1
|
Yarrarapu SNS, Goyal A, Venkata VS, Panchal V, Sivasubramanian BP, Du DT, Jakulla RS, Pamulapati H, Afaq MA, Owens S, Dalia T. Comprehensive review of statin-intolerance and the practical application of Bempedoic Acid. J Cardiol 2024; 84:22-29. [PMID: 38521120 DOI: 10.1016/j.jjcc.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
Statin-intolerance (SI) has prevalence between 8.0 % and 10 %, and muscular complaints are the most common reason for discontinuation. Bempedoic acid (BA), an ATP citrate lyase inhibitor, decreases hepatic generation of cholesterol, upregulates low-density lipoprotein (LDL) receptor expression in the liver, and eventually clears circulating LDL-cholesterol from the blood. Multiple randomized clinical trials studying BA demonstrate a reduction in LDL levels by 17-28 % in SI. The CLEAR OUTCOME trial established significant cardiovascular benefits with BA. A dose of 180 mg/day of BA showed promising results. BA alone or in combination with ezetimibe is US Food and Drug Administration-approved for use in adults with heterozygous familial hypercholesterolemia and/or established atherosclerotic cardiovascular disease. BA reduced HbA1c by 0.12 % (p < 0.0001) in patients with diabetes. Adverse events of BA include myalgia (4.7 %), anemia (3.4 %), and increased aminotransferases (0.3 %). BA can cause up to four times higher risk of gout in those with a previous gout diagnosis or high serum uric acid levels. Reports of increased blood urea nitrogen and serum creatinine were noted. Current evidence does not demonstrate a reduction in deaths from cardiovascular causes. More studies that include a diverse population and patients with both high and low LDL levels should be conducted. We recommend that providers consider BA as an adjunct to statin therapy in patients with a maximally tolerated dosage to specifically target LDL levels.
Collapse
Affiliation(s)
- Siva Naga S Yarrarapu
- Department of Internal Medicine, Rutgers/Monmouth Medical Center, Long Branch, NJ, USA
| | - Amandeep Goyal
- Department of Cardiology, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Viraj Panchal
- Department of Medicine, Smt. NHL Municipal Medical College, Ahmedabad, Gujarat, India
| | | | - Doantrang T Du
- Department of Internal Medicine, Rutgers/Monmouth Medical Center, Long Branch, NJ, USA
| | - Roopesh Sai Jakulla
- Department of Internal Medicine, University of Missouri, Kansas City, MO, USA
| | - Hema Pamulapati
- Department of Cardiology, Hays Medical Center, Hays, KS, USA
| | - Mazhar A Afaq
- Department of Cardiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Steven Owens
- Department of Cardiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Tarun Dalia
- Department of Cardiology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
2
|
Kee PS, Chin PKL, Kennedy MA, Maggo SDS. Pharmacogenetics of Statin-Induced Myotoxicity. Front Genet 2020; 11:575678. [PMID: 33193687 PMCID: PMC7596698 DOI: 10.3389/fgene.2020.575678] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022] Open
Abstract
Statins, a class of lipid-lowering medications, have been a keystone treatment in cardiovascular health. However, adverse effects associated with statin use impact patient adherence, leading to statin discontinuation. Statin-induced myotoxicity (SIM) is one of the most common adverse effects, prevalent across all ages, genders, and ethnicities. Although certain demographic cohorts carry a higher risk, the impaired quality of life attributed to SIM is significant. The pathogenesis of SIM remains to be fully elucidated, but it is clear that SIM is multifactorial. These factors include drug-drug interactions, renal or liver dysfunction, and genetics. Genetic-inferred risk for SIM was first reported by a landmark genome-wide association study, which reported a higher risk of SIM with a polymorphism in the SLCO1B1 gene. Since then, research associating genetic factors with SIM has expanded widely and has become one of the foci in the field of pharmacogenomics. This review provides an update on the genetic risk factors associated with SIM.
Collapse
Affiliation(s)
- Ping Siu Kee
- Gene Structure and Function Laboratory, Carney Centre for Pharmacogenomics, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | | | - Martin A. Kennedy
- Gene Structure and Function Laboratory, Carney Centre for Pharmacogenomics, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Simran D. S. Maggo
- Gene Structure and Function Laboratory, Carney Centre for Pharmacogenomics, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|
3
|
Attalah Nee Rezkallah C, Thongkum A, Zhu C, Chen QM. Resveratrol for protection against statin toxicity in C2C12 and H9c2 cells. J Biochem Mol Toxicol 2020; 34:e22484. [PMID: 32196851 DOI: 10.1002/jbt.22484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/02/2020] [Accepted: 02/14/2020] [Indexed: 01/01/2023]
Abstract
Statins are among the most commonly prescribed drugs for the treatment of high blood cholesterol. Myotoxicity of statins in certain individuals is often a severe side effect leading to withdrawal. Using C2C12 and H9c2 cells, both exhibiting characteristics of skeletal muscle cells, we addressed whether resveratrol (RSV) can prevent statin toxicity. Statins decreased cell viability in a dose and time-dependent manner. Among the five statins tested, atorvastatin, simvastatin, lovastatin, pravastatin, and fluvastatin, simvastatin is the most toxic one. Simvastatin at 10 µM caused about 65% loss of metabolic activity as measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays in C2C12 cells or H9c2 cells. Inhibition of metabolic activity correlates with an increase in caspase activity. RSV was found to protect H9c2 cells from simvastatin-induced activation of caspase-3/7. However, such protection was not found in C2C12 cells. This cell type-dependent effect of RSV adds to the complexity in muscle cell toxicity of statins.
Collapse
Affiliation(s)
| | - Angkana Thongkum
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Chao Zhu
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona.,Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Qin M Chen
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona.,Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, Arizona
| |
Collapse
|
4
|
Chen TL, Lin CS, Lin JA, Yeh CC, Sung LC, Chang YC, Shih CC, Liao CC. Evaluating Risk of Incident Diabetes Between Patients Who Used Lovastatin and Red Yeast Rice Prescriptions (LipoCol Forte): A Retrospective Cohort Study Based on a Real-World Database. Diabetes Metab Syndr Obes 2020; 13:89-98. [PMID: 32021355 PMCID: PMC6956995 DOI: 10.2147/dmso.s223833] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 12/09/2019] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE This study aimed to evaluate the risk of incident diabetes between people who used lovastatin and red yeast rice (RYR) prescriptions. METHODS A retrospective cohort study was performed to analyze the real-world database of Taiwan's National Health Insurance. We identified the RYR cohort, which included 34,504 persons age 20 years or older who began their use of a RYR prescription in 2010-2014. A comparison cohort of 34,504 adults beginning the use of lovastatin was selected from the same dataset, which was matched by age and sex. Both cohorts had no diabetes before the use of the medications. Events of incident diabetes in 2000-2015 were ascertained from medical claims. Adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) of incident diabetes associated with the use of RYR prescriptions were calculated. RESULTS The incidences of diabetes for the RYR cohort and the lovastatin cohort were 1.01 and 2.59 per 100 person-years, respectively (P < 0.0001). Compared with the lovastatin cohort, the adjusted HR of incident diabetes was 0.46 (95% CI 0.43-0.50) for people who used RYR prescriptions. The association between reduced incident diabetes and use of RYR prescriptions was significant in various subgroups. There was a dose-response relationship between RYR prescriptions and the reduced risk of incident diabetes. CONCLUSION We raised the possibility that people who used RYR prescriptions may have a lower risk of incident diabetes compared with the lovastatin cohort.
Collapse
Affiliation(s)
- Ta-Liang Chen
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chao-Shun Lin
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Jui-An Lin
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Chieh Yeh
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Surgery, University of Illinois, Chicago, IL, USA
| | - Li-Chin Sung
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Cheng Chang
- Division of Endocrinology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Chuan Shih
- School of Chinese Medicine for Post-Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chien-Chang Liao
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Research Center of Big Data and Meta-Analysis, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
5
|
Bielecka-Dabrowa A, Fabis J, Mikhailidis DP, von Haehling S, Sahebkar A, Rysz J, Banach M. Prosarcopenic Effects of Statins May Limit Their Effectiveness in Patients with Heart Failure. Trends Pharmacol Sci 2018; 39:331-353. [DOI: 10.1016/j.tips.2018.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 12/25/2022]
|
6
|
Oxidative stress as a possible mechanism of statin-induced myopathy. Inflammopharmacology 2018; 26:667-674. [PMID: 29574631 DOI: 10.1007/s10787-018-0469-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/15/2018] [Indexed: 01/05/2023]
Abstract
Statins, inhibitors of hydroxy methyl glutaryl coenzyme-A (HMG-CoA) reductase, are the most widely used drugs for treating hypercholesterolemia. However, statins can cause disabling myopathy as their main adverse effect. Several molecular mechanisms underlie the statin-induced myopathy including the decrease in the levels of essential mevalonate and cholesterol derivatives. This review discusses a further mechanism involving the loss of other anti-oxidant defenses besides ubiquinone (Co-Q) in skeletal muscles which produce a significant amount of reactive oxygen species (ROS). Therefore, to maintain their function, skeletal muscles need a high level of anti-oxidants.
Collapse
|
7
|
Hou T, Li Y, Chen W, Heffner RR, Vladutiu GD. Histopathologic and Biochemical Evidence for Mitochondrial Disease Among 279 Patients with Severe Statin Myopathy. J Neuromuscul Dis 2018; 4:77-87. [PMID: 28269789 DOI: 10.3233/jnd-160184] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Statins have well-known benefits in the prevention of cardiovascular disease, however, 7-29% of patients develop muscle side effects and up to 0.5% develop severe symptoms. Mitochondrial dysfunction has been associated with severe statin-induced myopathy (SM); however, there is a paucity of systematic studies in affected individuals. OBJECTIVES The goal of this study was to combine clinical and laboratory features with quantitative biochemical and histopathologic studies of skeletal muscle biopsies from SM cases to determine what proportion could be attributed to mitochondrial dysfunction and how many of these had primary respiratory chain defects. METHODS A retrospective analysis was performed on patient records derived from 279 SM patients whose muscle biopsies were referred to our clinical diagnostic laboratory for analysis. Clinical, histopathologic and biochemical features were compared with two myopathic control groups unexposed to statins: individuals with idiopathic mitochondrial myopathy (MMP; n = 94) and with unknown metabolic myopathy (UMP; n = 86); normal controls were unavailable for this record review study. RESULTS More SM patients had significantly elevated plasma CK than in the other two groups (p < 0.01). A subset of SM patients (67 of 279; 24%) had histopathologic and/or electron microscopic (EM) evidence for mitochondrial dysfunction in skeletal muscle; more cases were identified by EM than by histochemical analysis. Of 279 cases, 29 (10%) were confirmed to have respiratory chain defects by biochemical analysis; 4 of these had mitochondrial abnormalities by EM. An additional 20 cases had mitochondrial abnormalities by EM without a biochemical diagnosis. CONCLUSIONS Both primary and secondary mitochondrial dysfunction was found in subsets of SM patients. The fact that respiratory chain defects were not found in most cases with histopathologic mitochondrial abnormalities does not rule out primary mitochondrial disease in these cases, however, it is more likely that secondary effects on mitochondrial structure and function have occurred; molecular analysis may be helpful only in a small number of cases.
Collapse
Affiliation(s)
- Tieying Hou
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Yilan Li
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Weiwei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Reid R Heffner
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Georgirene D Vladutiu
- Departments of Pediatrics, Neurology, and Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.,Kaleida Health Laboratories, Buffalo, NY, USA
| |
Collapse
|
8
|
Gutierrez-Mariscal FM, Yubero-Serrano EM, Villalba JM, Lopez-Miranda J. Coenzyme Q10: From bench to clinic in aging diseases, a translational review. Crit Rev Food Sci Nutr 2018; 59:2240-2257. [DOI: 10.1080/10408398.2018.1442316] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Francisco M. Gutierrez-Mariscal
- Lipid and Atherosclerosis Unit, Department of Internal Medicine/IMIBIC/Reina Sofia University Hospital/University of Córdoba, Córdoba, Spain; CIBER Fisiología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Elena M. Yubero-Serrano
- Lipid and Atherosclerosis Unit, Department of Internal Medicine/IMIBIC/Reina Sofia University Hospital/University of Córdoba, Córdoba, Spain; CIBER Fisiología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose M. Villalba
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, ceiA3, Córdoba, Spain
| | - Jose Lopez-Miranda
- Lipid and Atherosclerosis Unit, Department of Internal Medicine/IMIBIC/Reina Sofia University Hospital/University of Córdoba, Córdoba, Spain; CIBER Fisiología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
9
|
Elsaid O, Taylor B, Zaleski A, Panza G, Thompson PD. Rationale for investigating metformin as a protectant against statin-associated muscle symptoms. J Clin Lipidol 2017; 11:1145-1151. [DOI: 10.1016/j.jacl.2017.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/23/2017] [Accepted: 06/29/2017] [Indexed: 11/30/2022]
|
10
|
Powers JM, Murphy G, Ralph N, O'Gorman SM, Murphy JEJ. Polypharmacy and sun exposure: Implications for mitochondrial DNA deletions in skin. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017. [PMID: 28649007 DOI: 10.1016/j.jphotobiol.2017.06.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Most somatic cells contain many copies of mitochondrial DNA (mtDNA). Because of both the high copy number and the lack of repair mechanisms available to mtDNA, damage to it largely goes unrepaired, and can accumulate over time. Large scale deletions are a recognised type of damage sustained by mtDNA as a consequence of exposure to the ultraviolet light in sunlight. A group of patients were identified as having abnormally high levels of either a 4977 base pair deletion (mtDNA4977) or 3895 base pair deletion (mtDNA3895), in mtDNA from sun exposed skin or skin suspected to be a non-melanoma skin cancer, but not in their non-sun exposed skin biopsies. In three of the four cases, skin cancer was ruled out due to histological testing. Additional factors from these patients' medical histories were studied, and it was noted that they shared diagnoses for multiple pathologies common to an older population, and that they were being treated with the same or related pharmaceuticals, including some that had been known to cause dermal side effects. Investigation into the biochemistry underlying the symptoms, the effects of sun exposure and side effects of the prescribed pharmaceuticals revealed a possible synergistic relationship leading to the localised high levels of mtDNA deletions.
Collapse
Affiliation(s)
- Julia Montelin Powers
- Mitochondrial Biology & Radiation Research Centre, Dept Life Sciences, IT Sligo, Sligo, Ireland.
| | | | - Nikki Ralph
- Dept of Dermatology, Beaumont Hospital, Dublin, Ireland
| | | | - James E J Murphy
- Mitochondrial Biology & Radiation Research Centre, Dept Life Sciences, IT Sligo, Sligo, Ireland
| |
Collapse
|
11
|
Murad HAS. L-Carnitine, but not coenzyme Q10, enhances the anti-osteoporotic effect of atorvastatin in ovariectomized rats. J Zhejiang Univ Sci B 2016; 17:43-53. [PMID: 26739525 DOI: 10.1631/jzus.b1500065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Statins' therapy in osteoporosis can aggravate muscle damage. This study was designed to assess which agent, L-carnitine or coenzyme Q10, could enhance the anti-osteoporotic effect of atorvastatin while antagonizing myopathy in ovariectomized rats. METHODS Forty-eight female Sprague Dawley rats were used; forty rats were ovariectomized while eight were sham-operated. Eight weeks post-ovariectomy, rats were divided into ovariectomized-untreated group and four ovariectomized-treated groups (n=8) which received by gavage (mg/(kg∙d), for 8 weeks) 17β-estradiol (0.1), atorvastatin (50), atorvastatin (50)+L-carnitine (100), or atorvastatin (50)+coenzyme Q10 (20). At the end of therapy, bone mineral density (BMD), bone mineral content (BMC), and serum levels of bone metabolic markers (BMMs) and creatine kinase (CK) were measured. Femurs were used for studying the breaking strength and histopathological changes. RESULTS Treatment with atorvastatin+L-carnitine restored BMD, BMC, and bone strength to near normal levels. Estrogen therapy restored BMD and BMC to near normal levels, but failed to increase bone strength. Although atorvastatin and atorvastatin+coenzyme Q10 improved BMD, BMC, and bone strength, they failed to restore levels to normal. All treatments decreased BMMs and improved histopathological changes maximally with atorvastatin+L-carnitine which restored levels to near normal. Atorvastatin aggravated the ovariectomy-induced increase in CK level while estrogen, atorvastatin+L-carnitine, and atorvastatin+coenzyme Q10 decreased its level mainly with atorvastatin+L-carnitine which restored the level to near normal. CONCLUSIONS Co-administration of L-carnitine, but not coenzyme Q10, enhances the anti-osteoporotic effect of atorvastatin while antagonizing myopathy in ovariectomized rats. This could be valuable in treatment of osteoporotic patients. However, further confirmatory studies are needed.
Collapse
Affiliation(s)
- Hussam A S Murad
- Department of Pharmacology, Faculty of Medicine, Rabigh, King Abdulaziz University, Jeddah 21589, Saudi Arabia.,Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo 11562, Egypt
| |
Collapse
|
12
|
Gluba-Brzozka A, Franczyk B, Toth PP, Rysz J, Banach M. Molecular mechanisms of statin intolerance. Arch Med Sci 2016; 12:645-58. [PMID: 27279860 PMCID: PMC4889699 DOI: 10.5114/aoms.2016.59938] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 01/13/2023] Open
Abstract
Statins reduce cardiovascular morbidity and mortality in primary and secondary prevention. Despite their efficacy, many persons are unable to tolerate statins due to adverse events such as hepatotoxicity and myalgia/myopathy. In the case of most patients, it seems that mild-to-moderate abnormalities in liver and muscle enzymes are not serious adverse effects and do not outweigh the benefits of coronary heart disease risk reduction. The risk for mortality or permanent organ damage ascribed to statin use is very small and limited to cases of myopathy and rhabdomyolysis. Statin-induced muscle-related adverse events comprise a highly heterogeneous clinical disorder with numerous, complex etiologies and a variety of genetic backgrounds. Every patient who presents with statin-related side effects cannot undergo the type of exhaustive molecular characterization that would include all of these mechanisms. Frequently the only solution is to either discontinue statin therapy/reduce the dose or attempt intermittent dosing strategies at a low dose.
Collapse
Affiliation(s)
- Anna Gluba-Brzozka
- Department of Nephrology, Hypertension and Family Medicine, WAM University Hospital, Lodz, Poland
- Healthy Aging Research Center, Medical University of Lodz, Lodz, Poland
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Lodz, Poland
| | - Peter P. Toth
- CGH Medical Center, Sterling, Illinois, and Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jacek Rysz
- Healthy Aging Research Center, Medical University of Lodz, Lodz, Poland
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Lodz, Poland
| | - Maciej Banach
- Healthy Aging Research Center, Medical University of Lodz, Lodz, Poland
- Department of Hypertension, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
13
|
Stanley FM, Linder KM, Cardozo TJ. Statins Increase Plasminogen Activator Inhibitor Type 1 Gene Transcription through a Pregnane X Receptor Regulated Element. PLoS One 2015; 10:e0138097. [PMID: 26379245 PMCID: PMC4574702 DOI: 10.1371/journal.pone.0138097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/25/2015] [Indexed: 12/15/2022] Open
Abstract
Plasminogen activator inhibitor type 1 (PAI-1) is a multifunctional protein that has important roles in inflammation and wound healing. Its aberrant regulation may contribute to many disease processes such as heart disease. The PAI-1 promoter is responsive to multiple inputs including cytokines, growth factors, steroids and oxidative stress. The statin drugs, atorvastatin, mevastatin and rosuvastatin, increased basal and stimulated expression of the PAI-1 promoter 3-fold. A statin-responsive, nuclear hormone response element was previously identified in the PAI-1 promoter, but it was incompletely characterized. We characterized this direct repeat (DR) of AGGTCA with a 3-nucleotide spacer at -269/-255 using deletion and directed mutagenesis. Deletion or mutation of this element increased basal transcription from the promoter suggesting that it repressed PAI-1 transcription in the unliganded state. The half-site spacing and the ligand specificity suggested that this might be a pregnane X receptor (PXR) responsive element. Computational molecular docking showed that atorvastatin, mevastatin and rosuvastatin were structurally compatible with the PXR ligand-binding pocket in its agonist conformation. Experiments with Gal4 DNA binding domain fusion proteins showed that Gal4-PXR was activated by statins while other DR + 3 binding nuclear receptor fusions were not. Overexpression of PXR further enhanced PAI-1 transcription in response to statins. Finally, ChIP experiments using Halo-tagged PXR and RXR demonstrated that both components of the PXR-RXR heterodimer bound to this region of the PAI-1 promoter.
Collapse
Affiliation(s)
- Frederick M. Stanley
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York, United States of America
| | - Kathryn M. Linder
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
| | - Timothy J. Cardozo
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York, United States of America
| |
Collapse
|
14
|
Suneja M, Fox DK, Fink BD, Herlein JA, Adams CM, Sivitz WI. Evidence for metabolic aberrations in asymptomatic persons with type 2 diabetes after initiation of simvastatin therapy. Transl Res 2015; 166:176-87. [PMID: 25683525 PMCID: PMC4509977 DOI: 10.1016/j.trsl.2015.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 01/03/2015] [Accepted: 01/20/2015] [Indexed: 01/14/2023]
Abstract
Hydroxymethylglutaryl coenzyme A reductase inhibitors (statins) prevent vascular events and are widely prescribed, particularly in persons with type 2 diabetes. However, intolerability because of myopathic symptoms often limits their use. We investigated the effects of simvastatin on parameters of mitochondrial function and muscle gene expression in 11 subjects with type 2 diabetes, none of whom had statin intolerance. After withdrawal of statins for 2 months, we obtained blood samples, performed vastus lateralis muscle biopsies, and assessed whole body resting energy expenditure (REE). We then reinitiated therapy using simvastatin, 20 mg/d, for 1 month before repeating these studies. As expected, simvastatin lowered low-density lipoprotein, but did not induce myalgias or significant increases in serum creatine kinase. However, we found subtle but significant reductions in muscle citrate synthase activity and REE. In addition, quantitative polymerase chain reaction and gene set enrichment analysis of muscle samples revealed significantly repressed gene sets involved in mitochondrial function and induced gene sets involved in remodeling of the extracellular matrix. Furthermore, the effects of simvastatin on muscle gene sets showed some similarities to previously described changes that occur in Duchenne muscular dystrophy, polymyositis, and dermatomyositis. Although statins inhibit an early step in coenzyme Q (CoQ) biosynthesis, we observed no differences in CoQ content within skeletal muscle mitochondria, muscle tissue, or circulating platelets. In summary, we report subtle changes in whole body energetics, mitochondrial citrate synthase activity, and microarray data consistent with subclinical myopathy. Although the benefits of statin therapy are clear, further understanding of muscular perturbations should help guide safety and tolerability.
Collapse
Affiliation(s)
- Manish Suneja
- Division of Nephrology, Department of Internal Medicine, University of Iowa and the Iowa City Veterans Affairs Health Care System, Iowa City VA, Iowa City, Iowa
| | - Daniel K Fox
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Brian D Fink
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa and the Iowa City Veterans Affairs Health Care System, Iowa City VA, Iowa City, Iowa
| | - Judy A Herlein
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa and the Iowa City Veterans Affairs Health Care System, Iowa City VA, Iowa City, Iowa
| | - Christopher M Adams
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa and the Iowa City Veterans Affairs Health Care System, Iowa City VA, Iowa City, Iowa
| | - William I Sivitz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa and the Iowa City Veterans Affairs Health Care System, Iowa City VA, Iowa City, Iowa.
| |
Collapse
|
15
|
Apostolopoulou M, Corsini A, Roden M. The role of mitochondria in statin-induced myopathy. Eur J Clin Invest 2015; 45:745-54. [PMID: 25991405 DOI: 10.1111/eci.12461] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 05/13/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Statins inhibit hydroxymethylglutaryl-coenzyme A reductase, decrease plasma low-density lipoprotein cholesterol and reduce cardiovascular morbidity and mortality. They can also exert adverse effects, mostly affecting skeletal muscle, ranging from mild myalgia to rhabdomyolysis. MATERIALS AND METHODS Based on a PubMed search until December 2014, this review summarizes studies on statin effects on muscle mitochondrial morphology and function in the context of myopathy. RESULTS Possible mechanisms of statin-induced myopathy include lower cholesterol synthesis and production of prenylated proteins, reduced dolichols and increased atrogin-1 expression. Statin-treated patients frequently feature decreased muscle coenzyme Q10 (CoQ10) contents, suggesting that statins might impair mitochondrial function. In cell cultures, statins diminish muscle oxygen consumption, promote mitochondrial permeability transient pore opening and generate apoptotic proteins. Animal models confirm the statin-induced decrease in muscle CoQ10, but reveal no changes in mitochondrial enzyme activities. Human studies yield contradictory results, with decreased CoQ10, elevated lipids, decreased enzyme activities in muscle and impaired maximal oxygen uptake in several but not all studies. Some patients are susceptible to statin-induced myopathy due to variations in genes encoding proteins involved in statin uptake and biotransformation such as the solute carrier organic anion transporter family member 1B1 (SLCO1B1) or cytochrome P450 (CYP2D6, CYP3A4, CYP3A5). Carriers for carnitine palmitoyltransferase II deficiency and McArdle disease also present with higher prevalence of statin-induced myopathy. CONCLUSIONS Despite the widespread use of statins, the pathogenesis of statin-induced myopathy remains unclear, requiring prospective randomized controlled trials with intensive phenotyping also for identifying strategies for its risk assessment, prevention and treatment.
Collapse
Affiliation(s)
- Maria Apostolopoulou
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| | - Alberto Corsini
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DISFeB), Università degli Studi di Milano, Milan, Italy.,IRCCS Multimedica, Milan, Italy
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany.,Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
16
|
Patnaik S, Patnaik AN. Management of dyslipidaemia in statin-intolerant patients in light of AHA and NPA guidelines of 2013–2014. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.jicc.2015.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
17
|
Feingold KR. Statin myopathy: a problem looking for a solution. Atherosclerosis 2015; 239:85-6. [PMID: 25576849 DOI: 10.1016/j.atherosclerosis.2014.12.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 12/30/2014] [Indexed: 10/24/2022]
Affiliation(s)
- Kenneth R Feingold
- Metabolism Section (111F), Department of Veterans Affairs Medical Center, University of California San Francisco, 4150 Clement Street, San Francisco, CA 94121, USA.
| |
Collapse
|
18
|
Ragbourne SC, Crook MA. Use of lipid-lowering medications in myasthenia gravis: a case report and literature review. J Clin Lipidol 2014; 9:256-9. [PMID: 25911083 DOI: 10.1016/j.jacl.2014.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 12/03/2014] [Accepted: 12/08/2014] [Indexed: 11/18/2022]
Abstract
We present a patient with myasthenia gravis (MG) who developed worsening of his condition after starting ezetimibe. We review the literature concerning lipid-modifying medications and MG. The use of bile acid sequestrant agents may have a place in the lipid management of MG patients because they did not seem to cause muscle-related side effects or worsening of MG.
Collapse
Affiliation(s)
- Sophie C Ragbourne
- Department of Clinical Biochemistry, Lewisham Hospital NHS Trust, London, UK; Guy's Hospital, Southwark, London, UK
| | - Martin A Crook
- Department of Clinical Biochemistry, Lewisham Hospital NHS Trust, London, UK; Department of Metabolic Medicine, University Hospital Lewishsam, London, UK; Guy's Hospital, Southwark, London, UK; St Thomas' Hospital, Lambeth, UK; University of Greenwich, London, UK.
| |
Collapse
|
19
|
Flowers N, Hartley L, Todkill D, Stranges S, Rees K, Cochrane Heart Group. Co-enzyme Q10 supplementation for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev 2014; 2014:CD010405. [PMID: 25474484 PMCID: PMC9759150 DOI: 10.1002/14651858.cd010405.pub2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cardiovascular disease (CVD) remains the number one cause of death and disability worldwide and public health interventions focus on modifiable risk factors, such as diet. Coenzyme Q10 (CoQ10) is an antioxidant that is naturally synthesised by the body and can also be taken as a dietary supplement. Studies have shown that a CoQ10 deficiency is associated with cardiovascular disease. OBJECTIVES To determine the effects of coenzyme Q10 supplementation as a single ingredient for the primary prevention of CVD. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL 2013, Issue 11); MEDLINE (Ovid, 1946 to November week 3 2013); EMBASE (Ovid, 1947 to 27 November 2013) and other relevant resources on 2 December 2013. We applied no language restrictions. SELECTION CRITERIA Randomised controlled trials (RCTs) lasting at least three months involving healthy adults or those at high risk of CVD but without a diagnosis of CVD. Trials investigated the supplementation of CoQ10 alone as a single supplement. The comparison group was no intervention or placebo. The outcomes of interest were CVD clinical events and major CVD risk factors, adverse effects and costs. We excluded any trials involving multifactorial lifestyle interventions to avoid confounding. DATA COLLECTION AND ANALYSIS Two authors independently selected trials for inclusion, abstracted data and assessed the risk of bias.We contacted authors for additional information where necessary. MAIN RESULTS We identified six RCTs with a total of 218 participants randomised, one trial awaiting classification and five ongoing trials. All trials were conducted in participants at high risk of CVD, two trials examined CoQ10 supplementation alone and four examined CoQ10 supplementation in patients on statin therapy; we analysed these separately. All six trials were small-scale, recruiting between 20 and 52 participants; one trial was at high risk of bias for incomplete outcome data and one for selective reporting; all studies were unclear in the method of allocation and therefore for selection bias. The dose of CoQ10 varied between 100 mg/day and 200 mg/day and the duration of the interventions was similar at around three months.No studies reported mortality or non-fatal cardiovascular events. None of the included studies provided data on adverse events.Two trials examined the effect of CoQ10 on blood pressure. For systolic blood pressure we did not perform a meta-analysis due to significant heterogeneity. In one trial CoQ10 supplementation had no effect on systolic blood pressure (mean difference (MD) -1.90 mmHg, 95% confidence interval (CI) -13.17 to 9.37, 51 patients randomised). In the other trial there was a statistically significant reduction in systolic blood pressure (MD -15.00 mmHg, 95% CI -19.06 to -10.94, 20 patients randomised). For diastolic blood pressure we performed a random-effects meta-analysis, which showed no evidence of effect of CoQ10 supplementation when these two small trials were pooled (MD -1.62 mmHg, 95% CI -5.2 to 1.96).One trial (51 patients randomised) looked at the effect of CoQ10 on lipid levels. The trial showed no evidence of effect of CoQ10 supplementation on total cholesterol (MD 0.30 mmol/L, 95% CI -0.10 to 0.70), high-density lipoprotein (HDL)-cholesterol (MD 0.02 mmol/L, 95% CI -0.13 to 0.17) or triglycerides (MD 0.05 mmol/L, 95% CI -0.42 to 0.52).Of the four trials that investigated CoQ10 supplementation in patients on statin therapy, three of them showed that simultaneous administration of CoQ10 did not significantly influence lipid levels or systolic blood pressure levels between the two groups. The fourth trial showed a significant increase in the change in total and low-density lipoprotein (LDL)-cholesterol at three months across the four arms of the trial (α-tocopherol, CoQ10, CoQ10 + α-tocopherol and placebo), however the way in which the data were presented meant that we were unable to determine if there was any significant difference between the CoQ10 only and placebo arms. In contrast, there was no significant difference in the change in HDL-cholesterol and triglycerides after three months between the four arms of the trial. AUTHORS' CONCLUSIONS There are very few studies to date examining CoQ10 for the primary prevention of CVD. The results from the ongoing studies will add to the evidence base. Due to the small number of underpowered trials contributing to the analyses, the results presented should be treated with caution and further high quality trials with longer-term follow-up are needed to determine the effects on cardiovascular events.
Collapse
Affiliation(s)
- Nadine Flowers
- Warwick Medical School, University of WarwickDivision of Health SciencesCoventryUKCV4 7AL
| | - Louise Hartley
- Warwick Medical School, University of WarwickDivision of Health SciencesCoventryUKCV4 7AL
| | - Daniel Todkill
- Warwick Medical School, University of WarwickDivision of Health SciencesCoventryUKCV4 7AL
| | - Saverio Stranges
- Warwick Medical School, University of WarwickDivision of Health SciencesCoventryUKCV4 7AL
| | - Karen Rees
- Warwick Medical School, University of WarwickDivision of Health SciencesCoventryUKCV4 7AL
| | | |
Collapse
|
20
|
Klug EQ, Raal FJ, Marais AD, Taskinen MR, Dalby AJ, Schamroth C, Rapeport N, Jankelow D, Blom DJ, Catsicas R, Webb DA. South African Dyslipidaemia Guideline Consensus Statement. S Afr Fam Pract (2004) 2014. [DOI: 10.1080/20786204.2013.10874296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
21
|
Norata GD, Tibolla G, Catapano AL. Statins and skeletal muscles toxicity: from clinical trials to everyday practice. Pharmacol Res 2014; 88:107-13. [PMID: 24835295 DOI: 10.1016/j.phrs.2014.04.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/23/2014] [Accepted: 04/27/2014] [Indexed: 12/26/2022]
Abstract
The mechanism(s) underlying the occurrence of statin-induced myopathy are ill defined, but the results of observational studies and clinical trials provide compelling evidence that skeletal muscle toxicity is a frequent, dose-dependent, adverse event associated with all statins. It has been suggested that reduced availability of metabolites produced by the mevalonate pathway rather than intracellular cholesterol lowering per se might be the primary trigger of toxicity, however other alternative explanations have gained credibility in recent years. Aim of this review is: (i) to describe the molecular mechanisms associated to statin induced myopathy including defects in isoprenoids synthesis followed by altered prenylation of small GTPase, such as Ras and Rab proteins; (ii) to present the emerging aspects on pharmacogenetics, including CYP3A4, OATP1B1 and glycine amidinotransferase (GATM) polymorphisms impacting either statin bioavailability or creatine synthesis; (iii) to summarize the available epidemiological evidences; and (iii) to discuss the concepts that would be of interest to the clinicians for the daily management of patients with statin induced myopathy. The interplay between drug-environment and drug-drug interaction in the context of different genetic settings contribute to statins and skeletal muscles toxicity. Until specific assays/algorithms able to combine genetic scores with drug-drug-environment interaction to identify patients at risk of myopathies will become available, clinicians should continue to monitor carefully patients on polytherapy which include statins and be ready to reconsider dose, statin or switching to alternative treatments. The beneficial effects of adding agents to provide the muscle with the metabolites, such as CoQ10, affected by statin treatment will also be addressed.
Collapse
Affiliation(s)
- Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy; Center for the Study of Atherosclerosis, Società Italiana Studio Aterosclerosi, Bassini Hospital, Cinisello Balsamo, Italy
| | - Gianpaolo Tibolla
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy; I.R.C.C.S. Multimedica, Milan, Italy
| | - Alberico Luigi Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy; I.R.C.C.S. Multimedica, Milan, Italy.
| |
Collapse
|
22
|
Littlefield N, Beckstrand RL, Luthy KE. Statins’ effect on plasma levels of Coenzyme Q10 and improvement in myopathy with supplementation. J Am Assoc Nurse Pract 2014; 26:85-90. [DOI: 10.1002/2327-6924.12046] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 06/14/2012] [Indexed: 11/09/2022]
|
23
|
Abstract
Statins are the revolutionary drugs in the cardiovascular pharmacotherapy. But they also possess several adverse effects like myopathy with elevation of hepatic transaminases (>3 times the upper limit of normal) or creatine kinase (>10 times the upper limit of normal) and some rare side-effects, including peripheral neuropathy, memory loss, sleep disturbances, and erectile dysfunction. Due to these adverse effects, patients abruptly withdrew statins without consulting physicians. This abrupt discontinuation of statins is termed as statin intolerance. Statin-induced myopathy constitutes two third of all side-effects from statins and is the primary reason for statin intolerance. Though statin intolerance has considerably impacted cardiovascular outcomes in the high-risk patients, it has been well effectively managed by prescribing statins either as alternate-day or once weekly dosage regimen, as combination therapy with a non-statin therapy or and by dietary intervention. The present article reviews the causes, clinical implications of statin withdrawal and management of statin intolerance.
Collapse
Affiliation(s)
- Soma B. Raju
- Care Hospital, The Institute of Medical Sciences, Hyderabad, India
| | - Kiron Varghese
- Department of Cardiology, St John's Medical College and Hospital, Bangalore, India
| | - K. Madhu
- Medical Affairs, AstraZeneca India, Bangalore, India
| |
Collapse
|
24
|
Barakat A, Shegokar R, Dittgen M, Müller RH. Coenzyme Q10 oral bioavailability: effect of formulation type. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2013. [DOI: 10.1007/s40005-013-0101-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
25
|
Affiliation(s)
- Rafael Bitzur
- Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel.
| | | | | | | |
Collapse
|
26
|
The mitochondrial unfolded protein response activator ATFS-1 protects cells from inhibition of the mevalonate pathway. Proc Natl Acad Sci U S A 2013; 110:5981-6. [PMID: 23530189 DOI: 10.1073/pnas.1218778110] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Statins are cholesterol-lowering drugs that inhibit 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase, the rate-limiting enzyme in the synthesis of cholesterol via the mevalonate pathway. This pathway also produces coenzyme Q (a component of the respiratory chain), dolichols (important for protein glycosylation), and isoprenoids (lipid moieties responsible for the membrane association of small GTPases). We previously showed that the nematode Caenorhabditis elegans is useful to study the noncholesterol effects of statins because its mevalonate pathway lacks the sterol synthesis branch but retains all other branches. Here, from a screen of 150,000 mutagenized genomes, we isolated four C. elegans mutants resistant to statins by virtue of gain-of-function mutations within the first six amino acids of the protein ATFS-1, the key regulator of the mitochondrial unfolded protein response that includes activation of the chaperones HSP-6 and HSP-60. The atfs-1 gain-of-function mutants are also resistant to ibandronate, an inhibitor of an enzyme downstream of HMG-CoA reductase, and to gliotoxin, an inhibitor acting on a subbranch of the pathway important for protein prenylation, and showed improved mitochondrial function and protein prenylation in the presence of statins. Additionally, preinduction of the mitochondrial unfolded protein response in wild-type worms using ethidium bromide or paraquat triggered statin resistance, and similar observations were made in Schizosaccharomyces pombe and in a mammalian cell line. We conclude that statin resistance through maintenance of mitochondrial homeostasis is conserved across species, and that the cell-lethal effects of statins are caused primarily through impaired protein prenylation that results in mitochondria dysfunction.
Collapse
|
27
|
Rallidis LS, Fountoulaki K, Anastasiou-Nana M. Managing the underestimated risk of statin-associated myopathy. Int J Cardiol 2012; 159:169-76. [DOI: 10.1016/j.ijcard.2011.07.048] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 07/07/2011] [Accepted: 07/10/2011] [Indexed: 12/20/2022]
|
28
|
Abstract
Statins are an effective treatment for the prevention of cardiovascular diseases and used extensively worldwide. However, myotoxicity induced by statins is a common adverse event and a major barrier to maximising cardiovascular risk reduction. The clinical spectrum of statin induced myotoxicity includes asymptomatic rise in creatine kinase concentration, myalgia, myositis and rhabdomyolysis. In certain cases, the cessation of statin therapy does not result in the resolution of muscular symptoms or the normalization of creatine kinase, raising the possibility of necrotizing autoimmune myopathy. There is increasing understanding and recognition of the pathophysiology and risk factors of statin induced myotoxicity. Careful history and physical examination in conjunction with selected investigations such as creatine kinase measurement, electromyography and muscle biopsy in appropriate clinical scenario help diagnose the condition. The management of statin induced myotoxicity involves statin cessation, the use of alternative lipid lowering agents or treatment regimes, and in the case of necrotizing autoimmune myopathy, immunosuppression.
Collapse
Affiliation(s)
- Sivakumar Sathasivam
- The Walton Centre NHS Foundation Trust, Lower Lane, Liverpool L9 7LJ, United Kingdom.
| |
Collapse
|
29
|
Abstract
Coenzyme Q10 has emerged as a valuable molecule for pharmaceutical and cosmetic applications. Therefore, research into producing and optimizing coenzyme Q10 via microbial fermentation is ongoing. There are two major paths being explored for maximizing production of this molecule to commercially advantageous levels. The first entails using microbes that naturally produce coenzyme Q10 as fermentation biocatalysts and optimizing the fermentation parameters in order to reach industrial levels of production. However, the natural coenzyme Q10-producing microbes tend to be intractable for industrial fermentation settings. The second path to coenzyme Q10 production being explored is to engineer Escherichia coli with the ability to biosynthesize this molecule in order to take advantage of its more favourable fermentation characteristics and the well-understood array of genetic tools available for this bacteria. Although many studies have attempted to over-produce coenzyme Q10 in E. coli through genetic engineering, production titres still remain below those of the natural coenzyme Q10-producing microorganisms. Current research is providing the knowledge needed to alleviate the bottlenecks involved in producing coenzyme Q10 from an E. coli strain platform and the fermentation parameters that could dramatically increase production titres from natural microbial producers. Synthesizing the lessons learned from both approaches may be the key towards a more cost-effective coenzyme Q10 industry.
Collapse
Affiliation(s)
- Corinne P Cluis
- Department of Biology, Concordia University, 7141 Sherbrooke West, Montréal, H4B 1R6, Québec, Canada
| | | | | |
Collapse
|
30
|
Kwak HB, Thalacker-Mercer A, Anderson EJ, Lin CT, Kane DA, Lee NS, Cortright RN, Bamman MM, Neufer PD. Simvastatin impairs ADP-stimulated respiration and increases mitochondrial oxidative stress in primary human skeletal myotubes. Free Radic Biol Med 2012; 52:198-207. [PMID: 22080086 PMCID: PMC3313473 DOI: 10.1016/j.freeradbiomed.2011.10.449] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 10/14/2011] [Accepted: 10/17/2011] [Indexed: 11/19/2022]
Abstract
Statins, the widely prescribed cholesterol-lowering drugs for the treatment of cardiovascular disease, cause adverse skeletal muscle side effects ranging from fatigue to fatal rhabdomyolysis. The purpose of this study was to determine the effects of simvastatin on mitochondrial respiration, oxidative stress, and cell death in differentiated primary human skeletal muscle cells (i.e., myotubes). Simvastatin induced a dose-dependent decrease in viability of proliferating and differentiating primary human muscle precursor cells, and a similar dose-dependent effect was noted in differentiated myoblasts and myotubes. Additionally, there were decreases in myotube number and size following 48 h of simvastatin treatment (5 μM). In permeabilized myotubes, maximal ADP-stimulated oxygen consumption, supported by palmitoylcarnitine+malate (PCM, complex I and II substrates) and glutamate+malate (GM, complex I substrates), was 32-37% lower (P<0.05) in simvastatin-treated (5 μM) vs control myotubes, providing evidence of impaired respiration at complex I. Mitochondrial superoxide and hydrogen peroxide generation were significantly greater in the simvastatin-treated human skeletal myotube cultures compared to control. In addition, simvastatin markedly increased protein levels of Bax (proapoptotic, +53%) and Bcl-2 (antiapoptotic, +100%, P<0.05), mitochondrial PTP opening (+44%, P<0.05), and TUNEL-positive nuclei in human skeletal myotubes, demonstrating up-regulation of mitochondrial-mediated myonuclear apoptotic mechanisms. These data demonstrate that simvastatin induces myotube atrophy and cell loss associated with impaired ADP-stimulated maximal mitochondrial respiratory capacity, mitochondrial oxidative stress, and apoptosis in primary human skeletal myotubes, suggesting that mitochondrial dysfunction may underlie human statin-induced myopathy.
Collapse
Affiliation(s)
- Hyo-Bum Kwak
- Department of Kinesiology, East Carolina University, Greenville, NC 27834
- Department of Physiology, East Carolina University, Greenville, NC 27834
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834
| | - Anna Thalacker-Mercer
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, AL 35294
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Ethan J. Anderson
- Department of Cardiovascular Sciences, East Carolina University, Greenville, NC 27834
- Department of Pharmacology, East Carolina University, Greenville, NC 27834
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834
| | - Chien-Te Lin
- Department of Kinesiology, East Carolina University, Greenville, NC 27834
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834
| | - Daniel A. Kane
- Department of Kinesiology, East Carolina University, Greenville, NC 27834
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834
| | - Nam-Sihk Lee
- Department of Internal Medicine, East Carolina University, Greenville, NC 27834
| | - Ronald N. Cortright
- Department of Kinesiology, East Carolina University, Greenville, NC 27834
- Department of Physiology, East Carolina University, Greenville, NC 27834
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834
| | - Marcas M. Bamman
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, AL 35294
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - P. Darrell Neufer
- Department of Kinesiology, East Carolina University, Greenville, NC 27834
- Department of Physiology, East Carolina University, Greenville, NC 27834
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834
- Corresponding Author: P. Darrell Neufer, PhD, Department of Physiology, Brody School of Medicine, 6N98, East Carolina University, Greenville, NC 27834, PH: (252) 744-2780, Fax: (252) 744-3460,
| |
Collapse
|
31
|
|
32
|
Blom DJ. Statins: adherence and side-effects. S Afr Fam Pract (2004) 2011. [DOI: 10.1080/20786204.2011.10874087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Affiliation(s)
- DJ Blom
- Division of Lipidology, Department of Medicine, University of Cape Town
| |
Collapse
|
33
|
Fung EC, Crook MA. Statin myopathy: a lipid clinic experience on the tolerability of statin rechallenge. Cardiovasc Ther 2011; 30:e212-8. [PMID: 21884002 DOI: 10.1111/j.1755-5922.2011.00267.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION Statin myopathy is a generally encountered side effect of statin usage. Both muscle symptoms and a raised serum creatine kinase (CK) are used in case definition, but these are common manifestations of other conditions, which may not be statin related. Statin rechallenge assuming no contraindication in selected cases is an option before considering a different class of lipid-lowering agent. AIMS We aim to characterize retrospectively the patients referred to our Lipid Clinic with a diagnosis of statin myopathy. The tolerability of different statins was assessed to determine a strategy for rechallenging statins in such patients in the future. RESULTS Patients with statin myopathy constitute 10.2% of our Lipid Clinic workload. They are predominantly female (62.0%), Caucasian (63.9%), with a mean age of 58.3 years and mean body mass index (BMI) of 29.3 kg/m(2). The serum CK and erythrocyte sedimentation rate (ESR) were statistically higher compared to patients with statin intolerances with no muscular component or CK elevations. Secondary causes of statin myopathy were implicated in 2.7% of cases. Following statin myopathy to simvastatin we found no statistical difference between the tolerability rates between atorvastatin, rosuvastatin, pravastatin, and fluvastatin. Fibrates, cholestyramine, and ezetimibe were statistically better tolerated in these patients. CONCLUSIONS Statin rechallenge is a real treatment option in patients with statin myopathy. Detailed history and examination is required to exclude muscle diseases unrelated to statin usage. In patients developing statin myopathy on simvastatin, we did not find any statistical difference between subsequent tolerability rates to rosuvastatin, pravastatin, and fluvastatin.
Collapse
Affiliation(s)
- En C Fung
- Department of Clinical Biochemistry and Metabolic Medicine, University Hospital Lewisham, London, UK.
| | | |
Collapse
|
34
|
Bank G, Kagan D, Madhavi D. Coenzyme Q10: Clinical Update and Bioavailability. J Evid Based Complementary Altern Med 2011. [DOI: 10.1177/2156587211399438] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Coenzyme Q10 (CoQ 10) supplementation has been reported to be beneficial in treating a variety of health conditions and diseases, with more than 200 clinical trials investigating its use as a drug or dietary supplement. Numerous reviews of the safety and clinical potential of CoQ10 have been published. Successful treatment and efficacy is dependent on the bioavailability of CoQ 10, which is well known to be poor because of its lipophilic nature and large molecular weight. A number of recent clinical trials on CoQ 10 have investigated new formulations of CoQ10 for improvements in absorption and bioavailability. This review provides an update of clinical efficacy trials using CoQ10 and describes recent advances in formulation technology to improve the bioavailability of CoQ10. The authors also discuss a new method to improve the standards of reporting the bioavailability results of such advanced CoQ10 formulations to help clinicians and consumers make informed decisions.
Collapse
Affiliation(s)
- Ginny Bank
- Full Spectrum Consulting, Boulder, CO, USA
| | | | | |
Collapse
|
35
|
Abstract
Statins lower cholesterol by inhibiting 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, the rate-limiting enzyme in the biosynthesis of cholesterol. However, severe adverse events, including myalgias and rhabdomyolysis, have been reported with statin treatment. Different mechanisms have been proposed to explain statin-induced myopathy, including reduction of mevalonate pathway products, induction of apoptosis, mitochondrial dysfunction, and genetic predisposition. A decrease in coenzyme Q(10) (CoQ), a product of the mevalonate pathway, could contribute to statin induced myopathy. This article reviews the clinical and biochemical features of statin-induced myopathy, the inter-relationship between statins and the concentration of CoQ in plasma and tissues, and whether there is a role for supplementation with CoQ to attenuate statin-induced myopathy.
Collapse
Affiliation(s)
- Emilie Mas
- School of Medicine and Pharmacology, Royal Perth Hospital Unit, University of Western Australia, Medical Research Foundation Building Level 4, Rear 50, Murray Street, Perth, WA, 6847, Australia.
| | | |
Collapse
|
36
|
Acimovic J, Korosec T, Seliskar M, Bjorkhem I, Monostory K, Szabo P, Pascussi JM, Belic A, Urleb U, Kocjan D, Rozman D. Inhibition of human sterol Δ7-reductase and other postlanosterol enzymes by LK-980, a novel inhibitor of cholesterol synthesis. Drug Metab Dispos 2011; 39:39-46. [PMID: 20952551 DOI: 10.1124/dmd.110.035840] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Novel potential inhibitors of the postsqualene portion of cholesterol synthesis were screened in HepG2 cells. 2-(4-Phenethylpiperazin-1-yl)-1-(pyridine-3-yl)ethanol (LK-980) was identified as a prospective compound and was characterized further in cultures of human primary hepatocytes from seven donors. In vitro kinetic measurements show that the half-life of LK-980 is at least 4.3 h. LK-980 does not induce CYP3A4 mRNA nor enzyme activity. Target prediction was performed by gas chromatography-mass spectrometry, allowing simultaneous separation and quantification of nine late cholesterol intermediates. Experiments indicated that human sterol Δ(7)-reductase (DHCR7) is the major target of LK-980 (34-fold increase of 7-dehydrocholesterol), whereas human sterol Δ(14)-reductase (DHCR14), human sterol Δ(24)-reductase (DHCR24), and human sterol C5-desaturase (SC5DL) represent minor targets. In the absence of purified enzymes, we used the mathematical model of cholesterol synthesis to evaluate whether indeed more than a single enzyme is inhibited. In silico inhibition of only DHCR7 modifies the flux of cholesterol intermediates, resulting in a sterol profile that does not support experimental data. Partial inhibition of the DHCR14, DHCR24, and SC5DL steps, in addition to DHCR7, supports the experimental sterol profile. In conclusion, we provide experimental and computational evidence that LK-980, a novel inhibitor from the late portion of cholesterol synthesis, inhibits primarily DHCR7 and to a lesser extent three other enzymes from this pathway.
Collapse
Affiliation(s)
- Jure Acimovic
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Gupta A, Thompson PD. The relationship of vitamin D deficiency to statin myopathy. Atherosclerosis 2010; 215:23-9. [PMID: 21185021 DOI: 10.1016/j.atherosclerosis.2010.11.039] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 11/24/2010] [Accepted: 11/25/2010] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Our goal was to examine the interaction between vitamin D and statins and the possible role of vitamin D deficiency in statin myopathy. BACKGROUND The vitamin D receptor is present in skeletal muscle and vitamin D deficiency can cause myopathy. Statins (3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitors) are generally well tolerated, but have been associated with a spectrum of skeletal muscle complaints, ranging from myalgia and asymptomatic mild elevations of creatine kinase (CK) to rhabdomyolysis. There has been recent interest in the possible interaction between statin myopathy and vitamin D deficiency. We performed a systematic medical literature review to examine this possible relationship. METHODS We identified English language articles relating statins, vitamin D and statin myopathy via a PubMed search through July 2010. Articles pertinent to the topic were reviewed in detail. RESULTS/CONCLUSIONS Our review suggests that some but not all statins increase 25(OH) D levels. Two cross sectional studies have associated vitamin D deficiency with statin-associated myalgias, and suggested that that increasing vitamin D levels can reverse the myalgia. Nevertheless, given the quality and paucity of studies examining this possibility, additional studies are needed to examine the potential role of vitamin D deficiency in statin myopathy. It is presently premature to recommend vitamin D supplementation as treatment for statin associated muscle complaints in the absence of low vitamin D levels although such supplementation could be tried in patients with deficient or reduced vitamin D levels.
Collapse
Affiliation(s)
- Ankur Gupta
- Cardiology, Hartford Hospital, 80 Seymour Street, Hartford, CT 06102, United States
| | | |
Collapse
|
38
|
McMurray JJV, Dunselman P, Wedel H, Cleland JGF, Lindberg M, Hjalmarson A, Kjekshus J, Waagstein F, Apetrei E, Barrios V, Böhm M, Kamenský G, Komajda M, Mareev V, Wikstrand J. Coenzyme Q10, rosuvastatin, and clinical outcomes in heart failure: a pre-specified substudy of CORONA (controlled rosuvastatin multinational study in heart failure). J Am Coll Cardiol 2010; 56:1196-204. [PMID: 20883926 DOI: 10.1016/j.jacc.2010.02.075] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2009] [Revised: 01/26/2010] [Accepted: 02/15/2010] [Indexed: 11/20/2022]
Abstract
OBJECTIVES The purpose of this study was to determine whether coenzyme Q₁₀ is an independent predictor of prognosis in heart failure. BACKGROUND Blood and tissue concentrations of the essential cofactor coenzyme Q₁₀ are decreased by statins, and this could be harmful in patients with heart failure. METHODS We measured serum coenzyme Q₁₀ in 1,191 patients with ischemic systolic heart failure enrolled in CORONA (Controlled Rosuvastatin Multinational Study in Heart Failure) and related this to clinical outcomes. RESULTS Patients with lower coenzyme Q₁₀ concentrations were older and had more advanced heart failure. Mortality was significantly higher among patients in the lowest compared to the highest coenzyme Q₁₀ tertile in a univariate analysis (hazard ratio: 1.50, 95% confidence interval: 1.04 to 2.6, p = 0.03) but not in a multivariable analysis. Coenzyme Q₁₀ was not an independent predictor of any other clinical outcome. Rosuvastatin reduced coenzyme Q₁₀ but there was no interaction between coenzyme Q₁₀ and the effect of rosuvastatin. CONCLUSIONS Coenzyme Q₁₀ is not an independent prognostic variable in heart failure. Rosuvastatin reduced coenzyme Q₁₀, but even in patients with a low baseline coenzyme Q₁₀, rosuvastatin treatment was not associated with a significantly worse outcome. (Controlled Rosuvastatin Multinational Study in Heart Failure [CORONA]; NCT00206310).
Collapse
Affiliation(s)
- John J V McMurray
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Abstract
Myopathy occurs in approximately 10% of statin-treated patients and is most commonly manifested by myalgias with or without plasma creatine kinase (CK) elevations. Predisposition exists in patients treated with high doses of potent statins and those who are older, female, have a genetic predisposition, and when statins are coadministered with drugs that compete with or inhibit drug metabolism. In symptomatic patients, CK levels may assist in guiding management. If less than five times the upper limit of normal, the existing statin should be titrated to achieve cholesterol goals and the CK repeated when symptoms appear or worsen. In patients with moderate to severe symptoms and any patient with CK elevated to more than 5-fold the upper limit of normal, the statin should be stopped. Once asymptomatic and CK is reduced (if elevated previously), cholesterol goals can be approached by: 1) a different statin (e.g. fluvastatin or pravastatin), starting with a low dose and titrating up; 2) an alternate daily or weekly more potent statin (e.g. rosuvastatin or atorvastatin); or 3) the combination of the lowest tolerated statin with a cholesterol absorption inhibitor (ezetimibe) and/or bile acid sequestrant. Over-the-counter preparations, e.g. red yeast rice, containing natural statin-like agents, or plant sterols can also lower cholesterol. These, however, have limited efficacy to achieve targeted cholesterol levels for most patients. In patients without CK elevations and symptoms, progress can be followed clinically, but in patients who show CK elevations, CK should be monitored. At present, the superiority of one approach has not been demonstrated, and the need for clinical trials in well-characterized patients with statin intolerance cannot be dismissed.
Collapse
Affiliation(s)
- Robert H Eckel
- University of Colorado Denver, Anschutz Medical Campus, Mail Stop 8106, 12801 East 17th Avenue, Aurora, Colorado 80045, USA.
| |
Collapse
|
41
|
Shinozaki S, Inoue Y, Yang W, Fukaya M, Carter EA, Yu YM, Ming-Yu Y, Fischman A, Tompkins R, Kaneki M. Farnesyltransferase inhibitor improved survival following endotoxin challenge in mice. Biochem Biophys Res Commun 2009; 391:1459-64. [PMID: 20034462 DOI: 10.1016/j.bbrc.2009.12.094] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 12/16/2009] [Indexed: 01/09/2023]
Abstract
Endotoxemia plays an important role in the pathogenesis of sepsis and is accompanied by dysregulated apoptosis of immune and non-immune cells. Treatment with statins reduces mortality in rodent models of sepsis and endotoxemia. Inhibition of protein isoprenylation, including farnesylation, has been proposed as a mechanism to mediate the lipid-lowering-independent effects of statins. Nonetheless, the effects of the inhibition of isoprenylation have not yet been studied. To investigate the role of farnesylation, we evaluated the effects of farnesyltransferase inhibitor and statin on survival following lipopolysaccharide (LPS) challenge in mice. Both simvastatin (2mg/kg BW) and FTI-277 (20mg/kg BW) treatment improved survival by twofold after LPS injection, as compared with vehicle alone (p<0.01). LPS-induced cleavage (activation) of caspase-3, an indicator of apoptotic change, and increased protein expression of proapoptotic molecules, Bax and Bim, and activation of c-Jun NH(2)-terminal kinase (JNK/SAPK) in the liver and spleen were attenuated by both simvastatin and FTI-277. These results demonstrate that farnesyltransferase inhibitor as well as statin significantly reduced LPS-induced mortality in mice. Our findings also suggest that inhibition of protein farnesylation may contribute to the lipid-lowering-independent protective effects of statins in endotoxemia, and that protein farnesylation may play a role in LPS-induced stress response, including JNK/SAPK activation, and apoptotic change. Our data argue that farnesyltransferase may be a potential molecular target for treating patients with endotoxemia.
Collapse
|
42
|
|
43
|
|
44
|
Achieving optimum lipid-lowering goals in patients with vascular disease. CURRENT CARDIOVASCULAR RISK REPORTS 2009. [DOI: 10.1007/s12170-009-0048-1] [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]
|
45
|
Abstract
The use of statins is widespread and many patients presenting for surgery are regularly taking them. There is evidence that statins have beneficial effects beyond those of lipid lowering, including reducing the perioperative risk of cardiac complications and sepsis. This review addresses the cellular mechanisms by which statins may produce these effects. Statins appear to have actions on vascular nitric oxide through the balance of inducible and endothelial nitric oxide synthase. The clinical evidence for these benefits is also briefly reviewed with the objective of clarifying the current status of statin use in the perioperative period. There is reasonably strong evidence that patients already taking statins should continue on them perioperatively. However, the evidence for the prophylactic use of statins perioperatively is weak and lacks prospective controlled studies.
Collapse
Affiliation(s)
- Z L S Brookes
- Microcirculation Research Group, School of Medicine and Biomedical Sciences, Royal Hallamshire Hospital, University of Sheffield, K Floor, Beech Hill Road, Sheffield, UK
| | | | | |
Collapse
|