1
|
Assis LHDP, Dorighello GDG, Rentz T, de Souza JC, Vercesi AE, de Oliveira HCF. In Vivo Pravastatin Treatment Reverses Hypercholesterolemia Induced Mitochondria-Associated Membranes Contact Sites, Foam Cell Formation, and Phagocytosis in Macrophages. Front Mol Biosci 2022; 9:839428. [PMID: 35372506 PMCID: PMC8965079 DOI: 10.3389/fmolb.2022.839428] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Statins are successful drugs used to treat hypercholesterolemia, a primary cause of atherosclerosis. In this work, we investigated how hypercholesterolemia and pravastatin treatment impact macrophage and mitochondria functions, the key cell involved in atherogenesis. By comparing bone marrow-derived macrophages (BMDM) of wild-type (WT) and LDL receptor knockout (LDLr−/−) mice, we observed hypercholesterolemia increased the number of contact sites at mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs), enhanced mitochondrial hydrogen peroxide release, altered the gene expression of inflammatory markers, and increased oxidized LDL (ox-LDL) uptake and phagocytic activity. Three months of in vivo pravastatin treatment of LDLr−/− mice reversed the number of contact sites at the MAM, ox-LDL uptake, and phagocytosis in LDLr−/− BMDM. Additionally, pravastatin increased BMDM mitochondrial network branching. In peritoneal macrophages (PMs), hypercholesterolemia did not change MAM stability, but stimulated hydrogen peroxide production and modulated gene expression of pro- and anti-inflammatory markers. It also increased mitochondrial branching degree and had no effects on ox-LDL uptake and phagocytosis in PM. Pravastatin treatment increased superoxide anion production and changed inflammation-related gene expression in LDLr−/− PM. In addition, pravastatin increased markedly the expression of the mitochondrial dynamics-related genes Mfn2 and Fis1 in both macrophages. In summary, our results show that hypercholesterolemia and pravastatin treatment affect macrophage mitochondria network structure as well as their interaction with the endoplasmic reticulum (ER). These effects impact on macrophage conversion rates to foam cell and macrophage phagocytic capacity. These findings associate MAM stability changes with known mechanisms involved in atherosclerosis progression and resolution.
Collapse
Affiliation(s)
| | | | - Thiago Rentz
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Jane Cristina de Souza
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Aníbal Eugênio Vercesi
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Helena Coutinho Franco de Oliveira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
- *Correspondence: Helena Coutinho Franco de Oliveira,
| |
Collapse
|
2
|
Imai S, Kashiwagi H, Sato Y, Miyai T, Sugawara M, Takekuma Y. Factors affecting creatine phosphokinase elevation during daptomycin therapy using a combination of machine learning and conventional methods. Br J Clin Pharmacol 2021; 88:1211-1222. [PMID: 34436795 DOI: 10.1111/bcp.15063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/10/2021] [Accepted: 08/16/2021] [Indexed: 01/04/2023] Open
Abstract
AIMS Musculoskeletal toxicity is a typical side effect of daptomycin (DAP). However, the risk factors have not been well established. Here, we aimed to identify independent factors affecting DAP-induced musculoskeletal toxicity using a combination of machine learning and conventional statistical methods. METHODS A population-based, retrospective, observational cohort study was conducted using the Japanese electronic medical record database. Patients who received DAP between October 2011 and December 2020 were enrolled. Two definitions of musculoskeletal toxicity were employed: (1) elevation of creatine phosphokinase (CPK) value more than twice from baseline and >200 IU/L, and (2) >1000 IU/L. First, multiple logistic regression analyses (a conventional statistical method) were performed to identify independent factors affecting CPK elevation. Then, decision tree analyses, a machine learning method, were performed to detect combinations of factors that change CPK elevation risk. RESULTS Of the 2970 patients who received DAP, 706 were included. Elevation of CPK values >200 IU/L and >1000 IU/L occurred in 83 (11.8%) and 17 (2.41%) patients, respectively. In multiple logistic regression analysis, baseline CPK value and concomitant use of hydrophobic statins were commonly extracted as independent factors affecting each CPK elevation, but concomitant use of hydrophilic statins was not. In decision tree analysis, patients who received hydrophobic statins and had high baseline CPK values were classified into the high-risk group. CONCLUSION Our novel approach revealed new risk factors for CPK elevation. Our findings suggest that high-risk patients require frequent CPK monitoring.
Collapse
Affiliation(s)
- Shungo Imai
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hitoshi Kashiwagi
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yuki Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Takayuki Miyai
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Mitsuru Sugawara
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.,Department of Pharmacy, Hokkaido University Hospital, Sapporo, Japan.,Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
| | - Yoh Takekuma
- Department of Pharmacy, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
3
|
Zi L, Zhou W, Xu J, Li J, Li N, Xu J, You C, Wang C, Tian M. Rosuvastatin Nanomicelles Target Neuroinflammation and Improve Neurological Deficit in a Mouse Model of Intracerebral Hemorrhage. Int J Nanomedicine 2021; 16:2933-2947. [PMID: 33907400 PMCID: PMC8068519 DOI: 10.2147/ijn.s294916] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/01/2021] [Indexed: 02/05/2023] Open
Abstract
Background Intracerebral hemorrhage (ICH), a devastating subtype of stroke, has a poor prognosis. However, there is no effective therapy currently available due to its complex pathological progression, in which neuroinflammation plays a pivotal role in secondary brain injury. In this work, the use of statin-loaded nanomicelles to target the neuroinflammation and improve the efficacy was studied in a mouse model of ICH. Methods Rosuvastatin-loaded nanomicelles were prepared by a co-solvent evaporation method using polyethylene glycol-poly(ε-caprolactone) (PEG-PCL) copolymer as a carrier. The prepared nanomicelles were characterized by transmission electron microscopy (TEM) and dynamic light scattering (DLS), and then in vitro and in vivo studies were performed. Results TEM shows that the nanomicelles are spherical with a diameter of about 19.41 nm, and DLS shows that the size, zeta potential, and polymer dispersity index of the nanomicelles were 23.37 nm, −19.2 mV, and 0.221, respectively. The drug loading content is 8.28%. The in vivo study showed that the nanomicelles significantly reduced neuron degeneration, inhibited the inflammatory cell infiltration, reduced the brain edema, and improved neurological deficit. Furthermore, it was observed that the nanomicelles promoted the polarization of microglia/macrophages to M2 phenotype, and also the expression of the proinflammatory cytokines, such as IL-1β and TNF-α, was significantly down-regulated, while the expression of the anti-inflammatory cytokine IL-10 was significantly up-regulated. The related mechanism was proposed and discussed. Conclusion The nanomicelles treatment suppressed the neuroinflammation that might contribute to the promoted nerve functional recovery of the ICH mouse, making it potential to be applied in clinic.
Collapse
Affiliation(s)
- Liu Zi
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Wencheng Zhou
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jiake Xu
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junshu Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Ning Li
- Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jianguo Xu
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Chao You
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Chengwei Wang
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Meng Tian
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
4
|
Zhang Q, Dong J, Yu Z. Pleiotropic use of Statins as non-lipid-lowering drugs. Int J Biol Sci 2020; 16:2704-2711. [PMID: 33110390 PMCID: PMC7586431 DOI: 10.7150/ijbs.42965] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 08/01/2020] [Indexed: 12/14/2022] Open
Abstract
Statins, known as HMG-CoA reductase (HMGCR) inhibitors, have primarily been utilized for metabolic and angiographic medical applications because of their cholesterol-lowering effects. Similar to other drugs, statins may also induce a series of potential side effects. Statins inhibit the HMGCR (rate-limiting enzyme) activity in early stages of mevalonate pathway and then indirectly affect a number of intermediate products, including non-sterol isoprenoids (coenzyme Q10, dolichol etc.), which can result in impaired functions of body organs. Recently, scores of studies have uncovered additional functional mechanisms of statins in other diseases, such as diabetes mellitus, nervous system diseases, coronary heart disease, inflammation and cancers. This review aims to summarize the positive and adverse mechanisms of statin therapy. Statin care should be taken in the treatment of many diseases including cancers. Since the underlying mechanisms are not fully elucidated, future studies should spend more time and efforts on basic research to explore the mechanisms of statins.
Collapse
Affiliation(s)
- Qijia Zhang
- Digestive internal medicine and Department of infectious diseases, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China
| | - Jianlong Dong
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Ze Yu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, China
| |
Collapse
|
5
|
Durmuş M, Bahçecioğlu ÖF, Gök S. Daptomycin in combination with rosuvastatin induced blood creatine phosphokinase elevation. Eur J Hosp Pharm 2020; 28:234-236. [PMID: 34162676 DOI: 10.1136/ejhpharm-2020-002218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 11/04/2022] Open
Abstract
We present the case of a 73-year-old male patient who was hospitalised with infective endocarditis, and report an elevation in his blood creatine phosphokinase (CPK) after receiving daptomycin and rosuvastatin therapy concomitantly. His previous home-scheduled medications included apixaban, ivabradine, metformin, rosuvastatin 20 mg, ginkgo biloba and trimetazidine, and he continued to receive these medications at the hospital. After three sets of blood cultures were taken, empirical treatment was started with vancomycin and gentamicin. On the eighth day of treatment, daptomycin and ampicillin-sulbactam were initiated due to ampicillin-resistant Enterococcus faecalis growth in the patient's blood culture. Daptomycin and rosuvastatin were discontinued on the 23rd day of treatment because of blood CPK elevation (2416 U/L) and linezolid was started instead of daptomycin. Six days after discontinuation of daptomycin and rosuvastatin, the CPK concentrations returned to normal range.
Collapse
Affiliation(s)
| | | | - Selim Gök
- Clinical pharmacy, Inonu University, Malatya, Turkey
| |
Collapse
|
6
|
Iwaki Y, Lee W, Sugiyama Y. Comparative and quantitative assessment on statin efficacy and safety: insights into inter-statin and inter-individual variability via dose- and exposure-response relationships. Expert Opin Drug Metab Toxicol 2019; 15:897-911. [PMID: 31648563 DOI: 10.1080/17425255.2019.1681399] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Introduction: Statins are prescribed widely for cholesterol-lowering therapy, but it is known that their efficacy and safety profiles vary, despite the shared pharmacophore and pharmacological target. The immense body of related clinical and preclinical data offers a unique opportunity to explore the possible factors underlying inter-statin and inter-individual variabilities.Area covered: Clinical and preclinical data from various statins were compiled with regard to the efficacy (cholesterol-lowering effect) and safety (muscle toxicity). Based on the compiled data, dose- and exposure-response relationships were explored to obtain mechanistic and quantitative insights into the variations in the efficacy and safety profiles of statins.Expert opinion: Our analyses indicated that the inter-statin variability in the cholesterol-lowering effect may be mainly attributable to variations in potency of inhibition of the pharmacological target, rather than variations in drug exposure at the site of drug action. However, the drug exposure at the sites of drug action (i.e., the liver for efficacy and the muscle for safety) may contribute to the differences in the efficacy and safety observed in individual patients.
Collapse
Affiliation(s)
- Yuki Iwaki
- Clinical Pharmacology, Janssen Pharmaceutical K.K., Tokyo, Japan
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama, Kanagawa, Japan
| |
Collapse
|
7
|
Lorza-Gil E, García-Arevalo M, Favero BC, Gomes-Marcondes MCC, Oliveira HCF. Diabetogenic effect of pravastatin is associated with insulin resistance and myotoxicity in hypercholesterolemic mice. J Transl Med 2019; 17:285. [PMID: 31455371 PMCID: PMC6712816 DOI: 10.1186/s12967-019-2045-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/20/2019] [Indexed: 12/18/2022] Open
Abstract
Background HMG-CoA reductase inhibitors (statins) are cholesterol-lowering drugs widely used to treat hypercholesterolemia and prevent cardiovascular disease. Statins are generally well tolerated, but adverse reactions may occur, particularly myopathy and new onset of diabetes. The exact mechanism of statin-induced myopathy and diabetes has not been fully elucidated. We have previously shown that treatment of hypercholesterolemic (LDLr−/−) mice with pravastatin for 2 months decreased pancreatic islet insulin secretion and increased oxidative stress and cell death, but no glucose intolerance was observed. The purpose of the current work was to study long-term pravastatin effects on glucose homeostasis, insulin sensitivity, muscle protein turnover and cell viability. Methods LDLr−/− mice were treated with pravastatin for 3, 6 and 10 months. Glucose tolerance, insulin resistance and glucose-stimulated insulin secretion were evaluated. The rates of protein synthesis and degradation were determined in gastrocnemius muscle after 10 months of treatment. Insulin signalling, oxidative stress and cell death were analysed in vitro using C2C12 myotubes. Results After 6 and 10 months of treatment, these mice became glucose intolerant, and after 10 months, they exhibited marked insulin resistance. Reduced islet glucose-stimulated insulin secretion was observed after the 3rd month of treatment. Mice treated for 10 months showed significantly decreased body weight and increased muscle protein degradation. In addition, muscle chymotrypsin-like proteasomal activity and lysosomal cathepsin were markedly elevated. C2C12 myotubes exposed to increasing concentrations of pravastatin presented dose-dependent impairment of insulin-induced Akt phosphorylation, increased apoptotic markers (Bax protein and cleaved caspase-3) and augmented superoxide anion production. Conclusions In addition to reduced insulin secretion, long-term pravastatin treatment induces insulin resistance and muscle wasting. These results suggest that the diabetogenic effect of statins is linked to the appearance of myotoxicity induced by oxidative stress, impaired insulin signalling, proteolysis and apoptosis.
Collapse
Affiliation(s)
- Estela Lorza-Gil
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Cidade Universitária Zeferino Vaz, Rua Monteiro Lobato, 255, Campinas, SP, CEP 13083-862, Brazil
| | - Marta García-Arevalo
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Cidade Universitária Zeferino Vaz, Rua Monteiro Lobato, 255, Campinas, SP, CEP 13083-862, Brazil
| | - Bianca Cristine Favero
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Cidade Universitária Zeferino Vaz, Rua Monteiro Lobato, 255, Campinas, SP, CEP 13083-862, Brazil
| | - Maria Cristina C Gomes-Marcondes
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Cidade Universitária Zeferino Vaz, Rua Monteiro Lobato, 255, Campinas, SP, CEP 13083-862, Brazil
| | - Helena C F Oliveira
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Cidade Universitária Zeferino Vaz, Rua Monteiro Lobato, 255, Campinas, SP, CEP 13083-862, Brazil.
| |
Collapse
|
8
|
Bouitbir J, Sanvee GM, Panajatovic MV, Singh F, Krähenbühl S. Mechanisms of statin-associated skeletal muscle-associated symptoms. Pharmacol Res 2019; 154:104201. [PMID: 30877064 DOI: 10.1016/j.phrs.2019.03.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/24/2019] [Accepted: 03/10/2019] [Indexed: 12/25/2022]
Abstract
Statins lower the serum low-density lipoprotein cholesterol and prevent cardiovascular events by inhibiting 3-hydroxy-3-methyl-glutaryl-CoA reductase. Although the safety of statins is documented, many patients ingesting statins may suffer from skeletal muscle-associated symptoms (SAMS). Importantly, SAMS are a common reason for stopping the treatment with statins. Statin-associated muscular symptoms include fatigue, weakness and pain, possibly accompanied by elevated serum creatine kinase activity. The most severe muscular adverse reaction is the potentially fatal rhabdomyolysis. The frequency of SAMS is variable but in up to 30% of the patients ingesting statins, depending on the population treated and the statin used. The mechanisms leading to SAMS are currently not completely clarified. Over the last 15 years, several research articles focused on statin-induced mitochondrial dysfunction as a reason for SAMS. Statins can impair the function of the mitochondrial respiratory chain, thereby reducing ATP and increasing ROS production. This can induce mitochondrial membrane permeability transition, release of cytochrome c into the cytosol and induce apoptosis. In parallel, statins inhibit activation of Akt, mainly due to reduced function of mTORC2, which may be related to mitochondrial dysfunction. Mitochondrial dysfunction by statins is also responsible for activation of AMPK, which is associated with impaired activation of mTORC1. Reduced activation of mTORC1 leads to increased skeletal muscle protein degradation, impaired protein synthesis and stimulation of apoptosis. In this paper, we discuss some of the different hypotheses how statins affect skeletal muscle in more detail, focusing particularly on those related to mitochondrial dysfunction and the impairment of the Akt/mTOR pathway.
Collapse
Affiliation(s)
- Jamal Bouitbir
- Division of Clinical Pharmacology & Toxicology, University Hospital, 4031, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland
| | - Gerda M Sanvee
- Division of Clinical Pharmacology & Toxicology, University Hospital, 4031, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland
| | - Miljenko V Panajatovic
- Division of Clinical Pharmacology & Toxicology, University Hospital, 4031, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland
| | - François Singh
- Division of Clinical Pharmacology & Toxicology, University Hospital, 4031, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology & Toxicology, University Hospital, 4031, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland.
| |
Collapse
|
9
|
Statins for the Primary Prevention of Coronary Heart Disease. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4870350. [PMID: 30834266 PMCID: PMC6374814 DOI: 10.1155/2019/4870350] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 11/06/2018] [Accepted: 12/03/2018] [Indexed: 11/17/2022]
Abstract
Object The purpose of this study was to fully assess the role of statins in the primary prevention of coronary heart disease (CHD). Methods We searched six databases (PubMed, the Cochrane Library, Web of Science, China National Knowledge Infrastructure, Wanfang Database, and Chinese Scientific Journal Database) to identify relevant randomized controlled trials (RCTs) from inception to 31 October 2017. Two review authors independently assessed the methodological quality and analysed the data using Rev Man 5.3 software. Risk ratios and 95% confidence intervals (95% CI) were pooled using fixed/random-effects models. Funnel plots and Begg's test were conducted to assess publication bias. The quality of the evidence was evaluated using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. Results Sixteen RCTs with 69159 participants were included in this review. Statins can effectively decrease the occurrence of angina (RR=0.70, 95% CI: 0.58~0.85, I2 =0%), nonfatal myocardial infarction (MI) (RR=0.60, 95% CI: 0.51~0.69, I2 =14%), fatal MI (RR=0.49, 95% CI: 0.24~0.98, I2 =0%), any MI (RR=0.53, 95% CI: 0.42~0.67, I2 =0%), any coronary heart events (RR=0.73, 95% CI: 0.68~0.78, I2=0%), coronary revascularization (RR=0.66, 95% CI: 0.55~0.78, I2 = 0%), and any cardiovascular events (RR=0.77, 95% CI: 0.72~82, I2 = 0%). However, based on the current evidence, there were no significant differences in CHD deaths (RR=0.82, 95% CI: 0.66~1.02, I2=0%) and all-cause mortality (RR=0.88, 95% CI: 0.76 ~1.01, I2 =58%) between the two groups. Additionally, statins were more likely to result in diabetes (RR=1.21, 95% CI: 1.05~1.39, I2 =0%). There was no evidence of publication biases, and the quality of the evidence was considered moderate. Conclusion Statins seemed to be beneficial for the primary prevention of CHDs but have no effect on CHD death and all-cause mortality.
Collapse
|
10
|
Godoy JC, Niesman IR, Busija AR, Kassan A, Schilling JM, Schwarz A, Alvarez EA, Dalton ND, Drummond JC, Roth DM, Kararigas G, Patel HH, Zemljic-Harpf AE. Atorvastatin, but not pravastatin, inhibits cardiac Akt/mTOR signaling and disturbs mitochondrial ultrastructure in cardiac myocytes. FASEB J 2018; 33:1209-1225. [PMID: 30169110 DOI: 10.1096/fj.201800876r] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Statins, which reduce LDL-cholesterol by inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, are among the most widely prescribed drugs. Skeletal myopathy is a known statin-induced adverse effect associated with mitochondrial changes. We hypothesized that similar effects would occur in cardiac myocytes in a lipophilicity-dependent manner between 2 common statins: atorvastatin (lipophilic) and pravastatin (hydrophilic). Neonatal cardiac ventricular myocytes were treated with atorvastatin and pravastatin for 48 h. Both statins induced endoplasmic reticular (ER) stress, but only atorvastatin inhibited ERK1/2T202/Y204, AktSer473, and mammalian target of rapamycin signaling; reduced protein abundance of caveolin-1, dystrophin, epidermal growth factor receptor, and insulin receptor-β; decreased Ras homolog gene family member A activation; and induced apoptosis. In cardiomyocyte-equivalent HL-1 cells, atorvastatin, but not pravastatin, reduced mitochondrial oxygen consumption. When male mice underwent atorvastatin and pravastatin administration per os for up to 7 mo, only long-term atorvastatin, but not pravastatin, induced elevated serum creatine kinase; swollen, misaligned, size-variable, and disconnected cardiac mitochondria; alteration of ER structure; repression of mitochondria- and endoplasmic reticulum-related genes; and a 21% increase in mortality in cardiac-specific vinculin-knockout mice during the first 2 months of administration. To our knowledge, we are the first to demonstrate in vivo that long-term atorvastatin administration alters cardiac ultrastructure, a finding with important clinical implications.-Godoy, J. C., Niesman, I. R., Busija, A. R., Kassan, A., Schilling, J. M., Schwarz, A., Alvarez, E. A., Dalton, N. D., Drummond, J. C., Roth, D. M., Kararigas, G., Patel, H. H., Zemljic-Harpf, A. E. Atorvastatin, but not pravastatin, inhibits cardiac Akt/mTOR signaling and disturbs mitochondrial ultrastructure in cardiac myocytes.
Collapse
Affiliation(s)
- Joseph C Godoy
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Ingrid R Niesman
- Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Anna R Busija
- Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Adam Kassan
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, West Coast University, North Hollywood, California, USA
| | - Jan M Schilling
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Anna Schwarz
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Erika A Alvarez
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Nancy D Dalton
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - John C Drummond
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Georgios Kararigas
- Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Alice E Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| |
Collapse
|
11
|
Ingelman-Sundberg M, Mkrtchian S, Zhou Y, Lauschke VM. Integrating rare genetic variants into pharmacogenetic drug response predictions. Hum Genomics 2018; 12:26. [PMID: 29793534 PMCID: PMC5968569 DOI: 10.1186/s40246-018-0157-3] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/08/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Variability in genes implicated in drug pharmacokinetics or drug response can modulate treatment efficacy or predispose to adverse drug reactions. Besides common genetic polymorphisms, recent sequencing projects revealed a plethora of rare genetic variants in genes encoding proteins involved in drug metabolism, transport, and response. RESULTS To understand the global importance of rare pharmacogenetic gene variants, we mapped the variability in 208 pharmacogenes by analyzing exome sequencing data from 60,706 unrelated individuals and estimated the importance of rare and common genetic variants using a computational prediction framework optimized for pharmacogenetic assessments. Our analyses reveal that rare pharmacogenetic variants were strongly enriched in mutations predicted to cause functional alterations. For more than half of the pharmacogenes, rare variants account for the entire genetic variability. Each individual harbored on average a total of 40.6 putatively functional variants, rare variants accounting for 10.8% of these. Overall, the contribution of rare variants was found to be highly gene- and drug-specific. Using warfarin, simvastatin, voriconazole, olanzapine, and irinotecan as examples, we conclude that rare genetic variants likely account for a substantial part of the unexplained inter-individual differences in drug metabolism phenotypes. CONCLUSIONS Combined, our data reveal high gene and drug specificity in the contributions of rare variants. We provide a proof-of-concept on how this information can be utilized to pinpoint genes for which sequencing-based genotyping can add important information to predict drug response, which provides useful information for the design of clinical trials in drug development and the personalization of pharmacological treatment.
Collapse
Affiliation(s)
- Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Souren Mkrtchian
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
12
|
Leung YH, Turgeon J, Michaud V. Study of Statin- and Loratadine-Induced Muscle Pain Mechanisms Using Human Skeletal Muscle Cells. Pharmaceutics 2017; 9:pharmaceutics9040042. [PMID: 28994701 PMCID: PMC5750648 DOI: 10.3390/pharmaceutics9040042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/30/2017] [Accepted: 10/01/2017] [Indexed: 12/15/2022] Open
Abstract
Many drugs can cause unexpected muscle disorders, often necessitating the cessation of an effective medication. Inhibition of monocarboxylate transporters (MCTs) may potentially lead to perturbation of l-lactic acid homeostasis and muscular toxicity. Previous studies have shown that statins and loratadine have the potential to inhibit l-lactic acid efflux by MCTs (MCT1 and 4). The main objective of this study was to confirm the inhibitory potentials of atorvastatin, simvastatin (acid and lactone forms), rosuvastatin, and loratadine on l-lactic acid transport using primary human skeletal muscle cells (SkMC). Loratadine (IC50 31 and 15 µM) and atorvastatin (IC50 ~130 and 210 µM) demonstrated the greatest potency for inhibition of l-lactic acid efflux at pH 7.0 and 7.4, respectively (~2.5-fold l-lactic acid intracellular accumulation). Simvastatin acid exhibited weak inhibitory potency on l-lactic acid efflux with an intracellular lactic acid increase of 25–35%. No l-lactic acid efflux inhibition was observed for simvastatin lactone or rosuvastatin. Pretreatment studies showed no change in inhibitory potential and did not affect lactic acid transport for all tested drugs. In conclusion, we have demonstrated that loratadine and atorvastatin can inhibit the efflux transport of l-lactic acid in SkMC. Inhibition of l-lactic acid efflux may cause an accumulation of intracellular l-lactic acid leading to the reported drug-induced myotoxicity.
Collapse
Affiliation(s)
- Yat Hei Leung
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H2X 0A9, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada.
| | - Jacques Turgeon
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H2X 0A9, Canada.
| | - Veronique Michaud
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H2X 0A9, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada.
| |
Collapse
|
13
|
Association of miR-145 With Statin-Induced Skeletal Muscle Toxicity in Human Rhabdomyosarcoma RD Cells. J Pharm Sci 2017; 106:2873-2880. [DOI: 10.1016/j.xphs.2017.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/30/2017] [Accepted: 04/07/2017] [Indexed: 12/15/2022]
|
14
|
Affiliation(s)
- Sharon Rees
- Assistant professor Therapeutics and Prescribing, University of Nottingham
| |
Collapse
|
15
|
El-Ganainy SO, El-Mallah A, Abdallah D, Khattab MM, Mohy El-Din MM, El-Khatib AS. Rosuvastatin safety: An experimental study of myotoxic effects and mitochondrial alterations in rats. Toxicol Lett 2016; 265:23-29. [PMID: 27815113 DOI: 10.1016/j.toxlet.2016.10.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 10/28/2016] [Accepted: 10/31/2016] [Indexed: 01/14/2023]
Abstract
Myopathy is the most commonly reported adverse effect of statins. All statins are associated with myopathy, though with different rates. Rosuvastatin is a potent statin reported to induce myopathy comparable to earlier statins. However, in clinical practice most patients could tolerate rosuvastatin over other statins. This study aimed to evaluate the myopathic pattern of rosuvastatin in rats using biochemical, functional and histopathological examinations. The possible deleterious effects of rosuvastatin on muscle mitochondria were also examined. The obtained results were compared to myopathy induced by atorvastatin in equimolar dose. Results showed that rosuvastatin induced a rise in CK, a slight increase in myoglobin level together with mild muscle necrosis. Motor activity, assessed by rotarod, showed that rosuvastatin decreased rats' performance. All these manifestations were obviously mild compared to the prominent effects of atorvastatin. Parallel results were obtained in mitochondrial dysfunction parameters. Rosuvastatin only induced a slight increase in LDH and a minor decrease in ATP (∼14%) and pAkt (∼12%). On the other hand, atorvastatin induced an increase in LDH, lactate/pyruvate ratio and a pronounced decline in ATP (∼80%) and pAkt (∼65%). These findings showed that rosuvastatin was associated with mild myotoxic effects in rats, especially when compared to atorvastatin.
Collapse
Affiliation(s)
- Samar O El-Ganainy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Pharos University, Alexandria, Egypt.
| | - Ahmed El-Mallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Pharos University, Alexandria, Egypt
| | - Dina Abdallah
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mahmoud M Mohy El-Din
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Pharos University, Alexandria, Egypt
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
16
|
Taha DA, De Moor CH, Barrett DA, Lee JB, Gandhi RD, Hoo CW, Gershkovich P. The role of acid-base imbalance in statin-induced myotoxicity. Transl Res 2016; 174:140-160.e14. [PMID: 27083388 PMCID: PMC4967449 DOI: 10.1016/j.trsl.2016.03.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/15/2016] [Accepted: 03/21/2016] [Indexed: 12/02/2022]
Abstract
Disturbances in acid-base balance, such as acidosis and alkalosis, have potential to alter the pharmacologic and toxicologic outcomes of statin therapy. Statins are commonly prescribed for elderly patients who have multiple comorbidities such as diabetes mellitus, cardiovascular, and renal diseases. These patients are at risk of developing acid-base imbalance. In the present study, the effect of disturbances in acid-base balance on the interconversion of simvastatin and pravastatin between lactone and hydroxy acid forms have been investigated in physiological buffers, human plasma, and cell culture medium over pH ranging from 6.8-7.8. The effects of such interconversion on cellular uptake and myotoxicity of statins were assessed in vitro using C2C12 skeletal muscle cells under conditions relevant to acidosis, alkalosis, and physiological pH. Results indicate that the conversion of the lactone forms of simvastatin and pravastatin to the corresponding hydroxy acid is strongly pH dependent. At physiological and alkaline pH, substantial proportions of simvastatin lactone (SVL; ∼87% and 99%, respectively) and pravastatin lactone (PVL; ∼98% and 99%, respectively) were converted to the active hydroxy acid forms after 24 hours of incubation at 37°C. At acidic pH, conversion occurs to a lower extent, resulting in greater proportion of statin remaining in the more lipophilic lactone form. However, pH alteration did not influence the conversion of the hydroxy acid forms of simvastatin and pravastatin to the corresponding lactones. Furthermore, acidosis has been shown to hinder the metabolism of the lactone form of statins by inhibiting hepatic microsomal enzyme activities. Lipophilic SVL was found to be more cytotoxic to undifferentiated and differentiated skeletal muscle cells compared with more hydrophilic simvastatin hydroxy acid, PVL, and pravastatin hydroxy acid. Enhanced cytotoxicity of statins was observed under acidic conditions and is attributed to increased cellular uptake of the more lipophilic lactone or unionized hydroxy acid form. Consequently, our results suggest that comorbidities associated with acid-base imbalance can play a substantial role in the development and potentiation of statin-induced myotoxicity.
Collapse
Key Words
- cdna, complementary dna
- ct, cycle threshold
- dmem, dulbecco's modified eagle medium
- gapdh, glyceraldehyde-3-phosphate dehydrogenase
- hprt, hypoxanthine phosphoribosyl transferase
- hqc, high concentration quality control
- is, internal standard
- ldh, lactate dehydrogenase
- lloq, lower limit of quantification
- lov-a, lovastatin hydroxy acid
- lov-l, lovastatin lactone
- lqc, low concentration quality control
- mhc, myosin heavy chain
- mqc, medium concentration quality control
- mrna, messenger rna
- mrp, multiresistant protein
- mtt, thiazolyl blue tetrazolium bromide
- na, nonapplicable
- oatp, organic anionic transporting polypeptide
- pbs, phosphate buffer saline
- pva, pravastatin hydroxy acid
- pvl, pravastatin lactone
- rsd, relative standard deviation
- re, relative error
- rps12, ribosomal protein s12
- sva, simvastatin hydroxy acid
- svl, simvastatin lactone
- tbp, tata box-binding protein
Collapse
Affiliation(s)
- Dhiaa A Taha
- Division of Medicinal Chemistry and Structural Biology, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Cornelia H De Moor
- Division of Molecular and Cellular Science, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - David A Barrett
- Division of Molecular and Cellular Science, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Jong Bong Lee
- Division of Medicinal Chemistry and Structural Biology, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Raj D Gandhi
- Division of Molecular and Cellular Science, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Chee Wei Hoo
- Division of Medicinal Chemistry and Structural Biology, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Pavel Gershkovich
- Division of Medicinal Chemistry and Structural Biology, School of Pharmacy, University of Nottingham, Nottingham, UK.
| |
Collapse
|
17
|
Abstract
The concept of a pharmacokinetics-pharmacodynamics (PK/PD) assessment of drug development candidates is well established in pharmaceutical research and development, and PK/PD modeling is common practice in all pharmaceutical companies. A recent analysis (Morgan et al., Drug Discov Today 17(9-10):419-424, 2012) revealed however that insufficient certainty in the integrity of the causal chain of fundamental pharmacological steps from drug dosing through systemic exposure, target tissue exposure, and engagement of molecular target to pharmacological response is still the major driver of failure in phase II of clinical drug development. Despite the rise of molecular biomarkers, ethical, scientific, and practical constraints very often still prevent a direct assessment of each necessary step ultimately leading to an intended drug effect or an unintended adverse reaction. Yet, incomplete investigation of the causality of drug responses is a major risk for translational assessments and the prediction of drug responses in different species or other populations. Mechanism-based modeling and simulation (M&S) offers a means to investigate complex physiological and pharmacological processes and to complement experimental data for non-accessible steps in the pharmacological causal chain. With the help of two examples, it is illustrated, what level of physiological detail, state-of-the-art models can represent, how predictive these models are and how mechanism-based approaches can be combined with empirical correlation-based concepts.
Collapse
|
18
|
Involvement of Monocarboxylate Transporter 4 Expression in Statin-Induced Cytotoxicity. J Pharm Sci 2016; 105:1544-9. [PMID: 26935883 DOI: 10.1016/j.xphs.2016.01.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/11/2015] [Accepted: 01/05/2016] [Indexed: 11/24/2022]
Abstract
Statins, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, are the most widely used cholesterol-lowering agents for prevention of obstructive cardiovascular events. However, statins can cause a variety of skeletal muscle problems, and exercise leads to an increase in statin-induced muscle injury. Exercise induces the protein content of monocarboxylate transporter 4 (MCT4), which is expressed strongly in skeletal muscle and is thought to play a major role in the transport of metabolically important monocarboxylates such as l-lactate. We previously reported that α-cyano-4-hydroxycinnamate, an MCT4 inhibitor, increased the inhibition of growth of RD cells, a prototypic embryonal rhabdomyosarcoma cell line (an RD cell line), as a model of in vitro skeletal muscle, induced by a statin. However, it is unclear whether statin-induced RD cell cytotoxicity is associated with MCT4 expression. We, therefore, examined the relationship between statin-induced cytotoxicity and MCT4 expression in RD cells. Atorvastatin reduced the number of viable cells and upregulated MCT4, but not MCT1, mRNA level in a concentration-dependent manner. MCT4 knockdown suppressed atorvastatin-, simvastatin-, and fluvastatin-induced reduction of cell viability and apoptosis compared with negative control-treated cells. In this study, we demonstrated that MCT4 expression is associated with statin-induced cytotoxicity.
Collapse
|
19
|
Gavronski M, Volmer D, Hartikainen S, Zharkovsky A. Potential drug interactions with statins: Estonian register-based study. Open Med (Wars) 2015; 10:254-260. [PMID: 28352703 PMCID: PMC5152992 DOI: 10.1515/med-2015-0038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/31/2015] [Indexed: 01/05/2023] Open
Abstract
In Estonia, HMG-CoA reductase inhibitors are widely used to modify lipid levels but there are no current data on additional medicines prescribed alongside the statins. The aim of this study was to identify the frequency of potential clinically relevant interactions at a national level among an outpatient population treated with statins between January and June 2008, based on the prescription database of the Estonian Health Insurance Fund. This retrospective prevalence study included 203,646 outpatients aged 50 years or older, of whom 29,367 received statin therapy. The study analysed individuals who had used at least one prescription medicine for a minimum of 7 days concomitantly with statins. Potential drug interactions were analysed using Epocrates online, Stockley's Drug Interactions, and the drug interaction database developed in Estonia. Statins metabolised by the CYP3A4 isoenzyme were prescribed to 64% of all statin users. Medicines known to have potentially clinically significant interactions with statins were prescribed to 4.6% of patients. The drugs prescribed concomitantly most often with simvastatin were warfarin (5.7%) and amiodarone (3.9%), whereas digoxin (1.2%) and ethinylestradiol (2%) were prescribed with atorvastatin. Potential interactions were not detected in the treatment regimens of rosuvastatin, pravastatin, and fluvastatin users.
Collapse
Affiliation(s)
- Maia Gavronski
- Institute of Biomedicine and Translational Medicine, Tartu, Tartumaa Estonia
| | - Daisy Volmer
- Department of Pharmacy, Medical Faculty, University of Tartu, Estonia
| | - Sirpa Hartikainen
- School of Pharmacy, University of Eastern Finland, Kuopio Campus, Estonia
| | - Alexander Zharkovsky
- Department of Pharmacology, Centre of Excellence for Translational Medicine, Medical Faculty, University of Tartu, Estonia
| |
Collapse
|
20
|
Ferreira ES, Amaral ALS, Demonte A, Zanelli CF, Capraro J, Duranti M, Neves VA. Hypocholesterolaemic effect of rat-administered oral doses of the isolated 7S globulins from cowpeas and adzuki beans. J Nutr Sci 2015; 4:e7. [PMID: 26090103 PMCID: PMC4463012 DOI: 10.1017/jns.2014.70] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 09/16/2014] [Accepted: 11/11/2014] [Indexed: 02/01/2023] Open
Abstract
The role of seed proteins, especially soyabean 7S globulins, in controlling dyslipidaemia is widely acknowledged. Amino acid sequence homology among the proteins of this family could reflect similar biological functions in other species. The aim of the present study was to unveil a hypolipidaemic effect of the 7S globulins from cowpeas (7S-C) and adzuki beans (7S-A), administered orally to rats fed a hypercholesterolaemic (HC; high cholesterol and TAG) diet for 28 d. A total of forty-five rats were divided into five groups (nine rats per group): (1) standard (STD) diet; (2) HC diet; (3) HC diet + 7S-C (300 mg/kg per d); (4) HC diet + 7S-A (300 mg/kg per d); and (5) HC diet + simvastatin (SVT; 50 mg/kg per d), as a control. Significant decreases in food intake and final body weight of rats receiving HC + 7S-C and HC + 7S-A diets compared with groups fed the HC and STD diets were observed. Significant decreases in serum total and non-HDL-cholesterol of 7S-C, 7S-A and SVT groups were also observed. HDL-cholesterol levels increased in the 7S-C, 7S-A and SVT groups, while hepatic cholesterol and TAG concentrations were significantly lower than in the HC diet group for the 7S-C-supplemented group only. Faecal excretions of fat and cholesterol in HC diet groups were considerably higher in animals consuming the 7S globulins. The results show that cowpea and adzuki bean 7S globulins promote cholesterol-decreasing effects in hypercholesterolaemic rats even at low dosages, as already observed for other legume seed storage proteins of this family. This main effect is discussed in relation to the possible mechanisms of action.
Collapse
Affiliation(s)
- Ederlan S. Ferreira
- Department of Bromatological Analysis,
School of Pharmacy, Federal University of Bahia,
Barão de Jeremoabo Road, 147, 40170-115,
Salvador, BA, Brazil
| | - Ana Lucia S. Amaral
- Department of Food and Nutrition,
São Paulo State University, Rodovia Araraquara –
Jaú, km 1, 14801-902,
Araraquara, SP, Brazil
| | - Aureluce Demonte
- Department of Food and Nutrition,
São Paulo State University, Rodovia Araraquara –
Jaú, km 1, 14801-902,
Araraquara, SP, Brazil
| | - Cleslei F. Zanelli
- Department of Biological Sciences,
School of Pharmaceutical Sciences, São Paulo State University,
Rodovia Araraquara – Jaú, km 1,
14801-902, Araraquara,
SP, Brazil
| | - Jessica Capraro
- Department of Food, Environmental and Nutritional
Sciences (DeFENS), Università degli Studi di
Milano, Via G. Celoria, 2,
20133, Milan, Italy
| | - Marcello Duranti
- Department of Food, Environmental and Nutritional
Sciences (DeFENS), Università degli Studi di
Milano, Via G. Celoria, 2,
20133, Milan, Italy
| | - Valdir A. Neves
- Department of Food and Nutrition,
São Paulo State University, Rodovia Araraquara –
Jaú, km 1, 14801-902,
Araraquara, SP, Brazil
| |
Collapse
|
21
|
Kobayashi M. Role of Monocarboxylate Transporter in Statin-induced Cytotoxicity. YAKUGAKU ZASSHI 2015; 135:1227-33. [DOI: 10.1248/yakushi.15-00199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| |
Collapse
|
22
|
Mandas A, Mereu RM, Catte O, Saba A, Serchisu L, Costaggiu D, Peiretti E, Caminiti G, Vinci M, Casu M, Piludu S, Fossarello M, Manconi PE, Dessí S. Cognitive Impairment and Age-Related Vision Disorders: Their Possible Relationship and the Evaluation of the Use of Aspirin and Statins in a 65 Years-and-Over Sardinian Population. Front Aging Neurosci 2014; 6:309. [PMID: 25426067 PMCID: PMC4224124 DOI: 10.3389/fnagi.2014.00309] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 10/22/2014] [Indexed: 11/13/2022] Open
Abstract
Neurological disorders (Alzheimer’s disease, vascular and mixed dementia) and visual loss (cataract, age-related macular degeneration, glaucoma, and diabetic retinopathy) are among the most common conditions that afflict people of at least 65 years of age. An increasing body of evidence is emerging, which demonstrates that memory and vision impairment are closely, significantly, and positively linked and that statins and aspirin may lessen the risk of developing age-related visual and neurological problems. However, clinical studies have produced contradictory results. Thus, the intent of the present study was to reliably establish whether a relationship exist between various types of dementia and age-related vision disorders, and to establish whether statins and aspirin may or may not have beneficial effects on these two types of disorders. We found that participants with dementia and/or vision problems were more likely to be depressed and displayed worse functional ability in basic and instrumental activities of daily living than controls. Mini mental state examination scores were significantly lower in patients with vision disorders compared to subjects without vision disorders. A closer association with macular degeneration was found in subjects with Alzheimer’s disease than in subjects without dementia or with vascular dementia, mixed dementia, or other types of age-related vision disorders. When we considered the associations between different types of dementia and vision disorders and the use of statins and aspirin, we found a significant positive association between Alzheimer’s disease and statins on their own or in combination with aspirin, indicating that these two drugs do not appear to reduce the risk of Alzheimer’s disease or improve its clinical evolution and may, on the contrary, favor its development. No significant association in statin use alone, aspirin use alone, or the combination of these was found in subjects without vision disorders but with dementia, and, similarly, none in subjects with vision disorders but without dementia. Overall, these results confirm the general impression so far; namely, that macular degeneration may contribute to cognitive disorders (Alzheimer’s disease in particular). In addition, they also suggest that, while statin and aspirin use may undoubtedly have some protective effects, they do not appear to be magic pills against the development of cognitive impairment or vision disorders in the elderly.
Collapse
Affiliation(s)
- Antonella Mandas
- Dipartimento di Scienze Mediche, Università degli Studi di Cagliari , Cagliari , Italy
| | - Rosa Maria Mereu
- Divisione Geriatria, Centro Alzheimer e disturbi della Memoria, P.O. SS. Trinità , Cagliari , Italy
| | - Olga Catte
- Divisione Geriatria, Centro Alzheimer e disturbi della Memoria, P.O. SS. Trinità , Cagliari , Italy
| | - Antonio Saba
- Divisione Geriatria, Centro Alzheimer e disturbi della Memoria, P.O. SS. Trinità , Cagliari , Italy
| | - Luca Serchisu
- Dipartimento di Scienze Mediche, Università degli Studi di Cagliari , Cagliari , Italy
| | - Diego Costaggiu
- Dipartimento di Scienze Mediche, Università degli Studi di Cagliari , Cagliari , Italy
| | - Enrico Peiretti
- Dipartimento di Scienze Chirurgiche e Odontoiatriche, Clinica Oculistica, Università degli Studi di Cagliari , Cagliari , Italy
| | - Giulia Caminiti
- Dipartimento di Scienze Chirurgiche e Odontoiatriche, Clinica Oculistica, Università degli Studi di Cagliari , Cagliari , Italy
| | - Michela Vinci
- Dipartimento di Scienze Chirurgiche e Odontoiatriche, Clinica Oculistica, Università degli Studi di Cagliari , Cagliari , Italy
| | - Maura Casu
- Dipartimento di Scienze Chirurgiche e Odontoiatriche, Clinica Oculistica, Università degli Studi di Cagliari , Cagliari , Italy
| | - Stefania Piludu
- Dipartimento di Scienze Chirurgiche e Odontoiatriche, Clinica Oculistica, Università degli Studi di Cagliari , Cagliari , Italy
| | - Maurizio Fossarello
- Dipartimento di Scienze Chirurgiche e Odontoiatriche, Clinica Oculistica, Università degli Studi di Cagliari , Cagliari , Italy
| | - Paolo Emilio Manconi
- Dipartimento di Scienze Mediche, Università degli Studi di Cagliari , Cagliari , Italy
| | - Sandra Dessí
- Dipartimento di Scienze Mediche, Università degli Studi di Cagliari , Cagliari , Italy
| |
Collapse
|
23
|
Uremic toxins enhance statin-induced cytotoxicity in differentiated human rhabdomyosarcoma cells. Toxins (Basel) 2014; 6:2612-25. [PMID: 25192420 PMCID: PMC4179151 DOI: 10.3390/toxins6092612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/29/2014] [Accepted: 08/29/2014] [Indexed: 01/14/2023] Open
Abstract
The risk of myopathy and rhabdomyolysis is considerably increased in statin users with end-stage renal failure (ESRF). Uremic toxins, which accumulate in patients with ESRF, exert cytotoxic effects that are mediated by various mechanisms. Therefore, accumulation of uremic toxins might increase statin-induced cytotoxicity. The purpose of this study was to determine the effect of four uremic toxins-hippuric acid, 3-carboxy-4-methyl-5-propyl-2-furanpropionate, indole-3-acetic acid, and 3-indoxyl sulfate-on statin-induced myopathy. Differentiated rhabdomyosarcoma cells were pre-treated with the uremic toxins for seven days, and then the cells were treated with pravastatin or simvastatin. Cell viability and apoptosis were assessed by viability assays and flow cytometry. Pre-treatment with uremic toxins increased statin- but not cisplatin-induced cytotoxicity (p < 0.05 vs. untreated). In addition, the pre-treatment increased statin-induced apoptosis, which is one of the cytotoxic factors (p < 0.05 vs. untreated). However, mevalonate, farnesol, and geranylgeraniol reversed the effects of uremic toxins and lowered statin-induced cytotoxicity (p < 0.05 vs. untreated). These results demonstrate that uremic toxins enhance statin-induced apoptosis and cytotoxicity. The mechanism underlying this effect might be associated with small G-protein geranylgeranylation. In conclusion, the increased severity of statin-induced rhabdomyolysis in patients with ESRF is likely due to the accumulation of uremic toxins.
Collapse
|
24
|
Translational insight into statin-induced muscle toxicity: from cell culture to clinical studies. Transl Res 2014; 164:85-109. [PMID: 24530275 DOI: 10.1016/j.trsl.2014.01.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 01/15/2014] [Accepted: 01/17/2014] [Indexed: 02/06/2023]
Abstract
Statins are lipid-lowering drugs used widely to prevent and treat cardiovascular and coronary heart diseases. These drugs are among the most commonly prescribed medicines intended for long-term use. In general, statins are well tolerated. However, muscular adverse effects appear to be the most common obstacle that limits their use, resulting in poor patient compliance or even drug discontinuation. In addition, rare but potentially fatal cases of rhabdomyolysis have been reported with the use of these drugs, especially in the presence of certain risk factors. Previous reports have investigated statin-induced myotoxicity in vivo and in vitro using a number of cell lines, muscle tissues, and laboratory animals, in addition to randomized clinical trials, observational studies, and case reports. None of them have compared directly results from laboratory investigations with clinical observations of statin-related muscular adverse effects. To the best of our knowledge this is the first review article that combines laboratory investigation with clinical aspects of statin-induced myotoxicity. By reviewing published literature of in vivo, in vitro, and clinically relevant studies of statin myotoxicity, we aim to translate this important drug-related problem to establish a clear picture of proposed mechanisms that explain the risk factors and describe the diagnostic approaches currently used for evaluating the degree of muscle damage induced by these agents. This review provides baseline novel translational insight that can be used to enhance the safety profile, to minimize the chance of progression of these adverse effects to more severe and potentially fatal rhabdomyolysis, and to improve the overall patient compliance and adherence to long-term statin therapy.
Collapse
|
25
|
Murlasits Z, Radák Z. The Effects of Statin Medications on Aerobic Exercise Capacity and Training Adaptations. Sports Med 2014; 44:1519-30. [DOI: 10.1007/s40279-014-0224-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
26
|
Naderi S, Cho L. Statin intolerance: diagnosis, treatment and alternative therapies. ACTA ACUST UNITED AC 2014. [DOI: 10.2217/clp.14.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
27
|
Silva PM, Cardoso SM, Ferreira AM. Persistent lipid abnormalities in patients with hypertension and dyslipidemia treated with statins: results of the Portuguese hypertensive subpopulation of the Dyslipidemia International Study (DYSIS). Clin Exp Hypertens 2014; 37:116-21. [DOI: 10.3109/10641963.2014.913605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
28
|
Abstract
Statin drugs are highly effective in lowering blood concentrations of LDL-cholesterol, with concomitant reduction in risk of major cardiovascular events. Although statins are generally regarded as safe and well-tolerated, some users develop muscle symptoms that are mostly mild but in rare cases can lead to life-threatening rhabdomyolysis. The SEARCH genome-wide association study, which has been independently replicated, found a significant association between the rs4149056 (c.521T>C) single-nucleotide polymorphism (SNP) in the SLCO1B1 gene, and myopathy in individuals taking 80 mg simvastatin per day, with an odds ratio of 4.5 per rs4149056 C allele. The purpose of this paper is to assemble evidence relating to the analytical validity, clinical validity and clinical utility of using SLCO1B1 rs4149056 genotyping to inform choice and dose of statin treatment, with the aim of minimising statin-induced myopathy and increasing adherence to therapy. Genotyping assays for the rs4149056 SNP appear to be robust and accurate, though direct evidence for the performance of array-based platforms in genotyping individual SNPs was not found. Using data from the SEARCH study, calculated values for the clinical sensitivity, specificity, positive- and negative-predictive values of a test for the C allele to predict definite or incipient myopathy during 5 years of 80 mg/day simvastatin use were 70.4%, 73.7%, 4.1% and 99.4% respectively. There is a need for studies comparing the clinical validity of SLCO1B1 rs4149056 genotyping with risk scores for myopathy based on other factors such as racial background, statin type and dose, gender, body mass index, co-medications and co-morbidities. No direct evidence was found for clinical utility of statin prescription guided by SLCO1B1 genotype.
Collapse
Affiliation(s)
- Alison Stewart
- McKing Consulting Corp., Atlanta, Georgia, USA; Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
29
|
Regulation of multidrug resistance protein 2 (MRP2, ABCC2) expression by statins: Involvement of SREBP-mediated gene regulation. Int J Pharm 2013; 452:36-41. [DOI: 10.1016/j.ijpharm.2013.04.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 03/13/2013] [Accepted: 04/08/2013] [Indexed: 11/18/2022]
|
30
|
Han L, Li M, Liu X. Effects of long-term atorvastatin treatment on cardiac aging. Exp Ther Med 2013; 6:721-726. [PMID: 24137254 PMCID: PMC3787009 DOI: 10.3892/etm.2013.1208] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 06/05/2013] [Indexed: 01/14/2023] Open
Abstract
A number of studies have reported that atorvastatin (AVT) may have an important role in the delay of cardiac aging. However, the mechanism by which AVT affects cardiac aging has not been established. In this study, a series of experiments were performed to investigate the effects of AVT treatment on the cardiovascular system and the associated mechanism. Wistar rats were administered AVT or saline for 4 months. Age-related changes in the hearts were measured at the end of the experiment. The results showed that compared with young rats, the aged rats had significant changes indicative of myocardial aging, including increased blood lipid 1evelss, increased body weight, cardiac hypertrophy, larger myocardial cells, irregular muscle fibers, fewer deeply stained nuclei, smaller intercellular spaces, a larger number of apoptotic cells and increased levels of lipofuscin in myocardial tissue. However, long-term AVT treatment was able to significantly delay or even reverse these aging-related changes. In addition, these effects showed a certain dose-dependence. In general, long-term AVT treatment reduces blood lipids, inhibits cardiac hypertrophy, suppresses cardiomyocyte apoptosis and lowers the level of oxidative stress to protect the heart from aging.
Collapse
Affiliation(s)
- Lei Han
- Aerospace and Diving Medical Center, Navy General Hospital of Chinese PLA, Beijing 100037, P.R. China
| | | | | |
Collapse
|
31
|
Lippert J, Brosch M, von Kampen O, Meyer M, Siegmund HU, Schafmayer C, Becker T, Laffert B, Görlitz L, Schreiber S, Neuvonen PJ, Niemi M, Hampe J, Kuepfer L. A mechanistic, model-based approach to safety assessment in clinical development. CPT Pharmacometrics Syst Pharmacol 2012; 1:e13. [PMID: 23835795 PMCID: PMC3600730 DOI: 10.1038/psp.2012.14] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 09/14/2012] [Indexed: 12/28/2022] Open
Abstract
Assessing the safety of pharmacotherapies is a primary goal of clinical trials in drug development. The low frequency of relevant side effects, however, often poses a significant challenge for risk assessment. Methodologies allowing robust extrapolation of safety statistics based on preclinical data and information from clinical trials with limited numbers of patients are hence needed to further improve safety and efficacy in the drug development process. Here, we present a generic systems pharmacology approach integrating prior physiological and pharmacological knowledge, preclinical data, and clinical trial results, which allows predicting adverse event rates related to drug exposure. Possible fields of application involve high-risk populations, novel drug candidates, and different dosing scenarios. As an example, the approach is applied to simvastatin and pravastatin and the prediction of myopathy rates in a population with a genotype leading to a significantly increased myopathy risk.CPT: Pharmacometrics & Systems Pharmacology (2012) 1, e13; doi:10.1038/psp.2012.14; advance online publication 7 November 2012.
Collapse
Affiliation(s)
- J Lippert
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - M Brosch
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Campus Kiel, Christian-Albrechts-University, Kiel, Germany
| | - O von Kampen
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Campus Kiel, Christian-Albrechts-University, Kiel, Germany
| | - M Meyer
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - H.-U Siegmund
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - C Schafmayer
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Christian-Albrechts-University, Kiel, Germany
| | - T Becker
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Christian-Albrechts-University, Kiel, Germany
| | - B Laffert
- Cell Culture Service, Hamburg, Germany
| | - L Görlitz
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - S Schreiber
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Campus Kiel, Christian-Albrechts-University, Kiel, Germany
| | - P J Neuvonen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - M Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - J Hampe
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Campus Kiel, Christian-Albrechts-University, Kiel, Germany
| | - L Kuepfer
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| |
Collapse
|
32
|
Kobayashi M, Hidaka K, Chisaki I, Takahashi N, Ogura J, Itagaki S, Hirano T, Yamaguchi H, Iseki K. Effects of Acidification and Alkalinization Agent on Statins-induced Muscle Toxicity. YAKUGAKU ZASSHI 2012; 132:609-15. [DOI: 10.1248/yakushi.132.609] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Kazuhiro Hidaka
- Laboratory of Clinical Pharmaceutics and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Ikumi Chisaki
- Laboratory of Clinical Pharmaceutics and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | | | - Jiro Ogura
- Laboratory of Clinical Pharmaceutics and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Shirou Itagaki
- Laboratory of Clinical Pharmaceutics and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Takeshi Hirano
- Laboratory of Clinical Pharmaceutics and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Hiroaki Yamaguchi
- Laboratory of Clinical Pharmaceutics and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Ken Iseki
- Laboratory of Clinical Pharmaceutics and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| |
Collapse
|
33
|
Ferreira ES, Silva MA, Demonte A, Neves VA. β-conglycinin combined with fenofibrate or rosuvastatin have exerted distinct hypocholesterolemic effects in rats. Lipids Health Dis 2012; 11:11. [PMID: 22243700 PMCID: PMC3285525 DOI: 10.1186/1476-511x-11-11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 01/13/2012] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND There is increasing interest in non-pharmacological control of cholesterol and triglyceride levels in the plasma and diet-drug association represent an important area of studies. The objective of this study was to observe the hypocholesterolemic effect of soybean β-conglycinin (7S protein) alone and combined with fenofibrate and rosuvastatin, two hypolipidemic drugs. METHODS The protein and drugs were administered orally once a day to rats and the effects were evaluated after 28 days. Wistar rats were divided into six groups (n = 9): hypercholesterolemic diet (HC), HC+7S protein (300 mg.kg-1 day-1) (HC-7S), HC+fenofibrate (30 mg.kg-1 day-1)(HC-FF), HC+rosuvastatin (10 mg.kg-1 day-1)(HC-RO), HC+7S+fenofibrate (HC-7S-FF) and HC+7S+rosuvastatin (HC-7S-RO). RESULTS Animals in HC-7S, HC-FF and HC-RO exhibited reductions of 22.9, 35.8 and 18.8% in total plasma cholesterol, respectively. In HC-7S-FF, animals did not show significant alteration of the level in HC+FF while the group HC-7S-RO showed a negative effect in comparison with groups taking only protein (HC-7S) or drug (HC-RO). The administration of the protein, fenofibrate and rosuvastatin alone caused increases in the plasma HDL-C of the animals, while the protein-drug combinations led to an increase compared to HC-FF and HC-RO. The plasma concentration of triacylgycerides was significantly reduced in the groups without association, while HC-7S-FF showed no alteration and HC-7S-RO a little reduction. CONCLUSION The results of our study indicate that conglycinin has effects comparable to fenofibrate and rosuvastatin on the control of plasma cholesterol, HDL-C and triacylglycerides, when given to hypercholesterolemic rats, and suggests that the association of this protein with rosuvastatin alters the action of drug in the homeostasis of cholesterol.
Collapse
Affiliation(s)
- Ederlan S Ferreira
- Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University -UNESP, Araraquara, SP, Brazil
| | - Maraiza A Silva
- Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University -UNESP, Araraquara, SP, Brazil
| | - Aureluce Demonte
- Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University -UNESP, Araraquara, SP, Brazil
| | - Valdir A Neves
- Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University -UNESP, Araraquara, SP, Brazil
| |
Collapse
|
34
|
Gee RH, Plant NJ, Johnston JD, Plant KE. Differential sensitivity to toxicity of statins: A comparison of liver and muscle cell lines. Toxicology 2011. [DOI: 10.1016/j.tox.2011.09.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
35
|
Gazzerro P, Proto MC, Gangemi G, Malfitano AM, Ciaglia E, Pisanti S, Santoro A, Laezza C, Bifulco M. Pharmacological actions of statins: a critical appraisal in the management of cancer. Pharmacol Rev 2011; 64:102-46. [PMID: 22106090 DOI: 10.1124/pr.111.004994] [Citation(s) in RCA: 316] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Statins, among the most commonly prescribed drugs worldwide, are cholesterol-lowering agents used to manage and prevent cardiovascular and coronary heart diseases. Recently, a multifaceted action in different physiological and pathological conditions has been also proposed for statins, beyond anti-inflammation and neuroprotection. Statins have been shown to act through cholesterol-dependent and -independent mechanisms and are able to affect several tissue functions and modulate specific signal transduction pathways that could account for statin pleiotropic effects. Typically, statins are prescribed in middle-aged or elderly patients in a therapeutic regimen covering a long life span during which metabolic processes, aging, and concomitant novel diseases, including cancer, could occur. In this context, safety, toxicity, interaction with other drugs, and the state of health have to be taken into account in subjects treated with statins. Some evidence has shown a dichotomous effect of statins with either cancer-inhibiting or -promoting effects. To date, clinical trials failed to demonstrate a reduced cancer occurrence in statin users and no sufficient data are available to define the long-term effects of statin use over a period of 10 years. Moreover, results from clinical trials performed to evaluate the therapeutic efficacy of statins in cancer did not suggest statin use as chemotherapeutic or adjuvant agents. Here, we reviewed the pharmacology of the statins, providing a comprehensive update of the current knowledge of their effects on tissues, biological processes, and pathological conditions, and we dissected the disappointing evidence on the possible future use of statin-based drugs in cancer therapy.
Collapse
Affiliation(s)
- Patrizia Gazzerro
- Department of Pharmaceutical and Biomedical Sciences, University of Salerno, Via Ponte Don Melillo, 84084 Fisciano (Salerno), Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kobayashi M, Gouda K, Chisaki I, Ochiai M, Itagaki S, Iseki K. Regulation mechanism of ABCA1 expression by statins in hepatocytes. Eur J Pharmacol 2011; 662:9-14. [DOI: 10.1016/j.ejphar.2011.04.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 03/28/2011] [Accepted: 04/15/2011] [Indexed: 10/18/2022]
|
37
|
Mor A, Wortmann RL, Mitnick HJ, Pillinger MH. Drugs Causing Muscle Disease. Rheum Dis Clin North Am 2011; 37:219-31, vi. [DOI: 10.1016/j.rdc.2011.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
38
|
Generaux GT, Bonomo FM, Johnson M, Mahar Doan KM. Impact ofSLCO1B1(OATP1B1) andABCG2(BCRP) genetic polymorphisms and inhibition on LDL-C lowering and myopathy of statins. Xenobiotica 2011; 41:639-51. [DOI: 10.3109/00498254.2011.562566] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
39
|
Baker WL, Datta R. Pitavastatin: a new 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitor for the treatment of hyperlipidemia. Adv Ther 2011; 28:13-27. [PMID: 21170619 DOI: 10.1007/s12325-010-0092-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Indexed: 10/18/2022]
Abstract
Statins have proven beneficial for reducing both primary and secondary events in patients with coronary heart disease. Tight control of serum lipid parameters in these patients is recommended by the most recent clinical guidelines. Although numerous lipid-lowering treatments are available, only a small percentage of eligible patients receive therapy and fewer achieve their lipid-lowering goals. Thus it is clear that new treatment strategies to manage patients with lipid abnormalities are warranted. Pitavastatin (Lival; Kowa Pharmaceuticals America, Montgomery, AL, USA) has been recently approved for the treatment of hypercholesterolemia and combined dyslipidemia. Pitavastatin 1-4 mg/day has shown similar low-density lipoprotein-reducing activity to other commercially available statins, including simvastatin and atorvastatin. Adverse events occurred at similar rates to other statins in clinical trials with favorable effects seen in patients with dyslipidemia and metabolic syndrome. Pharmacokinetic drug-drug interactions are minimized due to the lack of significant metabolism of pitavastatin by the cytochrome P450 enzyme system, although some drugs affect its uptake into hepatocytes and should be avoided. In addition to its higher acquisition cost, pitavastatin has not been shown to improve clinical outcomes in high-risk patient populations and thus may not be the agent of choice in many patients at this time in lieu of cheaper, clinically proven alternatives.
Collapse
|
40
|
The effects of time of treatment on statin muscle toxicity: Preliminary characterization of a human muscle cell line. Toxicology 2010. [DOI: 10.1016/j.tox.2010.08.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
Furugen A, Kobayashi M, Narumi K, Watanabe M, Otake S, Itagaki S, Iseki K. AMP-activated protein kinase regulates the expression of monocarboxylate transporter 4 in skeletal muscle. Life Sci 2010; 88:163-8. [PMID: 21070787 DOI: 10.1016/j.lfs.2010.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 10/18/2010] [Accepted: 11/01/2010] [Indexed: 11/17/2022]
Abstract
AIMS The aim of this study was to determine the effect of 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR), an AMP-activated protein kinase (AMPK) activator, on monocarboxylate transporter 4 (MCT4) expression in rat skeletal muscle and a prototypic embryonal rhabdomyosarcoma cell line (RD cells). MAIN METHODS We examined the alteration in Glucose transporter 4 (GLUT4) and MCT4 mRNA levels by quantitative real-time PCR. Alteration in GLUT4 and MCT4 protein levels was examined by Western blotting. KEY FINDINGS In an in vivo study, AICAR increased MCT4 mRNA and protein levels in a fiber-type specific manner. In an in vitro study, AICAR increased MCT4 mRNA and protein levels. Moreover, AICAR-induced MCT4 expression was blocked by Compound C, an AMPK inhibitor. SIGNIFICANCE In this study, we found that AMPK activation induced expression of MCT4 in RD cells and rat skeletal muscle in a fiber-type specific manner. These results indicate the possible involvement of an AMPK-mediated pathway associated with MCT4 expression in skeletal muscle.
Collapse
Affiliation(s)
- Ayako Furugen
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Hyperlipidemia, also known as high blood cholesterol, is a cardiovascular health risk that affects more than one third of adults in the United States. Statins are commonly prescribed and successful lipid-lowering medications that reduce the risks associated with cardiovascular disease. The side effects most commonly associated with statin use involve muscle cramping, soreness, fatigue, weakness, and, in rare cases, rapid muscle breakdown that can lead to death. Often, these side effects can become apparent during or after strenuous bouts of exercise. Although the mechanisms by which statins affect muscle performance are not entirely understood, recent research has identified some common causative factors. As musculoskeletal and exercise specialists, physical therapists have a unique opportunity to identify adverse effects related to statin use. The purposes of this perspective article are: (1) to review the metabolism and mechanisms of actions of statins, (2) to discuss the effects of statins on skeletal muscle function, (3) to detail the clinical presentation of statin-induced myopathies, (4) to outline the testing used to diagnose statin-induced myopathies, and (5) to introduce a role for the physical therapist for the screening and detection of suspected statin-induced skeletal muscle myopathy.
Collapse
|
43
|
|
44
|
UDP-Glucuronosyltransferase (UGT) Polymorphisms Affect Atorvastatin Lactonization In Vitro and In Vivo. Clin Pharmacol Ther 2009; 87:65-73. [DOI: 10.1038/clpt.2009.181] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
45
|
Chisaki I, Kobayashi M, Itagaki S, Hirano T, Iseki K. Liver X receptor regulates expression of MRP2 but not that of MDR1 and BCRP in the liver. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:2396-403. [PMID: 19728987 DOI: 10.1016/j.bbamem.2009.08.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 08/18/2009] [Accepted: 08/25/2009] [Indexed: 01/16/2023]
Abstract
Liver X receptors (LXRs) belong to the nuclear hormone receptor superfamily. Multidrug resistance-associated protein 2 (MRP2), multidrug resistance 1 (MDR1) and breast cancer resistance protein (BCRP) play an important role in the efflux of a broad range of endogenous and xenobiotic compounds from hepatocytes. Since the effects of LXR activation on there transporters have been obscure, we investigated the effects of LXR agonists, TO901317 and 25-hydroxycholesterol, on MRP2, MDR1, BCRP expression in HepG2 cells and the rat liver. In an in vitro study, TO901317 increased ABCA1, an LXR target gene, and MRP2 mRNA and protein levels. On the other hand, TO901317 had little effect on MDR1 and BCRP mRNA levels. In an in vivo study, Abca1 and Mrp2 mRNA and protein levels were increased by TO901317, but TO901317 had no effect on Mdr1a and Bcrp mRNA levels in the rat liver. Moreover, TO901317-induced MRP2 mRNA expression was blocked by LXRalpha knockdown. In this study, we demonstrated that LXR activation induced expression of MRP2 but not that of MDR1 and BCRP in hepatocytes. The results suggest that agonists for LXR activate transcription of the MRP2 gene in order to promote excretion of endogenous and xenobiotic compounds from hepatocytes into bile.
Collapse
Affiliation(s)
- Ikumi Chisaki
- Laboratory of Clinical Pharmaceutics and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | | | | | | | | |
Collapse
|
46
|
Fukami T, Ishii T, Io T, Suzuki N, Suzuki T, Yamamoto K, Xu J, Ramamoorthy A, Tomono K. Nanoparticle Processing in the Solid State Dramatically Increases the Cell Membrane Permeation of a Cholesterol-Lowering Drug, Probucol. Mol Pharm 2009; 6:1029-35. [DOI: 10.1021/mp9000487] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Toshiro Fukami
- College of Pharmacy, Nihon University, Chiba 274-8555, Japan, and Department of Pharmaceutical Sciences, College of Pharmacy, Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Tatsuya Ishii
- College of Pharmacy, Nihon University, Chiba 274-8555, Japan, and Department of Pharmaceutical Sciences, College of Pharmacy, Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Takeshi Io
- College of Pharmacy, Nihon University, Chiba 274-8555, Japan, and Department of Pharmaceutical Sciences, College of Pharmacy, Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Naoto Suzuki
- College of Pharmacy, Nihon University, Chiba 274-8555, Japan, and Department of Pharmaceutical Sciences, College of Pharmacy, Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Toyofumi Suzuki
- College of Pharmacy, Nihon University, Chiba 274-8555, Japan, and Department of Pharmaceutical Sciences, College of Pharmacy, Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Kazutoshi Yamamoto
- College of Pharmacy, Nihon University, Chiba 274-8555, Japan, and Department of Pharmaceutical Sciences, College of Pharmacy, Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Jiadi Xu
- College of Pharmacy, Nihon University, Chiba 274-8555, Japan, and Department of Pharmaceutical Sciences, College of Pharmacy, Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Ayyalusamy Ramamoorthy
- College of Pharmacy, Nihon University, Chiba 274-8555, Japan, and Department of Pharmaceutical Sciences, College of Pharmacy, Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Kazuo Tomono
- College of Pharmacy, Nihon University, Chiba 274-8555, Japan, and Department of Pharmaceutical Sciences, College of Pharmacy, Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
47
|
Current world literature. Curr Opin Rheumatol 2008; 20:729-35. [PMID: 18946335 DOI: 10.1097/bor.0b013e328317a234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|