PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 20214592-6 2009 CYP2C8 is a major catalyst in the metabolism of paclitaxel, amodiaquine, troglitazone, amiodarone, verapamil and ibuprofen, with a secondary role in the biotransformation of cerivastatin and fluvastatin. Amodiaquine 60-71 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 0-6 19926036-5 2009 For example, there is a clear association between concentrations of proguanil within plasma and certain genetic polymorphisms of CYP2C19, and genetically established levels of CYP2C8 might have important clinical implications in the toxicity of amodiaquine. Amodiaquine 245-256 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 129-136 19926036-5 2009 For example, there is a clear association between concentrations of proguanil within plasma and certain genetic polymorphisms of CYP2C19, and genetically established levels of CYP2C8 might have important clinical implications in the toxicity of amodiaquine. Amodiaquine 245-256 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 176-182 20126327-2 2009 Both subjects were homozygous for the wild type allele of cytochrome P450 2C8, the main enzyme responsible for amodiaquine metabolism. Amodiaquine 111-122 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 58-77 19074972-0 2009 Novel metabolites of amodiaquine formed by CYP1A1 and CYP1B1: structure elucidation using electrochemistry, mass spectrometry, and NMR. Amodiaquine 21-32 cytochrome P450, family 1, subfamily a, polypeptide 1 Rattus norvegicus 43-49 19184353-6 2009 Histamine fluxes accounted for <25 % of hist-rad fluxes, but this percentage increased after blocking HMT (100 microM amodiaquin) or DAO (100 microM aminoguanidine). Amodiaquine 121-131 histamine N-methyltransferase Sus scrofa 105-108 19074972-0 2009 Novel metabolites of amodiaquine formed by CYP1A1 and CYP1B1: structure elucidation using electrochemistry, mass spectrometry, and NMR. Amodiaquine 21-32 cytochrome P450, family 1, subfamily b, polypeptide 1 Rattus norvegicus 54-60 19074972-9 2009 Three additional new metabolites of amodiaquine and desethylamodiaquine were formed via rCYP1A1 and rCYP1B1. Amodiaquine 36-47 cytochrome P450, family 1, subfamily a, polypeptide 1 Rattus norvegicus 88-95 19074972-9 2009 Three additional new metabolites of amodiaquine and desethylamodiaquine were formed via rCYP1A1 and rCYP1B1. Amodiaquine 36-47 cytochrome P450, family 1, subfamily b, polypeptide 1 Rattus norvegicus 100-107 29788622-4 2008 Amodiaquine and isoniazid are metabolized polymorphically by CYP2C8 and N-acetyltransferase 2, respectively. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 61-67 18779360-0 2008 Effect of concomitant artesunate administration and cytochrome P4502C8 polymorphisms on the pharmacokinetics of amodiaquine in Ghanaian children with uncomplicated malaria. Amodiaquine 112-123 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 52-70 29788622-4 2008 Amodiaquine and isoniazid are metabolized polymorphically by CYP2C8 and N-acetyltransferase 2, respectively. Amodiaquine 0-11 N-acetyltransferase 2 Homo sapiens 72-93 18855526-2 2008 Amodiaquine is mainly metabolized hepatically towards its major active metabolite desethylamodiaquine, by the polymorphic P450 isoform CYP2C8. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 135-141 17361129-0 2007 Amodiaquine metabolism is impaired by common polymorphisms in CYP2C8: implications for malaria treatment in Africa. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 62-68 18444659-2 2008 We investigated in this study the down-regulative effects of 5-caffeoylquinic acid (CQA), the predominant isomer of CHA, on the H(2)O(2-) or TNF-alpha-induced secretion of interleukin (IL)-8, a central pro-inflammatory chemokine involved in the pathogenesis of inflammatory bowel diseases, in human intestinal epithelial Caco-2 cells. Amodiaquine 84-87 tumor necrosis factor Homo sapiens 141-150 18444659-2 2008 We investigated in this study the down-regulative effects of 5-caffeoylquinic acid (CQA), the predominant isomer of CHA, on the H(2)O(2-) or TNF-alpha-induced secretion of interleukin (IL)-8, a central pro-inflammatory chemokine involved in the pathogenesis of inflammatory bowel diseases, in human intestinal epithelial Caco-2 cells. Amodiaquine 84-87 C-X-C motif chemokine ligand 8 Homo sapiens 172-190 18444659-3 2008 After the cells had been pre- and simultaneously treated with CQA, the oversecretion of IL-8 and overexpression of its mRNA induced by H(2)O(2) were significantly suppressed in a dose-dependent manner in the range of 0.25-2.00 mmol/L. Amodiaquine 62-65 C-X-C motif chemokine ligand 8 Homo sapiens 88-92 18444659-4 2008 We further found that a metabolite of CQA, caffeic acid (CA), but not quinic acid, significantly inhibited the H(2)O(2)-induced IL-8 secretion and its mRNA expression in the same dose-dependent manner. Amodiaquine 38-41 C-X-C motif chemokine ligand 8 Homo sapiens 128-132 18255358-1 2008 The antimalarial drug amodiaquine is extensively metabolized to N-desethylamodiaquine (DEAQ) by cytochrome P450 2C8 (CYP2C8). Amodiaquine 22-33 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 96-115 18255358-1 2008 The antimalarial drug amodiaquine is extensively metabolized to N-desethylamodiaquine (DEAQ) by cytochrome P450 2C8 (CYP2C8). Amodiaquine 22-33 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 117-123 18758509-6 2008 The alkalinizing lysosomotropic drugs chloroquine, hydroxychloroquine, amodiaquine, and azithromycin had a similar effect on the overall production of mature bioactive TGFbeta. Amodiaquine 71-82 transforming growth factor beta 1 Homo sapiens 168-175 17361129-1 2007 Metabolism of the antimalarial drug amodiaquine (AQ) into its primary metabolite, N-desethylamodiaquine, is mediated by CYP2C8. Amodiaquine 36-47 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 120-126 17361129-1 2007 Metabolism of the antimalarial drug amodiaquine (AQ) into its primary metabolite, N-desethylamodiaquine, is mediated by CYP2C8. Amodiaquine 49-51 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 120-126 17361129-2 2007 We studied the frequency of CYP2C8 variants in 275 malaria-infected patients in Burkina Faso, the metabolism of AQ by CYP2C8 variants, and the impact of other drugs on AQ metabolism. Amodiaquine 112-114 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 118-124 17361129-5 2007 The variant most common in Africans, CYP2C8(*)2, showed defective metabolism of AQ (threefold higher K(m) and sixfold lower intrinsic clearance), and CYP2C8(*)3 had markedly decreased activity. Amodiaquine 80-82 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 37-43 17361129-6 2007 Considering drugs likely to be coadministered with AQ, the antiretroviral drugs efavirenz, saquinavir, lopinavir, and tipranavir were potent CYP2C8 inhibitors at clinically relevant concentrations. Amodiaquine 51-53 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 141-147 17361129-7 2007 Variable CYP2C8 activity owing to genetic variation and drug interactions may have important clinical implications for the efficacy and toxicity of AQ. Amodiaquine 148-150 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 9-15 17222819-5 2007 In conclusion, the present findings suggested that the elevation of endogenous histamine by amodiaquine may thus play a protective role through the regulation of TNF-alpha production in endotoxin-induced hepatic injury mice. Amodiaquine 92-103 tumor necrosis factor Mus musculus 162-171 17417075-7 2007 For example, the parameter values for HF alone and with AQ were: Cmax 144 +/- 53 versus 164 +/- 58 microg/L; T1/2beta 142 +/- 23 versus 139 +/- 28 hours; Cl/F 37.3 +/- 13.9 versus 32.3 +/- 11.4 L/h; and metabolic ratio 1.2 +/- 0.5 vs 1.1 +/- 0.6 Similarly, the disposition of HFM was not significantly altered (P > 0.05) after an earlier exposure to amodiaquine. Amodiaquine 56-58 interleukin 1 receptor like 1 Homo sapiens 109-121 17286541-5 2007 Amodiaquine is almost entirely metabolized by the polymorphic cytochrome P450 (CYP) isoform 2C8 to the pharmacologically active desethylamodiaquine. Amodiaquine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 17222819-0 2007 Effect of amodiaquine, a histamine N-methyltransferase inhibitor, on, Propionibacterium acnes and lipopolysaccharide-induced hepatitis in mice. Amodiaquine 10-21 histamine N-methyltransferase Mus musculus 25-54 17222819-1 2007 We examined whether treatment with amodiaquine, a potent inhibitor of histamine N-methyltransferase protects mice from Propionibacterium acnes (P. acnes)-primed and lipopolysaccharide (LPS)-induced hepatitis. Amodiaquine 35-46 histamine N-methyltransferase Mus musculus 70-99 17222819-3 2007 Pretreatment with amodiaquine also improved the survival rate of the hepatitis mice, and this improvement was partially associated with the decrease in serum levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). Amodiaquine 18-29 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 168-194 17222819-3 2007 Pretreatment with amodiaquine also improved the survival rate of the hepatitis mice, and this improvement was partially associated with the decrease in serum levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). Amodiaquine 18-29 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 196-199 17222819-3 2007 Pretreatment with amodiaquine also improved the survival rate of the hepatitis mice, and this improvement was partially associated with the decrease in serum levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). Amodiaquine 18-29 glutamic pyruvic transaminase, soluble Mus musculus 205-229 17222819-3 2007 Pretreatment with amodiaquine also improved the survival rate of the hepatitis mice, and this improvement was partially associated with the decrease in serum levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). Amodiaquine 18-29 glutamic pyruvic transaminase, soluble Mus musculus 231-234 17222819-4 2007 Amodiaquine partially suppressed increases of tumor necrosis factor (TNF)-alpha in the serum and TNF-alpha mRNA expression in the liver, whereas the expression of interleukin (IL)-18, interferon (IFN)-gamma and IL-12 in the liver was not changed by amodiaquine treatment. Amodiaquine 0-11 tumor necrosis factor Mus musculus 46-79 17222819-4 2007 Amodiaquine partially suppressed increases of tumor necrosis factor (TNF)-alpha in the serum and TNF-alpha mRNA expression in the liver, whereas the expression of interleukin (IL)-18, interferon (IFN)-gamma and IL-12 in the liver was not changed by amodiaquine treatment. Amodiaquine 0-11 tumor necrosis factor Mus musculus 97-106 17286541-5 2007 Amodiaquine is almost entirely metabolized by the polymorphic cytochrome P450 (CYP) isoform 2C8 to the pharmacologically active desethylamodiaquine. Amodiaquine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 16536934-1 2006 OBJECTIVES: To assess the relationship between the genetic and phenotypic factors linked to the cytochrome P-450 enzyme system and the response to the antimalarial drugs chloroquine, amodiaquine, mefloquine, and proguanil, as well as to determine how certain biological and social factors of the host influence the behavior of this enzymatic complex. Amodiaquine 183-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 16783563-0 2006 Amodiaquine, its desethylated metabolite, or both, inhibit the metabolism of debrisoquine (CYP2D6) and losartan (CYP2C9) in vivo. Amodiaquine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 91-97 16783563-0 2006 Amodiaquine, its desethylated metabolite, or both, inhibit the metabolism of debrisoquine (CYP2D6) and losartan (CYP2C9) in vivo. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 16783563-1 2006 OBJECTIVE: To study the extent of in vivo inhibition by the antimalarial drug amodiaquine, its active metabolite N-desethylamodiaquine, or both, of the metabolism of four probe drugs of the enzymes CYP2D6, CYP2C19, CYP2C9 and CYP1A2. Amodiaquine 78-89 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 198-204 16783563-1 2006 OBJECTIVE: To study the extent of in vivo inhibition by the antimalarial drug amodiaquine, its active metabolite N-desethylamodiaquine, or both, of the metabolism of four probe drugs of the enzymes CYP2D6, CYP2C19, CYP2C9 and CYP1A2. Amodiaquine 78-89 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 206-213 16783563-1 2006 OBJECTIVE: To study the extent of in vivo inhibition by the antimalarial drug amodiaquine, its active metabolite N-desethylamodiaquine, or both, of the metabolism of four probe drugs of the enzymes CYP2D6, CYP2C19, CYP2C9 and CYP1A2. Amodiaquine 78-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 215-221 16783563-1 2006 OBJECTIVE: To study the extent of in vivo inhibition by the antimalarial drug amodiaquine, its active metabolite N-desethylamodiaquine, or both, of the metabolism of four probe drugs of the enzymes CYP2D6, CYP2C19, CYP2C9 and CYP1A2. Amodiaquine 78-89 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 226-232 16783563-10 2006 The effects on CYP2D6 and CYP2C9 activities subsided within a week after intake of amodiaquine as tested by the phenotyping cocktail. Amodiaquine 83-94 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 15-21 16783563-10 2006 The effects on CYP2D6 and CYP2C9 activities subsided within a week after intake of amodiaquine as tested by the phenotyping cocktail. Amodiaquine 83-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 16783563-12 2006 CONCLUSION: A single dose of amodiaquine decreased CYP2D6 and CYP2C9 activities significantly compared to baseline values. Amodiaquine 29-40 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 51-57 16783563-12 2006 CONCLUSION: A single dose of amodiaquine decreased CYP2D6 and CYP2C9 activities significantly compared to baseline values. Amodiaquine 29-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 16168438-3 2005 The histamine receptor H1 antagonist diphenhydramine, the antimalarial drug amodiaquine, the antifolate drug metoprine, and the anticholinesterase drug tacrine (an early drug for Alzheimer"s disease) are surprisingly all potent HNMT inhibitors, having inhibition constants in the range of 10-100nM. Amodiaquine 76-87 histamine N-methyltransferase Homo sapiens 228-232 16359408-3 2005 The pro-drug AQ is transformed by cytochrome P450 CYP2C8 to the active metabolite N-desethylaminodiaquine. Amodiaquine 13-15 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 50-56 16359408-7 2005 AQ metabolism in patients with CYP2C8*2 may be impaired, and with an increase of AQ based treatment the risk of severe adverse events may mount. Amodiaquine 0-2 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 31-37 14681337-6 2004 Other classes of drugs that demonstrated inhibition of aldehyde oxidase included phenothiazines, tricyclic antidepressants, tricyclic atypical antipsychotic agents, and dihydropyridine calcium channel blockers, along with some other drugs, including loratadine, cyclobenzaprine, amodiaquine, maprotiline, ondansetron, propafenone, domperidone, quinacrine, ketoconazole, verapamil, tacrine, and salmeterol. Amodiaquine 279-290 aldehyde oxidase 1 Homo sapiens 55-71 15785959-1 2005 OBJECTIVE: The determination of the prevalence of the CYP2C8 main alleles in a typical set of malaria patients in Zanzibar, as these patients represent a typical population exposed to amodiaquine, an antimalarial mainly metabolized by CYP2C8. Amodiaquine 184-195 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 54-60 15601807-3 2005 A validated sensitive, moderate-throughput high-performance liquid chromatography/tandem mass spectrometry (HPLC/MS/MS) assay was used to detect N-desethylamodiaquine, the CYP2C8-derived major metabolite of amodiaquine metabolism, using heterologously expressed recombinant CYP2C8 (rhCYP2C8) and pooled human liver microsomes. Amodiaquine 155-166 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 172-178 15601807-3 2005 A validated sensitive, moderate-throughput high-performance liquid chromatography/tandem mass spectrometry (HPLC/MS/MS) assay was used to detect N-desethylamodiaquine, the CYP2C8-derived major metabolite of amodiaquine metabolism, using heterologously expressed recombinant CYP2C8 (rhCYP2C8) and pooled human liver microsomes. Amodiaquine 155-166 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 274-280 12082016-9 2002 Further exploration of antimalarial compounds identified the common medicinals chloroquine, quinacrine, and amodiaquine as Tp53-inducers. Amodiaquine 108-119 tumor protein p53 Homo sapiens 123-127 12903974-1 2003 The non-covalent interactions between the monomeric phenolic compound chlorogenic acid (5-CQA) and bovine serum albumin (BSA), lysozyme, and alpha-lactalbumin were characterized, and their effect on protein properties was examined. Amodiaquine 90-93 albumin Homo sapiens 106-119 12903974-1 2003 The non-covalent interactions between the monomeric phenolic compound chlorogenic acid (5-CQA) and bovine serum albumin (BSA), lysozyme, and alpha-lactalbumin were characterized, and their effect on protein properties was examined. Amodiaquine 90-93 lactalbumin alpha Homo sapiens 141-158 12397395-7 2002 Blockade of histamine N-methyltransferase by amodiaquin (100 microM) abolished catabolism completely, whereas blockade of diamine oxidase by aminoguanidine (100 microM) was less effective. Amodiaquine 45-55 histamine N-methyltransferase Sus scrofa 12-41 11124226-7 2001 Proguanil, cycloguanil, amodiaquine, and desethylamodiaquine inhibited CYP2D6 (K(i) = 6.76, 5.97, 2.1, and 4.13 microM, respectively). Amodiaquine 24-35 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 71-77 11805197-0 2002 Amodiaquine clearance and its metabolism to N-desethylamodiaquine is mediated by CYP2C8: a new high affinity and turnover enzyme-specific probe substrate. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 81-87 11805197-3 2002 CYP2C8 was the main hepatic isoform that cleared AQ and catalyzed the formation of DEAQ. The extrahepatic P450s, 1A1 and 1B1, also cleared AQ and catalyzed the formation of an unknown metabolite M2. Amodiaquine 49-51 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 0-6 11805197-3 2002 CYP2C8 was the main hepatic isoform that cleared AQ and catalyzed the formation of DEAQ. The extrahepatic P450s, 1A1 and 1B1, also cleared AQ and catalyzed the formation of an unknown metabolite M2. Amodiaquine 49-51 cytochrome P450 family 1 subfamily B member 1 Homo sapiens 106-124 11805197-3 2002 CYP2C8 was the main hepatic isoform that cleared AQ and catalyzed the formation of DEAQ. The extrahepatic P450s, 1A1 and 1B1, also cleared AQ and catalyzed the formation of an unknown metabolite M2. Amodiaquine 85-87 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 0-6 11805197-3 2002 CYP2C8 was the main hepatic isoform that cleared AQ and catalyzed the formation of DEAQ. The extrahepatic P450s, 1A1 and 1B1, also cleared AQ and catalyzed the formation of an unknown metabolite M2. Amodiaquine 85-87 cytochrome P450 family 1 subfamily B member 1 Homo sapiens 106-124 11805197-7 2002 Both the formation of DEAQ and the clearance of AQ showed excellent correlations (r(2) = 0.98 and 0.95) with 6alpha-hydroxylation of paclitaxel, a marker substrate for CYP2C8. Amodiaquine 24-26 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 168-174 11805197-9 2002 Docking of AQ into the active site homology models of the CYP2C isoforms showed favorable interactions with CYP2C8, which supported the likelihood of an N-desethylation reaction. Amodiaquine 11-13 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 108-114 11805197-10 2002 These data show that CYP2C8 is the main hepatic isoform responsible for the metabolism of AQ. Amodiaquine 90-92 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 21-27 11805197-11 2002 The specificity, high affinity, and high turnover make AQ desethylation an excellent marker reaction for CYP2C8 activity. Amodiaquine 55-57 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 105-111 10831384-1 2000 A multidrug-resistant strain of Plasmodium yoelii nigeriensis (MDR) showing a wide spectrum of resistance to chloroquine, amodiaquine, mepacrine, mefloquine, halofantrine, quinine, and quinidine was used in this study for in vivo evaluation of the blood schizontocidal activity of pyronaridine, a topoisomerase II inhibitor, in Swiss mice. Amodiaquine 122-133 malic enzyme complex, mitochondrial Mus musculus 63-66 11140821-2 2000 Treatment of rats with 10 mg/kg (single dose) amodiaquine resulted in 10% and 63% increases, respectively in the activities of liver superoxide dismutase and glutathione peroxidase (P<0.01), while the activity of liver catalase was significantly reduced by 26% compared to control. Amodiaquine 46-57 catalase Rattus norvegicus 222-230 11140821-4 2000 Following multiple dose (10 mg/kg for 4 consecutive days) amodiaquine treatment, activities of superoxide dismutase and glutathione peroxidase were increased by 30% and 133% respectively while catalase activity was decreased by 45%. Amodiaquine 58-69 catalase Rattus norvegicus 193-201 11140821-9 2000 The activities of serum aspartate amino transferase, alanine amino transferase, ornithine carbamyl transferase and gamma-glutamyl transferase were significantly increased by both single and multiple doses of amodiaquine treatment (P<0.01). Amodiaquine 208-219 gamma-glutamyltransferase 1 Rattus norvegicus 115-141 9736572-4 1998 The inclusion of a group at the C-5" position of amodiaquine reduced or eliminated bioactivation, as determined by glutathione conjugate formation in vivo. Amodiaquine 49-60 hemolytic complement Mus musculus 32-35 9736572-6 1998 Chemical substitution at the C-5" position also resulted in compounds which underwent slower elimination (<5% of the dose excreted into bile and urine, compared with 50% for amodiaquine) and increased levels of accumulation in tissue (10% of the dose in the liver at 48 h compared with 1% with amodiaquine). Amodiaquine 177-188 hemolytic complement Mus musculus 29-32 9736572-6 1998 Chemical substitution at the C-5" position also resulted in compounds which underwent slower elimination (<5% of the dose excreted into bile and urine, compared with 50% for amodiaquine) and increased levels of accumulation in tissue (10% of the dose in the liver at 48 h compared with 1% with amodiaquine). Amodiaquine 297-308 hemolytic complement Mus musculus 29-32 9736572-8 1998 The alteration in the disposition following the introduction of the C-5" substituent resulted in an increased duration of antimalarial activity in the mouse compared with that for amodiaquine. Amodiaquine 180-191 hemolytic complement Mus musculus 68-71 1346997-6 1992 Cyclosporine A was a potent competitive inhibitor of CQA metabolism, providing initial evidence that formation of metabolite I was catalyzed by proteins of the CYP3 gene family. Amodiaquine 53-56 peptidylprolyl isomerase F Homo sapiens 160-164 7714794-3 1995 Glutathione conjugates of AQ and desethylAQ were eliminated in bile after intraportal administration of [3H]AQ (54 mumol/kg, 20 microCi/kg) to anesthetized male CD1 mice. Amodiaquine 26-28 CD1 antigen complex Mus musculus 161-164 7816881-5 1994 The cholinesterase inhibitors tacrine (THA) and, to a lesser extent, physostigmine (PHYSO) and amodiaquine (AMDQ), caused an upward shift in the frequency of the resting electrocortical activity, although scopolamine significantly slowed the activity below baseline levels. Amodiaquine 95-106 butyrylcholinesterase Homo sapiens 4-18 7816881-5 1994 The cholinesterase inhibitors tacrine (THA) and, to a lesser extent, physostigmine (PHYSO) and amodiaquine (AMDQ), caused an upward shift in the frequency of the resting electrocortical activity, although scopolamine significantly slowed the activity below baseline levels. Amodiaquine 108-112 butyrylcholinesterase Homo sapiens 4-18 8145732-13 1994 IC50 values for the HNMT inhibitor amodiaquine were 0.50, 0.48, and 0.40 microM, respectively, for enzyme from these same three sources. Amodiaquine 35-46 histamine N-methyltransferase Homo sapiens 20-24 1346997-7 1992 This was substantiated by the finding that CQA metabolism was completely inhibited by a polyclonal antibody directed against a pregnenolone 16 alpha-carbonitrile-inducible cytochrome P-450 of rat liver. Amodiaquine 43-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 1346997-8 1992 The rate of CQA metabolism correlated significantly to the level of CYP3A4 expression, the rate of cyclosporine A metabolism to each of the primary metabolites (M-1, M-17, and M-21), and the rate of midazolam 4-hydroxylation. Amodiaquine 12-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-74 8329010-1 1993 The pharmacokinetics of amodiaquine (AQ, Flavoquine, CAS 6398-98-7) and its metabolites, mono (AQml) and bis-desethyl amodiaquine (AQm2) were investigated in 8 healthy volunteers after an oral dose of 306.2 mg of AQ. Amodiaquine 24-35 BCAR1 scaffold protein, Cas family member Homo sapiens 53-56 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). Amodiaquine 36-47 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). Amodiaquine 36-47 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 210-216 1907425-6 1991 IC50 values for the inhibition of mouse kidney HNMT by amodiaquine and S-adenosyl-L-homocysteine were 1.67 and 11.8 microM, respectively. Amodiaquine 55-66 histamine N-methyltransferase Mus musculus 47-51 34438008-0 2021 Development and Characterization of Inhalable Transferrin Functionalized Amodiaquine Nanoparticles - Efficacy in Non-small Cell Lung Cancer (NSCLC) Treatment. Amodiaquine 73-84 transferrin Homo sapiens 46-57 34592572-4 2021 We found that de-glycosylation at N331 and N343 drastically reduces the RBD binding to ACE2. Amodiaquine 34-38 angiotensin converting enzyme 2 Homo sapiens 87-91 34438008-4 2021 In this study, targeting potential of transferrin ligand conjugated inhalable AQ-loaded nanoparticles (Tf-AMQ NPs) was investigated against NSCLC. Amodiaquine 78-80 transferrin Homo sapiens 38-49 34438008-6 2021 Cytotoxicity studies in NSCLC cell line with overexpressed transferrin receptors shown significant reduction in IC50 values with Tf-decorated AQ-loaded nanoparticles compared to AQ or non-targeted NPs, along with significant apoptosis induction (caspase assay) and reduced % colony growth in A549 and H1299 cells with Tf-AMQ NP. Amodiaquine 142-144 transferrin Homo sapiens 59-70 34438008-6 2021 Cytotoxicity studies in NSCLC cell line with overexpressed transferrin receptors shown significant reduction in IC50 values with Tf-decorated AQ-loaded nanoparticles compared to AQ or non-targeted NPs, along with significant apoptosis induction (caspase assay) and reduced % colony growth in A549 and H1299 cells with Tf-AMQ NP. Amodiaquine 178-180 transferrin Homo sapiens 59-70 34438008-8 2021 AQ"s autophagy inhibition ability significantly increased with nanoparticle encapsulation and transferrin conjugation. Amodiaquine 0-2 transferrin Homo sapiens 94-105 34438008-9 2021 In conclusion, amodiaquine can be an assuring candidate for repurposing to consider for NSCLC treatment while delivering inhalable transferrin conjugated nanoparticles developed using a scalable HPH process to the target site, thus reducing the dose, side effects. Amodiaquine 15-26 transferrin Homo sapiens 131-142 35429395-7 2022 The mdr1-YY haplotype, associated with chloroquine and amodiaquine resistance, decreased over time, while the NY (wildtype) and the NF (modulates response to lumefantrine) haplotypes increased. Amodiaquine 55-66 ATP binding cassette subfamily B member 1 Homo sapiens 4-8 34808194-5 2021 We found that AQ effectively and significantly increased testosterone production in TM3 and primary Leydig cells through enhanced expression of StAR, CYP11A1, CYP17A1, and 3betaHSD. Amodiaquine 14-16 steroidogenic acute regulatory protein Mus musculus 144-148 34808194-5 2021 We found that AQ effectively and significantly increased testosterone production in TM3 and primary Leydig cells through enhanced expression of StAR, CYP11A1, CYP17A1, and 3betaHSD. Amodiaquine 14-16 cytochrome P450, family 11, subfamily a, polypeptide 1 Mus musculus 150-157 34808194-5 2021 We found that AQ effectively and significantly increased testosterone production in TM3 and primary Leydig cells through enhanced expression of StAR, CYP11A1, CYP17A1, and 3betaHSD. Amodiaquine 14-16 cytochrome P450, family 17, subfamily a, polypeptide 1 Mus musculus 159-166 34808194-6 2021 Concurrently, AQ dose-dependently increased the expression of 3-hydroxy-3-methylglutaryl-CoA reductase, a key enzyme in the cholesterol synthesis pathway, through induction of the transcriptional and DNA-binding activities of NR4A1, contributing to increased cholesterol synthesis in Leydig cells. Amodiaquine 14-16 nuclear receptor subfamily 4, group A, member 1 Mus musculus 226-231 34808194-7 2021 Furthermore, AQ increased the expression of fatty acid synthase and diacylglycerol acyltransferase and potentiated de novo synthesis of fatty acids and triglycerides (TG). Amodiaquine 13-15 fatty acid synthase Mus musculus 44-63 35132775-0 2022 Scaffold hopping from amodiaquine to novel Nurr1 agonist chemotypes via microscale analogue libraries. Amodiaquine 22-33 nuclear receptor subfamily 4 group A member 2 Homo sapiens 43-48 35132775-3 2022 To identify novel Nurr1 agonist chemotypes, we have employed the Nurr1 activator amodiaquine as template for microscale analogue library synthesis. Amodiaquine 81-92 nuclear receptor subfamily 4 group A member 2 Homo sapiens 18-23 35132775-3 2022 To identify novel Nurr1 agonist chemotypes, we have employed the Nurr1 activator amodiaquine as template for microscale analogue library synthesis. Amodiaquine 81-92 nuclear receptor subfamily 4 group A member 2 Homo sapiens 65-70 35474783-9 2022 Moreover, myricetin caused considerable elevation in the oral exposure of amodiaquine as a CYP2C8 substrate via a slowdown of amodiaquine clearance in the rat model. Amodiaquine 74-85 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 91-97 35474783-5 2022 In the in vitro study, myricetin showed a substantial effect on CYP2C8 inhibition in human liver microsomes using CYP2C8-catalyzed amodiaquine-N-deethylation as an index reaction. Amodiaquine 131-142 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 64-70 35474783-5 2022 In the in vitro study, myricetin showed a substantial effect on CYP2C8 inhibition in human liver microsomes using CYP2C8-catalyzed amodiaquine-N-deethylation as an index reaction. Amodiaquine 131-142 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 114-120 35474783-9 2022 Moreover, myricetin caused considerable elevation in the oral exposure of amodiaquine as a CYP2C8 substrate via a slowdown of amodiaquine clearance in the rat model. Amodiaquine 126-137 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 91-97 35110426-0 2022 Inhibition of stearoyl-CoA desaturase 1 potentiates anti-tumor activity of amodiaquine in non-small cell lung cancer. Amodiaquine 75-86 stearoyl-Coenzyme A desaturase 1 Mus musculus 14-39 35110426-4 2022 Here, we found that amodiaquine (AQ) increased autophagosome numbers and LC3BII and p62 at protein levels in A549 lung cancer cells suggesting the blockade of autophagic flux by AQ. Amodiaquine 20-31 nucleoporin 62 Mus musculus 84-87 35110426-4 2022 Here, we found that amodiaquine (AQ) increased autophagosome numbers and LC3BII and p62 at protein levels in A549 lung cancer cells suggesting the blockade of autophagic flux by AQ. Amodiaquine 33-35 nucleoporin 62 Mus musculus 84-87 35110426-4 2022 Here, we found that amodiaquine (AQ) increased autophagosome numbers and LC3BII and p62 at protein levels in A549 lung cancer cells suggesting the blockade of autophagic flux by AQ. Amodiaquine 178-180 nucleoporin 62 Mus musculus 84-87 35110426-5 2022 To identify the key metabolic vulnerability associated with autophagy inhibition by AQ treatment, we then performed transcriptomics analysis in the presence or absence of AQ in A549 lung cancer cells and found stearoyl-CoA desaturase 1 (SCD) 1 was one of the most highly upregulated with AQ exposure. Amodiaquine 84-86 stearoyl-Coenzyme A desaturase 1 Mus musculus 210-243 35110426-5 2022 To identify the key metabolic vulnerability associated with autophagy inhibition by AQ treatment, we then performed transcriptomics analysis in the presence or absence of AQ in A549 lung cancer cells and found stearoyl-CoA desaturase 1 (SCD) 1 was one of the most highly upregulated with AQ exposure. Amodiaquine 171-173 stearoyl-Coenzyme A desaturase 1 Mus musculus 210-243 35110426-5 2022 To identify the key metabolic vulnerability associated with autophagy inhibition by AQ treatment, we then performed transcriptomics analysis in the presence or absence of AQ in A549 lung cancer cells and found stearoyl-CoA desaturase 1 (SCD) 1 was one of the most highly upregulated with AQ exposure. Amodiaquine 288-290 stearoyl-Coenzyme A desaturase 1 Mus musculus 210-243 35110426-6 2022 The induction of SCD1 by AQ exposure at both protein and mRNA level suggests that SCD1 could represent a potential therapeutic target of AQ treatment. Amodiaquine 25-27 stearoyl-Coenzyme A desaturase 1 Mus musculus 17-21 35110426-6 2022 The induction of SCD1 by AQ exposure at both protein and mRNA level suggests that SCD1 could represent a potential therapeutic target of AQ treatment. Amodiaquine 25-27 stearoyl-Coenzyme A desaturase 1 Mus musculus 82-86 35110426-6 2022 The induction of SCD1 by AQ exposure at both protein and mRNA level suggests that SCD1 could represent a potential therapeutic target of AQ treatment. Amodiaquine 137-139 stearoyl-Coenzyme A desaturase 1 Mus musculus 17-21 35110426-6 2022 The induction of SCD1 by AQ exposure at both protein and mRNA level suggests that SCD1 could represent a potential therapeutic target of AQ treatment. Amodiaquine 137-139 stearoyl-Coenzyme A desaturase 1 Mus musculus 82-86 35110426-8 2022 Taken together, our study identified SCD1 could be a new therapeutic target upon autophagy inhibition by AQ exposure. Amodiaquine 105-107 stearoyl-Coenzyme A desaturase 1 Mus musculus 37-41 3337739-2 1988 Amodiaquine was clearly the most potent HMT inhibitor followed by quinacrine, chlorhexidine, alcuronium and chloroquine. Amodiaquine 0-11 histamine N-methyltransferase Rattus norvegicus 40-43 6380785-5 1984 The mefloquine-resistant Camp strain remained sensitive to chloroquine and amodiaquine, and became slightly more resistant to quinine; there was increased sensitivity to pyrimethamine. Amodiaquine 75-86 cathelicidin antimicrobial peptide Homo sapiens 25-29 4028443-7 1985 The Kii for inhibition of the RBC enzyme by amodiaquine, an HNMT inhibitor, was 1.0 X 10(-7) mol/l, while the Kis value was 0.48 X 10(-7) mol/l. Amodiaquine 44-55 histamine N-methyltransferase Homo sapiens 60-64 3832018-1 1985 Four compounds: amodiaquine, quinacrine, 1,4-(tele)-methylhistamine and metoprine, which in vitro effectively inhibit histamine N-methyltransferase (HMT) activity, were tested for their effects on histamine (HI) levels in the rat brain and ex vivo HMT activity. Amodiaquine 16-27 histamine N-methyltransferase Rattus norvegicus 118-147 3832018-1 1985 Four compounds: amodiaquine, quinacrine, 1,4-(tele)-methylhistamine and metoprine, which in vitro effectively inhibit histamine N-methyltransferase (HMT) activity, were tested for their effects on histamine (HI) levels in the rat brain and ex vivo HMT activity. Amodiaquine 16-27 histamine N-methyltransferase Rattus norvegicus 149-152 3832018-3 1985 Amodiaquine, quinacrine and 1,4-(tele)-methyl-HI weakly inhibited HMT activity ex vivo and they failed to alter HI levels in the rat brain. Amodiaquine 0-11 histamine N-methyltransferase Rattus norvegicus 66-69 33507465-9 2021 In the same vein, we found that amodiaquine substantially increased the level of phosphorylated P38 Mapk. Amodiaquine 32-43 mitogen-activated protein kinase 14 Mus musculus 96-104 7110523-1 1982 The effects of histamine and amodiaquine (a drug known to inhibit histamine-N-methyltransferase) on pulsatile growth hormone (GH) secretion were determined in unanaesthetized male rats. Amodiaquine 29-40 gonadotropin releasing hormone receptor Rattus norvegicus 110-124 7110523-1 1982 The effects of histamine and amodiaquine (a drug known to inhibit histamine-N-methyltransferase) on pulsatile growth hormone (GH) secretion were determined in unanaesthetized male rats. Amodiaquine 29-40 gonadotropin releasing hormone receptor Rattus norvegicus 126-128 7256355-1 1981 The antimalarial agent, amodiaquine, is a potent inhibitor of AChE (Ki = 1.50 x 10(-9) M, pH 7.4, 25 degrees C). Amodiaquine 24-35 acetylcholinesterase (Cartwright blood group) Homo sapiens 62-66 7256355-2 1981 Both the protonated diethylamino and phenolic hydroxyl functions of amodiaquine are necessary for interaction with AChE. Amodiaquine 68-79 acetylcholinesterase (Cartwright blood group) Homo sapiens 115-119 7256355-3 1981 This suggests that the inhibition of AChE by amodiaquine may involve binding of the protonated diethylamino and phenolic hydroxyl functions to the anionic and esteric sites of the enzyme respectively. Amodiaquine 45-56 acetylcholinesterase (Cartwright blood group) Homo sapiens 37-41 7256355-4 1981 The anti-AChE property of amodiaquine may be related in some way to the gastrointestinal and central nervous system disturbances frequently encountered when large doses of amodiaquine are used for the treatment of malaria. Amodiaquine 26-37 acetylcholinesterase (Cartwright blood group) Homo sapiens 9-13 7256355-4 1981 The anti-AChE property of amodiaquine may be related in some way to the gastrointestinal and central nervous system disturbances frequently encountered when large doses of amodiaquine are used for the treatment of malaria. Amodiaquine 172-183 acetylcholinesterase (Cartwright blood group) Homo sapiens 9-13 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. Amodiaquine 180-191 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 85-91 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. Amodiaquine 180-191 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. Amodiaquine 180-191 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. Amodiaquine 180-191 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 112-118 6789797-1 1980 Amodiaquine, 10(-5) M, totally inhibited the activity of histamine-methyltransferase (HMT) in the rat brain. Amodiaquine 0-11 histamine N-methyltransferase Rattus norvegicus 86-89 33507465-11 2021 These results collectively suggest that amodiaquine can augment tyrosine hydroxylase expression via phosphorylated P38 Mapk while negatively regulating the phosphorylated Akt in protein expression. Amodiaquine 40-51 tyrosine hydroxylase Mus musculus 64-84 33507465-11 2021 These results collectively suggest that amodiaquine can augment tyrosine hydroxylase expression via phosphorylated P38 Mapk while negatively regulating the phosphorylated Akt in protein expression. Amodiaquine 40-51 mitogen-activated protein kinase 14 Mus musculus 115-123 33507465-11 2021 These results collectively suggest that amodiaquine can augment tyrosine hydroxylase expression via phosphorylated P38 Mapk while negatively regulating the phosphorylated Akt in protein expression. Amodiaquine 40-51 thymoma viral proto-oncogene 1 Mus musculus 171-174 33629841-2 2021 We have systematically studied the structure-activity relationship of the 4-amino-7-chloroquinoline scaffold contained in Nurr1 activators amodiaquine and chloroquine and discovered fragment-like analogues that activated Nurr1 in several cellular settings. Amodiaquine 139-150 nuclear receptor subfamily 4 group A member 2 Homo sapiens 122-127 33629841-2 2021 We have systematically studied the structure-activity relationship of the 4-amino-7-chloroquinoline scaffold contained in Nurr1 activators amodiaquine and chloroquine and discovered fragment-like analogues that activated Nurr1 in several cellular settings. Amodiaquine 139-150 nuclear receptor subfamily 4 group A member 2 Homo sapiens 221-226 32924342-0 2020 Beneficial Effect of Chloroquine and Amodiaquine on Type 1 Diabetic Tubulopathy by Attenuating Mitochondrial Nox4 and Endoplasmic Reticulum Stress. Amodiaquine 37-48 NADPH oxidase 4 Mus musculus 109-113 33441761-6 2021 Further studies on cytochrome b5 (CYB5), an alternative NADH-dependent electron donor indicated particularly strong support of CYP2C8-dependent amodiaquine N-deethylation by CYB5 and this was confirmed by genetic CYB5 single- and POR/CYB5 double-knockout. Amodiaquine 144-155 cytochrome b5 type A Homo sapiens 19-32 33441761-6 2021 Further studies on cytochrome b5 (CYB5), an alternative NADH-dependent electron donor indicated particularly strong support of CYP2C8-dependent amodiaquine N-deethylation by CYB5 and this was confirmed by genetic CYB5 single- and POR/CYB5 double-knockout. Amodiaquine 144-155 cytochrome b5 type A Homo sapiens 34-38 33441761-6 2021 Further studies on cytochrome b5 (CYB5), an alternative NADH-dependent electron donor indicated particularly strong support of CYP2C8-dependent amodiaquine N-deethylation by CYB5 and this was confirmed by genetic CYB5 single- and POR/CYB5 double-knockout. Amodiaquine 144-155 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 127-133 33441761-6 2021 Further studies on cytochrome b5 (CYB5), an alternative NADH-dependent electron donor indicated particularly strong support of CYP2C8-dependent amodiaquine N-deethylation by CYB5 and this was confirmed by genetic CYB5 single- and POR/CYB5 double-knockout. Amodiaquine 144-155 cytochrome b5 type A Homo sapiens 174-178 33441761-6 2021 Further studies on cytochrome b5 (CYB5), an alternative NADH-dependent electron donor indicated particularly strong support of CYP2C8-dependent amodiaquine N-deethylation by CYB5 and this was confirmed by genetic CYB5 single- and POR/CYB5 double-knockout. Amodiaquine 144-155 cytochrome b5 type A Homo sapiens 174-178 33441761-6 2021 Further studies on cytochrome b5 (CYB5), an alternative NADH-dependent electron donor indicated particularly strong support of CYP2C8-dependent amodiaquine N-deethylation by CYB5 and this was confirmed by genetic CYB5 single- and POR/CYB5 double-knockout. Amodiaquine 144-155 cytochrome p450 oxidoreductase Homo sapiens 230-233 33441761-6 2021 Further studies on cytochrome b5 (CYB5), an alternative NADH-dependent electron donor indicated particularly strong support of CYP2C8-dependent amodiaquine N-deethylation by CYB5 and this was confirmed by genetic CYB5 single- and POR/CYB5 double-knockout. Amodiaquine 144-155 cytochrome b5 type A Homo sapiens 174-178 33289551-4 2020 Protein NMR structural footprinting data show that amodiaquine, chloroquine, and cytosporone B bind the Nurr1 LBD; ligands that do not bind include C-DIM12, celastrol, camptothecin, IP7e, isoalantolactone, ethyl 2-[2,3,4-trimethoxy-6-(1-octanoyl)phenyl]acetate (TMPA), and three high-throughput screening hit derivatives. Amodiaquine 51-62 nuclear receptor subfamily 4 group A member 2 Homo sapiens 104-109 33507465-0 2021 The orphan nuclear receptor Nurr1 agonist amodiaquine mediates neuroprotective effects in 6-OHDA Parkinson"s disease animal model by enhancing the phosphorylation of P38 mitogen-activated kinase but not PI3K/AKT signaling pathway. Amodiaquine 42-53 nuclear receptor subfamily 4, group A, member 2 Mus musculus 28-33 33507465-0 2021 The orphan nuclear receptor Nurr1 agonist amodiaquine mediates neuroprotective effects in 6-OHDA Parkinson"s disease animal model by enhancing the phosphorylation of P38 mitogen-activated kinase but not PI3K/AKT signaling pathway. Amodiaquine 42-53 thymoma viral proto-oncogene 1 Mus musculus 208-211 33507465-2 2021 In an attempt to corroborate the treatment-modifying disease that would replicate the effect of Nurr1, it has been found that amodiaquine and Nurr1 had the same chemical scaffolding, indicating a crucial structure-activity relationship. Amodiaquine 126-137 nuclear receptor subfamily 4, group A, member 2 Mus musculus 96-101 33507465-3 2021 Interestingly, amodiaquine stimulate the transcriptional function of Nurr1 by physical interaction with its ligand-binding domain (LBD). Amodiaquine 15-26 nuclear receptor subfamily 4, group A, member 2 Mus musculus 69-74 33507465-4 2021 However, the signaling route by which Nurr1 is activated by amodiaquine to cause the protective effect remains to be elucidated. Amodiaquine 60-71 nuclear receptor subfamily 4, group A, member 2 Mus musculus 38-43 33507465-5 2021 We first demonstrated that amodiaquine treatment ameliorated behavioural deficits in 6-OHDA Parkinson"s disease mouse model, and it promoted dopaminergic neurons protection signified by Tyrosine hydroxylase (TH) and dopamine transporter (DAT) mRNA; Tyrosine hydroxylase (TH) protein expression level and the immunoreactivity in the substantia nigra compacta. Amodiaquine 27-38 tyrosine hydroxylase Mus musculus 186-206 33507465-5 2021 We first demonstrated that amodiaquine treatment ameliorated behavioural deficits in 6-OHDA Parkinson"s disease mouse model, and it promoted dopaminergic neurons protection signified by Tyrosine hydroxylase (TH) and dopamine transporter (DAT) mRNA; Tyrosine hydroxylase (TH) protein expression level and the immunoreactivity in the substantia nigra compacta. Amodiaquine 27-38 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 216-236 33507465-5 2021 We first demonstrated that amodiaquine treatment ameliorated behavioural deficits in 6-OHDA Parkinson"s disease mouse model, and it promoted dopaminergic neurons protection signified by Tyrosine hydroxylase (TH) and dopamine transporter (DAT) mRNA; Tyrosine hydroxylase (TH) protein expression level and the immunoreactivity in the substantia nigra compacta. Amodiaquine 27-38 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 238-241 33507465-5 2021 We first demonstrated that amodiaquine treatment ameliorated behavioural deficits in 6-OHDA Parkinson"s disease mouse model, and it promoted dopaminergic neurons protection signified by Tyrosine hydroxylase (TH) and dopamine transporter (DAT) mRNA; Tyrosine hydroxylase (TH) protein expression level and the immunoreactivity in the substantia nigra compacta. Amodiaquine 27-38 tyrosine hydroxylase Mus musculus 249-269 33507465-6 2021 Subsequently, we used inhibitors to ascertain the effect of amodiaquine on Akt and P38 Mapk as crucial signaling pathways for neuroprotection. Amodiaquine 60-71 thymoma viral proto-oncogene 1 Mus musculus 75-78 33507465-6 2021 Subsequently, we used inhibitors to ascertain the effect of amodiaquine on Akt and P38 Mapk as crucial signaling pathways for neuroprotection. Amodiaquine 60-71 mitogen-activated protein kinase 14 Mus musculus 83-91 33507465-8 2021 Western blot analysis confirmed that the phosphorylated Akt decreased significantly in the amodiaquine group compared to the control group. Amodiaquine 91-102 thymoma viral proto-oncogene 1 Mus musculus 56-59 33570383-8 2021 A subset of these potent RXR agonists can synergize with a presumed Nurr1 ligand and antimalarial drug (amodiaquine) to further enhance Nurr1/NBREs-directed transcription. Amodiaquine 104-115 retinoid X receptor alpha Homo sapiens 25-28 33570383-8 2021 A subset of these potent RXR agonists can synergize with a presumed Nurr1 ligand and antimalarial drug (amodiaquine) to further enhance Nurr1/NBREs-directed transcription. Amodiaquine 104-115 nuclear receptor subfamily 4 group A member 2 Homo sapiens 136-141 33475021-5 2021 In our study, the antimalarial compounds have been screened and docked against SARS-CoV-2-RdRp (PDB ID: 7BTF), and it was observed that the antimalarials chloroquine, hydroxychloroquine, and amodiaquine exhibit good affinity. Amodiaquine 191-202 ORF1a polyprotein;ORF1ab polyprotein Severe acute respiratory syndrome coronavirus 2 90-94 33209048-2 2020 Amodiaquine, which is mainly bio-transformed by CYP2C8, is known to be associated with adverse events of different severity. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 48-54 32924342-3 2020 We designed this study to explore the therapeutic potential of chloroquine (CQ) and amodiaquine (AQ) for inhibiting mitochondrial Nox4 and diabetic tubular injury. Amodiaquine 84-95 NADPH oxidase 4 Mus musculus 130-134 32924342-3 2020 We designed this study to explore the therapeutic potential of chloroquine (CQ) and amodiaquine (AQ) for inhibiting mitochondrial Nox4 and diabetic tubular injury. Amodiaquine 97-99 NADPH oxidase 4 Mus musculus 130-134 32924342-7 2020 RESULTS: CQ and AQ inhibited mitochondrial Nox4 and increased mitochondrial mass in hRPTCs under high-glucose conditions. Amodiaquine 16-18 NADPH oxidase 4 Mus musculus 43-47 32924342-9 2020 Notably, CQ and AQ treatment diminished Nox4 activation and ER stress in the kidneys of STZ-induced diabetic mice. Amodiaquine 16-18 NADPH oxidase 4 Mus musculus 40-44 32924342-11 2020 CONCLUSION: We substantiated the protective actions of CQ and AQ in diabetic tubulopathy associated with reduced mitochondrial Nox4 activation and ER stress alleviation. Amodiaquine 62-64 NADPH oxidase 4 Mus musculus 127-131 32291412-10 2020 Finally, through virtual screening, we identified amodiaquine as a potential inhibitor targeting the Fyn/CD147 axis. Amodiaquine 50-61 FYN proto-oncogene, Src family tyrosine kinase Homo sapiens 101-104 32600728-11 2020 AQ"s autophagy inhibition ability significantly increased (increased LC3B-II levels) with nanoparticle encapsulation, along with moderate improvement in apoptosis induction (Caspase-3 levels). Amodiaquine 0-2 caspase 3 Homo sapiens 174-183 32291412-10 2020 Finally, through virtual screening, we identified amodiaquine as a potential inhibitor targeting the Fyn/CD147 axis. Amodiaquine 50-61 basigin (Ok blood group) Homo sapiens 105-110 32291412-11 2020 Amodiaquine treatment dramatically inhibited the phosphorylation of CD147 by Fyn, thus attenuating melanoma cell growth and invasion in vitro and in vivo, suggesting that amodiaquine is a promising inhibitor for melanoma treatment. Amodiaquine 0-11 basigin (Ok blood group) Homo sapiens 68-73 32291412-11 2020 Amodiaquine treatment dramatically inhibited the phosphorylation of CD147 by Fyn, thus attenuating melanoma cell growth and invasion in vitro and in vivo, suggesting that amodiaquine is a promising inhibitor for melanoma treatment. Amodiaquine 0-11 FYN proto-oncogene, Src family tyrosine kinase Homo sapiens 77-80 32291412-11 2020 Amodiaquine treatment dramatically inhibited the phosphorylation of CD147 by Fyn, thus attenuating melanoma cell growth and invasion in vitro and in vivo, suggesting that amodiaquine is a promising inhibitor for melanoma treatment. Amodiaquine 171-182 basigin (Ok blood group) Homo sapiens 68-73 32291412-11 2020 Amodiaquine treatment dramatically inhibited the phosphorylation of CD147 by Fyn, thus attenuating melanoma cell growth and invasion in vitro and in vivo, suggesting that amodiaquine is a promising inhibitor for melanoma treatment. Amodiaquine 171-182 FYN proto-oncogene, Src family tyrosine kinase Homo sapiens 77-80 31642009-0 2020 Influence of MAMA decoction, an Herbal Antimalarial, on the Pharmacokinetics of Amodiaquine in Mice. Amodiaquine 80-91 lectin, galactoside-binding, soluble, 3 binding protein Mus musculus 13-17 32238479-3 2020 In a second cohort, rats received the Nurr1 agonist amodiaquine (AQ) together with L-DOPA or ropinirole. Amodiaquine 52-63 nuclear receptor subfamily 4, group A, member 2 Rattus norvegicus 38-43 32238479-3 2020 In a second cohort, rats received the Nurr1 agonist amodiaquine (AQ) together with L-DOPA or ropinirole. Amodiaquine 65-67 nuclear receptor subfamily 4, group A, member 2 Rattus norvegicus 38-43 32238479-7 2020 Nurr1 agonism with AQ exacerbated LID in F344 rats. Amodiaquine 19-21 nuclear receptor subfamily 4, group A, member 2 Rattus norvegicus 0-5 31285544-0 2020 The antimalarial drug amodiaquine stabilizes p53 through ribosome biogenesis stress, independently of its autophagy-inhibitory activity. Amodiaquine 22-33 tumor protein p53 Homo sapiens 45-48 31285544-3 2020 Amodiaquine triggers degradation of the catalytic subunit of RNA polymerase I (Pol I), with ensuing RPL5/RPL11-dependent stabilization of p53. Amodiaquine 0-11 DNA polymerase iota Homo sapiens 79-84 31285544-3 2020 Amodiaquine triggers degradation of the catalytic subunit of RNA polymerase I (Pol I), with ensuing RPL5/RPL11-dependent stabilization of p53. Amodiaquine 0-11 ribosomal protein L5 Homo sapiens 100-104 31285544-3 2020 Amodiaquine triggers degradation of the catalytic subunit of RNA polymerase I (Pol I), with ensuing RPL5/RPL11-dependent stabilization of p53. Amodiaquine 0-11 ribosomal protein L11 Homo sapiens 105-110 31285544-3 2020 Amodiaquine triggers degradation of the catalytic subunit of RNA polymerase I (Pol I), with ensuing RPL5/RPL11-dependent stabilization of p53. Amodiaquine 0-11 tumor protein p53 Homo sapiens 138-141 31285544-5 2020 RNAseq analysis revealed mechanistic similarities of amodiaquine with BMH-21, the first-in-class Pol I inhibitor, and with chloroquine, the antimalarial analog of amodiaquine, with well-established autophagy-inhibitory activity. Amodiaquine 53-64 DNA polymerase iota Homo sapiens 97-102 31629065-6 2020 Metabolic studies using microsomes obtained from HepG2 cell lines overexpressing human CYPs demonstrated that CYP1A1, 2C8, and 3A4 were the major enzymes that metabolized ADQ to NADQ and that CYP1A2, 1B1, 2C19, and 3A5 also metabolized ADQ, but to a lesser extent. Amodiaquine 171-174 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 110-116 31629065-6 2020 Metabolic studies using microsomes obtained from HepG2 cell lines overexpressing human CYPs demonstrated that CYP1A1, 2C8, and 3A4 were the major enzymes that metabolized ADQ to NADQ and that CYP1A2, 1B1, 2C19, and 3A5 also metabolized ADQ, but to a lesser extent. Amodiaquine 171-174 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 192-198 31629065-6 2020 Metabolic studies using microsomes obtained from HepG2 cell lines overexpressing human CYPs demonstrated that CYP1A1, 2C8, and 3A4 were the major enzymes that metabolized ADQ to NADQ and that CYP1A2, 1B1, 2C19, and 3A5 also metabolized ADQ, but to a lesser extent. Amodiaquine 179-182 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 110-116 31629065-7 2020 The cytotoxicity of ADQ was increased in CYP2C8 and 3A4 overexpressing HepG2 cells compared to HepG2/CYP vector cells, confirming that NADQ was more toxic than ADQ. Amodiaquine 20-23 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 41-55 31629065-7 2020 The cytotoxicity of ADQ was increased in CYP2C8 and 3A4 overexpressing HepG2 cells compared to HepG2/CYP vector cells, confirming that NADQ was more toxic than ADQ. Amodiaquine 20-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 31629065-7 2020 The cytotoxicity of ADQ was increased in CYP2C8 and 3A4 overexpressing HepG2 cells compared to HepG2/CYP vector cells, confirming that NADQ was more toxic than ADQ. Amodiaquine 136-139 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 41-55 31629065-8 2020 Moreover, treatment of CYP2C8 and 3A4 overexpressing HepG2 cells with ADQ increased the phosphorylation of JNK, ERK1/2, and p38, but not the expression of Bcl-2 family proteins. Amodiaquine 70-73 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 23-37 31629065-8 2020 Moreover, treatment of CYP2C8 and 3A4 overexpressing HepG2 cells with ADQ increased the phosphorylation of JNK, ERK1/2, and p38, but not the expression of Bcl-2 family proteins. Amodiaquine 70-73 mitogen-activated protein kinase 8 Homo sapiens 107-110 31629065-8 2020 Moreover, treatment of CYP2C8 and 3A4 overexpressing HepG2 cells with ADQ increased the phosphorylation of JNK, ERK1/2, and p38, but not the expression of Bcl-2 family proteins. Amodiaquine 70-73 mitogen-activated protein kinase 3 Homo sapiens 112-118 31629065-8 2020 Moreover, treatment of CYP2C8 and 3A4 overexpressing HepG2 cells with ADQ increased the phosphorylation of JNK, ERK1/2, and p38, but not the expression of Bcl-2 family proteins. Amodiaquine 70-73 mitogen-activated protein kinase 1 Homo sapiens 124-127 30709838-4 2019 Inactivation of CYP2C8 by amiodarone (100 muM), clopidogrel acyl-beta-d-glucuronide (100 muM), gemfibrozil 1-O-beta-glucuronide (100 muM), and phenelzine (100 muM) was investigated in HLMs and three recombinant human CYP2C8 preparations (Supersomes, Bactosomes, and EasyCYP Bactosomes) using amodiaquine N-deethylation as the marker reaction. Amodiaquine 292-303 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 16-22 31861329-6 2019 In addition, AQ treatment increased the expression of cell proliferation markers MCM5 and PCNA, and transcription factor E2F1. Amodiaquine 13-15 E2F transcription factor 1 Mus musculus 121-125 31861329-7 2019 Furthermore, pharmacological stimulation of Nurr1 by AQ increased the expression levels of positive cell cycle regulators such as cyclin A and cyclin-dependent kinases (CDK) 2. Amodiaquine 53-55 nuclear receptor subfamily 4, group A, member 2 Mus musculus 44-49 31861329-7 2019 Furthermore, pharmacological stimulation of Nurr1 by AQ increased the expression levels of positive cell cycle regulators such as cyclin A and cyclin-dependent kinases (CDK) 2. Amodiaquine 53-55 cyclin A2 Mus musculus 130-138 31861329-7 2019 Furthermore, pharmacological stimulation of Nurr1 by AQ increased the expression levels of positive cell cycle regulators such as cyclin A and cyclin-dependent kinases (CDK) 2. Amodiaquine 53-55 cyclin-dependent kinase 2 Mus musculus 143-175 31861329-8 2019 In contrast, levels of CDK inhibitors p27KIP1 and p57KIP2 were reduced upon treatment with AQ. Amodiaquine 91-93 cyclin-dependent kinase 2 Mus musculus 23-26 31861329-8 2019 In contrast, levels of CDK inhibitors p27KIP1 and p57KIP2 were reduced upon treatment with AQ. Amodiaquine 91-93 cyclin-dependent kinase inhibitor 1B Mus musculus 38-45 31861329-8 2019 In contrast, levels of CDK inhibitors p27KIP1 and p57KIP2 were reduced upon treatment with AQ. Amodiaquine 91-93 cyclin-dependent kinase inhibitor 1C (P57) Mus musculus 50-57 31861329-9 2019 Similar to the in vitro results, RT-qPCR analysis of AQ-administered mice brains revealed an increase in the levels of markers of cell cycle progression, PCNA, MCM5, and Cdc25a. Amodiaquine 53-55 proliferating cell nuclear antigen Mus musculus 154-158 31861329-9 2019 Similar to the in vitro results, RT-qPCR analysis of AQ-administered mice brains revealed an increase in the levels of markers of cell cycle progression, PCNA, MCM5, and Cdc25a. Amodiaquine 53-55 minichromosome maintenance complex component 5 Mus musculus 160-164 31861329-9 2019 Similar to the in vitro results, RT-qPCR analysis of AQ-administered mice brains revealed an increase in the levels of markers of cell cycle progression, PCNA, MCM5, and Cdc25a. Amodiaquine 53-55 cell division cycle 25A Mus musculus 170-176 31861329-10 2019 Finally, AQ administration resulted in decreased p27KIP1 and increased CDK2 levels in the dentate gyrus of the mouse hippocampus, as quantified immunohistochemically. Amodiaquine 9-11 cyclin-dependent kinase inhibitor 1B Mus musculus 49-56 31861329-10 2019 Finally, AQ administration resulted in decreased p27KIP1 and increased CDK2 levels in the dentate gyrus of the mouse hippocampus, as quantified immunohistochemically. Amodiaquine 9-11 cyclin-dependent kinase 2 Mus musculus 71-75 31861329-11 2019 Our results demonstrate that the pharmacological stimulation of Nurr1 in adult hNSCs by AQ promotes the cell cycle by modulating cell cycle-related molecules. Amodiaquine 88-90 nuclear receptor subfamily 4, group A, member 2 Mus musculus 64-69 31102570-5 2019 In contrast, CQ and AQ were significantly less potent than the bis-indole derivatives and, for some of the NR4A2-regulated genes, CQ and AQ were inactive as inducers. Amodiaquine 137-139 nuclear receptor subfamily 4 group A member 2 Homo sapiens 107-112 30825859-0 2019 A Nurr1 agonist amodiaquine attenuates inflammatory events and neurological deficits in a mouse model of intracerebral hemorrhage. Amodiaquine 16-27 nuclear receptor subfamily 4, group A, member 2 Mus musculus 2-7 30825859-3 2019 Daily administration of a Nurr1 agonist amodiaquine (40 mg/kg, i.p.) Amodiaquine 40-51 nuclear receptor subfamily 4, group A, member 2 Mus musculus 26-31 30825859-5 2019 Amodiaquine also suppressed ICH-induced mRNA expression of IL-1beta, CCL2 and CXCL2, and ameliorated motor dysfunction of mice. Amodiaquine 0-11 interleukin 1 beta Mus musculus 59-67 30825859-5 2019 Amodiaquine also suppressed ICH-induced mRNA expression of IL-1beta, CCL2 and CXCL2, and ameliorated motor dysfunction of mice. Amodiaquine 0-11 chemokine (C-C motif) ligand 2 Mus musculus 69-73 30825859-5 2019 Amodiaquine also suppressed ICH-induced mRNA expression of IL-1beta, CCL2 and CXCL2, and ameliorated motor dysfunction of mice. Amodiaquine 0-11 chemokine (C-X-C motif) ligand 2 Mus musculus 78-83 31861329-2 2019 We previously demonstrated that the pharmacological stimulation of Nurr1 by amodiaquine (AQ) promotes spatial memory by enhancing adult hippocampal neurogenesis. Amodiaquine 76-87 nuclear receptor subfamily 4, group A, member 2 Mus musculus 67-72 31861329-2 2019 We previously demonstrated that the pharmacological stimulation of Nurr1 by amodiaquine (AQ) promotes spatial memory by enhancing adult hippocampal neurogenesis. Amodiaquine 89-91 nuclear receptor subfamily 4, group A, member 2 Mus musculus 67-72 31861329-6 2019 In addition, AQ treatment increased the expression of cell proliferation markers MCM5 and PCNA, and transcription factor E2F1. Amodiaquine 13-15 minichromosome maintenance complex component 5 Mus musculus 81-85 31861329-6 2019 In addition, AQ treatment increased the expression of cell proliferation markers MCM5 and PCNA, and transcription factor E2F1. Amodiaquine 13-15 proliferating cell nuclear antigen Mus musculus 90-94 30663942-0 2019 Involvement of CCL2/CCR2 macrophage recruitment in amodiaquine-induced liver injury. Amodiaquine 51-62 chemokine (C-C motif) ligand 2 Mus musculus 15-19 30663942-0 2019 Involvement of CCL2/CCR2 macrophage recruitment in amodiaquine-induced liver injury. Amodiaquine 51-62 chemokine (C-C motif) receptor 2 Mus musculus 20-24 30663942-5 2019 Specifically, amodiaquine (AQ), which is known to cause IDILI in humans, causes mild liver injury in wild-type C57BL/6 mice that resolves despite continued AQ treatment, but it causes more severe liver injury that does not resolve in PD-1-/- mice co-treated with anti-CTLA-4 to impair immune tolerance. Amodiaquine 14-25 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 268-274 30663942-5 2019 Specifically, amodiaquine (AQ), which is known to cause IDILI in humans, causes mild liver injury in wild-type C57BL/6 mice that resolves despite continued AQ treatment, but it causes more severe liver injury that does not resolve in PD-1-/- mice co-treated with anti-CTLA-4 to impair immune tolerance. Amodiaquine 27-29 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 268-274 30663942-6 2019 CCR2-/- mice treated with AQ were not protected from the expected AQ-induced liver injury seen in wild-type C57BL/6 mice. Amodiaquine 26-28 chemokine (C-C motif) receptor 2 Mus musculus 0-4 30663942-7 2019 In contrast, anti-CCL2 antibodies attenuated the liver injury caused by AQ in the impaired immune tolerance model. Amodiaquine 72-74 chemokine (C-C motif) ligand 2 Mus musculus 18-22 31078747-3 2019 Individuals carrying the CYP2C8*2 c.805A > T (CYP2C8*2; rs11572103) allele have impaired amodiaquine metabolism, increased risk of amodiaquine-related adverse events, and may promote the selection of drug-resistant parasite strains. Amodiaquine 92-103 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 25-31 31078747-3 2019 Individuals carrying the CYP2C8*2 c.805A > T (CYP2C8*2; rs11572103) allele have impaired amodiaquine metabolism, increased risk of amodiaquine-related adverse events, and may promote the selection of drug-resistant parasite strains. Amodiaquine 92-103 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 49-55 31078747-3 2019 Individuals carrying the CYP2C8*2 c.805A > T (CYP2C8*2; rs11572103) allele have impaired amodiaquine metabolism, increased risk of amodiaquine-related adverse events, and may promote the selection of drug-resistant parasite strains. Amodiaquine 134-145 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 25-31 31078747-3 2019 Individuals carrying the CYP2C8*2 c.805A > T (CYP2C8*2; rs11572103) allele have impaired amodiaquine metabolism, increased risk of amodiaquine-related adverse events, and may promote the selection of drug-resistant parasite strains. Amodiaquine 134-145 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 49-55 31078747-10 2019 CONCLUSIONS: This study gives the first description of CYP2C8*2 allele distribution in the Republic of Congo and highlights the potential risk of amodiaquine-related adverse events. Amodiaquine 146-157 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 55-61 30208056-8 2018 The prototypical 4-aminoquinoline, amodiaquine was found to be a selective, non-competitive APJ antagonist that inhibited apelin signaling in a concentration-dependent manner. Amodiaquine 35-46 apelin receptor Homo sapiens 92-95 30976185-3 2019 The effect of pterostilbene on cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzyme activities were studied using the enzyme-selective substrates amodiaquine (CYP2C8), midazolam (CYP3A4), estradiol (UGT1A1), serotonin (UGT1A6) and mycophenolic acid (UGT1A8/9/10). Amodiaquine 160-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 30976185-3 2019 The effect of pterostilbene on cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzyme activities were studied using the enzyme-selective substrates amodiaquine (CYP2C8), midazolam (CYP3A4), estradiol (UGT1A1), serotonin (UGT1A6) and mycophenolic acid (UGT1A8/9/10). Amodiaquine 160-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 30976185-3 2019 The effect of pterostilbene on cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzyme activities were studied using the enzyme-selective substrates amodiaquine (CYP2C8), midazolam (CYP3A4), estradiol (UGT1A1), serotonin (UGT1A6) and mycophenolic acid (UGT1A8/9/10). Amodiaquine 160-171 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 57-84 30976185-3 2019 The effect of pterostilbene on cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzyme activities were studied using the enzyme-selective substrates amodiaquine (CYP2C8), midazolam (CYP3A4), estradiol (UGT1A1), serotonin (UGT1A6) and mycophenolic acid (UGT1A8/9/10). Amodiaquine 160-171 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 86-89 30976185-3 2019 The effect of pterostilbene on cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzyme activities were studied using the enzyme-selective substrates amodiaquine (CYP2C8), midazolam (CYP3A4), estradiol (UGT1A1), serotonin (UGT1A6) and mycophenolic acid (UGT1A8/9/10). Amodiaquine 160-171 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 173-179 30515963-8 2019 Importantly, 5XFAD mice treated with amodiaquine, a highly selective synthetic Nurr1 agonist, showed robust reduction in typical AD features including deposition of Abeta plaques, neuronal loss, microgliosis, and impairment of adult hippocampal neurogenesis, leading to significant improvement of cognitive impairment. Amodiaquine 37-48 nuclear receptor subfamily 4, group A, member 2 Mus musculus 79-84 30208056-8 2018 The prototypical 4-aminoquinoline, amodiaquine was found to be a selective, non-competitive APJ antagonist that inhibited apelin signaling in a concentration-dependent manner. Amodiaquine 35-46 apelin Homo sapiens 122-128 30208056-9 2018 Additionally, amodiaquine suppressed both apelin-and VGEF-induced endothelial tube formation. Amodiaquine 14-25 apelin Homo sapiens 42-48 29943426-6 2018 This P450 was identified by screening of actinobacterial strains for amodiaquine and ritonavir metabolizing activities, followed by genome sequencing and expression of the annotated S. platensis P450s in Escherichia coli. Amodiaquine 69-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 30206341-3 2018 By examining gene-edited parasites, we report that the ability of the wide-spread Dd2 PfCRT isoform to mediate chloroquine and amodiaquine resistance is substantially reduced by the addition of the PfCRT L272F mutation, which arose under blasticidin selection. Amodiaquine 127-138 aldo-keto reductase family 1 member C2 Homo sapiens 82-85 29516607-7 2018 CONCLUSION: Our findings suggest that amodiaquine exerts beneficial effects on glucose and lipid metabolism by concurrent activation of PPARalpha/gamma. Amodiaquine 38-49 peroxisome proliferator activated receptor alpha Mus musculus 136-145 29938495-2 2018 We have previously shown that modulating the immune system by impairing programmed cell death protein (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling, both receptors involved in immune tolerance, was capable of producing an animal model of amodiaquine (AQ) IDILI with characteristics very similar to IDILI in humans. Amodiaquine 266-277 spermatogenesis associated 2 Homo sapiens 103-107 29938495-2 2018 We have previously shown that modulating the immune system by impairing programmed cell death protein (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling, both receptors involved in immune tolerance, was capable of producing an animal model of amodiaquine (AQ) IDILI with characteristics very similar to IDILI in humans. Amodiaquine 266-277 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 113-156 29938495-2 2018 We have previously shown that modulating the immune system by impairing programmed cell death protein (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling, both receptors involved in immune tolerance, was capable of producing an animal model of amodiaquine (AQ) IDILI with characteristics very similar to IDILI in humans. Amodiaquine 266-277 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 158-164 29938495-2 2018 We have previously shown that modulating the immune system by impairing programmed cell death protein (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling, both receptors involved in immune tolerance, was capable of producing an animal model of amodiaquine (AQ) IDILI with characteristics very similar to IDILI in humans. Amodiaquine 279-281 spermatogenesis associated 2 Homo sapiens 103-107 29938495-2 2018 We have previously shown that modulating the immune system by impairing programmed cell death protein (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling, both receptors involved in immune tolerance, was capable of producing an animal model of amodiaquine (AQ) IDILI with characteristics very similar to IDILI in humans. Amodiaquine 279-281 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 113-156 29938495-2 2018 We have previously shown that modulating the immune system by impairing programmed cell death protein (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling, both receptors involved in immune tolerance, was capable of producing an animal model of amodiaquine (AQ) IDILI with characteristics very similar to IDILI in humans. Amodiaquine 279-281 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 158-164 29938495-6 2018 Addition of anti-LAG3 or anti-TGF-beta antibodies produced a small increase ALT in AQ-treated wild-type mice. Amodiaquine 83-85 lymphocyte-activation gene 3 Mus musculus 17-21 29938495-6 2018 Addition of anti-LAG3 or anti-TGF-beta antibodies produced a small increase ALT in AQ-treated wild-type mice. Amodiaquine 83-85 transforming growth factor, beta 1 Mus musculus 30-38 29938495-6 2018 Addition of anti-LAG3 or anti-TGF-beta antibodies produced a small increase ALT in AQ-treated wild-type mice. Amodiaquine 83-85 glutamic pyruvic transaminase, soluble Mus musculus 76-79 29938495-7 2018 There was a significant increase in ALT in PD-1-/- mice co-treated with anti-LAG3 or anti-TGF-beta relative to AQ-treated wild-type mice. Amodiaquine 111-113 glutamic pyruvic transaminase, soluble Mus musculus 36-39 29938495-7 2018 There was a significant increase in ALT in PD-1-/- mice co-treated with anti-LAG3 or anti-TGF-beta relative to AQ-treated wild-type mice. Amodiaquine 111-113 programmed cell death 1 Mus musculus 43-47 29938495-7 2018 There was a significant increase in ALT in PD-1-/- mice co-treated with anti-LAG3 or anti-TGF-beta relative to AQ-treated wild-type mice. Amodiaquine 111-113 lymphocyte-activation gene 3 Mus musculus 77-81 29938495-7 2018 There was a significant increase in ALT in PD-1-/- mice co-treated with anti-LAG3 or anti-TGF-beta relative to AQ-treated wild-type mice. Amodiaquine 111-113 transforming growth factor, beta 1 Mus musculus 90-98 29663414-1 2018 Amodiaquine (AQ) and pioglitazone (PGZ) are both metabolized by CYP2C8, an enzyme also inhibited by PGZ. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 64-70 29663414-1 2018 Amodiaquine (AQ) and pioglitazone (PGZ) are both metabolized by CYP2C8, an enzyme also inhibited by PGZ. Amodiaquine 13-15 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 64-70 29516607-2 2018 We aimed to demonstrate that amodiaquine, an antimalarial agent, has potential as a PPARalpha/gamma dual agonist with low risk of adverse effects. Amodiaquine 29-40 peroxisome proliferator activated receptor alpha Mus musculus 84-93 29516607-3 2018 METHODS: We screened a Prestwick library (Prestwick Chemical; Illkirch, France) to identify novel PPARalpha/gamma dual agonists and selected amodiaquine (4-[(7-chloroquinolin-4-yl)amino]-2-[(diethylamino)methyl]phenol), which activated both PPAR-alpha & -gamma, for further investigation. Amodiaquine 154-217 peroxisome proliferator activated receptor alpha Mus musculus 241-251 29516607-5 2018 RESULTS: Amodiaquine selectively activated the transcriptional activities of PPARalpha/gamma and enhanced both fatty acid oxidation and glucose uptake without altering insulin secretion in vitro. Amodiaquine 9-20 peroxisome proliferator activated receptor alpha Mus musculus 77-86 29514826-9 2018 The formation of M14 was inhibited by amodiaquine (an HNMT inhibitor) and DCMB, providing additional evidence that both HNMT and TMT catalyzed M14 formation. Amodiaquine 38-49 histamine N-methyltransferase Homo sapiens 54-58 29884802-5 2018 Treatment with CQ or AQ abolished high-glucose-induced phospho-AMPK and phosph-PGC1alpha down-regulation in HKC8 cells. Amodiaquine 21-23 PPARG coactivator 1 alpha Homo sapiens 79-88 29514826-9 2018 The formation of M14 was inhibited by amodiaquine (an HNMT inhibitor) and DCMB, providing additional evidence that both HNMT and TMT catalyzed M14 formation. Amodiaquine 38-49 histamine N-methyltransferase Homo sapiens 120-124 29720942-0 2018 Glutathione S-Transferase P1 Protects Against Amodiaquine Quinoneimines-Induced Cytotoxicity but Does Not Prevent Activation of Endoplasmic Reticulum Stress in HepG2 Cells. Amodiaquine 46-57 glutathione S-transferase pi 1 Homo sapiens 0-28 29720942-2 2018 Glutathione conjugation protects against AQ-induced toxicity and GSTP1 is able to conjugate its quinoneimine metabolites AQ-QI and DEA-QI with glutathione. Amodiaquine 41-43 glutathione S-transferase pi 1 Homo sapiens 65-70 29720942-2 2018 Glutathione conjugation protects against AQ-induced toxicity and GSTP1 is able to conjugate its quinoneimine metabolites AQ-QI and DEA-QI with glutathione. Amodiaquine 121-123 glutathione S-transferase pi 1 Homo sapiens 65-70 29281794-5 2018 Using recombinant NQO1, we showed that low nM concentrations of NQO1 are sufficient to reduce synthetic amodiaquine and carbamazepine quinone-like metabolites in vitro. Amodiaquine 104-115 NAD(P)H quinone dehydrogenase 1 Homo sapiens 18-22 29281794-5 2018 Using recombinant NQO1, we showed that low nM concentrations of NQO1 are sufficient to reduce synthetic amodiaquine and carbamazepine quinone-like metabolites in vitro. Amodiaquine 104-115 NAD(P)H quinone dehydrogenase 1 Homo sapiens 64-68 29281794-7 2018 NQO2 catalyzed the reduction of quinone-like metabolites derived from acetaminophen, clozapine, 4"-hydroxydiclofenac, mefenamic acid, amodiaquine, and carbamazepine. Amodiaquine 134-145 N-ribosyldihydronicotinamide:quinone reductase 2 Homo sapiens 0-4 29087505-1 2018 We have developed an animal model of amodiaquine-induced liver injury that has characteristics very similar to idiosyncratic drug-induced liver injury (IDILI) in humans by impairing immune tolerance using a PD1-/- mouse and cotreatment with anti-CTLA-4. Amodiaquine 37-48 programmed cell death 1 Homo sapiens 207-210 29087505-1 2018 We have developed an animal model of amodiaquine-induced liver injury that has characteristics very similar to idiosyncratic drug-induced liver injury (IDILI) in humans by impairing immune tolerance using a PD1-/- mouse and cotreatment with anti-CTLA-4. Amodiaquine 37-48 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 246-252 29208963-0 2017 Enhanced CD25+Foxp3+ regulatory T cell development by amodiaquine through activation of nuclear receptor 4A. Amodiaquine 54-65 interleukin 2 receptor subunit alpha Homo sapiens 9-13 29208963-0 2017 Enhanced CD25+Foxp3+ regulatory T cell development by amodiaquine through activation of nuclear receptor 4A. Amodiaquine 54-65 forkhead box P3 Homo sapiens 14-19 29208963-5 2017 Despite the anti-proliferative activity of AQ, AQ only moderately decreased iTreg cell proliferation but substantially increased IL-2 production by iTreg cells. Amodiaquine 47-49 interleukin 2 Homo sapiens 129-133 29208963-6 2017 Furthermore, AQ dose-dependently increased iTreg cell development and significantly upregulated iTreg cell markers including CD25. Amodiaquine 13-15 interleukin 2 receptor subunit alpha Homo sapiens 125-129 29208963-7 2017 Interestingly, CD25 expression was decreased at later stages of iTreg cell development but was sustained in the presence of AQ, which was independent of IL-2 signaling pathway. Amodiaquine 124-126 interleukin 2 receptor subunit alpha Homo sapiens 15-19 29208963-8 2017 AQ directly increased CD25 gene transcription by enhancing the DNA-binding and transcriptional activity of nuclear receptor 4 A. Amodiaquine 0-2 interleukin 2 receptor subunit alpha Homo sapiens 22-26 28900272-0 2017 From malaria to cancer: Computational drug repositioning of amodiaquine using PLIP interaction patterns. Amodiaquine 60-71 protein tyrosine phosphatase mitochondrial 1 Homo sapiens 78-82 29061131-8 2017 RESULTS: Amodiaquine, proguanil, pyrimethamine and quinine were the most potent inhibitors of 4-(4-(dimethylamino)styryl)-N-methylpyridinium iodide (ASP) transport, a known substrate of OCT1/2, resulting in half maximal inhibitory concentrations (IC50) of 11, 13, 1.6, and 3.4 microM, respectively. Amodiaquine 9-20 solute carrier family 22 member 1 Homo sapiens 186-190 28934153-8 2017 Selaginellin and selaginellin M had high inhibitory potential for CYP2C8-mediated amodiaquine O-demethylation with IC50 values of 0.5 and 0.9 muM, respectively. Amodiaquine 82-93 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 66-72 28070879-5 2017 Adverse effects of amodiaquine were more common in patients with decreased CYP2C8 metabolism. Amodiaquine 19-30 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 75-81 28900272-5 2017 These patterns characterise BVDU"s interaction with its target s. Using PLIP we performed an in silico screen of all structural data currently available and identified the FDA approved malaria drug amodiaquine as a promising repositioning candidate. Amodiaquine 198-209 protein tyrosine phosphatase mitochondrial 1 Homo sapiens 72-76 28900272-6 2017 We validated our prediction by showing that amodiaquine suppresses chemoresistance in a multiple myeloma cancer cell line by inhibiting the chaperone function of the cancer target Hsp27. Amodiaquine 44-55 heat shock protein family B (small) member 1 Homo sapiens 180-185 28525267-11 2017 This is presumably because the myeloperoxidase in THP-1 cells can bioactivate amodiaquine to a reactive metabolite. Amodiaquine 78-89 myeloperoxidase Homo sapiens 31-46 27788475-4 2016 We then examined the effect of Nurr1 activation on adult hippocampal NPCs using amodiaquine (AQ), an anti-malarial drug that was recently discovered to be a Nurr1 agonist. Amodiaquine 80-91 nuclear receptor subfamily 4, group A, member 2 Mus musculus 157-162 28484907-5 2017 MAM-2201 exhibited mechanism-based inhibition of CYP2C8-catalyzed amodiaquine N-de-ethylation with K i and k inact values of 1.0 microM and 0.0738 min-1, respectively. Amodiaquine 66-77 sarcoglycan gamma Homo sapiens 0-3 28484907-5 2017 MAM-2201 exhibited mechanism-based inhibition of CYP2C8-catalyzed amodiaquine N-de-ethylation with K i and k inact values of 1.0 microM and 0.0738 min-1, respectively. Amodiaquine 66-77 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 49-55 28179134-3 2017 The results demonstrate that most of the unsaturated fatty acids showed marked inhibition towards CYP2C8 mediated amodiaquine N-deethylation followed by inhibition of CYP2C9 and CYP2B6 mediated activities. Amodiaquine 114-125 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 98-104 28478157-0 2017 Human glutathione S-transferases- and NAD(P)H:quinone oxidoreductase 1-catalyzed inactivation of reactive quinoneimines of amodiaquine and N-desethylamodiaquine: Possible implications for susceptibility to amodiaquine-induced liver toxicity. Amodiaquine 123-134 glutathione S-transferase kappa 1 Homo sapiens 6-32 28478157-0 2017 Human glutathione S-transferases- and NAD(P)H:quinone oxidoreductase 1-catalyzed inactivation of reactive quinoneimines of amodiaquine and N-desethylamodiaquine: Possible implications for susceptibility to amodiaquine-induced liver toxicity. Amodiaquine 149-160 glutathione S-transferase kappa 1 Homo sapiens 6-32 28478157-3 2017 Oxidative bioactivation to protein-reactive quinonimines (QIs) by hepatic cytochrome P450s and myeloperoxidase (MPO) have been suggested to be important mechanisms underlying AQ idiosyncratic toxicity. Amodiaquine 175-177 myeloperoxidase Homo sapiens 95-110 28478157-3 2017 Oxidative bioactivation to protein-reactive quinonimines (QIs) by hepatic cytochrome P450s and myeloperoxidase (MPO) have been suggested to be important mechanisms underlying AQ idiosyncratic toxicity. Amodiaquine 175-177 myeloperoxidase Homo sapiens 112-115 28478157-5 2017 In the present study, the activities of 15 recombinant human GSTs and NQO1 in the inactivation of reactive QIs of AQ and its pharmacological active metabolite, N-desethylamodiaquine (DEAQ) were investigated. Amodiaquine 114-116 glutathione S-transferase kappa 1 Homo sapiens 61-65 28478157-5 2017 In the present study, the activities of 15 recombinant human GSTs and NQO1 in the inactivation of reactive QIs of AQ and its pharmacological active metabolite, N-desethylamodiaquine (DEAQ) were investigated. Amodiaquine 114-116 NAD(P)H quinone dehydrogenase 1 Homo sapiens 70-74 28478157-6 2017 The results showed that GSTP1-1, GSTA4-4, GSTM4-4, GSTM2-2 and GSTA2-2 (activity in decreasing order) were active isoforms in catalyzing GSH conjugation of reactive QIs of AQ and DEAQ. Amodiaquine 172-174 glutathione S-transferase pi 1 Homo sapiens 24-31 28478157-6 2017 The results showed that GSTP1-1, GSTA4-4, GSTM4-4, GSTM2-2 and GSTA2-2 (activity in decreasing order) were active isoforms in catalyzing GSH conjugation of reactive QIs of AQ and DEAQ. Amodiaquine 172-174 glutathione S-transferase alpha 4 Homo sapiens 33-40 28478157-6 2017 The results showed that GSTP1-1, GSTA4-4, GSTM4-4, GSTM2-2 and GSTA2-2 (activity in decreasing order) were active isoforms in catalyzing GSH conjugation of reactive QIs of AQ and DEAQ. Amodiaquine 172-174 glutathione S-transferase mu 4 Homo sapiens 42-49 28478157-6 2017 The results showed that GSTP1-1, GSTA4-4, GSTM4-4, GSTM2-2 and GSTA2-2 (activity in decreasing order) were active isoforms in catalyzing GSH conjugation of reactive QIs of AQ and DEAQ. Amodiaquine 172-174 glutathione S-transferase mu 2 Homo sapiens 51-58 28478157-6 2017 The results showed that GSTP1-1, GSTA4-4, GSTM4-4, GSTM2-2 and GSTA2-2 (activity in decreasing order) were active isoforms in catalyzing GSH conjugation of reactive QIs of AQ and DEAQ. Amodiaquine 172-174 glutathione S-transferase alpha 2 Homo sapiens 63-70 27718269-0 2017 Characterization of human cytochrome P450 mediated bioactivation of amodiaquine and its major metabolite N-desethylamodiaquine. Amodiaquine 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 27718269-1 2017 AIMS: Oxidative bioactivation of amodiaquine (AQ) by cytochrome P450s to a reactive quinoneimine is considered as an important mechanism underlying its idiosyncratic hepatotoxicity. Amodiaquine 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 27718269-1 2017 AIMS: Oxidative bioactivation of amodiaquine (AQ) by cytochrome P450s to a reactive quinoneimine is considered as an important mechanism underlying its idiosyncratic hepatotoxicity. Amodiaquine 46-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 27718269-7 2017 For AQ bioactivation, enzyme kinetical parameters were Km , 11.5 +- 2.0 mumol l-1 , Vmax , 59.2 +- 3.2 pmol min-1 mg-1 and CLint , 5.15 mul min-1 mg-1 . Amodiaquine 4-6 CD59 molecule (CD59 blood group) Homo sapiens 108-119 27718269-7 2017 For AQ bioactivation, enzyme kinetical parameters were Km , 11.5 +- 2.0 mumol l-1 , Vmax , 59.2 +- 3.2 pmol min-1 mg-1 and CLint , 5.15 mul min-1 mg-1 . Amodiaquine 4-6 CD59 molecule (CD59 blood group) Homo sapiens 141-152 27718269-11 2017 High expression of CYP3A4, CYP2C8, CYP2C9, and CYP2D6 may be risk factors for hepatotoxicity caused by AQ-therapy. Amodiaquine 103-105 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 27-33 27718269-11 2017 High expression of CYP3A4, CYP2C8, CYP2C9, and CYP2D6 may be risk factors for hepatotoxicity caused by AQ-therapy. Amodiaquine 103-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 27718269-11 2017 High expression of CYP3A4, CYP2C8, CYP2C9, and CYP2D6 may be risk factors for hepatotoxicity caused by AQ-therapy. Amodiaquine 103-105 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 47-53 27788475-6 2016 AQ-treated NPCs showed increased levels of phosphorylation of Akt and ERK1/2 whereas AQ-treated Nurr1 siRNA-transfected NPCs showed no changes in those levels. Amodiaquine 0-2 thymoma viral proto-oncogene 1 Mus musculus 62-65 27788475-6 2016 AQ-treated NPCs showed increased levels of phosphorylation of Akt and ERK1/2 whereas AQ-treated Nurr1 siRNA-transfected NPCs showed no changes in those levels. Amodiaquine 0-2 mitogen-activated protein kinase 3 Mus musculus 70-76 27788475-6 2016 AQ-treated NPCs showed increased levels of phosphorylation of Akt and ERK1/2 whereas AQ-treated Nurr1 siRNA-transfected NPCs showed no changes in those levels. Amodiaquine 85-87 nuclear receptor subfamily 4, group A, member 2 Mus musculus 96-101 26581561-4 2016 Amodiaquine, montelukast, quercetin and rosiglitazone, known as substrates or competitive inhibitors of human CYP2C8, were metabolically depleted by recombinant monkey CYP2C8 at relatively high rates. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 110-116 27695271-0 2016 Inhibition of Human Cytochrome P450 2c8-catalyzed Amodiaquine N-desethylation: Effect of Five Traditionally and Commonly Used Herbs. Amodiaquine 50-61 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 20-39 27057621-3 2016 OBJECTIVE: The study was designed to investigate the possible enhancement of the antimalarial potency as well as possible herb-drug interaction resulting from concurrent administration of MAMA decoction with amodiaquine (AQ). Amodiaquine 208-219 lectin, galactoside-binding, soluble, 3 binding protein Mus musculus 188-192 27057621-3 2016 OBJECTIVE: The study was designed to investigate the possible enhancement of the antimalarial potency as well as possible herb-drug interaction resulting from concurrent administration of MAMA decoction with amodiaquine (AQ). Amodiaquine 221-223 lectin, galactoside-binding, soluble, 3 binding protein Mus musculus 188-192 26581561-4 2016 Amodiaquine, montelukast, quercetin and rosiglitazone, known as substrates or competitive inhibitors of human CYP2C8, were metabolically depleted by recombinant monkey CYP2C8 at relatively high rates. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 168-174 26581561-5 2016 Taken together with our reported findings of the slow eliminations of amodiaquine and montelukast by monkey CYP2C9, CYP2C19 and CYP2C76, the present results suggest that these at least four chemicals may be good marker substrates for monkey CYP2C8. Amodiaquine 70-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 26581561-5 2016 Taken together with our reported findings of the slow eliminations of amodiaquine and montelukast by monkey CYP2C9, CYP2C19 and CYP2C76, the present results suggest that these at least four chemicals may be good marker substrates for monkey CYP2C8. Amodiaquine 70-81 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 116-123 26581561-5 2016 Taken together with our reported findings of the slow eliminations of amodiaquine and montelukast by monkey CYP2C9, CYP2C19 and CYP2C76, the present results suggest that these at least four chemicals may be good marker substrates for monkey CYP2C8. Amodiaquine 70-81 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 241-247 27128896-3 2016 Aschantin potently inhibited CYP2C8-mediated amodiaquine N-de-ethylation, CYP2C9-mediated diclofenac 4"-hydroxylation, CYP2C19-mediated [S]-mephenytoin 4"-hydroxylation, and CYP3A4-mediated midazolam 1"-hydroxylation, with Ki values of 10.2, 3.7, 5.8, and 12.6 microM, respectively. Amodiaquine 45-56 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 29-35 27109480-7 2016 Both CQ and AQ elevated the transcription level of p21 though the activation of p53, but also blocked p21 protein degradation in the presence of cycloheximide, causing p21 protein accumulation mainly in the nucleus. Amodiaquine 12-14 tumor protein p53 Homo sapiens 80-83 27109480-7 2016 Both CQ and AQ elevated the transcription level of p21 though the activation of p53, but also blocked p21 protein degradation in the presence of cycloheximide, causing p21 protein accumulation mainly in the nucleus. Amodiaquine 12-14 cyclin dependent kinase inhibitor 1A Homo sapiens 102-105 27109480-7 2016 Both CQ and AQ elevated the transcription level of p21 though the activation of p53, but also blocked p21 protein degradation in the presence of cycloheximide, causing p21 protein accumulation mainly in the nucleus. Amodiaquine 12-14 cyclin dependent kinase inhibitor 1A Homo sapiens 102-105 27109480-8 2016 Sustained treatment of developing T cells with either CQ or AQ suppressed IFN-gamma production in a dose dependent manner and potently inhibited the differentiation of IFN-gamma-producing Th1 cells. Amodiaquine 60-62 interferon gamma Homo sapiens 74-83 27109480-8 2016 Sustained treatment of developing T cells with either CQ or AQ suppressed IFN-gamma production in a dose dependent manner and potently inhibited the differentiation of IFN-gamma-producing Th1 cells. Amodiaquine 60-62 interferon gamma Homo sapiens 168-177 27109480-9 2016 These results demonstrate that CQ and AQ increase the expression level of p21 and inhibit T cell proliferation and the development of IFN-gamma-producing Th1 cells, thereby revealing beneficial roles in treating a wide range of chronic inflammatory diseases mediated by inflammatory T cells. Amodiaquine 38-40 cyclin dependent kinase inhibitor 1A Homo sapiens 74-77 27109480-9 2016 These results demonstrate that CQ and AQ increase the expression level of p21 and inhibit T cell proliferation and the development of IFN-gamma-producing Th1 cells, thereby revealing beneficial roles in treating a wide range of chronic inflammatory diseases mediated by inflammatory T cells. Amodiaquine 38-40 interferon gamma Homo sapiens 134-143 27109480-0 2016 Anti-inflammatory activity of chloroquine and amodiaquine through p21-mediated suppression of T cell proliferation and Th1 cell differentiation. Amodiaquine 46-57 cyclin dependent kinase inhibitor 1A Homo sapiens 66-69 27109480-6 2016 Interestingly, the cyclin-dependent kinase inhibitor p21 was significantly and dose-dependently increased by CQ, and more potently by AQ, while other cell cycle regulators were unchanged. Amodiaquine 134-136 cyclin dependent kinase inhibitor 1A Homo sapiens 53-56 27109480-7 2016 Both CQ and AQ elevated the transcription level of p21 though the activation of p53, but also blocked p21 protein degradation in the presence of cycloheximide, causing p21 protein accumulation mainly in the nucleus. Amodiaquine 12-14 cyclin dependent kinase inhibitor 1A Homo sapiens 51-54 26647924-7 2016 Interestingly, AQ-induced damage of spermatogenesis recovered over time, based on the survival of promyelocytic leukemia zinc-finger (PLZF)-positive, undifferentiated spermatogonia. Amodiaquine 15-17 zinc finger and BTB domain containing 16 Rattus norvegicus 98-132 27045516-12 2016 An upregulation of P-gp was observed when artemisone and dihydroartemisinin were co-incubated with mefloquine and amodiaquine. Amodiaquine 114-125 ATP binding cassette subfamily B member 1 Homo sapiens 19-23 26851641-3 2016 Recently, reactivation of OP-inhibited AChE by the antimalarial drug amodiaquine was reported. Amodiaquine 69-80 acetylcholinesterase (Cartwright blood group) Homo sapiens 39-43 26851641-5 2016 Thereby, reversible inhibition of human cholinesterases by amodiaquine (AChE >> BChE) was observed. Amodiaquine 59-70 acetylcholinesterase (Cartwright blood group) Homo sapiens 72-76 26851641-5 2016 Thereby, reversible inhibition of human cholinesterases by amodiaquine (AChE >> BChE) was observed. Amodiaquine 59-70 butyrylcholinesterase Homo sapiens 86-90 26851641-6 2016 Additionally, a mixed competitive-non-competitive inhibition type of amodiaquine with human AChE was determined. Amodiaquine 69-80 acetylcholinesterase (Cartwright blood group) Homo sapiens 92-96 26851641-8 2016 Amodiaquine, being a potent, reversible AChE inhibitor, was tested for its potential benefit as a pretreatment to prevent complete irreversible AChE inhibition by the nerve agent soman. Amodiaquine 0-11 acetylcholinesterase (Cartwright blood group) Homo sapiens 40-44 26851641-8 2016 Amodiaquine, being a potent, reversible AChE inhibitor, was tested for its potential benefit as a pretreatment to prevent complete irreversible AChE inhibition by the nerve agent soman. Amodiaquine 0-11 acetylcholinesterase (Cartwright blood group) Homo sapiens 144-148 26851641-9 2016 Hereby, amodiaquine failed to prevent phosphonylation and resulted only in a slight increase of AChE activity after removal of amodiaquine and soman. Amodiaquine 8-19 acetylcholinesterase (Cartwright blood group) Homo sapiens 96-100 26851641-9 2016 Hereby, amodiaquine failed to prevent phosphonylation and resulted only in a slight increase of AChE activity after removal of amodiaquine and soman. Amodiaquine 127-138 acetylcholinesterase (Cartwright blood group) Homo sapiens 96-100 26851641-10 2016 At present the molecular mechanism of amodiaquine-induced reactivation of OP-inhibited AChE is not known, nevertheless amodiaquine could be considered as a template for the design of more potent non-oxime reactivators. Amodiaquine 38-49 acetylcholinesterase (Cartwright blood group) Homo sapiens 87-91 26647924-7 2016 Interestingly, AQ-induced damage of spermatogenesis recovered over time, based on the survival of promyelocytic leukemia zinc-finger (PLZF)-positive, undifferentiated spermatogonia. Amodiaquine 15-17 zinc finger and BTB domain containing 16 Rattus norvegicus 134-138 26641046-4 2015 Tissue was pretreated with or without aminoguanidine and amodiaquine to block the histamine-degrading enzymes diamine oxidase (DAO) and histamine -methyltransferase (HMT), respectively. Amodiaquine 57-68 amine oxidase copper containing 1 Sus scrofa 110-125 26721703-3 2016 CYP2C8 substrate drugs include amodiaquine, cerivastatin, dasabuvir, enzalutamide, imatinib, loperamide, montelukast, paclitaxel, pioglitazone, repaglinide, and rosiglitazone, and the number is increasing. Amodiaquine 31-42 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 0-6 26641046-4 2015 Tissue was pretreated with or without aminoguanidine and amodiaquine to block the histamine-degrading enzymes diamine oxidase (DAO) and histamine -methyltransferase (HMT), respectively. Amodiaquine 57-68 amine oxidase copper containing 1 Sus scrofa 127-130 26124091-6 2015 In particular, we found that two antimalarial drugs, amodiaquine and chloroquine stimulate the transcriptional function of Nurr1 through physical interaction with its ligand binding domain (LBD). Amodiaquine 53-64 nuclear receptor subfamily 4, group A, member 2 Rattus norvegicus 123-128 26310922-4 2015 We show that a clinically used antimalarial drug, Amodiaquine, discovered by this strategy, protects host cells against infection by multiple toxins and viruses by inhibiting host cathepsin B. Amodiaquine 50-61 cathepsin B Homo sapiens 180-191 26154582-0 2015 The Role of CD8 T Cells in Amodiaquine-Induced Liver Injury in PD1-/- Mice Cotreated with Anti-CTLA-4. Amodiaquine 27-38 CD8a molecule Homo sapiens 12-15 26154582-0 2015 The Role of CD8 T Cells in Amodiaquine-Induced Liver Injury in PD1-/- Mice Cotreated with Anti-CTLA-4. Amodiaquine 27-38 programmed cell death 1 Mus musculus 63-66 26154582-4 2015 Recently our laboratory reported a model of amodiaquine (AQ)-induced IDILI using PD1-/- mice and an anti-CTLA4 antibody. Amodiaquine 44-55 programmed cell death 1 Mus musculus 81-84 26154582-4 2015 Recently our laboratory reported a model of amodiaquine (AQ)-induced IDILI using PD1-/- mice and an anti-CTLA4 antibody. Amodiaquine 44-55 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 105-110 26154582-4 2015 Recently our laboratory reported a model of amodiaquine (AQ)-induced IDILI using PD1-/- mice and an anti-CTLA4 antibody. Amodiaquine 57-59 programmed cell death 1 Mus musculus 81-84 26154582-4 2015 Recently our laboratory reported a model of amodiaquine (AQ)-induced IDILI using PD1-/- mice and an anti-CTLA4 antibody. Amodiaquine 57-59 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 105-110 26154582-8 2015 Mice treated with AQ and anti-CTLA4 had a significant increase in percentage of hepatic CD4, CD8, Th17, and Treg cells after 10 weeks of AQ treatment, as well as significantly decreased NK cells. Amodiaquine 18-20 CD4 antigen Mus musculus 88-91 26154582-8 2015 Mice treated with AQ and anti-CTLA4 had a significant increase in percentage of hepatic CD4, CD8, Th17, and Treg cells after 10 weeks of AQ treatment, as well as significantly decreased NK cells. Amodiaquine 18-20 CD8a molecule Homo sapiens 93-96 26154582-8 2015 Mice treated with AQ and anti-CTLA4 had a significant increase in percentage of hepatic CD4, CD8, Th17, and Treg cells after 10 weeks of AQ treatment, as well as significantly decreased NK cells. Amodiaquine 137-139 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 30-35 26154582-10 2015 We found that depletion of CD8 T cells protected mice from AQ-induced liver injury in this model, which strongly suggests that they are responsible for the liver damage. Amodiaquine 59-61 CD8a molecule Homo sapiens 27-30 25959621-3 2015 The changes on G-6-Pase activity and liver glucose output induced by 5-CQA were also investigated. Amodiaquine 71-74 glucose-6-phosphatase catalytic subunit 1 Homo sapiens 15-23 25046026-10 2015 Treatment of Rag1(-/-) mice with AQ resulted in higher ALT activities than C57BL/6 mice, which suggested that the adaptive immune response was responsible for immune tolerance. Amodiaquine 33-35 recombination activating 1 Mus musculus 13-17 25046026-10 2015 Treatment of Rag1(-/-) mice with AQ resulted in higher ALT activities than C57BL/6 mice, which suggested that the adaptive immune response was responsible for immune tolerance. Amodiaquine 33-35 glutamic pyruvic transaminase, soluble Mus musculus 55-58 25046026-11 2015 In contrast, depletion of NK cells significantly attenuated the increase in ALT, which implied a role for NK cells in mild AQ-induced IDILI. Amodiaquine 123-125 glutamic pyruvic transaminase, soluble Mus musculus 76-79 24588327-0 2014 Involvement of myeloperoxidase and NADPH oxidase in the covalent binding of amodiaquine and clozapine to neutrophils: implications for drug-induced agranulocytosis. Amodiaquine 76-87 myeloperoxidase Mus musculus 15-30 24713129-3 2014 Amodiaquine N-deethylation and midazolam 1"-hydroxylation were used as marker reactions for CYP2C8 and CYP3A activity, respectively. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 92-98 24713129-3 2014 Amodiaquine N-deethylation and midazolam 1"-hydroxylation were used as marker reactions for CYP2C8 and CYP3A activity, respectively. Amodiaquine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 103-108 25283142-5 2015 In this study we found that AQ treatment of Cbl-b(-/-) and PD-1(-/-) mice, which have impaired immune tolerance, resulted in a slightly greater injury. Amodiaquine 28-30 Casitas B-lineage lymphoma b Mus musculus 44-49 25283142-6 2015 Cotreatment of C57BL/6 with AQ and anti-CTLA4 also resulted in a greater increase in ALT than treatment with AQ alone; however, these mice also had an increase in T regulatory (Treg) cells and T helper cells expressing PD-1 and CTLA4. Amodiaquine 28-30 glutamic pyruvic transaminase, soluble Mus musculus 85-88 25283142-6 2015 Cotreatment of C57BL/6 with AQ and anti-CTLA4 also resulted in a greater increase in ALT than treatment with AQ alone; however, these mice also had an increase in T regulatory (Treg) cells and T helper cells expressing PD-1 and CTLA4. Amodiaquine 28-30 cytotoxic T-lymphocyte protein 4 Sus scrofa 228-233 25283142-6 2015 Cotreatment of C57BL/6 with AQ and anti-CTLA4 also resulted in a greater increase in ALT than treatment with AQ alone; however, these mice also had an increase in T regulatory (Treg) cells and T helper cells expressing PD-1 and CTLA4. Amodiaquine 109-111 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 40-45 25283142-8 2015 Cotreatment of PD-1(-/-) mice with anti-CTLA4 antibody and AQ resulted in the greatest increase in ALT (200-300 U/L), and necroinflammatory responses characterized by portal infiltration of lymphocytes with interface hepatitis. Amodiaquine 59-61 glutamic pyruvic transaminase, soluble Mus musculus 99-102 25283142-10 2015 In addition, the ALT activity in PD-1(-/-) mice cotreated with anti-CTLA4 antibody and AQ did not return to normal, as it had in other mice. Amodiaquine 87-89 glutamic pyruvic transaminase, soluble Mus musculus 17-20 25388906-9 2014 CONCLUSION: Independent association of SNPs in IL22 and IL-4 with clearance of amodiaquine- and sulphadoxine/pyrimethamine-resistant parasites did not reach statistical significance, but may suggest that not all drug-resistant mutants are adversely affected by the same immune-mediated mechanisms of clearance. Amodiaquine 79-90 interleukin 22 Homo sapiens 47-51 25388906-9 2014 CONCLUSION: Independent association of SNPs in IL22 and IL-4 with clearance of amodiaquine- and sulphadoxine/pyrimethamine-resistant parasites did not reach statistical significance, but may suggest that not all drug-resistant mutants are adversely affected by the same immune-mediated mechanisms of clearance. Amodiaquine 79-90 interleukin 4 Homo sapiens 56-60 24588327-2 2014 Previous studies have shown that both AQ and CLZ are oxidized to reactive intermediates in vitro by activated neutrophils or by the combination of hydrogen peroxide and myeloperoxidase (MPO). Amodiaquine 38-40 myeloperoxidase Mus musculus 169-184 24588327-2 2014 Previous studies have shown that both AQ and CLZ are oxidized to reactive intermediates in vitro by activated neutrophils or by the combination of hydrogen peroxide and myeloperoxidase (MPO). Amodiaquine 38-40 myeloperoxidase Mus musculus 186-189 24588327-5 2014 In this study, we found that the binding of both AQ and CLZ to neutrophils from MPO knockout mice ex vivo decreased approximately 2-fold compared to neutrophils from wild-type mice, whereas binding to activated neutrophils from gp91 knockout (NADPH oxidase null) mice decreased 6-7-fold. Amodiaquine 49-51 myeloperoxidase Mus musculus 80-83 24588327-5 2014 In this study, we found that the binding of both AQ and CLZ to neutrophils from MPO knockout mice ex vivo decreased approximately 2-fold compared to neutrophils from wild-type mice, whereas binding to activated neutrophils from gp91 knockout (NADPH oxidase null) mice decreased 6-7-fold. Amodiaquine 49-51 paired Ig-like receptor B Mus musculus 228-232 24588327-6 2014 When the AQ studies were performed in vivo, again the binding was decreased in MPO knockout mice to about 50% of the binding in wild-type mice; however, covalent binding was significant in the absence of MPO. Amodiaquine 9-11 myeloperoxidase Mus musculus 79-82 24113242-5 2013 Monitoring an established set of protein markers (LAMP1, LC3-II, SQSTM1) and cell ultrastructural changes detected by electron microscopy, we observed that AQ treatment caused autophagic-lysosomal blockade in malignant A375 melanoma cells, a finding substantiated by detection of rapid inactivation of lysosomal cathepsins (CTSB, CTSL, CTSD). Amodiaquine 156-158 lysosomal associated membrane protein 1 Homo sapiens 50-55 24910237-3 2014 The metabolic pathways examined were the CYP1A2-catalyzed tacrine 1-hydroxylation, CYP2B6-catalyzed bupropion hydroxylation, CYP2C8-catalyzed amodiaquine N-deethylation, CYP2C9- catalyzed diclofenac 4"-hydroxylation, CYP2D6-catalyzed dextromethorphan O-demethylation, and CYP3A4-catalyzed midazolam 1"-hydroxylation. Amodiaquine 142-153 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 125-131 24113242-5 2013 Monitoring an established set of protein markers (LAMP1, LC3-II, SQSTM1) and cell ultrastructural changes detected by electron microscopy, we observed that AQ treatment caused autophagic-lysosomal blockade in malignant A375 melanoma cells, a finding substantiated by detection of rapid inactivation of lysosomal cathepsins (CTSB, CTSL, CTSD). Amodiaquine 156-158 sequestosome 1 Homo sapiens 65-71 24113242-5 2013 Monitoring an established set of protein markers (LAMP1, LC3-II, SQSTM1) and cell ultrastructural changes detected by electron microscopy, we observed that AQ treatment caused autophagic-lysosomal blockade in malignant A375 melanoma cells, a finding substantiated by detection of rapid inactivation of lysosomal cathepsins (CTSB, CTSL, CTSD). Amodiaquine 156-158 cathepsin B Homo sapiens 324-328 24113242-5 2013 Monitoring an established set of protein markers (LAMP1, LC3-II, SQSTM1) and cell ultrastructural changes detected by electron microscopy, we observed that AQ treatment caused autophagic-lysosomal blockade in malignant A375 melanoma cells, a finding substantiated by detection of rapid inactivation of lysosomal cathepsins (CTSB, CTSL, CTSD). Amodiaquine 156-158 cathepsin L Homo sapiens 330-334 24113242-5 2013 Monitoring an established set of protein markers (LAMP1, LC3-II, SQSTM1) and cell ultrastructural changes detected by electron microscopy, we observed that AQ treatment caused autophagic-lysosomal blockade in malignant A375 melanoma cells, a finding substantiated by detection of rapid inactivation of lysosomal cathepsins (CTSB, CTSL, CTSD). Amodiaquine 156-158 cathepsin D Homo sapiens 336-340 24113242-7 2013 AQ displayed potent antiproliferative effects, and gene expression array analysis revealed changes at the mRNA (CDKN1A, E2F1) and protein level (TP53, CDKN1A, CCND1, phospho-RB1 [Ser 780]/[Ser 807/811], E2F1) consistent with the observed proliferative blockade in S-phase. Amodiaquine 0-2 E2F transcription factor 1 Homo sapiens 203-207 23204183-0 2013 CYP2C8 status of patients with malaria influences selection of Plasmodium falciparum pfmdr1 alleles after amodiaquine-artesunate treatment. Amodiaquine 106-117 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 0-6 24591743-5 2013 This article deals with the development of a 2D QSAR model based on the inhibitory potential of gemfibrozil, its analogues and corresponding glucuronide conjugates in inhibiting the CYP2C8-catalysed amodiaquine N-deethylation. Amodiaquine 199-210 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 182-188 22014153-3 2012 EXPERIMENTAL APPROACH: Amodiaquine N-deethylation and midazolam 1"-hydroxylation were used as marker reactions for CYP2C8 and CYP3A4/5 activity. Amodiaquine 23-34 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 115-121 22658498-0 2012 Investigation of the interaction between amodiaquine and human serum albumin by fluorescence spectroscopy and molecular modeling. Amodiaquine 41-52 albumin Homo sapiens 63-76 22658498-1 2012 The interaction of amodiaquine (AQ) with human serum albumin (HSA) has been studied by fluorescence spectroscopy. Amodiaquine 19-30 albumin Homo sapiens 47-60 22658498-1 2012 The interaction of amodiaquine (AQ) with human serum albumin (HSA) has been studied by fluorescence spectroscopy. Amodiaquine 32-34 albumin Homo sapiens 47-60 22531455-2 2012 CYP2C8 enzyme is involved in the metabolism of the anti-malarials amodiaquine and chloroquine. Amodiaquine 66-77 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 0-6 22014153-3 2012 EXPERIMENTAL APPROACH: Amodiaquine N-deethylation and midazolam 1"-hydroxylation were used as marker reactions for CYP2C8 and CYP3A4/5 activity. Amodiaquine 23-34 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 126-132 21726541-3 2011 In this study, the contribution of the redox partners cytochrome P450 reductase (CPR) and cytochrome b5 to the substrate dependent activity of CYP2C8.3 (R139K, K399R) was investigated in human liver microsomes (HLMs) and Escherichia coli expressed recombinant CYP2C8 proteins using amodiaquine, paclitaxel, rosiglitazone and cerivastatin as probe substrates. Amodiaquine 282-293 cytochrome p450 oxidoreductase Homo sapiens 54-79 22003869-1 2012 CONTEXT: Artesunate (AS) and amodiaquine (AQ) are two prodrugs widely used as antimalarial agents and are metabolized by the CYP P450 2A6 (CYP 2A6) and CYP P450 2C8 (CYP 2C8) enzymes, respectively. Amodiaquine 29-40 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 125-137 22003869-1 2012 CONTEXT: Artesunate (AS) and amodiaquine (AQ) are two prodrugs widely used as antimalarial agents and are metabolized by the CYP P450 2A6 (CYP 2A6) and CYP P450 2C8 (CYP 2C8) enzymes, respectively. Amodiaquine 29-40 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 139-146 22003869-1 2012 CONTEXT: Artesunate (AS) and amodiaquine (AQ) are two prodrugs widely used as antimalarial agents and are metabolized by the CYP P450 2A6 (CYP 2A6) and CYP P450 2C8 (CYP 2C8) enzymes, respectively. Amodiaquine 29-40 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 152-164 22003869-1 2012 CONTEXT: Artesunate (AS) and amodiaquine (AQ) are two prodrugs widely used as antimalarial agents and are metabolized by the CYP P450 2A6 (CYP 2A6) and CYP P450 2C8 (CYP 2C8) enzymes, respectively. Amodiaquine 29-40 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 166-173 22003869-1 2012 CONTEXT: Artesunate (AS) and amodiaquine (AQ) are two prodrugs widely used as antimalarial agents and are metabolized by the CYP P450 2A6 (CYP 2A6) and CYP P450 2C8 (CYP 2C8) enzymes, respectively. Amodiaquine 42-44 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 125-137 22003869-1 2012 CONTEXT: Artesunate (AS) and amodiaquine (AQ) are two prodrugs widely used as antimalarial agents and are metabolized by the CYP P450 2A6 (CYP 2A6) and CYP P450 2C8 (CYP 2C8) enzymes, respectively. Amodiaquine 42-44 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 139-146 22003869-1 2012 CONTEXT: Artesunate (AS) and amodiaquine (AQ) are two prodrugs widely used as antimalarial agents and are metabolized by the CYP P450 2A6 (CYP 2A6) and CYP P450 2C8 (CYP 2C8) enzymes, respectively. Amodiaquine 42-44 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 152-164 22003869-1 2012 CONTEXT: Artesunate (AS) and amodiaquine (AQ) are two prodrugs widely used as antimalarial agents and are metabolized by the CYP P450 2A6 (CYP 2A6) and CYP P450 2C8 (CYP 2C8) enzymes, respectively. Amodiaquine 42-44 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 166-173 22122444-11 2011 IPT in children with artesunate (AS + SP), amodiaquine (AQ) + SPQ or SP alone is a cost effective and safe intervention for reducing the burden of malaria in children in areas with markedly seasonal malaria transmission. Amodiaquine 43-54 tRNA isopentenyltransferase 1 Homo sapiens 0-3 22122444-11 2011 IPT in children with artesunate (AS + SP), amodiaquine (AQ) + SPQ or SP alone is a cost effective and safe intervention for reducing the burden of malaria in children in areas with markedly seasonal malaria transmission. Amodiaquine 56-58 tRNA isopentenyltransferase 1 Homo sapiens 0-3 21726541-3 2011 In this study, the contribution of the redox partners cytochrome P450 reductase (CPR) and cytochrome b5 to the substrate dependent activity of CYP2C8.3 (R139K, K399R) was investigated in human liver microsomes (HLMs) and Escherichia coli expressed recombinant CYP2C8 proteins using amodiaquine, paclitaxel, rosiglitazone and cerivastatin as probe substrates. Amodiaquine 282-293 cytochrome p450 oxidoreductase Homo sapiens 81-84 21726541-3 2011 In this study, the contribution of the redox partners cytochrome P450 reductase (CPR) and cytochrome b5 to the substrate dependent activity of CYP2C8.3 (R139K, K399R) was investigated in human liver microsomes (HLMs) and Escherichia coli expressed recombinant CYP2C8 proteins using amodiaquine, paclitaxel, rosiglitazone and cerivastatin as probe substrates. Amodiaquine 282-293 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 143-149 21726541-7 2011 In HLMs, carriers of the CYP2C8*1/*3 genotype were as active as CYP2C8*1/*1 towards the CYP2C8 specific reaction amodiaquine N-deethylation. Amodiaquine 113-124 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 25-31 21726541-7 2011 In HLMs, carriers of the CYP2C8*1/*3 genotype were as active as CYP2C8*1/*1 towards the CYP2C8 specific reaction amodiaquine N-deethylation. Amodiaquine 113-124 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 64-70 21726541-7 2011 In HLMs, carriers of the CYP2C8*1/*3 genotype were as active as CYP2C8*1/*1 towards the CYP2C8 specific reaction amodiaquine N-deethylation. Amodiaquine 113-124 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 64-70 20382755-5 2010 Both dog CYP2A13 and CYP2A25 exhibited little or no catalytic activity toward other common cytochrome P450 probe substrates, including bupropion, amodiaquine, diclofenac, S-mephenytoin, bufuralol, dextromethorphan, midazolam, and testosterone. Amodiaquine 146-157 cytochrome P450 family 2 subfamily A polypeptide 13 Canis lupus familiaris 9-16 21529379-5 2011 RESULTS: The combination therapy of AQ and SP results in more pronounced hepatotoxicity as revealed by elevated level of serum ALT, AST with respect to their individual drug exposure regimen. Amodiaquine 36-38 glutamic pyruvic transaminase, soluble Mus musculus 127-130 21529379-5 2011 RESULTS: The combination therapy of AQ and SP results in more pronounced hepatotoxicity as revealed by elevated level of serum ALT, AST with respect to their individual drug exposure regimen. Amodiaquine 36-38 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 132-135 21597194-1 2011 Chlorogenic acid (CQA) is one of the major polyphenols in apple and a good substrate for the polyphenol oxidase (PPO) in apple. Amodiaquine 18-21 polyphenol oxidase, chloroplastic Malus domestica 93-111 21597194-1 2011 Chlorogenic acid (CQA) is one of the major polyphenols in apple and a good substrate for the polyphenol oxidase (PPO) in apple. Amodiaquine 18-21 polyphenol oxidase, chloroplastic Malus domestica 113-116 21597194-5 2011 Although catechins and CQA were oxidized by PPO, some catechins seemed to be non-enzymatically oxidized by CQA quinone. Amodiaquine 23-26 polyphenol oxidase, chloroplastic Malus domestica 44-47 20877236-5 2010 Eupatilin and jaceosidin were also found to moderately inhibit CYP2C19-catalyzed [S]-mephenytoin 4"-hydroxylation, CYP2D6-catalyzed bufuralol 1"-hydroxylation, and CYP2C8-catalyzed amodiaquine N-deethylation. Amodiaquine 181-192 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 63-70 20382755-5 2010 Both dog CYP2A13 and CYP2A25 exhibited little or no catalytic activity toward other common cytochrome P450 probe substrates, including bupropion, amodiaquine, diclofenac, S-mephenytoin, bufuralol, dextromethorphan, midazolam, and testosterone. Amodiaquine 146-157 cytochrome P450, family 2, subfamily A, polypeptide 7 Canis lupus familiaris 21-28 20212335-10 2010 CONCLUSION: The research provides the evidence that mutations present in dhfr and mdr1 86 has a significant effect on the type of treatment following SP and AQ chemotherapy. Amodiaquine 157-159 ATP binding cassette subfamily B member 1 Homo sapiens 82-86