catalytically competent in metabolizing artemisinin, although the rate was 10% of that for recombinant CYP2B6. artemisinin 63-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-18 10583023-10 1999 The rate of artemisinin metabolism in recombinant CYP2A6 was 15% of that for recombinant CYP2B6. artemisinin 12-23 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 50-56 10583023-10 1999 The rate of artemisinin metabolism in recombinant CYP2A6 was 15% of that for recombinant CYP2B6. artemisinin 12-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-95 10583023-11 1999 The inhibition of artemisinin metabolism in human liver microsomes by 8-methoxypsoralen (a CYP2A6 inhibitor) was 82% but CYP2A6 activity was not included in the regression tree. artemisinin 18-29 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 91-97 10583023-12 1999 CONCLUSIONS: Artemisinin metabolism in human liver microsomes is mediated primarily by CYP2B6 with probable secondary contribution of CYP3A4 in individuals with low CYP2B6 expression. artemisinin 13-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 10583023-12 1999 CONCLUSIONS: Artemisinin metabolism in human liver microsomes is mediated primarily by CYP2B6 with probable secondary contribution of CYP3A4 in individuals with low CYP2B6 expression. artemisinin 13-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 134-140 10583023-12 1999 CONCLUSIONS: Artemisinin metabolism in human liver microsomes is mediated primarily by CYP2B6 with probable secondary contribution of CYP3A4 in individuals with low CYP2B6 expression. artemisinin 13-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-171 10583023-0 1999 Identification of the human cytochrome P450 enzymes involved in the in vitro metabolism of artemisinin. artemisinin 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 10583023-1 1999 AIMS: The study aimed to identify the specific human cytochrome P450 (CYP450) enzymes involved in the metabolism of artemisinin. artemisinin 116-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 10583023-1 1999 AIMS: The study aimed to identify the specific human cytochrome P450 (CYP450) enzymes involved in the metabolism of artemisinin. artemisinin 116-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-76 10583023-2 1999 METHODS: Microsomes from human B-lymphoblastoid cell lines transformed with individual CYP450 cDNAs were investigated for their capacity to metabolize artemisinin. artemisinin 151-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-93 10583023-4 1999 The relative contribution of individual CYP450 isoenzymes to artemisinin metabolism in human liver microsomes was evaluated with a tree-based regression model of artemisinin disappearance rate and specific CYP450 activities. artemisinin 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-46 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 9929507-1 1999 The objective of this study was to investigate whether the decrease in artemisinin bioavailability after repeated oral dosing in humans can be a result of increased efflux of artemisinin by P-glycoprotein or decreased membrane transport at the intestinal barrier. artemisinin 71-82 ATP binding cassette subfamily B member 1 Homo sapiens 190-204 10626755-1 1999 The purpose of the study was to determine the enantiomer pharmacokinetics of omeprazole and 5-hydroxy-omeprazole before and after administration of the antimalarial artemisinin to confirm artemisinin"s ability to induce CYP2C19. artemisinin 188-199 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 220-227 10626755-11 1999 Artemisinin decreased the AUC of S-omeprazole to the same extent as that of R-omeprazole in extensive CYP2C19 metabolizers. artemisinin 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 102-109 10626755-12 1999 suggesting that artemisinin induces a different enzyme in addition to CYP2C19. artemisinin 16-27 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 70-77 10626755-13 1999 These results support and strengthen earlier findings that artemisinin induces CYP2C19 as well as at least one enzyme other than CYP3A4. artemisinin 59-70 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 79-86 10626755-13 1999 These results support and strengthen earlier findings that artemisinin induces CYP2C19 as well as at least one enzyme other than CYP3A4. artemisinin 59-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-135 9357485-1 1997 Bach Mai-Amsterdam Research Group on Artemisinin. artemisinin 37-48 acyl-CoA thioesterase 7 Homo sapiens 0-4 9728896-1 1998 OBJECTIVE: This study investigated whether time-dependent artemisinin pharmacokinetics correlated to CYP3A4 or CYP2C19 activity in vivo. artemisinin 58-69 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 101-107 9728896-1 1998 OBJECTIVE: This study investigated whether time-dependent artemisinin pharmacokinetics correlated to CYP3A4 or CYP2C19 activity in vivo. artemisinin 58-69 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 111-118 9728896-13 1998 The increased elimination of omeprazole in both poor and extensive CYP2C19 metabolizers suggests artemisinin induces both CYP2C19 and another enzyme. artemisinin 97-108 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 67-74 9728896-13 1998 The increased elimination of omeprazole in both poor and extensive CYP2C19 metabolizers suggests artemisinin induces both CYP2C19 and another enzyme. artemisinin 97-108 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 122-129 9604124-8 1998 Artemisinin oral plasma clearance was about 400 L h-1 exhibiting a slight decrease with dose, although the effect was weak. artemisinin 0-11 H1.5 linker histone, cluster member Homo sapiens 50-53 11324528-2 1998 By using patch-clamp whole cell recording configuration, the effects of five artemisinin-derivatives on the voltage-gated INa and IK have been studied on differentiated NG108-15 cells with reference to procaine. artemisinin 77-88 internexin neuronal intermediate filament protein, alpha Mus musculus 122-132 11324528-3 1998 The results showed that all the five artemisinin-derivatives clearly inhibited the voltage-gated sodium current (INa) of the cells in a dose-dependent manner and the effect was partially reversibly. artemisinin 37-48 internexin neuronal intermediate filament protein, alpha Mus musculus 131-135 9373657-0 1997 Severe and complicated malaria treated with artemisinin, artesunate or artemether in Viet Nam. artemisinin 44-55 SH3 and cysteine rich domain 3 Homo sapiens 90-93 9134741-1 1997 We determined the cytotoxicity of some artemisinin derivatives against EN2 tumor cells using the MTT assay. artemisinin 39-50 engrailed homeobox 2 Homo sapiens 71-74 7709742-6 1994 The highest rcross2 of alignment D, indicated that the side chain of -C6-O2-O1-C10-O3-C7-O4-C12-O5- and atom C16 are important groups of artemisinin analogs for antimalarial activity. artemisinin 137-148 chromosome 12 open reading frame 57 Homo sapiens 79-82 8080446-7 1994 However, artemisinin enhanced the inhibitory effect of iron (50 microM)-ascorbate (500 microM) on ATPase activity (46.3 +/- 3.9 vs 63 +/- 2.1% for basal; 57.2 +/- 2.5 vs 74.8 +/- 2.1% for CaM-activated). artemisinin 9-20 dynein axonemal heavy chain 8 Homo sapiens 98-104 8080446-10 1994 Artemisinin enhanced hemin (10 microM)-induced inhibition of basal (36.0 +/- 6.0 vs 73.7 +/- 3.0%) and CaM-activated Ca2+ pump ATPase (31.6 +/- 2.8 vs 70.0 +/- 1.5%). artemisinin 0-11 dynein axonemal heavy chain 8 Homo sapiens 127-133 34766463-8 2022 In addition, the synergistic effect of artemisinin and intracranial immunosuppression mediated by PD-L1 was more efficacious than either treatment alone. artemisinin 39-50 CD274 molecule Homo sapiens 98-103 33811898-0 2021 Artemisinin protects DPSC from hypoxia and TNF-alpha mediated osteogenesis impairments through CA9 and Wnt signaling pathway. artemisinin 0-11 tumor necrosis factor Homo sapiens 43-52 33811898-0 2021 Artemisinin protects DPSC from hypoxia and TNF-alpha mediated osteogenesis impairments through CA9 and Wnt signaling pathway. artemisinin 0-11 carbonic anhydrase 9 Homo sapiens 95-98 33811898-8 2021 Moreover, the beneficial effect of artemisinin was dependent on upregulated expression of CA9 and CA9-mediated antioxidant responses, as CA9 knockdown abolished the protective role of artemisinin on DPSC osteogenesis. artemisinin 35-46 carbonic anhydrase 9 Homo sapiens 90-93 33811898-8 2021 Moreover, the beneficial effect of artemisinin was dependent on upregulated expression of CA9 and CA9-mediated antioxidant responses, as CA9 knockdown abolished the protective role of artemisinin on DPSC osteogenesis. artemisinin 35-46 carbonic anhydrase 9 Homo sapiens 98-101 33811898-8 2021 Moreover, the beneficial effect of artemisinin was dependent on upregulated expression of CA9 and CA9-mediated antioxidant responses, as CA9 knockdown abolished the protective role of artemisinin on DPSC osteogenesis. artemisinin 35-46 carbonic anhydrase 9 Homo sapiens 98-101 33811898-8 2021 Moreover, the beneficial effect of artemisinin was dependent on upregulated expression of CA9 and CA9-mediated antioxidant responses, as CA9 knockdown abolished the protective role of artemisinin on DPSC osteogenesis. artemisinin 184-195 carbonic anhydrase 9 Homo sapiens 90-93 33811898-8 2021 Moreover, the beneficial effect of artemisinin was dependent on upregulated expression of CA9 and CA9-mediated antioxidant responses, as CA9 knockdown abolished the protective role of artemisinin on DPSC osteogenesis. artemisinin 184-195 carbonic anhydrase 9 Homo sapiens 98-101 33811898-8 2021 Moreover, the beneficial effect of artemisinin was dependent on upregulated expression of CA9 and CA9-mediated antioxidant responses, as CA9 knockdown abolished the protective role of artemisinin on DPSC osteogenesis. artemisinin 184-195 carbonic anhydrase 9 Homo sapiens 98-101 34606334-0 2022 Mutation in Plasmodium falciparum BTB/POZ domain of K13 protein confers artemisinin resistance. artemisinin 72-83 keratin 13 Homo sapiens 52-55 33588856-1 2021 BACKGROUND: The anti-malarial drug, amodiaquine, a commonly used, long-acting partner drug in artemisinin-based combination therapy, is metabolized to active desethyl-amodiaquine (DEAQ) by cytochrome P450 2C8 (CYP2C8). artemisinin 94-105 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 189-208 33588856-1 2021 BACKGROUND: The anti-malarial drug, amodiaquine, a commonly used, long-acting partner drug in artemisinin-based combination therapy, is metabolized to active desethyl-amodiaquine (DEAQ) by cytochrome P450 2C8 (CYP2C8). artemisinin 94-105 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 210-216 34900697-0 2021 Artemisinin Mediates Its Tumor-Suppressive Activity in Hepatocellular Carcinoma Through Targeted Inhibition of FoxM1. artemisinin 0-11 forkhead box M1 Homo sapiens 111-116 34931134-0 2021 Artemisinin Inhibits the Migration and Invasion in Uveal Melanoma via Inhibition of the PI3K/AKT/mTOR Signaling Pathway. artemisinin 0-11 AKT serine/threonine kinase 1 Homo sapiens 93-96 34931134-0 2021 Artemisinin Inhibits the Migration and Invasion in Uveal Melanoma via Inhibition of the PI3K/AKT/mTOR Signaling Pathway. artemisinin 0-11 mechanistic target of rapamycin kinase Homo sapiens 97-101 34931134-5 2021 Assessment of the mechanisms underlying artemisinin anticancer action revealed that its use dramatically reduced the phosphorylation of PI3K, AKT, and mTOR in UM cells. artemisinin 40-51 AKT serine/threonine kinase 1 Homo sapiens 142-145 34931134-5 2021 Assessment of the mechanisms underlying artemisinin anticancer action revealed that its use dramatically reduced the phosphorylation of PI3K, AKT, and mTOR in UM cells. artemisinin 40-51 mechanistic target of rapamycin kinase Homo sapiens 151-155 34774530-6 2021 Micromolar concentrations of several DIF derivatives strongly suppressed the growth of the four laboratory strains, including strains that exhibited resistance to chloroquine and artemisinin, as well as strains that were susceptible to these drugs. artemisinin 179-190 tumor necrosis factor Homo sapiens 37-40 34942304-6 2022 More importantly, the ART and PC co-loaded nanocomplexes showed the high efficacy against atherosclerosis of ApoE-/- mice model with both 8-week low dosage treatment or 1-week high dosage treatment. artemisinin 22-25 apolipoprotein E Mus musculus 109-113 34808366-6 2022 The overall results showed that artemisinin or its derivatives could significantly reduce fasting plasma glucose, 2-h plasma glucose (2hPG) in the intraperitoneal glucose tolerance test (IPGTT), 2hPG in the intraperitoneal insulin tolerance test (IPITT), glycated hemoglobin A1c, under the curve in the IPGTT/IPITT, total cholesterol, triglyceride, low-density lipoprotein cholesterol, free fatty acid, and urine volume. artemisinin 32-43 B cell leukemia/lymphoma 2 related protein A1c Mus musculus 275-278 34930267-5 2021 Further, the R561H mutation in the k13 gene, which confers artemisinin-resistance, was associated with delayed parasite clearance following treatment with artemether-lumefantrine in Rwanda in this study. artemisinin 59-70 keratin 13 Homo sapiens 35-38 34931134-7 2021 In contrast, AKT or mTOR activator (Sc79 and MHY1485, respectively) attenuated the inhibitory effect of artemisinin on the migration and invasion abilities of UM cells, further validating that artemisinin"s anticancer effect is likely to be mediated via inhibition of the PI3K/AKT/mTOR pathway. artemisinin 104-115 AKT serine/threonine kinase 1 Homo sapiens 13-16 34931134-7 2021 In contrast, AKT or mTOR activator (Sc79 and MHY1485, respectively) attenuated the inhibitory effect of artemisinin on the migration and invasion abilities of UM cells, further validating that artemisinin"s anticancer effect is likely to be mediated via inhibition of the PI3K/AKT/mTOR pathway. artemisinin 104-115 mechanistic target of rapamycin kinase Homo sapiens 20-24 34931134-7 2021 In contrast, AKT or mTOR activator (Sc79 and MHY1485, respectively) attenuated the inhibitory effect of artemisinin on the migration and invasion abilities of UM cells, further validating that artemisinin"s anticancer effect is likely to be mediated via inhibition of the PI3K/AKT/mTOR pathway. artemisinin 104-115 AKT serine/threonine kinase 1 Homo sapiens 277-280 34931134-7 2021 In contrast, AKT or mTOR activator (Sc79 and MHY1485, respectively) attenuated the inhibitory effect of artemisinin on the migration and invasion abilities of UM cells, further validating that artemisinin"s anticancer effect is likely to be mediated via inhibition of the PI3K/AKT/mTOR pathway. artemisinin 104-115 mechanistic target of rapamycin kinase Homo sapiens 281-285 34931134-7 2021 In contrast, AKT or mTOR activator (Sc79 and MHY1485, respectively) attenuated the inhibitory effect of artemisinin on the migration and invasion abilities of UM cells, further validating that artemisinin"s anticancer effect is likely to be mediated via inhibition of the PI3K/AKT/mTOR pathway. artemisinin 193-204 AKT serine/threonine kinase 1 Homo sapiens 13-16 34931134-7 2021 In contrast, AKT or mTOR activator (Sc79 and MHY1485, respectively) attenuated the inhibitory effect of artemisinin on the migration and invasion abilities of UM cells, further validating that artemisinin"s anticancer effect is likely to be mediated via inhibition of the PI3K/AKT/mTOR pathway. artemisinin 193-204 mechanistic target of rapamycin kinase Homo sapiens 281-285 34900697-3 2021 In our study, we have identified the antimalarial natural product, Artemisinin, to efficiently curb FoxM1 expression and activity in hepatic cancer cells, thereby exhibiting potential anticancer efficacy. artemisinin 67-78 forkhead box M1 Homo sapiens 100-105 34900697-4 2021 Here, we demonstrated that Artemisinin considerably mitigates FoxM1 transcriptional activity by disrupting its interaction with the promoter region of its downstream targets, thereby suppressing the expression of numerous oncogenic drivers. artemisinin 27-38 forkhead box M1 Homo sapiens 62-67 34900697-6 2021 Notably, FoxM1 overexpression rendered HCC cells poorly responsive to Artemisinin-mediated cytotoxicity while FoxM1 depletion in resistant liver cancer cells sensitized them to Artemisinin treatment, manifested in lower proliferative and growth index, drop in invasive potential and repressed expression of EMT markers with a concomitantly increased apoptosis. artemisinin 70-81 forkhead box M1 Homo sapiens 9-14 34900697-6 2021 Notably, FoxM1 overexpression rendered HCC cells poorly responsive to Artemisinin-mediated cytotoxicity while FoxM1 depletion in resistant liver cancer cells sensitized them to Artemisinin treatment, manifested in lower proliferative and growth index, drop in invasive potential and repressed expression of EMT markers with a concomitantly increased apoptosis. artemisinin 177-188 forkhead box M1 Homo sapiens 9-14 34900697-6 2021 Notably, FoxM1 overexpression rendered HCC cells poorly responsive to Artemisinin-mediated cytotoxicity while FoxM1 depletion in resistant liver cancer cells sensitized them to Artemisinin treatment, manifested in lower proliferative and growth index, drop in invasive potential and repressed expression of EMT markers with a concomitantly increased apoptosis. artemisinin 177-188 forkhead box M1 Homo sapiens 110-115 34900697-7 2021 Moreover, Artemisinin, when used in combination with Thiostrepton, an established FoxM1 inhibitor, markedly reduced anchorage-independent growth and displayed more pronounced death in liver cancer cells. artemisinin 10-21 forkhead box M1 Homo sapiens 82-87 34900697-9 2021 Altogether, our findings provide insight into the pivotal involvement of FoxM1 in the tumor suppressive activities of Artemisinin and shed light on the potential application of Artemisinin for improved therapeutic response, especially in resistant hepatic malignancies. artemisinin 118-129 forkhead box M1 Homo sapiens 73-78 34764635-7 2021 Incubation with artemisinin decreased basal vascular tension, NAD(P)H oxidase activity and reactive oxygen species (ROS) levels, but it also increased endothelial nitric oxide (NO) synthase (eNOS) activity and NO levels in the mesenteric artery, coronary artery, and pulmonary artery of SHRs. artemisinin 16-27 nitric oxide synthase 3 Rattus norvegicus 191-195 34832620-0 2021 Artemisinin Binds and Inhibits the Activity of Plasmodium falciparum Ddi1, a Retroviral Aspartyl Protease. artemisinin 0-11 DNA damage inducible 1 homolog 1 Homo sapiens 69-73 34592395-4 2021 In this work, we report a stable, safe and potent alternative artemisinin-based injectable nanocomplex consisting of dimeric artesunate-choline conjugate (dACC) micelles coated with hyaluronic acid (HA). artemisinin 62-73 Acetyl-CoA carboxylase Drosophila melanogaster 155-159 34764635-8 2021 Artemisinin chronic administration to SHRs increased the protein expression of eNOS and decreased the protein expression of the NAD(P)H oxidase subunits NOX-2 and NOX-4 in the mesenteric artery. artemisinin 0-11 nitric oxide synthase 3 Rattus norvegicus 79-83 34764635-8 2021 Artemisinin chronic administration to SHRs increased the protein expression of eNOS and decreased the protein expression of the NAD(P)H oxidase subunits NOX-2 and NOX-4 in the mesenteric artery. artemisinin 0-11 cytochrome b-245 beta chain Rattus norvegicus 153-158 34764635-8 2021 Artemisinin chronic administration to SHRs increased the protein expression of eNOS and decreased the protein expression of the NAD(P)H oxidase subunits NOX-2 and NOX-4 in the mesenteric artery. artemisinin 0-11 NADPH oxidase 4 Rattus norvegicus 163-168 34764635-9 2021 Conclusion: These results indicate that treatment with artemisinin has beneficial effects on reducing the heart rate and basal vascular tension and improving endothelium-dependent vascular relaxation in hypertension, which might occur by increasing eNOS activation and NO release and inhibiting NAD(P)H oxidase derived ROS production. artemisinin 55-66 nitric oxide synthase 3 Rattus norvegicus 249-253 34773836-1 2021 Decreased susceptibilities of the human malaria parasite Plasmodium falciparum towards the endoperoxide antimalarial artemisinin are linked to mutations of residue C580 of PfKelch13, a homologue of the redox sensor Keap1 and other vertebrate BTB-Kelch proteins. artemisinin 117-128 kelch like ECH associated protein 1 Homo sapiens 215-220 34288018-7 2021 Numerous NF-kappaB-regulated downstream genes are downregulated by artemisinin and its derivatives, for example, cytokines, chemokines, and immune receptors, which regulate immune cell differentiation, apoptosis genes, proliferation-regulating genes, signal transducers, and genes involved in antioxidant stress response. artemisinin 67-78 nuclear factor kappa B subunit 1 Homo sapiens 9-18 34270452-3 2021 Combining a published barcoding system of geographically restricted single-nucleotide polymorphisms (SNPs), mainly mitochondria of P. falciparum with SNPs in the K13 artemisinin resistance marker, could elucidate the parasite population structure and provide insight regarding the spread of drug resistance. artemisinin 166-177 keratin 13 Homo sapiens 162-165 34577169-0 2021 Molecular Basis of Artemisinin Derivatives Inhibition of Myeloid Differentiation Protein 2 by Combined in Silico and Experimental Study. artemisinin 19-30 lymphocyte antigen 96 Homo sapiens 57-90 34577169-3 2021 Recently, several reports mentioned that myeloid differentiation factor 2 (MD-2, also known as lymphocyte antigen 96) may be the endogenous target of artemisinin in the inhibition of lipopolysaccharide signaling. artemisinin 150-161 lymphocyte antigen 96 Homo sapiens 95-116 34577169-7 2021 Molecular docking results showed that artemisinin, artesunate, and artemether had similar binding poses, and all complexes remained stable throughout the whole molecular dynamics simulations, whereas the binding of artemisinin and its derivatives to MD-2 decreased the TLR4(Toll-Like Receptor 4)/MD-2 stability. artemisinin 38-49 toll like receptor 4 Homo sapiens 269-273 34577169-7 2021 Molecular docking results showed that artemisinin, artesunate, and artemether had similar binding poses, and all complexes remained stable throughout the whole molecular dynamics simulations, whereas the binding of artemisinin and its derivatives to MD-2 decreased the TLR4(Toll-Like Receptor 4)/MD-2 stability. artemisinin 38-49 toll like receptor 4 Homo sapiens 274-294 34577169-7 2021 Molecular docking results showed that artemisinin, artesunate, and artemether had similar binding poses, and all complexes remained stable throughout the whole molecular dynamics simulations, whereas the binding of artemisinin and its derivatives to MD-2 decreased the TLR4(Toll-Like Receptor 4)/MD-2 stability. artemisinin 215-226 toll like receptor 4 Homo sapiens 269-273 34577169-7 2021 Molecular docking results showed that artemisinin, artesunate, and artemether had similar binding poses, and all complexes remained stable throughout the whole molecular dynamics simulations, whereas the binding of artemisinin and its derivatives to MD-2 decreased the TLR4(Toll-Like Receptor 4)/MD-2 stability. artemisinin 215-226 toll like receptor 4 Homo sapiens 274-294 34279219-0 2021 Plasmodium falciparum K13 mutations in Africa and Asia impact artemisinin resistance and parasite fitness. artemisinin 62-73 keratin 13 Homo sapiens 22-25 34279219-1 2021 The emergence of mutant K13-mediated artemisinin (ART) resistance in Plasmodium falciparum malaria parasites has led to widespread treatment failure across Southeast Asia. artemisinin 37-48 keratin 13 Homo sapiens 24-27 34279219-1 2021 The emergence of mutant K13-mediated artemisinin (ART) resistance in Plasmodium falciparum malaria parasites has led to widespread treatment failure across Southeast Asia. artemisinin 50-53 keratin 13 Homo sapiens 24-27 34585460-9 2022 The 50% effective concentration (EC50 ) of artemisinin against SARS-CoV-2 Spike pseudovirus was found greater than 50 muM (EC45 ) in HEK293T cell line whereas the cell viability was 94% of the control (P < 0.01). artemisinin 43-54 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 74-79 34585460-10 2022 The immunosuppressive effects of artemisinin on TNF-alpha production on both pseudovirus and lipopolysaccharide (LPS)-induced THP-1 cells were found significant in a dose dependent manner. artemisinin 33-44 tumor necrosis factor Homo sapiens 48-57 34567410-0 2021 Artemisinin Reverses Glucocorticoid-Induced Injury in Bone Marrow-Derived Mesenchymal Stem Cells through Regulation of ERK1/2-CREB Signaling Pathway. artemisinin 0-11 mitogen-activated protein kinase 3 Homo sapiens 119-125 34567410-0 2021 Artemisinin Reverses Glucocorticoid-Induced Injury in Bone Marrow-Derived Mesenchymal Stem Cells through Regulation of ERK1/2-CREB Signaling Pathway. artemisinin 0-11 cAMP responsive element binding protein 1 Homo sapiens 126-130 34567410-4 2021 At relatively low concentrations, artemisinin treatment improved BMSC survival by promoting a decline of reactive oxygen species (ROS) production that correlated with the decrease of caspase-3 activation, LDH release, mitochondrial membrane potential (Deltapsim) loss, and apoptosis induced by dexamethasone (DEXA). artemisinin 34-45 caspase 3 Homo sapiens 183-192 34567410-7 2021 PD98059, the specific inhibitor of the ERK1/2 pathway, blocked ERK1/2 phosphorylation and artemisinin protection. artemisinin 90-101 mitogen-activated protein kinase 3 Homo sapiens 39-45 34567410-8 2021 Similarly, siCREB attenuated the protective effect of artemisinin, strongly suggesting the involvement of the ERK1/2-CREB pathway in the protective action of artemisinin against DEXA-induced damage in BMSCs. artemisinin 54-65 mitogen-activated protein kinase 3 Homo sapiens 110-116 34567410-8 2021 Similarly, siCREB attenuated the protective effect of artemisinin, strongly suggesting the involvement of the ERK1/2-CREB pathway in the protective action of artemisinin against DEXA-induced damage in BMSCs. artemisinin 54-65 cAMP responsive element binding protein 1 Homo sapiens 117-121 34567410-8 2021 Similarly, siCREB attenuated the protective effect of artemisinin, strongly suggesting the involvement of the ERK1/2-CREB pathway in the protective action of artemisinin against DEXA-induced damage in BMSCs. artemisinin 158-169 mitogen-activated protein kinase 3 Homo sapiens 110-116 34567410-8 2021 Similarly, siCREB attenuated the protective effect of artemisinin, strongly suggesting the involvement of the ERK1/2-CREB pathway in the protective action of artemisinin against DEXA-induced damage in BMSCs. artemisinin 158-169 cAMP responsive element binding protein 1 Homo sapiens 117-121 34567410-9 2021 In addition, we found that the expression of antiapoptotic protein B-cell lymphoma 2 protein (BCL-2) was also upregulated by artemisinin. artemisinin 125-136 BCL2 apoptosis regulator Homo sapiens 94-99 34567410-10 2021 These studies demonstrate the therapeutic potential of artemisinin in the survival improvement of BMSCs exposed to glucocorticoid-induced apoptosis and suggest that artemisinin-mediated protection may occur via the activation of ERK1/2-CREB signaling pathway. artemisinin 55-66 mitogen-activated protein kinase 3 Homo sapiens 229-235 34567410-10 2021 These studies demonstrate the therapeutic potential of artemisinin in the survival improvement of BMSCs exposed to glucocorticoid-induced apoptosis and suggest that artemisinin-mediated protection may occur via the activation of ERK1/2-CREB signaling pathway. artemisinin 55-66 cAMP responsive element binding protein 1 Homo sapiens 236-240 34567410-10 2021 These studies demonstrate the therapeutic potential of artemisinin in the survival improvement of BMSCs exposed to glucocorticoid-induced apoptosis and suggest that artemisinin-mediated protection may occur via the activation of ERK1/2-CREB signaling pathway. artemisinin 165-176 mitogen-activated protein kinase 3 Homo sapiens 229-235 34567410-10 2021 These studies demonstrate the therapeutic potential of artemisinin in the survival improvement of BMSCs exposed to glucocorticoid-induced apoptosis and suggest that artemisinin-mediated protection may occur via the activation of ERK1/2-CREB signaling pathway. artemisinin 165-176 cAMP responsive element binding protein 1 Homo sapiens 236-240 34515013-0 2022 Inactivation of PDH can Reduce Anaplastic Thyroid Cancer Cells" Sensitivity to Artemisinin. artemisinin 79-90 pyruvate dehydrogenase phosphatase catalytic subunit 1 Homo sapiens 16-19 34515013-7 2022 RESULTS: Artemisinin treatment significantly decreased the expression levels of COX2 and COX7A2 and increased that of COX14, YEM1l1, ALAS1, and OAT after 48h. artemisinin 9-20 mitochondrially encoded cytochrome c oxidase II Homo sapiens 80-84 34515013-7 2022 RESULTS: Artemisinin treatment significantly decreased the expression levels of COX2 and COX7A2 and increased that of COX14, YEM1l1, ALAS1, and OAT after 48h. artemisinin 9-20 cytochrome c oxidase subunit 7A2 Homo sapiens 89-95 34515013-7 2022 RESULTS: Artemisinin treatment significantly decreased the expression levels of COX2 and COX7A2 and increased that of COX14, YEM1l1, ALAS1, and OAT after 48h. artemisinin 9-20 cytochrome c oxidase assembly factor COX14 Homo sapiens 118-123 34515013-7 2022 RESULTS: Artemisinin treatment significantly decreased the expression levels of COX2 and COX7A2 and increased that of COX14, YEM1l1, ALAS1, and OAT after 48h. artemisinin 9-20 5'-aminolevulinate synthase 1 Homo sapiens 133-138 34515013-9 2022 The CAL-62 cells showed transient and reversible resistance to artemisinin, which was correlated to time-dependent changes in HIF1 , PDK1, and PDHA levels. artemisinin 63-74 hypoxia inducible factor 1 subunit alpha Homo sapiens 126-130 34515013-9 2022 The CAL-62 cells showed transient and reversible resistance to artemisinin, which was correlated to time-dependent changes in HIF1 , PDK1, and PDHA levels. artemisinin 63-74 pyruvate dehydrogenase kinase 1 Homo sapiens 133-137 34515013-9 2022 The CAL-62 cells showed transient and reversible resistance to artemisinin, which was correlated to time-dependent changes in HIF1 , PDK1, and PDHA levels. artemisinin 63-74 pyruvate dehydrogenase E1 subunit alpha 1 Homo sapiens 143-147 34515013-11 2022 CAL-62 cells show transient resistance to artemisinin via PDH downregulation, indicating that PDH activation may enhance the cytotoxic effects of artemisinin on ATC cells. artemisinin 42-53 pyruvate dehydrogenase phosphatase catalytic subunit 1 Homo sapiens 58-61 34515013-11 2022 CAL-62 cells show transient resistance to artemisinin via PDH downregulation, indicating that PDH activation may enhance the cytotoxic effects of artemisinin on ATC cells. artemisinin 42-53 pyruvate dehydrogenase phosphatase catalytic subunit 1 Homo sapiens 94-97 34515013-11 2022 CAL-62 cells show transient resistance to artemisinin via PDH downregulation, indicating that PDH activation may enhance the cytotoxic effects of artemisinin on ATC cells. artemisinin 146-157 pyruvate dehydrogenase phosphatase catalytic subunit 1 Homo sapiens 58-61 34515013-11 2022 CAL-62 cells show transient resistance to artemisinin via PDH downregulation, indicating that PDH activation may enhance the cytotoxic effects of artemisinin on ATC cells. artemisinin 146-157 pyruvate dehydrogenase phosphatase catalytic subunit 1 Homo sapiens 94-97 34358442-0 2022 Artemisinin inhibits NRas palmitoylation by targeting the protein acyltransferase ZDHHC6. artemisinin 0-11 NRAS proto-oncogene, GTPase Homo sapiens 21-25 34358442-0 2022 Artemisinin inhibits NRas palmitoylation by targeting the protein acyltransferase ZDHHC6. artemisinin 0-11 zinc finger DHHC-type palmitoyltransferase 6 Homo sapiens 82-88 34358442-2 2022 Here, we identify artemisinin (ART), a clinically approved antimalarial endoperoxide natural product with promising anticancer activities, as an inhibitor of the ER-residing palmitoyl transferase ZDHHC6 in cancer cells using a chemoproteomic approach. artemisinin 18-29 zinc finger DHHC-type palmitoyltransferase 6 Homo sapiens 196-202 34270452-13 2021 This study supports the use of mitochondrial SNPs to determine the origin of the parasite and suggests that the P. falciparum populations studied were susceptible to artemisinin during sampling because all K13 SNPs observed were outside the propeller domain for artemisinin resistance. artemisinin 262-273 keratin 13 Homo sapiens 206-209 34306988-3 2021 Our in silico approach suggested that unique phytocompounds such as emodin, thymol and carvacrol, and artemisinin could physically bind SARS-CoV-2 spike glycoproteins (6VXX and 6VYB), SARS-CoV-2 B.1.351 South Africa variant of Spike glycoprotein (7NXA), and even with ACE2 and prevent the SARS-CoV-2 binding to the host ACE2, TMPRSS2 and neutrapilin-1 receptors. artemisinin 102-113 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 147-152 34306988-3 2021 Our in silico approach suggested that unique phytocompounds such as emodin, thymol and carvacrol, and artemisinin could physically bind SARS-CoV-2 spike glycoproteins (6VXX and 6VYB), SARS-CoV-2 B.1.351 South Africa variant of Spike glycoprotein (7NXA), and even with ACE2 and prevent the SARS-CoV-2 binding to the host ACE2, TMPRSS2 and neutrapilin-1 receptors. artemisinin 102-113 angiotensin converting enzyme 2 Homo sapiens 320-324 34306988-3 2021 Our in silico approach suggested that unique phytocompounds such as emodin, thymol and carvacrol, and artemisinin could physically bind SARS-CoV-2 spike glycoproteins (6VXX and 6VYB), SARS-CoV-2 B.1.351 South Africa variant of Spike glycoprotein (7NXA), and even with ACE2 and prevent the SARS-CoV-2 binding to the host ACE2, TMPRSS2 and neutrapilin-1 receptors. artemisinin 102-113 transmembrane serine protease 2 Homo sapiens 326-333 34306988-4 2021 Since Chloroquine has been looked as potential therapy against COVID-19, we also compared the binding of chloroquine and artemisinin for its interaction with spike proteins (6VXX, 6VYB) and its variant 7NXA, respectively. artemisinin 121-132 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 158-163 34306988-9 2021 Emodin showed best interactions with TMPRSS 2 and ACE2 with Etotal of -7.1 and -7.3 KJ mol-1 respectively, whereas artemisinin interacts with TMPRSS 2 and ACE2 with Etotal of -6.9 and -7.4 KJ mol-1 respectively. artemisinin 115-126 transmembrane serine protease 2 Homo sapiens 142-150 34306988-9 2021 Emodin showed best interactions with TMPRSS 2 and ACE2 with Etotal of -7.1 and -7.3 KJ mol-1 respectively, whereas artemisinin interacts with TMPRSS 2 and ACE2 with Etotal of -6.9 and -7.4 KJ mol-1 respectively. artemisinin 115-126 angiotensin converting enzyme 2 Homo sapiens 155-159 34216470-1 2022 In Southeast Asia, mutations in the Plasmodium falciparum k13 gene have led to delayed parasite clearance and treatment failures in malaria patients receiving artemisinin combination therapies. artemisinin 159-170 keratin 13 Homo sapiens 58-61 34350834-7 2021 Connectivity Map and Comparative Toxicogenomics Database searches predicted dexamethasone may treat AF by reversing the expression of MIR100HG; artemisinin may reverse the expression of hub DEGs. artemisinin 144-155 mir-100-let-7a-2-mir-125b-1 cluster host gene Homo sapiens 134-142 34350834-7 2021 Connectivity Map and Comparative Toxicogenomics Database searches predicted dexamethasone may treat AF by reversing the expression of MIR100HG; artemisinin may reverse the expression of hub DEGs. artemisinin 144-155 delta 4-desaturase, sphingolipid 1 Homo sapiens 190-194 34073895-0 2021 Artemisinin Protects Porcine Mammary Epithelial Cells against Lipopolysaccharide-Induced Inflammatory Injury by Regulating the NF-kappaB and MAPK Signaling Pathways. artemisinin 0-11 nuclear factor kappa B subunit 1 Homo sapiens 127-136 34073895-4 2021 Pretreatment with 20 muM artemisinin weakened LPS-induced inflammatory damage in pMECs and decreased mRNA expression abundance and the content of inflammatory factors (IL-1beta, IL-6, and TNF-alpha) in pMECs (p < 0.05). artemisinin 25-36 interleukin 1 alpha Homo sapiens 168-176 34073895-4 2021 Pretreatment with 20 muM artemisinin weakened LPS-induced inflammatory damage in pMECs and decreased mRNA expression abundance and the content of inflammatory factors (IL-1beta, IL-6, and TNF-alpha) in pMECs (p < 0.05). artemisinin 25-36 interleukin 6 Homo sapiens 178-182 34073895-4 2021 Pretreatment with 20 muM artemisinin weakened LPS-induced inflammatory damage in pMECs and decreased mRNA expression abundance and the content of inflammatory factors (IL-1beta, IL-6, and TNF-alpha) in pMECs (p < 0.05). artemisinin 25-36 tumor necrosis factor Homo sapiens 188-197 34073895-5 2021 Mechanistically, artemisinin inhibited LPS-induced activation of the mitogen-activated protein kinase (MAPK) and nuclear factor-kappaB (NF-kappaB) signaling pathways. artemisinin 17-28 nuclear factor kappa B subunit 1 Homo sapiens 113-134 34073895-5 2021 Mechanistically, artemisinin inhibited LPS-induced activation of the mitogen-activated protein kinase (MAPK) and nuclear factor-kappaB (NF-kappaB) signaling pathways. artemisinin 17-28 nuclear factor kappa B subunit 1 Homo sapiens 136-145 34514004-7 2021 Results: Molecular docking studies indicated that hybrids of artemisinin and thymoquinone showed a relevant interaction with the active fraction of the enzyme Mpro, when compared to the reference drugs. artemisinin 61-72 NEWENTRY Severe acute respiratory syndrome-related coronavirus 159-163 34170261-10 2021 Simultaneous or separate administration of artemisinin and TSP-1-hEDSCs ameliorated this influence by considerably reducing Abeta plaque formation in the hippocampus, reducing glucose, MDA, ROS, and TNF-alpha levels, and increasing TAC levels. artemisinin 43-54 tumor necrosis factor Rattus norvegicus 199-208 34514004-11 2021 Conclusions: Hybrid products of artemisinin and thymoquinone have the potential to inhibit Mpro, with desirable pharmacokinetic and toxicity characteristics compared to commercially available drugs, being indicated for preclinical and subsequent clinical studies against SARS-CoV-2. artemisinin 32-43 NEWENTRY Severe acute respiratory syndrome-related coronavirus 91-95 35490765-1 2022 The current work aimed to enhance the oral bioavailability of water-insoluble drug Artemisinin (ART) by the inclusion of ART with hydroxypropyl-beta-cyclodextrin (HP-beta-CD) and then loaded with porous starch (PS). artemisinin 83-94 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 166-173 35348414-10 2022 CONCLUSIONS: Intervention with artemisinin in maternal HFD resulted in reduced islet size, decreased number of beta-cells and improved islet microcirculation, insulin processing shear process, decreased insulinogen/insulin ratio, and restored islet function through increased expression of PC1/3. artemisinin 31-42 proprotein convertase subtilisin/kexin type 1 Rattus norvegicus 290-295 35367585-1 2022 Dimeric artesunate phospholipid (ART-GPC), an amphiphilic derivative of artemisinin dimer reported in our previous work, can be applied to treat malaria effectively. artemisinin 72-83 glycophorin C (Gerbich blood group) Homo sapiens 37-40 35511795-2 2022 Genetic and epigenetic factors play a role in predisposing Plasmodium falciparum parasites to acquiring Pfkelch13 (K13) mutations associated with delayed artemisinin parasite clearance as reported in Southeast Asia. artemisinin 154-165 keratin 13 Homo sapiens 115-118 35511795-3 2022 In this study, we report on the prevalence of mutations in the K13, pfmdr-2 (P. falciparum multidrug resistance protein 2), fd (ferredoxin), pfcrt (P. falciparum chloroquine resistance transporter), and arps10 (apicoplast ribosomal protein S10) genes in Plasmodium falciparum parasites prior to (2005) and after (2013) introduction of artemisinin combination therapies for malaria treatment in Uganda. artemisinin 335-346 keratin 13 Homo sapiens 63-66 35349764-0 2022 Artemisinin upregulates neural cell adhesion molecule L1 to attenuate neurological deficits after intracerebral hemorrhage in mice. artemisinin 0-11 L1 cell adhesion molecule Mus musculus 24-56 35183684-3 2022 Here, we characterized the Plasmodium spp populations in wild Anopheles vectors by analyzing the genetic diversity of the P. falciparum kelch13 and mdr1 gene fragments implicated in artemisinin and partner drug resistance across Cameroon in three major malaria vectors. artemisinin 182-193 ATP binding cassette subfamily B member 1 Homo sapiens 148-152 35497913-8 2022 Finally, we verified the IL-17 signaling pathway and key cytokines, and ELISA and immunohistochemical results showed that artemisinin could downregulate the expression of proinflammatory cytokines such as IL-1beta and IL-17 in the IL-17 signaling pathway and upregulate the expression of the anti-inflammatory cytokine PPAR-gamma. artemisinin 122-133 interleukin 17A Rattus norvegicus 25-30 35497913-8 2022 Finally, we verified the IL-17 signaling pathway and key cytokines, and ELISA and immunohistochemical results showed that artemisinin could downregulate the expression of proinflammatory cytokines such as IL-1beta and IL-17 in the IL-17 signaling pathway and upregulate the expression of the anti-inflammatory cytokine PPAR-gamma. artemisinin 122-133 interleukin 1 alpha Rattus norvegicus 205-213 35497913-8 2022 Finally, we verified the IL-17 signaling pathway and key cytokines, and ELISA and immunohistochemical results showed that artemisinin could downregulate the expression of proinflammatory cytokines such as IL-1beta and IL-17 in the IL-17 signaling pathway and upregulate the expression of the anti-inflammatory cytokine PPAR-gamma. artemisinin 122-133 interleukin 17A Rattus norvegicus 218-223 35497913-8 2022 Finally, we verified the IL-17 signaling pathway and key cytokines, and ELISA and immunohistochemical results showed that artemisinin could downregulate the expression of proinflammatory cytokines such as IL-1beta and IL-17 in the IL-17 signaling pathway and upregulate the expression of the anti-inflammatory cytokine PPAR-gamma. artemisinin 122-133 interleukin 17A Rattus norvegicus 231-236 35497913-8 2022 Finally, we verified the IL-17 signaling pathway and key cytokines, and ELISA and immunohistochemical results showed that artemisinin could downregulate the expression of proinflammatory cytokines such as IL-1beta and IL-17 in the IL-17 signaling pathway and upregulate the expression of the anti-inflammatory cytokine PPAR-gamma. artemisinin 122-133 peroxisome proliferator-activated receptor gamma Rattus norvegicus 319-329 35370688-5 2022 The networks were constructed based on 50 artemisinin-disease intersection targets related to inflammation, cytokines, proliferation and apoptosis, showing the importance of GALNT2, BMP7 and TGFBR2 in the treatment of disease, which may be due to the occupation of the ricin B-type lectin domain of GALNT2 by artemisinin compounds or de novo designed candidates. artemisinin 42-53 polypeptide N-acetylgalactosaminyltransferase 2 Homo sapiens 174-180 35422868-9 2022 It was concluded that artemisinin treatment can reduce oxidative stress damage and alleviate CIRI through the SIRT1/FOXO1 signaling pathway, thereby achieving neuroprotective effects. artemisinin 22-33 sirtuin 1 Mus musculus 110-115 35422868-9 2022 It was concluded that artemisinin treatment can reduce oxidative stress damage and alleviate CIRI through the SIRT1/FOXO1 signaling pathway, thereby achieving neuroprotective effects. artemisinin 22-33 forkhead box O1 Mus musculus 116-121 35370688-5 2022 The networks were constructed based on 50 artemisinin-disease intersection targets related to inflammation, cytokines, proliferation and apoptosis, showing the importance of GALNT2, BMP7 and TGFBR2 in the treatment of disease, which may be due to the occupation of the ricin B-type lectin domain of GALNT2 by artemisinin compounds or de novo designed candidates. artemisinin 42-53 bone morphogenetic protein 7 Homo sapiens 182-186 35370688-5 2022 The networks were constructed based on 50 artemisinin-disease intersection targets related to inflammation, cytokines, proliferation and apoptosis, showing the importance of GALNT2, BMP7 and TGFBR2 in the treatment of disease, which may be due to the occupation of the ricin B-type lectin domain of GALNT2 by artemisinin compounds or de novo designed candidates. artemisinin 42-53 transforming growth factor beta receptor 2 Homo sapiens 191-197 35370688-5 2022 The networks were constructed based on 50 artemisinin-disease intersection targets related to inflammation, cytokines, proliferation and apoptosis, showing the importance of GALNT2, BMP7 and TGFBR2 in the treatment of disease, which may be due to the occupation of the ricin B-type lectin domain of GALNT2 by artemisinin compounds or de novo designed candidates. artemisinin 42-53 polypeptide N-acetylgalactosaminyltransferase 2 Homo sapiens 299-305 35370688-5 2022 The networks were constructed based on 50 artemisinin-disease intersection targets related to inflammation, cytokines, proliferation and apoptosis, showing the importance of GALNT2, BMP7 and TGFBR2 in the treatment of disease, which may be due to the occupation of the ricin B-type lectin domain of GALNT2 by artemisinin compounds or de novo designed candidates. artemisinin 309-320 polypeptide N-acetylgalactosaminyltransferase 2 Homo sapiens 174-180 35370688-5 2022 The networks were constructed based on 50 artemisinin-disease intersection targets related to inflammation, cytokines, proliferation and apoptosis, showing the importance of GALNT2, BMP7 and TGFBR2 in the treatment of disease, which may be due to the occupation of the ricin B-type lectin domain of GALNT2 by artemisinin compounds or de novo designed candidates. artemisinin 309-320 bone morphogenetic protein 7 Homo sapiens 182-186 35370688-5 2022 The networks were constructed based on 50 artemisinin-disease intersection targets related to inflammation, cytokines, proliferation and apoptosis, showing the importance of GALNT2, BMP7 and TGFBR2 in the treatment of disease, which may be due to the occupation of the ricin B-type lectin domain of GALNT2 by artemisinin compounds or de novo designed candidates. artemisinin 309-320 transforming growth factor beta receptor 2 Homo sapiens 191-197 35370688-5 2022 The networks were constructed based on 50 artemisinin-disease intersection targets related to inflammation, cytokines, proliferation and apoptosis, showing the importance of GALNT2, BMP7 and TGFBR2 in the treatment of disease, which may be due to the occupation of the ricin B-type lectin domain of GALNT2 by artemisinin compounds or de novo designed candidates. artemisinin 309-320 polypeptide N-acetylgalactosaminyltransferase 2 Homo sapiens 299-305 35293664-8 2022 The therapeutic dose of ART reduced the CAT activity and the GSH level, nonsignificantly. artemisinin 24-27 catalase Mus musculus 40-43 35293664-9 2022 The toxic dose of ART reduced the CAT activity and increased the POX activity. artemisinin 18-21 catalase Mus musculus 34-37 35215242-0 2022 Antiviral Effects of Artemisinin and Its Derivatives against SARS-CoV-2 Main Protease: Computational Evidences and Interactions with ACE2 Allelic Variants. artemisinin 21-32 angiotensin converting enzyme 2 Homo sapiens 133-137 35345364-0 2022 Design and Development of Nanostructured Co Delivery of Artemisinin and Chrysin for Targeting hTERT Gene Expression in Breast Cancer Cell Line: Possible Clinical Application in Cancer Treatment. artemisinin 56-67 telomerase reverse transcriptase Homo sapiens 94-99 34978886-5 2022 In this study, we compared the asexual and sexual stage activities of cipargamin, ganaplacide and artesunate in artemisinin resistant P. falciparum isolates (N=7, K13 mutation; C580Y, G449A and R539T) from Thailand and Cambodia. artemisinin 112-123 keratin 13 Homo sapiens 163-166 35045938-0 2022 Corrigendum to "Artemisinin protects DPSC from hypoxia and TNF-alpha mediated osteogenesis impairments through CA9 and Wnt signaling pathway" (Life Sci. artemisinin 16-27 tumor necrosis factor Homo sapiens 59-68 35045938-0 2022 Corrigendum to "Artemisinin protects DPSC from hypoxia and TNF-alpha mediated osteogenesis impairments through CA9 and Wnt signaling pathway" (Life Sci. artemisinin 16-27 carbonic anhydrase 9 Homo sapiens 111-114 35151247-9 2022 To verify the mechanism of cell death induced by artemisinin in A-253 cells, we found an increased level of Bax, Bim, Bad, Bak and reduced level of antiapoptotic protein Bcl-2, Bcl-XL with concomitant release of mitochondrial resident protein cytochrome c into the cytoplasm. artemisinin 49-60 BCL2 associated X, apoptosis regulator Homo sapiens 108-111 35151247-9 2022 To verify the mechanism of cell death induced by artemisinin in A-253 cells, we found an increased level of Bax, Bim, Bad, Bak and reduced level of antiapoptotic protein Bcl-2, Bcl-XL with concomitant release of mitochondrial resident protein cytochrome c into the cytoplasm. artemisinin 49-60 BCL2 like 11 Homo sapiens 113-116 35151247-9 2022 To verify the mechanism of cell death induced by artemisinin in A-253 cells, we found an increased level of Bax, Bim, Bad, Bak and reduced level of antiapoptotic protein Bcl-2, Bcl-XL with concomitant release of mitochondrial resident protein cytochrome c into the cytoplasm. artemisinin 49-60 BCL2 antagonist/killer 1 Homo sapiens 123-126 35151247-9 2022 To verify the mechanism of cell death induced by artemisinin in A-253 cells, we found an increased level of Bax, Bim, Bad, Bak and reduced level of antiapoptotic protein Bcl-2, Bcl-XL with concomitant release of mitochondrial resident protein cytochrome c into the cytoplasm. artemisinin 49-60 BCL2 apoptosis regulator Homo sapiens 170-175 35151247-9 2022 To verify the mechanism of cell death induced by artemisinin in A-253 cells, we found an increased level of Bax, Bim, Bad, Bak and reduced level of antiapoptotic protein Bcl-2, Bcl-XL with concomitant release of mitochondrial resident protein cytochrome c into the cytoplasm. artemisinin 49-60 BCL2 like 1 Homo sapiens 177-183 35151247-9 2022 To verify the mechanism of cell death induced by artemisinin in A-253 cells, we found an increased level of Bax, Bim, Bad, Bak and reduced level of antiapoptotic protein Bcl-2, Bcl-XL with concomitant release of mitochondrial resident protein cytochrome c into the cytoplasm. artemisinin 49-60 cytochrome c, somatic Homo sapiens 243-255 35151247-10 2022 Additionally, we found that artemisinin augments the production of reactive oxygen species which further leads to the activation of proapoptotic proteins PARP1, and caspase-3, in a concentration-dependent manner thereby triggering apoptosis. artemisinin 28-39 poly(ADP-ribose) polymerase 1 Homo sapiens 154-159 35151247-10 2022 Additionally, we found that artemisinin augments the production of reactive oxygen species which further leads to the activation of proapoptotic proteins PARP1, and caspase-3, in a concentration-dependent manner thereby triggering apoptosis. artemisinin 28-39 caspase 3 Homo sapiens 165-174 35204156-0 2022 Effect of Artemisinin on the Redox System of NADPH/FNR/Ferredoxin from Malaria Parasites. artemisinin 10-21 2,4-dienoyl-CoA reductase 1 Homo sapiens 45-65 35215242-8 2022 Furthermore, artemisinin interactions with angiotensin converting enzyme 2 (ACE2) were dependent on the ACE2 allelic variants. artemisinin 13-24 angiotensin converting enzyme 2 Homo sapiens 43-74 35215242-8 2022 Furthermore, artemisinin interactions with angiotensin converting enzyme 2 (ACE2) were dependent on the ACE2 allelic variants. artemisinin 13-24 angiotensin converting enzyme 2 Homo sapiens 76-80 35215242-8 2022 Furthermore, artemisinin interactions with angiotensin converting enzyme 2 (ACE2) were dependent on the ACE2 allelic variants. artemisinin 13-24 angiotensin converting enzyme 2 Homo sapiens 104-108 35215242-10 2022 A molecular dynamic simulation showed sufficient stability of the artemisinin-Mpro complex on the trajectory of 100 ns simulation frame. artemisinin 66-77 NEWENTRY Severe acute respiratory syndrome-related coronavirus 78-82 35215242-11 2022 These binding interactions, together with drug-likeness and pharmacokinetic findings, confirmed that artemisinin might inhibit Mpro activity and explain the ethnopharmacological use of the herb and its possible antiviral activity against SARS-CoV-2 infection inducing COVID-19. artemisinin 101-112 NEWENTRY Severe acute respiratory syndrome-related coronavirus 127-131 35002180-3 2022 Herein, we investigated the potential of artemisinin and its derivatives as possible inhibitors of SARS-CoV-2 Nsp1 through various computational approaches. artemisinin 41-52 SH2 domain containing 3A Homo sapiens 110-114 35047402-5 2021 Recently, we have shown that artemisinin derivatives enhance TRAIL-induced apoptosis in colon cancer cells. artemisinin 29-40 TNF superfamily member 10 Homo sapiens 61-66 35002180-4 2022 Molecular docking results show that artemisinin (CID68827) binds to Nsp1 with a binding energy of -6.53 kcal/mol and an inhibition constant of 16.43 microM. artemisinin 36-47 SH2 domain containing 3A Homo sapiens 68-72 35002180-9 2022 Thus, the findings of our research highlight the importance of artemisinin and its derivatives in the development of drugs to inhibit SARS-CoV-2 Nsp1 protein. artemisinin 63-74 SH2 domain containing 3A Homo sapiens 145-149 34975311-3 2022 Specifically, both in vitro and in vivo experiments showed that HF or HF-ATS induces apoptosis via activation of caspase-9 and caspase-8 while only caspase-9 is involved in ATS-induced apoptosis. artemisinin 173-176 caspase 9 Homo sapiens 148-157