PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 2504855-4 1989 Apolipoprotein B concentrations were lowered by 25.4% (pravastatin) and 22.0% (gemfibrozil). Gemfibrozil 79-90 apolipoprotein B Homo sapiens 0-16 2504855-8 1989 Gemfibrozil, in contrast, increased HDL3 cholesterol level by 9% because of an enrichment of HDL3 particles in both free cholesterol and cholesteryl esters and lowered the flotation rate of HDL3 (p less than 0.05). Gemfibrozil 0-11 HDL3 Homo sapiens 36-40 2504855-8 1989 Gemfibrozil, in contrast, increased HDL3 cholesterol level by 9% because of an enrichment of HDL3 particles in both free cholesterol and cholesteryl esters and lowered the flotation rate of HDL3 (p less than 0.05). Gemfibrozil 0-11 HDL3 Homo sapiens 93-97 2504855-8 1989 Gemfibrozil, in contrast, increased HDL3 cholesterol level by 9% because of an enrichment of HDL3 particles in both free cholesterol and cholesteryl esters and lowered the flotation rate of HDL3 (p less than 0.05). Gemfibrozil 0-11 HDL3 Homo sapiens 93-97 3477829-2 1987 Three antilipidemic drugs, clofibrate, bezafibrate, and gemfibrozil, two of which had previously been shown to reduce Hb/O2 affinity in vitro, were tested in mice for their ability to affect Hb/O2 affinity and to alter the radiosensitivity of the RIF-1 sarcoma. Gemfibrozil 56-67 replication timing regulatory factor 1 Mus musculus 247-252 3164977-3 1988 Type III HLP reasserted itself following an 8-week interruption of gemfibrozil therapy. Gemfibrozil 67-78 HLP Homo sapiens 9-12 2917701-6 1989 Addition of lovastatin to gemfibrozil effectively reduced total cholesterol (25%), LDL-chol (30%), and LDL-apoB (19%). Gemfibrozil 26-37 apolipoprotein B Homo sapiens 107-111 3162679-7 1988 In animals receiving the standard diet, fenofibrate, gemfibrozil and nicotinic acid decreased the triglyceride (TG) level (-28%, -31%, and -38%) by lowering VLDL-TG (-37%, -42%, and -49%), fenofibrate and gemfibrozil increased HDL-cholesterol by 18% and 31%, respectively. Gemfibrozil 53-64 CD320 antigen Mus musculus 157-161 3162679-7 1988 In animals receiving the standard diet, fenofibrate, gemfibrozil and nicotinic acid decreased the triglyceride (TG) level (-28%, -31%, and -38%) by lowering VLDL-TG (-37%, -42%, and -49%), fenofibrate and gemfibrozil increased HDL-cholesterol by 18% and 31%, respectively. Gemfibrozil 205-216 CD320 antigen Mus musculus 157-161 3464282-0 1986 Gemfibrozil treatment and the relationship between HDL cholesterol and apoE distribution in cholesterol-fed rats. Gemfibrozil 0-11 apolipoprotein E Rattus norvegicus 71-75 3555908-0 1987 Normalization of lipoprotein lipase and hepatic lipase by gemfibrozil results in correction of lipoprotein abnormalities in chronic renal failure. Gemfibrozil 58-69 lipoprotein lipase Homo sapiens 17-35 3555908-0 1987 Normalization of lipoprotein lipase and hepatic lipase by gemfibrozil results in correction of lipoprotein abnormalities in chronic renal failure. Gemfibrozil 58-69 lipase C, hepatic type Homo sapiens 40-54 3923042-5 1985 Gemfibrozil significantly increased plasma HDL cholesterol, apolipoprotein (apo) AI, and apo AII by 36%, 29%, and 38% from base line, respectively. Gemfibrozil 0-11 apolipoprotein A1 Homo sapiens 60-83 3081014-0 1986 Gemfibrozil increases both apo A-I and apo E concentrations. Gemfibrozil 0-11 apolipoprotein A1 Rattus norvegicus 27-44 3081014-5 1986 Only gemfibrozil increased plasma apo E levels which are characteristically low in this rat model. Gemfibrozil 5-16 apolipoprotein E Rattus norvegicus 34-39 3862732-0 1985 Apolipoprotein changes associated with the plasma lipid-regulating activity of gemfibrozil in cholesterol-fed rats. Gemfibrozil 79-90 apolipoprotein E Rattus norvegicus 0-14 3862732-6 1985 Whole plasma apoE and apoA-I concentrations were decreased and apoB increased due to cholesterol feeding as determined by electroimmunoassay, but again gemfibrozil treatment prevented these diet-induced alterations. Gemfibrozil 152-163 apolipoprotein E Rattus norvegicus 13-17 3862732-6 1985 Whole plasma apoE and apoA-I concentrations were decreased and apoB increased due to cholesterol feeding as determined by electroimmunoassay, but again gemfibrozil treatment prevented these diet-induced alterations. Gemfibrozil 152-163 apolipoprotein B Rattus norvegicus 63-67 3862732-8 1985 Gemfibrozil lowered the concentration of both apoB variants and prevented the shift of apoE from HDL to lower density lipoproteins. Gemfibrozil 0-11 apolipoprotein B Rattus norvegicus 46-50 3862732-8 1985 Gemfibrozil lowered the concentration of both apoB variants and prevented the shift of apoE from HDL to lower density lipoproteins. Gemfibrozil 0-11 apolipoprotein E Rattus norvegicus 87-91 3862732-10 1985 These methods also revealed a shift of apoA-IV from HDL to the d greater than 1.21 g/ml, lipoprotein-free fraction with gemfibrozil treatment when blood was taken from fasted or postabsorptive animals. Gemfibrozil 120-131 apolipoprotein A4 Rattus norvegicus 39-46 3862732-11 1985 Since it was also noted that in chow-fed rats more apoA-IV was present in the d greater than 1.21 g/ml fraction in the postabsorptive or fed state compared to fasted animals, it could be postulated that the shift of apoA-IV into this fraction in gemfibrozil-treated rats is related to an accelerated clearance of chylomicrons. Gemfibrozil 246-257 apolipoprotein A4 Rattus norvegicus 51-58 3862732-11 1985 Since it was also noted that in chow-fed rats more apoA-IV was present in the d greater than 1.21 g/ml fraction in the postabsorptive or fed state compared to fasted animals, it could be postulated that the shift of apoA-IV into this fraction in gemfibrozil-treated rats is related to an accelerated clearance of chylomicrons. Gemfibrozil 246-257 apolipoprotein A4 Rattus norvegicus 216-223 3923042-8 1985 Gemfibrozil increased synthetic rates of apo AI and apo AII by 27% and 34%, respectively, without changing the fractional catabolic rates. Gemfibrozil 0-11 apolipoprotein A1 Homo sapiens 41-47 3923042-9 1985 Stimulation of apo AI and apo AII synthesis by gemfibrozil was associated with the appearance in plasma of smaller (and heavier) HDL particles as assessed by gradient gel electrophoresis and HDL composition. Gemfibrozil 47-58 apolipoprotein A1 Homo sapiens 15-21 7018466-1 1981 The lipid lowering drug Gemfibrozil significantly increased the levels of HDL-apoproteins, apoA-I and apoA-II in a group of 20 Swedish hyperlipemic males who were given 1200 mg daily of the drug for 8 weeks and thereafter submitted to a 4 weeks placebo period. Gemfibrozil 24-35 apolipoprotein A1 Homo sapiens 91-97 6188330-2 1982 During gemfibrozil use, plasma prekallikrein and kininogen were increased significantly while kallikrein inhibitors increased only slightly. Gemfibrozil 7-18 kallikrein B1 Homo sapiens 24-44 6188330-2 1982 During gemfibrozil use, plasma prekallikrein and kininogen were increased significantly while kallikrein inhibitors increased only slightly. Gemfibrozil 7-18 kallikrein related peptidase 4 Homo sapiens 34-44 3907298-2 1985 Gemfibrozil also produces increases in both Apo AI and AII, and in kininogen and prekallikrein. Gemfibrozil 0-11 apolipoprotein A1 Homo sapiens 44-50 3907298-2 1985 Gemfibrozil also produces increases in both Apo AI and AII, and in kininogen and prekallikrein. Gemfibrozil 0-11 NLR family pyrin domain containing 3 Homo sapiens 55-58 6867010-4 1983 Gemfibrozil at 250 mg/kg/day for 4 weeks increased catalase by 86%. Gemfibrozil 0-11 catalase Rattus norvegicus 51-59 7010929-11 1981 Apoprotein A-I remained unchanged, but A-II increased by 20% during gemfibrozil treatment. Gemfibrozil 68-79 NLR family pyrin domain containing 3 Homo sapiens 39-43 7018466-1 1981 The lipid lowering drug Gemfibrozil significantly increased the levels of HDL-apoproteins, apoA-I and apoA-II in a group of 20 Swedish hyperlipemic males who were given 1200 mg daily of the drug for 8 weeks and thereafter submitted to a 4 weeks placebo period. Gemfibrozil 24-35 apolipoprotein A2 Homo sapiens 102-109 190608-9 1976 The postheparin plasma lipoprotein lipase and hepatic lipase activities, determined separately by an immunochemical method, increased during four weeks of gemfibrozil treatment (+18.1% and +20.6% respectively), but neither of these changes was significantly correlated with the changes in any of the serum lipid or lipoprotein levels. Gemfibrozil 155-166 lipoprotein lipase Homo sapiens 23-41 190608-9 1976 The postheparin plasma lipoprotein lipase and hepatic lipase activities, determined separately by an immunochemical method, increased during four weeks of gemfibrozil treatment (+18.1% and +20.6% respectively), but neither of these changes was significantly correlated with the changes in any of the serum lipid or lipoprotein levels. Gemfibrozil 155-166 lipase C, hepatic type Homo sapiens 46-60 32997809-1 2021 AIMS: The strong cytochrome P450 (CYP) 2C8 inhibitor gemfibrozil has been demonstrated to increase the area under the plasma concentration-time curve from 0 to infinity (AUC0- ) of ACT-333679, an active metabolite of selexipag, by 11-fold. Gemfibrozil 53-64 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 17-42 32997809-2 2021 Similarly as gemfibrozil, the CYP2C8 inhibitor clopidogrel also increases clopidogrel increased ACT-333679 concentration by 1.9-fold after a single loading dose (300 mg once daily) and 2.7-fold after repeated treatment with the maintenance dose (75 mg once daily) in Europeans. Gemfibrozil 13-24 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 30-36 33514677-9 2021 These findings highlight the importance of PPARalpha-dependent astroglial GDNF pathway in gemfibrozil-mediated protection of dopaminergic neurons in an animal model of PD and suggest possible therapeutic use of gemfibrozil in PD patients.SIGNIFICANCE STATEMENT:Increasing the level of glial cell-derived neurotrophic factor (GDNF) in the brain is important for the protection of dopamine neurons in Parkinson"s disease (PD). Gemfibrozil 90-101 peroxisome proliferator activated receptor alpha Homo sapiens 43-52 33514677-9 2021 These findings highlight the importance of PPARalpha-dependent astroglial GDNF pathway in gemfibrozil-mediated protection of dopaminergic neurons in an animal model of PD and suggest possible therapeutic use of gemfibrozil in PD patients.SIGNIFICANCE STATEMENT:Increasing the level of glial cell-derived neurotrophic factor (GDNF) in the brain is important for the protection of dopamine neurons in Parkinson"s disease (PD). Gemfibrozil 90-101 glial cell derived neurotrophic factor Homo sapiens 74-78 33514677-0 2021 Gemfibrozil Protects Dopaminergic Neurons in a Mouse Model of Parkinson"s disease via PPARalpha-Dependent Astrocytic GDNF Pathway. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 86-95 33514677-9 2021 These findings highlight the importance of PPARalpha-dependent astroglial GDNF pathway in gemfibrozil-mediated protection of dopaminergic neurons in an animal model of PD and suggest possible therapeutic use of gemfibrozil in PD patients.SIGNIFICANCE STATEMENT:Increasing the level of glial cell-derived neurotrophic factor (GDNF) in the brain is important for the protection of dopamine neurons in Parkinson"s disease (PD). Gemfibrozil 211-222 peroxisome proliferator activated receptor alpha Homo sapiens 43-52 33514677-0 2021 Gemfibrozil Protects Dopaminergic Neurons in a Mouse Model of Parkinson"s disease via PPARalpha-Dependent Astrocytic GDNF Pathway. Gemfibrozil 0-11 glial cell line derived neurotrophic factor Mus musculus 117-121 33514677-9 2021 These findings highlight the importance of PPARalpha-dependent astroglial GDNF pathway in gemfibrozil-mediated protection of dopaminergic neurons in an animal model of PD and suggest possible therapeutic use of gemfibrozil in PD patients.SIGNIFICANCE STATEMENT:Increasing the level of glial cell-derived neurotrophic factor (GDNF) in the brain is important for the protection of dopamine neurons in Parkinson"s disease (PD). Gemfibrozil 211-222 glial cell derived neurotrophic factor Homo sapiens 74-78 33514677-7 2021 Gemfibrozil-mediated protection of the nigrostriatal and locomotor activities in WT but not PPARalpha-/- mice from MPTP intoxication suggests that gemfibrozil needs the involvement of PPARalpha in protecting dopaminergic neurons. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 184-193 33514677-11 2021 Here, we delineate that gemfibrozil, a lipid-lowering drug, stimulates GDNF in astrocytes via peroxisome proliferator-activated receptor alpha (PPARalpha). Gemfibrozil 24-35 glial cell derived neurotrophic factor Homo sapiens 71-75 33514677-8 2021 While investigating further mechanisms, we found that gemfibrozil stimulated the transcription of glial-derived neurotrophic factor (GDNF) gene in astrocytes via PPARalpha and that gemfibrozil protected nigral neurons, normalized striatal fibers and neurotransmitters, and improved locomotor activities in MPTP-intoxicated Gfaf cre mice, but not Gdnf Deltaastro mice lacking GDNF in astrocytes. Gemfibrozil 54-65 glial cell line derived neurotrophic factor Mus musculus 133-137 33514677-11 2021 Here, we delineate that gemfibrozil, a lipid-lowering drug, stimulates GDNF in astrocytes via peroxisome proliferator-activated receptor alpha (PPARalpha). Gemfibrozil 24-35 peroxisome proliferator activated receptor alpha Mus musculus 94-142 33514677-8 2021 While investigating further mechanisms, we found that gemfibrozil stimulated the transcription of glial-derived neurotrophic factor (GDNF) gene in astrocytes via PPARalpha and that gemfibrozil protected nigral neurons, normalized striatal fibers and neurotransmitters, and improved locomotor activities in MPTP-intoxicated Gfaf cre mice, but not Gdnf Deltaastro mice lacking GDNF in astrocytes. Gemfibrozil 54-65 peroxisome proliferator activated receptor alpha Mus musculus 162-171 33514677-8 2021 While investigating further mechanisms, we found that gemfibrozil stimulated the transcription of glial-derived neurotrophic factor (GDNF) gene in astrocytes via PPARalpha and that gemfibrozil protected nigral neurons, normalized striatal fibers and neurotransmitters, and improved locomotor activities in MPTP-intoxicated Gfaf cre mice, but not Gdnf Deltaastro mice lacking GDNF in astrocytes. Gemfibrozil 54-65 glial cell line derived neurotrophic factor Mus musculus 346-350 33514677-8 2021 While investigating further mechanisms, we found that gemfibrozil stimulated the transcription of glial-derived neurotrophic factor (GDNF) gene in astrocytes via PPARalpha and that gemfibrozil protected nigral neurons, normalized striatal fibers and neurotransmitters, and improved locomotor activities in MPTP-intoxicated Gfaf cre mice, but not Gdnf Deltaastro mice lacking GDNF in astrocytes. Gemfibrozil 54-65 glial cell line derived neurotrophic factor Mus musculus 375-379 33514677-11 2021 Here, we delineate that gemfibrozil, a lipid-lowering drug, stimulates GDNF in astrocytes via peroxisome proliferator-activated receptor alpha (PPARalpha). Gemfibrozil 24-35 peroxisome proliferator activated receptor alpha Homo sapiens 144-153 33514677-8 2021 While investigating further mechanisms, we found that gemfibrozil stimulated the transcription of glial-derived neurotrophic factor (GDNF) gene in astrocytes via PPARalpha and that gemfibrozil protected nigral neurons, normalized striatal fibers and neurotransmitters, and improved locomotor activities in MPTP-intoxicated Gfaf cre mice, but not Gdnf Deltaastro mice lacking GDNF in astrocytes. Gemfibrozil 181-192 glial cell line derived neurotrophic factor Mus musculus 346-350 33514677-8 2021 While investigating further mechanisms, we found that gemfibrozil stimulated the transcription of glial-derived neurotrophic factor (GDNF) gene in astrocytes via PPARalpha and that gemfibrozil protected nigral neurons, normalized striatal fibers and neurotransmitters, and improved locomotor activities in MPTP-intoxicated Gfaf cre mice, but not Gdnf Deltaastro mice lacking GDNF in astrocytes. Gemfibrozil 181-192 glial cell line derived neurotrophic factor Mus musculus 375-379 33514677-12 2021 Moreover, gemfibrozil protected nigral neurons, normalized striatal fibers and neurotransmitters, and improved locomotor activities from MPTP toxicity via PPARalpha-dependent astroglial GDNF pathway. Gemfibrozil 10-21 peroxisome proliferator activated receptor alpha Homo sapiens 155-164 33514677-12 2021 Moreover, gemfibrozil protected nigral neurons, normalized striatal fibers and neurotransmitters, and improved locomotor activities from MPTP toxicity via PPARalpha-dependent astroglial GDNF pathway. Gemfibrozil 10-21 glial cell derived neurotrophic factor Homo sapiens 186-190 32708962-9 2020 Erythropoietin levels were increased in the serum of gemfibrozil-treated animals, and we also observed an increased expression of hypoxia-inducible factor-2 alpha (HIF-2alpha) and erythropoietin in renal tissue, while PPAR-alpha knockout mice treated with gemfibrozil did not present increased levels of serum erythropoietin or tissue HIF-2alpha and erythropoietin mRNA levels in the kidneys. Gemfibrozil 53-64 erythropoietin Mus musculus 310-324 32540489-0 2020 Gemfibrozil improves lipid metabolism in Nile tilapia Oreochromis niloticus fed a high-carbohydrate diet through peroxisome proliferator activated receptor-alpha activation. Gemfibrozil 0-11 peroxisome proliferator-activated receptor alpha Oreochromis niloticus 113-161 32540489-10 2020 This study indicates that gemfibrozil can improve lipid metabolism and maintain high antioxidant and anti-inflammatory capacity through activating PPARalpha in Nile tilapia fed a high carbohydrate diet. Gemfibrozil 26-37 peroxisome proliferator-activated receptor alpha Oreochromis niloticus 147-156 32708962-0 2020 Gemfibrozil Induces Anemia, Leukopenia and Reduces Hematopoietic Stem Cells via PPAR-alpha in Mice. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 80-90 32708962-4 2020 Gemfibrozil is a fibrate that acts via peroxisome proliferator-activated receptor alpha to promote changes in lipid metabolism and decrease serum triglyceride levels. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 39-87 32708962-9 2020 Erythropoietin levels were increased in the serum of gemfibrozil-treated animals, and we also observed an increased expression of hypoxia-inducible factor-2 alpha (HIF-2alpha) and erythropoietin in renal tissue, while PPAR-alpha knockout mice treated with gemfibrozil did not present increased levels of serum erythropoietin or tissue HIF-2alpha and erythropoietin mRNA levels in the kidneys. Gemfibrozil 53-64 endothelial PAS domain protein 1 Mus musculus 335-345 32708962-6 2020 Considering that gemfibrozil may lead to anemia and that gemfibrozil acts via peroxisome proliferator-activated receptor alpha, we treated wild-type and peroxisome proliferator-activated receptor alpha-knockout mice with gemfibrozil for four consecutive days. Gemfibrozil 57-68 peroxisome proliferator activated receptor alpha Mus musculus 78-126 32708962-6 2020 Considering that gemfibrozil may lead to anemia and that gemfibrozil acts via peroxisome proliferator-activated receptor alpha, we treated wild-type and peroxisome proliferator-activated receptor alpha-knockout mice with gemfibrozil for four consecutive days. Gemfibrozil 57-68 peroxisome proliferator activated receptor alpha Mus musculus 78-126 32708962-8 2020 PPAR-alpha-knockout mice were capable of reversing all of those reduced parameters induced by gemfibrozil treatment. Gemfibrozil 94-105 peroxisome proliferator activated receptor alpha Mus musculus 0-10 32708962-9 2020 Erythropoietin levels were increased in the serum of gemfibrozil-treated animals, and we also observed an increased expression of hypoxia-inducible factor-2 alpha (HIF-2alpha) and erythropoietin in renal tissue, while PPAR-alpha knockout mice treated with gemfibrozil did not present increased levels of serum erythropoietin or tissue HIF-2alpha and erythropoietin mRNA levels in the kidneys. Gemfibrozil 53-64 erythropoietin Mus musculus 310-324 32708962-9 2020 Erythropoietin levels were increased in the serum of gemfibrozil-treated animals, and we also observed an increased expression of hypoxia-inducible factor-2 alpha (HIF-2alpha) and erythropoietin in renal tissue, while PPAR-alpha knockout mice treated with gemfibrozil did not present increased levels of serum erythropoietin or tissue HIF-2alpha and erythropoietin mRNA levels in the kidneys. Gemfibrozil 256-267 erythropoietin Mus musculus 0-14 32708962-9 2020 Erythropoietin levels were increased in the serum of gemfibrozil-treated animals, and we also observed an increased expression of hypoxia-inducible factor-2 alpha (HIF-2alpha) and erythropoietin in renal tissue, while PPAR-alpha knockout mice treated with gemfibrozil did not present increased levels of serum erythropoietin or tissue HIF-2alpha and erythropoietin mRNA levels in the kidneys. Gemfibrozil 53-64 erythropoietin Mus musculus 0-14 32708962-9 2020 Erythropoietin levels were increased in the serum of gemfibrozil-treated animals, and we also observed an increased expression of hypoxia-inducible factor-2 alpha (HIF-2alpha) and erythropoietin in renal tissue, while PPAR-alpha knockout mice treated with gemfibrozil did not present increased levels of serum erythropoietin or tissue HIF-2alpha and erythropoietin mRNA levels in the kidneys. Gemfibrozil 256-267 endothelial PAS domain protein 1 Mus musculus 164-174 32708962-9 2020 Erythropoietin levels were increased in the serum of gemfibrozil-treated animals, and we also observed an increased expression of hypoxia-inducible factor-2 alpha (HIF-2alpha) and erythropoietin in renal tissue, while PPAR-alpha knockout mice treated with gemfibrozil did not present increased levels of serum erythropoietin or tissue HIF-2alpha and erythropoietin mRNA levels in the kidneys. Gemfibrozil 53-64 endothelial PAS domain protein 1 Mus musculus 130-162 32708962-9 2020 Erythropoietin levels were increased in the serum of gemfibrozil-treated animals, and we also observed an increased expression of hypoxia-inducible factor-2 alpha (HIF-2alpha) and erythropoietin in renal tissue, while PPAR-alpha knockout mice treated with gemfibrozil did not present increased levels of serum erythropoietin or tissue HIF-2alpha and erythropoietin mRNA levels in the kidneys. Gemfibrozil 53-64 endothelial PAS domain protein 1 Mus musculus 164-174 32708962-11 2020 Here, we show for the first time that gemfibrozil treatment leads to anemia and leukopenia via peroxisome proliferator-activated receptor alpha in mice. Gemfibrozil 38-49 peroxisome proliferator activated receptor alpha Mus musculus 95-143 32708962-9 2020 Erythropoietin levels were increased in the serum of gemfibrozil-treated animals, and we also observed an increased expression of hypoxia-inducible factor-2 alpha (HIF-2alpha) and erythropoietin in renal tissue, while PPAR-alpha knockout mice treated with gemfibrozil did not present increased levels of serum erythropoietin or tissue HIF-2alpha and erythropoietin mRNA levels in the kidneys. Gemfibrozil 53-64 erythropoietin Mus musculus 180-194 32708962-9 2020 Erythropoietin levels were increased in the serum of gemfibrozil-treated animals, and we also observed an increased expression of hypoxia-inducible factor-2 alpha (HIF-2alpha) and erythropoietin in renal tissue, while PPAR-alpha knockout mice treated with gemfibrozil did not present increased levels of serum erythropoietin or tissue HIF-2alpha and erythropoietin mRNA levels in the kidneys. Gemfibrozil 53-64 peroxisome proliferator activated receptor alpha Mus musculus 218-228 32155958-5 2020 Notably, administration of gemfibrozil (GEM), a PPARalpha activator, significantly reduced the in vivo Mabc load and inflammatory response in mice. Gemfibrozil 27-38 peroxisome proliferator activated receptor alpha Mus musculus 48-57 32155958-5 2020 Notably, administration of gemfibrozil (GEM), a PPARalpha activator, significantly reduced the in vivo Mabc load and inflammatory response in mice. Gemfibrozil 40-43 peroxisome proliferator activated receptor alpha Mus musculus 48-57 30898012-6 2020 We found that pharmacological activation of PPARA by the PPARA agonists gemfibrozil and Wy14643 induces autophagy in human microglia (HM) cells and U251 human glioma cells stably expressing the human APP (amyloid beta precursor protein) mutant (APP-p.M671L) and this effect is PPARA-dependent. Gemfibrozil 72-83 peroxisome proliferator activated receptor alpha Homo sapiens 44-49 31287236-2 2020 A 3-arm, open-label, randomized, single-dose, fixed-sequence crossover study was conducted to characterize the effects of the strong inhibitors gemfibrozil (of CYP2C8) and itraconazole (of CYP3A) and the strong inducer rifampin (of CYP3A) on the single-dose pharmacokinetics of brigatinib. Gemfibrozil 144-155 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 160-166 31963528-7 2020 Conversely, gemfibrozil, an agonist of PPARalpha, elicited the increase of PPARalpha, OCT-2, and MATE-1 expression by 115%, 144%, and 376%, respectively. Gemfibrozil 12-23 peroxisome proliferator activated receptor alpha Mus musculus 39-48 31963528-7 2020 Conversely, gemfibrozil, an agonist of PPARalpha, elicited the increase of PPARalpha, OCT-2, and MATE-1 expression by 115%, 144%, and 376%, respectively. Gemfibrozil 12-23 peroxisome proliferator activated receptor alpha Mus musculus 75-84 31963528-7 2020 Conversely, gemfibrozil, an agonist of PPARalpha, elicited the increase of PPARalpha, OCT-2, and MATE-1 expression by 115%, 144%, and 376%, respectively. Gemfibrozil 12-23 POU domain, class 2, transcription factor 2 Mus musculus 86-91 31963528-7 2020 Conversely, gemfibrozil, an agonist of PPARalpha, elicited the increase of PPARalpha, OCT-2, and MATE-1 expression by 115%, 144%, and 376%, respectively. Gemfibrozil 12-23 solute carrier family 47, member 1 Mus musculus 97-103 31926091-9 2020 Drug interactions may occur in patients who use premixed insulin with glimepiride, lisinopril, fenofibrate, candesartan, irbesartan, and gemfibrozil. Gemfibrozil 137-148 insulin Homo sapiens 57-64 30898012-6 2020 We found that pharmacological activation of PPARA by the PPARA agonists gemfibrozil and Wy14643 induces autophagy in human microglia (HM) cells and U251 human glioma cells stably expressing the human APP (amyloid beta precursor protein) mutant (APP-p.M671L) and this effect is PPARA-dependent. Gemfibrozil 72-83 peroxisome proliferator activated receptor alpha Homo sapiens 57-62 30898012-6 2020 We found that pharmacological activation of PPARA by the PPARA agonists gemfibrozil and Wy14643 induces autophagy in human microglia (HM) cells and U251 human glioma cells stably expressing the human APP (amyloid beta precursor protein) mutant (APP-p.M671L) and this effect is PPARA-dependent. Gemfibrozil 72-83 peroxisome proliferator activated receptor alpha Homo sapiens 57-62 30898012-9 2020 These results indicated that PPARA is an important factor regulating autophagy in the clearance of Abeta and suggested gemfibrozil be assessed as a possible treatment for AD. Gemfibrozil 119-130 peroxisome proliferator activated receptor alpha Homo sapiens 29-34 30074417-0 2019 Combined gemfibrozil (peroxisome proliferator-activated receptor alpha agonist) with reduced steroid dose gives a similar management picture as the full steroid dose in a rat adjuvant-induced arthritis model. Gemfibrozil 9-20 peroxisome proliferator activated receptor alpha Rattus norvegicus 22-70 31127527-5 2019 Our mRNA analyses followed by different immunoassays clearly indicated that PPARalpha agonist gemfibrozil strongly upregulated the expression of Nurr1 in wild-type, but not PPARalpha-/-, DA neurons. Gemfibrozil 94-105 peroxisome proliferator activated receptor alpha Mus musculus 76-85 31127527-5 2019 Our mRNA analyses followed by different immunoassays clearly indicated that PPARalpha agonist gemfibrozil strongly upregulated the expression of Nurr1 in wild-type, but not PPARalpha-/-, DA neurons. Gemfibrozil 94-105 nuclear receptor subfamily 4, group A, member 2 Mus musculus 145-150 31127527-7 2019 Finally, oral administration of gemfibrozil increased Nurr1 expression in vivo in nigra of wild-type, but not PPARalpha-/-, mice identifying PPARalpha as a novel regulator of Nurr1 expression and associated protection of DA neurons. Gemfibrozil 32-43 nuclear receptor subfamily 4, group A, member 2 Mus musculus 54-59 31127527-7 2019 Finally, oral administration of gemfibrozil increased Nurr1 expression in vivo in nigra of wild-type, but not PPARalpha-/-, mice identifying PPARalpha as a novel regulator of Nurr1 expression and associated protection of DA neurons. Gemfibrozil 32-43 peroxisome proliferator activated receptor alpha Mus musculus 141-150 31127527-7 2019 Finally, oral administration of gemfibrozil increased Nurr1 expression in vivo in nigra of wild-type, but not PPARalpha-/-, mice identifying PPARalpha as a novel regulator of Nurr1 expression and associated protection of DA neurons. Gemfibrozil 32-43 nuclear receptor subfamily 4, group A, member 2 Mus musculus 175-180 31357780-12 2019 Four drugs (gemfibrozil, bezafibrate, omega-3 carboxylic acid and glycyrrhizic acid) were screened by L1000 and DrugBank to activate lipoprotein lipase. Gemfibrozil 12-23 lipoprotein lipase Homo sapiens 133-151 31129789-0 2019 Physiologically Based Pharmacokinetic Models for Prediction of Complex CYP2C8 and OATP1B1 (SLCO1B1) Drug-Drug-Gene Interactions: A Modeling Network of Gemfibrozil, Repaglinide, Pioglitazone, Rifampicin, Clarithromycin and Itraconazole. Gemfibrozil 151-162 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 71-77 31129789-0 2019 Physiologically Based Pharmacokinetic Models for Prediction of Complex CYP2C8 and OATP1B1 (SLCO1B1) Drug-Drug-Gene Interactions: A Modeling Network of Gemfibrozil, Repaglinide, Pioglitazone, Rifampicin, Clarithromycin and Itraconazole. Gemfibrozil 151-162 solute carrier organic anion transporter family member 1B1 Homo sapiens 82-89 31129789-0 2019 Physiologically Based Pharmacokinetic Models for Prediction of Complex CYP2C8 and OATP1B1 (SLCO1B1) Drug-Drug-Gene Interactions: A Modeling Network of Gemfibrozil, Repaglinide, Pioglitazone, Rifampicin, Clarithromycin and Itraconazole. Gemfibrozil 151-162 solute carrier organic anion transporter family member 1B1 Homo sapiens 91-98 31129789-5 2019 METHODS: PBPK models of the CYP2C8 and organic-anion-transporting polypeptide (OATP) 1B1 perpetrator drug gemfibrozil (parent-metabolite model) and the CYP2C8 victim drugs repaglinide (also an OATP1B1 substrate) and pioglitazone were developed using a total of 103 clinical studies. Gemfibrozil 106-117 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 28-34 31129789-5 2019 METHODS: PBPK models of the CYP2C8 and organic-anion-transporting polypeptide (OATP) 1B1 perpetrator drug gemfibrozil (parent-metabolite model) and the CYP2C8 victim drugs repaglinide (also an OATP1B1 substrate) and pioglitazone were developed using a total of 103 clinical studies. Gemfibrozil 106-117 solute carrier organic anion transporter family member 1B1 Homo sapiens 39-88 31129789-5 2019 METHODS: PBPK models of the CYP2C8 and organic-anion-transporting polypeptide (OATP) 1B1 perpetrator drug gemfibrozil (parent-metabolite model) and the CYP2C8 victim drugs repaglinide (also an OATP1B1 substrate) and pioglitazone were developed using a total of 103 clinical studies. Gemfibrozil 106-117 solute carrier organic anion transporter family member 1B1 Homo sapiens 193-200 31336317-2 2019 GFZ may affect the metabolism of various drugs, including statins, by inhibiting the sinusoidal influx transporter OATP1B1 and also CYP2C9 and CYP2C8 enzymes. Gemfibrozil 0-3 solute carrier organic anion transporter family member 1B1 Homo sapiens 115-122 31336317-2 2019 GFZ may affect the metabolism of various drugs, including statins, by inhibiting the sinusoidal influx transporter OATP1B1 and also CYP2C9 and CYP2C8 enzymes. Gemfibrozil 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 31336317-2 2019 GFZ may affect the metabolism of various drugs, including statins, by inhibiting the sinusoidal influx transporter OATP1B1 and also CYP2C9 and CYP2C8 enzymes. Gemfibrozil 0-3 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 143-149 31259309-0 2019 Gemfibrozil, a Lipid-Lowering Drug, Lowers Amyloid Plaque Pathology and Enhances Memory in a Mouse Model of Alzheimer"s Disease via Peroxisome Proliferator-Activated Receptor alpha. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 132-180 31259309-6 2019 Finally, we delineate that gemfibrozil requires the transcription factor peroxisome proliferator-activated receptor alpha (PPARalpha) to exhibit its amyloid lowering and memory enhancing effects in 5XFAD mice. Gemfibrozil 27-38 peroxisome proliferator activated receptor alpha Mus musculus 73-121 31259309-6 2019 Finally, we delineate that gemfibrozil requires the transcription factor peroxisome proliferator-activated receptor alpha (PPARalpha) to exhibit its amyloid lowering and memory enhancing effects in 5XFAD mice. Gemfibrozil 27-38 peroxisome proliferator activated receptor alpha Mus musculus 123-132 29240983-6 2018 The formation of CAG was significantly inhibited by azidothymidine and gemfibrozil (well-characterized UGT2B7 substrates) in a concentration-dependent manner, or by fluconazole (a typical UGT2B7-selective inhibitor) in a time-dependent manner, for both HLMs and rUGT2B7, respectively. Gemfibrozil 71-82 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 103-109 30500419-6 2019 Moreover, CS-induced (sub-chronic) ceramide-accumulation was significantly (p < 0.05) reduced by treatment with TFEB/autophagy-inducing drug, gemfibrozil (GEM), suggesting that autophagy regulates CS-induced ceramide-accumulation. Gemfibrozil 142-153 transcription factor EB Homo sapiens 112-116 30500419-6 2019 Moreover, CS-induced (sub-chronic) ceramide-accumulation was significantly (p < 0.05) reduced by treatment with TFEB/autophagy-inducing drug, gemfibrozil (GEM), suggesting that autophagy regulates CS-induced ceramide-accumulation. Gemfibrozil 155-158 transcription factor EB Homo sapiens 112-116 30260990-0 2018 Testing the PPAR hypothesis of tobacco use disorder in humans: A randomized trial of the impact of gemfibrozil (a partial PPARalpha agonist) in smokers. Gemfibrozil 99-110 peroxisome proliferator activated receptor alpha Homo sapiens 122-131 30260990-2 2018 Our goal was to investigate the potential of gemfibrozil, a PPARalpha agonist, on reducing tobacco smoking in humans. Gemfibrozil 45-56 peroxisome proliferator activated receptor alpha Homo sapiens 60-69 30023335-1 2018 Purpose: Gemfibrozil (GEM) apart from agonist activity at peroxisome proliferator-activated receptor-alpha (PPAR-alpha) has antioxidant and anti-inflammatory properties. Gemfibrozil 9-20 peroxisome proliferator activated receptor alpha Mus musculus 108-118 30023335-8 2018 Results: GEM led to significant decrease in serum ALT and AST activities and increase in catalase activity and hepatic GSH level and reduces malondialdehyde and ROS levels in the liver tissue. Gemfibrozil 9-12 glutamic pyruvic transaminase, soluble Mus musculus 50-53 30023335-8 2018 Results: GEM led to significant decrease in serum ALT and AST activities and increase in catalase activity and hepatic GSH level and reduces malondialdehyde and ROS levels in the liver tissue. Gemfibrozil 9-12 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 58-61 29662003-9 2018 The PPAR system thus offers an important hope in the management of atherogenic dyslipidemias, although concerns regarding potential adverse events such as the rise of plasma creatinine, gallstone formation, drug-drug interactions (i.e., gemfibrozil) and myopathy should also be acknowledged. Gemfibrozil 237-248 peroxisome proliferator activated receptor alpha Homo sapiens 4-8 30630815-0 2019 Clopidogrel and Gemfibrozil Strongly Inhibit the CYP2C8-Dependent Formation of 3-Hydroxydesloratadine and Increase Desloratadine Exposure In Humans. Gemfibrozil 16-27 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 49-55 30630815-3 2019 Glucuronide metabolites of clopidogrel and gemfibrozil act as time-dependent inhibitors of CYP2C8, but they have not been compared clinically. Gemfibrozil 43-54 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 91-97 30680004-0 2019 Gemfibrozil reduces lipid accumulation in SMMC-7721 cells via the involvement of PPARalpha and SREBP1. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Homo sapiens 81-90 30680004-0 2019 Gemfibrozil reduces lipid accumulation in SMMC-7721 cells via the involvement of PPARalpha and SREBP1. Gemfibrozil 0-11 sterol regulatory element binding transcription factor 1 Homo sapiens 95-101 30680004-6 2019 Upregulation of peroxisome proliferator-activated receptor alpha (PPARalpha) protein and sterol regulatory element-binding protein 1 (SREBP1) was detected following treatment with GEM. Gemfibrozil 180-183 peroxisome proliferator activated receptor alpha Homo sapiens 16-64 30680004-6 2019 Upregulation of peroxisome proliferator-activated receptor alpha (PPARalpha) protein and sterol regulatory element-binding protein 1 (SREBP1) was detected following treatment with GEM. Gemfibrozil 180-183 peroxisome proliferator activated receptor alpha Homo sapiens 66-75 30680004-6 2019 Upregulation of peroxisome proliferator-activated receptor alpha (PPARalpha) protein and sterol regulatory element-binding protein 1 (SREBP1) was detected following treatment with GEM. Gemfibrozil 180-183 sterol regulatory element binding transcription factor 1 Homo sapiens 89-132 30680004-6 2019 Upregulation of peroxisome proliferator-activated receptor alpha (PPARalpha) protein and sterol regulatory element-binding protein 1 (SREBP1) was detected following treatment with GEM. Gemfibrozil 180-183 sterol regulatory element binding transcription factor 1 Homo sapiens 134-140 30251457-3 2018 METHODS: In this study, wild-type and Ppara-null mice fed a medium-fat diet (MFD) were administered gemfibrozil and fenofibrate for 3 months respectively, to explore the effect and action mechanism. Gemfibrozil 100-111 peroxisome proliferator activated receptor alpha Mus musculus 38-43 30251457-6 2018 The STAT3 pathway was activated in adipose and hepatic tissues in positive control, and inhibited in groups treated with gemfibrozil and fenofibrate. Gemfibrozil 121-132 signal transducer and activator of transcription 3 Mus musculus 4-9 30335192-10 2018 In the tDDI verification, the model reasonably predicted pravastatin tDDI caused by rifampicin and gemfibrozil OATP1B1/3 inhibition but under-predicted tDDI caused by cyclosporine. Gemfibrozil 99-110 solute carrier organic anion transporter family member 1B1 Homo sapiens 111-118 29864903-6 2018 The data suggest that combined treatment with EPA and ATM is beneficial to endothelial function and was unique to EPA and ATM since similar improvements could not be recapitulated by substituting another O3FA docosahexaenoic acid (DHA) or other TG-lowering agents such as fenofibrate, niacin, or gemfibrozil. Gemfibrozil 296-307 ATM serine/threonine kinase Homo sapiens 54-57 29712725-8 2018 The DDIs were then simulated using the established PBPK models with previously obtained in vitro inhibition constants of CsA or GEM/GEM-glu against the OATP1B1 and cytochrome P450s. Gemfibrozil 128-131 solute carrier organic anion transporter family member 1B1 Homo sapiens 152-159 29534628-0 2018 Gemfibrozil has antidepressant effects in mice: Involvement of the hippocampal brain-derived neurotrophic factor system. Gemfibrozil 0-11 brain derived neurotrophic factor Mus musculus 79-112 29534628-3 2018 Besides being a lipid-regulating agent, gemfibrozil is an agonist of peroxisome proliferator-activated receptor-alpha (PPAR-alpha). Gemfibrozil 40-51 peroxisome proliferator activated receptor alpha Mus musculus 69-117 29534628-3 2018 Besides being a lipid-regulating agent, gemfibrozil is an agonist of peroxisome proliferator-activated receptor-alpha (PPAR-alpha). Gemfibrozil 40-51 peroxisome proliferator activated receptor alpha Homo sapiens 119-129 29534628-5 2018 The changes in brain-derived neurotrophic factor (BDNF) signaling cascade in the brain after CUMS and gemfibrozil treatment were further assessed. Gemfibrozil 102-113 brain derived neurotrophic factor Mus musculus 50-54 29534628-9 2018 Gemfibrozil treatment also restored CUMS-induced inhibition of the hippocampal BDNF signaling pathway. Gemfibrozil 0-11 brain derived neurotrophic factor Mus musculus 79-83 29534628-10 2018 Blocking PPAR-alpha and BDNF but not the serotonergic system abolished the antidepressant effects of gemfibrozil on mice. Gemfibrozil 101-112 peroxisome proliferator activated receptor alpha Mus musculus 9-19 29534628-10 2018 Blocking PPAR-alpha and BDNF but not the serotonergic system abolished the antidepressant effects of gemfibrozil on mice. Gemfibrozil 101-112 brain derived neurotrophic factor Mus musculus 24-28 29534628-11 2018 Gemfibrozil produced antidepressant effects in mice by promoting the hippocampal BDNF system. Gemfibrozil 0-11 brain derived neurotrophic factor Mus musculus 81-85 29240983-6 2018 The formation of CAG was significantly inhibited by azidothymidine and gemfibrozil (well-characterized UGT2B7 substrates) in a concentration-dependent manner, or by fluconazole (a typical UGT2B7-selective inhibitor) in a time-dependent manner, for both HLMs and rUGT2B7, respectively. Gemfibrozil 71-82 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 188-194 29240983-6 2018 The formation of CAG was significantly inhibited by azidothymidine and gemfibrozil (well-characterized UGT2B7 substrates) in a concentration-dependent manner, or by fluconazole (a typical UGT2B7-selective inhibitor) in a time-dependent manner, for both HLMs and rUGT2B7, respectively. Gemfibrozil 71-82 UDP glucuronosyltransferase family 2 member B7 Rattus norvegicus 262-269 28444890-2 2017 Prescribing information for VIEKIRA PAK contraindicates gemfibrozil, a strong CYP2C8 inhibitor, because coadministration significantly increases dasabuvir exposures, which may increase the risk of QT prolongation. Gemfibrozil 56-67 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 78-84 28653847-5 2017 As interaction perpetrators with CYP2C8, glucuronides of gemfibrozil and clopidogrel showed marked clinical drug-drug interactions (e.g., with cerivastatin and repaglinide), which are more than expected from the parent drug. Gemfibrozil 57-68 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 33-39 29874752-7 2018 In this study, there was significant induction of CYP19A2 in individual exposures of diltiazem, fluoxetine, gemfibrozil and metformin at concentrations measured in Lake Michigan. Gemfibrozil 108-119 cytochrome P450, family 19, subfamily A, polypeptide 1b Danio rerio 50-57 28752027-3 2017 PPARs constitute a recognized druggable target and indeed several classes of drugs used in the treatment of metabolic disease symptoms, such as dyslipidemia (fibrates, e.g. fenofibrate and gemfibrozil) and diabetes (thiazolidinediones, e.g. rosiglitazone and pioglitazone) are ligands for the various PPAR isoforms. Gemfibrozil 189-200 peroxisome proliferator activated receptor alpha Homo sapiens 0-4 28374976-0 2017 Peroxisome Proliferator-Activated Receptor alpha Activation Suppresses Cytochrome P450 Induction Potential in Mice Treated with Gemfibrozil. Gemfibrozil 128-139 peroxisome proliferator activated receptor alpha Mus musculus 0-48 28374976-0 2017 Peroxisome Proliferator-Activated Receptor alpha Activation Suppresses Cytochrome P450 Induction Potential in Mice Treated with Gemfibrozil. Gemfibrozil 128-139 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 71-86 28374976-1 2017 Gemfibrozil, a peroxisome proliferator-activated receptor alpha (PPARalpha) agonist, is widely used for hypertriglyceridaemia and mixed hyperlipidaemia. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 15-63 28374976-1 2017 Gemfibrozil, a peroxisome proliferator-activated receptor alpha (PPARalpha) agonist, is widely used for hypertriglyceridaemia and mixed hyperlipidaemia. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 65-74 28374976-2 2017 Drug-drug interaction of gemfibrozil and other PPARalpha agonists has been reported. Gemfibrozil 25-36 peroxisome proliferator activated receptor alpha Mus musculus 47-56 28374976-4 2017 In this study, wild-type mice were first fed gemfibrozil-containing diets (0.375%, 0.75% and 1.5%) for 14 days to establish a dose-response relationship for CYP induction. Gemfibrozil 45-56 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 157-160 28374976-6 2017 CYP3a, CYP2b and CYP2c were induced in a dose-dependent manner by gemfibrozil. Gemfibrozil 66-77 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 0-5 28374976-6 2017 CYP3a, CYP2b and CYP2c were induced in a dose-dependent manner by gemfibrozil. Gemfibrozil 66-77 cytochrome P450, family 2, subfamily b, polypeptide 10 Mus musculus 7-12 28374976-6 2017 CYP3a, CYP2b and CYP2c were induced in a dose-dependent manner by gemfibrozil. Gemfibrozil 66-77 cytochrome P450, family 2, subfamily c, polypeptide 29 Mus musculus 17-22 28374976-8 2017 So, gemfibrozil induced CYP, and this action was inhibited by activated PPARalpha. Gemfibrozil 4-15 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 24-27 28374976-8 2017 So, gemfibrozil induced CYP, and this action was inhibited by activated PPARalpha. Gemfibrozil 4-15 peroxisome proliferator activated receptor alpha Mus musculus 72-81 28479356-5 2017 The time profiles of RPG and the inhibitors were analyzed by PBPK models, considering the inhibition of OATP1B1 and CYP3A4 by CsA or OATP1B1 inhibition by GEM and its glucuronide and the mechanism-based inhibition of CYP2C8 by GEM glucuronide. Gemfibrozil 155-158 solute carrier organic anion transporter family member 1B1 Homo sapiens 104-111 28479356-5 2017 The time profiles of RPG and the inhibitors were analyzed by PBPK models, considering the inhibition of OATP1B1 and CYP3A4 by CsA or OATP1B1 inhibition by GEM and its glucuronide and the mechanism-based inhibition of CYP2C8 by GEM glucuronide. Gemfibrozil 155-158 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 28479356-5 2017 The time profiles of RPG and the inhibitors were analyzed by PBPK models, considering the inhibition of OATP1B1 and CYP3A4 by CsA or OATP1B1 inhibition by GEM and its glucuronide and the mechanism-based inhibition of CYP2C8 by GEM glucuronide. Gemfibrozil 155-158 solute carrier organic anion transporter family member 1B1 Homo sapiens 133-140 27858342-6 2017 RESULTS: The statin models reasonably predicted the observed exposure change due to Organic Anion Transporting Polypeptide (OATP) 1B1 polymorphism or clinical DDIs with itraconazole, erythromycin, and gemfibrozil, while under-predicted the observed DDIs caused by rifampin and cyclosporine. Gemfibrozil 201-212 solute carrier organic anion transporter family member 1B1 Homo sapiens 84-133 28199020-5 2017 Therefore, we administered gemfibrozil (gem), an food and drug administration (FDA)-approved lipid-lowering drug, which has been shown to stimulate lysosomal biogenesis and induce anti-inflammation, orally, at a dose of 7.5 mg/kg body wt/day to Cln2(-/-) mice. Gemfibrozil 27-38 tripeptidyl peptidase I Mus musculus 245-249 28623936-6 2017 Gemfibrozil has shown efficacy in an animal model of NCL known as CLN2 (late infantile classic juvenile) and has been shown to be safe for lowering lipids in children. Gemfibrozil 0-11 tripeptidyl peptidase 1 Homo sapiens 66-70 28343425-5 2017 Expert opinion: In addition to the aim of improving lipid profile with fibrates, the interest in new PPAR-alpha activators stems from the need to overcome some of the clinical problems encountered with dose-dependent adverse events; a rise of plasma creatinine, gallstone formation, drug-drug interactions (i.e. gemfibrozil), and myopathy. Gemfibrozil 312-323 peroxisome proliferator activated receptor alpha Homo sapiens 101-111 28199020-6 2017 We observed that gem-fed Cln2(-/-) mice lived longer by more than 10 weeks and had better motor activity compared to vehicle (0.1% Methyl cellulose) treatment. Gemfibrozil 17-20 tripeptidyl peptidase I Mus musculus 25-29 27665778-5 2016 PPARalpha agonist activity of HDBB was elucidated by comparison with gene expression data of typical PPARalpha agonist, i.e. clofibrate, WY-14643, gemfibrozil, and fenofibrate, from TG GATEs database. Gemfibrozil 147-158 peroxisome proliferator activated receptor alpha Rattus norvegicus 0-9 28296193-6 2017 The predicted effects of gemfibrozil and its metabolite were moderate (1.88-fold increase in rosuvastatin AUC) and mediated primarily via inhibition of hepatic OATP1B1 and renal organic cation transporter 3. Gemfibrozil 25-36 solute carrier organic anion transporter family member 1B1 Homo sapiens 160-167 27648490-5 2017 Clinical DDIs of montelukast were evaluated using physiologically based pharmacokinetic modeling; and simulation of the interactions with gemfibrozil-CYP2C8 and OATP1B1/1B3 inhibitor, clarithromycin-CYP3A and OATP1B1/1B3 inhibitor, and itraconazole-CYP3A inhibitor, implicated OATPs-CYP2C8-CYP2C8 interplay as the primary determinant of montelukast pharmacokinetics. Gemfibrozil 138-149 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 150-156 27918198-0 2017 PPARalpha-dependent increase of mouse urine output by gemfibrozil and fenofibrate. Gemfibrozil 54-65 peroxisome proliferator activated receptor alpha Mus musculus 0-9 27918198-11 2017 These results revealed a new pharmacodynamic effect of clinically prescribed PPARalpha agonists and suggested the potential value of gemfibrozil in modification of blood pressure. Gemfibrozil 133-144 peroxisome proliferator activated receptor alpha Mus musculus 77-86 30090481-0 2017 Application research on PPARalpha-transgenic mice in preclinical safety evaluation of gemfibrozil. Gemfibrozil 86-97 peroxisome proliferator activated receptor alpha Mus musculus 24-33 27624558-7 2016 Antioxidants (glutathione and N-Acetylcysteine), Cyp3a inhibitor ketoconazole and SLCO1B1 inhibitor gemfibrozil suppressed cytotoxicity and ROS formation in primary hepatocytes of diabetic rats. Gemfibrozil 100-111 solute carrier organic anion transporter family member 1B1 Homo sapiens 82-89 28340451-5 2017 Since enzalutamide is extensively metabolized by CYP2C8, its plasma levels are likely to be raised if coadministered with strong CYP2C8 inhibitors such as gemfibrozil or pioglitazone. Gemfibrozil 155-166 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 49-55 28340451-5 2017 Since enzalutamide is extensively metabolized by CYP2C8, its plasma levels are likely to be raised if coadministered with strong CYP2C8 inhibitors such as gemfibrozil or pioglitazone. Gemfibrozil 155-166 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 129-135 28065762-5 2017 We then developed a high-throughput screen and tested for correction of deficient cells with lentiviral-mediated CLN5 or CLN6 gene transfer and fibrate drugs, gemfibrozil and fenofibrate in CLN6 deficient neural cultures. Gemfibrozil 159-170 ceroid-lipofuscinosis neuronal protein 6 Ovis aries 190-194 27259818-1 2016 CYP2C8 is involved in the metabolic clearance of several important drugs and recent reports have shown that acyl glucuronides of gemfibrozil and clopidogrel are potent time-dependent inhibitors of CYP2C8 activity. Gemfibrozil 129-140 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 0-6 27259818-1 2016 CYP2C8 is involved in the metabolic clearance of several important drugs and recent reports have shown that acyl glucuronides of gemfibrozil and clopidogrel are potent time-dependent inhibitors of CYP2C8 activity. Gemfibrozil 129-140 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 197-203 27462272-11 2016 Furthermore, incubation of zebrafish with another PPARalpha agonist, gemfibrozil, also increased expression of the mbp promoter-driven fluorescent reporter in an SREBF-dependent manner. Gemfibrozil 69-80 peroxisome proliferator-activated receptor alpha b Danio rerio 50-59 27462272-11 2016 Furthermore, incubation of zebrafish with another PPARalpha agonist, gemfibrozil, also increased expression of the mbp promoter-driven fluorescent reporter in an SREBF-dependent manner. Gemfibrozil 69-80 myelin basic protein a Danio rerio 115-118 27209701-0 2016 Gemfibrozil not fenofibrate decreases systemic glucose level via PPARalpha. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 65-74 27209701-8 2016 Secondary regulation on the transcription of pyruvate kinase, and gluconolactonase were observed following gemfibrozil treatment, which was differential between WT mice and KO mice. Gemfibrozil 107-118 regucalcin Mus musculus 66-82 27209701-9 2016 CONCLUSIONS: Gemfibrozil, not fenofibrate, down-regulates systemic glucose level and glycogen storage in the liver dependent on PPARalpha, suggesting its potential value for treatment of dyslipidemia with concurrent diabetes or high glucose levels. Gemfibrozil 13-24 peroxisome proliferator activated receptor alpha Mus musculus 128-137 26080426-7 2015 Gemfibrozil, an agonist of PPARalpha, induced the recruitment of PPARalpha:retinoid x receptor alpha, but not PPARgamma coactivator 1alpha (PGC1alpha), to the Adam10 promoter in wild-type mouse hippocampal neurons and shifted APP processing toward the alpha-secretase, as determined by augmented soluble APPalpha and decreased Abeta production. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 27-36 26721703-5 2016 In vivo, already a small dose of gemfibrozil, i.e., 10% of its therapeutic dose, is a strong, irreversible inhibitor of CYP2C8. Gemfibrozil 33-44 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 120-126 26721703-6 2016 Interestingly, recent findings indicate that the acyl-beta-glucuronides of gemfibrozil and clopidogrel cause metabolism-dependent inactivation of CYP2C8, leading to a strong potential for drug interactions. Gemfibrozil 75-86 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 146-152 26195223-2 2015 In the present study, gemfibrozil preincubation time-dependently inhibited the metabolism of pioglitazone in the cytochrome P450 (CYP)- and UDP-glucuronosyltransferase (UGT)-activated human liver microsomes. Gemfibrozil 22-33 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 140-167 26195223-2 2015 In the present study, gemfibrozil preincubation time-dependently inhibited the metabolism of pioglitazone in the cytochrome P450 (CYP)- and UDP-glucuronosyltransferase (UGT)-activated human liver microsomes. Gemfibrozil 22-33 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 169-172 26080426-7 2015 Gemfibrozil, an agonist of PPARalpha, induced the recruitment of PPARalpha:retinoid x receptor alpha, but not PPARgamma coactivator 1alpha (PGC1alpha), to the Adam10 promoter in wild-type mouse hippocampal neurons and shifted APP processing toward the alpha-secretase, as determined by augmented soluble APPalpha and decreased Abeta production. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 65-74 26080426-7 2015 Gemfibrozil, an agonist of PPARalpha, induced the recruitment of PPARalpha:retinoid x receptor alpha, but not PPARgamma coactivator 1alpha (PGC1alpha), to the Adam10 promoter in wild-type mouse hippocampal neurons and shifted APP processing toward the alpha-secretase, as determined by augmented soluble APPalpha and decreased Abeta production. Gemfibrozil 0-11 retinoid X receptor alpha Mus musculus 75-100 26080426-7 2015 Gemfibrozil, an agonist of PPARalpha, induced the recruitment of PPARalpha:retinoid x receptor alpha, but not PPARgamma coactivator 1alpha (PGC1alpha), to the Adam10 promoter in wild-type mouse hippocampal neurons and shifted APP processing toward the alpha-secretase, as determined by augmented soluble APPalpha and decreased Abeta production. Gemfibrozil 0-11 a disintegrin and metallopeptidase domain 10 Mus musculus 159-165 26080426-7 2015 Gemfibrozil, an agonist of PPARalpha, induced the recruitment of PPARalpha:retinoid x receptor alpha, but not PPARgamma coactivator 1alpha (PGC1alpha), to the Adam10 promoter in wild-type mouse hippocampal neurons and shifted APP processing toward the alpha-secretase, as determined by augmented soluble APPalpha and decreased Abeta production. Gemfibrozil 0-11 amyloid beta (A4) precursor protein Mus musculus 327-332 25595597-6 2015 Other inhibitors of CYP2C8 (gemfibrozil, montelukast, clopidogrel glucuronide, repaglinide, and cerivastatin) also caused extensive inhibition of 3-hydroxydesloratadine formation (73%-100%). Gemfibrozil 28-39 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 20-26 25750174-5 2015 This study demonstrates that gemfibrozil, an agonist of peroxisome proliferator-activated receptor (PPAR) alpha, alone and in conjunction with all-trans-retinoic acid is capable of enhancing TFEB in brain cells. Gemfibrozil 29-40 peroxisome proliferator activated receptor alpha Homo sapiens 56-111 25750174-5 2015 This study demonstrates that gemfibrozil, an agonist of peroxisome proliferator-activated receptor (PPAR) alpha, alone and in conjunction with all-trans-retinoic acid is capable of enhancing TFEB in brain cells. Gemfibrozil 29-40 transcription factor EB Homo sapiens 191-195 25750174-6 2015 We also observed that PPARalpha, but not PPARbeta and PPARgamma, is involved in gemfibrozil-mediated up-regulation of TFEB. Gemfibrozil 80-91 peroxisome proliferator activated receptor alpha Homo sapiens 22-31 25750174-6 2015 We also observed that PPARalpha, but not PPARbeta and PPARgamma, is involved in gemfibrozil-mediated up-regulation of TFEB. Gemfibrozil 80-91 transcription factor EB Homo sapiens 118-122 25499606-7 2015 Our data show that the CoA esters of bezafibrate and gemfibrozil reduce chain elongation by specifically inhibiting ELOVL1. Gemfibrozil 53-64 ELOVL fatty acid elongase 1 Homo sapiens 116-122 25667670-6 2015 The LDH, AST and ALT values were significantly elevated following CLP compared with those in the sham group, and GFZ treatment was able to reduce these elevations. Gemfibrozil 113-116 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 9-12 25667670-7 2015 GFZ also reduced the sepsis-induced elevations of TNF-alpha and IL-1. Gemfibrozil 0-3 tumor necrosis factor Rattus norvegicus 50-59 25414411-4 2015 Effects of cyclosporine A (CsA), rifampin, and gemfibrozil on OATP1B1-mediated uptake of 12 substrate drugs were examined in OATP1B1-expressing human embryonic kidney 293 cells. Gemfibrozil 47-58 solute carrier organic anion transporter family member 1B1 Homo sapiens 62-69 25653502-4 2015 The increased uptake of catalposide via the OAT3, OATP1B1, and OATP1B3 transporters was decreased to basal levels in the presence of representative inhibitors such as probenecid, furosemide, and cimetidine (for OAT3) and cyclosporin A, gemfibrozil, and rifampin (for OATP1B1 and OATP1B3). Gemfibrozil 236-247 solute carrier family 22 member 8 Homo sapiens 44-48 25653502-4 2015 The increased uptake of catalposide via the OAT3, OATP1B1, and OATP1B3 transporters was decreased to basal levels in the presence of representative inhibitors such as probenecid, furosemide, and cimetidine (for OAT3) and cyclosporin A, gemfibrozil, and rifampin (for OATP1B1 and OATP1B3). Gemfibrozil 236-247 solute carrier organic anion transporter family member 1B3 Homo sapiens 63-70 25119429-6 2014 PPARalpha agonist Gemfibrozil improved the vacuolization but did not rescue CCR5(-/-) infected mice from high serum triglycerides levels and enhanced mortality. Gemfibrozil 18-29 peroxisome proliferator activated receptor alpha Mus musculus 0-9 25088042-7 2014 Oatp1d1 mediated the transport of diclofenac with very high affinity, followed by high affinity towards perfluorooctanesulfonic acid (PFOS), nonylphenol, gemfibrozil and 17alpha-ethinylestradiol; moderate affinity towards carbaryl, diazinon and caffeine; and low affinity towards metolachlor. Gemfibrozil 154-165 solute carrier organic anion transporter family, member 1D1 Danio rerio 0-7 23657159-0 2013 Gemfibrozil impairs imatinib absorption and inhibits the CYP2C8-mediated formation of its main metabolite. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 57-63 24306327-5 2014 Efflux in bacteria (NorA [S. aureus], Lde [L. monocytogenes]) and in macrophages (Mrp4) was assessed using the corresponding inhibitors reserpine and gemfibrozil, respectively. Gemfibrozil 150-161 prolactin family 2, subfamily c, member 5 Mus musculus 82-86 24385052-0 2014 Gemfibrozil disrupts lysophosphatidylcholine and bile acid homeostasis via PPARalpha and its relevance to hepatotoxicity. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 75-84 24385052-1 2014 Gemfibrozil, a ligand of peroxisome proliferator-activated receptor alpha (PPARalpha), is one of the most widely prescribed anti-dyslipidemia fibrate drugs. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 25-73 24385052-1 2014 Gemfibrozil, a ligand of peroxisome proliferator-activated receptor alpha (PPARalpha), is one of the most widely prescribed anti-dyslipidemia fibrate drugs. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 75-84 24385052-4 2014 In this study, wild-type and Ppara-null mice were dosed with a gemfibrozil-containing diet for 14 days. Gemfibrozil 63-74 peroxisome proliferator activated receptor alpha Mus musculus 29-34 24385052-10 2014 These data suggest that PPARalpha mediates gemfibrozil-induced hepatotoxicity in part by disrupting phospholipid and bile acid homeostasis. Gemfibrozil 43-54 peroxisome proliferator activated receptor alpha Mus musculus 24-33 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. Gemfibrozil 96-107 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 182-188 24271013-7 2014 Fenofibrate and gemfibrozil, previously reported to induce TPP1 activity in control cells, failed to increase TPP1 activity in patient iPSC-derived neural progenitor cells. Gemfibrozil 16-27 tripeptidyl peptidase 1 Homo sapiens 59-63 24491572-2 2014 OBJECTIVE: The goal of these studies was to investigate potential drug-drug interactions between empagliflozin and gemfibrozil (an organic anion-transporting polypeptide 1B1 [OATP1B1]/1B3 and organic anion transporter 3 [OAT3] inhibitor), rifampicin (an OATP1B1/1B3 inhibitor), or probenecid (an OAT3 and uridine diphosphate glucuronosyltransferase inhibitor). Gemfibrozil 115-126 solute carrier organic anion transporter family member 1B1 Homo sapiens 131-173 24491572-2 2014 OBJECTIVE: The goal of these studies was to investigate potential drug-drug interactions between empagliflozin and gemfibrozil (an organic anion-transporting polypeptide 1B1 [OATP1B1]/1B3 and organic anion transporter 3 [OAT3] inhibitor), rifampicin (an OATP1B1/1B3 inhibitor), or probenecid (an OAT3 and uridine diphosphate glucuronosyltransferase inhibitor). Gemfibrozil 115-126 solute carrier organic anion transporter family member 1B1 Homo sapiens 175-182 24491572-2 2014 OBJECTIVE: The goal of these studies was to investigate potential drug-drug interactions between empagliflozin and gemfibrozil (an organic anion-transporting polypeptide 1B1 [OATP1B1]/1B3 and organic anion transporter 3 [OAT3] inhibitor), rifampicin (an OATP1B1/1B3 inhibitor), or probenecid (an OAT3 and uridine diphosphate glucuronosyltransferase inhibitor). Gemfibrozil 115-126 solute carrier family 22 member 8 Homo sapiens 192-219 24491572-2 2014 OBJECTIVE: The goal of these studies was to investigate potential drug-drug interactions between empagliflozin and gemfibrozil (an organic anion-transporting polypeptide 1B1 [OATP1B1]/1B3 and organic anion transporter 3 [OAT3] inhibitor), rifampicin (an OATP1B1/1B3 inhibitor), or probenecid (an OAT3 and uridine diphosphate glucuronosyltransferase inhibitor). Gemfibrozil 115-126 solute carrier family 22 member 8 Homo sapiens 221-225 24491572-2 2014 OBJECTIVE: The goal of these studies was to investigate potential drug-drug interactions between empagliflozin and gemfibrozil (an organic anion-transporting polypeptide 1B1 [OATP1B1]/1B3 and organic anion transporter 3 [OAT3] inhibitor), rifampicin (an OATP1B1/1B3 inhibitor), or probenecid (an OAT3 and uridine diphosphate glucuronosyltransferase inhibitor). Gemfibrozil 115-126 solute carrier organic anion transporter family member 1B1 Homo sapiens 254-261 24491572-2 2014 OBJECTIVE: The goal of these studies was to investigate potential drug-drug interactions between empagliflozin and gemfibrozil (an organic anion-transporting polypeptide 1B1 [OATP1B1]/1B3 and organic anion transporter 3 [OAT3] inhibitor), rifampicin (an OATP1B1/1B3 inhibitor), or probenecid (an OAT3 and uridine diphosphate glucuronosyltransferase inhibitor). Gemfibrozil 115-126 solute carrier family 22 member 8 Homo sapiens 296-300 24180432-4 2014 Chemical-specific and mechanistic data support concordance of temporal and dose-response relationships for the key events associated with many PPARalpha activators including a phthalate ester plasticizer di(2-ethylhexyl) phthalate (DEHP) and the drug gemfibrozil. Gemfibrozil 251-262 peroxisome proliferator activated receptor alpha Homo sapiens 143-152 25147562-2 2014 The present study aimed at evaluating how 3 PPARalpha agonists, namely, fenofibrate, gemfibrozil, and Wy14,643, affect bilirubin synthesis and metabolism. Gemfibrozil 85-96 peroxisome proliferator activated receptor alpha Homo sapiens 44-53 23111552-0 2013 Gemfibrozil and its combination with metformin on pleiotropic effect on IL-10 and adiponectin and anti-atherogenic treatment in insulin resistant type 2 diabetes mellitus rats. Gemfibrozil 0-11 interleukin 10 Rattus norvegicus 72-77 23288702-6 2013 While the expression of NRF-1 and TFAM were induced in gemfibrozil-pretreated met-estrous females, they were suppressed in males. Gemfibrozil 55-66 transcription factor A, mitochondrial Rattus norvegicus 34-38 23288702-8 2013 In the mitogen-activated protein kinase (MAPKs) pathway, gemfibrozil pretreatment induced the expression of extracellular signal-regulated kinases (ERK1/2) in met-estrous females and reduced it in males. Gemfibrozil 57-68 mitogen activated protein kinase 3 Rattus norvegicus 148-154 24575330-5 2013 After polarization of MBP-primed Th1 cells to Th2 by gemfibrozil and other drugs, we observed that MBP-primed Th2 cells dose dependently inhibited the production of interleukin-1beta (IL-1beta) and nitric oxide (NO) in LPS-stimulated microglia via cell-to-cell contact. Gemfibrozil 53-64 myelin basic protein Homo sapiens 22-25 24575330-5 2013 After polarization of MBP-primed Th1 cells to Th2 by gemfibrozil and other drugs, we observed that MBP-primed Th2 cells dose dependently inhibited the production of interleukin-1beta (IL-1beta) and nitric oxide (NO) in LPS-stimulated microglia via cell-to-cell contact. Gemfibrozil 53-64 negative elongation factor complex member C/D Homo sapiens 33-36 24575330-5 2013 After polarization of MBP-primed Th1 cells to Th2 by gemfibrozil and other drugs, we observed that MBP-primed Th2 cells dose dependently inhibited the production of interleukin-1beta (IL-1beta) and nitric oxide (NO) in LPS-stimulated microglia via cell-to-cell contact. Gemfibrozil 53-64 myelin basic protein Homo sapiens 99-102 24575330-5 2013 After polarization of MBP-primed Th1 cells to Th2 by gemfibrozil and other drugs, we observed that MBP-primed Th2 cells dose dependently inhibited the production of interleukin-1beta (IL-1beta) and nitric oxide (NO) in LPS-stimulated microglia via cell-to-cell contact. Gemfibrozil 53-64 interleukin 1 beta Homo sapiens 165-182 23288702-2 2013 The ability of Gemfibrozil, a fibrate, is investigated for the first time to modulate mitochondrial pro-survival factors involved in the mitochondrial biogenesis signaling pathway, including peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha), nuclear respiratory factor (NRF-1), and mitochondrial transcription factor A (TFAM) in the brain. Gemfibrozil 15-26 PPARG coactivator 1 alpha Rattus norvegicus 254-264 23288702-2 2013 The ability of Gemfibrozil, a fibrate, is investigated for the first time to modulate mitochondrial pro-survival factors involved in the mitochondrial biogenesis signaling pathway, including peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha), nuclear respiratory factor (NRF-1), and mitochondrial transcription factor A (TFAM) in the brain. Gemfibrozil 15-26 nuclear respiratory factor 1 Rattus norvegicus 295-300 23288702-2 2013 The ability of Gemfibrozil, a fibrate, is investigated for the first time to modulate mitochondrial pro-survival factors involved in the mitochondrial biogenesis signaling pathway, including peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha), nuclear respiratory factor (NRF-1), and mitochondrial transcription factor A (TFAM) in the brain. Gemfibrozil 15-26 transcription factor A, mitochondrial Rattus norvegicus 345-349 23288702-6 2013 While the expression of NRF-1 and TFAM were induced in gemfibrozil-pretreated met-estrous females, they were suppressed in males. Gemfibrozil 55-66 nuclear respiratory factor 1 Rattus norvegicus 24-29 23805127-6 2013 Inhibition assays using human liver microsomes (HLM) incubated with 19-norandrosterone and selective inhibitors confirmed that UGT2B7 and UGT2B15 are involved in 19-norandrosterone glucuronidation, since the presence of the specific UGT2B7 and UGT2B15 inhibitors gemfibrozil and valproic acid inhibited the 19-norandrosterone glucuronidation by 35 and 45%, respectively. Gemfibrozil 263-274 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 127-133 23805127-6 2013 Inhibition assays using human liver microsomes (HLM) incubated with 19-norandrosterone and selective inhibitors confirmed that UGT2B7 and UGT2B15 are involved in 19-norandrosterone glucuronidation, since the presence of the specific UGT2B7 and UGT2B15 inhibitors gemfibrozil and valproic acid inhibited the 19-norandrosterone glucuronidation by 35 and 45%, respectively. Gemfibrozil 263-274 UDP glucuronosyltransferase family 2 member B15 Homo sapiens 138-145 23111552-0 2013 Gemfibrozil and its combination with metformin on pleiotropic effect on IL-10 and adiponectin and anti-atherogenic treatment in insulin resistant type 2 diabetes mellitus rats. Gemfibrozil 0-11 adiponectin, C1Q and collagen domain containing Rattus norvegicus 82-93 23111552-1 2013 AIM: Gemfibrozil is a PPAR-alpha ligand that inhibits the progression of atherosclerosis in insulin resistance type 2 diabetes mellitus (IR type 2 DM). Gemfibrozil 5-16 peroxisome proliferator activated receptor alpha Rattus norvegicus 22-32 23111552-8 2013 OVERALL CONCLUSIONS: Gemfibrozil plus metformin decrease MMP-9, increase IL-10 and adiponectin acting as anti-atherogenic, anti-inflammatory and immunomodulatory in IR type 2 DM. Gemfibrozil 21-32 matrix metallopeptidase 9 Rattus norvegicus 57-62 23111552-8 2013 OVERALL CONCLUSIONS: Gemfibrozil plus metformin decrease MMP-9, increase IL-10 and adiponectin acting as anti-atherogenic, anti-inflammatory and immunomodulatory in IR type 2 DM. Gemfibrozil 21-32 interleukin 10 Rattus norvegicus 73-78 23111552-8 2013 OVERALL CONCLUSIONS: Gemfibrozil plus metformin decrease MMP-9, increase IL-10 and adiponectin acting as anti-atherogenic, anti-inflammatory and immunomodulatory in IR type 2 DM. Gemfibrozil 21-32 adiponectin, C1Q and collagen domain containing Rattus norvegicus 83-94 22625877-1 2013 AIM: The objective of this study was to determine the extent to which the CYP2C8*3 allele influences pharmacokinetic variability in the drug-drug interaction between gemfibrozil (CYP2C8 inhibitor) and pioglitazone (CYP2C8 substrate). Gemfibrozil 166-177 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 74-80 22773136-10 2013 In contrast, gemfibrozil pretreatment was toxic to males, enhancing the expression of inflammatory factors such as tumor necrosis factor-alpha, nuclear factor-kappaB, and cyclooxygenase-2, and decreasing Nrf-2 expression and SOD activity, leading to hippocampal neurodegeneration. Gemfibrozil 13-24 tumor necrosis factor Rattus norvegicus 115-142 22773136-10 2013 In contrast, gemfibrozil pretreatment was toxic to males, enhancing the expression of inflammatory factors such as tumor necrosis factor-alpha, nuclear factor-kappaB, and cyclooxygenase-2, and decreasing Nrf-2 expression and SOD activity, leading to hippocampal neurodegeneration. Gemfibrozil 13-24 prostaglandin-endoperoxide synthase 2 Rattus norvegicus 171-187 22773136-10 2013 In contrast, gemfibrozil pretreatment was toxic to males, enhancing the expression of inflammatory factors such as tumor necrosis factor-alpha, nuclear factor-kappaB, and cyclooxygenase-2, and decreasing Nrf-2 expression and SOD activity, leading to hippocampal neurodegeneration. Gemfibrozil 13-24 NFE2 like bZIP transcription factor 2 Rattus norvegicus 204-209 22625877-0 2013 Impact of the CYP2C8 *3 polymorphism on the drug-drug interaction between gemfibrozil and pioglitazone. Gemfibrozil 74-85 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 14-20 22625877-1 2013 AIM: The objective of this study was to determine the extent to which the CYP2C8*3 allele influences pharmacokinetic variability in the drug-drug interaction between gemfibrozil (CYP2C8 inhibitor) and pioglitazone (CYP2C8 substrate). Gemfibrozil 166-177 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 179-185 22625877-1 2013 AIM: The objective of this study was to determine the extent to which the CYP2C8*3 allele influences pharmacokinetic variability in the drug-drug interaction between gemfibrozil (CYP2C8 inhibitor) and pioglitazone (CYP2C8 substrate). Gemfibrozil 166-177 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 179-185 22625877-7 2013 The relative change in pioglitazone plasma exposure following gemfibrozil administration was significantly influenced by CYP2C8 genotype. Gemfibrozil 62-73 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 121-127 22625877-9 2013 CONCLUSION: CYP2C8*3 is associated with decreased pioglitazone plasma exposure in vivo and significantly influences the pharmacokinetic magnitude of the gemfibrozil-pioglitazone drug-drug interaction. Gemfibrozil 153-164 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 12-18 22528839-4 2012 This study underlines the importance of PPARbeta/delta in mediating the anti-inflammatory effect of gemfibrozil, an FDA-approved lipid-lowering drug, in primary human microglia. Gemfibrozil 100-111 peroxisome proliferator activated receptor delta Homo sapiens 40-48 22989886-0 2012 Gemfibrozil and fenofibrate, Food and Drug Administration-approved lipid-lowering drugs, up-regulate tripeptidyl-peptidase 1 in brain cells via peroxisome proliferator-activated receptor alpha: implications for late infantile Batten disease therapy. Gemfibrozil 0-11 tripeptidyl peptidase I Mus musculus 101-124 22989886-0 2012 Gemfibrozil and fenofibrate, Food and Drug Administration-approved lipid-lowering drugs, up-regulate tripeptidyl-peptidase 1 in brain cells via peroxisome proliferator-activated receptor alpha: implications for late infantile Batten disease therapy. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 144-192 22989886-4 2012 This study reveals a novel use of gemfibrozil and fenofibrate, Food and Drug Administration-approved lipid-lowering drugs, in up-regulating TPP1 in brain cells. Gemfibrozil 34-45 tripeptidyl peptidase I Mus musculus 140-144 22989886-5 2012 Both gemfibrozil and fenofibrate up-regulated mRNA, protein, and enzymatic activity of TPP1 in primary mouse neurons and astrocytes as well as human astrocytes and neuronal cells. Gemfibrozil 5-16 tripeptidyl peptidase I Mus musculus 87-91 22989886-6 2012 Because gemfibrozil and fenofibrate are known to activate peroxisome proliferator-activated receptor-alpha (PPARalpha), the role of PPARalpha in gemfibrozil- and fenofibrate-mediated up-regulation of TPP1 was investigated revealing that both drugs up-regulated TPP1 mRNA, protein, and enzymatic activity both in vitro and in vivo in wild type (WT) and PPARbeta(-/-), but not PPARalpha(-/-), mice. Gemfibrozil 8-19 peroxisome proliferator activated receptor alpha Mus musculus 58-106 22989886-6 2012 Because gemfibrozil and fenofibrate are known to activate peroxisome proliferator-activated receptor-alpha (PPARalpha), the role of PPARalpha in gemfibrozil- and fenofibrate-mediated up-regulation of TPP1 was investigated revealing that both drugs up-regulated TPP1 mRNA, protein, and enzymatic activity both in vitro and in vivo in wild type (WT) and PPARbeta(-/-), but not PPARalpha(-/-), mice. Gemfibrozil 8-19 peroxisome proliferator activated receptor alpha Mus musculus 108-117 22989886-6 2012 Because gemfibrozil and fenofibrate are known to activate peroxisome proliferator-activated receptor-alpha (PPARalpha), the role of PPARalpha in gemfibrozil- and fenofibrate-mediated up-regulation of TPP1 was investigated revealing that both drugs up-regulated TPP1 mRNA, protein, and enzymatic activity both in vitro and in vivo in wild type (WT) and PPARbeta(-/-), but not PPARalpha(-/-), mice. Gemfibrozil 145-156 peroxisome proliferator activated receptor alpha Mus musculus 132-141 22989886-6 2012 Because gemfibrozil and fenofibrate are known to activate peroxisome proliferator-activated receptor-alpha (PPARalpha), the role of PPARalpha in gemfibrozil- and fenofibrate-mediated up-regulation of TPP1 was investigated revealing that both drugs up-regulated TPP1 mRNA, protein, and enzymatic activity both in vitro and in vivo in wild type (WT) and PPARbeta(-/-), but not PPARalpha(-/-), mice. Gemfibrozil 145-156 peroxisome proliferator activated receptor alpha Mus musculus 132-141 22989886-8 2012 Accordingly, all-trans-retinoic acid, alone or together with gemfibrozil, up-regulated TPP1. Gemfibrozil 61-72 tripeptidyl peptidase I Mus musculus 87-91 22879602-0 2012 Gemfibrozil, a lipid-lowering drug, increases myelin genes in human oligodendrocytes via peroxisome proliferator-activated receptor-beta. Gemfibrozil 0-11 peroxisome proliferator activated receptor delta Homo sapiens 89-136 22879602-3 2012 The current study identified another novel property of gemfibrozil in stimulating the expression of myelin-specific genes (myelin basic protein, myelin oligodendrocyte glycoprotein, 2",3"-cyclic-nucleotide 3"-phosphodiesterase, and proteolipid protein (PLP)) in primary human oligodendrocytes, mixed glial cells, and spinal cord organotypic cultures. Gemfibrozil 55-66 myelin basic protein Homo sapiens 123-143 22879602-3 2012 The current study identified another novel property of gemfibrozil in stimulating the expression of myelin-specific genes (myelin basic protein, myelin oligodendrocyte glycoprotein, 2",3"-cyclic-nucleotide 3"-phosphodiesterase, and proteolipid protein (PLP)) in primary human oligodendrocytes, mixed glial cells, and spinal cord organotypic cultures. Gemfibrozil 55-66 myelin oligodendrocyte glycoprotein Homo sapiens 145-180 22879602-3 2012 The current study identified another novel property of gemfibrozil in stimulating the expression of myelin-specific genes (myelin basic protein, myelin oligodendrocyte glycoprotein, 2",3"-cyclic-nucleotide 3"-phosphodiesterase, and proteolipid protein (PLP)) in primary human oligodendrocytes, mixed glial cells, and spinal cord organotypic cultures. Gemfibrozil 55-66 proteolipid protein 1 Homo sapiens 232-257 22879602-4 2012 Although gemfibrozil is a known activator of peroxisome proliferator-activated receptor-alpha (PPAR-alpha), we were unable to detect PPAR-alpha in either gemfibrozil-treated or untreated human oligodendrocytes, and gemfibrozil increased the expression of myelin genes in oligodendrocytes isolated from both wild type and PPAR-alpha(-/-) mice. Gemfibrozil 9-20 peroxisome proliferator activated receptor alpha Homo sapiens 45-93 22879602-4 2012 Although gemfibrozil is a known activator of peroxisome proliferator-activated receptor-alpha (PPAR-alpha), we were unable to detect PPAR-alpha in either gemfibrozil-treated or untreated human oligodendrocytes, and gemfibrozil increased the expression of myelin genes in oligodendrocytes isolated from both wild type and PPAR-alpha(-/-) mice. Gemfibrozil 9-20 peroxisome proliferator activated receptor alpha Homo sapiens 95-105 22879602-5 2012 On the other hand, gemfibrozil markedly increased the expression of PPAR-beta but not PPAR-gamma. Gemfibrozil 19-30 peroxisome proliferator activated receptor delta Homo sapiens 68-77 22879602-6 2012 Consistently, antisense knockdown of PPAR-beta, but not PPAR-gamma, abrogated the stimulatory effect of gemfibrozil on myelin genes in human oligodendrocytes. Gemfibrozil 104-115 peroxisome proliferator activated receptor delta Homo sapiens 37-46 22879602-8 2012 Chromatin immunoprecipitation analysis showed that gemfibrozil induced the recruitment of PPAR-beta to the promoter of PLP and myelin oligodendrocyte glycoprotein genes in human oligodendrocytes. Gemfibrozil 51-62 peroxisome proliferator activated receptor delta Homo sapiens 90-99 22879602-8 2012 Chromatin immunoprecipitation analysis showed that gemfibrozil induced the recruitment of PPAR-beta to the promoter of PLP and myelin oligodendrocyte glycoprotein genes in human oligodendrocytes. Gemfibrozil 51-62 proteolipid protein 1 Homo sapiens 119-122 22879602-8 2012 Chromatin immunoprecipitation analysis showed that gemfibrozil induced the recruitment of PPAR-beta to the promoter of PLP and myelin oligodendrocyte glycoprotein genes in human oligodendrocytes. Gemfibrozil 51-62 myelin oligodendrocyte glycoprotein Homo sapiens 127-162 22879602-9 2012 Furthermore, gemfibrozil treatment also led to the recruitment of PPAR-beta to the PLP promoter in vivo in the spinal cord of experimental autoimmune encephalomyelitis mice and suppression of experimental autoimmune encephalomyelitis symptoms in PLP-T cell receptor transgenic mice. Gemfibrozil 13-24 peroxisome proliferator activator receptor delta Mus musculus 66-75 22879602-9 2012 Furthermore, gemfibrozil treatment also led to the recruitment of PPAR-beta to the PLP promoter in vivo in the spinal cord of experimental autoimmune encephalomyelitis mice and suppression of experimental autoimmune encephalomyelitis symptoms in PLP-T cell receptor transgenic mice. Gemfibrozil 13-24 proteolipid protein (myelin) 1 Mus musculus 83-86 22879602-9 2012 Furthermore, gemfibrozil treatment also led to the recruitment of PPAR-beta to the PLP promoter in vivo in the spinal cord of experimental autoimmune encephalomyelitis mice and suppression of experimental autoimmune encephalomyelitis symptoms in PLP-T cell receptor transgenic mice. Gemfibrozil 13-24 proteolipid protein (myelin) 1 Mus musculus 246-249 22879602-10 2012 These results suggest that gemfibrozil stimulates the expression of myelin genes via PPAR-beta and that gemfibrozil, a prescribed drug for humans, may find further therapeutic use in demyelinating diseases. Gemfibrozil 27-38 peroxisome proliferator activated receptor delta Homo sapiens 85-94 22538052-8 2012 Rifamycin SV strongly, and gemfibrozil weakly, inhibited the OATP1B1-mediated transport of substrates. Gemfibrozil 27-38 solute carrier organic anion transporter family member 1B1 Homo sapiens 61-68 22685291-0 2012 Gemfibrozil, a lipid-lowering drug, induces suppressor of cytokine signaling 3 in glial cells: implications for neurodegenerative disorders. Gemfibrozil 0-11 suppressor of cytokine signaling 3 Mus musculus 44-78 22685291-4 2012 This study underlines the importance of gemfibrozil, a Food and Drug Administration-approved lipid-lowering drug, in up-regulating the expression of SOCS3 in glial cells. Gemfibrozil 40-51 suppressor of cytokine signaling 3 Mus musculus 149-154 22685291-5 2012 Gemfibrozil increased the expression of Socs3 mRNA and protein in mouse astroglia and microglia in both a time- and dose-dependent manner. Gemfibrozil 0-11 suppressor of cytokine signaling 3 Mus musculus 40-45 22685291-6 2012 Interestingly, gemfibrozil induced the activation of type IA phosphatidylinositol (PI) 3-kinase and AKT. Gemfibrozil 15-26 thymoma viral proto-oncogene 1 Mus musculus 100-103 22685291-7 2012 Accordingly, inhibition of PI 3-kinase and AKT by chemical inhibitors abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 80-91 thymoma viral proto-oncogene 1 Mus musculus 43-46 22685291-7 2012 Accordingly, inhibition of PI 3-kinase and AKT by chemical inhibitors abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 80-91 suppressor of cytokine signaling 3 Mus musculus 118-123 22685291-8 2012 Furthermore, we demonstrated that gemfibrozil induced the activation of Kruppel-like factor 4 (KLF4) via the PI 3-kinase-AKT pathway and that siRNA knockdown of KLF4 abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 34-45 Kruppel-like factor 4 (gut) Mus musculus 72-93 22685291-8 2012 Furthermore, we demonstrated that gemfibrozil induced the activation of Kruppel-like factor 4 (KLF4) via the PI 3-kinase-AKT pathway and that siRNA knockdown of KLF4 abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 34-45 Kruppel-like factor 4 (gut) Mus musculus 95-99 22685291-8 2012 Furthermore, we demonstrated that gemfibrozil induced the activation of Kruppel-like factor 4 (KLF4) via the PI 3-kinase-AKT pathway and that siRNA knockdown of KLF4 abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 34-45 thymoma viral proto-oncogene 1 Mus musculus 121-124 22685291-8 2012 Furthermore, we demonstrated that gemfibrozil induced the activation of Kruppel-like factor 4 (KLF4) via the PI 3-kinase-AKT pathway and that siRNA knockdown of KLF4 abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 34-45 Kruppel-like factor 4 (gut) Mus musculus 161-165 22685291-8 2012 Furthermore, we demonstrated that gemfibrozil induced the activation of Kruppel-like factor 4 (KLF4) via the PI 3-kinase-AKT pathway and that siRNA knockdown of KLF4 abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 34-45 suppressor of cytokine signaling 3 Mus musculus 214-219 22685291-8 2012 Furthermore, we demonstrated that gemfibrozil induced the activation of Kruppel-like factor 4 (KLF4) via the PI 3-kinase-AKT pathway and that siRNA knockdown of KLF4 abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 176-187 Kruppel-like factor 4 (gut) Mus musculus 72-93 22685291-8 2012 Furthermore, we demonstrated that gemfibrozil induced the activation of Kruppel-like factor 4 (KLF4) via the PI 3-kinase-AKT pathway and that siRNA knockdown of KLF4 abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 176-187 Kruppel-like factor 4 (gut) Mus musculus 95-99 22685291-8 2012 Furthermore, we demonstrated that gemfibrozil induced the activation of Kruppel-like factor 4 (KLF4) via the PI 3-kinase-AKT pathway and that siRNA knockdown of KLF4 abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 176-187 Kruppel-like factor 4 (gut) Mus musculus 161-165 22685291-8 2012 Furthermore, we demonstrated that gemfibrozil induced the activation of Kruppel-like factor 4 (KLF4) via the PI 3-kinase-AKT pathway and that siRNA knockdown of KLF4 abrogated gemfibrozil-mediated up-regulation of SOCS3. Gemfibrozil 176-187 suppressor of cytokine signaling 3 Mus musculus 214-219 22685291-9 2012 Gemfibrozil also induced the recruitment of KLF4 to the distal, but not proximal, KLF4-binding site of the Socs3 promoter. Gemfibrozil 0-11 Kruppel-like factor 4 (gut) Mus musculus 44-48 22685291-9 2012 Gemfibrozil also induced the recruitment of KLF4 to the distal, but not proximal, KLF4-binding site of the Socs3 promoter. Gemfibrozil 0-11 Kruppel-like factor 4 (gut) Mus musculus 82-86 22685291-9 2012 Gemfibrozil also induced the recruitment of KLF4 to the distal, but not proximal, KLF4-binding site of the Socs3 promoter. Gemfibrozil 0-11 suppressor of cytokine signaling 3 Mus musculus 107-112 22685291-10 2012 This study delineates a novel property of gemfibrozil in up-regulating SOCS3 in glial cells via PI 3-kinase-AKT-mediated activation of KLF4 and suggests that gemfibrozil may find therapeutic application in neuroinflammatory and neurodegenerative disorders. Gemfibrozil 42-53 suppressor of cytokine signaling 3 Mus musculus 71-76 22685291-10 2012 This study delineates a novel property of gemfibrozil in up-regulating SOCS3 in glial cells via PI 3-kinase-AKT-mediated activation of KLF4 and suggests that gemfibrozil may find therapeutic application in neuroinflammatory and neurodegenerative disorders. Gemfibrozil 42-53 thymoma viral proto-oncogene 1 Mus musculus 108-111 22685291-10 2012 This study delineates a novel property of gemfibrozil in up-regulating SOCS3 in glial cells via PI 3-kinase-AKT-mediated activation of KLF4 and suggests that gemfibrozil may find therapeutic application in neuroinflammatory and neurodegenerative disorders. Gemfibrozil 42-53 Kruppel-like factor 4 (gut) Mus musculus 135-139 22685291-10 2012 This study delineates a novel property of gemfibrozil in up-regulating SOCS3 in glial cells via PI 3-kinase-AKT-mediated activation of KLF4 and suggests that gemfibrozil may find therapeutic application in neuroinflammatory and neurodegenerative disorders. Gemfibrozil 158-169 suppressor of cytokine signaling 3 Mus musculus 71-76 22685291-10 2012 This study delineates a novel property of gemfibrozil in up-regulating SOCS3 in glial cells via PI 3-kinase-AKT-mediated activation of KLF4 and suggests that gemfibrozil may find therapeutic application in neuroinflammatory and neurodegenerative disorders. Gemfibrozil 158-169 thymoma viral proto-oncogene 1 Mus musculus 108-111 22685291-10 2012 This study delineates a novel property of gemfibrozil in up-regulating SOCS3 in glial cells via PI 3-kinase-AKT-mediated activation of KLF4 and suggests that gemfibrozil may find therapeutic application in neuroinflammatory and neurodegenerative disorders. Gemfibrozil 158-169 Kruppel-like factor 4 (gut) Mus musculus 135-139 22538270-4 2012 The aim of the present study was to investigate the effect of gemfibrozil (CYP2C9 inhibitor) and its 1-O-beta-glucuronide (CYP2C8 inhibitor) on BRV disposition both in vivo (healthy participants) and in vitro (human liver microsomes and hepatocytes). Gemfibrozil 62-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22538270-8 2012 In human hepatocytes and/or liver microsomes, gemfibrozil potently inhibited the hydroxylation of BRV-AC into BRV-OHAC (K(I) 12 muM) while having a marginal effect on BRV-OH formation (K(I) >=153 muM). Gemfibrozil 46-57 latexin Homo sapiens 128-131 22538270-8 2012 In human hepatocytes and/or liver microsomes, gemfibrozil potently inhibited the hydroxylation of BRV-AC into BRV-OHAC (K(I) 12 muM) while having a marginal effect on BRV-OH formation (K(I) >=153 muM). Gemfibrozil 46-57 latexin Homo sapiens 199-202 22528839-8 2012 Interestingly, either antisense knockdown of PPAR-beta or antagonism of PPAR-beta by a specific chemical antagonist abrogated gemfibrozil-mediated inhibition of microglial activation. Gemfibrozil 126-137 peroxisome proliferator activated receptor delta Homo sapiens 45-54 22528839-8 2012 Interestingly, either antisense knockdown of PPAR-beta or antagonism of PPAR-beta by a specific chemical antagonist abrogated gemfibrozil-mediated inhibition of microglial activation. Gemfibrozil 126-137 peroxisome proliferator activated receptor delta Homo sapiens 72-81 22528839-10 2012 These results highlight the fact that gemfibrozil regulates microglial activation by inhibiting inflammatory gene expression in a PPAR-beta dependent pathway and further reinforce its therapeutic application in several neuroinflammatory and neurodegenerative diseases. Gemfibrozil 38-49 peroxisome proliferator activated receptor delta Homo sapiens 130-139 23153186-8 2012 The strong CYP2C8 and OATP1B1 inhibitor gemfibrozil increases substantially the plasma concentrations of repaglinide and TZDs. Gemfibrozil 40-51 solute carrier organic anion transporter family member 1B1 Homo sapiens 22-29 22528839-6 2012 However, gemfibrozil markedly suppressed proinflammatory molecules and CD11b in LPS-stimulated microglia. Gemfibrozil 9-20 integrin subunit alpha M Homo sapiens 71-76 22706077-4 2012 The present study underlines a novel application of gemfibrozil (gem), a Food and Drug Administration-approved lipid-lowering drug, in increasing the expression of IL-1Ra in primary mouse and human neurons. Gemfibrozil 52-63 interleukin 1 receptor antagonist Mus musculus 164-170 22706077-4 2012 The present study underlines a novel application of gemfibrozil (gem), a Food and Drug Administration-approved lipid-lowering drug, in increasing the expression of IL-1Ra in primary mouse and human neurons. Gemfibrozil 52-55 interleukin 1 receptor antagonist Mus musculus 164-170 22706077-10 2012 Taken together, these results highlight the importance of the PI3K-Akt-CREB pathway in mediating gem-induced upregulation of IL-1Ra in neurons and suggest gem as a possible therapeutic treatment for propagating neuronal self-defense in neuroinflammatory and neurodegenerative disorders. Gemfibrozil 97-100 cAMP responsive element binding protein 1 Mus musculus 71-75 22706077-6 2012 Activation of type IA p110alpha PI3K and Akt by gem and abrogation of gem-induced upregulation of IL-1Ra by inhibitors of PI3K and Akt indicate a role of the PI3K-Akt pathway in the upregulation of IL-1Ra. Gemfibrozil 48-51 thymoma viral proto-oncogene 1 Mus musculus 41-44 22706077-10 2012 Taken together, these results highlight the importance of the PI3K-Akt-CREB pathway in mediating gem-induced upregulation of IL-1Ra in neurons and suggest gem as a possible therapeutic treatment for propagating neuronal self-defense in neuroinflammatory and neurodegenerative disorders. Gemfibrozil 97-100 interleukin 1 receptor antagonist Mus musculus 125-131 22706077-6 2012 Activation of type IA p110alpha PI3K and Akt by gem and abrogation of gem-induced upregulation of IL-1Ra by inhibitors of PI3K and Akt indicate a role of the PI3K-Akt pathway in the upregulation of IL-1Ra. Gemfibrozil 70-73 interleukin 1 receptor antagonist Mus musculus 98-104 22706077-6 2012 Activation of type IA p110alpha PI3K and Akt by gem and abrogation of gem-induced upregulation of IL-1Ra by inhibitors of PI3K and Akt indicate a role of the PI3K-Akt pathway in the upregulation of IL-1Ra. Gemfibrozil 70-73 thymoma viral proto-oncogene 1 Mus musculus 131-134 22706077-6 2012 Activation of type IA p110alpha PI3K and Akt by gem and abrogation of gem-induced upregulation of IL-1Ra by inhibitors of PI3K and Akt indicate a role of the PI3K-Akt pathway in the upregulation of IL-1Ra. Gemfibrozil 70-73 thymoma viral proto-oncogene 1 Mus musculus 131-134 22706077-6 2012 Activation of type IA p110alpha PI3K and Akt by gem and abrogation of gem-induced upregulation of IL-1Ra by inhibitors of PI3K and Akt indicate a role of the PI3K-Akt pathway in the upregulation of IL-1Ra. Gemfibrozil 70-73 interleukin 1 receptor antagonist Mus musculus 198-204 22706077-7 2012 Gem also induced the activation of CREB via the PI3K-Akt pathway, and small interfering RNA attenuation of CREB abolished the gem-mediated increase in IL-1Ra. Gemfibrozil 126-129 cAMP responsive element binding protein 1 Mus musculus 35-39 22706077-7 2012 Gem also induced the activation of CREB via the PI3K-Akt pathway, and small interfering RNA attenuation of CREB abolished the gem-mediated increase in IL-1Ra. Gemfibrozil 126-129 cAMP responsive element binding protein 1 Mus musculus 107-111 22706077-7 2012 Gem also induced the activation of CREB via the PI3K-Akt pathway, and small interfering RNA attenuation of CREB abolished the gem-mediated increase in IL-1Ra. Gemfibrozil 126-129 interleukin 1 receptor antagonist Mus musculus 151-157 22706077-8 2012 Furthermore, gem was able to protect neurons from IL-1beta insult. Gemfibrozil 13-16 interleukin 1 alpha Mus musculus 50-58 22706077-9 2012 However, small interfering RNA knockdown of neuronal IL-1Ra abrogated the protective effect of gem against IL-1beta, suggesting that this drug increases the defense mechanism of cortical neurons via upregulation of IL-1Ra. Gemfibrozil 95-98 interleukin 1 receptor antagonist Mus musculus 53-59 22706077-9 2012 However, small interfering RNA knockdown of neuronal IL-1Ra abrogated the protective effect of gem against IL-1beta, suggesting that this drug increases the defense mechanism of cortical neurons via upregulation of IL-1Ra. Gemfibrozil 95-98 interleukin 1 alpha Mus musculus 107-115 22706077-9 2012 However, small interfering RNA knockdown of neuronal IL-1Ra abrogated the protective effect of gem against IL-1beta, suggesting that this drug increases the defense mechanism of cortical neurons via upregulation of IL-1Ra. Gemfibrozil 95-98 interleukin 1 receptor antagonist Mus musculus 215-221 21838784-5 2012 Plasma concentrations of montelukast, gemfibrozil, itraconazole and their metabolites were measured up to 72 h. RESULTS: The CYP2C8 inhibitor gemfibrozil increased the AUC(0, ) of montelukast 4.3-fold and its t(1/2) 2.1-fold (P < 0.001). Gemfibrozil 38-49 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 125-131 21838784-5 2012 Plasma concentrations of montelukast, gemfibrozil, itraconazole and their metabolites were measured up to 72 h. RESULTS: The CYP2C8 inhibitor gemfibrozil increased the AUC(0, ) of montelukast 4.3-fold and its t(1/2) 2.1-fold (P < 0.001). Gemfibrozil 142-153 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 125-131 21917557-10 2011 The adjusted IRR for a statin and fenofibrate was 3.26 (95% CI 1.21 to 8.80), while the adjusted IRR for a statin and gemfibrozil was 11.93 (95% CI 3.96 to 35.93) versus statin therapy alone. Gemfibrozil 118-129 insulin receptor related receptor Homo sapiens 97-100 21903140-6 2012 The importance of TG in producing these changes was supported by the finding that the TG-lowering medication gemfibrozil as compared to vehicle, when peripherally administered before consumption of a HiSat meal, significantly decreased the expression of OX, while increasing the expression of NPY and AgRP. Gemfibrozil 109-120 hypocretin neuropeptide precursor Rattus norvegicus 254-256 21903140-6 2012 The importance of TG in producing these changes was supported by the finding that the TG-lowering medication gemfibrozil as compared to vehicle, when peripherally administered before consumption of a HiSat meal, significantly decreased the expression of OX, while increasing the expression of NPY and AgRP. Gemfibrozil 109-120 neuropeptide Y Rattus norvegicus 293-296 21903140-6 2012 The importance of TG in producing these changes was supported by the finding that the TG-lowering medication gemfibrozil as compared to vehicle, when peripherally administered before consumption of a HiSat meal, significantly decreased the expression of OX, while increasing the expression of NPY and AgRP. Gemfibrozil 109-120 agouti related neuropeptide Rattus norvegicus 301-305 22028316-6 2012 Because the haloperidol O-glucuronidation in a panel of 17 HLM was significantly correlated (r = 0.732, p < 0.01) with zidovudine O-glucuronidation, a probe activity of UGT2B7, and the activity in the pooled HLM was prominently inhibited (58% of control) by gemfibrozil, an inhibitor of UGT2B7, we surmised that the reaction would mainly be catalyzed by UGT2B7. Gemfibrozil 261-272 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 172-178 21911547-0 2011 Studies to further investigate the inhibition of human liver microsomal CYP2C8 by the acyl-beta-glucuronide of gemfibrozil. Gemfibrozil 111-122 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 72-78 21911547-1 2011 In previous studies, gemfibrozil acyl-beta-glucuronide, but not gemfibrozil, was found to be a mechanism-based inhibitor of cytochrome P450 2C8. Gemfibrozil 21-32 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 124-143 21911547-5 2011 When the aromatic methyl groups on the gemfibrozil moiety were substituted with trifluoromethyls, the resulting glucuronide conjugate was a weaker inhibitor of CYP2C8 and mechanism-based inhibition was abolished. Gemfibrozil 39-50 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 160-166 21963672-0 2011 The PPAR alpha agonist gemfibrozil is an ineffective treatment for spinal cord injured mice. Gemfibrozil 23-34 peroxisome proliferator activated receptor alpha Mus musculus 4-14 21963672-2 2011 Gemfibrozil, an agonist of PPAR-alpha, is a FDA approved drug for hyperlipidemia and has been shown to inhibit clinical signs in a rodent model of multiple sclerosis. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 27-37 21881934-6 2011 Gemfibrozil was discontinued and in a few days AST and ALT levels returned to normal. Gemfibrozil 0-11 solute carrier family 17 member 5 Homo sapiens 47-50 21889235-0 2011 Discovery of gemfibrozil analogues that activate PPARalpha and enhance the expression of gene CPT1A involved in fatty acids catabolism. Gemfibrozil 13-24 peroxisome proliferator activated receptor alpha Homo sapiens 49-58 21889235-0 2011 Discovery of gemfibrozil analogues that activate PPARalpha and enhance the expression of gene CPT1A involved in fatty acids catabolism. Gemfibrozil 13-24 carnitine palmitoyltransferase 1A Homo sapiens 94-99 21889235-1 2011 A new series of gemfibrozil analogues conjugated with alpha-asarone, trans-stilbene, chalcone, and their bioisosteric modifications were synthesized and evaluated to develop PPARalpha agonists. Gemfibrozil 16-27 peroxisome proliferator activated receptor alpha Homo sapiens 174-183 21778352-0 2011 Dose-dependent interaction between gemfibrozil and repaglinide in humans: strong inhibition of CYP2C8 with subtherapeutic gemfibrozil doses. Gemfibrozil 122-133 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 95-101 21778352-2 2011 We investigated the effect of gemfibrozil dose on CYP2C8 activity in humans using repaglinide as a probe drug. Gemfibrozil 30-41 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 50-56 21778352-7 2011 The gemfibrozil-repaglinide interaction could be mainly explained by gemfibrozil 1-O-beta-glucuronide concentration-dependent, mechanism-based inhibition of CYP2C8, with a minor contribution by competitive inhibition of organic anion-transporting polypeptide 1B1 at the highest gemfibrozil dose. Gemfibrozil 4-15 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 157-163 21778352-8 2011 The findings are consistent with ~50% inhibition of CYP2C8 already with a single 30-mg dose of gemfibrozil and >95% inhibition with 900 mg. Gemfibrozil 95-106 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 52-58 21778352-9 2011 In clinical drug-drug interaction studies, a single 900-mg dose of gemfibrozil can be used to achieve nearly complete inactivation of CYP2C8. Gemfibrozil 67-78 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 134-140 21368757-0 2011 Mechanism-based inactivation of CYP2C8 by gemfibrozil occurs rapidly in humans. Gemfibrozil 42-53 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 32-38 21652050-5 2011 Gemfibrozil exposure induced biomarkers of stress (glutathione S-transferase and metallothionein) at both concentrations 24h and 96 h after exposure, respectively. Gemfibrozil 0-11 glutathione S-transferase kappa 1 Homo sapiens 51-76 21297316-6 2011 Gemfibrozil displays DDIs with some OATP1B1 substrates, although their extent is small. Gemfibrozil 0-11 solute carrier organic anion transporter family member 1B1 Homo sapiens 36-43 21223511-3 2011 Because serum paraoxonase 1 (PON1) is an important enzyme with specific protective function on metabolism of lipid peroxides, we examined the influence of GEM on PON1 activity in liver and serum. Gemfibrozil 155-158 paraoxonase 1 Rattus norvegicus 14-27 21223511-3 2011 Because serum paraoxonase 1 (PON1) is an important enzyme with specific protective function on metabolism of lipid peroxides, we examined the influence of GEM on PON1 activity in liver and serum. Gemfibrozil 155-158 paraoxonase 1 Rattus norvegicus 162-166 21381897-8 2011 The present results demonstrated complex interactions between IFN-alpha2b and hepatocytes and the observed down-regulation of CYP1A2, OAT2 and UGT2B7 is consistent with reports of drug interactions between IFN-alpha2b and drugs such as theophylline, clozapine and gemfibrozil. Gemfibrozil 264-275 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 126-132 21381897-8 2011 The present results demonstrated complex interactions between IFN-alpha2b and hepatocytes and the observed down-regulation of CYP1A2, OAT2 and UGT2B7 is consistent with reports of drug interactions between IFN-alpha2b and drugs such as theophylline, clozapine and gemfibrozil. Gemfibrozil 264-275 solute carrier family 22 member 7 Homo sapiens 134-138 21381897-8 2011 The present results demonstrated complex interactions between IFN-alpha2b and hepatocytes and the observed down-regulation of CYP1A2, OAT2 and UGT2B7 is consistent with reports of drug interactions between IFN-alpha2b and drugs such as theophylline, clozapine and gemfibrozil. Gemfibrozil 264-275 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 143-149 21368757-1 2011 To study the time to onset of mechanism-based inactivation of cytochrome P450 (CYP) 2C8 by gemfibrozil in vivo, we conducted a randomized five-phase crossover study in 10 healthy volunteers. Gemfibrozil 91-102 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 62-87 21368757-5 2011 The strong inactivation of CYP2C8, evident as soon as 1 h after gemfibrozil dosing, has implications in clinical practice and in studies with gemfibrozil as a CYP2C8 model inhibitor. Gemfibrozil 64-75 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 27-33 21368757-5 2011 The strong inactivation of CYP2C8, evident as soon as 1 h after gemfibrozil dosing, has implications in clinical practice and in studies with gemfibrozil as a CYP2C8 model inhibitor. Gemfibrozil 142-153 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 27-33 21368757-5 2011 The strong inactivation of CYP2C8, evident as soon as 1 h after gemfibrozil dosing, has implications in clinical practice and in studies with gemfibrozil as a CYP2C8 model inhibitor. Gemfibrozil 142-153 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 159-165 19628752-7 2009 In MRP3-expressing membrane vesicles, ATP-dependent uptakes of the glucuronide conjugates of estradiol, gemfibrozil, E3040, and troglitazone were markedly greater than those in controls, whereas MRP4-expressing membrane vesicles exhibited significant ATP-dependent uptake of gemfibrozil glucuronide and estradiol glucuronide. Gemfibrozil 104-115 ATP-binding cassette, sub-family C (CFTR/MRP), member 3 Mus musculus 3-7 20931329-1 2011 PURPOSE: Gemfibrozil, a strong inhibitor of cytochrome P450 (CYP) 2C8 in vivo, was recently found to markedly increase the plasma concentrations of montelukast in humans. Gemfibrozil 9-20 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 44-69 21175442-12 2010 CONCLUSIONS: Inhibition of UGT enzymes, especially UGT1A1, by gemfibrozil and its glucuronide is an additional mechanism to consider when rationalizing the interaction between repaglinide and gemfibrozil. Gemfibrozil 62-73 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 27-30 21175442-12 2010 CONCLUSIONS: Inhibition of UGT enzymes, especially UGT1A1, by gemfibrozil and its glucuronide is an additional mechanism to consider when rationalizing the interaction between repaglinide and gemfibrozil. Gemfibrozil 62-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 21175442-12 2010 CONCLUSIONS: Inhibition of UGT enzymes, especially UGT1A1, by gemfibrozil and its glucuronide is an additional mechanism to consider when rationalizing the interaction between repaglinide and gemfibrozil. Gemfibrozil 192-203 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 27-30 21175442-12 2010 CONCLUSIONS: Inhibition of UGT enzymes, especially UGT1A1, by gemfibrozil and its glucuronide is an additional mechanism to consider when rationalizing the interaction between repaglinide and gemfibrozil. Gemfibrozil 192-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 19779008-5 2010 This is of great interest since we have recently shown that gemfibrozil, a drug used to treat hyperlipidemia in humans and a synthetic ligand of PPAR alpha, significantly reduces the mortality associated with influenza infections in mice. Gemfibrozil 60-71 peroxisome proliferator activated receptor alpha Homo sapiens 145-155 20810571-8 2010 The relative potency of the compounds was different with each chemokine; for example, gemfibrozil exerted a 55% inhibition on CXCL11, whereas it had a weaker activity on CXCL9 (40% inhibition). Gemfibrozil 86-97 C-X-C motif chemokine ligand 11 Homo sapiens 126-132 20949026-8 2010 A beneficial effect on aortic plaques was also obtained by treating ApoE(-/-) mice with gemfibrozil, a PPARalpha agonist. Gemfibrozil 88-99 apolipoprotein E Mus musculus 68-72 20949026-8 2010 A beneficial effect on aortic plaques was also obtained by treating ApoE(-/-) mice with gemfibrozil, a PPARalpha agonist. Gemfibrozil 88-99 peroxisome proliferator activated receptor alpha Mus musculus 103-112 20516252-9 2010 Further studies with this novel model demonstrated that OATP and cytochrome P450 have a synergistic effect on cerivastatin-gemfibrozil interactions. Gemfibrozil 123-134 solute carrier organic anion transporter family member 1A2 Homo sapiens 56-60 20548972-0 2010 Effects of high sucrose diet, gemfibrozil, and their combination on plasma paraoxonase 1 activity and lipid levels in rats. Gemfibrozil 30-41 paraoxonase 1 Rattus norvegicus 75-88 20953357-7 2010 These events are causally related to hepatotoxicity and hepatocarcinogenicity of gemfibrozil in rodents via peroxisome proliferator activated receptor-alpha (PPARalpha) activation; however, there is widespread evidence that activation of PPARalpha in humans results in expression of genes involved in lipid metabolism, but not in hepatocellular proliferation. Gemfibrozil 81-92 peroxisome proliferator activated receptor alpha Homo sapiens 108-156 20953357-7 2010 These events are causally related to hepatotoxicity and hepatocarcinogenicity of gemfibrozil in rodents via peroxisome proliferator activated receptor-alpha (PPARalpha) activation; however, there is widespread evidence that activation of PPARalpha in humans results in expression of genes involved in lipid metabolism, but not in hepatocellular proliferation. Gemfibrozil 81-92 peroxisome proliferator activated receptor alpha Homo sapiens 158-167 20953357-7 2010 These events are causally related to hepatotoxicity and hepatocarcinogenicity of gemfibrozil in rodents via peroxisome proliferator activated receptor-alpha (PPARalpha) activation; however, there is widespread evidence that activation of PPARalpha in humans results in expression of genes involved in lipid metabolism, but not in hepatocellular proliferation. Gemfibrozil 81-92 peroxisome proliferator activated receptor alpha Homo sapiens 238-247 19919292-10 2010 Bezafibrate and gemfibrozil exhibited concentration-dependent inhibition of OATP1B1-mediated uptake of [(3)H]-oestradiol 17beta-glucuronide with mean IC(50) values of 16 and 27 microM, respectively. Gemfibrozil 16-27 solute carrier organic anion transporter family member 1B1 Homo sapiens 76-83 21966330-2 2011 Fibrates, including fenofibrate, gemfibrozil, benzafibrate, ciprofibrate, and clofibrate act on PPAR alpha to reduce the level of hypertriglyceridemia. Gemfibrozil 33-44 peroxisome proliferator activated receptor alpha Homo sapiens 96-106 20592724-0 2010 Gemfibrozil markedly increases the plasma concentrations of montelukast: a previously unrecognized role for CYP2C8 in the metabolism of montelukast. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 108-114 20592724-6 2010 In conclusion, gemfibrozil markedly increases the plasma concentrations of montelukast, indicating that CYP2C8 is crucial in the elimination of montelukast. Gemfibrozil 15-26 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 104-110 20006695-6 2010 (iii) Parallel changes of prestin mRNA and Gata-3,-2 mRNA levels were observed in response to thyroid hormone and to gemfibrozil application. Gemfibrozil 117-128 solute carrier family 26 member 5 Rattus norvegicus 26-33 20006695-6 2010 (iii) Parallel changes of prestin mRNA and Gata-3,-2 mRNA levels were observed in response to thyroid hormone and to gemfibrozil application. Gemfibrozil 117-128 GATA binding protein 3 Rattus norvegicus 43-52 20548972-4 2010 In the rats on CD and HSD, GEM caused a significant decrease of PON1 activity by 44% and 33%, while a significant decrease of TGs level by 38% (P<0.05) was measured only in rats on CD. Gemfibrozil 27-30 paraoxonase 1 Rattus norvegicus 64-68 20208386-4 2010 Studies with HLM under-predict the ability of gemfibrozil and bupropion to cause clinically significant inhibition of CYP2C8 and CYP2D6, respectively, and over-predict the ability of ezetimibe to cause clinically significant inhibition of CYP3A4. Gemfibrozil 46-57 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 118-124 20208386-4 2010 Studies with HLM under-predict the ability of gemfibrozil and bupropion to cause clinically significant inhibition of CYP2C8 and CYP2D6, respectively, and over-predict the ability of ezetimibe to cause clinically significant inhibition of CYP3A4. Gemfibrozil 46-57 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 129-135 20208386-5 2010 Gemfibrozil and bupropion represent examples of glucuronidation-dependent and reduction-dependent activation to metabolites that inhibit CYP2C8 and CYP2D6, respectively, whereas ezetimibe represents an example of glucuronidation-dependent protection against metabolism-dependent inhibition of CYP3A4. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 137-143 20208386-5 2010 Gemfibrozil and bupropion represent examples of glucuronidation-dependent and reduction-dependent activation to metabolites that inhibit CYP2C8 and CYP2D6, respectively, whereas ezetimibe represents an example of glucuronidation-dependent protection against metabolism-dependent inhibition of CYP3A4. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 148-154 20208386-5 2010 Gemfibrozil and bupropion represent examples of glucuronidation-dependent and reduction-dependent activation to metabolites that inhibit CYP2C8 and CYP2D6, respectively, whereas ezetimibe represents an example of glucuronidation-dependent protection against metabolism-dependent inhibition of CYP3A4. Gemfibrozil 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 293-299 19628752-10 2009 Significant reductions in the basal-to-apical flux of the glucuronide conjugates of estradiol, gemfibrozil, E3040, and troglitazone were observed in the OATP1B1/MRP2/MRP3 triple transfectants compared with those in the double transfectants, whereas significant reduction was observed only for gemfibrozil glucuronide and estradiol glucuronide in the OATP1B1/MRP2/MRP4 triple transfectants. Gemfibrozil 95-106 ATP-binding cassette, sub-family C (CFTR/MRP), member 2 Mus musculus 161-165 19628752-10 2009 Significant reductions in the basal-to-apical flux of the glucuronide conjugates of estradiol, gemfibrozil, E3040, and troglitazone were observed in the OATP1B1/MRP2/MRP3 triple transfectants compared with those in the double transfectants, whereas significant reduction was observed only for gemfibrozil glucuronide and estradiol glucuronide in the OATP1B1/MRP2/MRP4 triple transfectants. Gemfibrozil 95-106 ATP-binding cassette, sub-family C (CFTR/MRP), member 3 Mus musculus 166-170 19628752-10 2009 Significant reductions in the basal-to-apical flux of the glucuronide conjugates of estradiol, gemfibrozil, E3040, and troglitazone were observed in the OATP1B1/MRP2/MRP3 triple transfectants compared with those in the double transfectants, whereas significant reduction was observed only for gemfibrozil glucuronide and estradiol glucuronide in the OATP1B1/MRP2/MRP4 triple transfectants. Gemfibrozil 95-106 ATP-binding cassette, sub-family C (CFTR/MRP), member 2 Mus musculus 358-362 19628752-10 2009 Significant reductions in the basal-to-apical flux of the glucuronide conjugates of estradiol, gemfibrozil, E3040, and troglitazone were observed in the OATP1B1/MRP2/MRP3 triple transfectants compared with those in the double transfectants, whereas significant reduction was observed only for gemfibrozil glucuronide and estradiol glucuronide in the OATP1B1/MRP2/MRP4 triple transfectants. Gemfibrozil 95-106 ATP-binding cassette, sub-family C (CFTR/MRP), member 4 Mus musculus 363-367 19801273-11 2009 In Experiment 4, the TG-lowering drug gemfibrozil also caused a significant reduction in the expression of the orexigenic peptide, OX, in the perifornical lateral hypothalamus. Gemfibrozil 38-49 hypocretin neuropeptide precursor Rattus norvegicus 131-133 19376321-4 2009 The objective of this study was to investigate whether the PPARalpha activator gemfibrozil prevents or delays the development of AF in patients with coronary heart disease. Gemfibrozil 79-90 peroxisome proliferator activated receptor alpha Homo sapiens 59-68 19307362-3 2009 In the present paper, we first show that ciprofloxacin-resistant cells display a faster efflux of ciprofloxacin which is inhibited by gemfibrozil (an unspecific MRP inhibitor). Gemfibrozil 134-145 ATP-binding cassette, sub-family C (CFTR/MRP), member 1 Mus musculus 161-164 19492148-7 2009 In cultured preadipocytes and adipocytes gemfibrozil and fenofibrate significantly reduced PAI-1 protein expression by up to 55 +/- 5% and 34 +/- 4% under basal conditions and up to 56 +/- 6% and 31 +/- 6% under conditions of stimulation of the cells with 40 pM transforming growth factor (TGF)beta, respectively. Gemfibrozil 41-52 serine (or cysteine) peptidase inhibitor, clade E, member 1 Mus musculus 91-96 19492148-7 2009 In cultured preadipocytes and adipocytes gemfibrozil and fenofibrate significantly reduced PAI-1 protein expression by up to 55 +/- 5% and 34 +/- 4% under basal conditions and up to 56 +/- 6% and 31 +/- 6% under conditions of stimulation of the cells with 40 pM transforming growth factor (TGF)beta, respectively. Gemfibrozil 41-52 transforming growth factor, beta 1 Mus musculus 290-298 19492148-10 2009 The decrease in PAI-1 expression induced by gemfibrozil was inhibited by MK886, a PPARalpha inhibitor. Gemfibrozil 44-55 serine (or cysteine) peptidase inhibitor, clade E, member 1 Mus musculus 16-21 19492148-10 2009 The decrease in PAI-1 expression induced by gemfibrozil was inhibited by MK886, a PPARalpha inhibitor. Gemfibrozil 44-55 peroxisome proliferator activated receptor alpha Mus musculus 82-91 19299749-2 2009 In this study, we investigated the mechanism by which the PPARalpha agonist gemfibrozil induces immune deviation and protects mice from EAE. Gemfibrozil 76-87 peroxisome proliferator activated receptor alpha Mus musculus 58-67 19299749-3 2009 We demonstrated that treatment with gemfibrozil increases expression of the Th2 transcription factor GATA-3 and decreases expression of the Th1 transcription factor T-bet in vitro and directly ex vivo. Gemfibrozil 36-47 GATA binding protein 3 Mus musculus 101-107 19299749-3 2009 We demonstrated that treatment with gemfibrozil increases expression of the Th2 transcription factor GATA-3 and decreases expression of the Th1 transcription factor T-bet in vitro and directly ex vivo. Gemfibrozil 36-47 negative elongation factor complex member C/D, Th1l Mus musculus 140-143 19299749-3 2009 We demonstrated that treatment with gemfibrozil increases expression of the Th2 transcription factor GATA-3 and decreases expression of the Th1 transcription factor T-bet in vitro and directly ex vivo. Gemfibrozil 36-47 delta/notch-like EGF repeat containing Mus musculus 167-170 19166694-6 2009 The combination of nicotinic acid and gemfibrozil resulted in a further 17.8% reduction in apolipoprotein B (p = 0.06), a further 33.8% reduction in IDL (p = 0.06), and a greater reduction in the apolipoprotein B/apolipoprotein A-I ratio (p = 0.02). Gemfibrozil 38-49 apolipoprotein B Homo sapiens 91-107 19166694-6 2009 The combination of nicotinic acid and gemfibrozil resulted in a further 17.8% reduction in apolipoprotein B (p = 0.06), a further 33.8% reduction in IDL (p = 0.06), and a greater reduction in the apolipoprotein B/apolipoprotein A-I ratio (p = 0.02). Gemfibrozil 38-49 apolipoprotein B Homo sapiens 196-212 19166694-6 2009 The combination of nicotinic acid and gemfibrozil resulted in a further 17.8% reduction in apolipoprotein B (p = 0.06), a further 33.8% reduction in IDL (p = 0.06), and a greater reduction in the apolipoprotein B/apolipoprotein A-I ratio (p = 0.02). Gemfibrozil 38-49 apolipoprotein A1 Homo sapiens 213-231 19166694-7 2009 The combination of nicotinic acid and gemfibrozil reduced atherogenic by IDL 71%, dense LDL-III by 52%, and apolipoprotein B by 37% and increased protective HDL(2) by 90%. Gemfibrozil 38-49 apolipoprotein B Homo sapiens 108-124 19166694-7 2009 The combination of nicotinic acid and gemfibrozil reduced atherogenic by IDL 71%, dense LDL-III by 52%, and apolipoprotein B by 37% and increased protective HDL(2) by 90%. Gemfibrozil 38-49 junctophilin 3 Homo sapiens 157-163 19238654-2 2008 The mean increase in the repaglinide area under the plasma concentration-time curve from 0 h to infinity (AUC(0-infinity)) produced by gemfibrozil was larger in individuals with the SLCO1B1 c.521CC genotype (n = 6) than in those with the c.521TC (n = 6) and c.521TT (n = 12) genotypes, by factors of 1.56 (P = 0.004) and 1.54 (P = 0.002), respectively. Gemfibrozil 135-146 solute carrier organic anion transporter family member 1B1 Homo sapiens 182-189 18799592-1 2008 The first generation peroxisome proliferator-activated receptor (PPAR) alpha agonist gemfibrozil reduces the risk of major cardiovascular events; therefore, more potent PPARalpha agonists for the treatment of cardiovascular diseases have been actively sought. Gemfibrozil 85-96 peroxisome proliferator activated receptor alpha Homo sapiens 65-69 18799592-1 2008 The first generation peroxisome proliferator-activated receptor (PPAR) alpha agonist gemfibrozil reduces the risk of major cardiovascular events; therefore, more potent PPARalpha agonists for the treatment of cardiovascular diseases have been actively sought. Gemfibrozil 85-96 peroxisome proliferator activated receptor alpha Mus musculus 169-178 19238654-6 2008 In conclusion, the extent of gemfibrozil-repaglinide interaction depends on SLCO1B1 genotype. Gemfibrozil 29-40 solute carrier organic anion transporter family member 1B1 Homo sapiens 76-83 18325492-7 2008 Despite these differences, the C299S AKR1B10 mutant still manifests kinetics similar to the wild-type protein with other fibrates including zopolrestat, fenofibrate, Wy 14,346, gemfibrozil and ciprofibrate that show mixed non-competitive inhibition kinetics. Gemfibrozil 177-188 aldo-keto reductase family 1 member B10 Homo sapiens 37-44 18678301-1 2008 The weak peroxisome proliferator activated receptor-alpha (PPAR-alpha) agonists gemfibrozil and fenofibrate achieve only small increases in high-density lipoprotein (HDL) cholesterol. Gemfibrozil 80-91 peroxisome proliferator activated receptor alpha Homo sapiens 9-57 18678301-1 2008 The weak peroxisome proliferator activated receptor-alpha (PPAR-alpha) agonists gemfibrozil and fenofibrate achieve only small increases in high-density lipoprotein (HDL) cholesterol. Gemfibrozil 80-91 peroxisome proliferator activated receptor alpha Homo sapiens 59-69 18420459-6 2008 An experimental study has shown that the fibrate gemfibrozil, a peroxisome proliferator-activated receptor (PPAR) alpha agonist, reduces mortality in H2N2 influenza virus-infected mice. Gemfibrozil 49-60 peroxisome proliferator activated receptor alpha Homo sapiens 108-112 18547436-7 2008 Changes in plasma PCSK9 following fenofibrate or gemfibrozil treatments (fibric acid derivatives) were inversely correlated with changes in LDLC levels (r = -0.558, p = 0.013). Gemfibrozil 49-60 proprotein convertase subtilisin/kexin type 9 Homo sapiens 18-23 18547436-7 2008 Changes in plasma PCSK9 following fenofibrate or gemfibrozil treatments (fibric acid derivatives) were inversely correlated with changes in LDLC levels (r = -0.558, p = 0.013). Gemfibrozil 49-60 component of oligomeric golgi complex 2 Homo sapiens 140-144 18356553-6 2008 Although gemfibrozil reduced Lp-PLA(2) only modestly (6.6%), at higher levels of Lp-PLA(2) gemfibrozil produced a significant reduction in CV events. Gemfibrozil 9-20 phospholipase A2 group VII Homo sapiens 29-37 18356553-6 2008 Although gemfibrozil reduced Lp-PLA(2) only modestly (6.6%), at higher levels of Lp-PLA(2) gemfibrozil produced a significant reduction in CV events. Gemfibrozil 9-20 phospholipase A2 group VII Homo sapiens 29-38 18356553-7 2008 CONCLUSIONS: In VA-HIT, a population with low HDL-C and LDL-C, high Lp-PLA(2) independently predicted CV events that were reduced by gemfibrozil. Gemfibrozil 133-144 component of oligomeric golgi complex 2 Homo sapiens 56-61 18356553-7 2008 CONCLUSIONS: In VA-HIT, a population with low HDL-C and LDL-C, high Lp-PLA(2) independently predicted CV events that were reduced by gemfibrozil. Gemfibrozil 133-144 phospholipase A2 group VII Homo sapiens 68-76 17901929-5 2008 Gemfibrozil and cyclosporine inhibition data obtained in human embryonic kidney cells expressing OATP1B1 and hepatic input concentration ([I]in) were used for qualitative zoning of 14 transporter-mediated DDIs. Gemfibrozil 0-11 solute carrier organic anion transporter family member 1B1 Homo sapiens 97-104 17335828-8 2008 CONCLUSIONS: In VA-HIT, gemfibrozil resulted in weight loss associated with reductions in insulin. Gemfibrozil 24-35 insulin Homo sapiens 90-97 18502109-5 2008 Gemfibrozil produces major modifications in fatty acid composition, which are both fatty acid and lipid class specific but generally decreases SFA and increases PUFA (mainly n6) and increases the proportion of fatty acids with chain length of 18C or more. Gemfibrozil 0-11 pumilio RNA binding family member 3 Homo sapiens 161-165 18356690-5 2008 While fenofibrate and LY518674 showed exclusive dependency on PPARalpha for these activities, bezafibrate and gemfibrozil exhibited dependency on PPARbeta/delta and PPARgamma as well. Gemfibrozil 110-121 peroxisome proliferator activated receptor delta Homo sapiens 146-154 18356690-5 2008 While fenofibrate and LY518674 showed exclusive dependency on PPARalpha for these activities, bezafibrate and gemfibrozil exhibited dependency on PPARbeta/delta and PPARgamma as well. Gemfibrozil 110-121 peroxisome proliferator activated receptor gamma Homo sapiens 165-174 17625103-7 2007 The histological and immunohistochemical analyses also demonstrate the inhibitory effect of gemfibrozil on the invasion of T-bet-positive T cells into the spinal cord of EAE mice. Gemfibrozil 92-103 T-box 21 Mus musculus 123-128 18092840-13 2008 In clinical trials of gemfibrozil, a PPARalpha ligand, significantly fewer patients treated with this lipid-lowering drug were diagnosed with melanoma as compared to those in the control group. Gemfibrozil 22-33 peroxisome proliferator activated receptor alpha Homo sapiens 37-46 17823123-3 2007 Here we describe that after Th2 polarization by gemfibrozil or other drugs, MBP-primed T cells induced the expression of neurotrophic molecules such as brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), but not proinflammatory molecules in microglia and astroglia via cell-to-cell contact. Gemfibrozil 48-59 myelin basic protein Homo sapiens 76-79 17823123-3 2007 Here we describe that after Th2 polarization by gemfibrozil or other drugs, MBP-primed T cells induced the expression of neurotrophic molecules such as brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), but not proinflammatory molecules in microglia and astroglia via cell-to-cell contact. Gemfibrozil 48-59 brain derived neurotrophic factor Homo sapiens 152-185 17965553-6 2007 In fact, WY-14,643, clofibrate, gemfibrozil and benzbromarone, reported to be PPARalpha activators, distributed to the former, whereas propylthiouracil, omeprazole, phenobarbital, thioacetamide, methapyrilene, sulfasalazine and coumarin did to the latter. Gemfibrozil 32-43 peroxisome proliferator activated receptor alpha Rattus norvegicus 78-87 18078862-5 2008 Analyses in the gemfibrozil arm showed that subjects with recurrent CVD events had significantly higher prebeta-1 and had significantly lower alpha-1 and alpha-2 HDL levels than those without such events. Gemfibrozil 16-27 adrenoceptor alpha 1D Homo sapiens 142-149 18078862-8 2008 Gemfibrozil treatment was associated with 3% to 6% decreases in the small, lipid-poor prebeta-1 HDL and in the large, lipid-rich alpha-1 and alpha-2 HDL and with increases in the small alpha-3 (3%) and prealpha-3 (16%) HDLs. Gemfibrozil 0-11 adrenoceptor alpha 1D Homo sapiens 129-136 17670842-0 2007 The UDP-glucuronosyltransferase 2B7 isozyme is responsible for gemfibrozil glucuronidation in the human liver. Gemfibrozil 63-74 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 4-35 17670842-3 2007 Comprehensive assessment of the drug interaction between gemfibrozil and cytochrome P450 2C8 substrates requires a clear understanding of gemfibrozil glucuronidation. Gemfibrozil 138-149 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 73-92 17670842-4 2007 However, the primary UDP-glucuronosyltransferase (UGT) isozymes responsible for gemfibrozil glucuronidation remain to be determined. Gemfibrozil 80-91 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 21-48 17670842-4 2007 However, the primary UDP-glucuronosyltransferase (UGT) isozymes responsible for gemfibrozil glucuronidation remain to be determined. Gemfibrozil 80-91 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 50-53 17670842-5 2007 Here, we identified the main UGT isozymes involved in gemfibrozil glucuronidation. Gemfibrozil 54-65 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 29-32 17670842-6 2007 Evaluation of 12 recombinant human UGT isozymes shows gemfibrozil glucuronidation activity in UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, with UGT2B7 showing the highest activity. Gemfibrozil 54-65 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 35-38 17670842-6 2007 Evaluation of 12 recombinant human UGT isozymes shows gemfibrozil glucuronidation activity in UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, with UGT2B7 showing the highest activity. Gemfibrozil 54-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 17670842-6 2007 Evaluation of 12 recombinant human UGT isozymes shows gemfibrozil glucuronidation activity in UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, with UGT2B7 showing the highest activity. Gemfibrozil 54-65 UDP glucuronosyltransferase family 1 member A3 Homo sapiens 102-108 17670842-6 2007 Evaluation of 12 recombinant human UGT isozymes shows gemfibrozil glucuronidation activity in UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, with UGT2B7 showing the highest activity. Gemfibrozil 54-65 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 110-116 17670842-6 2007 Evaluation of 12 recombinant human UGT isozymes shows gemfibrozil glucuronidation activity in UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, with UGT2B7 showing the highest activity. Gemfibrozil 54-65 UDP glucuronosyltransferase family 2 member B4 Homo sapiens 118-124 17670842-6 2007 Evaluation of 12 recombinant human UGT isozymes shows gemfibrozil glucuronidation activity in UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, with UGT2B7 showing the highest activity. Gemfibrozil 54-65 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 126-132 17670842-6 2007 Evaluation of 12 recombinant human UGT isozymes shows gemfibrozil glucuronidation activity in UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, with UGT2B7 showing the highest activity. Gemfibrozil 54-65 UDP glucuronosyltransferase family 2 member B17 Homo sapiens 138-145 17670842-6 2007 Evaluation of 12 recombinant human UGT isozymes shows gemfibrozil glucuronidation activity in UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, with UGT2B7 showing the highest activity. Gemfibrozil 54-65 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 152-158 17670842-8 2007 The high affinity K(m) value was 2.5 microM, which is similar to the K(m) value of gemfibrozil glucuronidation in recombinant UGT2B7 (2.2 microM). Gemfibrozil 83-94 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 126-132 17670842-11 2007 These results suggest that UGT2B7 is the main isozyme responsible for gemfibrozil glucuronidation in humans. Gemfibrozil 70-81 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 27-33 17625103-1 2007 The present study underlines the importance of gemfibrozil, a lipid-lowering drug and an activator of peroxisome proliferator-activated receptor-alpha (PPAR-alpha), in inhibiting the disease process of adoptively transferred experimental allergic encephalomyelitis (EAE), an animal model of relapsing-remitting multiple sclerosis. Gemfibrozil 47-58 peroxisome proliferator activated receptor alpha Mus musculus 102-150 17625103-1 2007 The present study underlines the importance of gemfibrozil, a lipid-lowering drug and an activator of peroxisome proliferator-activated receptor-alpha (PPAR-alpha), in inhibiting the disease process of adoptively transferred experimental allergic encephalomyelitis (EAE), an animal model of relapsing-remitting multiple sclerosis. Gemfibrozil 47-58 peroxisome proliferator activated receptor alpha Mus musculus 152-162 17625103-4 2007 Gemfibrozil also inhibited the encephalitogenicity of MBP-primed T cells and switched the immune response from a Th1 to a Th2 profile independent of PPAR-alpha. Gemfibrozil 0-11 myelin basic protein Mus musculus 54-57 17625103-4 2007 Gemfibrozil also inhibited the encephalitogenicity of MBP-primed T cells and switched the immune response from a Th1 to a Th2 profile independent of PPAR-alpha. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 149-159 17625103-5 2007 Gemfibrozil consistently inhibited the expression and DNA-binding activity of T-bet, a key regulator of interferon-gamma (IFN-gamma) expression and stimulated the expression and DNA-binding activity of GATA3, a key regulator of IL-4. Gemfibrozil 0-11 T-box 21 Mus musculus 78-83 17625103-5 2007 Gemfibrozil consistently inhibited the expression and DNA-binding activity of T-bet, a key regulator of interferon-gamma (IFN-gamma) expression and stimulated the expression and DNA-binding activity of GATA3, a key regulator of IL-4. Gemfibrozil 0-11 interferon gamma Mus musculus 104-120 17625103-8 2007 Furthermore, we demonstrate that the differential effect of gemfibrozil on the expression of T-bet and GATA3 was due to its inhibitory effect on NO production. Gemfibrozil 60-71 T-box 21 Mus musculus 93-98 17625103-5 2007 Gemfibrozil consistently inhibited the expression and DNA-binding activity of T-bet, a key regulator of interferon-gamma (IFN-gamma) expression and stimulated the expression and DNA-binding activity of GATA3, a key regulator of IL-4. Gemfibrozil 0-11 interferon gamma Mus musculus 122-131 17625103-5 2007 Gemfibrozil consistently inhibited the expression and DNA-binding activity of T-bet, a key regulator of interferon-gamma (IFN-gamma) expression and stimulated the expression and DNA-binding activity of GATA3, a key regulator of IL-4. Gemfibrozil 0-11 GATA binding protein 3 Mus musculus 202-207 17625103-5 2007 Gemfibrozil consistently inhibited the expression and DNA-binding activity of T-bet, a key regulator of interferon-gamma (IFN-gamma) expression and stimulated the expression and DNA-binding activity of GATA3, a key regulator of IL-4. Gemfibrozil 0-11 interleukin 4 Mus musculus 228-232 17625103-8 2007 Furthermore, we demonstrate that the differential effect of gemfibrozil on the expression of T-bet and GATA3 was due to its inhibitory effect on NO production. Gemfibrozil 60-71 GATA binding protein 3 Mus musculus 103-108 17470528-0 2007 Substrate-dependent drug-drug interactions between gemfibrozil, fluvastatin and other organic anion-transporting peptide (OATP) substrates on OATP1B1, OATP2B1, and OATP1B3. Gemfibrozil 51-62 solute carrier organic anion transporter family member 1A2 Homo sapiens 86-120 17625103-6 2007 Gemfibrozil treatment decreased the number of T-bet-positive T cells and increased the number of GATA3-positive T cells in spleen of donor mice. Gemfibrozil 0-11 T-box transcription factor 21 Homo sapiens 46-51 17625103-6 2007 Gemfibrozil treatment decreased the number of T-bet-positive T cells and increased the number of GATA3-positive T cells in spleen of donor mice. Gemfibrozil 0-11 GATA binding protein 3 Homo sapiens 97-102 17470528-8 2007 Recombinant OATP1B1-, OATP2B1-, and OATP1B3-mediated fluvastatin transport was inhibited to 97, 70, and 62% by gemfibrozil (200 microM), respectively, whereas only a small inhibitory effect by gemfibrozil (200 microM) on fluvastatin uptake into primary human hepatocytes was observed (27% inhibition). Gemfibrozil 193-204 solute carrier organic anion transporter family member 1B1 Homo sapiens 12-19 17785853-0 2007 Involvement of phosphatidylinositol 3-kinase-mediated up-regulation of I kappa B alpha in anti-inflammatory effect of gemfibrozil in microglia. Gemfibrozil 118-129 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 71-86 17785853-2 2007 Gemfibrozil inhibited LPS-induced expression of inducible NO synthase (iNOS) and proinflammatory cytokines in mouse BV-2 microglial cells and primary microglia. Gemfibrozil 0-11 nitric oxide synthase 2, inducible Mus musculus 48-69 17785853-2 2007 Gemfibrozil inhibited LPS-induced expression of inducible NO synthase (iNOS) and proinflammatory cytokines in mouse BV-2 microglial cells and primary microglia. Gemfibrozil 0-11 nitric oxide synthase 2, inducible Mus musculus 71-75 17785853-3 2007 By overexpressing wild-type and dominant-negative constructs of peroxisome proliferator-activated receptor-alpha (PPAR-alpha) in microglial cells and isolating primary microglia from PPAR-alpha-/- mice, we have demonstrated that gemfibrozil inhibits the activation of microglia independent of PPAR-alpha. Gemfibrozil 229-240 peroxisome proliferator activated receptor alpha Mus musculus 64-112 17785853-3 2007 By overexpressing wild-type and dominant-negative constructs of peroxisome proliferator-activated receptor-alpha (PPAR-alpha) in microglial cells and isolating primary microglia from PPAR-alpha-/- mice, we have demonstrated that gemfibrozil inhibits the activation of microglia independent of PPAR-alpha. Gemfibrozil 229-240 peroxisome proliferator activated receptor alpha Mus musculus 114-124 17785853-4 2007 Interestingly, gemfibrozil induced the activation of p85alpha-associated PI3K (p110beta but not p110alpha) and inhibition of that PI3K by either chemical inhibitors or dominant-negative mutants abrogated the inhibitory effect of gemfibrozil. Gemfibrozil 15-26 phosphoinositide-3-kinase regulatory subunit 1 Mus musculus 53-61 17785853-4 2007 Interestingly, gemfibrozil induced the activation of p85alpha-associated PI3K (p110beta but not p110alpha) and inhibition of that PI3K by either chemical inhibitors or dominant-negative mutants abrogated the inhibitory effect of gemfibrozil. Gemfibrozil 15-26 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta Mus musculus 79-87 17785853-4 2007 Interestingly, gemfibrozil induced the activation of p85alpha-associated PI3K (p110beta but not p110alpha) and inhibition of that PI3K by either chemical inhibitors or dominant-negative mutants abrogated the inhibitory effect of gemfibrozil. Gemfibrozil 229-240 phosphoinositide-3-kinase regulatory subunit 1 Mus musculus 53-61 17785853-5 2007 Conversely, overexpression of the constitutively active mutant of p110 enhanced the inhibitory effect of gemfibrozil on LPS-induced expression of proinflammatory molecules. Gemfibrozil 105-116 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha Mus musculus 66-70 17785853-6 2007 Similarly, gemfibrozil also inhibited fibrillar amyloid beta (Abeta)-, prion peptide (PrP)-, dsRNA (poly IC)-, HIV-1 Tat-, and 1-methyl-4-phenylpyridinium (MPP+)-, but not IFN-gamma-, induced microglial expression of iNOS. Gemfibrozil 11-22 prion protein Mus musculus 86-89 17785853-6 2007 Similarly, gemfibrozil also inhibited fibrillar amyloid beta (Abeta)-, prion peptide (PrP)-, dsRNA (poly IC)-, HIV-1 Tat-, and 1-methyl-4-phenylpyridinium (MPP+)-, but not IFN-gamma-, induced microglial expression of iNOS. Gemfibrozil 11-22 interferon gamma Mus musculus 172-181 17785853-6 2007 Similarly, gemfibrozil also inhibited fibrillar amyloid beta (Abeta)-, prion peptide (PrP)-, dsRNA (poly IC)-, HIV-1 Tat-, and 1-methyl-4-phenylpyridinium (MPP+)-, but not IFN-gamma-, induced microglial expression of iNOS. Gemfibrozil 11-22 nitric oxide synthase 2, inducible Mus musculus 217-221 17785853-7 2007 Inhibition of PI3K also abolished the inhibitory effect of gemfibrozil on Abeta-, PrP-, poly IC-, Tat-, and MPP+-induced microglial expression of iNOS. Gemfibrozil 59-70 prion protein Mus musculus 82-85 17785853-7 2007 Inhibition of PI3K also abolished the inhibitory effect of gemfibrozil on Abeta-, PrP-, poly IC-, Tat-, and MPP+-induced microglial expression of iNOS. Gemfibrozil 59-70 nitric oxide synthase 2, inducible Mus musculus 146-150 17470528-8 2007 Recombinant OATP1B1-, OATP2B1-, and OATP1B3-mediated fluvastatin transport was inhibited to 97, 70, and 62% by gemfibrozil (200 microM), respectively, whereas only a small inhibitory effect by gemfibrozil (200 microM) on fluvastatin uptake into primary human hepatocytes was observed (27% inhibition). Gemfibrozil 193-204 solute carrier organic anion transporter family member 2B1 Homo sapiens 22-29 17470528-8 2007 Recombinant OATP1B1-, OATP2B1-, and OATP1B3-mediated fluvastatin transport was inhibited to 97, 70, and 62% by gemfibrozil (200 microM), respectively, whereas only a small inhibitory effect by gemfibrozil (200 microM) on fluvastatin uptake into primary human hepatocytes was observed (27% inhibition). Gemfibrozil 193-204 solute carrier organic anion transporter family member 1B3 Homo sapiens 36-43 17470528-0 2007 Substrate-dependent drug-drug interactions between gemfibrozil, fluvastatin and other organic anion-transporting peptide (OATP) substrates on OATP1B1, OATP2B1, and OATP1B3. Gemfibrozil 51-62 solute carrier organic anion transporter family member 1A2 Homo sapiens 122-126 17470528-0 2007 Substrate-dependent drug-drug interactions between gemfibrozil, fluvastatin and other organic anion-transporting peptide (OATP) substrates on OATP1B1, OATP2B1, and OATP1B3. Gemfibrozil 51-62 solute carrier organic anion transporter family member 1B1 Homo sapiens 142-149 17470528-0 2007 Substrate-dependent drug-drug interactions between gemfibrozil, fluvastatin and other organic anion-transporting peptide (OATP) substrates on OATP1B1, OATP2B1, and OATP1B3. Gemfibrozil 51-62 solute carrier organic anion transporter family member 2B1 Homo sapiens 151-158 17470528-0 2007 Substrate-dependent drug-drug interactions between gemfibrozil, fluvastatin and other organic anion-transporting peptide (OATP) substrates on OATP1B1, OATP2B1, and OATP1B3. Gemfibrozil 51-62 solute carrier organic anion transporter family member 1B3 Homo sapiens 164-171 17470528-5 2007 For OATP1B1 the inhibition by gemfibrozil was substrate-dependent as the transport of fluvastatin (IC(50) of 63 microM), pravastatin, simvastatin, and taurocholate was inhibited by gemfibrozil, whereas the transport of estrone-3-sulfate and troglitazone sulfate (both used at 3 microM) was not affected. Gemfibrozil 30-41 solute carrier organic anion transporter family member 1B1 Homo sapiens 4-11 17470528-5 2007 For OATP1B1 the inhibition by gemfibrozil was substrate-dependent as the transport of fluvastatin (IC(50) of 63 microM), pravastatin, simvastatin, and taurocholate was inhibited by gemfibrozil, whereas the transport of estrone-3-sulfate and troglitazone sulfate (both used at 3 microM) was not affected. Gemfibrozil 181-192 solute carrier organic anion transporter family member 1B1 Homo sapiens 4-11 17470528-8 2007 Recombinant OATP1B1-, OATP2B1-, and OATP1B3-mediated fluvastatin transport was inhibited to 97, 70, and 62% by gemfibrozil (200 microM), respectively, whereas only a small inhibitory effect by gemfibrozil (200 microM) on fluvastatin uptake into primary human hepatocytes was observed (27% inhibition). Gemfibrozil 111-122 solute carrier organic anion transporter family member 1B1 Homo sapiens 12-19 17470528-8 2007 Recombinant OATP1B1-, OATP2B1-, and OATP1B3-mediated fluvastatin transport was inhibited to 97, 70, and 62% by gemfibrozil (200 microM), respectively, whereas only a small inhibitory effect by gemfibrozil (200 microM) on fluvastatin uptake into primary human hepatocytes was observed (27% inhibition). Gemfibrozil 111-122 solute carrier organic anion transporter family member 2B1 Homo sapiens 22-29 17470528-8 2007 Recombinant OATP1B1-, OATP2B1-, and OATP1B3-mediated fluvastatin transport was inhibited to 97, 70, and 62% by gemfibrozil (200 microM), respectively, whereas only a small inhibitory effect by gemfibrozil (200 microM) on fluvastatin uptake into primary human hepatocytes was observed (27% inhibition). Gemfibrozil 111-122 solute carrier organic anion transporter family member 1B3 Homo sapiens 36-43 17633905-7 2007 Other lipid-lowering agents reported to reduce CRP include niacin, fibrates, and gemfibrozil. Gemfibrozil 81-92 C-reactive protein Homo sapiens 47-50 17333159-2 2007 We studied the possible effect of gemfibrozil, an in vivo inhibitor of CYP2C8, on the pharmacokinetics of ibuprofen in healthy volunteers. Gemfibrozil 34-45 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 71-77 17333159-10 2007 CONCLUSIONS: Gemfibrozil moderately increases the AUC(0-infinity) of R-ibuprofen and prolongs its t (1/2), indicating that R-ibuprofen is partially metabolised by CYP2C8. Gemfibrozil 13-24 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 163-169 17333159-0 2007 Stereoselective interaction between the CYP2C8 inhibitor gemfibrozil and racemic ibuprofen. Gemfibrozil 57-68 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 40-46 16782410-4 2006 PPARalpha is activated by polyunsaturated fatty acids and by fibrate drugs (fenofibrate and gemfibrozil) and controls expression of genes involved in lipid metabolism. Gemfibrozil 92-103 peroxisome proliferator activated receptor alpha Homo sapiens 0-9 17897018-4 2007 Fenofibrate and gemfibrozil are two drugs that act as PPARalpha agonists and are currently in use in the clinical setting. Gemfibrozil 16-27 peroxisome proliferator activated receptor alpha Homo sapiens 54-63 17897018-8 2007 These studies have shown both PPARalpha agonists, gemfibrozil and fenofibrate, confer anti-atherosclerotic effects, partly independent of their metabolic effects. Gemfibrozil 50-61 peroxisome proliferator activated receptor alpha Homo sapiens 30-39 17202761-2 2006 In the present study, the data of three ligands of peroxisome proliferator activated receptor alpha (PPARalpha), i.e., clofibrate, WY-14643 and gemfibrozil in our database were analyzed. Gemfibrozil 144-155 peroxisome proliferator activated receptor alpha Rattus norvegicus 51-99 17202761-2 2006 In the present study, the data of three ligands of peroxisome proliferator activated receptor alpha (PPARalpha), i.e., clofibrate, WY-14643 and gemfibrozil in our database were analyzed. Gemfibrozil 144-155 peroxisome proliferator activated receptor alpha Rattus norvegicus 101-110 16764943-2 2006 Recently, we demonstrated that administration of the PPAR-alpha agonists gemfibrozil and fenofibrate, inhibit the clinical signs of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Gemfibrozil 73-84 peroxisome proliferator activated receptor alpha Mus musculus 53-63 16764943-8 2006 Interestingly, a combination of 9-cis RA and the PPAR-alpha agonists fenofibrate or gemfibrozil cooperatively inhibited NO, TNF-alpha, IL-1beta, IL-6, and MCP-1 production by these cells. Gemfibrozil 84-95 peroxisome proliferator activated receptor alpha Mus musculus 49-59 16764943-8 2006 Interestingly, a combination of 9-cis RA and the PPAR-alpha agonists fenofibrate or gemfibrozil cooperatively inhibited NO, TNF-alpha, IL-1beta, IL-6, and MCP-1 production by these cells. Gemfibrozil 84-95 tumor necrosis factor Mus musculus 124-133 16764943-8 2006 Interestingly, a combination of 9-cis RA and the PPAR-alpha agonists fenofibrate or gemfibrozil cooperatively inhibited NO, TNF-alpha, IL-1beta, IL-6, and MCP-1 production by these cells. Gemfibrozil 84-95 interleukin 1 beta Mus musculus 135-143 16764943-8 2006 Interestingly, a combination of 9-cis RA and the PPAR-alpha agonists fenofibrate or gemfibrozil cooperatively inhibited NO, TNF-alpha, IL-1beta, IL-6, and MCP-1 production by these cells. Gemfibrozil 84-95 interleukin 6 Mus musculus 145-149 16764943-8 2006 Interestingly, a combination of 9-cis RA and the PPAR-alpha agonists fenofibrate or gemfibrozil cooperatively inhibited NO, TNF-alpha, IL-1beta, IL-6, and MCP-1 production by these cells. Gemfibrozil 84-95 mast cell protease 1 Mus musculus 155-160 17523051-0 2007 Gemfibrozil and its glucuronide inhibit the hepatic uptake of pravastatin mediated by OATP1B1. Gemfibrozil 0-11 solute carrier organic anion transporter family member 1B1 Homo sapiens 86-93 17523051-7 2007 Considering the plasma concentrations of gemfibrozil and gem-glu in humans, the inhibition of OATP1B1-mediated hepatic uptake of pravastatin by gem-glu would contribute, at least in part, to the elevation of plasma concentration of pravastatin by the concomitant use of gemfibrozil. Gemfibrozil 41-52 solute carrier organic anion transporter family member 1B1 Homo sapiens 94-101 17523051-7 2007 Considering the plasma concentrations of gemfibrozil and gem-glu in humans, the inhibition of OATP1B1-mediated hepatic uptake of pravastatin by gem-glu would contribute, at least in part, to the elevation of plasma concentration of pravastatin by the concomitant use of gemfibrozil. Gemfibrozil 270-281 solute carrier organic anion transporter family member 1B1 Homo sapiens 94-101 17455113-0 2007 Inhibition of human organic anion transporter 3 mediated pravastatin transport by gemfibrozil and the metabolites in humans. Gemfibrozil 82-93 solute carrier family 22 member 8 Homo sapiens 20-47 17455113-5 2007 Gemfibrozil and its glucuronide and carboxylic metabolite forms inhibited the uptake of pravastatin by hOAT3 with IC(50) values of 6.8 microM, 19.7 microM and 5.4 microM, respectively. Gemfibrozil 0-11 solute carrier family 22 member 8 Homo sapiens 103-108 17455113-6 2007 Considering the plasma concentrations of gemfibrozil and its metabolites in humans, the inhibition of hOAT3-mediated pravastatin transport by gemfibrozil and its metabolites would lead to a decrease in the renal clearance of pravastatin in clinical settings. Gemfibrozil 41-52 solute carrier family 22 member 8 Homo sapiens 102-107 17455113-6 2007 Considering the plasma concentrations of gemfibrozil and its metabolites in humans, the inhibition of hOAT3-mediated pravastatin transport by gemfibrozil and its metabolites would lead to a decrease in the renal clearance of pravastatin in clinical settings. Gemfibrozil 142-153 solute carrier family 22 member 8 Homo sapiens 102-107 17253883-15 2007 The most impressive pharmacokinetic interaction was reported with combined administration of gemfibrozil (a strong CYP2C8 inhibitor) and repaglinide (8-fold increase in repaglinide AUC). Gemfibrozil 93-104 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 115-121 17178259-10 2006 Gemfibrozil and its glucuronide inhibit CYP2C8 and OATP1B1. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 40-46 17178259-10 2006 Gemfibrozil and its glucuronide inhibit CYP2C8 and OATP1B1. Gemfibrozil 0-11 solute carrier organic anion transporter family member 1B1 Homo sapiens 51-58 17178262-8 2006 Furthermore, estrone-3-sulfate transport into OATP2B1-overexpressing Madin-Darby canine kidney II cells was inhibited by various drugs, including atorvastatin, simvastatin, cerivastatin, glyburide (INN, glibenclamide), and gemfibrozil, with the most pronounced effect being found for atorvastatin (inhibition constant, 0.7 +/- 0.4 micromol/L). Gemfibrozil 223-234 solute carrier organic anion transporter family member 2B1 Homo sapiens 46-53 16828346-2 2006 Treatment with a fibrate such as gemfibrozil (a PPAR alpha agonist) has been shown to be especially effective in people with low HDL-C and other features of the metabolic syndrome. Gemfibrozil 33-44 peroxisome proliferator activated receptor alpha Homo sapiens 48-58 16832679-0 2006 The CYP2C8 inhibitor gemfibrozil does not increase the plasma concentrations of zopiclone. Gemfibrozil 21-32 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 4-10 16832679-2 2006 We studied the possible effect of gemfibrozil, an inhibitor of CYP2C8, on the pharmacokinetics and pharmacodynamics of zopiclone. Gemfibrozil 34-45 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 63-69 16221474-1 2006 BACKGROUND: The Veterans Affairs HDL Intervention Trial (VA-HIT) showed that gemfibrozil, which activates peroxisome proliferator-activator receptor alpha (PPARalpha), significantly reduced the risk of cardiovascular (CV) events in men with low HDL cholesterol (< 40 mg/dl) and established coronary heart disease. Gemfibrozil 77-88 peroxisome proliferator activated receptor alpha Homo sapiens 156-165 16299161-0 2006 Glucuronidation converts gemfibrozil to a potent, metabolism-dependent inhibitor of CYP2C8: implications for drug-drug interactions. Gemfibrozil 25-36 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 84-90 16463048-10 2006 In addition, gemfibrozil reduced the expression of the genes encoding the NADPH oxidase subunits p47phox, gp91phox and Rac-1. Gemfibrozil 13-24 neutrophil cytosolic factor 1 Mus musculus 97-104 16463048-10 2006 In addition, gemfibrozil reduced the expression of the genes encoding the NADPH oxidase subunits p47phox, gp91phox and Rac-1. Gemfibrozil 13-24 cytochrome b-245, beta polypeptide Mus musculus 106-114 16463048-10 2006 In addition, gemfibrozil reduced the expression of the genes encoding the NADPH oxidase subunits p47phox, gp91phox and Rac-1. Gemfibrozil 13-24 Rac family small GTPase 1 Mus musculus 119-124 16463048-11 2006 In addition, gemfibrozil reduced the expression of the genes encoding nuclear factor kappa B (NF-kappaB) subunit, p65, the NF-kappaB-dependent chemokine monocyte chemoattractant protein-1, and tissue factor. Gemfibrozil 13-24 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 70-92 16463048-11 2006 In addition, gemfibrozil reduced the expression of the genes encoding nuclear factor kappa B (NF-kappaB) subunit, p65, the NF-kappaB-dependent chemokine monocyte chemoattractant protein-1, and tissue factor. Gemfibrozil 13-24 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 94-103 16463048-11 2006 In addition, gemfibrozil reduced the expression of the genes encoding nuclear factor kappa B (NF-kappaB) subunit, p65, the NF-kappaB-dependent chemokine monocyte chemoattractant protein-1, and tissue factor. Gemfibrozil 13-24 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 114-117 16463048-11 2006 In addition, gemfibrozil reduced the expression of the genes encoding nuclear factor kappa B (NF-kappaB) subunit, p65, the NF-kappaB-dependent chemokine monocyte chemoattractant protein-1, and tissue factor. Gemfibrozil 13-24 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 123-132 16463048-11 2006 In addition, gemfibrozil reduced the expression of the genes encoding nuclear factor kappa B (NF-kappaB) subunit, p65, the NF-kappaB-dependent chemokine monocyte chemoattractant protein-1, and tissue factor. Gemfibrozil 13-24 coagulation factor III Mus musculus 193-206 16299161-1 2006 Gemfibrozil more potently inhibits CYP2C9 than CYP2C8 in vitro, and yet the opposite inhibitory potency is observed in the clinic. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 16299161-1 2006 Gemfibrozil more potently inhibits CYP2C9 than CYP2C8 in vitro, and yet the opposite inhibitory potency is observed in the clinic. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 47-53 16299161-3 2006 Gemfibrozil most potently inhibited CYP2C9 (IC50 of 30 microM), whereas gemfibrozil glucuronide most potently inhibited CYP2C8 (IC50 of 24 microM). Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 16299161-4 2006 Unexpectedly, gemfibrozil glucuronide, but not gemfibrozil, was found to be a metabolism-dependent inhibitor of CYP2C8 only. Gemfibrozil 14-25 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 112-118 16299161-7 2006 Potent inhibition of CYP2C8 was also achieved by first incubating gemfibrozil with alamethicin-activated human liver microsomes and UDP-glucuronic acid (to form gemfibrozil glucuronide), followed by a second incubation with NADPH. Gemfibrozil 66-77 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 21-27 16299161-9 2006 The results described have important implications for the mechanism of the clinical interaction reported between gemfibrozil and CYP2C8 substrates such as cerivastatin, repaglinide, rosiglitazone, and pioglitazone. Gemfibrozil 113-124 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 129-135 16883221-10 2006 vWF activity did not change in controls and significantly increased in gemfibrozil group by days 7, 14, but from lower baseline level. Gemfibrozil 71-82 von Willebrand factor Homo sapiens 0-3 17668698-6 2006 RESULTS: Gemfibrozil elevated significantly apolipoprotein A-I, but decreased the total cholesterol, LDL cholesterol, triglycerides, the lipid indices, the ADP-induced platelet aggregation, plasminogen, alpha2-antiplasmin and hematocrit. Gemfibrozil 9-20 apolipoprotein A1 Homo sapiens 44-62 17668698-6 2006 RESULTS: Gemfibrozil elevated significantly apolipoprotein A-I, but decreased the total cholesterol, LDL cholesterol, triglycerides, the lipid indices, the ADP-induced platelet aggregation, plasminogen, alpha2-antiplasmin and hematocrit. Gemfibrozil 9-20 serpin family F member 2 Homo sapiens 203-221 16042675-11 2005 The findings suggest that in vivo gemfibrozil, probably due to its metabolites, is a much more potent inhibitor of CYP2C8 than of CYP2C9. Gemfibrozil 34-45 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 115-121 16507380-0 2005 Potential drug-drug interaction between interferon alfa-2b and gemfibrozil in a patient with malignant melanoma. Gemfibrozil 63-74 interferon alpha 1 Homo sapiens 40-50 16507380-4 2005 OBJECTIVE: The aim of this report was to describe a potential drug-drug interaction between IFN alfa-2b and gemfibrozil in a patient with malignant melanoma. Gemfibrozil 108-119 interferon alpha 1 Homo sapiens 92-95 16507380-10 2005 RESULTS: In this case of a possible drug-drug interaction between IFN alfa 11 MU TIW and gemfibrozil 600 mg BID in a patient undergoing treatment for IFN-induced hypertriglyceridemia, the Naranjo Adverse Drug Reactions (ADR) Probability Scale score was 7 (ie, ADR possibly related to treatment). Gemfibrozil 89-100 interferon alpha 1 Homo sapiens 66-69 16507380-10 2005 RESULTS: In this case of a possible drug-drug interaction between IFN alfa 11 MU TIW and gemfibrozil 600 mg BID in a patient undergoing treatment for IFN-induced hypertriglyceridemia, the Naranjo Adverse Drug Reactions (ADR) Probability Scale score was 7 (ie, ADR possibly related to treatment). Gemfibrozil 89-100 BH3 interacting domain death agonist Homo sapiens 108-111 16507380-10 2005 RESULTS: In this case of a possible drug-drug interaction between IFN alfa 11 MU TIW and gemfibrozil 600 mg BID in a patient undergoing treatment for IFN-induced hypertriglyceridemia, the Naranjo Adverse Drug Reactions (ADR) Probability Scale score was 7 (ie, ADR possibly related to treatment). Gemfibrozil 89-100 interferon alpha 1 Homo sapiens 150-153 16507380-11 2005 CONCLUSIONS: Both IFN and gemfibrozil inhibit the activity of the hepatic enzymes CYP1A2 and CYP2C19. Gemfibrozil 26-37 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 82-88 16507380-11 2005 CONCLUSIONS: Both IFN and gemfibrozil inhibit the activity of the hepatic enzymes CYP1A2 and CYP2C19. Gemfibrozil 26-37 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 93-100 16507380-12 2005 A possible drug-drug interaction between IFN alfa 11 MU TIW and gemfibrozil 600 mg BID was reported in a patient undergoing treatment for IFN-induced hypertriglyceridemia. Gemfibrozil 64-75 interferon alpha 1 Homo sapiens 41-44 16507380-12 2005 A possible drug-drug interaction between IFN alfa 11 MU TIW and gemfibrozil 600 mg BID was reported in a patient undergoing treatment for IFN-induced hypertriglyceridemia. Gemfibrozil 64-75 BH3 interacting domain death agonist Homo sapiens 83-86 16507380-12 2005 A possible drug-drug interaction between IFN alfa 11 MU TIW and gemfibrozil 600 mg BID was reported in a patient undergoing treatment for IFN-induced hypertriglyceridemia. Gemfibrozil 64-75 interferon alpha 1 Homo sapiens 138-141 16226225-1 2005 Fibrates such as bezafibrate, gemfibrozil, clofibric acid, ciprofibrate and fenofibrate, are ligands for peroxisome proliferator-activated receptor alpha (PPARalpha), and are used as therapeutic agents in the treatment of hyperlipidemia. Gemfibrozil 30-41 peroxisome proliferator activated receptor alpha Homo sapiens 105-153 16226225-1 2005 Fibrates such as bezafibrate, gemfibrozil, clofibric acid, ciprofibrate and fenofibrate, are ligands for peroxisome proliferator-activated receptor alpha (PPARalpha), and are used as therapeutic agents in the treatment of hyperlipidemia. Gemfibrozil 30-41 peroxisome proliferator activated receptor alpha Homo sapiens 155-164 16226225-4 2005 Bezafibrate and gemfibrozil displayed a mixed non-competitive inhibition pattern in the glyceraldehyde reduction activity and pure non-competitive inhibition pattern in the benzyl alcohol oxidation activity of AR. Gemfibrozil 16-27 aldo-keto reductase family 1 member B Homo sapiens 210-212 16235714-2 2005 evening dose) and gemfibrozil (600 mg/kg, po twice) for 30 days produced significant decrease in the level of reduced glutathione, superoxide dismutase, catalase and increase in the level of lipid peroxidation and various serum parameters (creatine phosphokinase, lactate dehydrogenase, serum glutamate oxaloacetate transaminase, creatinine, urea and blood urea nitrogen). Gemfibrozil 18-29 catalase Rattus norvegicus 153-161 16042675-0 2005 Coadministration of gemfibrozil and itraconazole has only a minor effect on the pharmacokinetics of the CYP2C9 and CYP3A4 substrate nateglinide. Gemfibrozil 20-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 16042675-0 2005 Coadministration of gemfibrozil and itraconazole has only a minor effect on the pharmacokinetics of the CYP2C9 and CYP3A4 substrate nateglinide. Gemfibrozil 20-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-121 16042675-1 2005 BACKGROUND AND AIMS: Gemfibrozil, and particularly its combination with itraconazole, greatly increases the area under the plasma concentration-time curve [AUC(0, infinity)] and response to the cytochrome P450 (CYP) 2C8 and 3A4 substrate repaglinide. Gemfibrozil 21-32 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 194-219 16042675-2 2005 In vitro, gemfibrozil is a more potent inhibitor of CYP2C9 than of CYP2C8. Gemfibrozil 10-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 16042675-2 2005 In vitro, gemfibrozil is a more potent inhibitor of CYP2C9 than of CYP2C8. Gemfibrozil 10-21 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 67-73 16042675-3 2005 Our aim was to investigate the effects of the gemfibrozil-itraconazole combination on the pharmacokinetics and pharmacodynamics of another meglitinide analogue, nateglinide, which is metabolized by CYP2C9 and CYP3A4. Gemfibrozil 46-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 16042675-3 2005 Our aim was to investigate the effects of the gemfibrozil-itraconazole combination on the pharmacokinetics and pharmacodynamics of another meglitinide analogue, nateglinide, which is metabolized by CYP2C9 and CYP3A4. Gemfibrozil 46-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 209-215 16283275-1 2005 OBJECTIVE: Our objective was to study the effects of gemfibrozil on the pharmacokinetics of pioglitazone and the active compounds, which are all the substrates of CYP2C8 and CYP3A4. Gemfibrozil 53-64 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 163-169 16283275-1 2005 OBJECTIVE: Our objective was to study the effects of gemfibrozil on the pharmacokinetics of pioglitazone and the active compounds, which are all the substrates of CYP2C8 and CYP3A4. Gemfibrozil 53-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 174-180 16176562-10 2005 Gemfibrozil is only a moderate inhibitor of CYP2C8 and does not inhibit CYP3A4; inhibition of CYP-enzymes by parent gemfibrozil alone does not explain its interaction with repaglinide in vivo. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 44-50 16193754-5 2005 Agonists of both PPARalpha (WY14,643 and gemfibrozil) and PPARbeta (bezafibrate) induced luciferase activity, while rosiglitazone, a PPARgamma agonist, was not effective. Gemfibrozil 41-52 peroxisome proliferator-activated receptor alpha b Oncorhynchus mykiss 17-26 16042675-11 2005 The findings suggest that in vivo gemfibrozil, probably due to its metabolites, is a much more potent inhibitor of CYP2C8 than of CYP2C9. Gemfibrozil 34-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 15968640-2 2005 Recently, we demonstrated that administration of the PPAR-alpha agonists gemfibrozil and fenofibrate, inhibit the clinical signs of experimental autoimmune encephalomyelitis (EAE), the animal model of multiple sclerosis (MS). Gemfibrozil 73-84 peroxisome proliferator activated receptor alpha Mus musculus 53-63 15968640-4 2005 Our studies demonstrated that the PPAR-alpha agonists ciprofibrate, fenofibrate, gemfibrozil, and WY 14,643 each inhibited NO production by cytokine-stimulated microglia in a dose-dependent manner. Gemfibrozil 81-92 peroxisome proliferator activated receptor alpha Mus musculus 34-44 16101410-2 2005 The physiological effects of several marketed drugs for the treatment of dyslipidemia (fenofibrate and gemfibrozil) and diabetes (rosiglitazone and pioglitazone) have now been shown to be mediated through PPARalpha and PPARgamma respectively. Gemfibrozil 103-114 peroxisome proliferator activated receptor alpha Homo sapiens 205-214 16101410-2 2005 The physiological effects of several marketed drugs for the treatment of dyslipidemia (fenofibrate and gemfibrozil) and diabetes (rosiglitazone and pioglitazone) have now been shown to be mediated through PPARalpha and PPARgamma respectively. Gemfibrozil 103-114 peroxisome proliferator activated receptor gamma Homo sapiens 219-228 15292370-5 2004 On gemfibrozil, carriers of the LPL N9 allele in VA-HIT had lower levels of large LDL (-32%; P < 0.01) but higher levels of small, dense LDL (+59%; P < 0.003) than did noncarriers. Gemfibrozil 3-14 lipoprotein lipase Homo sapiens 32-35 15998575-1 2005 Eighteen-month-old Sprague-Dawley rats present age-related alterations in lipid and glucose metabolism and are resistant to the effect of PPARalpha-activating hypolipidemic drugs, such as gemfibrozil. Gemfibrozil 188-199 peroxisome proliferator activated receptor alpha Rattus norvegicus 138-147 15998575-4 2005 Although gemfibrozil induced mild effects on hepatic PPARalpha, HNF-4, and LXRalpha, only rosiglitazone significantly reduced plasma TG (59%), glucose (19%), insulin (61%), and leptin (66%), and liver TG (43%), CE (49%), and NEFA (27%). Gemfibrozil 9-20 peroxisome proliferator activated receptor alpha Rattus norvegicus 53-62 16316932-11 2005 Considering the plasma protein binding and IC50 values for OATP1B1, only gemfibrozil appeared to have a potential to cause drug-drug interactions by inhibiting OATP1B1 at clinically relevant concentrations. Gemfibrozil 73-84 solute carrier organic anion transporter family member 1B1 Homo sapiens 160-167 15801938-2 2005 Because gemfibrozil inhibits CYP2C9 in vitro, we studied its effects on the pharmacokinetics and pharmacodynamics of racemic warfarin. Gemfibrozil 8-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 15618549-0 2005 Regulation of human apoA-I by gemfibrozil and fenofibrate through selective peroxisome proliferator-activated receptor alpha modulation. Gemfibrozil 30-41 apolipoprotein A1 Homo sapiens 20-26 15618549-0 2005 Regulation of human apoA-I by gemfibrozil and fenofibrate through selective peroxisome proliferator-activated receptor alpha modulation. Gemfibrozil 30-41 peroxisome proliferator activated receptor alpha Homo sapiens 76-124 15345331-7 2004 GFB increased also by 80% the cortex PPARalpha content in old males. Gemfibrozil 0-3 peroxisome proliferator activated receptor alpha Rattus norvegicus 37-46 15900215-11 2005 Patients who were homozygous for CYP3A5*3 demonstrated greater serum CK levels than patients who were heterozygous for CYP3A5*3, when concomitant lipid-lowering agents were sequentially removed from the analysis (P=0.025 without gemfibrozil, P=0.010 without gemfibrozil and niacin). Gemfibrozil 229-240 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 15900215-11 2005 Patients who were homozygous for CYP3A5*3 demonstrated greater serum CK levels than patients who were heterozygous for CYP3A5*3, when concomitant lipid-lowering agents were sequentially removed from the analysis (P=0.025 without gemfibrozil, P=0.010 without gemfibrozil and niacin). Gemfibrozil 258-269 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 15900286-11 2005 CONCLUSIONS: Gemfibrozil elevates the plasma concentrations of pioglitazone, probably by inhibition of its CYP2C8-mediated metabolism. Gemfibrozil 13-24 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 107-113 15618549-1 2005 OBJECTIVE: The objective of this trial was to study the effects of fenofibrate (FF) and gemfibrozil (GF), the most commonly used fibrates, on high-density lipoprotein (HDL) and apolipoprotein (apo) A-I. Gemfibrozil 88-99 apolipoprotein A1 Homo sapiens 177-201 15771232-8 2005 However, in contrast to the mRNA induction, marked decreases (>60%) in CYP2C8 activity were obtained with gemfibrozil treatment. Gemfibrozil 109-120 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 74-80 15771232-13 2005 Gemfibrozil is also an inducer of CYP3A4, but acts as both an inducer and an inhibitor of CYP2C8. Gemfibrozil 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-40 15771232-13 2005 Gemfibrozil is also an inducer of CYP3A4, but acts as both an inducer and an inhibitor of CYP2C8. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 90-96 15363580-4 2004 CYP2C7 gene expression was dramatically down-regulated in the livers of rats treated for 13 weeks by WY-14,643 (WY; 500 ppm) or gemfibrozil (GEM; 8000 ppm). Gemfibrozil 128-139 cytochrome P450, family 2, subfamily c, polypeptide 7 Rattus norvegicus 0-6 15292370-7 2004 In men with low HDL-C and CHD: 1) the LPL N9 and S291 alleles are more frequent than in CHD-free men with normal HDL-C, whereas the X447 allele is less frequent, and 2) the LPL N9 allele is associated with the LDL subclass response to gemfibrozil. Gemfibrozil 235-246 lipoprotein lipase Homo sapiens 38-41 15292370-7 2004 In men with low HDL-C and CHD: 1) the LPL N9 and S291 alleles are more frequent than in CHD-free men with normal HDL-C, whereas the X447 allele is less frequent, and 2) the LPL N9 allele is associated with the LDL subclass response to gemfibrozil. Gemfibrozil 235-246 lipoprotein lipase Homo sapiens 173-176 15322734-5 2004 In addition, the hypothesis that gemfibrozil-mediated inhibition of P-glycoprotein contributes to the interaction between gemfibrozil and cerivastatin was tested in Caco-2 cells. Gemfibrozil 33-44 ATP binding cassette subfamily B member 1 Homo sapiens 68-82 15194707-0 2004 Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. Gemfibrozil 0-11 solute carrier organic anion transporter family member 1B1 Homo sapiens 44-84 15194707-0 2004 Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. Gemfibrozil 0-11 solute carrier organic anion transporter family member 1B1 Homo sapiens 86-91 15194707-0 2004 Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. Gemfibrozil 0-11 solute carrier organic anion transporter family member 1B1 Homo sapiens 92-99 15194707-0 2004 Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. Gemfibrozil 0-11 solute carrier organic anion transporter family member 1B1 Homo sapiens 100-107 15194707-0 2004 Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 137-143 15194707-0 2004 Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. Gemfibrozil 281-292 solute carrier organic anion transporter family member 1B1 Homo sapiens 44-84 15194707-0 2004 Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. Gemfibrozil 281-292 solute carrier organic anion transporter family member 1B1 Homo sapiens 86-91 15194707-0 2004 Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. Gemfibrozil 281-292 solute carrier organic anion transporter family member 1B1 Homo sapiens 92-99 15194707-0 2004 Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. Gemfibrozil 281-292 solute carrier organic anion transporter family member 1B1 Homo sapiens 100-107 15194707-2 2004 In the present study, we examined the inhibitory effects of GEM and its metabolites, M3 and gemfibrozil 1-O-beta-glucuronide (GEM-1-O-glu), on the uptake of CER by human organic anion transporting polypeptide 2 (OATP2)-expressing cells and its metabolism in cytochrome P450 expression systems. Gemfibrozil 60-63 solute carrier organic anion transporter family member 1B1 Homo sapiens 170-210 15194707-2 2004 In the present study, we examined the inhibitory effects of GEM and its metabolites, M3 and gemfibrozil 1-O-beta-glucuronide (GEM-1-O-glu), on the uptake of CER by human organic anion transporting polypeptide 2 (OATP2)-expressing cells and its metabolism in cytochrome P450 expression systems. Gemfibrozil 60-63 solute carrier organic anion transporter family member 1B1 Homo sapiens 212-217 15194707-3 2004 Uptake studies showed that GEM and GEM-1-O-glu significantly inhibited the OATP2-mediated uptake of CER with IC(50) values of 72 and 24 microM, respectively. Gemfibrozil 27-30 solute carrier organic anion transporter family member 1B1 Homo sapiens 75-80 15194707-5 2004 GEM and GEM-1-O-glu minimally inhibited the CYP3A4-mediated metabolism of CER. Gemfibrozil 0-3 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 44-50 15194707-7 2004 However, considering the possibly concentrated high unbound concentrations of GEM-1-O-glu in the liver and its relatively larger plasma unbound fraction compared with GEM itself, the glucuronide inhibition of the CYP2C8-mediated metabolism of CER appears to be the main mechanism for the clinically relevant drug-drug interaction. Gemfibrozil 78-81 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 213-219 15289377-8 2004 A selective CFTR channel blocker, gemfibrozil, abrogated the protective effect of IPC. Gemfibrozil 34-45 cystic fibrosis transmembrane conductance regulator Mus musculus 12-16 15322734-5 2004 In addition, the hypothesis that gemfibrozil-mediated inhibition of P-glycoprotein contributes to the interaction between gemfibrozil and cerivastatin was tested in Caco-2 cells. Gemfibrozil 122-133 ATP binding cassette subfamily B member 1 Homo sapiens 68-82 15215125-6 2004 Cells made resistant to probenecid and showing a marked overexpression of MRP1 (by Western blot analysis and confocal microscopy) accumulated ciprofloxacin to almost the same extent as did control cells, but efflux was inhibited less by probenecid, gemfibrozil, and MK571. Gemfibrozil 249-260 ATP-binding cassette, sub-family C (CFTR/MRP), member 1 Mus musculus 74-78 15194003-0 2004 PPARalpha agonists clofibrate and gemfibrozil inhibit cell growth, down-regulate hCG and up-regulate progesterone secretions in immortalized human trophoblast cells. Gemfibrozil 34-45 peroxisome proliferator activated receptor alpha Homo sapiens 0-9 15194003-0 2004 PPARalpha agonists clofibrate and gemfibrozil inhibit cell growth, down-regulate hCG and up-regulate progesterone secretions in immortalized human trophoblast cells. Gemfibrozil 34-45 chorionic gonadotropin subunit beta 5 Homo sapiens 81-84 15116058-13 2004 The in vitro results indicate that the maximum gemfibrozil inhibition of rosuvastatin OATP2-mediated uptake was 50%; the inhibition constant for the inhibitory process was 4.0 +/- 1.3 micromol/L. Gemfibrozil 47-58 solute carrier organic anion transporter family member 1B1 Homo sapiens 86-91 15142970-4 2004 PPARalpha is activated by polyunsaturated fatty acids and oxidized derivatives and by lipid-modifying drugs of the fibrate family, including fenofibrate or gemfibrozil. Gemfibrozil 156-167 peroxisome proliferator activated receptor alpha Homo sapiens 0-9 15116058-15 2004 Gemfibrozil inhibition of OATP2-mediated rosuvastatin hepatic uptake may contribute to the mechanism of the drug-drug interaction. Gemfibrozil 0-11 solute carrier organic anion transporter family member 1B1 Homo sapiens 26-31 15100326-3 2004 The PPAR alpha agonists, gemfibrozil, ciprofibrate, and fenofibrate, have an excellent track history as oral agents used to treat hypertriglyceridemia. Gemfibrozil 25-36 peroxisome proliferator activated receptor alpha Homo sapiens 4-14 14698041-5 2004 We tested whether gemfibrozil, fenofibrate, fenofibric acid, and bezafibrate inhibit Pgp in vitro using a calcein acetoxymethylester (calcein-AM) uptake assay and confocal laser scanning microscopy with bodipy-verapamil as substrate in L-MDR1 cells, which overexpress human Pgp. Gemfibrozil 18-29 ATP binding cassette subfamily B member 1 Homo sapiens 85-88 14685799-5 2004 RESULTS: Bezafibrate, fenofibrate and gemfibrozil significantly reduced CYP7A1 mRNA levels. Gemfibrozil 38-49 cytochrome P450 family 7 subfamily A member 1 Homo sapiens 72-78 15100326-6 2004 More importantly, gemfibrozil was shown to shift the cytokine secretion of human T cell lines by inhibiting IFN-gamma and promoting IL-4 secretion. Gemfibrozil 18-29 interferon gamma Homo sapiens 108-117 15100326-6 2004 More importantly, gemfibrozil was shown to shift the cytokine secretion of human T cell lines by inhibiting IFN-gamma and promoting IL-4 secretion. Gemfibrozil 18-29 interleukin 4 Homo sapiens 132-136 15100326-7 2004 These results suggest that PPAR alpha agonists such as gemfibrozil and fenofibrate, may be attractive candidates for use in human inflammatory conditions such as multiple sclerosis. Gemfibrozil 55-66 peroxisome proliferator activated receptor alpha Homo sapiens 27-37 15283301-5 2004 In the presence of gemfibrozil, the CYP- or UGT-mediated metabolism of pitavastatin was inhibited in vitro. Gemfibrozil 19-30 peptidylprolyl isomerase G Homo sapiens 36-39 15283301-5 2004 In the presence of gemfibrozil, the CYP- or UGT-mediated metabolism of pitavastatin was inhibited in vitro. Gemfibrozil 19-30 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 44-47 14555339-0 2003 Metabolism and disposition of gemfibrozil in Wistar and multidrug resistance-associated protein 2-deficient TR- rats. Gemfibrozil 30-41 ATP binding cassette subfamily C member 2 Rattus norvegicus 56-97 14709251-10 2003 Ritonavir partially inhibited induction of PPAR target genes by gemfibrozil and rosiglitazone. Gemfibrozil 64-75 peroxisome proliferator activated receptor alpha Mus musculus 43-47 14555339-2 2003 The roles of multidrug resistance-associated protein (Mrp) 2 deficiency and Mrp3 up-regulation were evaluated on the metabolism and disposition of gemfibrozil. Gemfibrozil 147-158 ATP binding cassette subfamily C member 3 Rattus norvegicus 76-80 14555339-4 2003 Results from in vitro studies in microsomes showed that the hepatic intrinsic clearance (CLint) for the oxidative metabolism of gemfibrozil was slightly higher (1.5-fold) in male TR- rats, which are deficient in Mrp2, than in wild-type Wistar rats, whereas CLint for glucuronidation was similar in both strains. Gemfibrozil 128-139 ATP binding cassette subfamily C member 2 Rattus norvegicus 212-216 14555339-11 2003 Overall, it was demonstrated that alterations in the expression of the transporters Mrp2 and Mrp3 significantly affected the excretion as well as the secondary metabolism and distribution of [14C]gemfibrozil. Gemfibrozil 196-207 ATP binding cassette subfamily C member 2 Rattus norvegicus 84-88 14555339-11 2003 Overall, it was demonstrated that alterations in the expression of the transporters Mrp2 and Mrp3 significantly affected the excretion as well as the secondary metabolism and distribution of [14C]gemfibrozil. Gemfibrozil 196-207 ATP binding cassette subfamily C member 3 Rattus norvegicus 93-97 12935323-6 2003 These data suggest that FLPs and gemfibrozil not only lower lipid intolerance but also reduce diabetic-dyslipidemic complications by activating peroxisome proliferator-activated receptors (PPAR). Gemfibrozil 33-44 peroxisome proliferator activated receptor alpha Rattus norvegicus 144-187 12935323-6 2003 These data suggest that FLPs and gemfibrozil not only lower lipid intolerance but also reduce diabetic-dyslipidemic complications by activating peroxisome proliferator-activated receptors (PPAR). Gemfibrozil 33-44 peroxisome proliferator activated receptor alpha Rattus norvegicus 189-193 12692010-0 2003 Gemfibrozil reduces plasma C-reactive protein levels in abdominally obese men with the atherogenic dyslipidemia of the metabolic syndrome. Gemfibrozil 0-11 C-reactive protein Homo sapiens 27-45 12763281-0 2003 Gemfibrozil reduces release of tumor necrosis factor-alpha in peripheral blood mononuclear cells from healthy subjects and patients with coronary heart disease. Gemfibrozil 0-11 tumor necrosis factor Homo sapiens 31-58 12763281-3 2003 But little is known about the effect of gemfibrozil on tumor necrosis factor (TNF)-alpha secretion in peripheral blood mononuclear cells (PBMC) from patients with coronary heart disease. Gemfibrozil 40-51 tumor necrosis factor Homo sapiens 55-76 12763281-3 2003 But little is known about the effect of gemfibrozil on tumor necrosis factor (TNF)-alpha secretion in peripheral blood mononuclear cells (PBMC) from patients with coronary heart disease. Gemfibrozil 40-51 tumor necrosis factor Homo sapiens 78-88 12763281-11 2003 Gemfibrozil (10(-5),10(-4) mol/l) significantly inhibited TNF-alpha secretion in both groups (P<0.05). Gemfibrozil 0-11 tumor necrosis factor Homo sapiens 58-67 12763281-12 2003 CONCLUSIONS: Our data demonstrated that gemfibrozil reduced release of TNF-alpha in PBMC both from CHD patients and controls. Gemfibrozil 40-51 tumor necrosis factor Homo sapiens 71-80 12746275-5 2003 PPARalpha potentiates fatty acid catabolism in the liver and is the molecular target of the lipid-lowering fibrates (e.g. fenofibrate and gemfibrozil), whereas PPARgamma is essential for adipocyte differentiation and mediates the activity of the insulin-sensitizing thiazolidinediones (e.g. rosiglitazone and pioglitazone). Gemfibrozil 138-149 peroxisome proliferator activated receptor alpha Homo sapiens 0-9 12834305-0 2003 Preparation of antiserum against rat delta6-desaturase and its use to evaluate the desaturase protein levels in rats treated with gemfibrozil, a ligand for peroxisome proliferator-activated receptor alpha. Gemfibrozil 130-141 fatty acid desaturase 2 Rattus norvegicus 37-54 12834305-0 2003 Preparation of antiserum against rat delta6-desaturase and its use to evaluate the desaturase protein levels in rats treated with gemfibrozil, a ligand for peroxisome proliferator-activated receptor alpha. Gemfibrozil 130-141 peroxisome proliferator activated receptor alpha Rattus norvegicus 156-204 12834305-3 2003 Subsequently, the antiserum was used to demonstrate that gemfibrozil, a ligand for peroxisome proliferator-activated receptor alpha, is involved in activating delta6-desaturase gene expression, thereby elevating the protein level and the activity. Gemfibrozil 57-68 peroxisome proliferator activated receptor alpha Rattus norvegicus 83-131 12834305-3 2003 Subsequently, the antiserum was used to demonstrate that gemfibrozil, a ligand for peroxisome proliferator-activated receptor alpha, is involved in activating delta6-desaturase gene expression, thereby elevating the protein level and the activity. Gemfibrozil 57-68 fatty acid desaturase 2 Rattus norvegicus 159-176 12716814-5 2003 Throughout both lower and higher ranges of HDL cholesterol and triglycerides, the rate of new cardiovascular events and the reduction of events with gemfibrozil was greater in subjects with insulin resistance than without, despite the finding that an increase in HDL cholesterol and a decrease in triglycerides with gemfibrozil was less with insulin resistance than without insulin resistance. Gemfibrozil 149-160 insulin Homo sapiens 190-197 12623126-5 2003 Paradoxically, under the same treatment conditions in male rats, liver P450R protein levels decreased after exposure to the PPs Wy-14,643 ([4-chloro-6-(2,3-xylidino)pyrimidynylthio]acetic acid) (WY) or gemfibrozil (GEM). Gemfibrozil 202-213 cytochrome p450 oxidoreductase Rattus norvegicus 71-76 12623126-5 2003 Paradoxically, under the same treatment conditions in male rats, liver P450R protein levels decreased after exposure to the PPs Wy-14,643 ([4-chloro-6-(2,3-xylidino)pyrimidynylthio]acetic acid) (WY) or gemfibrozil (GEM). Gemfibrozil 215-218 cytochrome p450 oxidoreductase Rattus norvegicus 71-76 12623126-6 2003 The down-regulation of the P450R protein was sex- and tissue-specific in that exposure to PPs led to increases in P450R protein in female rat livers [di-n-butyl phthalate (DBP) only] and male rat kidneys (WY, GEM, DBP). Gemfibrozil 209-212 cytochrome p450 oxidoreductase Rattus norvegicus 27-32 14727929-10 2003 The use of combined therapy with an HMG-CoA reductase inhibitor and a fibric acid derivative requires the respect of some rules such as avoiding the prescription in patients with concomitant conditions like renal failure and avoiding the use of gemfibrozil as a fibric acid derivative in such a combination. Gemfibrozil 245-256 3-hydroxy-3-methylglutaryl-CoA reductase Homo sapiens 36-53 12640339-11 2003 In addition, the percentage increment of MCP-1 was higher in patients treated with gemfibrozil than in patients who received bezafibrate. Gemfibrozil 83-94 C-C motif chemokine ligand 2 Homo sapiens 41-46 12640339-13 2003 This finding was associated with increased HDL-C. Fibrates, especially gemfibrozil, increased MCP-1 concentrations. Gemfibrozil 71-82 C-C motif chemokine ligand 2 Homo sapiens 94-99 12433802-0 2002 Gemfibrozil inhibits CYP2C8-mediated cerivastatin metabolism in human liver microsomes. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 21-27 14682608-4 2003 Several CYP enzymes were shown to be principally responsible for the metabolism of gemfibrozil in contrast to other fibrates. Gemfibrozil 83-94 peptidylprolyl isomerase G Homo sapiens 8-11 14682608-5 2003 In the presence of gemfibrozil, a focal point was obtained in Dixon plots, demonstrating that there was inhibition of CYP2C8-, CYP2C9- and CYP3A4-mediated metabolism. Gemfibrozil 19-30 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 118-124 14682608-5 2003 In the presence of gemfibrozil, a focal point was obtained in Dixon plots, demonstrating that there was inhibition of CYP2C8-, CYP2C9- and CYP3A4-mediated metabolism. Gemfibrozil 19-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 14682608-5 2003 In the presence of gemfibrozil, a focal point was obtained in Dixon plots, demonstrating that there was inhibition of CYP2C8-, CYP2C9- and CYP3A4-mediated metabolism. Gemfibrozil 19-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 139-145 14682608-6 2003 We propose that the increase of plasma concentration caused by co-administration of gemfibrozil and statins is at least partially due to CYP-mediated inhibition. Gemfibrozil 84-95 peptidylprolyl isomerase G Homo sapiens 137-140 12496749-7 2002 Gemfibrozil increased the AUC(0-infinity) of cerivastatin lactone, on average, to 440% (94% to 594%; P =.0024) and that of metabolite M-1 to 435% (216% to 802%; P =.0002) of the control (placebo) values, whereas the AUC(0-24) of metabolite M-23 was decreased to 22% (11% to 74%; P =.0017). Gemfibrozil 0-11 myoregulin Homo sapiens 134-137 12496749-8 2002 CONCLUSIONS: Gemfibrozil greatly increases plasma concentrations of cerivastatin, cerivastatin lactone, and metabolite M-1, whereas the level of metabolite M-23 is markedly reduced by gemfibrozil. Gemfibrozil 13-24 myoregulin Homo sapiens 119-122 12496749-9 2002 Gemfibrozil therefore inhibits the formation of M-23, which is thought to be dependent on CYP2C8. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 90-96 14650362-6 2003 The cytochrome P (CYP) enzyme CYP2C9 plays a major role in the metabolism of gemfibrozil. Gemfibrozil 77-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 12456232-2 2002 OBJECTIVES: To determine the efficacy of gemfibrozil in subjects with varying levels of glucose tolerance or hyperinsulinemia and to examine the association between diabetes status and glucose and insulin levels and risk of cardiovascular outcomes. Gemfibrozil 41-52 insulin Homo sapiens 114-121 12456232-10 2002 Among individuals without diabetes, gemfibrozil was most efficacious for those in the highest fasting plasma insulin level quartile (risk reduction, 35%; P =.04). Gemfibrozil 36-47 insulin Homo sapiens 109-116 12456232-11 2002 CONCLUSION: In men with CHD and a low high-density lipoprotein cholesterol level, gemfibrozil use was associated with a reduction in major cardiovascular events in persons with diabetes and in nondiabetic subjects with a high fasting plasma insulin level. Gemfibrozil 82-93 insulin Homo sapiens 241-248 12433802-5 2002 The present in vitro findings suggest that inhibition of CYP2C8 activity by gemfibrozil at least partially explains the interaction between gemfibrozil and cerivastatin. Gemfibrozil 76-87 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 57-63 12433802-5 2002 The present in vitro findings suggest that inhibition of CYP2C8 activity by gemfibrozil at least partially explains the interaction between gemfibrozil and cerivastatin. Gemfibrozil 140-151 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 57-63 12433802-7 2002 Inhibition of CYP2C8-mediated metabolism by gemfibrozil warrants further in vivo exploration. Gemfibrozil 44-55 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 14-20 12433802-4 2002 Additionally, gemfibrozil inhibited paclitaxel 6 alpha-hydroxylation [K(i) (IC(50)) = 75 micro M (91 micro M)], a CYP2C8 marker reaction, but did not inhibit testosterone 6 beta-hydroxylation (CYP3A4). Gemfibrozil 14-25 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 114-120 12433802-4 2002 Additionally, gemfibrozil inhibited paclitaxel 6 alpha-hydroxylation [K(i) (IC(50)) = 75 micro M (91 micro M)], a CYP2C8 marker reaction, but did not inhibit testosterone 6 beta-hydroxylation (CYP3A4). Gemfibrozil 14-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 193-199 12244038-1 2002 Gemfibrozil, a lipid-lowering drug, inhibited cytokine-induced production of NO and the expression of inducible nitric-oxide synthase (iNOS) in human U373MG astroglial cells and primary astrocytes. Gemfibrozil 0-11 nitric oxide synthase 2 Homo sapiens 102-133 12244038-4 2002 Since gemfibrozil is known to activate peroxisome proliferator-activated receptor-alpha (PPAR-alpha), we investigated the role of PPAR-alpha in gemfibrozil-mediated inhibition of iNOS. Gemfibrozil 144-155 nitric oxide synthase 2 Homo sapiens 179-183 12244038-1 2002 Gemfibrozil, a lipid-lowering drug, inhibited cytokine-induced production of NO and the expression of inducible nitric-oxide synthase (iNOS) in human U373MG astroglial cells and primary astrocytes. Gemfibrozil 0-11 nitric oxide synthase 2 Homo sapiens 135-139 12244038-5 2002 Gemfibrozil induced peroxisome proliferator-responsive element (PPRE)-dependent luciferase activity, which was inhibited by the expression of DeltahPPAR-alpha, the dominant-negative mutant of human PPAR-alpha. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Homo sapiens 148-158 12244038-2 2002 Similar to gemfibrozil, clofibrate, another fibrate drug, also inhibited the expression of iNOS. Gemfibrozil 11-22 nitric oxide synthase 2 Homo sapiens 91-95 12244038-6 2002 However, DeltahPPAR-alpha was unable to abrogate gemfibrozil-mediated inhibition of iNOS suggesting that gemfibrozil inhibits iNOS independent of PPAR-alpha. Gemfibrozil 105-116 nitric oxide synthase 2 Homo sapiens 126-130 12244038-3 2002 Inhibition of human iNOS promoter-driven luciferase activity by gemfibrozil in cytokine-stimulated U373MG astroglial cells suggests that this compound inhibits the transcription of iNOS. Gemfibrozil 64-75 nitric oxide synthase 2 Homo sapiens 20-24 12244038-9 2002 Interestingly, gemfibrozil strongly inhibited the activation of NF-kappaB, AP-1, and C/EBPbeta but not that of GAS in cytokine-stimulated astroglial cells. Gemfibrozil 15-26 nuclear factor kappa B subunit 1 Homo sapiens 64-73 12244038-3 2002 Inhibition of human iNOS promoter-driven luciferase activity by gemfibrozil in cytokine-stimulated U373MG astroglial cells suggests that this compound inhibits the transcription of iNOS. Gemfibrozil 64-75 nitric oxide synthase 2 Homo sapiens 181-185 12244038-9 2002 Interestingly, gemfibrozil strongly inhibited the activation of NF-kappaB, AP-1, and C/EBPbeta but not that of GAS in cytokine-stimulated astroglial cells. Gemfibrozil 15-26 JunB proto-oncogene, AP-1 transcription factor subunit Homo sapiens 75-79 12244038-4 2002 Since gemfibrozil is known to activate peroxisome proliferator-activated receptor-alpha (PPAR-alpha), we investigated the role of PPAR-alpha in gemfibrozil-mediated inhibition of iNOS. Gemfibrozil 6-17 peroxisome proliferator activated receptor alpha Homo sapiens 39-87 12244038-9 2002 Interestingly, gemfibrozil strongly inhibited the activation of NF-kappaB, AP-1, and C/EBPbeta but not that of GAS in cytokine-stimulated astroglial cells. Gemfibrozil 15-26 CCAAT enhancer binding protein beta Homo sapiens 85-94 12244038-4 2002 Since gemfibrozil is known to activate peroxisome proliferator-activated receptor-alpha (PPAR-alpha), we investigated the role of PPAR-alpha in gemfibrozil-mediated inhibition of iNOS. Gemfibrozil 6-17 peroxisome proliferator activated receptor alpha Homo sapiens 89-99 12244038-10 2002 These results suggest that gemfibrozil inhibits the induction of iNOS probably by inhibiting the activation of NF-kappaB, AP-1, and C/EBPbeta and that gemfibrozil, a prescribed drug for humans, may further find its therapeutic use in neuroinflammatory diseases. Gemfibrozil 27-38 nitric oxide synthase 2 Homo sapiens 65-69 12386136-2 2002 Gemfibrozil (GFZ) modestly affected the formation of beta-oxidative products and CYP3A4-mediated oxidative metabolites of simvastatin hydroxy acid (SVA) but markedly inhibited the glucuronidation-mediated lactonization of SVA and the glucuronidation of a beta-oxidation product (IC(50) approximately 50 and 15 microM, respectively). Gemfibrozil 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 12244038-10 2002 These results suggest that gemfibrozil inhibits the induction of iNOS probably by inhibiting the activation of NF-kappaB, AP-1, and C/EBPbeta and that gemfibrozil, a prescribed drug for humans, may further find its therapeutic use in neuroinflammatory diseases. Gemfibrozil 27-38 nuclear factor kappa B subunit 1 Homo sapiens 111-120 12244038-10 2002 These results suggest that gemfibrozil inhibits the induction of iNOS probably by inhibiting the activation of NF-kappaB, AP-1, and C/EBPbeta and that gemfibrozil, a prescribed drug for humans, may further find its therapeutic use in neuroinflammatory diseases. Gemfibrozil 27-38 JunB proto-oncogene, AP-1 transcription factor subunit Homo sapiens 122-126 12244038-10 2002 These results suggest that gemfibrozil inhibits the induction of iNOS probably by inhibiting the activation of NF-kappaB, AP-1, and C/EBPbeta and that gemfibrozil, a prescribed drug for humans, may further find its therapeutic use in neuroinflammatory diseases. Gemfibrozil 27-38 CCAAT enhancer binding protein beta Homo sapiens 132-141 12386136-2 2002 Gemfibrozil (GFZ) modestly affected the formation of beta-oxidative products and CYP3A4-mediated oxidative metabolites of simvastatin hydroxy acid (SVA) but markedly inhibited the glucuronidation-mediated lactonization of SVA and the glucuronidation of a beta-oxidation product (IC(50) approximately 50 and 15 microM, respectively). Gemfibrozil 13-16 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 12386136-6 2002 While GFZ is not an inhibitor of CYP3A4, it is a competitive inhibitor (K(i) = 87 microM) of CYP2C8, a major catalyzing enzyme for CVA oxidation. Gemfibrozil 6-9 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 93-99 12386136-7 2002 These results suggest that 1) the pharmacokinetic interaction observed between GFZ and statins was not likely mediated by the inhibitory effect of GFZ on the beta-oxidation, but rather by its effect primarily on the glucuronidation and non-CYP3A-mediated oxidation of statin hydroxy acids, and 2) there is a potential difference between fibrates in their ability to affect the pharmacokinetics of statins, and among statins in their susceptibility to metabolic interactions with GFZ in humans. Gemfibrozil 79-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 240-245 12172398-0 2002 Influences of the PPAR alpha-L162V polymorphism on plasma HDL(2)-cholesterol response of abdominally obese men treated with gemfibrozil. Gemfibrozil 124-135 peroxisome proliferator activated receptor alpha Homo sapiens 18-28 12204791-0 2002 Effect of gemfibrozil on apolipoprotein B secretion and diacylglycerol acyltransferase activity in human hepatoblastoma (HepG2) cells. Gemfibrozil 10-21 apolipoprotein B Homo sapiens 25-41 12204791-1 2002 The mechanism of action of a widely used drug gemfibrozil to reduce triglycerides (TG) and apolipoprotein B (apo B) is incompletely understood. Gemfibrozil 46-57 apolipoprotein B Homo sapiens 91-107 12204791-1 2002 The mechanism of action of a widely used drug gemfibrozil to reduce triglycerides (TG) and apolipoprotein B (apo B) is incompletely understood. Gemfibrozil 46-57 apolipoprotein B Homo sapiens 109-114 12204791-2 2002 Using human hepatoblastoma (HepG2) cells, we examined the effect of gemfibrozil on apo B secretion and TG synthesis catalyzed by diacylglycerol acyltransferase (DGAT), primary processes associated with the secretion of LDL. Gemfibrozil 68-79 apolipoprotein B Homo sapiens 83-88 12204791-2 2002 Using human hepatoblastoma (HepG2) cells, we examined the effect of gemfibrozil on apo B secretion and TG synthesis catalyzed by diacylglycerol acyltransferase (DGAT), primary processes associated with the secretion of LDL. Gemfibrozil 68-79 diacylglycerol O-acyltransferase 1 Homo sapiens 129-159 12204791-2 2002 Using human hepatoblastoma (HepG2) cells, we examined the effect of gemfibrozil on apo B secretion and TG synthesis catalyzed by diacylglycerol acyltransferase (DGAT), primary processes associated with the secretion of LDL. Gemfibrozil 68-79 diacylglycerol O-acyltransferase 1 Homo sapiens 161-165 12204791-3 2002 Gemfibrozil significantly decreased apo B secretion by HepG2 cells. Gemfibrozil 0-11 apolipoprotein B Homo sapiens 36-41 12204791-4 2002 It decreased oleate-induced stimulation of apo B secretion, suggesting that gemfibrozil-mediated inhibition of apo B secretion may be dependent on the synthesis of TG catalyzed by DGAT. Gemfibrozil 76-87 apolipoprotein B Homo sapiens 43-48 12204791-4 2002 It decreased oleate-induced stimulation of apo B secretion, suggesting that gemfibrozil-mediated inhibition of apo B secretion may be dependent on the synthesis of TG catalyzed by DGAT. Gemfibrozil 76-87 apolipoprotein B Homo sapiens 111-116 12204791-4 2002 It decreased oleate-induced stimulation of apo B secretion, suggesting that gemfibrozil-mediated inhibition of apo B secretion may be dependent on the synthesis of TG catalyzed by DGAT. Gemfibrozil 76-87 diacylglycerol O-acyltransferase 1 Homo sapiens 180-184 12204791-5 2002 Pre-incubation of HepG2 cells with gemfibrozil (200-400 micromol/l for 48 h) significantly inhibited microsomal DGAT activity. Gemfibrozil 35-46 diacylglycerol O-acyltransferase 1 Homo sapiens 112-116 12204791-6 2002 When added directly to the DGAT assay system containing control microsomes, gemfibrozil significantly inhibited the activity of DGAT by 14-25%. Gemfibrozil 76-87 diacylglycerol O-acyltransferase 1 Homo sapiens 27-31 12204791-6 2002 When added directly to the DGAT assay system containing control microsomes, gemfibrozil significantly inhibited the activity of DGAT by 14-25%. Gemfibrozil 76-87 diacylglycerol O-acyltransferase 1 Homo sapiens 128-132 12204791-8 2002 The data indicate that gemfibrozil inhibits DGAT activity resulting in decreased synthesis of TG and its availability for apo B lipidation rendering it susceptible to intracellular apo B degradation leading to the decreased secretion. Gemfibrozil 23-34 diacylglycerol O-acyltransferase 1 Homo sapiens 44-48 12204791-8 2002 The data indicate that gemfibrozil inhibits DGAT activity resulting in decreased synthesis of TG and its availability for apo B lipidation rendering it susceptible to intracellular apo B degradation leading to the decreased secretion. Gemfibrozil 23-34 apolipoprotein B Homo sapiens 122-127 12204791-8 2002 The data indicate that gemfibrozil inhibits DGAT activity resulting in decreased synthesis of TG and its availability for apo B lipidation rendering it susceptible to intracellular apo B degradation leading to the decreased secretion. Gemfibrozil 23-34 apolipoprotein B Homo sapiens 181-186 12204791-9 2002 These in-vitro data suggest a novel additional mechanism by which gemfibrozil lowers plasma TG and atherogenic apo B lipoproteins in dyslipidemic patients. Gemfibrozil 66-77 apolipoprotein B Homo sapiens 111-116 12147264-5 2002 Non-cross-reacting ACS1, ACS4, and ACS5 peptide antibodies showed that ACS4 was the only ACS isoform present in peroxisomes isolated from livers of gemfibrozil-treated rats. Gemfibrozil 148-159 acyl-CoA synthetase long-chain family member 4 Rattus norvegicus 71-75 12104084-13 2002 Lag time, the principle measure of lipoprotein susceptibility to oxidation, was decreased by gemfibrozil. Gemfibrozil 93-104 stathmin 1 Homo sapiens 0-3 12104084-15 2002 CONCLUSION: These data support the notion that gemfibrozil increases the proportion of polyunsaturated fatty acids in plasma lipids and that this increase is associated with an increase in lipoprotein oxidative susceptibility as measured by lag time in hypertriglyceridemia. Gemfibrozil 47-58 stathmin 1 Homo sapiens 241-244 12123487-8 2002 The results of this study suggest that the ACE I/D polymorphism influences the effect of gemfibrozil on plasma HDL-C levels. Gemfibrozil 89-100 angiotensin I converting enzyme Homo sapiens 43-46 12172398-1 2002 PURPOSE: The effect of gemfibrozil is mediated by the activation of peroxisome proliferator-activated receptor alpha (PPAR alpha). Gemfibrozil 23-34 peroxisome proliferator activated receptor alpha Homo sapiens 68-116 12172398-1 2002 PURPOSE: The effect of gemfibrozil is mediated by the activation of peroxisome proliferator-activated receptor alpha (PPAR alpha). Gemfibrozil 23-34 peroxisome proliferator activated receptor alpha Homo sapiens 118-128 12172398-2 2002 The objective of this study was to determine whether the lipid response to gemfibrozil therapy is influenced by the PPAR alpha-L162V polymorphism. Gemfibrozil 75-86 peroxisome proliferator activated receptor alpha Homo sapiens 116-126 12172398-5 2002 CONCLUSION: These results suggest that the HDL(2)-C response to gemfibrozil is modulated by the PPAR alpha-L162V polymorphism. Gemfibrozil 64-75 peroxisome proliferator activated receptor alpha Homo sapiens 96-106 12023536-5 2002 In dog and human liver microsomes, GFZ exerted a minimal inhibitory effect on CYP3A-mediated SVA oxidation, but did inhibit SVA glucuronidation. Gemfibrozil 35-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-83 12163133-7 2002 This suggests that ciprofibrate and gemfibrozil induce a different conformation to the receptor-PGC-1 and receptor-CBP complex, respectively. Gemfibrozil 36-47 PPARG coactivator 1 alpha Homo sapiens 96-101 12163133-7 2002 This suggests that ciprofibrate and gemfibrozil induce a different conformation to the receptor-PGC-1 and receptor-CBP complex, respectively. Gemfibrozil 36-47 CREB binding protein Homo sapiens 115-118 12065698-0 2002 Effect of albumin and cytosol on enzyme kinetics of tolbutamide hydroxylation and on inhibition of CYP2C9 by gemfibrozil in human liver microsomes. Gemfibrozil 109-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 12065698-6 2002 The K(i) (6 microM) of gemfibrozil for CYP2C9, calculated using total drug concentrations, was increased by Hlc (8 microM), Hsa (40 microM), or both (72 microM). Gemfibrozil 23-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 12023536-8 2002 Among six human UGT isozymes tested, UGT1A1 and 1A3 were capable of catalyzing the glucuronidation of both GFZ and SVA. Gemfibrozil 107-110 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 16-19 12023536-8 2002 Among six human UGT isozymes tested, UGT1A1 and 1A3 were capable of catalyzing the glucuronidation of both GFZ and SVA. Gemfibrozil 107-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-51 12023536-9 2002 Further studies conducted in human liver microsomes with atorvastatin (AVA) showed that, as with SVA, GFZ was a less potent inhibitor of the CYP3A4-mediated oxidation of this drug than its glucuronidation. Gemfibrozil 102-105 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 141-147 12023536-10 2002 However, with cerivastatin (CVA), the glucuronidation as well as the CYP2C8- and CYP3A4-mediated oxidation pathways were much more susceptible to inhibition by GFZ than was observed with SVA or AVA. Gemfibrozil 160-163 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 69-75 12023536-10 2002 However, with cerivastatin (CVA), the glucuronidation as well as the CYP2C8- and CYP3A4-mediated oxidation pathways were much more susceptible to inhibition by GFZ than was observed with SVA or AVA. Gemfibrozil 160-163 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 11858378-8 2002 At the end of the fourth month, gemfibrozil reduced total cholesterol by 34%, LDL by 30%, apo B by 21% and triglycerides by 53% (p < 0.05). Gemfibrozil 32-43 apolipoprotein B Homo sapiens 90-95 12025914-10 2002 Fibrates (with the exception of gemfibrozil) also consistently lower plasma fibrinogen levels. Gemfibrozil 32-43 fibrinogen beta chain Homo sapiens 76-86 11947905-5 2002 In low and high glucose media, gemfibrozil (100 micromol/l) reduced total PG production in the presence of TGF-beta 1, which was associated with a decrease in the apparent size of PGs. Gemfibrozil 31-42 transforming growth factor beta 1 Homo sapiens 107-117 11947905-6 2002 Gemfibrozil and another PPAR-alpha ligand, WY-14643, significantly inhibited basal and TGF-beta1 stimulated GAG synthesis. Gemfibrozil 0-11 transforming growth factor beta 1 Homo sapiens 87-96 11947915-3 2002 The reduction of total cholesterol (TC), betaquant LDL and LDL cholesterol (LDL-C) was significantly greater in patients treated with pravastatin than with gemfibrozil. Gemfibrozil 156-167 component of oligomeric golgi complex 2 Homo sapiens 59-74 11947915-3 2002 The reduction of total cholesterol (TC), betaquant LDL and LDL cholesterol (LDL-C) was significantly greater in patients treated with pravastatin than with gemfibrozil. Gemfibrozil 156-167 component of oligomeric golgi complex 2 Homo sapiens 76-81 12047397-6 2002 CONCLUSIONS: Gemfibrozil does not affect insulin sensitivity to glucose or fat metabolism in type 2 diabetes but enhances the lowering of plasma NEFA concentrations by insulin, probably by reducing hepatic fatty acid synthesis. Gemfibrozil 13-24 insulin Homo sapiens 168-175 11942772-9 2002 CONCLUSION: Fenofibrate and gemfibrozil induced similar variations from baseline values in triglycerides, HDL cholesterol, apolipoprotein B, and fibrinogen, but the decreases in total and LDL cholesterol levels were greater with fenofibrate, in this group of patients with primary hyperlipidemia. Gemfibrozil 28-39 apolipoprotein B Homo sapiens 123-139 11942772-9 2002 CONCLUSION: Fenofibrate and gemfibrozil induced similar variations from baseline values in triglycerides, HDL cholesterol, apolipoprotein B, and fibrinogen, but the decreases in total and LDL cholesterol levels were greater with fenofibrate, in this group of patients with primary hyperlipidemia. Gemfibrozil 28-39 fibrinogen beta chain Homo sapiens 145-155 11947915-6 2002 Both gemfibrozil and pravastatin reduced the concentrations of trigylceride-rich Lp-Bc (-12.2 and -13.3%, respectively) and Lp-A-II;B;C;D;E (-19 and -12.7%, respectively) particles and their characteristic apoC-III constituent (-10.0 and -7.0%, respectively). Gemfibrozil 5-16 apolipoprotein C3 Homo sapiens 206-214 11947915-8 2002 Both pravastatin and gemfibrozil significantly increased the levels of apoA-I and, with both drugs, the elevated concentrations of apoA-I were due to significantly increased levels of Lp-A-I;A-II particles. Gemfibrozil 21-32 apolipoprotein A1 Homo sapiens 71-77 11947915-8 2002 Both pravastatin and gemfibrozil significantly increased the levels of apoA-I and, with both drugs, the elevated concentrations of apoA-I were due to significantly increased levels of Lp-A-I;A-II particles. Gemfibrozil 21-32 apolipoprotein A1 Homo sapiens 131-137 12047397-0 2002 Effects of gemfibrozil on insulin resistance to fat metabolism in subjects with type 2 diabetes and hypertriglyceridaemia. Gemfibrozil 11-22 insulin Homo sapiens 26-33 12047397-1 2002 AIM: To examine whether lowering of plasma triglyceride concentrations using the fibrate peroxisome proliferator-activated receptor (PPAR)alpha agonist gemfibrozil would influence insulin sensitivity to various aspects of intermediary metabolism among subjects with type 2 diabetes mellitus. Gemfibrozil 152-163 insulin Homo sapiens 180-187 11914252-2 2002 METHODS AND RESULTS: We investigated the association between the leucine 162 to valine (L162V) polymorphism and a G to C transversion in intron 7 of the PPARalpha gene and progression of atherosclerosis in the Lopid Coronary Angiography Trial (LOCAT), a trial examining the effect of gemfibrozil treatment on progression of atherosclerosis after bypass surgery and on risk of IHD in the second Northwick Park Heart Study (NPHS2), a prospective study of healthy middle-aged men in the United Kingdom. Gemfibrozil 284-295 peroxisome proliferator activated receptor alpha Homo sapiens 153-162 11694861-8 2001 RESULTS: After three months, it was observed that under the effect of gemfibrozil, serum triglyceride level was significantly decreased (from median: 3.46 mmol/l quartiles: q1=2.92 q3=5.28 to median 2.20 mmol/l quartiles: q1=1.79 q3=3.65; p<0.001) while protective high-density lipoprotein (from 1.02 +/- 0.22 mmol/l to 1.13 +/- 0.28 mmol/l; p=0.05) and apolipoprotein A(1) (from 1.56 +/- 0.33 g/l to 1.72 +/- 0.29; p<0.01) levels were significantly increased. Gemfibrozil 70-81 apolipoprotein A1 Homo sapiens 357-376 11602509-0 2001 Gemfibrozil is a potent inhibitor of human cytochrome P450 2C9. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-62 11602509-3 2001 Gemfibrozil strongly and competitively inhibited CYP2C9 activity, with a K(i) (IC(50)) value of 5.8 (9.6) microM. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11602509-4 2001 In addition, gemfibrozil exhibited somewhat smaller inhibitory effects on CYP2C19 and CYP1A2 activities, with K(i) (IC(50)) values of 24 (47) microM and 82 (136) microM, respectively. Gemfibrozil 13-24 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 74-81 11602509-4 2001 In addition, gemfibrozil exhibited somewhat smaller inhibitory effects on CYP2C19 and CYP1A2 activities, with K(i) (IC(50)) values of 24 (47) microM and 82 (136) microM, respectively. Gemfibrozil 13-24 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 86-92 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Gemfibrozil 15-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Gemfibrozil 15-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Gemfibrozil 140-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Gemfibrozil 140-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 11602509-8 2001 Gemfibrozil may also impair clearance of CYP2C19 and CYP1A2 substrates, but inhibition of other CYP isoforms is unlikely. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 41-48 11602509-8 2001 Gemfibrozil may also impair clearance of CYP2C19 and CYP1A2 substrates, but inhibition of other CYP isoforms is unlikely. Gemfibrozil 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 53-59 11719730-1 2001 OBJECTIVE: Our objective was to study the effects of gemfibrozil on the pharmacokinetics and pharmacodynamics of glimepiride, a new sulfonylurea antidiabetic drug and a substrate of cytochrome P4502C9 (CYP2C9). Gemfibrozil 53-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-200 11712864-6 2001 PPAR-alpha activators (gemfibrozil) decrease the risk of coronary heart disease in patients with normal LDL-cholesterol and low HDL-cholesterol (VA-HIT) and they slow the progression of premature coronary atherosclerosis (BECAIT) (bezafibrate), particularly in patients with type 2 diabetes (DAIS) (fenofibrate). Gemfibrozil 23-34 peroxisome proliferator activated receptor alpha Homo sapiens 0-10 11816722-5 2001 Gemfibrozil inhibited basal PAI-1 expression by 23% (p = ns) and TGF-beta-induced PAI-1 expression by 52% (p = 0.017) whereas t-PA and total protein synthesis was not affected. Gemfibrozil 0-11 serpin family E member 1 Homo sapiens 28-33 11816722-5 2001 Gemfibrozil inhibited basal PAI-1 expression by 23% (p = ns) and TGF-beta-induced PAI-1 expression by 52% (p = 0.017) whereas t-PA and total protein synthesis was not affected. Gemfibrozil 0-11 transforming growth factor beta 1 Homo sapiens 65-73 11816722-5 2001 Gemfibrozil inhibited basal PAI-1 expression by 23% (p = ns) and TGF-beta-induced PAI-1 expression by 52% (p = 0.017) whereas t-PA and total protein synthesis was not affected. Gemfibrozil 0-11 serpin family E member 1 Homo sapiens 82-87 11816722-6 2001 Changes in PAI-1 protein accumulation reflected PAI-1 gene expression attributable to modulation of half-life of PAI-1 mRNA by gemfibrozil. Gemfibrozil 127-138 serpin family E member 1 Homo sapiens 11-16 11816722-8 2001 Gemfibrozil specifically attenuates TGF-beta-induced PAI-1 expression in human arterial smooth muscle cells. Gemfibrozil 0-11 transforming growth factor beta 1 Homo sapiens 36-44 11816722-8 2001 Gemfibrozil specifically attenuates TGF-beta-induced PAI-1 expression in human arterial smooth muscle cells. Gemfibrozil 0-11 serpin family E member 1 Homo sapiens 53-58 11385362-6 2001 Gemfibrozil effectively lowered concentrations of plasma lipid, apolipoprotein (apo) B, and apo E. Gemfibrozil 0-11 apolipoprotein B Homo sapiens 64-86 11780395-6 2001 PDGF-B detected by immuno-histochemistry and RT-PCR analysis showed that the release of PDGF-B was inhibited by simvastatin and gemfibrozil after de-endothelialization. Gemfibrozil 128-139 platelet-derived growth factor subunit B Oryctolagus cuniculus 0-6 11780395-6 2001 PDGF-B detected by immuno-histochemistry and RT-PCR analysis showed that the release of PDGF-B was inhibited by simvastatin and gemfibrozil after de-endothelialization. Gemfibrozil 128-139 platelet-derived growth factor subunit B Oryctolagus cuniculus 88-94 11522369-4 2001 Chronic exposure to the PP WY-14,643 (WY) and gemfibrozil (GEM), but not di-n-butyl phthalate (DBP), led to decreases in ES-4 in male rat livers. Gemfibrozil 46-57 carboxylesterase 1C Mus musculus 121-125 11522369-4 2001 Chronic exposure to the PP WY-14,643 (WY) and gemfibrozil (GEM), but not di-n-butyl phthalate (DBP), led to decreases in ES-4 in male rat livers. Gemfibrozil 59-62 carboxylesterase 1C Mus musculus 121-125 11551527-0 2001 Increase in hepatic expression of SREBP-2 by gemfibrozil administration to rats. Gemfibrozil 45-56 sterol regulatory element binding transcription factor 2 Rattus norvegicus 34-41 11551527-4 2001 As a part of a compensatory homeostatic response, we report for the first time that gemfibrozil administration to rats increased the hepatic sterol regulatory element binding protein-2 (SREBP-2) mRNA (2.9-fold) and mature protein (2.2-fold) levels. Gemfibrozil 84-95 sterol regulatory element binding transcription factor 2 Rattus norvegicus 141-184 11551527-4 2001 As a part of a compensatory homeostatic response, we report for the first time that gemfibrozil administration to rats increased the hepatic sterol regulatory element binding protein-2 (SREBP-2) mRNA (2.9-fold) and mature protein (2.2-fold) levels. Gemfibrozil 84-95 sterol regulatory element binding transcription factor 2 Rattus norvegicus 186-193 11551527-5 2001 An early increase in the transcriptional activity of SREBP-2 elicited by gemfibrozil administration might be responsible for the observed changes in HMG-CoA reductase, phosphatidate phosphohydrolase, and SREBP-2 expression. Gemfibrozil 73-84 sterol regulatory element binding transcription factor 2 Rattus norvegicus 53-60 11551527-5 2001 An early increase in the transcriptional activity of SREBP-2 elicited by gemfibrozil administration might be responsible for the observed changes in HMG-CoA reductase, phosphatidate phosphohydrolase, and SREBP-2 expression. Gemfibrozil 73-84 sterol regulatory element binding transcription factor 2 Rattus norvegicus 204-211 11385362-6 2001 Gemfibrozil effectively lowered concentrations of plasma lipid, apolipoprotein (apo) B, and apo E. Gemfibrozil 0-11 apolipoprotein E Homo sapiens 92-97 11385362-8 2001 The surface-to-core ratio and apo E/apo B ratio of VLDL particles were increased after gemfibrozil treatment. Gemfibrozil 87-98 apolipoprotein E Homo sapiens 30-35 11385362-8 2001 The surface-to-core ratio and apo E/apo B ratio of VLDL particles were increased after gemfibrozil treatment. Gemfibrozil 87-98 apolipoprotein B Homo sapiens 36-41 11474570-5 2001 PPAR-alpha activators (fibrates) improve plasma lipid levels and decrease CHD risk in patients with low HDL-cholesterol (gemfibrozil). Gemfibrozil 121-132 peroxisome proliferator activated receptor alpha Homo sapiens 0-10 11248139-9 2001 In gemfibrozil treated rats and hamsters, a decrease in alpha-tocopherol content and an increase in DT-diaphorase activity were observed. Gemfibrozil 3-14 NAD(P)H quinone dehydrogenase 1 Rattus norvegicus 100-113 11159800-4 2001 Intrinsic clearance values for human UGT1A1 and UGT2B7 substrates were an order of magnitude higher in DLM than in HLM (e.g., gemfibrozil: 31 microl/min/mg versus 3.0 microl/min/mg; ketoprofen: 2.4 microl/min/mg versus 0.2 microl/min/mg). Gemfibrozil 126-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 11159800-4 2001 Intrinsic clearance values for human UGT1A1 and UGT2B7 substrates were an order of magnitude higher in DLM than in HLM (e.g., gemfibrozil: 31 microl/min/mg versus 3.0 microl/min/mg; ketoprofen: 2.4 microl/min/mg versus 0.2 microl/min/mg). Gemfibrozil 126-137 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 48-54 11159800-4 2001 Intrinsic clearance values for human UGT1A1 and UGT2B7 substrates were an order of magnitude higher in DLM than in HLM (e.g., gemfibrozil: 31 microl/min/mg versus 3.0 microl/min/mg; ketoprofen: 2.4 microl/min/mg versus 0.2 microl/min/mg). Gemfibrozil 126-137 oxysterol binding protein 2 Homo sapiens 115-118 14728015-9 2001 Fibric acid derivatives and HMG-CoA reductase inhibitor therapy can be combined with care, provided that gemfibrozil is avoided, fibric acid derivatives are given in the mornings and shorter half -life HMG-CoA reductase inhibitors are used at night. Gemfibrozil 105-116 3-hydroxy-3-methylglutaryl-CoA reductase Homo sapiens 28-45 10657576-5 2000 Gemfibrozil treatment did not affect plasma fibrinolysis or fibrinogen levels, despite marked reduction of plasma triglycerides and improvement of the insulin sensitivity associated with triglyceride normalization. Gemfibrozil 0-11 insulin Homo sapiens 151-158 11120871-5 2000 Some evidence also indicates that APO E2 individuals are more likely to respond favorably to gemfibrozil and cholestyramine. Gemfibrozil 93-104 apolipoprotein E Homo sapiens 34-40 10789612-8 2000 Effects on plasma fibrinogen levels were significantly favorable for Niaspan compared with gemfibrozil (P<.02), as gemfibrozil increased the fibrinogen level (from 5% to 9%) and Niaspan tended to decrease the fibrinogen level (from -1% to -6%). Gemfibrozil 91-102 fibrinogen beta chain Homo sapiens 18-28 10789612-8 2000 Effects on plasma fibrinogen levels were significantly favorable for Niaspan compared with gemfibrozil (P<.02), as gemfibrozil increased the fibrinogen level (from 5% to 9%) and Niaspan tended to decrease the fibrinogen level (from -1% to -6%). Gemfibrozil 118-129 fibrinogen beta chain Homo sapiens 144-154 10789612-8 2000 Effects on plasma fibrinogen levels were significantly favorable for Niaspan compared with gemfibrozil (P<.02), as gemfibrozil increased the fibrinogen level (from 5% to 9%) and Niaspan tended to decrease the fibrinogen level (from -1% to -6%). Gemfibrozil 118-129 fibrinogen beta chain Homo sapiens 144-154 10744143-5 2000 tPA and PAI-1 antigen increased significantly in the gemfibrozil group (+11.3% and + 16.4%, respectively, p < 0.001), whereas there was no treatment effect on PAI-1 activity (median change 0.0%). Gemfibrozil 53-64 serpin family E member 1 Homo sapiens 8-13 10744143-7 2000 Furthermore, pronounced long-term lowering of serum triglycerides by gemfibrozil treatment does not significantly affect the plasma PAI-1 activity level but increases the plasma tPA and PAI-1 antigen concentrations. Gemfibrozil 69-80 serpin family E member 1 Homo sapiens 186-191 10910008-8 2000 After 6 months, gemfibrozil therapy versus placebo had a marked lipid-lowering effect, significantly decreased triglycerides (61%), VLDL cholesterol (72%), apoB (28%), and apoE (55%), and increased high-density lipoprotein (44%) and apoA-I (18%). Gemfibrozil 16-27 apolipoprotein B Homo sapiens 156-160 10910008-8 2000 After 6 months, gemfibrozil therapy versus placebo had a marked lipid-lowering effect, significantly decreased triglycerides (61%), VLDL cholesterol (72%), apoB (28%), and apoE (55%), and increased high-density lipoprotein (44%) and apoA-I (18%). Gemfibrozil 16-27 apolipoprotein E Homo sapiens 172-176 10910008-8 2000 After 6 months, gemfibrozil therapy versus placebo had a marked lipid-lowering effect, significantly decreased triglycerides (61%), VLDL cholesterol (72%), apoB (28%), and apoE (55%), and increased high-density lipoprotein (44%) and apoA-I (18%). Gemfibrozil 16-27 apolipoprotein A1 Homo sapiens 233-239 10910008-9 2000 Furthermore, gemfibrozil affected the apoprotein composition of VLDL: total protein increased by 28%, the molar ratio of apoE to apoB decreased 64%, and apoE content decreased 55%. Gemfibrozil 13-24 apolipoprotein E Homo sapiens 121-125 10910008-9 2000 Furthermore, gemfibrozil affected the apoprotein composition of VLDL: total protein increased by 28%, the molar ratio of apoE to apoB decreased 64%, and apoE content decreased 55%. Gemfibrozil 13-24 apolipoprotein B Homo sapiens 129-133 10910008-9 2000 Furthermore, gemfibrozil affected the apoprotein composition of VLDL: total protein increased by 28%, the molar ratio of apoE to apoB decreased 64%, and apoE content decreased 55%. Gemfibrozil 13-24 apolipoprotein E Homo sapiens 153-157 10657576-0 2000 Effects of gemfibrozil on insulin sensitivity and on haemostatic variables in hypertriglyceridemic patients. Gemfibrozil 11-22 insulin Homo sapiens 26-33 10656170-10 1999 Small fibrinogen (t0, t2, t6, t8), PAI-1 (t6) and AT III (t0-t8) increases were observed after gemfibrozil, while simvastatin did not significantly modify these parameters. Gemfibrozil 95-106 serpin family E member 1 Homo sapiens 35-40 11111964-6 2000 F and G combined treatment is safe and useful in patients who do not respond satisfactorily to monotherapy with F. Gemfibrozil augments the effect of F on TC, LDL-C, and triglyceride levels. Gemfibrozil 115-126 component of oligomeric golgi complex 2 Homo sapiens 159-164 10521386-6 1999 In the gemfibrozil group, there was a significant decrease in F1+2, while TAT complexes did not change. Gemfibrozil 7-18 coagulation factor XII Homo sapiens 62-66 10656170-10 1999 Small fibrinogen (t0, t2, t6, t8), PAI-1 (t6) and AT III (t0-t8) increases were observed after gemfibrozil, while simvastatin did not significantly modify these parameters. Gemfibrozil 95-106 serpin family C member 1 Homo sapiens 50-56 10494028-7 1999 Under serum conditions allowing apo CIII secretion, treatment with the peroxisome-proliferator activated receptor (PPAR)alpha activators fenofibrate, gemfibrozil and Wy-14643 result in a marked lowering of apo CIII secretion and gene expression, this effect being most pronounced with Wy-14643. Gemfibrozil 150-161 apolipoprotein C3 Homo sapiens 32-40 10364093-7 1999 In contrast, both fenofibric acid and gemfibrozil markedly decreased PAI-1 transcription and secretion from HUVECs and EA.hy926 cells. Gemfibrozil 38-49 serpin family E member 1 Homo sapiens 69-74 10494028-7 1999 Under serum conditions allowing apo CIII secretion, treatment with the peroxisome-proliferator activated receptor (PPAR)alpha activators fenofibrate, gemfibrozil and Wy-14643 result in a marked lowering of apo CIII secretion and gene expression, this effect being most pronounced with Wy-14643. Gemfibrozil 150-161 peroxisome proliferator activated receptor alpha Homo sapiens 115-125 10494028-7 1999 Under serum conditions allowing apo CIII secretion, treatment with the peroxisome-proliferator activated receptor (PPAR)alpha activators fenofibrate, gemfibrozil and Wy-14643 result in a marked lowering of apo CIII secretion and gene expression, this effect being most pronounced with Wy-14643. Gemfibrozil 150-161 apolipoprotein C3 Homo sapiens 206-214 9791843-7 1998 CONCLUSION: In patients with risk of pancreatitis due to severe hypertriglyceridemia, immediate reduction of the triglyceride level is achievable by using a single dose of regular insulin (0.1 unit/kg, subcutaneous) while long-term maintenance therapy can be provided by gemfibrozil. Gemfibrozil 271-282 insulin Homo sapiens 180-187 9853402-4 1998 At 1 mM, gemfibrozil decreased not only the activity of HMG-CoA reductase and cholesterol synthesis, but also the protein content of the cells and peroxisomal enzyme activity, indicating nonspecific inhibition at this concentration. Gemfibrozil 9-20 3-hydroxy-3-methylglutaryl-CoA reductase Rattus norvegicus 56-73 9853402-6 1998 With respect to the direct effect on HMG-CoA reductase in the cell homogenate, gemfibrozil at 0.25 mm did not affect the activity, but it clearly inhibited the activity at 2 mM and above. Gemfibrozil 79-90 3-hydroxy-3-methylglutaryl-CoA reductase Rattus norvegicus 37-54 9853402-11 1998 In contrast to whole body results, gemfibrozil may suppress cholesterol synthesis from [14C]acetate through the inhibition of HMG-CoA reductase at the culture cell level. Gemfibrozil 35-46 3-hydroxy-3-methylglutaryl-CoA reductase Rattus norvegicus 126-143 9853402-3 1998 Gemfibrozil at 0.25 mM increased the activity of some peroxisomal enzymes (catalase and the cyanide-insensitive fatty acyl-CoA oxidizing system) after incubation for 72 h. However, contrary to whole body experiments, gemfibrozil decreased the activity of HMG-CoA reductase and cholesterol synthesis from [14C]acetate. Gemfibrozil 0-11 3-hydroxy-3-methylglutaryl-CoA reductase Rattus norvegicus 255-272 9608883-5 1998 Gemfibrozil had no effect on plasma total cholesterol levels but gave significant dose-dependent decreases in plasma (VLDL) triglyceride levels (up to -53%). Gemfibrozil 0-11 CD320 antigen Mus musculus 118-122 9699891-0 1998 The 5A/6A polymorphism in the promoter of the stromelysin-1 (MMP-3) gene predicts progression of angiographically determined coronary artery disease in men in the LOCAT gemfibrozil study. Gemfibrozil 169-180 matrix metallopeptidase 3 Homo sapiens 46-59 9699891-0 1998 The 5A/6A polymorphism in the promoter of the stromelysin-1 (MMP-3) gene predicts progression of angiographically determined coronary artery disease in men in the LOCAT gemfibrozil study. Gemfibrozil 169-180 matrix metallopeptidase 3 Homo sapiens 61-66 9678787-13 1998 Serum apolipoprotein A1 (apo A1) was increased (P < 0.05) by bezafibrate compared to gemfibrozil (placebo 103 +/- 26, bezafibrate 111 +/- 28 and gemfibrozil 102 +/- 25 mg/dl) and CETA from HDL to VLDL and LDL was decreased (P < 0.05) by bezafibrate compared to placebo, but the apparent decrease with gemfibrozil did not achieve statistical significance (placebo 39.6 +/- 17.7, bezafibrate 32.3 +/- 14.7 and gemfibrozil 33.8 +/- 15.0 nmol/ml/h). Gemfibrozil 148-159 apolipoprotein A1 Homo sapiens 6-23 9678787-13 1998 Serum apolipoprotein A1 (apo A1) was increased (P < 0.05) by bezafibrate compared to gemfibrozil (placebo 103 +/- 26, bezafibrate 111 +/- 28 and gemfibrozil 102 +/- 25 mg/dl) and CETA from HDL to VLDL and LDL was decreased (P < 0.05) by bezafibrate compared to placebo, but the apparent decrease with gemfibrozil did not achieve statistical significance (placebo 39.6 +/- 17.7, bezafibrate 32.3 +/- 14.7 and gemfibrozil 33.8 +/- 15.0 nmol/ml/h). Gemfibrozil 148-159 apolipoprotein A1 Homo sapiens 25-31 9678787-13 1998 Serum apolipoprotein A1 (apo A1) was increased (P < 0.05) by bezafibrate compared to gemfibrozil (placebo 103 +/- 26, bezafibrate 111 +/- 28 and gemfibrozil 102 +/- 25 mg/dl) and CETA from HDL to VLDL and LDL was decreased (P < 0.05) by bezafibrate compared to placebo, but the apparent decrease with gemfibrozil did not achieve statistical significance (placebo 39.6 +/- 17.7, bezafibrate 32.3 +/- 14.7 and gemfibrozil 33.8 +/- 15.0 nmol/ml/h). Gemfibrozil 148-159 apolipoprotein A1 Homo sapiens 6-23 9678787-13 1998 Serum apolipoprotein A1 (apo A1) was increased (P < 0.05) by bezafibrate compared to gemfibrozil (placebo 103 +/- 26, bezafibrate 111 +/- 28 and gemfibrozil 102 +/- 25 mg/dl) and CETA from HDL to VLDL and LDL was decreased (P < 0.05) by bezafibrate compared to placebo, but the apparent decrease with gemfibrozil did not achieve statistical significance (placebo 39.6 +/- 17.7, bezafibrate 32.3 +/- 14.7 and gemfibrozil 33.8 +/- 15.0 nmol/ml/h). Gemfibrozil 148-159 apolipoprotein A1 Homo sapiens 25-31 9678787-13 1998 Serum apolipoprotein A1 (apo A1) was increased (P < 0.05) by bezafibrate compared to gemfibrozil (placebo 103 +/- 26, bezafibrate 111 +/- 28 and gemfibrozil 102 +/- 25 mg/dl) and CETA from HDL to VLDL and LDL was decreased (P < 0.05) by bezafibrate compared to placebo, but the apparent decrease with gemfibrozil did not achieve statistical significance (placebo 39.6 +/- 17.7, bezafibrate 32.3 +/- 14.7 and gemfibrozil 33.8 +/- 15.0 nmol/ml/h). Gemfibrozil 148-159 apolipoprotein A1 Homo sapiens 6-23 9678787-13 1998 Serum apolipoprotein A1 (apo A1) was increased (P < 0.05) by bezafibrate compared to gemfibrozil (placebo 103 +/- 26, bezafibrate 111 +/- 28 and gemfibrozil 102 +/- 25 mg/dl) and CETA from HDL to VLDL and LDL was decreased (P < 0.05) by bezafibrate compared to placebo, but the apparent decrease with gemfibrozil did not achieve statistical significance (placebo 39.6 +/- 17.7, bezafibrate 32.3 +/- 14.7 and gemfibrozil 33.8 +/- 15.0 nmol/ml/h). Gemfibrozil 148-159 apolipoprotein A1 Homo sapiens 25-31 9678787-15 1998 Plasma fibrinogen was increased (P < 0.05) by gemfibrozil (placebo 4.16 (3.38-4.71) and gemfibrozil 4.65 (4.05-5.77) g/l) and was significantly lower (P < 0.001) on bezafibrate (3.60 (3.18-4.54) g/l) than on gemfibrozil treatment. Gemfibrozil 49-60 fibrinogen beta chain Homo sapiens 7-17 9678787-15 1998 Plasma fibrinogen was increased (P < 0.05) by gemfibrozil (placebo 4.16 (3.38-4.71) and gemfibrozil 4.65 (4.05-5.77) g/l) and was significantly lower (P < 0.001) on bezafibrate (3.60 (3.18-4.54) g/l) than on gemfibrozil treatment. Gemfibrozil 91-102 fibrinogen beta chain Homo sapiens 7-17 9678787-15 1998 Plasma fibrinogen was increased (P < 0.05) by gemfibrozil (placebo 4.16 (3.38-4.71) and gemfibrozil 4.65 (4.05-5.77) g/l) and was significantly lower (P < 0.001) on bezafibrate (3.60 (3.18-4.54) g/l) than on gemfibrozil treatment. Gemfibrozil 91-102 fibrinogen beta chain Homo sapiens 7-17 9678787-20 1998 The two drugs also had different effects on the plasma fibrinogen levels, which increased with gemfibrozil and tended to decrea Gemfibrozil 95-106 fibrinogen beta chain Homo sapiens 55-65 18370512-1 1998 OBJECTIVE: This pilot study investigated the efficacy of high-dose gemfibrozil (2400 mg/day) in treating patients with elevated lipoprotein (a) [Lp(a)]. Gemfibrozil 67-78 lipoprotein(a) Homo sapiens 145-150 9514936-5 1998 The addition of gemfibrozil to cultures of monkey and human hepatocytes had no effect on apo(a) synthesis, but resulted in a dose- and time-dependent increase of apo A-I synthesis and mRNA. Gemfibrozil 16-27 apolipoprotein A1 Homo sapiens 162-169 9514936-7 1998 Similar results were obtained by using human hepatocytes; apo(a) synthesis remained unchanged, while apo A-I secretion was 2.0-fold increased at 1 mM gemfibrozil. Gemfibrozil 150-161 apolipoprotein A1 Homo sapiens 101-108 9484993-3 1998 Fibrates like gemfibrozil are well-known PPARalpha activators and are used in the treatment of hyperlipidemia. Gemfibrozil 14-25 peroxisome proliferator activated receptor alpha Homo sapiens 41-50 9484993-4 1998 We show that the RXR ligand LGD1069 (Targretin), like gemfibrozil, can activate the PPARalpha/RXR signal-transduction pathway, including transactivation of the bifunctional enzyme or acyl-CoA oxidase response elements in a cotransfection assay. Gemfibrozil 54-65 retinoid X receptor alpha Homo sapiens 17-20 9484993-4 1998 We show that the RXR ligand LGD1069 (Targretin), like gemfibrozil, can activate the PPARalpha/RXR signal-transduction pathway, including transactivation of the bifunctional enzyme or acyl-CoA oxidase response elements in a cotransfection assay. Gemfibrozil 54-65 peroxisome proliferator activated receptor alpha Homo sapiens 84-93 9484993-4 1998 We show that the RXR ligand LGD1069 (Targretin), like gemfibrozil, can activate the PPARalpha/RXR signal-transduction pathway, including transactivation of the bifunctional enzyme or acyl-CoA oxidase response elements in a cotransfection assay. Gemfibrozil 54-65 retinoid X receptor alpha Homo sapiens 94-97 9507986-1 1998 Previously, we demonstrated that protein-DNA interactions at the drug response element (DRE) in the human apoA-I promoter were important for the induction of apoA-I gene expression by gemfibrozil. Gemfibrozil 184-195 apolipoprotein A1 Homo sapiens 106-112 9507986-1 1998 Previously, we demonstrated that protein-DNA interactions at the drug response element (DRE) in the human apoA-I promoter were important for the induction of apoA-I gene expression by gemfibrozil. Gemfibrozil 184-195 apolipoprotein A1 Homo sapiens 158-164 9507986-9 1998 Together these observations indicate that HSmuBP2 acts as a transcription factor that regulates apoA-I gene expression in hepatoma cells and whose activity may be stimulated by gemfibrozil treatment. Gemfibrozil 177-188 apolipoprotein A1 Homo sapiens 96-102 9364979-8 1997 However, after twelve weeks, gemfibrozil enhanced functional fibrinogen levels by 7.2% (p <0.05) as assessed by the Clauss mechanical assay, but decreased functional fibrinogen levels by 12.5% (p <0.0001) when a Clauss assay based on turbidity was used. Gemfibrozil 29-40 fibrinogen beta chain Homo sapiens 169-179 9409246-0 1997 Upregulation of low density lipoprotein receptor by gemfibrozil, a hypolipidemic agent, in human hepatoma cells through stabilization of mRNA transcripts. Gemfibrozil 52-63 low density lipoprotein receptor Homo sapiens 16-48 9409246-3 1997 Exposure to gemfibrozil, 40 mumol/L, for 3 days increased the binding of 125I-LDL to the surface of three lines of human hepatoma cell, HepG2, HuH7, and HLE by 1.5- to 2.0-fold. Gemfibrozil 12-23 MIR7-3 host gene Homo sapiens 143-147 9409246-7 1997 Gemfibrozil increased the levels of LDL receptor mRNA and protein in HepG2 cells. Gemfibrozil 0-11 low density lipoprotein receptor Homo sapiens 36-48 9409246-11 1997 The expression of LDL receptor regulated by the sterol regulatory element was increased by pravastatin, but not by gemfibrozil. Gemfibrozil 115-126 low density lipoprotein receptor Homo sapiens 18-30 9409246-13 1997 Gemfibrozil prolonged the half-life of the mRNA for LDL receptor but not that for the EGF receptor. Gemfibrozil 0-11 low density lipoprotein receptor Homo sapiens 52-64 9409246-14 1997 Stabilization of the LDL receptor mRNA is suggested to be the novel mode of action of gemfibrozil. Gemfibrozil 86-97 low density lipoprotein receptor Homo sapiens 21-33 9361689-7 1997 The postprandial TRL apo B increase was reduced with gemfibrozil (from 11.6 +/- 2.8 to 20.7 +/- 5.0 mg/dL with therapy v 19.0 +/- 7.6 to 33.0 +/- 12.5 mg/dL before therapy, P < .05, respectively) with a proportionally greater increase in apo B-48 (119% and 169%, respectively) compared with apo B-100 (64% and 64%, respectively). Gemfibrozil 53-64 apolipoprotein B Homo sapiens 294-303 9364979-0 1997 Effects of gemfibrozil and ciprofibrate on plasma levels of tissue-type plasminogen activator, plasminogen activator inhibitor-1 and fibrinogen in hyperlipidaemic patients. Gemfibrozil 11-22 plasminogen activator, tissue type Homo sapiens 60-93 9497506-5 1997 RESULTS: Gemfibrozil reduced TC (-21%), LDL-C (-26%), triglycerides (TG)(-48%) and Lp(a) (-25%), increased HDL-C (+48%)(p < 0.001). Gemfibrozil 9-20 component of oligomeric golgi complex 2 Homo sapiens 40-45 9364979-3 1997 We sought to learn more about this variability by examining the effects of gemfibrozil and ciprofibrate on plasma levels of tissue-type plasminogen activator (t-PA), plasminogen activator inhibitor-1 (PAI-1) and fibrinogen in primary hyperlipidaemic patients after six and twelve weeks of treatment using different assay systems for PAI-1 and fibrinogen. Gemfibrozil 75-86 plasminogen activator, tissue type Homo sapiens 124-157 9364979-6 1997 Fibrinogen antigen levels were elevated by 17.6% (p <0.05) and 24.3% (p <0.001) with gemfibrozil after six and twelve weeks, respectively, whereas with ciprofibrate there was no effect. Gemfibrozil 91-102 fibrinogen beta chain Homo sapiens 0-10 9364979-8 1997 However, after twelve weeks, gemfibrozil enhanced functional fibrinogen levels by 7.2% (p <0.05) as assessed by the Clauss mechanical assay, but decreased functional fibrinogen levels by 12.5% (p <0.0001) when a Clauss assay based on turbidity was used. Gemfibrozil 29-40 fibrinogen beta chain Homo sapiens 61-71 9294990-12 1997 Simvastatin + ciprofibrate was more effective than pravastatin + gemfibrozil in reducing LDL, TG, and plasma fibrinogen levels. Gemfibrozil 65-76 fibrinogen beta chain Homo sapiens 109-119 9298501-2 1997 Several drugs and other compounds, e.g. phenobarbital, gemfibrozil, fenofibrate, prednisone, estrogen and alcohol, induce apolipoprotein A-I synthesis. Gemfibrozil 55-66 apolipoprotein A1 Homo sapiens 122-140 9497506-7 1997 CONCLUSION: Besides reducing plasma LDL-C, TG and increasing HDL-C, gemfibrozil effectively lowers Lp(a) levels. Gemfibrozil 68-79 lipoprotein(a) Homo sapiens 99-104 9327717-4 1997 In contrast, plasma plasminogen activator inhibitor-1 antigen (36.9 +/- 12.4 vs 27.3 +/- 11.4 ng/ml; p = 0.008) and activity (15.5 +/- 5.5 vs 11.8 +/- 3.0 IU/ml; p = 0.009) and tissue plasminogen activator antigen (13.2 +/- 4.0 vs 10.4 +/- 3.7 ng/ml; p = 0.007) were significantly depressed, and tissue plasimogen activator activity (0.57 +/- 0.31 vs 0.69 +/- 0.38 IU/ml; p = 0.015) was significantly elevated by gemfibrozil. Gemfibrozil 413-424 serpin family E member 1 Homo sapiens 20-53 9024157-0 1997 Attenuation by gemfibrozil of expression of plasminogen activator inhibitor type 1 induced by insulin and its precursors. Gemfibrozil 15-26 serpin family E member 1 Homo sapiens 44-82 9145201-5 1997 The hydroxymethylglutaryl-CoA (HMG-CoA) reductase (rate-limiting enzyme of cholesterol synthesis) activity of peroxisomes and microsomes was greatly increased by in vivo treatment with gemfibrozil, but was decreased by addition of the agent to the assay mixture of the enzyme. Gemfibrozil 185-196 3-hydroxy-3-methylglutaryl-CoA reductase Rattus norvegicus 4-49 9105566-12 1997 Lp(a) (P = 0.04) and plasma insulin concentrations (P = 0.03) were negative predictors of percentage triglyceride-lowering with gemfibrozil. Gemfibrozil 128-139 lipoprotein(a) Homo sapiens 0-5 9105566-12 1997 Lp(a) (P = 0.04) and plasma insulin concentrations (P = 0.03) were negative predictors of percentage triglyceride-lowering with gemfibrozil. Gemfibrozil 128-139 insulin Homo sapiens 28-35 9024157-0 1997 Attenuation by gemfibrozil of expression of plasminogen activator inhibitor type 1 induced by insulin and its precursors. Gemfibrozil 15-26 insulin Homo sapiens 94-101 9024157-4 1997 METHODS AND RESULTS: In Hep G2 cells, 500 (700) mumol/L gemfibrozil decreased basal secretion of PAI-1 by 26% (43%) (P = .012 and P = .021, respectively) and attenuated insulin-induced (10 nmol/L) augmentation of PAI-1 in conditioned media by 61% (109%) (P = .010) within 24 hours. Gemfibrozil 56-67 serpin family E member 1 Homo sapiens 97-102 9024157-4 1997 METHODS AND RESULTS: In Hep G2 cells, 500 (700) mumol/L gemfibrozil decreased basal secretion of PAI-1 by 26% (43%) (P = .012 and P = .021, respectively) and attenuated insulin-induced (10 nmol/L) augmentation of PAI-1 in conditioned media by 61% (109%) (P = .010) within 24 hours. Gemfibrozil 56-67 insulin Homo sapiens 169-176 9024157-4 1997 METHODS AND RESULTS: In Hep G2 cells, 500 (700) mumol/L gemfibrozil decreased basal secretion of PAI-1 by 26% (43%) (P = .012 and P = .021, respectively) and attenuated insulin-induced (10 nmol/L) augmentation of PAI-1 in conditioned media by 61% (109%) (P = .010) within 24 hours. Gemfibrozil 56-67 serpin family E member 1 Homo sapiens 213-218 9024157-6 1997 Gemfibrozil attenuated the augmentation of PAI-1 secretion induced by proinsulin (> 100%), by des(31,32)proinsulin (75%), and by des(64,65) proinsulin (77%) as well (10 nmol/L each). Gemfibrozil 0-11 serpin family E member 1 Homo sapiens 43-48 9024157-6 1997 Gemfibrozil attenuated the augmentation of PAI-1 secretion induced by proinsulin (> 100%), by des(31,32)proinsulin (75%), and by des(64,65) proinsulin (77%) as well (10 nmol/L each). Gemfibrozil 0-11 insulin Homo sapiens 70-80 9024157-6 1997 Gemfibrozil attenuated the augmentation of PAI-1 secretion induced by proinsulin (> 100%), by des(31,32)proinsulin (75%), and by des(64,65) proinsulin (77%) as well (10 nmol/L each). Gemfibrozil 0-11 insulin Homo sapiens 107-117 9024157-6 1997 Gemfibrozil attenuated the augmentation of PAI-1 secretion induced by proinsulin (> 100%), by des(31,32)proinsulin (75%), and by des(64,65) proinsulin (77%) as well (10 nmol/L each). Gemfibrozil 0-11 insulin Homo sapiens 107-117 9024157-10 1997 CONCLUSIONS: Gemfibrozil attenuated the augmentation of synthesis and secretion of PAI-1 induced by insulin and its precursors directly and specifically. Gemfibrozil 13-24 serpin family E member 1 Homo sapiens 83-88 9024157-10 1997 CONCLUSIONS: Gemfibrozil attenuated the augmentation of synthesis and secretion of PAI-1 induced by insulin and its precursors directly and specifically. Gemfibrozil 13-24 insulin Homo sapiens 100-107 9012633-4 1997 Different fibrates showed different potency in suppressing PAI-1 production: gemfibrozil and clofibric acid, at a concentration of 1 mmol/L, reduced PAI-1 synthesis over 24 hours to 52 +/- 20% and 60 +/- 5%, while clofibrate and bezafibrate lowered PAI-1 synthesis to only 86 +/- 17% and 84 +/- 15% of control values, respectively. Gemfibrozil 77-88 plasminogen activator inhibitor 1 Macaca fascicularis 59-64 9012633-4 1997 Different fibrates showed different potency in suppressing PAI-1 production: gemfibrozil and clofibric acid, at a concentration of 1 mmol/L, reduced PAI-1 synthesis over 24 hours to 52 +/- 20% and 60 +/- 5%, while clofibrate and bezafibrate lowered PAI-1 synthesis to only 86 +/- 17% and 84 +/- 15% of control values, respectively. Gemfibrozil 77-88 plasminogen activator inhibitor 1 Macaca fascicularis 149-154 9012633-4 1997 Different fibrates showed different potency in suppressing PAI-1 production: gemfibrozil and clofibric acid, at a concentration of 1 mmol/L, reduced PAI-1 synthesis over 24 hours to 52 +/- 20% and 60 +/- 5%, while clofibrate and bezafibrate lowered PAI-1 synthesis to only 86 +/- 17% and 84 +/- 15% of control values, respectively. Gemfibrozil 77-88 plasminogen activator inhibitor 1 Macaca fascicularis 149-154 9006809-2 1996 Although both drugs lowered plasma triglycerides (TG) to about the same extent in chow-fed rats, gemfibrozil lowered liver TG as well as plasma total and LDL-cholesterol (LDL-C), but elevated HDL-cholesterol (HDL-C) and plasma apo E concentrations. Gemfibrozil 97-108 apolipoprotein E Rattus norvegicus 227-232 9069450-14 1997 Furthermore, gemfibrozil treatment resulted in the reduction of plasma concentrations of complex (LP-Bc) and simple (LP-B) apoB-containing lipoprotein particles by 22 and 7%, respectively. Gemfibrozil 13-24 apolipoprotein B Homo sapiens 123-127 9069450-19 1997 CONCLUSIONS: Gemfibrozil treatment significantly reduces both plasma lipids and apoB, apoC-III and apoE concentrations in patients with moderately advanced renal insufficiency. Gemfibrozil 13-24 apolipoprotein B Homo sapiens 80-84 9069450-19 1997 CONCLUSIONS: Gemfibrozil treatment significantly reduces both plasma lipids and apoB, apoC-III and apoE concentrations in patients with moderately advanced renal insufficiency. Gemfibrozil 13-24 apolipoprotein C3 Homo sapiens 86-94 9069450-19 1997 CONCLUSIONS: Gemfibrozil treatment significantly reduces both plasma lipids and apoB, apoC-III and apoE concentrations in patients with moderately advanced renal insufficiency. Gemfibrozil 13-24 apolipoprotein E Homo sapiens 99-103 9069450-20 1997 The results of this study indicate that gemfibrozil enhances the clearance of apoB-containing triglyceride-rich lipoproteins. Gemfibrozil 40-51 apolipoprotein B Homo sapiens 78-82 9031453-0 1997 Modulation of plasma fibrinogen levels by ciprofibrate and gemfibrozil in primary hyperlipidaemia. Gemfibrozil 59-70 fibrinogen beta chain Homo sapiens 21-31 9006809-7 1996 From these data we postulate that, although similar with regard to TG lowering activity and mechanisms thereof, gemfibrozil and bezafibrate produce fundamentally different effects on LDL, HDL and apolipoprotein metabolism (apo B and apo E) in rats which may relate to potential differential effects on reverse cholesterol transport and atherogenesis. Gemfibrozil 112-123 apolipoprotein B Rattus norvegicus 188-238 8900208-1 1996 Previously, we demonstrated that when two human hepatoma cell lines, Hep3B and HepG2, were exposed to gemfibrozil, a hypolipidemic drug, a 2-fold induction in apolipoprotein A-I (apoA-I) mRNA levels resulted. Gemfibrozil 102-113 apolipoprotein A1 Homo sapiens 159-177 8902147-0 1996 Effects of gemfibrozil administration on very low density lipoprotein receptor mRNA levels in rabbits. Gemfibrozil 11-22 very low-density lipoprotein receptor Oryctolagus cuniculus 41-78 8902147-1 1996 To elucidate the regulation of very low density lipoprotein (VLDL) receptor gene expression, we administered to rabbits for 14 days gemfibrozil, a fabric acid derivative and a lipid lowering drug that is also included among peroxisome proliferators. Gemfibrozil 132-143 very low-density lipoprotein receptor Oryctolagus cuniculus 31-75 8902147-3 1996 The VLDL receptor mRNA levels in retroperitoneal adipose tissue and in gastrocnemius muscle were increased 6.9-fold and 3.7-fold, respectively, with gemfibrozil treatment, but no marked changes were observed in the heart, the organ in which VLDL receptor is most highly expressed. Gemfibrozil 149-160 very low-density lipoprotein receptor Oryctolagus cuniculus 4-17 8902147-3 1996 The VLDL receptor mRNA levels in retroperitoneal adipose tissue and in gastrocnemius muscle were increased 6.9-fold and 3.7-fold, respectively, with gemfibrozil treatment, but no marked changes were observed in the heart, the organ in which VLDL receptor is most highly expressed. Gemfibrozil 149-160 very low-density lipoprotein receptor Oryctolagus cuniculus 241-254 8900208-1 1996 Previously, we demonstrated that when two human hepatoma cell lines, Hep3B and HepG2, were exposed to gemfibrozil, a hypolipidemic drug, a 2-fold induction in apolipoprotein A-I (apoA-I) mRNA levels resulted. Gemfibrozil 102-113 apolipoprotein A1 Homo sapiens 179-185 8900208-4 1996 However, nuclear run-off assays indicated that the transcription rate of the apoA-I gene was increased 2-fold in gemfibrozil-treated cells. Gemfibrozil 113-124 apolipoprotein A1 Homo sapiens 77-83 8900208-5 1996 Transient transfection experiments also indicated that the induction of apoA-I mRNA level in response to gemfibrozil is mediated at the transcriptional level. Gemfibrozil 105-116 apolipoprotein A1 Homo sapiens 72-78 8900208-6 1996 We have identified two copies of the "drug-responsive element" (DRE) in the apoA-I promoter region that may be responsible for the increase in apoA-I transcriptional activity by gemfibrozil. Gemfibrozil 178-189 apolipoprotein A1 Homo sapiens 76-82 8900208-6 1996 We have identified two copies of the "drug-responsive element" (DRE) in the apoA-I promoter region that may be responsible for the increase in apoA-I transcriptional activity by gemfibrozil. Gemfibrozil 178-189 apolipoprotein A1 Homo sapiens 143-149 8900208-15 1996 These results implicate protein-DNA interactions at the DRE region in the transcriptional induction of human apoA-I gene expression by gemfibrozil. Gemfibrozil 135-146 apolipoprotein A1 Homo sapiens 109-115 8696946-3 1996 Since the liver is the major organ involved in HDL production and removal, we assessed the effect of gemfibrozil on the modulation of apoA-I (a major protein of HDL)-containing particles by a human hepatoblastoma cell line (Hep G2). Gemfibrozil 101-112 apolipoprotein A1 Homo sapiens 134-140 8790349-5 1996 SCD1 mRNA was analyzed from the livers of BALB/c mice that had been fed diets supplemented with clofibrate or gemfibrozil. Gemfibrozil 110-121 stearoyl-Coenzyme A desaturase 1 Mus musculus 0-4 8879439-4 1996 Gemfibrozil significantly affected VLDL composition: protein increased 26%, molar ratio of apoE to apoB reduced 48%, apoC-II increased 19%, and apoC-III decreased 9%. Gemfibrozil 0-11 apolipoprotein E Homo sapiens 91-95 8879439-4 1996 Gemfibrozil significantly affected VLDL composition: protein increased 26%, molar ratio of apoE to apoB reduced 48%, apoC-II increased 19%, and apoC-III decreased 9%. Gemfibrozil 0-11 apolipoprotein B Homo sapiens 99-103 8879439-4 1996 Gemfibrozil significantly affected VLDL composition: protein increased 26%, molar ratio of apoE to apoB reduced 48%, apoC-II increased 19%, and apoC-III decreased 9%. Gemfibrozil 0-11 apolipoprotein C2 Homo sapiens 117-124 8879439-4 1996 Gemfibrozil significantly affected VLDL composition: protein increased 26%, molar ratio of apoE to apoB reduced 48%, apoC-II increased 19%, and apoC-III decreased 9%. Gemfibrozil 0-11 apolipoprotein C3 Homo sapiens 144-152 8879439-9 1996 Taken together, these results suggest that treatment with gemfibrozil reduces plasma concentrations of VLDL and alters the apoprotein composition of VLDL in a manner that may favor LDL- and VLDL-receptor-mediated clearance of the apoE-rich VLDL subfraction, thereby reducing TG-rich particle concentrations, and possibly reducing risk for coronary heart disease. Gemfibrozil 58-69 very low density lipoprotein receptor Homo sapiens 190-203 8879439-9 1996 Taken together, these results suggest that treatment with gemfibrozil reduces plasma concentrations of VLDL and alters the apoprotein composition of VLDL in a manner that may favor LDL- and VLDL-receptor-mediated clearance of the apoE-rich VLDL subfraction, thereby reducing TG-rich particle concentrations, and possibly reducing risk for coronary heart disease. Gemfibrozil 58-69 apolipoprotein E Homo sapiens 230-234 8696946-5 1996 These data indicate that gemfibrozil stabilizes apoA-I mRNA transcripts, resulting in increased translation of functional apoA-I-containing particles capable of effluxing cellular cholesterol, thus defining a major mechanism by which gemfibrozil increases HDL. Gemfibrozil 25-36 apolipoprotein A1 Homo sapiens 48-54 8696946-5 1996 These data indicate that gemfibrozil stabilizes apoA-I mRNA transcripts, resulting in increased translation of functional apoA-I-containing particles capable of effluxing cellular cholesterol, thus defining a major mechanism by which gemfibrozil increases HDL. Gemfibrozil 25-36 apolipoprotein A1 Homo sapiens 122-128 8696946-5 1996 These data indicate that gemfibrozil stabilizes apoA-I mRNA transcripts, resulting in increased translation of functional apoA-I-containing particles capable of effluxing cellular cholesterol, thus defining a major mechanism by which gemfibrozil increases HDL. Gemfibrozil 234-245 apolipoprotein A1 Homo sapiens 48-54 8696946-5 1996 These data indicate that gemfibrozil stabilizes apoA-I mRNA transcripts, resulting in increased translation of functional apoA-I-containing particles capable of effluxing cellular cholesterol, thus defining a major mechanism by which gemfibrozil increases HDL. Gemfibrozil 234-245 apolipoprotein A1 Homo sapiens 122-128 8946270-0 1996 Effects of gemfibrozil therapy on glucose tolerance, insulin sensitivity and plasma plasminogen activator inhibitor activity in hypertriglyceridaemia. Gemfibrozil 11-22 insulin Homo sapiens 53-60 8824106-7 1996 In keeping with a more effective reduction of VLDL particles, a more pronounced reduction of apo-CIII also was observed after gemfibrozil, which correlated with the reduction in plasma triglycerides. Gemfibrozil 126-137 apolipoprotein C3 Homo sapiens 93-101 8865525-0 1996 Gemfibrozil reduces thrombin generation in patients with combined hyperlipidaemia, without influencing plasma fibrinogen, fibrin gel structure or coagulation factor VII. Gemfibrozil 0-11 coagulation factor II, thrombin Homo sapiens 20-28 8675576-0 1996 Gemfibrozil treatment of endogenous hypertriglyceridemia: effect on insulin-mediated glucose disposal and plasma insulin concentrations. Gemfibrozil 0-11 insulin Homo sapiens 68-75 8865525-4 1996 Gemfibrozil reduced the plasma concentrations of thrombin-antithrombin complex (TAT) and prothrombin fragment F1 + 2 (F1 + 2), both at rest and during mental stress. Gemfibrozil 0-11 coagulation factor XII Homo sapiens 49-124 8865525-7 1996 Thus, gemfibrozil reduced thrombin generation in men with combined hyperlipoproteinaemia, without influencing the plasma levels of fibrinogen, VIIa and XIIa, or fibrin gel structure. Gemfibrozil 6-17 coagulation factor II, thrombin Homo sapiens 26-34 8865525-8 1996 Attenuation of thrombin generation may contribute to the primary-preventive effects of gemfibrozil on coronary heart disease. Gemfibrozil 87-98 coagulation factor II, thrombin Homo sapiens 15-23 8831919-9 1996 Ciprofibrate therapy significantly reduced (-8.33%) and gemfibrozil therapy significantly increased (+6.97%) plasma fibrinogen levels (P < 0.001 compared with baseline in each case). Gemfibrozil 56-67 fibrinogen beta chain Homo sapiens 116-126 8675576-0 1996 Gemfibrozil treatment of endogenous hypertriglyceridemia: effect on insulin-mediated glucose disposal and plasma insulin concentrations. Gemfibrozil 0-11 insulin Homo sapiens 113-120 8647932-10 1996 When transgenic mice were treated with gemfibrozil (0.5% wt/wt) plasma human apo A-I and HDL-cholesterol levels increased 32 and 73%, respectively, above control levels. Gemfibrozil 39-50 apolipoprotein A1 Homo sapiens 77-84 8808764-8 1996 These results indicate i) that the levels of Lp[a] are about 2 times higher in type IIa than IIb subjects, and ii) that although gemfibrozil elicits a rather uniform decrease in fasting and post-prandial triglyceride levels in type IIa and IIb patients, the drug causes heterogeneous changes in Lp[a], suggesting that different metabolic mechanisms may be dominant in subjects showing opposing effects. Gemfibrozil 129-140 lipoprotein(a) Homo sapiens 45-49 8808764-8 1996 These results indicate i) that the levels of Lp[a] are about 2 times higher in type IIa than IIb subjects, and ii) that although gemfibrozil elicits a rather uniform decrease in fasting and post-prandial triglyceride levels in type IIa and IIb patients, the drug causes heterogeneous changes in Lp[a], suggesting that different metabolic mechanisms may be dominant in subjects showing opposing effects. Gemfibrozil 129-140 lipoprotein(a) Homo sapiens 295-299 8808764-2 1996 Previous studies have provided evidence that the levels of Lp[a] and triglyceride are related, suggesting that Lp[a] might be altered by gemfibrozil, a drug well known for its efficacy in reducing plasma triglycerides. Gemfibrozil 137-148 lipoprotein(a) Homo sapiens 59-63 8620056-3 1995 This study presents data of an ongoing study to dissect the role of the apolipoprotein E gene locus in the response to low fat/low cholesterol diet combined with gemfibrozil treatment. Gemfibrozil 162-173 apolipoprotein E Homo sapiens 72-88 8808764-2 1996 Previous studies have provided evidence that the levels of Lp[a] and triglyceride are related, suggesting that Lp[a] might be altered by gemfibrozil, a drug well known for its efficacy in reducing plasma triglycerides. Gemfibrozil 137-148 lipoprotein(a) Homo sapiens 111-115 9072407-6 1995 Gemfibrozil effectively increased high density lipoprotein (HDL) subfraction rich in apoE (apoE-HDL) and significantly decreased very low density lipoprotein (VLDL) in normocholesterolaemic SHRSP. Gemfibrozil 0-11 apolipoprotein E Rattus norvegicus 85-89 9072407-6 1995 Gemfibrozil effectively increased high density lipoprotein (HDL) subfraction rich in apoE (apoE-HDL) and significantly decreased very low density lipoprotein (VLDL) in normocholesterolaemic SHRSP. Gemfibrozil 0-11 apolipoprotein E Rattus norvegicus 91-99 9072407-10 1995 On the other hand, it significantly increased the contents of apoA-I, A-IV and E in the HDL fraction compared with the control group, suggesting that gemfibrozil effectively increases anti-atherogenic HDL subfractions rich in apoA-I, A-IV or E. In the liver of hypercholesterolaemic SHRSP, gemfibrozil markedly prevented lipid accumulation. Gemfibrozil 150-161 apolipoprotein A1 Rattus norvegicus 62-68 9072407-10 1995 On the other hand, it significantly increased the contents of apoA-I, A-IV and E in the HDL fraction compared with the control group, suggesting that gemfibrozil effectively increases anti-atherogenic HDL subfractions rich in apoA-I, A-IV or E. In the liver of hypercholesterolaemic SHRSP, gemfibrozil markedly prevented lipid accumulation. Gemfibrozil 150-161 apolipoprotein A1 Rattus norvegicus 226-232 8620056-0 1995 Apolipoprotein E and complement C3 polymorphism and their role in the response to gemfibrozil and low fat low cholesterol therapy. Gemfibrozil 82-93 apolipoprotein E Homo sapiens 0-16 8620056-0 1995 Apolipoprotein E and complement C3 polymorphism and their role in the response to gemfibrozil and low fat low cholesterol therapy. Gemfibrozil 82-93 complement C3 Homo sapiens 21-34 8615769-3 1996 Increased levels of mdr2 mRNA were observed in the liver of mice treated with different fibrates: ciprofibrate, 660+/-155% (as compared with control group); clofibrate, 611+/-77%; bezafibrate, 410+/-47%; fenofibrate, 310+/-52%; gemfibrozil, 190+/-25% (P <0.05 compared with control group). Gemfibrozil 228-239 ATP-binding cassette, sub-family B (MDR/TAP), member 4 Mus musculus 20-24 8847480-5 1995 In the current studies, we sought to determine whether the selected fibrates, bezafibrate, clofibrate, fenofibrate, and gemfibrozil, could reduce hepatic apoC-III mRNA and plasma apoC-III levels. Gemfibrozil 120-131 apolipoprotein C3 Homo sapiens 154-162 8847480-5 1995 In the current studies, we sought to determine whether the selected fibrates, bezafibrate, clofibrate, fenofibrate, and gemfibrozil, could reduce hepatic apoC-III mRNA and plasma apoC-III levels. Gemfibrozil 120-131 apolipoprotein C3 Homo sapiens 179-187 8847480-9 1995 Gemfibrozil strongly reduced plasma triglyceride levels and had an intermediate but significant effect on apoC-III mRNA and plasma apoC-III levels. Gemfibrozil 0-11 apolipoprotein C3 Homo sapiens 106-114 8847480-9 1995 Gemfibrozil strongly reduced plasma triglyceride levels and had an intermediate but significant effect on apoC-III mRNA and plasma apoC-III levels. Gemfibrozil 0-11 apolipoprotein C3 Homo sapiens 131-139 8847480-10 1995 Some of the fibrates, especially gemfibrozil also reduced plasma apoC-II levels, an effect that could contribute to the observed triglyceride-lowering effect. Gemfibrozil 33-44 apolipoprotein C2 Homo sapiens 65-72 7578896-5 1995 In the gemfibrozil-treated group a significant decrease of total cholesterol and triglycerides and a significant increase of HDL-C and HDL2-C were found. Gemfibrozil 7-18 junctophilin 3 Homo sapiens 135-139 7595182-6 1995 RESULTS: Gemfibrozil increased the concentration of HDL cholesterol (+11.1%) because of the rise of HDL3 cholesterol (34.5%, P < 0.01). Gemfibrozil 9-20 HDL3 Homo sapiens 100-104 7595182-9 1995 Gemfibrozil increased LPL and HL activities by 18.2% (P < 0.05) and by 19.6%, respectively. Gemfibrozil 0-11 lipoprotein lipase Homo sapiens 22-25 7595182-9 1995 Gemfibrozil increased LPL and HL activities by 18.2% (P < 0.05) and by 19.6%, respectively. Gemfibrozil 0-11 lipase C, hepatic type Homo sapiens 30-32 7595182-10 1995 Plasma CETP activity was also increased during gemfibrozil treatment (+15.8%, P < 0.05). Gemfibrozil 47-58 cholesteryl ester transfer protein Homo sapiens 7-11 7595182-11 1995 CONCLUSION: The gemfibrozil-induced elevation of HDL3 and apoA-II may reflect the combined action of LPL, HL and CETP on plasma HDL metabolism. Gemfibrozil 16-27 HDL3 Homo sapiens 49-53 7595182-11 1995 CONCLUSION: The gemfibrozil-induced elevation of HDL3 and apoA-II may reflect the combined action of LPL, HL and CETP on plasma HDL metabolism. Gemfibrozil 16-27 apolipoprotein A2 Homo sapiens 58-65 7595182-11 1995 CONCLUSION: The gemfibrozil-induced elevation of HDL3 and apoA-II may reflect the combined action of LPL, HL and CETP on plasma HDL metabolism. Gemfibrozil 16-27 lipoprotein lipase Homo sapiens 101-104 7595182-11 1995 CONCLUSION: The gemfibrozil-induced elevation of HDL3 and apoA-II may reflect the combined action of LPL, HL and CETP on plasma HDL metabolism. Gemfibrozil 16-27 lipase C, hepatic type Homo sapiens 106-108 7595182-11 1995 CONCLUSION: The gemfibrozil-induced elevation of HDL3 and apoA-II may reflect the combined action of LPL, HL and CETP on plasma HDL metabolism. Gemfibrozil 16-27 cholesteryl ester transfer protein Homo sapiens 113-117 7578896-7 1995 After 12 weeks of treatment in the gemfibrozil group the release of t-PA:Ag in response to venous occlusion was significantly higher and plasma PAI activity was significantly lower than in placebo group. Gemfibrozil 35-46 plasminogen activator, tissue type Homo sapiens 68-72 7578896-7 1995 After 12 weeks of treatment in the gemfibrozil group the release of t-PA:Ag in response to venous occlusion was significantly higher and plasma PAI activity was significantly lower than in placebo group. Gemfibrozil 35-46 serpin family B member 2 Homo sapiens 144-147 8595642-7 1995 Gemfibrozil was significantly more effective than lovastatin in raising total HDL and HDL3 levels and in lowering the IDL plus VLDL:HDL ratio. Gemfibrozil 0-11 HDL3 Homo sapiens 86-90 7755647-0 1995 Insulin action and glucose metabolism are improved by gemfibrozil treatment in hypertriglyceridemic patients. Gemfibrozil 54-65 insulin Homo sapiens 0-7 7641354-3 1995 We and others have observed attenuation of PAI-1 expression by gemfibrozil both in vivo and in vitro. Gemfibrozil 63-74 serpin family E member 1 Homo sapiens 43-48 7666007-3 1995 Lp[a] reduction was directly related to blood gemfibrozil concentration (range 36-428 microM, r = 0.969) and occurred without concomitant changes in apolipoprotein B. Gemfibrozil 46-57 apolipoprotein(a) Macaca fascicularis 0-4 7666007-7 1995 However, both Lp[a] and apo[a] mRNA in primary cynomolgus monkey hepatocytes were coordinately lowered in a dose-dependent fashion by gemfibrozil. Gemfibrozil 134-145 apolipoprotein(a) Macaca fascicularis 14-18 7666007-8 1995 Thus, Lp[a] can be regulated by gemfibrozil at the level of apo[a] mRNA expression. Gemfibrozil 32-43 apolipoprotein(a) Macaca fascicularis 6-10 7788039-15 1995 This beneficial effect of gemfibrozil, which was expressed by the third month and was evident for some time afterwards, was attributed to a significant reduction of triglyceride and fibrinogen levels, an increase of HDL cholesterol concentrations and a moderate decrease of total cholesterol and LDL cholesterol levels. Gemfibrozil 26-37 fibrinogen beta chain Homo sapiens 182-192 8595642-5 1995 Gemfibrozil significantly decreased triglyceride, very-low-density lipoprotein (VLDL), and intermediate-density lipoprotein (IDL) levels, the total cholesterol:HDL ratio, and the IDL plus VLDL;HDL ratio, and significantly increased levels of HDL, HDL2, and HDL3. Gemfibrozil 0-11 junctophilin 3 Homo sapiens 247-251 8595642-5 1995 Gemfibrozil significantly decreased triglyceride, very-low-density lipoprotein (VLDL), and intermediate-density lipoprotein (IDL) levels, the total cholesterol:HDL ratio, and the IDL plus VLDL;HDL ratio, and significantly increased levels of HDL, HDL2, and HDL3. Gemfibrozil 0-11 HDL3 Homo sapiens 257-261 7752906-7 1995 The rate of insulin-induced whole-body glucose disposal increased similarly both before (basal 10.8 +/- 1.8, low-dose insulin 10.5 +/- 2.1, and high-dose insulin 20.9 +/- 11.9 mumol.kg-1.min-1) and after (11.1 +/- 1.7, 10.7 +/- 1.2, and 18.6 +/- 7.9, respectively) gemfibrozil treatment. Gemfibrozil 265-276 insulin Homo sapiens 12-19 7662602-0 1995 Gemfibrozil and Mediterranean diet for patients with high plasma levels of lipoprotein [Lp(a)] and cholesterol--pilot study. Gemfibrozil 0-11 lipoprotein(a) Homo sapiens 88-93 7662602-3 1995 We have evaluated a combination of medical treatment with Gemfibrozil (600 mg bid) and a Mediterranean diet for 2 months in 15 patients with both hypercholesterolemia (> 240 mg/dl) and high levels of Lp(a) (> 30 mg/dl). Gemfibrozil 58-69 BH3 interacting domain death agonist Homo sapiens 78-81 7662602-3 1995 We have evaluated a combination of medical treatment with Gemfibrozil (600 mg bid) and a Mediterranean diet for 2 months in 15 patients with both hypercholesterolemia (> 240 mg/dl) and high levels of Lp(a) (> 30 mg/dl). Gemfibrozil 58-69 lipoprotein(a) Homo sapiens 203-208 7755644-9 1995 Furthermore, gemfibrozil increased HDL cholesterol (P < 0.05) and normalised the elevated HDL2 and HDL3 TG content. Gemfibrozil 13-24 junctophilin 3 Homo sapiens 93-97 7755644-9 1995 Furthermore, gemfibrozil increased HDL cholesterol (P < 0.05) and normalised the elevated HDL2 and HDL3 TG content. Gemfibrozil 13-24 HDL3 Homo sapiens 102-106 7749807-4 1995 Moreover, the urinary excretion of 11-dehydro-thromboxane-B2 was 54% higher during treatment with gemfibrozil (P < .001), whereas the excretion of beta-thromboglobulin in urine was unaltered. Gemfibrozil 98-109 pro-platelet basic protein Homo sapiens 150-170 7869918-0 1995 A comparison between the effects of gemfibrozil and simvastatin on insulin sensitivity in patients with non-insulin-dependent diabetes mellitus and hyperlipoproteinemia. Gemfibrozil 36-47 insulin Homo sapiens 67-74 7869918-2 1995 After gemfibrozil treatment, the insulin concentration was increased during the major part of the intravenous glucose tolerance test (IVGTT) and during the hyperinsulinemic euglycemic clamp. Gemfibrozil 6-17 insulin Homo sapiens 33-40 7869918-4 1995 Insulin sensitivity decreased by 27% and 28% during gemfibrozil and simvastatin treatment, respectively. Gemfibrozil 52-63 insulin Homo sapiens 0-7 7869918-8 1995 After gemfibrozil treatment, lipoprotein(a) [Lp(a)] was decreased by 24%, and the plasma free fatty acid (FFA) concentration was increased by 20% and skeletal muscle lipoprotein lipase activity (LPLA) by 37%. Gemfibrozil 6-17 lipoprotein(a) Homo sapiens 29-43 8071873-8 1994 Thus, in this rodent model of hypertension and dyslipidemia, gemfibrozil lowers plasma TG levels by 50% with no effect on TG secretion; the hypotriglyceridemic effect is due mainly to an increase in TG removal rate associated with a post-transcriptional increase in LPL activity in skeletal muscle. Gemfibrozil 61-72 lipoprotein lipase Rattus norvegicus 266-269 7826893-2 1994 We compare here the effects of gemfibrozil and placebo on the serum levels of dehydroepiandrosterone (DHEA), its sulfate (DHEAS), their metabolite androstanediol glucuronide (3 alpha-AdiolG), androstenedione, cortisol, testosterone, and sex-hormone binding globulin (SHBG) in non-smokers. Gemfibrozil 31-42 sex hormone binding globulin Homo sapiens 237-265 7826893-2 1994 We compare here the effects of gemfibrozil and placebo on the serum levels of dehydroepiandrosterone (DHEA), its sulfate (DHEAS), their metabolite androstanediol glucuronide (3 alpha-AdiolG), androstenedione, cortisol, testosterone, and sex-hormone binding globulin (SHBG) in non-smokers. Gemfibrozil 31-42 sex hormone binding globulin Homo sapiens 267-271 7826893-5 1994 The mean SHBG levels decreased from 46.4 to 41.7 nmol/l; P = 0.03 with gemfibrozil treatment. Gemfibrozil 71-82 sex hormone binding globulin Homo sapiens 9-13 8077896-0 1994 An interaction between Gemfibrozil and alpha 1-antitrypsin. Gemfibrozil 23-34 serpin family A member 1 Homo sapiens 39-58 8077896-3 1994 Gemfibrozil treatment in non-PiZ patients also resulted in a reduction of alpha 1-AT activity which could be reproduced in vitro. Gemfibrozil 0-11 serpin family A member 1 Homo sapiens 74-84 7917468-5 1994 When HDL of the subjects studied were recombined with the CETP fraction and VLDL/LDL isolated from the control plasma pool, the rate of EC transfer was decreased by 43% after gemfibrozil administration. Gemfibrozil 175-186 cholesteryl ester transfer protein Homo sapiens 58-62 8017467-6 1994 Gemfibrozil reduced LDL-C by 15.8%, TC by 13.4%, VLDL-C by 32.2%, LDL-C:HDL-C by 24.8%, and TG by 34.2%, and increased HDL-C by 13.9% (all changes were statistically significant, p < 0.001) compared with baseline. Gemfibrozil 0-11 component of oligomeric golgi complex 2 Homo sapiens 20-25 8017467-6 1994 Gemfibrozil reduced LDL-C by 15.8%, TC by 13.4%, VLDL-C by 32.2%, LDL-C:HDL-C by 24.8%, and TG by 34.2%, and increased HDL-C by 13.9% (all changes were statistically significant, p < 0.001) compared with baseline. Gemfibrozil 0-11 component of oligomeric golgi complex 2 Homo sapiens 50-55 8017467-7 1994 Gemfibrozil produced significantly greater changes in VLDL-C (p < 0.01), HDL-C (p < 0.001), and TG (p < 0.001), but not in LDL-C: HDL-C, compared with fluvastatin. Gemfibrozil 0-11 component of oligomeric golgi complex 2 Homo sapiens 55-60 8017467-9 1994 At the end of the study, 43.8% of fluvastatin patients and 45% of gemfibrozil patients achieved a reduction of > 20% in LDL-C levels. Gemfibrozil 66-77 component of oligomeric golgi complex 2 Homo sapiens 123-128 8017467-10 1994 Normalization of LDL-C levels was achieved (according to European Atherosclerosis Society guidelines) by 13.4% of fluvastatin- and 14.6% of gemfibrozil-treated patients. Gemfibrozil 140-151 component of oligomeric golgi complex 2 Homo sapiens 17-22 8304363-11 1994 CONCLUSION: Gemfibrozil treatment in patients with familial dysbetalipoproteinemia resulted in a marked reduction of the concentrations of large VLDL, small VLDL, and IDL, and an increase in the levels of HDL, apo A-I, and apo A-II. Gemfibrozil 12-23 apolipoprotein A2 Homo sapiens 223-231 8222487-5 1993 These differences may have a physiologic background because the main effect of gemfibrozil is in the stable HDL3 subfraction, rather than in the variable HDL2. Gemfibrozil 79-90 HDL3 Homo sapiens 108-112 8231846-0 1993 Effect of gemfibrozil on adipose tissue and muscle lipoprotein lipase. Gemfibrozil 10-21 lipoprotein lipase Homo sapiens 51-69 7509512-0 1993 Inhibition of endothelial cell expression of plasminogen activator inhibitor type-1 by gemfibrozil. Gemfibrozil 87-98 serpin family E member 1 Homo sapiens 45-83 7509512-2 1993 In pilot studies designed to identify pharmacologic approaches capable of diminishing such increases, we found that gemfibrozil attenuated the stimulation of synthesis of PAI-1 in a human, immortal, hepatoma cell line (Hep G2) induced by platelets. Gemfibrozil 116-127 serpin family E member 1 Homo sapiens 171-176 7509512-5 1993 Gemfibrozil, 100 microM, suppressed basal PAI-1 production by 15% and attenuated the augmentation of synthesis of PAI-1 induced by lysates from platelets (4 x 10(7)/ml) by 36% over 24 h without inhibiting overall protein synthesis. Gemfibrozil 0-11 serpin family E member 1 Homo sapiens 42-47 7509512-5 1993 Gemfibrozil, 100 microM, suppressed basal PAI-1 production by 15% and attenuated the augmentation of synthesis of PAI-1 induced by lysates from platelets (4 x 10(7)/ml) by 36% over 24 h without inhibiting overall protein synthesis. Gemfibrozil 0-11 serpin family E member 1 Homo sapiens 114-119 8236126-4 1993 After 1 month of therapy, plasma fibrinogen significantly decreased by 9% and 15% in fenofibrate and bezafibrate groups respectively and increased by 19% in gemfibrozil treated patients. Gemfibrozil 157-168 fibrinogen beta chain Homo sapiens 33-43 8458531-2 1993 In the present study we examined whether gemfibrozil, by lowering triglyceride levels, improves the glucoregulatory and antilipolytic action of insulin in Type 2 (non-insulin-dependent) diabetes mellitus. Gemfibrozil 41-52 insulin Homo sapiens 144-151 8509712-5 1993 However, immunoblot analysis of tissue homogenates revealed that SCP2 protein is decreased by 75% in the livers of gemfibrozil-treated animals and increased by 5-fold at 48 h in regenerating liver and in the remaining kidney after unilateral nephrectomy. Gemfibrozil 115-126 sterol carrier protein 2 Rattus norvegicus 65-69 8509712-6 1993 Taken together these results suggest that SCP2 gene expression is developmentally regulated and modulated translationally or post-translationally in the adult rat by gemfibrozil and compensatory cell growth. Gemfibrozil 166-177 sterol carrier protein 2 Rattus norvegicus 42-46 8318063-6 1993 Postheparin lipoprotein lipase (LPL) and hepatic lipase (HL) activities were increased by gemfibrozil therapy while only a mild elevation in LPL activity alone was seen on lovastatin therapy. Gemfibrozil 90-101 lipoprotein lipase Homo sapiens 12-30 8318063-6 1993 Postheparin lipoprotein lipase (LPL) and hepatic lipase (HL) activities were increased by gemfibrozil therapy while only a mild elevation in LPL activity alone was seen on lovastatin therapy. Gemfibrozil 90-101 lipoprotein lipase Homo sapiens 32-35 8318063-6 1993 Postheparin lipoprotein lipase (LPL) and hepatic lipase (HL) activities were increased by gemfibrozil therapy while only a mild elevation in LPL activity alone was seen on lovastatin therapy. Gemfibrozil 90-101 lipase C, hepatic type Homo sapiens 41-55 8257455-6 1993 Gemfibrozil increased the concentration of HDL cholesterol (P < 0.01), which was due to the rise of HDL3 cholesterol (+16%), while in the placebo group these values remained unchanged. Gemfibrozil 0-11 HDL3 Homo sapiens 103-107 8257455-7 1993 Gemfibrozil increased the concentrations of apo A-I(+12.6%, NS), apo A-II (+28.2%, P < 0.01) and Lp A-I:A-II particles (+21.6%, P < 0.06) but there were no changes in the placebo group. Gemfibrozil 0-11 apolipoprotein A1 Homo sapiens 44-51 8257455-7 1993 Gemfibrozil increased the concentrations of apo A-I(+12.6%, NS), apo A-II (+28.2%, P < 0.01) and Lp A-I:A-II particles (+21.6%, P < 0.06) but there were no changes in the placebo group. Gemfibrozil 0-11 apolipoprotein A2 Homo sapiens 65-73 8257455-7 1993 Gemfibrozil increased the concentrations of apo A-I(+12.6%, NS), apo A-II (+28.2%, P < 0.01) and Lp A-I:A-II particles (+21.6%, P < 0.06) but there were no changes in the placebo group. Gemfibrozil 0-11 lipoprotein(a) Homo sapiens 100-104 8257455-11 1993 Gemfibrozil increased LPL and HL activities by 14.7% (P < 0.05) and by 18.8% (P < 0.01), respectively, while in the placebo group LPL and HL activities remained unchanged. Gemfibrozil 0-11 lipoprotein lipase Homo sapiens 22-25 8257455-11 1993 Gemfibrozil increased LPL and HL activities by 14.7% (P < 0.05) and by 18.8% (P < 0.01), respectively, while in the placebo group LPL and HL activities remained unchanged. Gemfibrozil 0-11 lipase C, hepatic type Homo sapiens 30-32 8257455-12 1993 Plasma CETP activity was also increased during gemfibrozil treatment while in the placebo group it remained unchanged. Gemfibrozil 47-58 cholesteryl ester transfer protein Homo sapiens 7-11 8257455-14 1993 The gemfibrozil-induced elevation of HDL3 and dense HDL subpopulations may reflect the concerted action of LPL, HL and CETP on plasma HDL metabolism. Gemfibrozil 4-15 HDL3 Homo sapiens 37-41 8257455-14 1993 The gemfibrozil-induced elevation of HDL3 and dense HDL subpopulations may reflect the concerted action of LPL, HL and CETP on plasma HDL metabolism. Gemfibrozil 4-15 lipoprotein lipase Homo sapiens 107-110 8257455-14 1993 The gemfibrozil-induced elevation of HDL3 and dense HDL subpopulations may reflect the concerted action of LPL, HL and CETP on plasma HDL metabolism. Gemfibrozil 4-15 lipase C, hepatic type Homo sapiens 112-114 8257455-14 1993 The gemfibrozil-induced elevation of HDL3 and dense HDL subpopulations may reflect the concerted action of LPL, HL and CETP on plasma HDL metabolism. Gemfibrozil 4-15 cholesteryl ester transfer protein Homo sapiens 119-123 8458531-2 1993 In the present study we examined whether gemfibrozil, by lowering triglyceride levels, improves the glucoregulatory and antilipolytic action of insulin in Type 2 (non-insulin-dependent) diabetes mellitus. Gemfibrozil 41-52 insulin Homo sapiens 167-174 8458531-9 1993 Rate of whole body glucose disposal during a low-dose insulin infusion (serum insulin -90 pmol/l) (pre- vs post-gemfibrozil 11.9 +/- 1.1 vs 11.1 +/- 0.7, pre- vs post-placebo 9.9 +/- 1.1 vs 10.8 +/- 0.8 mumol.kg-1.min-1, NS for both) and a high-dose insulin infusion (serum insulin approximately 500 pmol/l) (16.2 +/- 1.7 vs 17.7 +/- 2.7, 17.1 +/- 4.2 vs 17.4 +/- 2.9 mumol.kg-1 x min-1, respectively, NS for both) remained unchanged. Gemfibrozil 112-123 insulin Homo sapiens 54-61 1397665-2 1992 After oral treatment with 450 mg gemfibrozil twice daily for 28 days, triglyceride concentrations were reduced in serum, chylomicrons, VLDL and low-density lipoprotein, and total cholesterol concentrations were reduced in serum, chylomicrons and VLDL, and increased in HDL2 plus HDL3. Gemfibrozil 33-44 HDL3 Homo sapiens 279-283 8418887-4 1993 A phosphocholine cytidylyltransferase-mediated rise in cellular CDP choline content may explain the gemfibrozil-dependent rise in phosphatidylcholine biosynthesis since homogenates of monolayers incubated with CDP choline preferentially incorporate labeled diacylglycerol into phosphatidylcholine rather than triacylglycerol. Gemfibrozil 100-111 cut-like homeobox 1 Rattus norvegicus 64-67 8418887-4 1993 A phosphocholine cytidylyltransferase-mediated rise in cellular CDP choline content may explain the gemfibrozil-dependent rise in phosphatidylcholine biosynthesis since homogenates of monolayers incubated with CDP choline preferentially incorporate labeled diacylglycerol into phosphatidylcholine rather than triacylglycerol. Gemfibrozil 100-111 cut-like homeobox 1 Rattus norvegicus 210-213 1613316-3 1992 Gemfibrozil and lovastatin caused primarily similar alterations in HDL components in HDL2 and HDL3 subfractions. Gemfibrozil 0-11 junctophilin 3 Homo sapiens 85-89 1613316-3 1992 Gemfibrozil and lovastatin caused primarily similar alterations in HDL components in HDL2 and HDL3 subfractions. Gemfibrozil 0-11 HDL3 Homo sapiens 94-98 1613316-6 1992 Gemfibrozil increased the cholesterol concentrations of HDL2 and HDL3 (p less than 0.05 for both), and lovastatin caused significant increases in HDL2 (p less than 0.05) and HDL3 phospholipids (p less than 0.01). Gemfibrozil 0-11 junctophilin 3 Homo sapiens 56-60 1613316-6 1992 Gemfibrozil increased the cholesterol concentrations of HDL2 and HDL3 (p less than 0.05 for both), and lovastatin caused significant increases in HDL2 (p less than 0.05) and HDL3 phospholipids (p less than 0.01). Gemfibrozil 0-11 HDL3 Homo sapiens 65-69 1397665-4 1992 It is concluded that gemfibrozil is effective in lowering triglycerides and cholesterol, particularly in triglyceride-rich particles, and raising the cholesterol content of HDL2 plus HDL3. Gemfibrozil 21-32 HDL3 Homo sapiens 183-187 1534991-4 1992 These new observations suggest that gemfibrozil treatment increases the production and turnover of DHEA and DHEAS and may in addition stimulate the 5 alpha-reduction of androgens. Gemfibrozil 36-47 sulfotransferase family 2A member 1 Homo sapiens 108-113 1519208-4 1992 Plasma prothrombin fragment F1 + 2 concentration, a marker of the in vivo rate of generation of thrombin, was 25 (12 to 37)% lower on average while on gemfibrozil than during the placebo phase. Gemfibrozil 151-162 coagulation factor II, thrombin Homo sapiens 10-18 1572044-6 1992 METHODS AND RESULTS: To determine whether pharmacological concentrations of gemfibrozil directly affect PAI-1 synthesis, we characterized its effects on a human hepatoma cell line (Hep G2) in vitro. Gemfibrozil 76-87 serpin family E member 1 Homo sapiens 104-109 1572044-7 1992 Gemfibrozil decreased basal PAI-1 secretion by 43% and attenuated the augmentation of PAI-1 synthesis over 24 hours induced by EGF and TGF-beta by 37% and 39% without altering overall protein synthesis. Gemfibrozil 0-11 serpin family E member 1 Homo sapiens 28-33 1572044-7 1992 Gemfibrozil decreased basal PAI-1 secretion by 43% and attenuated the augmentation of PAI-1 synthesis over 24 hours induced by EGF and TGF-beta by 37% and 39% without altering overall protein synthesis. Gemfibrozil 0-11 serpin family E member 1 Homo sapiens 86-91 1572044-7 1992 Gemfibrozil decreased basal PAI-1 secretion by 43% and attenuated the augmentation of PAI-1 synthesis over 24 hours induced by EGF and TGF-beta by 37% and 39% without altering overall protein synthesis. Gemfibrozil 0-11 epidermal growth factor Homo sapiens 127-130 1572044-10 1992 CONCLUSIONS: Beneficial effects of gemfibrozil in reducing coronary events in hypertriglyceridemic patients may depend, in part, on potentiation of fibrinolysis by direct diminution of synthesis of endogenous PAI-1. Gemfibrozil 35-46 serpin family E member 1 Homo sapiens 209-214 1534991-2 1992 Simultaneously we studied the effects of gemfibrozil treatment on the serum levels of dehydroepiandrosterone (DHEA), its sulfate (DHEAS), and their metabolite androstanediol glucuronide (3 alpha AdiolG) with those of placebo. Gemfibrozil 41-52 sulfotransferase family 2A member 1 Homo sapiens 130-135 1619364-4 1992 In contrast, hepatic LCAT mRNA levels decreased to 67%, 64%, and 46% of the control levels after treatment with the fibric acid derivatives clofibrate, gemfibrozil, and fenofibrate, respectively. Gemfibrozil 152-163 lecithin cholesterol acyltransferase Rattus norvegicus 21-25 1547188-9 1992 Both clofibrate and gemfibrozil had effects comparable to those of fenofibrate on liver and intestinal apolipoprotein mRNA levels except for liver apo A-II mRNA, which decreased only marginally. Gemfibrozil 20-31 apolipoprotein E Rattus norvegicus 103-117 1547188-10 1992 Compared with fenofibrate, clofibrate caused similar changes in plasma cholesterol, apo A-I, apo A-IV, and apo E concentrations, whereas gemfibrozil increased plasma cholesterol and apo E without changing apo A-I and apo A-IV concentrations. Gemfibrozil 137-148 apolipoprotein E Rattus norvegicus 182-187 1811554-1 1991 Exposure of HepG2 and Hep3B cells to gemfibrozil (40 micrograms/ml), a hypolipidemic drug, resulted in a 2-fold induction of apo AI mRNA and, a one-third reduction in apo B mRNA but had no significant effect on apo E mRNA levels. Gemfibrozil 37-48 apolipoprotein A1 Homo sapiens 125-131 1471924-2 1992 They were treated with gemfibrozil (600 mg BID) for 12 weeks. Gemfibrozil 23-34 BH3 interacting domain death agonist Homo sapiens 43-46 1789808-9 1991 It is suggested that the HMG-CoA reductase inhibition observed in mononuclear cells during gemfibrozil treatment is due to changes in LDL structure affecting LDL receptor binding rather than direct effects of the drug on cellular cholesterol metabolism. Gemfibrozil 91-102 low density lipoprotein receptor Homo sapiens 158-170 1811554-2 1991 The hypothesis that the mechanism of action of gemfibrozil involved the cytochrome P-450 system was tested by using ketoconazole, a P-450 inhibitor, which blocks the formation of endogenous polar sterols. Gemfibrozil 47-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 1811554-8 1991 The induction of apo AI mRNA by gemfibrozil was not apparent when the cells were simultaneously treated with ketoconazole. Gemfibrozil 32-43 apolipoprotein A1 Homo sapiens 17-23 1907081-10 1991 Gemfibrozil significantly increased plasma HDL cholesterol, apolipoprotein (apo) AI, and apo AII by 36%, 29%, and 38% from baseline, respectively. Gemfibrozil 0-11 apolipoprotein A1 Homo sapiens 60-83 1678324-0 1991 DNA polymorphisms of apolipoprotein B and AI/CIII genes and response to gemfibrozil treatment. Gemfibrozil 72-83 apolipoprotein B Homo sapiens 21-37 1678324-6 1991 These data indicate that common polymorphisms of the apolipoprotein B and apolipoprotein AI/CIII gene loci influence serum lipid levels by mechanisms that are amenable to an intervention with gemfibrozil. Gemfibrozil 192-203 apolipoprotein B Homo sapiens 53-91 1793455-0 1991 Effects of gemfibrozil on serum apolipoprotein E distribution in hypercholesterolemic patients. Gemfibrozil 11-22 apolipoprotein E Homo sapiens 32-48 1992957-5 1991 Filicol and gemfibrozil caused similar decrements of total cholesterol (14% for both), low-density lipoprotein cholesterol (20% and 18%, respectively), and apolipoprotein B (16% and 21%, respectively). Gemfibrozil 12-23 apolipoprotein B Homo sapiens 156-172 1992957-7 1991 Gemfibrozil, but not filicol, significantly increased plasma high-density lipoprotein cholesterol (16%) and apolipoprotein A-I (17%) levels and reduced triglyceride levels (35%). Gemfibrozil 0-11 apolipoprotein A1 Homo sapiens 108-126 1907081-12 1991 Gemfibrozil increased synthetic rates of apo AI and apo AII by 27% and 34%, respectively, without changing the FCR. Gemfibrozil 0-11 apolipoprotein A1 Homo sapiens 41-47 2073911-6 1990 Ultracentrifuge analysis in a subsample of the cohort revealed that gemfibrozil raised the level of HDL3-cholesterol but had little effect on HDL2-cholesterol. Gemfibrozil 68-79 HDL3 Homo sapiens 100-104 2025787-5 1991 Gemfibrozil lowered plasma triglycerides, VLDL cholesterol and apolipoprotein B (apoB), increased HDL cholesterol and apoAI levels in both groups, and induced a very substantial reduction in LDL cholesterol in type IIa patients only. Gemfibrozil 0-11 apolipoprotein B Homo sapiens 63-79 2025787-5 1991 Gemfibrozil lowered plasma triglycerides, VLDL cholesterol and apolipoprotein B (apoB), increased HDL cholesterol and apoAI levels in both groups, and induced a very substantial reduction in LDL cholesterol in type IIa patients only. Gemfibrozil 0-11 apolipoprotein B Homo sapiens 81-85 1898694-7 1991 Apolipoprotein B decrease was 21% with pravastatin and 13% with gemfibrozil. Gemfibrozil 64-75 apolipoprotein B Homo sapiens 0-16 1997786-2 1991 The most noteworthy finding was the 50% decrease in the cytoplasmic desmin fraction in cells treated with gemfibrozil in comparison to control cultures, and the 19% increase in the cytoskeletal fraction in cultures treated with gemfibrozil and with bezafibrate. Gemfibrozil 106-117 desmin Gallus gallus 68-74 1997786-3 1991 Vimentin accumulation by cells treated with bezafibrate was similar to that in control cultures, however the cytoskeletal vimentin fraction rose by 26% after treatment with gemfibrozil, and fell 13% after treatment with fenofibrate. Gemfibrozil 173-184 vimentin Gallus gallus 122-130 2203245-7 1990 Gemfibrozil additionally stimulates apolipoprotein AI synthesis. Gemfibrozil 0-11 apolipoprotein A1 Homo sapiens 36-53 2407250-6 1990 Gemfibrozil increased the level of HDL-cholesterol with the main effect on HDL3-subfraction. Gemfibrozil 0-11 HDL3 Homo sapiens 75-79 2256470-9 1990 The fibric acid derivative gemfibrozil inhibits adipose lipolysis and enhances lipoprotein lipase activity thus decreasing LDL synthesis and increasing its removal. Gemfibrozil 27-38 lipoprotein lipase Homo sapiens 79-97 2076620-6 1990 Furthermore, plasma fibrinogen levels increased by a mean of 17.6%, a potentially adverse effect of gemfibrozil that has not been previously reported. Gemfibrozil 100-111 fibrinogen beta chain Homo sapiens 20-30 34365128-2 2021 A series of new gemfibrozil derivatives were synthesized and evaluated for sGC activation. Gemfibrozil 16-27 serglycin Mus musculus 75-78 2343430-0 1990 [Post-heparin LPL and HL activities after two months" treatment with gemfibrozil. Gemfibrozil 69-80 lipoprotein lipase Homo sapiens 14-17 2343430-3 1990 Though lipidic parameters were not modified after treatment, gemfibrozil increased the LPL and HL plasmatic activities measured 10 minutes after heparin injection. Gemfibrozil 61-72 lipoprotein lipase Homo sapiens 87-90 23008696-2 2012 The PPARalpha agonists GW7647, WY14643, and gemfibrozil acutely relaxed aortas held under isometric tension and dilated pressurized MCAs with the following order of potency: GW7647>>WY14643>gemfibrozil. Gemfibrozil 44-55 peroxisome proliferator activated receptor alpha Mus musculus 4-13 23008696-2 2012 The PPARalpha agonists GW7647, WY14643, and gemfibrozil acutely relaxed aortas held under isometric tension and dilated pressurized MCAs with the following order of potency: GW7647>>WY14643>gemfibrozil. Gemfibrozil 199-210 peroxisome proliferator activated receptor alpha Mus musculus 4-13 34365128-3 2021 The structure-activity relationship study identified the positions in gemfibrozil"s scaffold that are detrimental for sGC activation and those that are amendable for optimizing modifications. Gemfibrozil 70-81 serglycin Mus musculus 118-121 34365128-4 2021 Compared with gemfibrozil, compounds 7c and 15b were more potent activators of cGMP-forming activity of purified sGC and exhibited enhanced relaxation of preconstricted mouse thoracic aorta rings. Gemfibrozil 14-25 serglycin Mus musculus 113-116 34365128-5 2021 These studies established the overall framework needed for futher improvement of sGC activators based on gemfibrozil scaffold. Gemfibrozil 105-116 serglycin Mus musculus 81-84 34499887-5 2021 Gemfibrozil, a pharmacological agonist of PPARalpha, increased PPARalpha levels and activity in podocytes. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 42-51 34499887-5 2021 Gemfibrozil, a pharmacological agonist of PPARalpha, increased PPARalpha levels and activity in podocytes. Gemfibrozil 0-11 peroxisome proliferator activated receptor alpha Mus musculus 63-72 34499887-11 2021 This study identified a novel mechanism of the PPARalpha agonist gemfibrozil that exerts its protective effects by inhibiting PAN-induced apoptosis and cytoskeleton rearrangements through inhibition of ANGPTL3 expression. Gemfibrozil 65-76 peroxisome proliferator activated receptor alpha Mus musculus 47-56 34499887-11 2021 This study identified a novel mechanism of the PPARalpha agonist gemfibrozil that exerts its protective effects by inhibiting PAN-induced apoptosis and cytoskeleton rearrangements through inhibition of ANGPTL3 expression. Gemfibrozil 65-76 angiopoietin-like 3 Mus musculus 202-209 35163693-0 2022 Structural Analysis of Human Serum Albumin in Complex with the Fibrate Drug Gemfibrozil. Gemfibrozil 76-87 albumin Homo sapiens 29-42 35163693-4 2022 Most of them appear to be related to the ability of GEM to bind with high affinity human serum albumin (HSA), the major drug-carrier protein in blood plasma. Gemfibrozil 52-55 albumin Homo sapiens 89-102 34303168-7 2021 In the presence of Cl-, we found that Cl2 - and in particular ClO were responsible for the enhanced degradation with increasing Cl- concentrations due to the considerable ClO reactivity of gemfibrozil (1.93 x 109 M-1 s-1) and naproxen (9.24 x 109 M-1 s-1) and the rapid transformation of Cl2 - to ClO . Gemfibrozil 191-202 endogenous retrovirus group W member 5 Homo sapiens 38-41