Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
1.
Pharm Biol ; 60(1): 282-293, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35138992

ABSTRACT

CONTEXT: Cordia dichotoma Forst. (Boraginaceae) has potent pharmacological impact. Meanwhile, its effect on fertility is unclear. OBJECTIVE: This study investigates the effect of Cordia fresh fruits hydroethanolic extract on fertility. MATERIALS AND METHODS: 120 Wistar albino male rats were divided into four groups (n = 30). The first group was negative control, and the second, third, and fourth groups received 125, 250, and 500 mg extract/kg bodyweight for 56 days. After 56 days, Cordia force-feeding stopped, and all groups were kept under laboratory conditions for another month to study the recovering effect. RESULTS: After day 56, extract at 500 mg/kg significantly reduced sperm total count, motility%, and alive%, to 47.60 ± 2.27 × 106 sperm/mL, 43.33% ± 1.49, and 63.67% ± 1.19, respectively, abnormalities% increased considerably (26.67% ± 0.54), compared to the negative control. Also, significant depletion on follicle-stimulating hormone (2.66 ± 0.21 mIU/L), luteinizing hormone (1.07 ± 0.06 mIU/L), and testosterone (2.69 ± 0.13 nmol/L) level was recorded, compared to the negative control. Cordia negative effect showed on histopathological studies of testes, prostate, and seminal vesicles. Fortunately, these adverse effects of Cordia recovered remarkably after stopping administration for one month. CONCLUSIONS: Cordia antifertility effect may be due to its hypocholesterolemic effect, where cholesterol, the steroid cycle precursor, was significantly reduced. This study can be incorporated in clinical research after being repeated on another small experimental animal, their offspring, and one large experimental animal, then going to a clinical study that we plan to do in the future.


Subject(s)
Cordia/chemistry , Plant Extracts/toxicity , Spermatozoa/drug effects , Testis/drug effects , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/isolation & purification , Anticholesteremic Agents/toxicity , Dose-Response Relationship, Drug , Follicle Stimulating Hormone/metabolism , Fruit , Luteinizing Hormone/metabolism , Male , Plant Extracts/administration & dosage , Rats , Rats, Wistar , Testis/pathology , Testosterone/metabolism
2.
Article in English | MEDLINE | ID: mdl-34756985

ABSTRACT

Atorvastatin (ATV) and gemfibrozil (GEM) are two typical lipid-lowering pharmaceuticals with different action modes, which are frequently detected in various water bodies owning to their wide usage. However, there is limited information about their effects on Daphnia magna. The present study addressed and compared the toxic effects of ATV and GEM on D. magna through determining the responses of the stress related genes (including Nrf2, Keap1, HO-1, GCLC, p53 and PIG3) in D. magna for 24 h and 48 h acute exposure and the changes of life history traits and swimming behaviors in a 21 days chronic exposure under different concentrations of ATV and GEM exposure (5 µg L-1, 50 µg L-1, 500 µg L-1 and 5000 µg L-1). Results showed that the expression of Nrf2, Keap1, HO-1, GCLC, p53 and PIG3 were induced to various degrees under the ATV exposure. There were similar performances for GEM. ATV and GEM caused the delay of first brooding and hatching time and decrease of eggs production number, especially in GEM exposure, reproduction of Daphnia was significantly inhibited, decreasing 38.51% compared to the control. ATV and GEM increased the heart rate of D. magna, and changed swimming behaviors of D. magna. In summary, two lipid-lowering pharmaceuticals caused oxidative stress on D. magna, subsequently brought about alterations in physiological traits. Comparatively, ATV pose more higher risks to D. magna than GEM, but the detailed action mechanisms of ATV and GEM on D. magna needs more investigations in future.


Subject(s)
Anticholesteremic Agents/toxicity , Atorvastatin/toxicity , Gemfibrozil/toxicity , Toxicity Tests , Animals , Daphnia , Gene Expression Regulation/drug effects , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism
3.
J Med Chem ; 64(18): 13215-13258, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34375108

ABSTRACT

Cholesteryl ester transfer protein (CETP) represents one of the key regulators of the homeostasis of lipid particles, including high-density lipoprotein (HDL) and low-density lipoprotein (LDL) particles. Epidemiological evidence correlates increased HDL and decreased LDL to coronary heart disease (CHD) risk reduction. This relationship is consistent with a clinical outcomes trial of a CETP inhibitor (anacetrapib) combined with standard of care (statin), which led to a 9% additional risk reduction compared to standard of care alone. We discuss here the discovery of MK-8262, a CETP inhibitor with the potential for being the best-in-class molecule. Novel in vitro and in vivo paradigms were integrated to drug discovery to guide optimization informed by a critical understanding of key clinical adverse effect profiles. We present preclinical and clinical evidence of MK-8262 safety and efficacy by means of HDL increase and LDL reduction as biomarkers for reduced CHD risk.


Subject(s)
Anticholesteremic Agents/therapeutic use , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Coronary Disease/drug therapy , Oxazolidinones/therapeutic use , Animals , Anticholesteremic Agents/chemical synthesis , Anticholesteremic Agents/pharmacokinetics , Anticholesteremic Agents/toxicity , Dogs , Humans , Macaca mulatta , Mice, Inbred C57BL , Molecular Structure , Oxazolidinones/chemical synthesis , Oxazolidinones/pharmacokinetics , Oxazolidinones/toxicity , Rats, Wistar , Structure-Activity Relationship
4.
Toxicol Appl Pharmacol ; 422: 115557, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33932462

ABSTRACT

CSL112 (apolipoprotein A-I, apo AI [human]) is an investigational drug in Phase 3 development for risk reduction of early recurrent cardiovascular events following an acute myocardial infarction (AMI). Although CSL112 is known to be well tolerated with a regimen of four weekly 6 g intravenous infusions after AMI, high doses of reconstituted apo AI preparations can transiently elevate liver enzymes in rats, raising the possibility of additive liver toxicity and toxicokinetic (TK) effects upon co-administration with cholesterol-lowering drugs, i.e., HMG-CoA reductase and proprotein convertase subtilisin/kexin type 9 inhibitors. We performed a toxicity and TK study in CD rats assigned to eleven treatment groups, including two dose levels of intravenous (IV) CSL112 (140 mg/kg, low-dose; 600 mg/kg, high-dose) administered as a single dose, alone or with intravenous alirocumab 50 mg/kg/week and/or oral atorvastatin 10 mg/kg/day. In addition, control groups of atorvastatin and alirocumab alone and in combination were investigated. Results showed some liver enzyme elevations (remaining <2-fold of baseline) related to administration of CSL112 alone. There was limited evidence of an additive effect of CSL112 on liver enzymes when combined, at either dose level, with alirocumab and/or atorvastatin, and histology revealed no evidence of an increased incidence or severity of hepatocyte vacuolation compared to the control treatments. Co-administration of the study drugs had minimal effect on their respective exposure levels, and on levels of total cholesterol and high-density lipoprotein cholesterol. These data support concomitant use of CSL112 with alirocumab and/or atorvastatin with no anticipated negative impact on liver safety and TK.


Subject(s)
Antibodies, Monoclonal, Humanized/toxicity , Anticholesteremic Agents/toxicity , Atorvastatin/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Lipoproteins, HDL/toxicity , Liver/drug effects , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Anticholesteremic Agents/pharmacokinetics , Atorvastatin/pharmacokinetics , Biomarkers/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cholesterol/blood , Drug Interactions , Female , Lipoproteins, HDL/pharmacokinetics , Liver/metabolism , Liver/pathology , Male , Rats, Sprague-Dawley , Risk Assessment , Toxicity Tests , Toxicokinetics
5.
J Appl Toxicol ; 41(7): 1076-1088, 2021 07.
Article in English | MEDLINE | ID: mdl-33073877

ABSTRACT

Statins belong to the most often prescribed medications, which efficiently normalise hyperlipidaemia and prevent cardiovascular complications in obese and diabetic patients. However, beside expected therapeutic results based on the inhibition of 3-hydroxyl-3-methylglutaryl-CoA reductase, these drugs exert multiple side effects of poorly understood characteristic. In this study, side effects of pravastatin and atorvastatin on EA.hy926 endothelial cell line were investigated. It was found that both statins activate proinflammatory response, elevate nitric oxide and reactive oxygen species (ROS) generation and stimulate antioxidative response in these cells. Moreover, only slight stimulation of the mitochondrial biogenesis and significant changes in the mitochondrial network organisation have been noted. Although biochemical bases behind these effects are not clear, they may partially be explained as an elevation of AMP-activated protein kinase (AMPK) activity and an increased activating phosphorylation of sirtuin 1 (Sirt1), which were observed in statins-treated cells. In addition, both statins increased nicotinamide N-methyltransferase (NNMT) protein level that may explain a reduced fraction of methylated histone H3. Interestingly, a substantial reduction of the total level of histone H3 in cells treated with pravastatin but not atorvastatin was also observed. These results indicate a potential additional biochemical target for statins related to reduced histone H3 methylation due to increased NNMT protein level. Thus, NNMT may directly modify gene activity.


Subject(s)
Anticholesteremic Agents/toxicity , Atorvastatin/toxicity , Nicotinamide N-Methyltransferase/metabolism , Nitric Oxide/metabolism , Pravastatin/toxicity , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Cell Line , Cells, Cultured , Endothelial Cells/drug effects , Histones/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Mitochondria/metabolism , Pyrroles
6.
Arterioscler Thromb Vasc Biol ; 40(10): 2481-2493, 2020 10.
Article in English | MEDLINE | ID: mdl-32847390

ABSTRACT

OBJECTIVE: HDL (high-density lipoprotein) role in atherosclerosis is controversial. Clinical trials with CETP (cholesterylester transfer protein)-inhibitors have not provided benefit. We have shown that HDL remodeling in hypercholesterolemia reduces HDL cardioprotective potential. We aimed to assess whether hypercholesterolemia affects HDL-induced atherosclerotic plaque regression. Approach and Results: Atherosclerosis was induced in New Zealand White rabbits for 3-months by combining a high-fat-diet and double-balloon aortic denudation. Then, animals underwent magnetic resonance imaging (basal plaque) and randomized to receive 4 IV infusions (1 infusion/wk) of HDL isolated from normocholesterolemic (NC-HDL; 75 mg/kg; n=10), hypercholesterolemic (HC-HDL; 75 mg/Kg; n=10), or vehicle (n=10) rabbits. Then, animals underwent a second magnetic resonance imaging (end plaque). Blood, aorta, and liver samples were obtained for analyses. Follow-up magnetic resonance imaging revealed that NC-HDL administration regressed atherosclerotic lesions by 4.3%, whereas, conversely, the administration of HC-HDLs induced a further 6.5% progression (P<0.05 versus basal). Plaque characterization showed that HC-HDL administered animals had a 2-fold higher lipid and cholesterol content versus those infused NC-HDL and vehicle (P<0.05). No differences were observed among groups in CD31 levels, nor in infiltrated macrophages or smooth muscle cells. Plaques from HC-HDL administered animals exhibited higher Casp3 (caspase 3) content (P<0.05 versus vehicle and NC-HDL) whereas plaques from NC-HDL infused animals showed lower expression of Casp3, Cox1 (cyclooxygenase 1), inducible nitric oxide synthase, and MMP (metalloproteinase) activity (P<0.05 versus HC-HDL and vehicle). HDLs isolated from animals administered HC-HDL displayed lower antioxidant potential and cholesterol efflux capacity as compared with HDLs isolated from NC-HDL-infused animal and vehicle or donor HDL (P<0.05). There were no differences in HDL-ApoA1 content, ABCA1 (ATP-binding cassette transporter A1) vascular expression, and SRB1 (scavenger receptor B1) and ABCA1 liver expression. CONCLUSIONS: HDL particles isolated from a hypercholesterolemic milieu lose their ability to regress and stabilize atherosclerotic lesions. Our data suggest that HDL remodeling in patients with co-morbidities may lead to the loss of HDL atheroprotective functions.


Subject(s)
Anticholesteremic Agents/administration & dosage , Aorta, Abdominal/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Cholesterol, HDL/administration & dosage , Hypercholesterolemia/drug therapy , Magnetic Resonance Imaging , Plaque, Atherosclerotic , Animals , Anticholesteremic Agents/toxicity , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/metabolism , Aortic Diseases/blood , Aortic Diseases/diagnostic imaging , Aortic Diseases/etiology , Atherosclerosis/blood , Atherosclerosis/diagnostic imaging , Atherosclerosis/etiology , Biomarkers/blood , Cholesterol, HDL/blood , Cholesterol, HDL/toxicity , Disease Models, Animal , Disease Progression , Hypercholesterolemia/blood , Hypercholesterolemia/complications , Infusions, Intravenous , Male , Rabbits
7.
Turk Kardiyol Dern Ars ; 48(4): 410-424, 2020 06.
Article in English | MEDLINE | ID: mdl-32519978

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the effectiveness of plants used in the formulations of traditional Chinese medicine (TCM), which were also used in clinical trials to treat patients with the novel coronavirus COVID-19, and to assess their effects on the cardiovascular system. METHODS: A literature review of PubMed, ResearchGate, ScienceDirect, the Cochrane Library, and TCM monographs was conducted and the effects of the plants on the cardiovascular system and the mechanisms of action in COVID-19 treatment were evaluated. RESULTS: The mechanism of action, cardiovascular effects, and possible toxicity of 10 plants frequently found in TCM formulations that were used in the clinical treatment of COVID-19 were examined. CONCLUSION: TCM formulations that had been originally developed for earlier viral diseases have been used in COVID-19 treatment. Despite the effectiveness seen in laboratory and animal studies with the most commonly used plants in these formulations, the clinical studies are currently insufficient according to standard operating procedures. More clinical studies are needed to understand the safe clinical use of traditional plants.


Subject(s)
Cardiovascular System/drug effects , Coronavirus Infections/therapy , Drugs, Chinese Herbal/therapeutic use , Medicine, Chinese Traditional , Pneumonia, Viral/therapy , Animals , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Anti-Arrhythmia Agents/toxicity , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/toxicity , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Anticholesteremic Agents/toxicity , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Antihypertensive Agents/toxicity , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Antiviral Agents/toxicity , COVID-19 , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channel Blockers/toxicity , Drug Interactions , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/toxicity , Humans , Pandemics , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation Inhibitors/toxicity , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use , Vasodilator Agents/toxicity
8.
J Toxicol Environ Health A ; 83(3): 113-125, 2020 02 01.
Article in English | MEDLINE | ID: mdl-32116137

ABSTRACT

Simvastatin (SIM), a hypocholesterolaemic drug belonging to the statins group, is a widely prescribed pharmaceutical for prevention of cardiovascular diseases. Several studies showed that lipophilic statins, as SIM, cross the blood-brain barrier and interfere with the energy metabolism of the central nervous system in humans and mammalian models. In fish and other aquatic organisms, the effects of SIM on the brain energy metabolism are unknown, particularly following exposure to low environmentally relevant concentrations. Therefore, the present study aimed at investigating the influence of SIM on gene signaling pathways involved in brain energy metabolism of adult zebrafish (Danio rerio) following chronic exposure (90 days) to environmentally relevant SIM concentrations ranging from 8 ng/L to 1000 ng/L. Real-time PCR was used to determine the transcript levels of several genes involved in different pathways of the brain energy metabolism (glut1b, gapdh, acadm, accα, fasn, idh3a, cox4i1, and cox5aa). The findings here reported integrated well with ecological and biochemical responses obtained in a parallel study. Data demonstrated that SIM modulates transcription of key genes involved in the mitochondrial electron transport chain, in glucose transport and metabolism, in fatty acid synthesis and ß-oxidation. Further, SIM exposure led to a sex-dependent transcription profile for some of the studied genes. Overall, the present study demonstrated, for the first time, that SIM modulates gene regulation of key pathways involved in the energy metabolism in fish brain at environmentally relevant concentrations.


Subject(s)
Brain/drug effects , Brain/metabolism , Gene Expression Regulation/drug effects , Simvastatin/toxicity , Water Pollutants, Chemical/toxicity , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/toxicity , Biological Assay , Drug Administration Schedule , Female , Humans , Male , Simvastatin/administration & dosage , Water Pollutants, Chemical/administration & dosage , Zebrafish
9.
Toxicol Lett ; 305: 58-64, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30735765

ABSTRACT

Myopathy is a well-known adverse effect of statins, affecting a large sector of statins users. The reported experimental data emphasized on mechanistic study of statin myopathy on large muscles. Clinically, both large muscles and respiratory muscles are reported to be involved in the myotoxic profile of statins. However, the experimental data investigating the myopathic mechanism on respiratory muscles are still lacking. The present work aimed to study the effect of atorvastatin treatment on respiratory muscles using rat isolated hemidiaphragm in normoxic & hypoxic conditions. The contractile activity of isolated hemidiaphragm in rats treated with atorvastatin for 21 days was investigated using nerve stimulated technique. Muscle twitches, train of four and tetanic stimulation was measured in normoxic, hypoxic and reoxygenation conditions. Atorvastatin significantly increased the tetanic fade, a measure of muscle fatigability, in hypoxic conditions. Upon reoxygenation, rat hemidiaphragm regains its normal contractile profile. Co-treatment with coenzyme Q10 showed significant improvement in defective diaphragmatic contractility in hypoxic conditions. This work showed that atorvastatin treatment rapidly deteriorates diaphragmatic activity in low oxygen environment. The mitochondrial respiratory dysfunction is probably the mechanism behind such finding. This was supported by the improvement of muscle contractile activity following CoQ10 co-treatment.


Subject(s)
Anticholesteremic Agents/toxicity , Atorvastatin/toxicity , Hypoxia/metabolism , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Animals , Male , Rats , Rats, Sprague-Dawley , Ubiquinone/metabolism
10.
Sci Total Environ ; 651(Pt 1): 399-409, 2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30240922

ABSTRACT

Simvastatin is one of the most commonly cholesterol-lowering prescribed drugs all over the world. With the increase of consumption of these pharmaceuticals and subsequent their discharge into the aquatic environment in recent years, they are present at detectable levels in most sewage effluents. Unfortunately, limited information is provided about their potential impacts on aquatic organisms, especially on the detoxification-related metabolism in fish. In the present study, one local native benthic fish (Mugilogobius abei) in southern China was employed as test species and exposed to SV (0.5 µg L-1, 5 µg L-1, 50 µg L-1 and 500 µg L-1) for 72 h. The transcriptional expression of nucleus transcriptional factor pregnane X receptor (PXR) and its downstream targeted genes including multixenobiotics resistance protein or permeability glycoprotein (P-gp), cytochrome 1A (CYP1A), cytochrome P450 3A (CYP3A), glutathione-S-transferase (GST) and the expression of associated microRNA such as miR-27, miR-34 and miR-148 in Mugilogobius abei were investigated. Result showed that the expressions of P-gp, CYP 1A, CYP 3A, GST and PXR were induced to some extend under simvastatin exposure for 72 h. A positive correlation was observed between PXR and CYP1A, CYP3A and P-gp. While for microRNA, a negative relationship was found between miR-34a and CYP3A, CYP1A. The expression of miR-148a was significantly induced under the exposure of SV (50 µg L-1), which was positive related to the transcriptional expression of PXR. For enzyme activity, erythromycin N-demethylase (ERND) significantly increased at 24 h and the activity of catalase (CAT) and superoxide dismutase (SOD) exhibited different trends. CAT was slightly inhibited at 24 h exposure but SOD was significantly induced in high concentration. Glutathione-S-transferase (GST) activity was significant inhibited after 72 h exposure. The reductive small molecule glutathione (GSH) content showed obvious decrease, while the quantity of malondialdehyde (MDA) increased significantly in high concentrations of SV exposure. GSH and MDA showed a typical negative correlation to some degree. Moreover, simvastatin caused histological changes in the liver tissues of M. abei, especially the size of adipocyte significantly decreased. The present study indicated that environmentally relevant concentration SV may affect the PXR signaling pathway in M. abei and pose potential ecological risks to non-target organisms like fish.


Subject(s)
Anticholesteremic Agents/toxicity , Fish Proteins/genetics , Gene Expression Regulation/drug effects , Liver/drug effects , Perciformes/genetics , Simvastatin/toxicity , Water Pollutants, Chemical/toxicity , Animals , Fish Proteins/metabolism , Liver/pathology , Perciformes/metabolism , Pregnane X Receptor/genetics , Pregnane X Receptor/metabolism , Signal Transduction/drug effects
11.
BMC Complement Altern Med ; 18(1): 258, 2018 Sep 24.
Article in English | MEDLINE | ID: mdl-30249222

ABSTRACT

BACKGROUND: For decades, various cardiovascular symptoms have been relieved by the use of Ya-Hom Navakot, which is a formulation comprising 54 herbal medicines. The Thailand Ministry of Public Health listed Ya-Hom Navakot's nine active principle and nomenclative herbal ingredients and termed them 'Phikud Navakot' (PN). Several reports have confirmed that PN has cardiovascular benefits similar to Ya-Hom Navakot. However, whether PN facilitates lipid-lowering activity remains unclear. METHODS: The present study investigated an in vitro model for examining the gene expression levels of 3-hydroxyl-3-methylglutaryl-CoA reductase (HMGCR) and low-density lipoprotein receptor (LDL-R) in HepG2 cells using qRT-PCR. The ethanol and water extractions of Ya-Hom Navakot, PN and Ya-Hom Navakot without PN were compared. RESULTS: One mg/ml of both NYEF and NYWF were found to significantly lower cholesterol by either the up-regulation of LDL-R or down-regulation of HMGCR compared with negative controls and 1 mg/ml simvastatin (p < 0.05). PNEF also up-regulated LDL-R gene expression, even more than NYEF (p < 0.05). In addition, the ethanol and water extracts of PN significantly down-regulated HMGCR gene expression compared with those of Ya-Hom Navakot without PN (p < 0.05). CONCLUSION: The use of Ya-Hom Navakot or PN may provide an alternative treatment to lower cholesterol through HMGCR gene inhibition and LDL-R gene enhancement.


Subject(s)
Anticholesteremic Agents/pharmacology , Gene Expression/drug effects , Hydroxymethylglutaryl CoA Reductases/metabolism , Plant Extracts/pharmacology , Receptors, LDL/metabolism , Anticholesteremic Agents/toxicity , Cholesterol/blood , Hep G2 Cells , Humans , Hydroxymethylglutaryl CoA Reductases/analysis , Hydroxymethylglutaryl CoA Reductases/genetics , Plant Extracts/toxicity , Receptors, LDL/analysis , Receptors, LDL/genetics , Simvastatin/pharmacology
12.
J Ethnopharmacol ; 224: 290-296, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-29772355

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Morus nigra L. is a plant native to Asia, and well adapted to the Brazilian climate. It is popularly known as "amoreira preta", and is part of the National List of Plants of Interest to the Brazilian Unified Health System. It is used in folk medicine mainly to soften the effects of menopause, as anti-inflammatory, antidiabetic and antihypertensive. However, information on safe doses and use is still precarious. AIM OF THE STUDY: To identify the chemical composition of the ethanolic extract of Morus nigra L. leaves (EEMN), as well as perform a toxicological study in male and female rats. MATERIALS AND METHODS: The chemical composition of the extract was performed by HPLC/DAD. In the acute study, the dose administered was 2000 mg/kg, and signs of toxicity and mortality was observed. In the sub-acute study, the extract was administered at doses of 500, 750 and 1000 mg/kg for 28 days. Behavioral changes, object recognition test, renal and hepatic tissue assessments, biochemical and hematological parameters were determined. The extract was administered orally to male and female rats in both studies. RESULTS: Quercetin and caffeic acid showed as major compounds in the extract. In the acute treatment, the extract was classified as safe (category 5), according to the protocol. In the subacute study, there was a decrease in AST in males (750 and 1000 mg/kg) and females (1000 mg/kg), reduction of total cholesterol in females (750 and 1000 mg/kg), and increase in renal and hepatic change the LPO levels. CONCLUSION: The present investigation showed that EEMN did not present significant toxic effects when administered orally. Moreover, presented a potentially protective action of organs and possesses hypocholesterolemic activity, thus, it is shown as a promising natural source to be used in pharmacology.


Subject(s)
Anticholesteremic Agents/toxicity , Morus , Plant Extracts/toxicity , Administration, Oral , Animals , Anticholesteremic Agents/analysis , Catalase/metabolism , Cholesterol/blood , Female , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Pattern Recognition, Visual/drug effects , Phytochemicals/analysis , Phytochemicals/toxicity , Plant Extracts/analysis , Plant Leaves/chemistry , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism , Toxicity Tests, Acute , Toxicity Tests, Subacute
13.
Atherosclerosis ; 257: 186-194, 2017 02.
Article in English | MEDLINE | ID: mdl-28152406

ABSTRACT

BACKGROUND AND AIMS: High-density lipoprotein cholesterol (HDL-C) is inversely related to cardiovascular risk. HDL-C raising ester transfer protein (CETP) inhibitors, are novel therapeutics. We studied the effects of CETP inhibitors anacetrapib and evacetrapib on triglycerides, cholesterol and lipoproteins, cholesterol efflux, paraoxonase activity (PON-1), reactive oxygen species (ROS), and endothelial function in E3L and E3L.CETP mice. METHODS: Triglycerides and cholesterol were measured at weeks 5, 14 and 21 in E3L.CETP mice on high cholesterol diet and treated with anacetrapib (3 mg/kg/day), evacetrapib (3 mg/kg/day) or placebo. Cholesterol efflux was assessed ex-vivo in mice treated with CETP inhibitors for 3 weeks on a normal chow diet. Endothelial function was analyzed at week 21 in isolated aortic rings, and serum lipoproteins assessed by fast-performance liquid chromatography. RESULTS: Anacetrapib and evacetrapib increased HDL-C levels (5- and 3.4-fold, resp.) and reduced triglycerides (-39% vs. placebo, p = 0.0174). Total cholesterol levels were reduced only in anacetrapib-treated mice (-32%, p = 0.0386). Cholesterol efflux and PON-1 activity (+45% and +35% vs. control, p < 0.005, resp.) were increased, while aortic ROS production was reduced with evacetrapib (-49% vs. control, p = 0.020). Anacetrapib, but not evacetrapib, impaired endothelium dependent vasorelaxation (p < 0.05). In contrast, no such effects were observed in E3L mice for all parameters tested. CONCLUSIONS: Notwithstanding a marked rise in HDL-C, evacetrapib did not improve endothelial function, while anacetrapib impaired it, suggesting that CETP inhibition does not provide vascular protection. Anacetrapib exerts unfavorable endothelial effects beyond CETP inhibition, which may explain the neutral results of large clinical trials in spite of increased HDL-C.


Subject(s)
Anticholesteremic Agents/pharmacology , Benzodiazepines/pharmacology , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol, HDL/blood , Dyslipidemias/drug therapy , Endothelium, Vascular/drug effects , Oxazolidinones/pharmacology , Vasodilation/drug effects , Animals , Anticholesteremic Agents/toxicity , Apolipoprotein E3/genetics , Aryldialkylphosphatase/blood , Benzodiazepines/toxicity , Biomarkers/blood , Cholesterol Ester Transfer Proteins/genetics , Cholesterol Ester Transfer Proteins/metabolism , Diet, High-Fat , Disease Models, Animal , Dyslipidemias/blood , Dyslipidemias/genetics , Dyslipidemias/physiopathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Female , Genetic Predisposition to Disease , Humans , Mice, Transgenic , Oxazolidinones/toxicity , Phenotype , Reactive Oxygen Species/metabolism , Triglycerides/blood , Up-Regulation
14.
Lipids Health Dis ; 16(1): 19, 2017 Jan 23.
Article in English | MEDLINE | ID: mdl-28115017

ABSTRACT

BACKGROUND: Efficacy and safety of proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitors, alirocumab (ALI) and evolocumab (EVO) have previously been evaluated through controlled clinical trials with selective patient groups. Post-commercially, in patients with heterozygous familial hypercholesterolemia (HeFH) and/or cardiovascular disease (CVD) with suboptimal LDL cholesterol (LDLC) lowering on maximal tolerated cholesterol lowering therapy, we assessed efficacy and safety of ALI and EVO. METHODS: Post-commercially, we started 25 patients on ALI 75 mg, 15 on ALI 150 mg, and 32 on EVO 140 mg bi-weekly added to entry LDLC lowering regimen, with follow-up for a median 24 weeks. History, physical exam, demographics, and adverse event data were collected. Changes in LDLC and AHA and NIH calculated 10-year CVD risks were assessed on ALI and EVO. RESULTS: Of 72 patients, 25 had HeFH only, 25 CVD only, 22 had both, median age was 65 years, 63% females, 38% males, 86% Caucasian, 11% African-Americans, 17% diabetics, 63% on anti-hypertensives, and 7% smokers. At entry, 30 (42%) were on a statin and 42 (58%) could not tolerate any statins. At 24-weeks, median LDLC decreased on ALI 75 mg from 117 to 62 mg/dL (-54%), on ALI 150 mg from 175 to 57 mg/dL (-63%), and on EVO 140 mg from 165 to 69 mg/dL (-63%), p <0.0001 for all. Absolute and percent LDLC reduction did not differ (p >.05) between ALI 150 and EVO 140 mg, but were less on ALI 75 mg vs ALI 150 mg and EVO 140 mg (p <.05). Percent reductions in 10-year CVD risks by AHA and NIH calculators, respectively were ALI 75 mg -22 and -44%, ALI 150 mg -31 and -50%, and EVO 140 mg -29 and -56%, p ≤.002 for all. The three most common adverse events included flu-like myositis 10%, respiratory tract symptoms 8%, and injection site reaction 6%. CONCLUSION: In patients with HeFH and/or CVD, LDLC was lowered by 63% on EVO and ALI 150 mg, and 54% on ALI 75 mg. Adverse events were minimal and tolerable. ALI and EVO represent paradigm shifts in LDLC lowering. Long term, post-commercial safety and efficacy remain to be determined.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Anticholesteremic Agents/administration & dosage , Cardiovascular Diseases/prevention & control , Hypercholesterolemia/drug therapy , Aged , Antibodies, Monoclonal/toxicity , Antibodies, Monoclonal, Humanized , Anticholesteremic Agents/toxicity , Cardiovascular Diseases/blood , Cholesterol, LDL/blood , Female , Humans , Hypercholesterolemia/blood , Male , Maximum Tolerated Dose , Middle Aged , Risk Factors , Treatment Outcome
15.
Cell Physiol Biochem ; 38(5): 1815-30, 2016.
Article in English | MEDLINE | ID: mdl-27160211

ABSTRACT

BACKGROUND/AIMS: To develop a suitable hepatocyte-like cell model that could be a substitute for primary hepatocytes with essential transporter expression and functions. Induced hepatocyte-like (iHep) cells directly reprogrammed from mice fibroblast cells were fully characterized. METHODS: Naïve iHep cells were transfected with nuclear hepatocyte factor 4 alpha (Hnf4α) and treated with selected small molecules. Sandwich cultured configuration was applied. The mRNA and protein expression of transporters were determined by Real Time PCR and confocal. The functional transporters were estimated by drug biliary excretion measurement. The inhibition of bile acid efflux transporters by cholestatic drugs were assessed. RESULTS: The expression and function of p-glycoprotein (P-gp), bile salt efflux pump (Bsep), multidrug resistance-associated protein 2 (Mrp2), Na+-dependent taurocholate cotransporting polypeptide (Ntcp), and organic anion transporter polypedtides (Oatps) in iHep cells were significantly improved after transfection of hepatocyte nuclear factor 4 alpha (Hnf4α) and treatment with selected inducers. In vitro intrinsic biliary clearances (CLb,int) of optimized iHep cells for rosuvastatin, methotrexate, d8-TCA (deuterium-labeled sodium taurocholate acid) and DPDPE ([D-Pen2,5] enkephalin hydrate) correlated well with that of sandwich-cultured primary mouse hepatocytes (SCMHs) (r2 = 0.984). Cholestatic drugs were evaluated and the results were compared well with primary mice hepatocytes. CONCLUSION: The optimized iHep cells expressed functional drug transporters and were comparable to primary mice hepatocytes. This study suggested direct reprogramming could provide a potential alternative to primary hepatocytes for drug candidate hepatobiliary disposition and hepatotoxicity screening.


Subject(s)
Anticholesteremic Agents/metabolism , Cellular Reprogramming , Hepatocyte Nuclear Factor 4/metabolism , Membrane Transport Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Anticholesteremic Agents/analysis , Anticholesteremic Agents/toxicity , Bile Acids and Salts/metabolism , Cell Culture Techniques , Cell Survival/drug effects , Cells, Cultured , Enkephalin, D-Penicillamine (2,5)-/analysis , Enkephalin, D-Penicillamine (2,5)-/metabolism , Enkephalin, D-Penicillamine (2,5)-/toxicity , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression/drug effects , Hepatocyte Nuclear Factor 4/genetics , Hepatocytes/cytology , Hepatocytes/metabolism , Membrane Transport Proteins/genetics , Methotrexate/analysis , Methotrexate/metabolism , Methotrexate/toxicity , Mice , Mice, Inbred ICR , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Rosuvastatin Calcium/analysis , Rosuvastatin Calcium/metabolism , Rosuvastatin Calcium/toxicity , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Symporters/genetics , Symporters/metabolism
16.
Bioorg Med Chem Lett ; 26(3): 849-853, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26783178

ABSTRACT

Cholesterol absorption inhibitor (CAI) targeting Niemann-Pick C1-like1 protein was developed for the treatment of hyperlipidaemia and only ezetimibe was approved so far. For developing novel CAIs, we synthesized sixteen 2-azetidinone derivatives and thirteen 1H-pyrrole-2,5-dione derivatives containing sulfonamide group at the side chain, and their inhibitory activity of cholesterol absorption was evaluated in Caco-2 cell line in vitro. Furthermore, top six compounds were measured by cytotoxicity and partition coefficient, and 2-azetidinone analogue 9e was selected for in vivo study. Finally, 9e considerably reduced total cholesterol, LDL-C, FFA and triglyceride in the serum and increased the rate of HDL-C to total cholesterol, suggesting it could regulate the lipid metabolism and act as a potent CAI.


Subject(s)
Azetidines/chemistry , Azetidines/pharmacology , Cholesterol/metabolism , Pyrroles/chemistry , Pyrroles/pharmacology , Sulfanilamides/chemistry , Animals , Anticholesteremic Agents/chemistry , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/toxicity , Apoptosis/drug effects , Azetidines/toxicity , Body Weight/drug effects , Caco-2 Cells , Cholesterol, LDL/blood , Cricetinae , Drug Evaluation, Preclinical , Fatty Acids/blood , HEK293 Cells , Humans , Nitric Oxide/metabolism , Pyrroles/chemical synthesis , Pyrroles/toxicity , Sulfanilamide , Triglycerides/blood
17.
Toxicol Appl Pharmacol ; 289(1): 117-23, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26386191

ABSTRACT

HMG-CoA reductase inhibitors (e.g., statins) are an important clinical option to lower cholesterol and treat co-morbidities. Atorvastatin is the most prescribed statin and has obtained generic status. We recently had a clinical development program evaluating a combination of atorvastatin with a GPR119 agonist as a treatment for dyslipidemia, where toxicological evaluations in dogs were completed. There were several challenges related to selecting doses for atorvastatin, including understanding the dose-exposure relationship from different drug forms used by the innovator in their general toxicology studies, bioanalytical assays that did not separate and quantify parent from metabolites, and high variability in the systemic exposures following oral dosing. The studies in this report characterized the toxicokinetics and toxicity of atorvastatin in the dog for up to 13-weeks. Overall, there were no notable differences in the toxicokinetics of atorvastatin or the two active hydroxylated metabolites between the sexes at Week 13. However, systemic exposures were markedly lower at Week 13 compared to that observed at Week 4, suggesting induction of metabolism or reduced absorption from the gastrointestinal tract following oral dosing. Changes in laboratory chemistries included increased liver enzyme levels and lower cholesterol levels. Histopathologic evaluation revealed multifocal minimal to slight hemorrhages in the submucosa of the gallbladder; all findings were reversible. The information from these studies along with the existing clinical experience with atorvastatin can be used to design robust toxicology studies in dogs and reduce animal use.


Subject(s)
Atorvastatin/pharmacokinetics , Atorvastatin/toxicity , Animals , Anticholesteremic Agents/pharmacokinetics , Anticholesteremic Agents/toxicity , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Male , Toxicity Tests
18.
Drug Des Devel Ther ; 9: 3695-704, 2015.
Article in English | MEDLINE | ID: mdl-26229434

ABSTRACT

The hERG gene encodes the pore-forming α-subunit of the rapidly activating delayed rectifier potassium channel (I Kr), which is important for cardiac repolarization. Reduction of I hERG due to genetic mutations or drug interferences causes long QT syndrome, leading to life-threatening cardiac arrhythmias (torsades de pointes) or sudden death. Probucol is a cholesterol-lowering drug that could reduce hERG current by decreasing plasma membrane hERG protein expression and eventually cause long QT syndrome. Here, we investigated the mechanisms of probucol effects on I hERG and hERG-channel expression. Our data demonstrated that probucol reduces SGK1 expression, known as SGK isoform, in a concentration-dependent manner, resulting in downregulation of phosphorylated E3 ubiquitin ligase Nedd4-2 expression, but not the total level of Nedd4-2. As a result, the hERG protein reduces, due to the enhanced ubiquitination level. On the contrary, carbachol could enhance the phosphorylation level of Nedd4-2 as an alternative to SGK1, and thus rescue the ubiquitin-mediated degradation of hERG channels caused by probucol. These discoveries provide a novel mechanism of probucol-induced hERG-channel deficiency, and imply that carbachol or its analog may serve as potential therapeutic compounds for the handling of probucol cardiotoxicity.


Subject(s)
Anticholesteremic Agents/toxicity , Ether-A-Go-Go Potassium Channels/genetics , Long QT Syndrome/chemically induced , Probucol/toxicity , Anticholesteremic Agents/administration & dosage , Carbachol/pharmacology , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Endosomal Sorting Complexes Required for Transport/metabolism , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Nedd4 Ubiquitin Protein Ligases , Phosphorylation/drug effects , Probucol/administration & dosage , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/metabolism
19.
PLoS One ; 9(9): e108890, 2014.
Article in English | MEDLINE | ID: mdl-25268751

ABSTRACT

Statins are increasingly being recognized as anti-cancer agents against various cancers including breast cancer. To understand the molecular pathways targeted by fluvastatin and its differential sensitivity against metastatic breast cancer cells, we analyzed protein alterations in MDA-MB-231 cells treated with fluvastatin using 2-DE in combination with LC-MS/MS. Results revealed dys-regulation of 39 protein spots corresponding to 35 different proteins. To determine the relevance of altered protein profiles with breast cancer cell death, we mapped these proteins to major pathways involved in the regulation of cell-to-cell signaling and interaction, cell cycle, Rho GDI and proteasomal pathways using IPA analysis. Highly interconnected sub networks showed that vimentin and ERK1/2 proteins play a central role in controlling the expression of altered proteins. Fluvastatin treatment caused proteolysis of vimentin, a marker of epithelial to mesenchymal transition. This effect of fluvastatin was reversed in the presence of mevalonate, a downstream product of HMG-CoA and caspase-3 inhibitor. Interestingly, fluvastatin neither caused an appreciable cell death nor did modulate vimentin expression in normal mammary epithelial cells. In conclusion, fluvastatin alters levels of cytoskeletal proteins, primarily targeting vimentin through increased caspase-3- mediated proteolysis, thereby suggesting a role for vimentin in statin-induced breast cancer cell death.


Subject(s)
Anticholesteremic Agents/toxicity , Apoptosis/drug effects , Electrophoresis, Gel, Two-Dimensional , Fatty Acids, Monounsaturated/toxicity , Indoles/toxicity , Proteome/analysis , Acyl Coenzyme A/metabolism , Breast Neoplasms , Caspase 3/chemistry , Caspase 3/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Epithelial-Mesenchymal Transition/drug effects , Female , Fluvastatin , Humans , Metabolic Networks and Pathways , Mevalonic Acid/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Tandem Mass Spectrometry , Vimentin/metabolism
20.
Lipids Health Dis ; 13: 153, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25273894

ABSTRACT

BACKGROUND: Many drugs are substrates for P-glycoprotein (P-gp) and interactions involving P-gp may be relevant to clinical practice. Co-administration with P-gp inhibitors or inducers changes the absorption profile as well as the risk for drug toxicity, therefore it is important to evaluate possible P-gp alterations. The purpose of this study was to investigate the effect of two novel cholesterol-lowering agents, disodium ascorbyl phytostanol phosphate (DAPP) and nanostructured aluminium silicate (NSAS), a protonated montmorillonite clay, on mdr-1 gene expression and its protein, P-glycoprotein (P-gp) within Caco-2 cells. METHODS: The effects of DAPP and NSAS on the regulation of mdr-1 gene, P-gp protein expression and activity within Caco-2 cells, were determined using cell viability and cytotoxicity tests, RT-PCR, Western Blot analysis and bi-directional transport studies. RESULTS: We observed a significant down-regulation of mdr-1 mRNA (e.g. 38.5 ± 17% decrease vs. control at 5 µM DAPP and 61.2 ± 25% versus control at 10 µM DAPP; n = 6, P* < 0.05) within Caco-2 cells. Western Blot analysis of P-gp expression showed that changes in mdr-1 gene expression lead to correlating changes in P-gp protein expression. This down-regulation of P-glycoprotein also resulted in decreased activity of P-glycoprotein compared to untreated control. In contrast, when Caco-2 cells were treated with NSAS, no changes in mdr-1 gene expression, P-gp protein expression nor P-gp activity were observed. CONCLUSIONS: DAPP but not NSAS decreases P-gp mediated drug efflux through decreased mdr-1 gene expression and consequently decreased P-gp protein expression. These findings have to be taken into consideration when DAPP is concurrently given with other drugs that are substrates for P-gp since drug-drug interactions harbour a safety issue and alter bioavailability profiles.NSAS does not have any P-gp altering properties and therefore might not affect drug-drug interactions. We conclude from this study that NSAS might make a safer drug candidate compared to DAPP for lowering LDL-cholesterol.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Aluminum Silicates/pharmacology , Anticholesteremic Agents/pharmacology , Gene Expression/drug effects , Phytosterols/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Aluminum Silicates/toxicity , Anticholesteremic Agents/toxicity , Biological Transport , Caco-2 Cells , Cell Survival/drug effects , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Fluorescent Dyes/metabolism , Humans , Nanostructures/toxicity , Phytosterols/toxicity , Rhodamine 123/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...