1
|
Kuretu A, Mothibe M, Ngubane P, Sibiya N. Elucidating the effect of drug-induced mitochondrial dysfunction on insulin signaling and glucose handling in skeletal muscle cell line (C2C12) in vitro. PLoS One 2024; 19:e0310406. [PMID: 39288128 PMCID: PMC11407670 DOI: 10.1371/journal.pone.0310406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/31/2024] [Indexed: 09/19/2024] Open
Abstract
Efavirenz, tenofovir, rifampicin, simvastatin, lamotrigine and clarithromycin are known potential mitochondrial toxicants. Mitochondrial toxicity has been reported to disrupt the chain of events in the insulin signalling pathway. Considering the upward trajectory of diabetes mellitus prevalence, studies which seek to uncover probable risk factors for developing diabetes should be encouraged. This study aimed to evaluate the intracellular mechanisms leading to the development of insulin resistance in the presence of various conventional pharmacological agents reported as potential mitochondrial toxicants in skeletal muscle cell line. Differentiated C2C12 preparations were exposed to multiple concentrations of efavirenz, tenofovir, rifampicin, simvastatin, lamotrigine, and clarithromycin, separately. Glucose handling was evaluated by observing the changes in insulin-stimulated glucose uptake and assessing the changes in GLUT4 translocation, GLUT4 expression and Akt expression. The changes in mitochondrial function were evaluated by assessing mitochondrial membrane integrity, cellular ATP production, generation of intracellular reactive oxygen species, expression of tafazzin and quantification of medium malonaldehyde. Insulin stimulated glucose uptake was perturbed in C2C12 pre-treated with potential mitotoxicants. Additionally, ATP synthesis, alterations in mitochondrial membrane potential, excessive accumulation of ROS and malonaldehyde were observed in the presence of potential mitotoxicants. Particularly, we observed suppression of proteins involved in the insulin signalling pathway and maintenance of mitochondrial function namely GLUT4, Akt and tafazzin. Mitochondrial toxicants can potentially induce insulin resistance emanating from mitochondrial dysfunction. These new findings will contribute to the understanding of underlying mechanisms involved in the development of insulin resistance linked to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Auxiliare Kuretu
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Mamosheledi Mothibe
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Phikelelani Ngubane
- School of Medical Sciences and Laboratory Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Ntethelelo Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| |
Collapse
|
2
|
Lee J, Choi JY, Choi BG, Choi YJ, Park S, Kang DO, Park EJ, Kim JB, Roh SY, Na JO, Choi CU, Kim EJ, Park CG, Jeong MH, Hwang JY, Hur SH, Jeong JO, Oh S, Rha SW. Different diabetogenic effect of statins according to intensity and dose in patients with acute myocardial infarction: a nationwide cohort study. Sci Rep 2024; 14:19438. [PMID: 39169014 PMCID: PMC11339444 DOI: 10.1038/s41598-024-67585-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/12/2024] [Indexed: 08/23/2024] Open
Abstract
Statin is crucial for acute myocardial infarction (AMI) patients. However, the risk of new-onset diabetes mellitus (NODM) associated with statin is a concern. This study aimed to determine the incremental diabetogenic effects of statins according to their intensity and dose in AMI patients undergoing percutaneous coronary intervention (PCI). Among 13,104 patients enrolled in the Korea AMI Registry between 2011 and 2015, 6152 patients without diabetes mellitus (DM) who underwent PCI and received moderate-to-high-intensity atorvastatin and rosuvastatin were selected for the study. The endpoints were NODM and major adverse cardiovascular events (MACE), composite of all-cause mortality, recurrent MI, and revascularization up to 3 years. Among the participants, 3747 and 2405 received moderate- and high-intensity statins, respectively. The Kaplan-Meier curves demonstrated a higher incidence of NODM in patients with high-intensity statins than those with moderate-intensity. High-intensity statin was a significant predictor of NODM after adjusting for other co-variables (HR = 1.316, 95% CI 1.024-1.692; P < 0.032). Higher dose of rosuvastatin was associated with a higher cumulative incidence of NODM, but this dose-dependency was not apparent with atorvastatin. Cumulative incidence of MACE decreased dose-dependently only with atorvastatin. High-intensity statin was associated with a higher cumulative incidence of NODM in AMI patients, and this association was more evident in rosuvastatin. The different diabetogenic effects of the two statins provide supporting evidence for understanding the nuanced nature of statin treatment in relation to NODM.
Collapse
Affiliation(s)
- Jieun Lee
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Jah Yeon Choi
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Byoung Geol Choi
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - You-Jung Choi
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Soohyung Park
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Dong Oh Kang
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Eun Jin Park
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Ji Bak Kim
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Seung Young Roh
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Jin Oh Na
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Cheol Ung Choi
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Eung Ju Kim
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Chang Gyu Park
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea
| | - Myung Ho Jeong
- Chonnam National University Hospital, Gwangju, South Korea
| | - Jin-Yong Hwang
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Jinju, South Korea
| | - Seung-Ho Hur
- Keimyung University Dongsan Medical Center, Cardiovascular Medicine, Deagu, South Korea
| | - Jin-Ok Jeong
- Department of Internal Medicine, College of Medicine, Chungnam National University Hospital, Chungnam National Univeresity, Daejeon, South Korea
| | - SeokKyu Oh
- Division of Cardiology, Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, South Korea
| | - Seung-Woon Rha
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, South Korea.
| |
Collapse
|
3
|
Alvarez-Jimenez L, Morales-Palomo F, Moreno-Cabañas A, Ortega JF, Mora-Rodríguez R. Effects of statin therapy on glycemic control and insulin resistance: A systematic review and meta-analysis. Eur J Pharmacol 2023; 947:175672. [PMID: 36965747 DOI: 10.1016/j.ejphar.2023.175672] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
AIMS To update the evidence about the diabetogenic effect of statins. METHODS We searched for randomized-controlled trials reporting the effects of statin therapy on glycosylated hemoglobin (HbA1c) and/or homeostatic model insulin resistance (i.e., HOMA-IR) as indexes of diabetes. Studies were classified between the ones testing normal vs individuals with already altered glycemic control (HbA1c ≥ 6.5%; and HOMA-IR ≥ 2.15). Furthermore, studies were separated by statin type and dosage prescribed. Data are presented as mean difference (MD) and 95% confidence intervals. RESULTS A total of 67 studies were included in the analysis (>25,000 individuals). In individuals with altered glycemic control, statins increased HbA1c levels (MD 0.21%, 95% CI 0.16-to-0.25) and HOMA-IR index (MD 0.31, 95% CI 0.24-to-0.38). In individuals with normal glycemic control, statin increased HbA1c (MD 1.33%, 95% CI 1.31-to-1.35) and HOMA-IR (MD 0.49, 95% CI 0.41-to-0.58) in comparison to the placebo groups. The dose or type of statins did not modulate the diabetogenic effect. CONCLUSIONS Statins, slightly but significantly raise indexes of diabetes in individuals with adequate or altered glycemic control. The diabetogenic effect does not seem to be influenced by the type or dosage of statin prescribed.
Collapse
Affiliation(s)
- Laura Alvarez-Jimenez
- Exercise Physiology Lab at Toledo, Sports Science Department, University of Castilla-La Mancha, 45004, Toledo, Spain
| | - Felix Morales-Palomo
- Exercise Physiology Lab at Toledo, Sports Science Department, University of Castilla-La Mancha, 45004, Toledo, Spain
| | - Alfonso Moreno-Cabañas
- Exercise Physiology Lab at Toledo, Sports Science Department, University of Castilla-La Mancha, 45004, Toledo, Spain
| | - Juan F Ortega
- Exercise Physiology Lab at Toledo, Sports Science Department, University of Castilla-La Mancha, 45004, Toledo, Spain
| | - Ricardo Mora-Rodríguez
- Exercise Physiology Lab at Toledo, Sports Science Department, University of Castilla-La Mancha, 45004, Toledo, Spain.
| |
Collapse
|
4
|
Statin-Induced Geranylgeranyl Pyrophosphate Depletion Promotes PCSK9-Dependent Adipose Insulin Resistance. Nutrients 2022; 14:nu14245314. [PMID: 36558473 PMCID: PMC9853319 DOI: 10.3390/nu14245314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Statin treatment is accepted to prevent adverse cardiovascular events. However, statin therapy has been reported to be dose-dependently associated with increased risk for new-onset type 2 diabetes mellitus (T2DM). Proprotein convertase subtilisin/kexin type 9 (PCSK9) is expressed in adipose tissue and is positively correlated with lipid metabolism. It is, however, unknown if PCSK9 participates in adipocyte insulin resistance occurring as a result of statin use. Our goal was to use an in vitro adipose tissue explant approach to support the hypothesis that PCSK9 regulates statin-induced new-onset T2DM. Studies were performed using Pcsk-/- and C57Bl/6J control mice. Pcsk9-/- and control mice were fed a high-fat diet to affect a state of chronically altered lipid metabolism and increased PCSK9. Epididymal fat was excised and incubated with atorvastatin (1 µmol/L) in the absence and presence of insulin or geranylgeranyl pyrophosphate (GGPP). PCSK9 mRNA was evaluated using quantitative rtPCR. We further examined the effects of atorvastatin on insulin-mediated AKT signaling in adipose tissue explants by immunoblotting. Atorvastatin was found to upregulate PCSK9 gene expression in adipose tissue. The metabolic intermediate GGPP is required to downregulate PCSK9 expression. PCSK9 deficiency protects against statin-induced impairments in insulin signaling. Moreover, supplementation with GGPP reversed atorvastatin-induced suppression of insulin signaling. Furthermore, the basal and atorvastatin-stimulated release of free fatty acids was observed in adipose tissue from wild-type mice but not PCSK9 deficient mice. Collectively, we describe a novel mechanism for PCSK9 expression in adipose tissue that could mediate statin-impaired adipose insulin resistance.
Collapse
|
5
|
English AR, Prasad B, McGuigan DH, Horigan G, O’Kane M, Bjourson AJ, Shukla P, Kelly C, McClean PL. Simvastatin is associated with superior lipid and glycaemic control to atorvastatin and reduced levels of incident Type 2 diabetes, in men and women, in the UK Biobank. Endocrinol Diabetes Metab 2022; 5:e00326. [PMID: 35243827 PMCID: PMC9094470 DOI: 10.1002/edm2.326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Cardiovascular disease (CVD) is the leading cause of mortality in people with Type 2 diabetes mellitus (T2DM). Statins reduce low-density lipoproteins and positively affect CVD outcomes. Statin type and dose have differential effects on glycaemia and risk of incident T2DM; however, the impact of gender, and of individual drugs within the statin class, remains unclear. AIM To compare effects of simvastatin and atorvastatin on lipid and glycaemic control in men and women with and without T2DM, and their association with incident T2DM. METHODS The effect of simvastatin and atorvastatin on lipid and glycaemic control was assessed in the T2DM DiaStrat cohort. Prescribed medications, gender, age, BMI, diabetes duration, blood lipid profile and HbA1c were extracted from Electronic Care Record, and compared in men and women prescribed simvastatin and atorvastatin. Analyses were replicated in the UKBiobank in those with and without T2DM. The association of simvastatin and atorvastatin with incident T2DM was also investigated in the UKBiobank. Cohorts where matched for age, BMI and diabetes duration in men and women, in the UKBioBank analysis, where possible. RESULTS Simvastatin was associated with better LDL (1.6 ± 0.6 vs 2.1 ± 0.9 mmol/L, p < .01) and total cholesterol (3.6 ± 0.7 vs 4.2 ± 1.0 mmol/L, p < .05), and glycaemic control (62 ± 17 vs 67 ± 19 mmol/mol, p < .059) than atorvastatin specifically in women in the DiaStrat cohort. In the UKBiobank, both men and women prescribed simvastatin had better LDL (Women: 2.6 ± 0.6 vs 2.6 ± 0.7 mmol/L, p < .05; Men: 2.4 ± 0.6 vs 2.4 ± 0.6, p < .01) and glycaemic control (Women:54 ± 14 vs 56 ± 15mmol/mol, p < .05; Men, 54 ± 14 vs 55 ± 15 mmol/mol, p < .01) than those prescribed atorvastatin. Simvastatin was also associated with reduced risk of incident T2DM in both men and women (p < .0001) in the UKBiobank. CONCLUSIONS Simvastatin is associated with superior lipid and glycaemic control to atorvastatin in those with and without T2DM, and with fewer incident T2DM cases. Given the importance of lipid and glycaemic control in preventing secondary complications of T2DM, these findings may help inform prescribing practices.
Collapse
Affiliation(s)
- Andrew R. English
- Northern Ireland Centre for Stratified Medicine School of Biomedical Sciences C‐TRIC Altnagelvin Hospital Ulster University Derry~Londonderry UK
| | - Bodhayan Prasad
- Northern Ireland Centre for Stratified Medicine School of Biomedical Sciences C‐TRIC Altnagelvin Hospital Ulster University Derry~Londonderry UK
| | - Declan H. McGuigan
- Northern Ireland Centre for Stratified Medicine School of Biomedical Sciences C‐TRIC Altnagelvin Hospital Ulster University Derry~Londonderry UK
| | - Geraldine Horigan
- Northern Ireland Centre for Stratified Medicine School of Biomedical Sciences C‐TRIC Altnagelvin Hospital Ulster University Derry~Londonderry UK
| | - Maurice O’Kane
- Clinical Chemistry Laboratory Altnagelvin Hospital Derry~Londonderry UK
- Centre for Personalised Medicine: Clinical Decision Making and Patient Safety C‐TRIC Altnagelvin Hospital Londonderry UK
| | - Anthony J. Bjourson
- Northern Ireland Centre for Stratified Medicine School of Biomedical Sciences C‐TRIC Altnagelvin Hospital Ulster University Derry~Londonderry UK
| | - Priyank Shukla
- Northern Ireland Centre for Stratified Medicine School of Biomedical Sciences C‐TRIC Altnagelvin Hospital Ulster University Derry~Londonderry UK
| | - Catriona Kelly
- Northern Ireland Centre for Stratified Medicine School of Biomedical Sciences C‐TRIC Altnagelvin Hospital Ulster University Derry~Londonderry UK
| | - Paula L. McClean
- Northern Ireland Centre for Stratified Medicine School of Biomedical Sciences C‐TRIC Altnagelvin Hospital Ulster University Derry~Londonderry UK
| |
Collapse
|
6
|
Ji Y, Song J, Su T, Gu X. Adipokine Retinol Binding Protein 4 and Cardiovascular Diseases. Front Physiol 2022; 13:856298. [PMID: 35309061 PMCID: PMC8924404 DOI: 10.3389/fphys.2022.856298] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 01/31/2022] [Indexed: 01/12/2023] Open
Abstract
The morbidity and mortality of cardiovascular diseases (CVDs) have been increasing year by year all over the world and expanding greatly to the younger population, which becomes the leading causes of death globally that threatens human life safety. Prediction of the occurrence of diseases by using risk related adverse events is crucial for screening and early detection of CVDs. Thus, the discovery of new biomarkers that related to risks of CVDs are of urgent in the field. Retinol-binding protein 4 (RBP4) is a 21-kDa adipokine, mainly secreted by adipocytes. Besides its well-established function in the induction of insulin resistance, it has also been found in recent years to be closely associated with CVDs and other risk factors, such as hypertension, coronary heart disease, heart failure, obesity, and hyperlipidemia. In this review, we mainly focus on the progress of research that establishes the correlation between RBP4 and CVDs and the corresponding major risk factors in recent years.
Collapse
|
7
|
Bahrami A, Bo S, Jamialahmadi T, Sahebkar A. Effects of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors on ageing: Molecular mechanisms. Ageing Res Rev 2020; 58:101024. [PMID: 32006687 DOI: 10.1016/j.arr.2020.101024] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 12/11/2019] [Accepted: 01/27/2020] [Indexed: 12/15/2022]
Abstract
Human ageing is determined by degenerative alterations and processes with different manifestations such as gradual organ dysfunction, tissue function loss, increased population of aged (senescent) cells, incapability of maintaining homeostasis and reduced repair capacity, which collectively lead to an increased risk of diseases and death. The inhibitors of HMG-CoA reductase (statins) are the most widely used lipid-lowering agents, which can reduce cardiovascular morbidity and mortality. Accumulating evidence has documented several pleiotropic effects of statins in addition to their lipid-lowering properties. Recently, several studies have highlighted that statins may have the potential to delay the ageing process and inhibit the onset of senescence. In this review, we focused on the anti-ageing mechanisms of statin drugs and their effects on cardiovascular and non-cardiovascular diseases.
Collapse
|
8
|
Sunjaya AP, Sunjaya AF, Halim S, Ferdinal F. Risk and Benefits of Statins in Glucose Control Management of Type II Diabetes. Int J Angiol 2018; 27:121-131. [PMID: 30154630 DOI: 10.1055/s-0036-1572523] [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] [Indexed: 01/12/2023] Open
Abstract
Worldwide statins are considered to be the first-line pharmacological treatment for dyslipidemia and reducing the risk of coronary heart disease. However, recently various studies have shown its adverse effect on glucose control among diabetic patients and the U.S. Food and Drug Administration have revised statin drug labels to include information that increases in fasting serum glucose and glycated hemoglobin levels have been reported. This systematic review objective is to evaluate the risks and benefits of statins in glucose control management of type 2 diabetes patients based on the 44 published journal articles included and obtained through MEDLINE full text, PubMed, Science Direct, Pro Quest, SAGE, Taylor and Francis Online, Google Scholar, High Wire, and Elsevier Clinical Key. Statins were found to affect glucose control through several ways, namely, by affecting insulin production and secretion by β-pancreatic cells, insulin resistance, insulin uptake by the muscles and adipocytes and production of adipokines. Current evidence available shows that most of the statins give unfavorable side effects with regards to glucose control among diabetic patients. A dose-dependent and time-dependent effect was also observed in some statins which may be present among other statins as well.
Collapse
Affiliation(s)
| | | | - Samuel Halim
- Department of Internal Medicine, Faculty of Medicine, Tarumanagara University, Jakarta, Indonesia
| | - Frans Ferdinal
- Department of Biochemistry, Faculty of Medicine, Tarumanagara University, Jakarta, Indonesia
| |
Collapse
|
9
|
Mach F, Ray KK, Wiklund O, Corsini A, Catapano AL, Bruckert E, De Backer G, Hegele RA, Hovingh GK, Jacobson TA, Krauss RM, Laufs U, Leiter LA, März W, Nordestgaard BG, Raal FJ, Roden M, Santos RD, Stein EA, Stroes ES, Thompson PD, Tokgözoğlu L, Vladutiu GD, Gencer B, Stock JK, Ginsberg HN, Chapman MJ, European Atherosclerosis Society Consensus Panel. Adverse effects of statin therapy: perception vs. the evidence - focus on glucose homeostasis, cognitive, renal and hepatic function, haemorrhagic stroke and cataract. Eur Heart J 2018; 39:2526-2539. [PMID: 29718253 PMCID: PMC6047411 DOI: 10.1093/eurheartj/ehy182] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/09/2017] [Accepted: 03/22/2018] [Indexed: 12/17/2022] Open
Abstract
Aims To objectively appraise evidence for possible adverse effects of long-term statin therapy on glucose homeostasis, cognitive, renal and hepatic function, and risk for haemorrhagic stroke or cataract. Methods and results A literature search covering 2000-2017 was performed. The Panel critically appraised the data and agreed by consensus on the categorization of reported adverse effects. Randomized controlled trials (RCTs) and genetic studies show that statin therapy is associated with a modest increase in the risk of new-onset diabetes mellitus (about one per thousand patient-years), generally defined by laboratory findings (glycated haemoglobin ≥6.5); this risk is significantly higher in the metabolic syndrome or prediabetes. Statin treatment does not adversely affect cognitive function, even at very low levels of low-density lipoprotein cholesterol and is not associated with clinically significant deterioration of renal function, or development of cataract. Transient increases in liver enzymes occur in 0.5-2% of patients taking statins but are not clinically relevant; idiosyncratic liver injury due to statins is very rare and causality difficult to prove. The evidence base does not support an increased risk of haemorrhagic stroke in individuals without cerebrovascular disease; a small increase in risk was suggested by the Stroke Prevention by Aggressive Reduction of Cholesterol Levels study in subjects with prior stroke but has not been confirmed in the substantive evidence base of RCTs, cohort studies and case-control studies. Conclusion Long-term statin treatment is remarkably safe with a low risk of clinically relevant adverse effects as defined above; statin-associated muscle symptoms were discussed in a previous Consensus Statement. Importantly, the established cardiovascular benefits of statin therapy far outweigh the risk of adverse effects.
Collapse
Affiliation(s)
- François Mach
- Division of Cardiology, Department of Medical Specialties, Foundation for Medical Researches, Geneva University Hospital, Rue Gabrielle-Perret-Gentil 4 1205 Geneva, Switzerland
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
| | - Olov Wiklund
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, University of Milan and IRCCS Multimedica, Milan, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan and IRCCS Multimedica, Milan, Italy
| | - Eric Bruckert
- National Institute for Health and Medical Research (INSERM) UMRS1166, Department of Endocrinology-Metabolism, ICAN—Institute of CardioMetabolism and Nutrition, AP-HP, Hôpital de la Pitié, Paris, France
| | - Guy De Backer
- Department of Public Health, University Hospital Ghent, Ghent, Belgium
| | - Robert A Hegele
- Department of Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Ronald M Krauss
- Department of Atherosclerosis Research, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - Ulrich Laufs
- Department of Cardiology, University of Leipzig, Leipzig, Germany
| | - Lawrence A Leiter
- Li Ka Shing Knowledge Institute of St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry and The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Frederick J Raal
- Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Michael Roden
- German Center for Diabetes Research (DZD), München-Neuherberg, Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Raul D Santos
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Evan A Stein
- Metabolic and Atherosclerosis Research Center, Cincinnati, OH, USA
| | - Erik S Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Lale Tokgözoğlu
- Department of Cardiology, Hacettepe University, Ankara, Turkey
| | - Georgirene D Vladutiu
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, The State University of New York, New York, USA
| | - Baris Gencer
- Division of Cardiology, Department of Medical Specialties, Foundation for Medical Researches, Geneva University Hospital, Rue Gabrielle-Perret-Gentil 4 1205 Geneva, Switzerland
| | - Jane K Stock
- European Atherosclerosis Society, Gothenburg, Sweden
| | - Henry N Ginsberg
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, USA
| | - M John Chapman
- National Institute for Health and Medical Research (INSERM), and University of Pierre and Marie Curie—Paris 6, Pitié Salpêtrière, Paris, France
| | | |
Collapse
|
10
|
van Stee MF, de Graaf AA, Groen AK. Actions of metformin and statins on lipid and glucose metabolism and possible benefit of combination therapy. Cardiovasc Diabetol 2018; 17:94. [PMID: 29960584 PMCID: PMC6026339 DOI: 10.1186/s12933-018-0738-4] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/20/2018] [Indexed: 12/13/2022] Open
Abstract
Patients with diabetes type 2 have an increased risk for cardiovascular disease and commonly use combination therapy consisting of the anti-diabetic drug metformin and a cholesterol-lowering statin. However, both drugs act on glucose and lipid metabolism which could lead to adverse effects when used in combination as compared to monotherapy. In this review, the proposed molecular mechanisms of action of statin and metformin therapy in patients with diabetes and dyslipidemia are critically assessed, and a hypothesis for mechanisms underlying interactions between these drugs in combination therapy is developed.
Collapse
Affiliation(s)
- Mariël F. van Stee
- Netherlands Organisation for Applied Scientific Research (TNO), Zeist, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert A. de Graaf
- Netherlands Organisation for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Albert K. Groen
- Amsterdam Diabetes Center and Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
11
|
Razali NR, Huri HZ, Ibrahim L, Vethakkan SR, Abdullah BM. Glycemic effects of simvastatin: Where do we stand? BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000117192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
| | - Hasniza Zaman Huri
- University of Malaya, Malaysia; University Malaya Medical Centre, Malaysia
| | | | | | | |
Collapse
|
12
|
Han KH. Functional Implications of HMG-CoA Reductase Inhibition on Glucose Metabolism. Korean Circ J 2018; 48:951-963. [PMID: 30334382 PMCID: PMC6196158 DOI: 10.4070/kcj.2018.0307] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023] Open
Abstract
HMG-CoA reductase inhibitors, i.e. statins, are effective in reducing cardiovascular disease events but also in cardiac-related and overall mortality. Statins are in general well-tolerated, but currently the concerns are raised if statins may increase the risk of new-onset diabetes mellitus (NOD). In this review, the possible effects of statins on organs/tissues being involved in glucose metabolism, i.e. liver, pancreas, adipose tissue, and muscles, had been discussed. The net outcome seems to be inconsistent and often contradictory, which may be largely affected by in vitro experimental settings or/and in vivo animal conditions. The majority of studies point out statin-induced changes of regulations of isoprenoid metabolites and cell-associated cholesterol contents as predisposing factors related to the statin-induced NOD. On the other hand, it should be considered that dysfunctions of isoprenoid pathway and mitochondrial ATP production and the cholesterol homeostasis are already developed under (pre)diabetic and hypercholesterolemic conditions. In order to connect the basic findings with the clinical manifestation more clearly, further research efforts are needed.
Collapse
Affiliation(s)
- Ki Hoon Han
- Department of Internal Medicine, College of Medicine Ulsan University, Asan Medical Center, Seoul, Korea.
| |
Collapse
|
13
|
Bril F, Cusi K. Management of Nonalcoholic Fatty Liver Disease in Patients With Type 2 Diabetes: A Call to Action. Diabetes Care 2017; 40:419-430. [PMID: 28223446 DOI: 10.2337/dc16-1787] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/08/2016] [Indexed: 02/03/2023]
Abstract
Traditionally a disease of hepatologists, nonalcoholic fatty liver disease (NAFLD) has recently become a major concern for a broad spectrum of health care providers. Endocrinologists and those caring for patients with type 2 diabetes mellitus (T2DM) are at center stage, as T2DM appears to worsen the course of NAFLD and the liver disease makes diabetes management more challenging. However, the nature of this relationship remains incompletely understood. Although the increasing prevalence of NAFLD is frequently attributed to the epidemic of obesity and is often oversimplified as the "hepatic manifestation of the metabolic syndrome," it is a much more complex disease process that may also be observed in nonobese individuals and in patients without clinical manifestations of the metabolic syndrome. It carries both metabolic and liver-specific complications that make its approach unique among medical conditions. Diabetes appears to promote the development of nonalcoholic steatohepatitis (NASH), the more severe form of the disease, and increases the risk of cirrhosis and hepatocellular carcinoma. Patients and physicians face many uncertainties, including fragmented information on the natural history of the disease, challenges in the diagnosis of NASH, and few pharmacological agents with proven efficacy. However, recent advances in diagnosis and treatment, combined with the risk of serious consequences from inaction, call for health care providers to be more proactive in the management of patients with T2DM and NASH.
Collapse
Affiliation(s)
- Fernando Bril
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, Florida
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, Florida .,Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida
| |
Collapse
|
14
|
Barb D, Cusi K. Reply to "statins and non-alcoholic steatohepatitis". Metabolism 2017; 66:e3-e5. [PMID: 27865560 DOI: 10.1016/j.metabol.2016.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/17/2016] [Indexed: 02/07/2023]
Affiliation(s)
- Diana Barb
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, Gainesville, FL; Division of Endocrinology, Diabetes, and Metabolism at Malcom Randall Veterans Affairs Medical Center, Gainesville, FL
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, Gainesville, FL; Division of Endocrinology, Diabetes, and Metabolism at Malcom Randall Veterans Affairs Medical Center, Gainesville, FL.
| |
Collapse
|
15
|
Cai R, Yuan Y, Sun J, Xia W, Huang R, Tian S, Dong X, Shen Y, Wang S. Statins worsen glycemic control of T2DM in target LDL-c level and LDL-c reduction dependent manners: a meta-analysis. Expert Opin Pharmacother 2016; 17:1839-49. [DOI: 10.1080/14656566.2016.1220539] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
16
|
Bernardi A, Rocha VZ, Faria-Neto JR. Use of statins and the incidence of type 2 diabetes mellitus. Rev Assoc Med Bras (1992) 2016; 61:375-80. [PMID: 26466221 DOI: 10.1590/1806-9282.61.04.375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 07/16/2015] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION the use of statins is associated with reduced cardiovascular risk in studies of primary and secondary prevention, and the reduction is directly proportional to the reduction of LDL-cholesterol. Recent evidence suggests that statins may be associated with a higher incidence of new cases of diabetes. The aim of this review is to explore this possibility, identifying factors associated with the increase in risk and the potential diabetogenic mechanisms of statins. In addition, we evaluated if the risk of diabetes interferes with the reduction in cardiovascular risk achieved with statins. METHODS we reviewed articles published in the Scielo and Pubmed databases, which assessed or described the association between use of statins and risk of diabetes up to June 2015. RESULTS use of statins is associated with a small increase in the incidence of new cases of diabetes. Age, potency of statin therapy, presence of metabolic syndrome, impaired fasting blood glucose, overweight and previously altered glycated hemoglobin levels are associated with increased risk of diabetes, but there is no consensus about the possible diabetogenic mechanisms of statins. In patients candidate to hypolipemiant drug therapy, the benefit of reducing cardiovascular risk outweighs any risk increase in the incidence of diabetes. CONCLUSION statins are associated with a small increase in incidence of diabetes in patients predisposed to glycemic alteration. However, since the benefit of cardiovascular risk reduction prevails even in this group, there is no evidence to date that this finding should change the recommendation of starting statin therapy.
Collapse
Affiliation(s)
- André Bernardi
- Centro de Epidemiologia e Pesquisa Clínica, Escola de Medicina, Pontifícia Universidade Católica do Paraná, Curitiba, PR, BR
| | | | - José Rocha Faria-Neto
- Centro de Epidemiologia e Pesquisa Clínica, Escola de Medicina, Pontifícia Universidade Católica do Paraná, Curitiba, PR, BR
| |
Collapse
|
17
|
Sahebkar A, Simental-Mendía LE, Pedone C, Ferretti G, Nachtigal P, Bo S, Derosa G, Maffioli P, Watts GF. Statin therapy and plasma free fatty acids: a systematic review and meta-analysis of controlled clinical trials. Br J Clin Pharmacol 2016; 81:807-18. [PMID: 26624855 DOI: 10.1111/bcp.12854] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 10/31/2015] [Accepted: 11/22/2015] [Indexed: 12/30/2022] Open
Abstract
AIM The aim of this meta-analysis was to evaluate the effect of statin therapy on plasma FFA concentrations in a systematic review and meta-analysis of controlled clinical trials. METHODS PubMed-Medline, SCOPUS, Web of Science and Google Scholar databases were searched (from inception to February 16 2015) to identify controlled trials evaluating the impact of statins on plasma FFA concentrations. A systematic assessment of bias in the included studies was performed using the Cochrane criteria. A random effects model and generic inverse variance method were used for quantitative data synthesis. Sensitivity analysis was conducted using the leave-one-out method. Random effects meta-regression was performed using unrestricted maximum likelihood method to evaluate the impact of potential moderators. RESULTS Meta-analysis of data from 14 treatment arms indicated a significant reduction in plasma FFA concentrations following treatment with statins (weighted mean difference (WMD) -19.42%, 95% CI -23.19, --15.64, P < 0.001). Subgroup analysis confirmed the significance of the effect with both atorvastatin (WMD -20.56%, 95% CI -24.51, -16.61, P < 0.01) and simvastatin (WMD -18.05%, 95% CI -28.12, -7.99, P < 0.001). Changes in plasma FFA concentrations were independent of treatment duration (slope -0.10, 95% CI -0.30, 0.11, P = 0.354) and magnitude of reduction in plasma low density lipoprotein cholesterol concentrations (slope 0.55, 95% CI -0.17, 1.27, P = 0.133) by statins. CONCLUSIONS The results of the present study suggest that statin therapy may lower plasma FFA concentrations. The cardiovascular and metabolic significance of this finding requires further investigation.
Collapse
Affiliation(s)
- Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran.,Metabolic Research Centre, Royal Perth Hospital, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - Luis E Simental-Mendía
- Biomedical Research Unit, Mexican Social Security Institute, Durango.,Universidad Autónoma España de Durango, Durango, Dgo., México
| | - Claudio Pedone
- Area di Geriatria, Università Campus Biomedico di Roma, Via Alvaro del Portillo 21, 00128, Roma
| | - Gianna Ferretti
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche (DISCO), Università Politecnica Delle Marche, Italy
| | - Petr Nachtigal
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University in Prague, Hradec Kralove, Czech Republic
| | - Simona Bo
- Department of Medical Sciences, University of Turin, Turin
| | - Giuseppe Derosa
- Center for the Study of Endocrine-Metabolic Pathophysiology and Clinical Research, University of Pavia, Pavia.,Department of Internal Medicine and Therapeutics, University of Pavia and Fondazione IRCCS Policlinico S. Matteo, Pavia, Pavia, Italy
| | - Pamela Maffioli
- Department of Internal Medicine and Therapeutics, University of Pavia and Fondazione IRCCS Policlinico S. Matteo, Pavia, Pavia, Italy
| | - Gerald F Watts
- Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
18
|
Fritsch M, Koliaki C, Livingstone R, Phielix E, Bierwagen A, Meisinger M, Jelenik T, Strassburger K, Zimmermann S, Brockmann K, Wolff C, Hwang JH, Szendroedi J, Roden M. Time course of postprandial hepatic phosphorus metabolites in lean, obese, and type 2 diabetes patients. Am J Clin Nutr 2015; 102:1051-8. [PMID: 26423389 DOI: 10.3945/ajcn.115.107599] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 08/26/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Impaired energy metabolism is a possible mechanism that contributes to insulin resistance and ectopic fat storage. OBJECTIVE We examined whether meal ingestion differently affects hepatic phosphorus metabolites in insulin-sensitive and insulin-resistant humans. DESIGN Young, lean, insulin-sensitive humans (CONs) [mean ± SD body mass index (BMI; in kg/m(2)): 23.2 ± 1.5]; insulin-resistant, glucose-tolerant, obese humans (OBEs) (BMI: 34.3 ± 1.7); and type 2 diabetes patients (T2Ds) (BMI: 32.0 ± 2.4) were studied (n = 10/group). T2Ds (61 ± 7 y old) were older (P < 0.001) than were OBEs (31 ± 7 y old) and CONs (28 ± 3 y old). We quantified hepatic γATP, inorganic phosphate (Pi), and the fat content [hepatocellular lipids (HCLs)] with the use of (31)P/(1)H magnetic resonance spectroscopy before and at 160 and 240 min after a high-caloric mixed meal. In a subset of volunteers, we measured the skeletal muscle oxidative capacity with the use of high-resolution respirometry. Whole-body insulin sensitivity (M value) was assessed with the use of hyperinsulinemic-euglycemic clamps. RESULTS OBEs and T2Ds were similarly insulin resistant (M value: 3.5 ± 1.4 and 1.9 ± 2.5 mg · kg(-1) · min(-1), respectively; P = 0.9) and had 12-fold (P = 0.01) and 17-fold (P = 0.002) higher HCLs, respectively, than those of lean persons. Despite comparable fasting hepatic γATP concentrations, the maximum postprandial increase of γATP was 6-fold higher in OBEs (0.7 ± 0.2 mmol/L; P = 0.03) but only tended to be higher in T2Ds (0.6 ± 0.2 mmol/L; P = 0.09) than in CONs (0.1 ± 0.1 mmol/L). However, in the fasted state, muscle complex I activity was 53% lower (P = 0.01) in T2Ds but not in OBEs (P = 0.15) than in CONs. CONCLUSIONS Young, obese, nondiabetic humans exhibit augmented postprandial hepatic energy metabolism, whereas elderly T2Ds have impaired fasting muscle energy metabolism. These findings support the concept of a differential and tissue-specific regulation of energy metabolism, which can occur independently of insulin resistance. This trial was registered at clinicaltrials.gov as NCT01229059.
Collapse
Affiliation(s)
- Maria Fritsch
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Department of Pediatric and Adolescent Medicine, Medical University of Vienna, Vienna, Austria; and
| | - Chrysi Koliaki
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Department of Endocrinology and Diabetology, Medical Faculty, and German Center of Diabetes Research, Partner Düsseldorf, Germany
| | - Roshan Livingstone
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research
| | - Esther Phielix
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research
| | - Alessandra Bierwagen
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, German Center of Diabetes Research, Partner Düsseldorf, Germany
| | - Markus Meisinger
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research
| | - Tomas Jelenik
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, German Center of Diabetes Research, Partner Düsseldorf, Germany
| | - Klaus Strassburger
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine University, Düsseldorf, Germany, German Center of Diabetes Research, Partner Düsseldorf, Germany
| | - Stefanie Zimmermann
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, German Center of Diabetes Research, Partner Düsseldorf, Germany
| | - Katharina Brockmann
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, German Center of Diabetes Research, Partner Düsseldorf, Germany
| | - Christina Wolff
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, German Center of Diabetes Research, Partner Düsseldorf, Germany
| | - Jong-Hee Hwang
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, German Center of Diabetes Research, Partner Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Department of Endocrinology and Diabetology, Medical Faculty, and German Center of Diabetes Research, Partner Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Department of Endocrinology and Diabetology, Medical Faculty, and German Center of Diabetes Research, Partner Düsseldorf, Germany
| |
Collapse
|
19
|
Abstract
Diabetes mellitus is accompanied by hormonal and neurochemical changes that can be associated with anxiety and depression. I investigated the antidepressant effect of simvastatin (SMV) on diabetic rats. Rats were divided into control (CTR) and streptozotocin-induced diabetic (STZ) groups and were orally administered 0, 5, or 10 mg/kg of SMV daily for 14 days, then exposed to the forced swimming test (FST). Our results showed that diabetic rats had higher immobility duration than the CTR rats, and SMV decreased this depressive-like behavior in the diabetic rats. However, clomipramine lowered the immobility time in the CTR and STZ rats. STZ decreased serotonin concentration in the hippocampus, which was reversed by SMV and clomipramine. The dopamine concentration in the hippocampus decreased in the STZ groups compared with the CTR groups. However, SMV and clomipramine had no significant effect on the dopamine levels in either the CTR or STZ groups. Corticosterone levels were increased in the untreated STZ group; SMV and clomipramine significantly decreased corticosterone levels in the STZ groups, but had no effect on the CTR groups. In conclusion, SMV exerts an antidepressant-like effect on diabetic rats that are submitted to the FST. The antidepressant-like effect of SMV in the FST appears to be mediated, at least in part, by the biochemical changes to the blood levels of corticosterone and of serotonin concentration in the hippocampus.
Collapse
Affiliation(s)
- Maha Mohamed ElBatsh
- Department of Clinical Pharmacology, Faculty of Medicine, Menoufia University, Shebin El-Kom 32511, Egypt
- Department of Clinical Pharmacology, Faculty of Medicine, Menoufia University, Shebin El-Kom 32511, Egypt
| |
Collapse
|
20
|
Begovatz P, Koliaki C, Weber K, Strassburger K, Nowotny B, Nowotny P, Müssig K, Bunke J, Pacini G, Szendrödi J, Roden M. Pancreatic adipose tissue infiltration, parenchymal steatosis and beta cell function in humans. Diabetologia 2015; 58:1646-55. [PMID: 25740696 DOI: 10.1007/s00125-015-3544-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/04/2015] [Indexed: 12/12/2022]
Abstract
AIMS/HYPOTHESIS This study aimed to perform a comprehensive analysis of interlobular, intralobular and parenchymal pancreatic fat in order to assess their respective effects on beta cell function. METHODS Fifty-six participants (normal glucose tolerance [NGT] (n = 28), impaired fasting glucose (IFG) and/or impaired glucose tolerance (IGT) (n = 14) and patients with type 2 diabetes (n = 14)) underwent a frequent-sampling OGTT and non-invasive magnetic resonance imaging (MRI; whole-body and pancreatic) and proton magnetic resonance spectroscopy ((1)H-MRS; liver and pancreatic fat). Total pancreatic fat was assessed by a standard 2 cm(3) (1)H-MRS method, intralobular fat by 1 cm(3) (1)H-MRS that avoided interlobular fat within modified DIXON (mDIXON) water images, and parenchymal fat by a validated mDIXON-MRI fat-fraction method. RESULTS Comparison of (1)H-MRS techniques revealed an inhomogeneous distribution of interlobular and intralobular adipose tissue, which increased with decreasing glucose tolerance. mDIXON-MRI measurements provided evidence against uniform steatosis, revealing regions of parenchymal tissue void of lipid accumulation in all participants. Total (r = 0.385, p < 0.01) and intralobular pancreas adipose tissue infiltration (r = 0.310, p < 0.05) positively associated with age, but not with fasting or 2 h glucose levels, BMI or visceral fat content (all p > 0.5). Furthermore, no associations were found between total and intralobular pancreatic adipose tissue infiltration and insulin secretion or beta cell function within NGT, IFG/IGT or patients with type 2 diabetes (all p > 0.2). CONCLUSIONS/INTERPRETATION The pancreas does not appear to be another target organ for abnormal endocrine function because of ectopic parenchymal fat storage. No relationship was found between pancreatic adipose tissue infiltration and beta cell function, regardless of glucose tolerance status.
Collapse
Affiliation(s)
- Paul Begovatz
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Auf`m Hennekamp 65, 40225, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Cederberg H, Stančáková A, Yaluri N, Modi S, Kuusisto J, Laakso M. Increased risk of diabetes with statin treatment is associated with impaired insulin sensitivity and insulin secretion: a 6 year follow-up study of the METSIM cohort. Diabetologia 2015; 58:1109-17. [PMID: 25754552 DOI: 10.1007/s00125-015-3528-5] [Citation(s) in RCA: 226] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/30/2015] [Indexed: 01/14/2023]
Abstract
AIMS/HYPOTHESIS The aim of this work was to investigate the mechanisms underlying the risk of type 2 diabetes associated with statin treatment in the population-based Metabolic Syndrome in Men (METSIM) cohort. METHODS A total of 8,749 non-diabetic participants, aged 45-73 years, were followed up for 5.9 years. New diabetes was diagnosed in 625 men by means of an OGTT, HbA1c ≥6.5% (48 mmol/mol) or glucose-lowering medication started during the follow-up. Insulin sensitivity and secretion were evaluated with OGTT-derived indices. RESULTS Participants on statin treatment (N = 2,142) had a 46% increased risk of type 2 diabetes (adjusted HR 1.46 [95% CI 1.22, 1.74]). The risk was dose dependent for simvastatin and atorvastatin. Statin treatment significantly increased 2 h glucose (2hPG) and glucose AUC of an OGTT at follow-up, with a nominally significant increase in fasting plasma glucose (FPG). Insulin sensitivity was decreased by 24% and insulin secretion by 12% in individuals on statin treatment (at FPG and 2hPG <5.0 mmol/l) compared with individuals without statin treatment (p < 0.01). Decreases in insulin sensitivity and insulin secretion were dose dependent for simvastatin and atorvastatin. CONCLUSIONS/INTERPRETATION Statin treatment increased the risk of type 2 diabetes by 46%, attributable to decreases in insulin sensitivity and insulin secretion.
Collapse
Affiliation(s)
- Henna Cederberg
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, P.O. Box 1627, 70211, Kuopio, Finland
| | | | | | | | | | | |
Collapse
|
22
|
Park ZH, Juska A, Dyakov D, Patel RV. Statin-associated incident diabetes: a literature review. ACTA ACUST UNITED AC 2015; 29:317-34. [PMID: 24849689 DOI: 10.4140/tcp.n.2014.317] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To evaluate available evidence for incident diabetes associated with statin use and offer some practical management considerations. DATA SOURCES A literature search was performed using MEDLINE from 2000 to October 2013. The following MESH terms and text key words alone or in combination were included: 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, HMG-CoA reductase inhibitors, statins, incident diabetes, new-onset diabetes, insulin resistance, impaired insulin secretion, meta-analysis, cohort study, and observational study. STUDY SELECTION Analyzed studies were published in English and investigated incident diabetes associated with statin use. DATA EXTRACTION Author consensus determined study inclusion in this review, focusing on observational studies and meta-analyses. DATA SYNTHESIS Since the report of incident diabetes associated with rosuvastatin, an unexpected finding in the Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin, safety concerns with statins have emerged. Results of observational studies and meta-analyses show association of incident diabetes with statin use in patients with concomitant risk factors for diabetes. A pharmacodynamic mechanism has yet to be delineated, and individual statins may behave differently. Whether cardiovascular (CV) risk will increase with statin-associated incident diabetes remains unclear. CONCLUSION Review of current, available clinical data suggest a possible association between statin use and incident diabetes in patients with underlying diabetes risk factors. Although study data may be insufficient to change the current practice paradigm, clinicians should vigilantly monitor for incident diabetes in patients on statins. Patients with a low risk of CV disease and high risk of diabetes should reconsider statin use and focus on lifestyle management.
Collapse
Affiliation(s)
- Zoon H Park
- Department of Pharmacy Services, Swedish Covenant Hospital, Chicago, Illinois
| | | | | | - Ramesh V Patel
- Pharmacy Services and Clinical Research, Swedish Covenant Hospital
| |
Collapse
|
23
|
Winhofer Y, Wolf P, Krššák M, Wolfsberger S, Tura A, Pacini G, Gessl A, Raber W, Kukurova IJ, Kautzky-Willer A, Knosp E, Trattnig S, Krebs M, Luger A. No evidence of ectopic lipid accumulation in the pathophysiology of the acromegalic cardiomyopathy. J Clin Endocrinol Metab 2014; 99:4299-306. [PMID: 25148232 DOI: 10.1210/jc.2014-2242] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT PATIENTS with acromegaly frequently display disturbances of glucose and lipid metabolism, which might contribute to their increased cardiovascular risk. Because insulin resistance and increased lipolysis have been linked to ectopic lipid deposition, altered lipid accumulation in the liver and the myocardium might contribute to metabolic and cardiac complications in these patients. OBJECTIVE The aim of this study was to investigate myocardial (MYCL) and hepatic lipid content (HCL), insulin sensitivity, and cardiac function in active acromegaly and after control of GH excess through transsphenoidal surgery. PATIENTS Ten patients with newly diagnosed acromegaly (ACRO_active) were compared with 12 healthy controls (CON), matched for age, body mass index, and gender. In seven patients GH excess was controlled, and they were compared with their active state. METHODS MYCL and HCL were assessed by (1)H-magnetic resonance spectroscopy, pericardial fat and cardiac function by (1)H-magnetic resonance imaging, and insulin sensitivity and secretion by an oral glucose tolerance test. RESULTS Although MYCL tended to be lower, HCL was significantly lower in ACRO_active compared with CON (HCL: 1.2% ± 1.2% vs 4.3% ± 3.5% of (1)H-magnetic resonance spectroscopy signal, P < .02). Parameters of systolic function and hypertrophy were significantly increased in ACRO_active compared with CON, as were insulin secretion and resistance. After the control of GH excess, HCL and MYCL remained unchanged, but pericardial fat was increased in the patients in whom GH excess was controlled (from 11.6 ± 5.5 to 14.7 ± 6.2 cm(2), P = .02). CONCLUSION Acromegaly represents a unique condition characterized by low myocardial and hepatic lipid content despite decreased insulin sensitivity, hyperinsulinemia, and hyperglycemia. Hence, ectopic lipid accumulation does not appear to contribute to cardiac morbidity, and increased lipid oxidation might counteract ectopic lipid accumulation in GH excess.
Collapse
Affiliation(s)
- Yvonne Winhofer
- Department of Internal Medicine III, Division of Endocrinology and Metabolism (Y.W., P.W., M.Krš., A.G., W.R., A.K.-W., M.Kre., A.L.), Department of Biomedical Imaging and Image Guided therapy, Centre of Excellence High-field MR (M.Krš., I.J.K., S.T.), and Department of Neurosurgery (S.W., E.K.), Medical University of Vienna, A-1090 Vienna, Austria; and Institute of Biomedical Engineering (A.T., G.P.), National Research Council, Metabolic Unit, I-35127 Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kahl S, Nowotny B, Piepel S, Nowotny PJ, Strassburger K, Herder C, Pacini G, Roden M. Estimates of insulin sensitivity from the intravenous-glucose-modified-clamp test depend on suppression of lipolysis in type 2 diabetes: a randomised controlled trial. Diabetologia 2014; 57:2094-102. [PMID: 25047649 DOI: 10.1007/s00125-014-3328-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/23/2014] [Indexed: 11/30/2022]
Abstract
AIMS/HYPOTHESIS The combined IVGTT-hyperinsulinaemic-euglycaemic clamp (Botnia clamp) allows the assessment of insulin secretion and sensitivity in one experiment. It remains unclear whether this clamp yields results comparable with those of the standard hyperinsulinaemic-euglycaemic clamp (SHEC) in diabetes patients. We hypothesised that the IVGTT induces responses affecting insulin sensitivity assessment. METHODS Of 22 randomised diet- or metformin-treated patients with well-controlled type 2 diabetes, 19 randomly underwent a Botnia clamp and an SHEC, spaced by 2 weeks, in one clinical research centre in a crossover study. The main outcomes were whole-body and hepatic insulin sensitivity as measured by the clamp and [6,6-(2)H2]glucose. Substrate utilisation was assessed from indirect calorimetry and beta cell function from insulin dynamics during IVGTT. RESULTS The values of whole-body insulin sensitivity obtained from Botnia clamp and SHEC were correlated (r = 0.87, p < 0.001), but also revealed intra-individual variations. Hepatic insulin sensitivity did not differ between experiments during the clamp, but differed after IVGTT. The contribution of glucose oxidation to glucose disposal increased by 2.2 ± 0.3 and 1.2 ± 0.4 mg kg fat-free mass (FFM)(-1) min(-1) (Botnia and SHEC, p < 0.05), whereas lipid oxidation decreased by 0.8 ± 0.1 and 0.4 ± 0.1 mg kg FFM(-1) min(-1) (p < 0.05) from baseline. Differences in NEFA (r = -0.60, p < 0.01), but not C-peptide (r = -0.16, p = 0.52) or hepatic insulin sensitivity between IVGTT and placebo before the clamps correlated with individual variations of insulin sensitivity. CONCLUSIONS/INTERPRETATION The Botnia clamp provides similar estimates of insulin sensitivity as SHEC in patients with type 2 diabetes, but changes in NEFA during IVGTT may affect insulin sensitivity and thereby the discrimination between insulin-sensitive and insulin-resistant individuals. TRIAL REGISTRATION ClinicalTrials.gov NCT01397279 FUNDING: The study was funded by the Ministry of Science and Research of the State of North Rhine-Westphalia and the German Federal Ministry of Health, and supported in part by grants from the Federal Ministry for Research to the Centers for Diabetes Research, Helmholtz Alliance Imaging and Curing Environmental Metabolic Diseases and the Schmutzler-Stiftung.
Collapse
Affiliation(s)
- Sabine Kahl
- Department of Endocrinology and Diabetology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Goyal A, Singh S, Tandon N, Gupta N, Gupta YK. Effect of atorvastatin on pancreatic Beta-cell function and insulin resistance in type 2 diabetes mellitus patients: a randomized pilot study. Can J Diabetes 2014; 38:466-72. [PMID: 24933106 DOI: 10.1016/j.jcjd.2014.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/17/2013] [Accepted: 01/13/2014] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Statins are commonly used for the management of dyslipidemia in type 2 diabetes mellitus patients. We hypothesized that atorvastatin could modulate the beta-cell function by altering the levels of proapoptotic and antiapoptotic lipoproteins and could also have an effect on insulin resistance. The aim of the present pilot study was to assess the effect of atorvastatin 10 mg on pancreatic beta-cell function and insulin resistance in patients with hyperlipidemia and type 2 diabetes by using the homeostasis model assessment-2 (HOMA2) index. METHODS Fifty-one type 2 diabetes patients receiving oral antidiabetes drugs, not taking statins, with baseline low-density lipoprotein cholesterol between 2.6 mmol/L and 4.1 mmol/L were included. Forty-three patients (21 in placebo group and 22 in atorvastatin group) completed the study and were taken up for final analysis. Fasting blood samples were obtained at baseline and at 12 weeks to determine levels of blood glucose, lipid profile, insulin, C-peptide and glycosylated hemoglobin (A1C). RESULTS Atorvastatin nonsignificantly increased fasting serum insulin (+14.29%, p=0.18), accompanied by marginal nonsignificant increases in fasting plasma glucose and A1C. There was a decrease in HOMA2 percent beta-cell function (-2.9%, p=0.72) and increase in HOMA2 insulin resistance (+14%, p=0.16) in the atorvastatin group as compared with baseline, but the difference was not statistically significant. CONCLUSIONS Atorvastatin in the dose used failed to produce significant change in pancreatic beta-cell function and insulin resistance in type 2 diabetes patients as assessed by the HOMA2 index. The possible explanations include absence of lipotoxicity at prevailing levels of dyslipidemia at baseline or inadequacy of statin dose used in the study. (Clinical Trials Registry-India: CTRI/2008/091/000099).
Collapse
Affiliation(s)
- Aman Goyal
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Surender Singh
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India.
| | - Nikhil Tandon
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Nandita Gupta
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Yogendra Kumar Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
26
|
Black RNA, Ennis CN, Young IS, Hunter SJ, Atkinson AB, Bell PM. The peroxisome proliferator-activated receptor alpha agonist fenofibrate has no effect on insulin sensitivity compared to atorvastatin in type 2 diabetes mellitus; a randomised, double-blind controlled trial. J Diabetes Complications 2014; 28:323-7. [PMID: 24560135 DOI: 10.1016/j.jdiacomp.2014.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/02/2013] [Accepted: 01/02/2014] [Indexed: 11/28/2022]
Abstract
AIMS Assess insulin sensitivity after treatment with a selective PPAR-alpha agonist compared to an HMG CoA reductase inhibitor in human subjects with type 2 diabetes mellitus. METHODS Thirteen subjects with Type 2 diabetes mellitus were studied in a double-blind crossover design with 4-week placebo run-in and washout and 12-week treatment periods, randomised to micronised fenofibrate 267 mg or atorvastatin 10mg daily followed by the alternate drug in the second period. Insulin resistance was measured using the isoglycaemic hyperinsulinaemic clamp method with isotope dilution. RESULTS Weight, physical activity and other medications did not change. Total cholesterol (mean +/- standard error) was 4.60+/-0.21 versus 3.9+/-0.22 mmol/L after fenofibrate and atorvastatin respectively, p<0.05. LDL was 2.70+/-0.19 versus 1.95+/-0.23 mmol/L, p<0.05 and triglyceride 1.64+/-0.23 versus 1.84+/-0.26 mmol/L, p<0.05. Insulin-stimulated whole-body glucose disposal (35.4+/-3.1 versus 33.2+/-3.0 μmol/kg/min) and nadir endogenous glucose production (6.2+/-1.4 versus 7.0+/-1.1 μmol/kg/min) revealed no significant differences in effects of the treatments. CONCLUSIONS In human subjects with Type 2 diabetes mellitus there were characteristic differences in lipid profile changes but no difference in insulin sensitivity after treatment with micronised fenofibrate compared to atorvastatin. This study finds no evidence of increased insulin sensitivity using this selective PPAR-alpha agonist over a commonly used statin at these doses.
Collapse
Affiliation(s)
- R Neil A Black
- Regional Centre for Endocrinology & Diabetes, Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK.
| | - Cieran N Ennis
- Regional Centre for Endocrinology & Diabetes, Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK
| | - Ian S Young
- Nutrition and Metabolism Group, The Queen's University of Belfast, Belfast, UK
| | - Steven J Hunter
- Regional Centre for Endocrinology & Diabetes, Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK
| | - A Brew Atkinson
- Regional Centre for Endocrinology & Diabetes, Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK
| | - Patrick M Bell
- Regional Centre for Endocrinology & Diabetes, Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK
| |
Collapse
|
27
|
Winhofer Y, Krššák M, Wolf P, Tura A, Anderwald CH, Kosi L, Reiter G, Pacini G, Trattnig S, Luger A, Krebs M, Kautzky-Willer A. Hepatic rather than cardiac steatosis relates to glucose intolerance in women with prior gestational diabetes. PLoS One 2014; 9:e91607. [PMID: 24621572 PMCID: PMC3951459 DOI: 10.1371/journal.pone.0091607] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/13/2014] [Indexed: 11/19/2022] Open
Abstract
Background Increased myocardial lipid accumulation has been described in patients with pre- and overt type 2 diabetes and could underlie the development of left-ventricular dysfunction in metabolic diseases (diabetic cardiomyopathy). Since women with prior gestational diabetes (pGDM) display a generally young population at high risk of developing diabetes and associated cardiovascular complications, we aimed to assess whether myocardial lipid accumulation can be detected at early stages of glucose intolerance and relates to markers of hepatic steatosis (Fatty Liver Index), cardiac function, insulin sensitivity and secretion. Methods Myocardial lipid content (MYCL), left-ventricular function (1H-magnetic-resonance-spectroscopy and -imaging), insulin sensitivity/secretion (oral glucose tolerance test) and the fatty liver index (FLI) were assessed in 35 pGDM (45.6±7.0 years, 28.3±4.8 kg/m2) and 14 healthy control females (CON; 44.7±9.8 years, 26.1±2.5 kg/m2), matching for age and body-mass-index (each p>0.1). Results Of 35 pGDM, 9 displayed normal glucose tolerance (NGT), 6 impaired glucose regulation (IGR) and 20 had been already diagnosed with type 2 diabetes (T2DM). MYCL and cardiac function were comparable between pGDM and CON; in addition, no evidence of left-ventricular dysfunction was observed. MYCL was inversely correlated with the ejection fraction in T2DM (R = −0.45, p<0.05), while the FLI was tightly correlated with metabolic parameters (such as HbA1C, fasting plasma glucose and HDL-cholesterol) and rose along GT-groups. Conclusions There is no evidence of cardiac steatosis in middle-aged women with prior gestational diabetes, suggesting that cardiac complications might develop later in the time-course of diabetes and may be accelerated by the co-existence of further risk factors, whereas hepatic steatosis remains a valid biomarker for metabolic diseases even in this rather young female cohort.
Collapse
Affiliation(s)
- Yvonne Winhofer
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
- * E-mail:
| | - Martin Krššák
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Peter Wolf
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Andrea Tura
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council, Padova, Italy
| | - Christian-Heinz Anderwald
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council, Padova, Italy
- Mariahilf Community Pharmacy, Arnoldstein, Austria
- Medical Direction, Specialized Hospital Complex Agathenhof, Micheldorf, Austria
| | - Lana Kosi
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | | | - Giovanni Pacini
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council, Padova, Italy
| | - Siegfried Trattnig
- Centre of Excellence, High-Field MR, Department of Radiodiagnostics, Medical University of Vienna, Vienna, Austria
| | - Anton Luger
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Michael Krebs
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Alexandra Kautzky-Willer
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Muscogiuri G, Sarno G, Gastaldelli A, Savastano S, Ascione A, Colao A, Orio F. The good and bad effects of statins on insulin sensitivity and secretion. Endocr Res 2014; 39:137-43. [PMID: 25208056 DOI: 10.3109/07435800.2014.952018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
AIMS Statins are the main lipid-lowering treatment in both primary and secondary prevention populations. Whether statins deteriorates glycemic control, predisposing to the onset of diabetes mellitus has been a matter of recent concern. Statins may accelerate progression to diabetes via molecular mechanisms that impact insulin sensitivity and secretion. In this review, we debate the relative effect of statins in driving insulin resistance and the impairment of insulin secretion. METHODS Narrative overview of the literature synthesizing the findings of literature was retrieved from searches of computerized databases, hand searches, and authoritative texts employing the key words "Statins", "Randomized Clinical Trial", "Insulin sensitivity", "Insulin resistance", "Insulin Secretion", "Diabetes Mellitus" alone and/or in combination. RESULTS The weight of clinical evidence suggests a worsening effect of statins on insulin resistance and secretion, anyway basic science studies did not find a clear molecular explanation, providing conflicting evidence regarding both the beneficial and the adverse effects of statin therapy on insulin sensitivity. CONCLUSIONS Although most of the clinical studies suggest a worsening of insulin resistance and secretion, the cardiovascular benefits of statin therapy outweigh the risk of developing insulin resistance, thus the data suggest the need to treat dyslipidemia and to make patients aware of the possible risk of developing type 2 diabetes or, if they already are diabetic, of worsening their metabolic control.
Collapse
Affiliation(s)
- Giovanna Muscogiuri
- Department of Clinical Medicine and Surgery, University "Federico II" , Naples , Italy
| | | | | | | | | | | | | |
Collapse
|
29
|
Zhou Y, Yuan Y, Cai RR, Huang Y, Xia WQ, Yang Y, Wang P, Wei Q, Wang SH. Statin therapy on glycaemic control in type 2 diabetes: a meta-analysis. Expert Opin Pharmacother 2013; 14:1575-84. [PMID: 23826679 DOI: 10.1517/14656566.2013.810210] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Randomised controlled trials (RCTs) indicate that statin therapy has cardiovascular benefit among patients with type 2 diabetes. Recently, statins were reported to increase risk of diabetes by 9%. The aim was to investigate by a meta-analysis whether statins deteriorate glycaemic control in type 2 diabetes. METHODS Medline, EMBASE and Cochrane Central Register of Controlled Trials from 1966 to 2012 were searched for RCTs of statins. Included were only trials with type 2 diabetes. MAIN OUTCOME MEASURES The I(2) statistic was used to measure heterogeneity between trials and calculated mean differences for glycaemic parameters with random-effect meta-analysis. RESULTS 26 eligible studies were identified with 3232 participants. Statin therapy had no remarkable influence on HbA1c (WMD 0.04%, 95% CI -0.08 to 0.16, I² = 45.7%, n = 3070), FPG (2.25 mg/dl, 95% CI -3.50 to 7.99, I² = 46%, n = 1176), BMI, fasting insulin or HOMA-IR. However, subgroup analysis showed significant, detrimental effect of atorvastatin on HbA1c, whereas simvastatin presented an ameliorative effect. Meta-regression presented that neither baseline age nor relative reduction in LDL-cholesterol concentrations accounted for residual heterogeneity. CONCLUSION Statin therapy showed non-significant effect on glycaemic control in type 2 diabetes. Statin therapy need not change among them with moderate or high cardiovascular risk or existing cardiovascular disease.
Collapse
Affiliation(s)
- Yi Zhou
- The Affiliated ZhongDa Hospital of Southeast University, Department of Endocrinology, No. 87 DingJiaQiao Road, Nanjing, 210009, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wang L, Duan G, Lu Y, Pang S, Huang X, Jiang Q, Dang N. The effect of simvastatin on glucose homeostasis in streptozotocin induced type 2 diabetic rats. J Diabetes Res 2013; 2013:274986. [PMID: 23671864 PMCID: PMC3647597 DOI: 10.1155/2013/274986] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 01/01/2013] [Accepted: 01/21/2013] [Indexed: 01/21/2023] Open
Abstract
Objective. To investigate the effect of simvastatin on glucose homeostasis in streptozotocin induced type 2 diabetic rats. Methods. Forty male Wistar rats were randomly divided into four groups. Normal control rats were fed with standard diet, others were fed with high-fat diet. Diabetic rats were induced by a single intraperitoneal injection of STZ. The simvastatin intervention rats were fed with simvastatin during the experiment process, and the simvastatin treatment rats were fed with simvastatin after diabetes rats were induced. We measured body weight, fasting plasma glucose, cholesterol, high-density lipoprotein cholesterol, and triglyceride after an overnight fast. Results. The FPG was higher in diabetic rats when compared to normal control ones; the simvastatin intervention rats had a higher FPG compared to the diabetic rats and were more easily be induced to diabetes at the end of 4 weeks, FPG level of simvastatin treatment rats was increased compared with diabetic model rats after 12 weeks. Conclusion. These data indicate that simvastatin intervention rats may cause hyperglycemia by impairing the function of islet β cells and have an adverse effect on glucose homeostasis, especially on FPG level.
Collapse
Affiliation(s)
- Lulu Wang
- School of Medicine, Shandong University, Jinan, Shandong 250013, China
- Department of Medicine, Jinan Central Hospital Shandong University, Jinan, Shandong 250013, China
| | - Guanglan Duan
- Department of Medicine, Jinan Central Hospital Shandong University, Jinan, Shandong 250013, China
| | - Yong Lu
- Department of Medicine, Jinan Central Hospital Shandong University, Jinan, Shandong 250013, China
| | - Shuguang Pang
- School of Medicine, Shandong University, Jinan, Shandong 250013, China
- Department of Medicine, Jinan Central Hospital Shandong University, Jinan, Shandong 250013, China
- *Shuguang Pang:
| | - Xianping Huang
- Department of Medicine, Jinan Central Hospital Shandong University, Jinan, Shandong 250013, China
| | - Qiang Jiang
- Department of Medicine, Jinan Central Hospital Shandong University, Jinan, Shandong 250013, China
| | - Ningning Dang
- Department of Medicine, Jinan Central Hospital Shandong University, Jinan, Shandong 250013, China
| |
Collapse
|
31
|
Abstract
Multiple clinical trials have demonstrated the beneficial effects of statins in lowering levels of low-density lipoprotein and in the prevention of cardiovascular disease. However, although statins have a good safety profile, a debate has been ongoing as to whether use of statins increases risk of new-onset diabetes. Recent large scale meta-analyses of statin trials support the concept of a diabetogenic effect of statins, as do some other small trials assessing changes in glycaemia parameters and insulin levels. However, a definitive mechanism of action has not yet been elucidated. Nevertheless, the level of evidence has been sufficient to lead the FDA to make a change to the labelling of statins. This review assesses the current available evidence and offers a clinical perspective.
Collapse
|
32
|
Bellia A, Rizza S, Lombardo MF, Donadel G, Fabiano R, Andreadi K, Quon MJ, Sbraccia P, Federici M, Tesauro M, Cardillo C, Lauro D. Deterioration of glucose homeostasis in type 2 diabetic patients one year after beginning of statins therapy. Atherosclerosis 2012; 223:197-203. [DOI: 10.1016/j.atherosclerosis.2012.04.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 03/23/2012] [Accepted: 04/15/2012] [Indexed: 12/16/2022]
|
33
|
Cholesterol synthesis is associated with hepatic lipid content and dependent on fructose/glucose intake in healthy humans. EXPERIMENTAL DIABETES RESEARCH 2011; 2012:361863. [PMID: 22203835 PMCID: PMC3235776 DOI: 10.1155/2012/361863] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 09/08/2011] [Indexed: 01/01/2023]
Abstract
Visceral obesity and fatty liver have been related to high synthesis and low absorption of cholesterol. This study aimed to investigate the associations of cholesterol metabolism with liver and visceral fat content in healthy humans. Another objective was to explore the effects of very-high-fructose and very-high-glucose diets on cholesterol homeostasis. We report on a cohort of 20 people (12 males, 8 females; age 30.5 ± 2.0 years; body mass index 25.9 ± 0.5 kg/m2) who completed a four-week dietary intervention study. Between the baseline and the followup examination the study participants in addition to a balanced weight-maintaining diet received 150 g of either fructose or glucose per day. Visceral and liver fat were measured with magnetic resonance (MR) imaging and 1H-MR spectroscopy, respectively. Cholesterol absorption and synthesis were estimated from the serum noncholesterol sterol concentrations. Performing cross-sectional analyses the lanosterol and desmosterol to cholesterol ratios were positively correlated with visceral and liver fat content (all P < .03). The lathosterol to cholesterol ratio decreased in response to high-fructose diet (P = .006) but not in response to high-glucose diet. To conclude, visceral and liver fat content are associated with cholesterol synthesis in healthy humans. Furthermore, cholesterol synthesis appears to be dependent on fructose/glucose intake.
Collapse
|
34
|
Maurantonio M, Ballestri S, Odoardi MR, Lonardo A, Loria P. Treatment of atherogenic liver based on the pathogenesis of nonalcoholic fatty liver disease: a novel approach to reduce cardiovascular risk? Arch Med Res 2011; 42:337-353. [PMID: 21843565 DOI: 10.1016/j.arcmed.2011.08.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 07/18/2011] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), which spans a spectrum of conditions ranging from simple steatosis to progressive nonalcoholic steatohepatitis (NASH), is the most common chronic liver disease and a relevant public health issue. The prevalence of NAFLD depends on adiposity, age, gender and ethnicity. The natural history of liver disease in those with NAFLD critically depends on liver histological changes. However, cardiovascular mortality is increased in NAFLD, particularly in middle-aged adults. Against such a background, this review consists of three sections. First, data on NAFLD as a novel mechanism of increased cardiovascular risk via hyperinsulinism, pro-thrombotic potential, and subclinical inflammation are summarized. Next, the role of atherogenic liver in the development of manifestations of oxidative stress and atherosclerosis is emphasized. Finally, whether and how treating NAFLD will mechanistically result in reduced cardiovascular risk through ameliorated metabolic syndrome is discussed.
Collapse
Affiliation(s)
- Mauro Maurantonio
- Department of Internal Medicine, Endocrinology, Metabolism and Geriatrics, Nuovo Ospedale Civile Sant'Agostino-Estense di Modena, University of Modena and Reggio Emilia, Modena, Italy.
| | | | | | | | | |
Collapse
|
35
|
Anderwald C, Tura A, Grassi A, Krebs M, Szendroedi J, Roden M, Bischof MG, Luger A, Pacini G. Insulin infusion during normoglycemia modulates insulin secretion according to whole-body insulin sensitivity. Diabetes Care 2011; 34:437-41. [PMID: 21216852 PMCID: PMC3024363 DOI: 10.2337/dc10-1137] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Glucose is the major stimulus for insulin release. Time course and amount of insulin secreted after glycemic stimulus are different between type 2 diabetes mellitus (T2DM) patients and healthy subjects. In rodents, it was demonstrated that insulin can modulate its own release. Previous studies in humans yielded contrasting results: Insulin was shown to have an enhancing effect, no effect, or a suppressive effect on its own secretion. Thus, we aimed to evaluate short-term effects of human insulin infusion on insulin secretion during normoglycemia in healthy humans and T2DM subjects of both sex. RESEARCH DESIGN AND METHODS Hyperinsulinemic-isoglycemic clamps with whole-body insulin-sensitivity (M) and C-peptide measurements for insulin secretion modeling were performed in 65 insulin-sensitive (IS) subjects (45 ± 1 year, BMI: 24.8 ± 0.5 kg/m(2)), 17 insulin-resistant (IR) subjects (46 ± 2 years, 28.1 ± 1.3 kg/m(2)), and 20 T2DM patients (56 ± 2 years, 28.0 ± 0.8 kg/m(2); HbA(1c) = 6.7 ± 0.1%). RESULTS IS subjects (M = 8.8 ± 0.3 mg · min(-1) · kg(-1)) had higher (P < 0.00001) whole-body insulin sensitivity than IR subjects (M = 4.0 ± 0.2) and T2DM patients (M = 4.3 ± 0.5). Insulin secretion profiles during clamp were different (P < 0.00001) among the groups, increasing in IS subjects (slope: 0.56 ± 0.11 pmol/min(2)) but declining in IR (-0.41 ± 0.14) and T2DM (-0.87 ± 0.12, P < 0.00002 IR and T2DM vs. IS) subjects. Insulin secretion changes during clamp directly correlated with M (r = 0.6, P < 0.00001). CONCLUSIONS Insulin release during normoglycemia can be modulated by exogenous insulin infusion and directly depends on whole-body insulin sensitivity. Thus, in highly sensitive subjects, insulin increases its own secretion. On the other hand, a suppressive effect of insulin on its own secretion occurs in IR and T2DM subjects.
Collapse
Affiliation(s)
- Christian Anderwald
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council, Padova, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
The use of statins potentiates the insulin-sensitizing effect of exercise training in obese males with and without Type 2 diabetes. Clin Sci (Lond) 2010; 119:293-301. [PMID: 20465545 DOI: 10.1042/cs20100153] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Exercise training is advocated in insulin resistance and statins are used to treat hyperlipidaemia, two cardiometabolic risk factors often presenting concurrently. Statin intake may blunt mitochondrial function and the adaptive response to exercise training. Thus combining exercise training with statin administration may have adverse effects. We examined whether improvements in cardiometabolic risk factors, insulin sensitivity and mitochondrial function mediated by progressive exercise training are affected by statin use. A group of 14 obese elderly males on statins (ST) and 22 matched control subjects (C) were examined. Results on in vivo mitochondrial function [MRS (magnetic resonance spectroscopy)], mitochondrial density (Western blotting), insulin sensitivity (clamp) and metabolic flexibility (indirect calorimetry) were compared before and after a 12-week combined progressive exercise training programme (3 x per week; 45 min per session). Except for LDL (low-density lipoprotein) cholesterol, all pre-training values were comparable between statin users and control subjects. In vivo mitochondrial function and mitochondrial density improved by training in both groups. Interestingly, blood-lipid profile, insulin sensitivity (+72%), non-oxidative and oxidative glucose disposal (+38% and +112%) and insulin-mediated suppression of fat oxidation (-62%) improved only in the ST group. We conclude that statin treatment did not impede exercise performance or tolerance, mitochondrial function or mass. In addition, training-induced improvements in glucose homoeostasis were preserved in the ST group. Strikingly, the insulin-sensitizing effect of training was more prominent in the ST group than in the C group. The combined prescription of statins along with exercise training is safe and should be considered for subjects prone to develop insulin resistance.
Collapse
|
37
|
Jones PH, Cusi K, Davidson MH, Kelly MT, Setze CM, Thakker K, Sleep DJ, Stolzenbach JC. Efficacy and safety of fenofibric acid co-administered with low- or moderate-dose statin in patients with mixed dyslipidemia and type 2 diabetes mellitus: results of a pooled subgroup analysis from three randomized, controlled, double-blind trials. Am J Cardiovasc Drugs 2010; 10:73-84. [PMID: 20136164 DOI: 10.2165/10061630-000000000-00000] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND Monotherapy with lipid-modifying medication is frequently insufficient to normalize lipid abnormalities in patients with mixed dyslipidemia and type 2 diabetes mellitus. OBJECTIVE To evaluate the efficacy and safety of fenofibric acid + statin combination therapy in this population. STUDY DESIGN A pooled, subgroup analysis of three randomized, controlled, double-blind, 12-week trials. SETTING Multiple clinical research facilities in the US and Canada. PATIENTS Patients with mixed dyslipidemia and type 2 diabetes (n = 586). INTERVENTION Fenofibric acid (Trilipix) 135 mg monotherapy; low-, moderate-, or high-dose statin monotherapy (rosuvastatin [Crestor] 10, 20, or 40 mg; simvastatin [Zocor] 20, 40, or 80 mg; or atorvastatin [Lipitor] 20, 40, or 80 mg); or fenofibric acid + low- or moderate-dose statin. MAIN OUTCOME MEASURE Mean percentage changes in lipid parameters, percentages of patients achieving optimal serum lipid/apolipoprotein levels, and incidence of adverse events. RESULTS Fenofibric acid + low-dose statin resulted in significantly (p < 0.001) greater mean percentage changes in high-density lipoprotein cholesterol (HDL-C) [16.8%] and triglycerides (-43.9%) than low-dose statin monotherapy (4.7% and -18.1%, respectively) and significantly (p < 0.001) greater reductions in low-density lipoprotein cholesterol (LDL-C) [-34.0%] than fenofibric acid monotherapy (-5.3%). Similarly, fenofibric acid + moderate-dose statin resulted in significantly (p < or = 0.011) greater mean percentage changes in HDL-C (16.3%) and triglycerides (-43.4%) than moderate-dose statin monotherapy (8.7% and -24.2%, respectively) and significantly (p < 0.001) greater reductions in LDL-C (-32.6%) than fenofibric acid monotherapy (-5.3%). Compared with low- or moderate-dose statin, fenofibric acid + low- or moderate-dose statin resulted in over 5-fold higher percentages of patients achieving optimal levels of LDL-C, non-HDL-C, apolipoprotein B, HDL-C, and triglycerides simultaneously. Incidence of adverse events was generally similar among treatments. CONCLUSION Fenofibric acid + statin combination therapy in patients with mixed dyslipidemia and type 2 diabetes was well tolerated and resulted in more comprehensive improvement in the lipid/apolipoprotein profile than either monotherapy. [Clinical trials are registered at www.clinicaltrials.gov: NCT00300482, NCT00300456, and NCT00300469].
Collapse
|
38
|
Bellia A, Rizza S, Galli A, Fabiano R, Donadel G, Lombardo MF, Cardillo C, Sbraccia P, Tesauro M, Lauro D. Early vascular and metabolic effects of rosuvastatin compared with simvastatin in patients with type 2 diabetes. Atherosclerosis 2010; 210:199-201. [DOI: 10.1016/j.atherosclerosis.2009.11.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 11/10/2009] [Accepted: 11/10/2009] [Indexed: 11/22/2022]
|
39
|
Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM, de Craen AJM, Seshasai SRK, McMurray JJ, Freeman DJ, Jukema JW, Macfarlane PW, Packard CJ, Stott DJ, Westendorp RG, Shepherd J, Davis BR, Pressel SL, Marchioli R, Marfisi RM, Maggioni AP, Tavazzi L, Tognoni G, Kjekshus J, Pedersen TR, Cook TJ, Gotto AM, Clearfield MB, Downs JR, Nakamura H, Ohashi Y, Mizuno K, Ray KK, Ford I. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 2010; 375:735-42. [PMID: 20167359 DOI: 10.1016/s0140-6736(09)61965-6] [Citation(s) in RCA: 1732] [Impact Index Per Article: 115.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Trials of statin therapy have had conflicting findings on the risk of development of diabetes mellitus in patients given statins. We aimed to establish by a meta-analysis of published and unpublished data whether any relation exists between statin use and development of diabetes. METHODS We searched Medline, Embase, and the Cochrane Central Register of Controlled Trials from 1994 to 2009, for randomised controlled endpoint trials of statins. We included only trials with more than 1000 patients, with identical follow-up in both groups and duration of more than 1 year. We excluded trials of patients with organ transplants or who needed haemodialysis. We used the I(2) statistic to measure heterogeneity between trials and calculated risk estimates for incident diabetes with random-effect meta-analysis. FINDINGS We identified 13 statin trials with 91 140 participants, of whom 4278 (2226 assigned statins and 2052 assigned control treatment) developed diabetes during a mean of 4 years. Statin therapy was associated with a 9% increased risk for incident diabetes (odds ratio [OR] 1.09; 95% CI 1.02-1.17), with little heterogeneity (I(2)=11%) between trials. Meta-regression showed that risk of development of diabetes with statins was highest in trials with older participants, but neither baseline body-mass index nor change in LDL-cholesterol concentrations accounted for residual variation in risk. Treatment of 255 (95% CI 150-852) patients with statins for 4 years resulted in one extra case of diabetes. INTERPRETATION Statin therapy is associated with a slightly increased risk of development of diabetes, but the risk is low both in absolute terms and when compared with the reduction in coronary events. Clinical practice in patients with moderate or high cardiovascular risk or existing cardiovascular disease should not change. FUNDING None.
Collapse
Affiliation(s)
- Naveed Sattar
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Garip S, Yapici E, Ozek NS, Severcan M, Severcan F. Evaluation and discrimination of simvastatin-induced structural alterations in proteins of different rat tissues by FTIR spectroscopy and neural network analysis. Analyst 2010; 135:3233-41. [DOI: 10.1039/c0an00540a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
41
|
Abstract
AIM In the recently published The Justification for the Use of statins in Primary prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER) mega-trial, rosuvastatin significantly reduced cardiovascular events at the expense of a small but significant increase in the risk of developing type 2 diabetes. The increased risk of new-onset diabetes was in keeping with a recent meta-analysis which suggested that statins, with the possible exception of pravastatin, marginally increase the risk of developing type 2 diabetes. METHODS Although the net effect of rosuvastatin was obviously very positive, we hypothesized that the addition of a bile aid sequestrant to a statin would not only further decrease lipid levels and potentially further decrease cardiovascular events but also protect against the development of diabetes. This is particularly relevant because the bile acid sequestrant, colesevelam, has recently been approved for therapy of diabetes. RESULTS Colesevelam like other bile acid sequestrants lowers low-density lipoprotein levels by 16% and C-reactive protein by 22% beyond the reductions that occur with statin therapy alone. Bile acid sequestrants confer lipid-lowering, glucose-lowering, and anti-inflammatory benefits, and have been shown to reduce risk of cardiovascular events. CONCLUSIONS Therefore, colesevelam should be the most effective and logical agent to add to a statin in the diabetic and insulin-resistant patient, because in addition to lowering cardiac risk it may prevent the development of diabetes, as well as improving glycaemic control in the established diabetic patient.
Collapse
Affiliation(s)
- D S H Bell
- Southside Endocrinology and University of Alabama Medical School-Birmingham, Alabama, USA
| | | |
Collapse
|