1
|
Liepinsh E, Zvejniece L, Clemensson L, Ozola M, Vavers E, Cirule H, Korzh S, Skuja S, Groma V, Briviba M, Grinberga S, Liu W, Olszewski P, Gentreau M, Fredriksson R, Dambrova M, Schiöth HB. Hydroxymethylglutaryl-CoA reductase activity is essential for mitochondrial β-oxidation of fatty acids to prevent lethal accumulation of long-chain acylcarnitines in the mouse liver. Br J Pharmacol 2024; 181:2750-2773. [PMID: 38641905 DOI: 10.1111/bph.16363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/16/2024] [Accepted: 01/30/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Statins are competitive inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (HMGCR), and exert adverse effects on mitochondrial function, although the mechanisms underlying these effects remain unclear. We used a tamoxifen-induced Hmgcr-knockout (KO) mouse model, a multi-omics approach and mitochondrial function assessments to investigate whether decreased HMGCR activity impacts key liver energy metabolism pathways. EXPERIMENTAL APPROACH We established a new mouse strain using the Cre/loxP system, which enabled whole-body deletion of Hmgcr expression. These mice were crossed with Rosa26Cre mice and treated with tamoxifen to delete Hmgcr in all cells. We performed transcriptomic and metabolomic analyses and thus evaluated time-dependent changes in metabolic functions to identify the pathways leading to cell death in Hmgcr-KO mice. KEY RESULTS Lack of Hmgcr expression resulted in lethality, due to acute liver damage caused by rapid disruption of mitochondrial fatty acid β-oxidation and very high accumulation of long-chain (LC) acylcarnitines in both male and female mice. Gene expression and KO-related phenotype changes were not observed in other tissues. The progression to liver failure was driven by diminished peroxisome formation, which resulted in impaired mitochondrial and peroxisomal fatty acid metabolism, enhanced glucose utilization and whole-body hypoglycaemia. CONCLUSION AND IMPLICATIONS Our findings suggest that HMGCR is crucial for maintaining energy metabolism balance, and its activity is necessary for functional mitochondrial β-oxidation. Moreover, statin-induced adverse reactions might be rescued by the prevention of LC acylcarnitine accumulation.
Collapse
Affiliation(s)
- Edgars Liepinsh
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | | | | | - Melita Ozola
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | - Edijs Vavers
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Helena Cirule
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | | | | | | | - Monta Briviba
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | - Wen Liu
- Uppsala University, Uppsala, Sweden
| | | | | | | | - Maija Dambrova
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | | |
Collapse
|
2
|
Al-Sabri MH, Ammar N, Korzh S, Alsehli AM, Hosseini K, Fredriksson R, Mwinyi J, Williams MJ, Boukhatmi H, Schiöth HB. Fluvastatin-induced myofibrillar damage is associated with elevated ROS, and impaired fatty acid oxidation, and is preceded by mitochondrial morphological changes. Sci Rep 2024; 14:3338. [PMID: 38336990 PMCID: PMC10858229 DOI: 10.1038/s41598-024-53446-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Previously, we showed that fluvastatin treatment induces myofibrillar damage and mitochondrial phenotypes in the skeletal muscles of Drosophila. However, the sequential occurrence of mitochondrial phenotypes and myofibril damage remains elusive. To address this, we treated flies with fluvastatin for two and five days and examined their thorax flight muscles using confocal microscopy. In the two-day fluvastatin group, compared to the control, thorax flight muscles exhibited mitochondrial morphological changes, including fragmentation, rounding up and reduced content, while myofibrils remained organized in parallel. In the five-day fluvastatin treatment, not only did mitochondrial morphological changes become more pronounced, but myofibrils became severely disorganized with significantly increased thickness and spacing, along with myofilament abnormalities, suggesting myofibril damage. These findings suggest that fluvastatin-induced mitochondrial changes precede myofibril damage. Moreover, in the five-day fluvastatin group, the mitochondria demonstrated elevated H2O2 and impaired fatty acid oxidation compared to the control group, indicating potential mitochondrial dysfunction. Surprisingly, knocking down Hmgcr (Drosophila homolog of HMGCR) showed normal mitochondrial respiration in all parameters compared to controls or five-day fluvastatin treatment, which suggests that fluvastatin-induced mitochondrial dysfunction might be independent of Hmgcr inhibition. These results provide insights into the sequential occurrence of mitochondria and myofibril damage in statin-induced myopathy for future studies.
Collapse
Affiliation(s)
- Mohamed H Al-Sabri
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24, Uppsala, Sweden.
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24, Uppsala, Sweden.
| | - Nourhane Ammar
- Institut de Génétique Et Développement de Rennes (IGDR), Université de Rennes, CNRS, UMR6290, 35065, Rennes, France
| | - Stanislava Korzh
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, 1006, Latvia
| | - Ahmed M Alsehli
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24, Uppsala, Sweden
- Faculty of Medicine, King Abdulaziz University and Hospital, Al Ehtifalat St., 21589, Jeddah, Saudi Arabia
| | - Kimia Hosseini
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24, Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24, Uppsala, Sweden
| | - Jessica Mwinyi
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24, Uppsala, Sweden
| | - Michael J Williams
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24, Uppsala, Sweden
| | - Hadi Boukhatmi
- Institut de Génétique Et Développement de Rennes (IGDR), Université de Rennes, CNRS, UMR6290, 35065, Rennes, France
| | - Helgi B Schiöth
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24, Uppsala, Sweden.
| |
Collapse
|
3
|
Gander J, Carrard J, Gallart-Ayala H, Borreggine R, Teav T, Infanger D, Colledge F, Streese L, Wagner J, Klenk C, Nève G, Knaier R, Hanssen H, Schmidt-Trucksäss A, Ivanisevic J. Metabolic Impairment in Coronary Artery Disease: Elevated Serum Acylcarnitines Under the Spotlights. Front Cardiovasc Med 2021; 8:792350. [PMID: 34977199 PMCID: PMC8716394 DOI: 10.3389/fcvm.2021.792350] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/09/2021] [Indexed: 12/26/2022] Open
Abstract
Coronary artery disease (CAD) remains the leading cause of death worldwide. Expanding patients' metabolic phenotyping beyond clinical chemistry investigations could lead to earlier recognition of disease onset and better prevention strategies. Additionally, metabolic phenotyping, at the molecular species level, contributes to unravel the roles of metabolites in disease development. In this cross-sectional study, we investigated clinically healthy individuals (n = 116, 65% male, 70.8 ± 8.7 years) and patients with CAD (n = 54, 91% male, 67.0 ± 11.5 years) of the COmPLETE study. We applied a high-coverage quantitative liquid chromatography-mass spectrometry approach to acquire a comprehensive profile of serum acylcarnitines, free carnitine and branched-chain amino acids (BCAAs), as markers of mitochondrial health and energy homeostasis. Multivariable linear regression analyses, adjusted for confounders, were conducted to assess associations between metabolites and CAD phenotype. In total, 20 short-, medium- and long-chain acylcarnitine species, along with L-carnitine, valine and isoleucine were found to be significantly (adjusted p ≤ 0.05) and positively associated with CAD. For 17 acylcarnitine species, associations became stronger as the number of affected coronary arteries increased. This implies that circulating acylcarnitine levels reflect CAD severity and might play a role in future patients' stratification strategies. Altogether, CAD is characterized by elevated serum acylcarnitine and BCAA levels, which indicates mitochondrial imbalance between fatty acid and glucose oxidation.
Collapse
Affiliation(s)
- Joséphine Gander
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Justin Carrard
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Rébecca Borreggine
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Tony Teav
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Denis Infanger
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Flora Colledge
- Division of Sports Science, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Lukas Streese
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Jonathan Wagner
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Christopher Klenk
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Gilles Nève
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Raphael Knaier
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Henner Hanssen
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Arno Schmidt-Trucksäss
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
- Arno Schmidt-Trucksäss
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- *Correspondence: Julijana Ivanisevic
| |
Collapse
|
4
|
Mollazadeh H, Tavana E, Fanni G, Bo S, Banach M, Pirro M, von Haehling S, Jamialahmadi T, Sahebkar A. Effects of statins on mitochondrial pathways. J Cachexia Sarcopenia Muscle 2021; 12:237-251. [PMID: 33511728 PMCID: PMC8061391 DOI: 10.1002/jcsm.12654] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/09/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Statins are a family of drugs that are used for treating hyperlipidaemia with a recognized capacity to prevent cardiovascular disease events. They inhibit β-hydroxy β-methylglutaryl-coenzyme A reductase, i.e. the rate-limiting enzyme in mevalonate pathway, reduce endogenous cholesterol synthesis, and increase low-density lipoprotein clearance by promoting low-density lipoprotein receptor expression mainly in the hepatocytes. Statins have pleiotropic effects including stabilization of atherosclerotic plaques, immunomodulation, anti-inflammatory properties, improvement of endothelial function, antioxidant, and anti-thrombotic action. Despite all beneficial effects, statins may elicit adverse reactions such as myopathy. Studies have shown that mitochondria play an important role in statin-induced myopathies. In this review, we aim to report the mechanisms of action of statins on mitochondrial function. Results have shown that statins have several effects on mitochondria including reduction of coenzyme Q10 level, inhibition of respiratory chain complexes, induction of mitochondrial apoptosis, dysregulation of Ca2+ metabolism, and carnitine palmitoyltransferase-2 expression. The use of statins has been associated with the onset of additional pathological conditions like diabetes and dementia as a result of interference with mitochondrial pathways by various mechanisms, such as reduction in mitochondrial oxidative phosphorylation, increase in oxidative stress, decrease in uncoupling protein 3 concentration, and interference in amyloid-β metabolism. Overall, data reported in this review suggest that statins may have major effects on mitochondrial function, and some of their adverse effects might be mediated through mitochondrial pathways.
Collapse
Affiliation(s)
- Hamid Mollazadeh
- Department of Physiology and Pharmacology, Faculty of MedicineNorth Khorasan University of Medical SciencesBojnurdIran
- Natural Products and Medicinal Plants Research CenterNorth Khorasan University of Medical SciencesBojnurdIran
| | - Erfan Tavana
- Student Research Committee, School of MedicineNorth Khorasan University of Medical SciencesBojnurdIran
| | - Giovanni Fanni
- Department of Medical SciencesUniversity of TurinTurinItaly
| | - Simona Bo
- Department of Medical Sciences, AOU Città della Salute e della Scienza di TorinoUniversity of TurinTurinItaly
| | - Maciej Banach
- Department of HypertensionWAM University Hospital in LodzMedical University of Lodz, LodzPoland
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), LodzPoland
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of MedicineUniversity of PerugiaPerugiaItaly
| | - Stephan von Haehling
- Department of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
- German Center for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Tannaz Jamialahmadi
- Department of Food Science and TechnologyIslamic Azad UniversityQuchanQuchanIran
- Department of Nutrition, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Neurogenic Inflammation Research CenterMashhad University of Medical SciencesMashhadIran
- Halal Research Center of IRIFDATehranIran
| |
Collapse
|
5
|
Rosuvastatin protects isolated hearts against ischemia-reperfusion injury: role of Akt-GSK-3β, metabolic environment, and mitochondrial permeability transition pore. J Physiol Biochem 2020; 76:85-98. [PMID: 31916218 DOI: 10.1007/s13105-019-00718-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023]
Abstract
The cardioprotective activity of rosuvastatin (R) is yet to be known. The objective of this study was to research whether R perfusion before global ischemia can mitigate myocardial ischemia-reperfusion damage, considering the metabolic condition in which these effects occur, and to contemplate potential mitochondrial benefits. Protein kinase B (Akt)/glycogen synthase kinase-3β (GSK-3β) and mitochondrial permeability transition pore (MPTP) are key elements in myocardial injury produced by ischemia-reperfusion. Isolated rat hearts were subjected to 25-min ischemia and 1-h reperfusion in the presence or absence of R, with or without Wortmannin (W), a phosphatidylinositol 3-kinase (PI3K)/Akt inhibitor. Akt and GSK-3β were measured by Western blot analysis; lactate, glycogen, and G6PDH were determined; and Ca2+-induced MPTP opening was evaluated using a spectrophotometric method. Contractility was assessed by left ventricular developed pressure (LVDP), and rate-pressure product (RPP), peak rate of contraction and peak rate of relaxation (± dP/dt), and left ventricular end-diastolic pressure (LVEDP) were determined. Tissue samples were extracted to evaluate mitochondrial damage by electron microscopy and to assess infarct size. Statistical analysis employed ANOVA (n = 6/per group). Myocardial infarct size was significantly reduced by R, which also improved cardiac function. MPTP opening was delayed to 300 μM CaCl2, while use of W resulted in MPTP opening at 200 μM CaCl2. Electron microscopy showed better mitochondrial preservation with R, which reduced lactic acid production, increased glycogen consumption and G6PDH activity, as well as phosphorylation of Akt and GSK-3β. R before ischemia is cardioprotective against ischemic and reperfusion damage, activating Akt and regulating GSK-3β negatively and attenuating the MPTP opening.
Collapse
|
6
|
Ruiz M, Labarthe F, Fortier A, Bouchard B, Thompson Legault J, Bolduc V, Rigal O, Chen J, Ducharme A, Crawford PA, Tardif JC, Des Rosiers C. Circulating acylcarnitine profile in human heart failure: a surrogate of fatty acid metabolic dysregulation in mitochondria and beyond. Am J Physiol Heart Circ Physiol 2017; 313:H768-H781. [PMID: 28710072 DOI: 10.1152/ajpheart.00820.2016] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 07/07/2017] [Accepted: 07/07/2017] [Indexed: 12/19/2022]
Abstract
Heart failure (HF) is associated with metabolic perturbations, particularly of fatty acids (FAs), which remain to be better understood in humans. This study aimed at testing the hypothesis that HF patients with reduced ejection fraction display systemic perturbations in levels of energy-related metabolites, especially those reflecting dysregulation of FA metabolism, namely, acylcarnitines (ACs). Circulating metabolites were assessed using mass spectrometry (MS)-based methods in two cohorts. The main cohort consisted of 72 control subjects and 68 HF patients exhibiting depressed left ventricular ejection fraction (25.9 ± 6.9%) and mostly of ischemic etiology with ≥2 comorbidities. HF patients displayed marginal changes in plasma levels of tricarboxylic acid cycle-related metabolites or indexes of mitochondrial or cytosolic redox status. They had, however, 22-79% higher circulating ACs, irrespective of chain length (P < 0.0001, adjusted for sex, age, renal function, and insulin resistance, determined by shotgun MS/MS), which reflects defective mitochondrial β-oxidation, and were significantly associated with levels of NH2-terminal pro-B-type natriuretic peptide levels, a disease severity marker. Subsequent extended liquid chromatography-tandem MS analysis of 53 plasma ACs in a subset group from the primary cohort confirmed and further substantiated with a comprehensive lipidomic analysis in a validation cohort revealed in HF patients a more complex circulating AC profile. The latter included dicarboxylic-ACs and dihydroxy-ACs as well as very long chain (VLC) ACs or sphingolipids with VLCFAs (>20 carbons), which are proxies of dysregulated FA metabolism in peroxisomes. Our study identified alterations in circulating ACs in HF patients that are independent of biological traits and associated with disease severity markers. These alterations reflect dysfunctional FA metabolism in mitochondria but also beyond, namely, in peroxisomes, suggesting a novel mechanism contributing to global lipid perturbations in human HF.NEW & NOTEWORTHY Mass spectrometry-based profiling of circulating energy metabolites, including acylcarnitines, in two cohorts of heart failure versus control subjects revealed multiple alterations in fatty acid metabolism in peroxisomes in addition to mitochondria, thereby highlighting a novel mechanism contributing to global lipid perturbations in heart failure.Listen to this article's corresponding podcast at http://ajpheart.podbean.com/e/acylcarnitines-in-human-heart-failure/.
Collapse
Affiliation(s)
- Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - François Labarthe
- CHRU de Tours, Université François Rabelais, Institut National de la Santé et de la Recherche Médicale U1069, Nutrition, Croissance et Cancer, Tours, France
| | - Annik Fortier
- Montreal Health Innovations Coordinating Center, Montreal, Quebec, Canada
| | - Bertrand Bouchard
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Julie Thompson Legault
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Virginie Bolduc
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Odile Rigal
- Laboratoire de Biochimie, Hôpital R. Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jane Chen
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Anique Ducharme
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Peter A Crawford
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | | | - Christine Des Rosiers
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; .,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Khelfi A, Azzouz M, Abtroun R, Reggabi M, Alamir B. [Direct mechanism of action in toxic myopathies]. ANNALES PHARMACEUTIQUES FRANÇAISES 2017; 75:323-343. [PMID: 28526123 DOI: 10.1016/j.pharma.2017.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/02/2017] [Accepted: 04/03/2017] [Indexed: 01/04/2023]
Abstract
Toxic myopathies are a large group of disorders generated by surrounding agents and characterized by structural and/or functional disturbances of muscles. The most recurrent are those induced by commonly used medications. Illicit drugs, environmental toxins from animals, vegetables, or produced by micro-organisms as well as chemical products commonly used are significant causes of such disorders. The muscle toxicity results from multiple mechanisms at different biological levels. Many agents can induce myotoxicity through a direct mechanism in which statins, glucocorticoids and ethyl alcohol are the most representative. Diverse mechanisms were highlighted as interaction with macromolecules and induction of metabolic and cellular dysfunctions. Muscle damage can be related to amphiphilic properties of some drugs (chloroquine, hydroxychloroquine, etc.) leading to specific lysosomal disruptions and autophagic dysfunctions. Some agents affect the whole muscle fiber by inducing oxidative stress (ethyl alcohol and some statins) or triggering cell death pathways (apoptosis or necrosis) resulting in extensive alterations. More studies on these mechanisms are needed. They would allow a better knowledge of the intracellular mediators involved in these pathologies in order to develop targeted therapies of high efficiency.
Collapse
Affiliation(s)
- A Khelfi
- Service de toxicologie, CHU Bab-El-Oued, rue Mohamed-Lamine-Debaghine, 16009 Alger, Algérie; Centre national de toxicologie, route du Petit-Staouali-Delly-Brahim, 16062 Alger, Algérie.
| | - M Azzouz
- Laboratoire central de biologie et de toxicologie, EHS Ait-Idir, rue Abderrezak-Hahad-Casbah, 16017 Alger, Algérie
| | - R Abtroun
- Service de toxicologie, CHU Bab-El-Oued, rue Mohamed-Lamine-Debaghine, 16009 Alger, Algérie
| | - M Reggabi
- Laboratoire central de biologie et de toxicologie, EHS Ait-Idir, rue Abderrezak-Hahad-Casbah, 16017 Alger, Algérie
| | - B Alamir
- Service de toxicologie, CHU Bab-El-Oued, rue Mohamed-Lamine-Debaghine, 16009 Alger, Algérie; Centre national de toxicologie, route du Petit-Staouali-Delly-Brahim, 16062 Alger, Algérie
| |
Collapse
|
8
|
Ekhart C, de Jong L, Gross-Martirosyan L, van Hunsel F. Muscle rupture associated with statin use. Br J Clin Pharmacol 2016; 82:473-7. [PMID: 27074553 DOI: 10.1111/bcp.12973] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/15/2016] [Accepted: 04/09/2016] [Indexed: 01/14/2023] Open
Abstract
AIM Statins are used in the treatment of hyperlipidaemia. They are among the most commonly prescribed drugs worldwide. Statins have been linked to musculoskeletal adverse drug reactions. However muscle rupture has not been discussed as an adverse drug reaction to statins so far. The aim of this article is to give an overview of cases of muscle rupture associated with the use of statins. METHOD We analyzed the cases of muscle rupture associated with the use of statins that were collected by the Netherlands Pharmacovigilance Centre Lareb complemented with the review of cases from the EudraVigilance database. RESULTS Fifteen cases of muscle rupture associated with statin use have been identified in the database of the Netherlands Pharmacovigilance Centre. Overall, there was a plausible temporal association of events in most cases. In addition, the EudraVigilance database contained 165 reports of muscle rupture reported in patients using statins. Muscle rupture was disproportionally associated with statin use in both databases. The reporting odds ratio was 23.4 (95% CI 11.9, 46.0) and 14.6 (95% CI 12.3, 17.2), respectively. CONCLUSION Data from spontaneous reporting systems suggest that use of statins is associated with muscle rupture. Physicians and patients should be aware that this can occur.
Collapse
Affiliation(s)
- Corine Ekhart
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
| | - Loek de Jong
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
| | | | - Florence van Hunsel
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
| |
Collapse
|
9
|
Abstract
Approximately 95% of statin-treated patients tolerate this form of cholesterol management without any adverse effects. However, given their efficacy in reducing low density lipoproteins and cardiovascular events large numbers of patients are selected for statin therapy. Therefore muscle complications are, in fact, quite common. Limited understanding of the underlying pathophysiology has hampered physicians' ability to identify patients at risk for developing statin myotoxicity. A growing number of published case reports/series have implicated statins in the exacerbation of both acquired and genetic myopathies. A clinical management algorithm is presented which outlines a variety of co-morbidities which can potentiate the adverse effects of statins on muscle. In addition, a rational approach to the selection of those patients most likely to benefit from skeletal muscle biopsy is discussed. Ongoing work will define the extent to which statin-intolerant patients represent carriers of recessive metabolic myopathies or pre-symptomatic acquired myopathies. The expanding importance of pharmacogenomics will undoubtedly be realized in the field of statin myopathy research within the next few years. Such critical information is needed to establish more definitive management and diagnostic strategies.
Collapse
|
10
|
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
|
11
|
Costa RAP, Fernandes MP, de Souza-Pinto NC, Vercesi AE. Protective effects of l-carnitine and piracetam against mitochondrial permeability transition and PC3 cell necrosis induced by simvastatin. Eur J Pharmacol 2013; 701:82-6. [PMID: 23333250 DOI: 10.1016/j.ejphar.2013.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 12/20/2012] [Accepted: 01/08/2013] [Indexed: 11/16/2022]
Abstract
Mitochondrial oxidative stress followed by membrane permeability transition (MPT) has been considered as a possible mechanism for statins cytotoxicity. Statins use has been associated with reduced risk of cancer incidence, especially prostate cancer. Here we investigated the pathways leading to simvastatin-induced prostate cancer cell death as well as the mechanisms of cell death protection by l-carnitine or piracetam. These compounds are known to prevent and/or protect against cell death mediated by oxidative mitochondrial damage induced by a variety of conditions, either in vivo or in vitro. The results provide evidence that simvastatin induced MPT and cell necrosis were sensitive to either l-carnitine or piracetam in a dose-dependent fashion and mediated by additive mechanisms. When combined, l-carnitine and piracetam acted at concentrations significantly lower than they act individually. These results shed new light into both the cytotoxic mechanisms of statins and the mechanisms underlying the protection against MPT and cell death by the compounds l-carnitine and piracetam.
Collapse
Affiliation(s)
- Rute A P Costa
- Departamento de Patologia Clínica, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | | | | | | |
Collapse
|
12
|
Ghatak A, Faheem O, Thompson PD. The genetics of statin-induced myopathy. Atherosclerosis 2009; 210:337-43. [PMID: 20042189 DOI: 10.1016/j.atherosclerosis.2009.11.033] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Revised: 11/20/2009] [Accepted: 11/20/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Our goal was to use genetic variants to identify factors contributing to the muscular side effects of statins. BACKGROUND Statins (3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors) are usually well tolerated medications, but muscle symptoms, ranging from mild myalgia to clinically important rhabdomyolysis are an important side effect of these drugs and a leading cause of noncompliance. Recent results suggest that genetic factors increase the risk of statin-related muscle complaints. We performed a systematic review of the medical literature to determine genetic factors associated with statin myopathy. METHODS We identified English language articles relating statin myopathy and genetic diseases and gene variants via a PubMed search. Articles pertinent to the topic were reviewed in detail. RESULTS/CONCLUSIONS Our review suggests that some patients are susceptible to statin myopathy because of pre-existing subclinical inherited muscular disorders, or genetic variation in statin uptake proteins encoded by SLCO1B1 or the cytochrome P enzyme system. Variations in genes affecting pain perception and polymorphism in vascular receptors may also contribute to statin myopathy. None of the variants identified in this review suggested novel metabolic mechanisms leading to statin myopathy.
Collapse
Affiliation(s)
- Abhijit Ghatak
- Department of Internal Medicine, University of Connecticut, Farmington, CT 06032, USA
| | | | | |
Collapse
|
13
|
Abstract
The statins have emerged as the dominant class of drug for the treatment of hypercholesterolemia. These medications are generally well tolerated. However, myalgias, the most frequent side-effect, occur in up to 7% of patients. Transaminitis and skeletal myotoxicity, with elevated serum creatine kinase (CK) levels (i.e., >10 times the upper limit of normal), occur with reported frequencies of 1% and 0.1%, respectively. Various hypotheses have been proposed to explain the relationship between statin therapy and the spectrum of muscle dysfunction manifested by myalgia, myopathy, and rhabdomyolysis.Statin-mediatd inhibition of mevalonate metabolism impairs the synthesis of isoprenylated products–the most notable of which is ubiquinone. However, isoprenylation is responsible for the post-translational modification of up to 2% of cellular proteins. Therefore, numerous metabolic pathways are potentially modified by statin-mediated hypoprenylation. Subclinical defects in one or more energy-deriving pathways may be unmasked upon exposure to the pleotropic effects of statins. Such pharmacogenomic synergism may underlie the development of “statin myopathy” in a subset of patients. In this regard, we describe four patients with mutations in the myophosphorylase (PYGM; MIM 232600), myoadenylate deaminase (AMPD1; MIM 102770), and carnitine palmitoyltransferase (CPT2; MIM 600650) genes whose diagnoses became apparent during the course of investigations for statin-induced myalgias and hyperCKemia.
Collapse
|
14
|
Antons KA, Williams CD, Baker SK, Phillips PS. Clinical perspectives of statin-induced rhabdomyolysis. Am J Med 2006; 119:400-9. [PMID: 16651050 DOI: 10.1016/j.amjmed.2006.02.007] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Revised: 01/26/2006] [Accepted: 02/08/2006] [Indexed: 11/30/2022]
Abstract
Fear of muscle toxicity remains a major reason that patients with hyperlipidemia are undertreated. Recent evaluations of statin-induced rhabdomyolysis offer new insights on the clinical management of both muscle symptoms and hyperlipidemia after rhabdomyolysis. The incidence of statin-induced rhabdomyolysis is higher in practice than in controlled trials in which high-risk subjects are excluded. Accepted risks include age; renal, hepatic, and thyroid dysfunction; and hypertriglyceridemia. New findings suggest that exercise, Asian race, and perioperative status also may increase the risk of statin muscle toxicity. The proposed causes and the relationship of drug levels to statin rhabdomyolysis are briefly reviewed along with the problems with the pharmacokinetic theory. Data suggesting that patients with certain metabolic abnormalities are predisposed to statin rhabdomyolysis are presented. The evaluation and treatment of patients' muscle symptoms and hyperlipidemia after statin rhabdomyolysis are presented. Patients whose symptoms are related to other disorders need to be identified. Lipid management of those whose symptoms are statin-related is reviewed including treatment suggestions.
Collapse
Affiliation(s)
- Kenneth A Antons
- Scripps Mercy Clinical Research Center, Scripps Mercy Hospital, San Diego, Calif 92103, USA
| | | | | | | |
Collapse
|
15
|
Vladutiu GD, Simmons Z, Isackson PJ, Tarnopolsky M, Peltier WL, Barboi AC, Sripathi N, Wortmann RL, Phillips PS. Genetic risk factors associated with lipid-lowering drug-induced myopathies. Muscle Nerve 2006; 34:153-62. [PMID: 16671104 DOI: 10.1002/mus.20567] [Citation(s) in RCA: 196] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Lipid-lowering drugs produce myopathic side effects in up to 7% of treated patients, with severe rhabdomyolysis occurring in as many as 0.5%. Underlying metabolic muscle diseases have not been evaluated extensively. In a cross-sectional study of 136 patients with drug-induced myopathies, we report a higher prevalence of underlying metabolic muscle diseases than expected in the general population. Control groups included 116 patients on therapy with no myopathic symptoms, 100 asymptomatic individuals from the general population never exposed to statins, and 106 patients with non-statin-induced myopathies. Of 110 patients who underwent mutation testing, 10% were heterozygous or homozygous for mutations causing three metabolic myopathies, compared to 3% testing positive among asymptomatic patients on therapy (P = 0.04). The actual number of mutant alleles found in the test group patients was increased fourfold over the control group (P < 0.0001) due to an increased presence of mutation homozygotes. The number of carriers for carnitine palmitoyltransferase II deficiency and for McArdle disease was increased 13- and 20-fold, respectively, over expected general population frequencies. Homozygotes for myoadenylate deaminase deficiency were increased 3.25-fold with no increase in carrier status. In 52% of muscle biopsies from patients, significant biochemical abnormalities were found in mitochondrial or fatty acid metabolism, with 31% having multiple defects. Variable persistent symptoms occurred in 68% of patients despite cessation of therapy. The effect of statins on energy metabolism combined with a genetic susceptibility to triggering of muscle symptoms may account for myopathic outcomes in certain high-risk groups.
Collapse
Affiliation(s)
- Georgirene D Vladutiu
- Department of Pediatrics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, 936 Delaware Avenue, Buffalo, New York 14209, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Arduini A, Peschechera A, Giannessi F, Carminati P. Improvement of statin-associated myotoxicity by L-carnitine. J Thromb Haemost 2004; 2:2270-1. [PMID: 15613049 DOI: 10.1111/j.1538-7836.2004.01048.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
17
|
Sato H, Takahashi N, Nakamoto M, Ohgami M, Yamazaki M, Fukui T. Effects of streptozotocin-induced diabetes on acetoacetyl-CoA synthetase activity in rats. Biochem Pharmacol 2002; 63:1851-5. [PMID: 12034369 DOI: 10.1016/s0006-2952(02)00911-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In order to investigate the physiological role of acetoacetyl-CoA synthetase (acetoacetate-CoA ligase, EC 6.2.1.16), a cytosolic acetoacetate-activating enzyme, effects of streptozotocin (STZ)-induced diabetes on the enzyme activity was investigated in rats. At 72 hr of the STZ administration (80 mg/kg body weight, injected intravenously), hepatic enzyme specific activity decreased to 23% of its initial activity. However, the enzyme activities in non-hepatic tissues were not significantly affected by the STZ treatment. Feeding of rats with both 4% cholestyramine and 0.4% pravastatin for 3 days remarkably increased the hepatic acetoacetyl-CoA synthetase activity and decreased the plasma ketone bodies level in the diabetic rats. These results suggest that acetoacetyl-CoA synthetase has important roles in the regulation of ketone body utilization in rat liver and that these hypocholesterolemic agents have the ability to remedy the impaired utilization of ketone bodies under the diabetic condition.
Collapse
Affiliation(s)
- Hiroki Sato
- Department of Health Chemistry, Hoshi University, Shinagawa-ku, Tokyo 142-8501, Japan
| | | | | | | | | | | |
Collapse
|
18
|
Yamamoto K, Todaka N, Goto H, Jayasooriya AP, Sakono M, Ogawa Y, Fukuda N. Effect of NK-104, a new synthetic HMG-CoA reductase inhibitor, on triglyceride secretion and fatty acid oxidation in rat liver. Life Sci 1999; 65:1493-502. [PMID: 10530801 DOI: 10.1016/s0024-3205(99)00390-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
For the investigation of the mechanism responsible for the hypotriglyceridemic effect of NK-104, a new synthetic inhibitor of HMG-CoA reductase, the rate-limiting enzyme for cholesterol synthesis, isolated rat liver was perfused with or without NK-104 in the presence of exogenous [1-(14)C]oleic acid substrate. Addition of NK-104 tended to increase the ketone body production while it caused a significant decrease in the secretion rate of triglyceride by the perfused liver without affecting uptake of exogenous [1-(14)C]oleic acid. The inhibitor also significantly decreased hepatic triglyceride concentration. The altered triglyceride secretion was accompanied by a concomitant decreased incorporation of exogenous [1-(14)C]oleate into triglyceride. The conversion of exogenous [1-(14)C]oleic acid substrate indicated an inverse relationship between the pathways of oxidation and esterification. No effect of NK-104 on hepatic secretion of cholesterol was observed. These results suggest that NK-104 exerts its hypotriglyceridemic action, primarily by diverting the exogenous free fatty acid to the pathways of oxidation at the expense of esterification.
Collapse
Affiliation(s)
- K Yamamoto
- Department of Internal Medicine, Saga Medical University School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
19
|
Christians U, Jacobsen W, Floren LC. Metabolism and drug interactions of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors in transplant patients: are the statins mechanistically similar? Pharmacol Ther 1998; 80:1-34. [PMID: 9804052 DOI: 10.1016/s0163-7258(98)00016-3] [Citation(s) in RCA: 160] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
3-Hydroxy-3-methylglutaryl coenzyme A reductase (EC 1.1.1.88) inhibitors are the most effective drugs to lower cholesterol in transplant patients. However, immunosuppressants and several other drugs used after organ transplantation are cytochrome P4503A (CYP3A, EC 1.14.14.1) substrates. Pharmacokinetic interaction with some of the 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, specifically lovastatin and simvastatin, leads to an increased incidence of muscle skeletal toxicity in transplant patients. It is our objective to review the role of drug metabolism and drug interactions of lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, and cerivastatin. In the treatment of transplant patients, from a drug interaction perspective, pravastatin, which is not significantly metabolized by CYP enzymes, and fluvastatin, presumably a CYP2C9 substrate, compare favorably with the other statins for which the major metabolic pathways are catalyzed by CYP3A.
Collapse
Affiliation(s)
- U Christians
- Department of Biopharmaceutical Sciences, School of Pharmacy, University of California at San Francisco, 94143-0446, USA
| | | | | |
Collapse
|