Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 196
Filter
1.
Article in English | MEDLINE | ID: mdl-38522713

ABSTRACT

Statins, widely prescribed for cholesterol management by inhibiting HMG-CoA reductase in the cholesterol biosynthesis pathway, may also influence vertebrate development. In this study, we investigated the developmental effects of two widely used statins, atorvastatin (ATO) and pravastatin (PRA), on zebrafish offspring. For ATO, we administered doses classified as low (1 µM), medium (5 µM), and high (10 µM), while for PRA, the corresponding concentrations were set at low (18 µM), medium (180 µM), and high (270 µM). Our results showed significant reductions in birth and hatching rates, along with decreased body length in offspring at all ATO concentrations and medium to high PRA concentrations. A notable increase in malformation rates, especially in the spine and heart, was observed across all ATO treatments and in medium and high PRA groups. Additionally, we observed reduced heart contraction rates, decreased heart size, lower bone volumes, and diminished expression of mRNA osteogenic markers. Elevated venous sinus-artery bulb (SV-BA) ratios, increased thoracic area, and abnormal cartilage development were also prominent in all ATO-treated groups. Transcriptome analysis revealed alterations in genes predominantly associated with ion channels. These findings provide insights into the potential impacts of specific concentrations of statins on offspring development and highlight potential gene interactions with statins.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Animals , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Zebrafish/genetics , Transcriptome , Pravastatin/pharmacology , Pravastatin/therapeutic use , Atorvastatin/toxicity , Ion Channels
2.
Toxicol Mech Methods ; 34(2): 130-147, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37771097

ABSTRACT

An increased risk of new-onset diabetes mellitus has been recently reported for statin therapy, and experimental studies have shown reduced glucose-stimulated insulin secretion (GSIS) and mitochondrial dysfunction in beta cells with effects differing among agents. Organic anion transporting polypeptide (OATP) 2B1 contributes to hepatic uptake of rosuvastatin, atorvastatin and pravastatin, three known substrates. Since OATP2B1 is present in beta cells of the human pancreas, we investigated if OATP2B1 facilitates the local accumulation of statins in a rat beta cell model INS-1 832/13 (INS-1) thereby amplifying statin-induced toxicity. OATP2B1 overexpression in INS-1 cells via adenoviral transduction showed 2.5-, 1.8- and 1.4-fold higher cellular retention of rosuvastatin, atorvastatin and pravastatin, respectively, relative to LacZ control, while absolute intracellular concentration was about twice as high for the lipophilic atorvastatin compared to the more hydrophilic rosuvastatin and pravastatin. After 24 h statin treatment at high concentrations, OATP2B1 enhanced statin toxicity involving activation of intrinsic apoptosis (caspase 3/7 activation) and mitochondrial dysfunction (NADH dehydrogenase activity) following rosuvastatin and atorvastatin, which was partly reversed by isoprenoids. OATP2B1 had no effect on statin-induced reduction in GSIS, mitochondrial electron transport chain complex expression or caspase 9 activation. We confirmed a dose-dependent reduction in insulin secretion by rosuvastatin and atorvastatin in native INS-1 with a modest change in cellular ATP. Collectively, our results indicate a role of OATP2B1, which is abundant in human beta cells, in statin accumulation and statin-induced toxicity but not insulin secretion of rosuvastatin and atorvastatin in INS-1 cells.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Mitochondrial Diseases , Humans , Rats , Animals , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Atorvastatin/toxicity , Rosuvastatin Calcium/toxicity , Pravastatin , Mitochondrial Diseases/chemically induced
3.
Environ Res ; 234: 116572, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37429398

ABSTRACT

Statins are the most widely used pharmacological agents for reducing blood cholesterol levels and treating atherosclerotic cardiovascular diseases. Most of the statins' derivatives have been limited by water solubility, bioavailability, and oral absorption, which has led to adverse effects on several organs, especially at high doses. As an approach to reducing statin intolerance, achieving a stable formulation with improved efficacy and bioavailability at low doses has been suggested. Nanotechnology-based formulations may provide a therapeutic benefit over traditional formulations in terms of potency and biosafety. Nanocarriers can provide tailored delivery platforms for statins, thereby enhancing the localized biological effects and lowering the risk of undesired side effects while boosting statin's therapeutic index. Furthermore, tailored nanoparticles can deliver the active cargo to the desired site, which culminates in reducing off-targeting and toxicity. Nanomedicine could also provide opportunities for therapeutic methods by personalized medicine. This review delves into the existing data on the potential improvement of statin therapy using nano-formulations.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Nanomedicine , Nanotechnology
5.
Muscle Nerve ; 67(6): 537-547, 2023 06.
Article in English | MEDLINE | ID: mdl-36975763

ABSTRACT

INTRODUCTION/AIMS: The mechanisms that underlie the pathogenesis of statin-associated muscle symptoms (SAMS) remain unclear. Pregnancy is associated with increased cholesterol levels. Statins may be useful during pregnancy, but their safety is uncertain. Hence, we investigated the postpartum effects of exposure to rosuvastatin and simvastatin during pregnancy in Wistar rats, targeting the neuromuscular structures. METHODS: Twenty-one pregnant Wistar rats were divided into three groups: control (C) treated with vehicle (dimethylsulfoxide + dH20), simvastatin (S) 62.5 mg/kg/day, and rosuvastatin (R) 10 mg/kg/day. Gavage was performed daily from the gestational days 8 to 20. At weaning, the postpartum mother tissues were collected and subjected to morphological and morphometric analysis of the soleus muscle, associated neuromuscular junctions (NMJs), and the sciatic nerve; protein quantification; quantification of the cholesterol and creatine kinase in the serum; and intramuscular collagen analysis. RESULTS: An increase in morphometric parameters (area, maximum and minimum diameters, Feret diameter, and minimum Feret) was observed in NMJs from the S and R groups in comparison with the C group, and there was also a loss of common NMJ circularity. The number of myofibers with central nuclei was higher in S (17 ± 3.9, P = .0083) and R (18.86 ± 14.42, P = .0498) than in C (6.8 ± 2.6). DISCUSSION: Gestational exposure to statins induced postpartum NMJ morphology alterations in soleus muscle, which may be caused by the remodeling of clusters of nicotinic acetylcholine receptors. This may be associated with the development and progression of SAMS observed in clinical practice.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Rats , Pregnancy , Humans , Female , Animals , Rats, Wistar , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Rosuvastatin Calcium , Neuromuscular Junction/metabolism , Muscle, Skeletal/metabolism , Simvastatin/adverse effects , Postpartum Period
6.
Chemosphere ; 313: 137574, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36528155

ABSTRACT

Cardiovascular pharmaceuticals (CVPs) are globally present in inland waters and have also been found in the sediment and plasma of fish from the Sava River, Croatia. Based on the previous research, CVPs amiodarone (AMI), ramipril (RAM), simvastatin (SIM), and verapamil (VER) have been selected for this study. Their effect has been investigated, individually and in a mixture, on the development of the zebrafish embryo Danio rerio (Hamilton, 1822) within the first 96 h of development. Upon exposure to environmentally relevant concentrations of tested CVPs (0.1, 1, and 10 µg/L) zebrafish survival and development as apparent from observed morphological abnormalities, heartbeat rates and changes in behavior, hatching success, larval length and oxidative stress level were monitored. The CVP causing the highest mortality and pathological changes was SIM (1 and 10 µg/L), which corresponds well with the observed effects during zebrafish exposure to CVPs' mixtures (4 and 40 µg/L). All pharmaceuticals affected cardiac function and decreased heart rate. SIM (1 µg/L), VER and RAM (10 µg/L) decreased larval length, while induced oxidative stress was recorded in the SIM- and VER-exposed specimens. Behavioral alterations of zebrafish were observed only in AMI-treated group (10 µg/L). Our amino acid sequence comparison and structural and docking analysis showed a highly conserved binding site between human and zebrafish HMG-CoA reductase for SIM and its main metabolite simvastatin acid. Using these ecotoxicological bioassays on a zebrafish model with particular emphasis on sublethal endpoints, the risk of CVPs, especially statins, for fish in inland waters has been identified.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Water Pollutants, Chemical , Humans , Animals , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Zebrafish/metabolism , Rivers , Larva , Pharmaceutical Preparations , Water Pollutants, Chemical/metabolism , Embryo, Nonmammalian
7.
Ecotoxicol Environ Saf ; 243: 113981, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36029576

ABSTRACT

Statins are prescribed widely as lipid-lowering agents. However, statins are associated with an increased harmful risk on public health and the ecosystem. Little is known about statins' toxicity on biological development and the underlying molecular mechanisms. We exposed zebrafish embryos to a series of statins to evaluate their development toxicity. Statins-induced embryonic developmental defects in a concentration-dependent manner. 72 h LC50 values for lovastatin, simvastatin, fluvastatin, atorvastatin, rosuvastatin, and pravastatin were 0.01 µM, 0.04 µM, 1.93 µM, 37.28 µM, 79.29 µM, and 2170 µM, respectively. Moreover, the expression of genes involved in heart contraction, calcium ion binding, transcription factors, nucleus, and G protein-coupled receptor signaling pathway was altered by statins. The early growth response gene (egr4) and transcription factor genes (fosab and fosb) were screened as potential toxicity targets due to their significant upregulation based on protein-protein interaction (PPI) and drug-gene interaction network analysis. Finally, the ecotoxicity profile of statins was predicted by in silico method, and statins were high or moderate risk to aquatic organisms. We provide a systems toxicology strategy to explore the toxicity of statins and illustrate the potential mechanisms of action.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Animals , Ecosystem , Fatty Acids, Monounsaturated , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Indoles , Simvastatin , Transcriptome , Zebrafish/genetics
8.
Drug Chem Toxicol ; 45(5): 2233-2245, 2022 Sep.
Article in English | MEDLINE | ID: mdl-33934680

ABSTRACT

Statins are 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase inhibitor drugs that lead to serum-cholesterol-lowering effects. Rosuvastatin, a third-generation statin, has shown better results in reducing cholesterol concentrations when compared to other widely prescribed statins. Recent studies by our group reported that rosuvastatin impairs reproductive function in rats possibly by disrupting the reproductive-endocrine axis. In this study, we evaluated whether rosuvastatin presents estrogenic or antiestrogenic effects, by an in vivo uterotrophic assay in rats, and investigated the direct effect of this drug upon rat uterine tissue contractility both in non-gravid and gravid periods. Rosuvastatin exposure in vivo at doses of 0 (control), 3, and 10 mg/kg/d was not associated with estrogenic or antiestrogenic effects on uterine tissue. However, in vivo (doses of 0, 3, and 10 mg/kg/d) and ex vivo (concentrations of 0, 1, 10, and 100 µg/mL) exposures to this drug were related to alterations in uterine basal contraction pattern. Furthermore, in vivo and ex vivo rosuvastatin exposures potentially modulate the action of uterine contraction inducers carbachol, norepinephrine, and prostaglandin E2. Thus, rosuvastatin can affect uterine physiology not necessarily by an endocrine mechanism related to the estrogen signaling, but possibly by its pleiotropic effects, with indirect tissue and cellular interactions, since in vivo and ex vivo exposures of uterine fragments to rosuvastatin presented different responses in uterine contractile parameters, which require further studies upon the precise mechanism of action of this drug in female reproductive function.


Subject(s)
Estrogens , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Animals , Cholesterol , Estrogens/toxicity , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Pregnancy , Rats , Rats, Wistar , Rosuvastatin Calcium/toxicity
9.
Pak J Pharm Sci ; 34(5(Supplementary)): 1939-1944, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34836863

ABSTRACT

Type 2 Diabetes Mellitus (T2DM) patients are at high risk of Coronary Heart Disease (CHD) and need a global therapeutic intervention. A fixed-dose combination prescription medication containing anti-diabetic drug (Sitagliptin) and lipid lowering (Simvastatin) has recently been approved. Present study was designed to explore the potential synergistic toxic effects of sitagliptin and simvastatin at cellular level. MTT assay revealed the potential synergistic cytotoxic effect whereas Comet assay spotlighted the genotoxicity. MTT assay conducted on Vero cell lines revealed no significant change in proliferative activity upon treatment with simvastatin but cell survival percentage (CSP) decreased upon treatment with sitagliptin (51% at 1000µg/mL). However, combination of both drugs exhibited a better survival percentage except highest dose combination (1000:500µg/mL) which augmented antiproliferative effects rendering CSP 71.6%. The genotoxic assay spotted that Simvastatin produced less damage to DNA with the threshold of 500µg/ml whereas Sitagliptin significantly damage above the 250µg/mL, However, combination of drugs produced lesser damage than Sitagliptin alone. The findings concluded a non-genotoxic combination of sitagliptin and simvastatin which possess a least cytotoxic potential suggesting the safe use of the combination both in T2DM and CHD.


Subject(s)
Cell Survival/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Hypoglycemic Agents/toxicity , Mutagens/toxicity , Simvastatin/toxicity , Sitagliptin Phosphate/toxicity , Animals , Cell Proliferation/drug effects , Chlorocebus aethiops , Comet Assay , DNA Damage , Diabetes Mellitus, Type 2/drug therapy , Drug Combinations , Drug Interactions , Drug Synergism , Vero Cells
10.
J Cardiovasc Pharmacol ; 78(4): 544-550, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34651601

ABSTRACT

INTRODUCTION: Our study aimed to investigate the effect of atorvastatin on plaque calcification by matching the results obtained by 18F-sodium fluoride (18F-NaF) positron emission tomography (PET)/computed tomography (CT) with data from histologic sections. METHODS AND RESULTS: The rabbits were divided into 2 groups as follows: an atherosclerosis group (n = 10) and an atorvastatin group (n = 10). All rabbits underwent an abdominal aortic operation and were fed a high-fat diet to induce atherosclerosis. Plasma samples were used to analyze serum inflammation markers and blood lipid levels. 18F-NaF PET/CT scans were performed twice. The plaque area, macrophage number and calcification were measured, and the data from the pathological sections were matched with the 18F-NaF PET/CT scan results. The mean standardized uptake value (0.725 ± 0.126 vs. 0.603 ± 0.071, P < 0.001) and maximum standardized uptake value (1.024 ± 0.116 vs. 0.854 ± 0.091, P < 0.001) significantly increased in the atherosclerosis group, but only slightly increased in the atorvastatin group (0.616 ± 0.103 vs. 0.613 ± 0.094, P = 0.384; 0.853 ± 0.099 vs.0.837 ± 0.089, P < 0.001, respectively). The total calcium density was significantly increased in rabbits treated with atorvastatin compared with rabbits not treated with atorvastatin (1.64 ± 0.90 vs. 0.49 ± 0.35, P < 0.001), but the microcalcification level was significantly lower. There were more microcalcification deposits in the areas with increased radioactive uptake of 18F-NaF. CONCLUSIONS: Our study suggests that the anti-inflammatory activity of atorvastatin may promote macrocalcification but not microcalcification within atherosclerotic plaques. 18F-NaF PET/CT can detect plaque microcalcifications.


Subject(s)
Aorta, Abdominal/drug effects , Atherosclerosis/drug therapy , Atorvastatin/toxicity , Fluorine Radioisotopes , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Sodium Fluoride , Vascular Calcification/chemically induced , Animals , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Atherosclerosis/diagnostic imaging , Atherosclerosis/metabolism , Atherosclerosis/pathology , Calcium/metabolism , Disease Models, Animal , Male , Plaque, Atherosclerotic , Rabbits , Vascular Calcification/diagnostic imaging , Vascular Calcification/metabolism , Vascular Calcification/pathology
11.
Biochem Pharmacol ; 192: 114750, 2021 10.
Article in English | MEDLINE | ID: mdl-34461118

ABSTRACT

Statins decrease the serum LDL-cholesterol concentration and reduce the risk for cardiovascular diseases but can cause myopathy, which may be related to mTORC inhibition. In the current study, we investigated which mTORC is inhibited by simvastatin and by which mechanisms. In C2C12 myoblasts and myotubes and mouse gastrocnemius, simvastatin was cytotoxic and inhibited S6rp and Akt Ser473 phosphorylation, indicating inhibition of mTORC1 and mTORC2, respectively. In contrast to simvastatin, the mTORC1 inhibitor rapamycin did not inhibit mTORC2 activity and was not cytotoxic. Like simvastatin, knock-down of Rictor, an essential component of mTORC2, impaired Akt Ser473 and S6rp phosphorylation and was cytotoxic for C2C12 myoblasts, suggesting that mTORC2 inhibition is an important myotoxic mechanism. The investigation of the mechanism of mTORC2 inhibition showed that simvastatin impaired Ras farnesylation, which was prevented by farnesol but without restoring mTORC2 activity. In comparison, Rap1 knock-down reduced mTORC2 activity and was cytotoxic for C2C12 myoblasts. Simvastatin impaired Rap1 geranylgeranylation and function, which was prevented by geranylgeraniol. In addition, simvastatin and the complex III inhibitor antimycin A caused mitochondrial superoxide accumulation and impaired the activity of mTORC2, which could partially be prevented by the antioxidant MitoTEMPO. In conclusion, mTORC2 inhibition is an important mechanism of simvastatin-induced myotoxicity. Simvastatin inhibits mTORC2 by impairing geranylgeranylation of Rap1 and by inducing mitochondrial dysfunction.


Subject(s)
Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mitochondria/drug effects , Muscle, Skeletal/drug effects , Prenylation/drug effects , Simvastatin/toxicity , rap1 GTP-Binding Proteins/antagonists & inhibitors , Animals , Cell Line , Drug Delivery Systems/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Male , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Prenylation/physiology , Simvastatin/administration & dosage , rap1 GTP-Binding Proteins/metabolism
12.
Pharmacol Res ; 167: 105528, 2021 05.
Article in English | MEDLINE | ID: mdl-33667685

ABSTRACT

Geranylgeranyl diphosphate synthase (GGDPS), an enzyme in the isoprenoid biosynthetic pathway (IBP), produces the isoprenoid (geranylgeranyl pyrophosphate, GGPP) used in protein geranylgeranylation reactions. Our prior studies utilizing triazole bisphosphonate-based GGDPS inhibitors (GGSIs) have revealed that these agents represent a novel strategy by which to induce cancer cell death, including multiple myeloma and pancreatic cancer. Statins inhibit the rate-limiting enzyme in the IBP and potentiate the effects of GGSIs in vitro. The in vivo effects of combination therapy with statins and GGSIs have not been determined. Here we evaluated the effects of combining VSW1198, a novel GGSI, with a statin (lovastatin or pravastatin) in CD-1 mice. Twice-weekly dosing with VSW1198 at the previously established maximally tolerated dose in combination with a statin led to hepatotoxicity, while once-weekly VSW1198-based combinations were feasible. No abnormalities in kidney, spleen, brain or skeletal muscle were observed with combination therapy. Combination therapy disrupted protein geranylgeranylation in vivo. Evaluation of hepatic isoprenoid levels revealed decreased GGPP levels in the single drug groups and undetectable GGPP levels in the combination groups. Additional studies with combinations using 50% dose-reductions of either VSW1198 or lovastatin revealed minimal hepatotoxicity with expected on-target effects of diminished GGPP levels and disruption of protein geranylgeranylation. Combination statin/GGSI therapy significantly slowed tumor growth in a myeloma xenograft model. Collectively, these studies are the first to demonstrate that combination IBP inhibitor therapy alters isoprenoid levels and disrupts protein geranylgeranylation in vivo as well as slows tumor growth in a myeloma xenograft model, thus providing the framework for future clinical exploration.


Subject(s)
Biosynthetic Pathways/drug effects , Diterpenes/administration & dosage , Drug Delivery Systems/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Protein Prenylation/drug effects , Terpenes/metabolism , Triazoles/administration & dosage , Animals , Biosynthetic Pathways/physiology , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Diterpenes/toxicity , Drug Evaluation, Preclinical/methods , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Lovastatin/administration & dosage , Lovastatin/toxicity , Mice , Mice, Inbred NOD , Mice, SCID , Pravastatin/administration & dosage , Pravastatin/toxicity , Protein Prenylation/physiology , Terpenes/antagonists & inhibitors , Triazoles/toxicity , Xenograft Model Antitumor Assays/methods
13.
Toxicol Lett ; 342: 85-94, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33600922

ABSTRACT

Triptolide (TP), an active component of Tripterygium wilfordii Hook. F, has been widely used in China for treating autoimmune and inflammatory diseases, and has also been validated by modern science and developed as a candidate anti-cancer treatment. However, liver toxicity of TP has seriously hindered its use and development, the clinical features and primary toxicological mechanism have been unclear. Considering the major target regulation mechanism of TP is the suppression of global transcription regulated by RNAPII, which is closed related with the detoxification of drugs. This paper tries to verify the synergistic liver injury and its mechanism of TP when co-administered with CYP3A4 substrate drug. The experiments showed that TP dose-dependently blocked transcriptional activation of CYP3A4 in both hPXR and hPXR-CYP3A4 reporter cell lines, lowered the mRNA and protein expression of PXR target genes such as CYP3A1, CYP2B1, and MDR1, and inhibited the functional activity of CYP3A in a time- and concentration-dependent manner in sandwich-cultured rat hepatocytes (SCRH) and female Sprague-Dawley (f-SD) rats. Furthermore, TP combined with atorvastatin (ATR), the substrate of CYP3A4, synergistically enhanced hepatotoxicity in cultured HepG2 and SCRH cells (CI is 0.38 and 0.29, respectively), as well as in f-SD rats, with higher exposure levels of both drugs. These results clearly indicate that TP inhibits PXR-mediated transcriptional activation of CYP3A4, leading to a blockade on the detoxification of itself and ATR, thereby greatly promoting liver injury. This study may implies the key cause of TP related liver injury and provides experimental data for the rational use of TP in a clinical scenario.


Subject(s)
Atorvastatin/toxicity , Cytochrome P-450 CYP3A/metabolism , Diterpenes/toxicity , Hepatocytes/drug effects , Phenanthrenes/toxicity , Pregnane X Receptor/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Atorvastatin/administration & dosage , Atorvastatin/pharmacokinetics , Cytochrome P-450 CYP3A/genetics , Diterpenes/administration & dosage , Diterpenes/pharmacokinetics , Drug Synergism , Epoxy Compounds/administration & dosage , Epoxy Compounds/pharmacokinetics , Epoxy Compounds/toxicity , Female , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Phenanthrenes/administration & dosage , Phenanthrenes/pharmacokinetics , Rats , Rats, Sprague-Dawley
14.
Somatosens Mot Res ; 38(1): 90-94, 2021 03.
Article in English | MEDLINE | ID: mdl-33263458

ABSTRACT

INTRODUCTION: Drug-induced myopathy is among the most common causes of muscle disease. Lipid-lowering drugs, primarily the statins as inhibitors of the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, are a common cause of myopathy. Statin-fibrate combination potentially increases risk for myopathy and rhabdomyolysis. Blood levels of the enzymes creatine kinase (CK), aldolase and lactate dehydrogenase (LDH) increase during myopathy. Exercise may be a trigger for statin-associated muscle symptoms (SAMS). METHODS: In this study a model of myopathy induction was designed via combination of oral atorvastatin, gemfibrozil and exercise for ten days in rats. To maximise exercise, the rats were placed in a pool of water and allowed to swim before sinking in the last three days. Finally, the mean of swimming tolerance times and blood levels of creatine kinase, aldolase and lactate dehydrogenase were measured. RESULTS: The results showed a significantly (p < 0.05) decreased swimming tolerance time and elevated enzyme levels in rats receiving atorvastatin (ATV) and gemfibrozil (GMF) plus exercise compared with those rats in other groups. This animal model can be used to evaluate the effects of medication on reduction of statin/fibrate-induced myopathy.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Muscular Diseases , Animals , Atorvastatin/toxicity , Disease Models, Animal , Fibric Acids , Gemfibrozil/toxicity , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Muscular Diseases/chemically induced , Rats
15.
PLoS One ; 15(11): e0242422, 2020.
Article in English | MEDLINE | ID: mdl-33237943

ABSTRACT

Statins are used to lower cholesterol and prevent cardiovascular disease. Musculoskeletal side effects known as statin associated musculoskeletal symptoms (SAMS), are reported in up to 10% of statin users, necessitating statin therapy interruption and increasing cardiovascular disease risk. We tested the hypothesis that, when exposed to statins ex vivo, engineered human skeletal myobundles derived from individuals with (n = 10) or without (n = 14) SAMS and elevated creatine-kinase levels exhibit statin-dependent muscle defects. Myoblasts were derived from muscle biopsies of individuals (median age range of 62-64) with hyperlipidemia with (n = 10) or without (n = 14) SAMS. Myobundles formed from myoblasts were cultured with growth media for 4 days, low amino acid differentiation media for 4 days, then dosed with 0 and 5µM of statins for 5 days. Tetanus forces were subsequently measured. To model the change of tetanus forces among clinical covariates, a mixed effect model with fixed effects being donor type, statin concentration, statin type and their two way interactions (donor type*statin concentration and donor type* statin type) and the random effect being subject ID was applied. The results indicate that statin exposure significantly contributed to decrease in force (P<0.001) and the variability in data (R2C [R square conditional] = 0.62). We found no significant differences in force between myobundles from patients with/without SAMS, many of whom had chronic diseases. Immunofluorescence quantification revealed a positive correlation between the number of straited muscle fibers and tetanus force (R2 = 0.81,P = 0.015) and negative correlation between number of fragmented muscle fibers and tetanus force (R2 = 0.482,P = 0.051) with no differences between donors with or without SAMS. There is also a correlation between statin exposure and presence of striated fibers (R2 = 0.833, P = 0.047). In patient-derived myobundles, statin exposure results in myotoxicity disrupting SAA organization and reducing force. We were unable to identify differences in ex vivo statin myotoxicity in this system. The results suggest that it is unlikely that there is inherent susceptibility to or persistent effects of statin myopathy using patient-derived myobundles.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Muscle, Skeletal/drug effects , Muscular Diseases/chemically induced , Aged , Amino Acids/pharmacology , Cells, Cultured , Culture Media/pharmacology , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hyperlipidemias/drug therapy , Hyperlipidemias/pathology , In Vitro Techniques , Male , Middle Aged , Muscle Contraction/drug effects , Muscle Fatigue/physiology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiopathology , Muscular Diseases/physiopathology , Myoblasts/drug effects , Quadriceps Muscle/cytology , Single-Blind Method , Tissue Engineering
16.
Bratisl Lek Listy ; 121(8): 589-599, 2020.
Article in English | MEDLINE | ID: mdl-32726123

ABSTRACT

AIM: The aim of the present study was to investigate the effect of apoptosis on rat skeletal muscle caused by chronic alcohol and statin consumption with modified liquid diet and to elucidate protective effects of betaine supplementation. METHODS: TNF-α (tumor necrosis factor), NF-kB (Nuclear Factor kappa B), cytochrome c and caspase-3 levels with or without betaine treatment in alcohol and/or statin-induced skeleton muscle apoptosis rats as well as in controls were measured in serum and tissue. Histologic examinations of the muscle tissues were also performed. RESULTS: In our study, betaine treated treatment groups we found that calpain and caspase activities and cytokine c release were decreased caused by alcohol, statin and more importantly alcohol+statin group and TNF and NF-kB levels were also close to the levels of control group. Similarly, significant improvements have been observed in our morphological and histological examination results also supporting our biochemical data. CONCLUSION: We found that combined consumption of ethanol and statin is capable of triggering apoptotic cell death in rat muscles more than the consumption of only alcohol or only statin. Betaine was able to reduced this muscle cell death induced by alcohol and/or statin consumption (Tab. 4, Fig. 4, Ref. 43).


Subject(s)
Apoptosis , Betaine , Ethanol , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Animals , Apoptosis/drug effects , Betaine/pharmacology , Ethanol/toxicity , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Muscle, Skeletal/drug effects , NF-kappa B , Rats , Tumor Necrosis Factor-alpha
17.
Toxicol Appl Pharmacol ; 401: 115076, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32479918

ABSTRACT

Statin induced myopathy (SIM) is a main deleterious effect leading to the poor treatment compliance, while the preventive or therapeutic treatments are absent. Mounting evidences demonstrated that vitamin D plays a vital role in muscle as a direct modulator. The deficiency of vitamin D was considered as a cause of muscle dysfunction, whereas the supplementation resulted in a remission. However, there is no causal proof that vitamin D supplementation rescues SIM. Here, using the mice model of simvastatin-induced myopathy, we investigated the role of vitamin D supplementation and the mechanisms associated with mitochondria. Results indicated that simvastatin administration (80 mg/kg) impaired skeletal muscle with the increased serum creatine kinase (CK) level and the declined grip strength, which were alleviated by vitamin D supplementation. Moreover, vitamin D supplementation rescued the energy metabolism dysfunction in simvastatin-treated mice gastrocnemius by reducing the abnormal aggregation of muscular glycogen and lactic acid. Mitochondrial homeostasis plays a key role in the process of energy metabolism. Thus, the mitochondrial dysfunction is a mortal damage for the highly energy-requiring tissue. In our study, the mitochondrial cristae observed under transmission electron microscope (TEM) were lytic in simvastatin-treated gastrocnemius. Interestingly, vitamin D supplementation improved the mitochondrial cristae shape by regulating the expression of mitofusin-1/2 (MFN1/2), optic atrophy 1 (OPA1) and dynamin-related protein 1 (Drp1). As expected, the mitochondrial dysfunction and oxidative stress was mitigated by vitamin D supplementation. In conclusion, these findings suggested that moderate vitamin D supplementation rescued simvastatin induced myopathy via improving the mitochondrial cristae shape and function.


Subject(s)
Dietary Supplements , Mitochondria/drug effects , Muscular Diseases/chemically induced , Muscular Diseases/drug therapy , Simvastatin/toxicity , Vitamin D/administration & dosage , Animals , Energy Metabolism/drug effects , Energy Metabolism/physiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/pathology , Muscular Diseases/metabolism , Random Allocation
19.
Continuum (Minneap Minn) ; 25(6): 1712-1731, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31794468

ABSTRACT

PURPOSE OF REVIEW: This article reviews the pathogenesis, clinical features, and management of toxic myopathy related to common medications, critical illness, and illicit substances. RECENT FINDINGS: Muscle symptoms are common among statin users and are usually reversible after discontinuation of the statin; rarely, however, statins trigger an immune-mediated necrotizing myopathy that persists and requires immunomodulatory therapy. Autoantibodies targeting 3-hydroxy-3-methylglutaryl coenzyme A reductase can distinguish the toxic and immune-mediated forms. Immune checkpoint inhibitors, increasingly used in the treatment of advanced cancer, have recently been associated with the development of inflammatory myositis. A reversible mitochondrial myopathy has long been associated with zidovudine, but recent reports elucidate the risk of myopathy with newer antivirals, such as telbivudine and raltegravir. SUMMARY: The medications most commonly associated with myopathy include statins, amiodarone, chloroquine, hydroxychloroquine, colchicine, certain antivirals, and corticosteroids, and myopathy can occur with chronic alcoholism. Certain clinical, electrodiagnostic, and histologic features can aid in early recognition. Stopping the use of the offending agent reverses symptoms in most cases, but specific and timely treatment may be required in cases related to agents that trigger immune-mediated muscle injury.


Subject(s)
Adrenal Cortex Hormones/toxicity , Anti-Retroviral Agents/toxicity , Enzyme Inhibitors/toxicity , Fibric Acids/toxicity , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Immunologic Factors/toxicity , Myotoxicity , Tubulin Modulators/toxicity , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Myotoxicity/etiology , Myotoxicity/genetics , Myotoxicity/physiopathology
20.
Curr Pharm Biotechnol ; 20(15): 1244-1259, 2019.
Article in English | MEDLINE | ID: mdl-31333127

ABSTRACT

Statins are drugs used for people with abnormal lipid levels (hyperlipidemia) and are among the best-selling medications in the United States. Thus, the aspects related to the production of these drugs are of extreme importance for the pharmaceutical industry. Herein, we provide a non-exhaustive review of fungal species used to produce statin and highlighted the major factors affecting the efficacy of this process. The current biotechnological approaches and the advances of a metabolic engineer to improve statins production are also emphasized. The biotechnological production of the main statins (lovastatin, pravastatin and simvastatin) uses different species of filamentous fungi, for example Aspergillus terreus. The statins production is influenced by different types of nutrients available in the medium such as the carbon and nitrogen sources, and several researches have focused their efforts to find the optimal cultivation conditions. Enzymes belonging to Lov class, play essential roles in statin production and have been targeted to genetic manipulations in order to improve the efficiency for Lovastatin and Simvastatin production. For instance, Escherichia coli strains expressing the LovD have been successfully used for lovastatin production. Other examples include the use of iRNA targeting LovF of A. terreus. Therefore, fungi are important allies in the fight against hyperlipidemias. Although many studies have been conducted, investigations on bioprocess optimization (using both native or genetic- modified strains) still necessary.


Subject(s)
Biotechnology/methods , Fungi/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/biosynthesis , Metabolic Engineering/methods , Pravastatin/biosynthesis , Animals , Fermentation , Fungi/genetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Hyperlipidemias/drug therapy , Lovastatin/pharmacology , Lovastatin/toxicity , Pravastatin/pharmacology , Pravastatin/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...