1
|
Lembo M, Trimarco V, Izzo R, Pacella D, Jankauskas SS, Gallo P, Piccinocchi R, Morisco C, Piccinocchi G, Bardi L, Cristiano S, Esposito G, Giugliano G, Varzideh F, Manzi MV, Trimarco B, Santulli G. Statin-induced risk of diabetes does not reduce cardiovascular benefits in primary prevention: a 6-year propensity-score matched study in a large population. Cardiovasc Diabetol 2025; 24:233. [PMID: 40450251 DOI: 10.1186/s12933-025-02798-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Accepted: 05/20/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND The long-term risk of cardiovascular (CV) events in individuals who develop new-onset type 2 diabetes (T2D) after having received statin therapy in primary prevention is mostly unknown. METHODS We designed a population-based cohort study in individuals without T2D and atherosclerotic CV disease (ASCVD), divided in two groups according to the presence or not of statin therapy. We also balanced the study groups for demographic and clinical factors using propensity score matching. RESULTS 119307 individuals without T2D and ASCVD were divided in statin users (N = 90906) or not (N = 28401) and followed-up for 70.1 ± 61.3 months. Yearly incidence of T2D rate was 0.3% in the control group and 2.2% in the statin treated group. A Cox regression analysis confirmed the association between incident T2D and statin therapy. In normotensive individuals, the presence of statin therapy led to a 2-fold risk to develop incident T2D with a HR 2.61 (95% CI 2.11-3.22, p < 0.001) which was also that of statin untreated hypertensive patients. In the hypertensive population statin therapy was associated with a HR of incident T2D of 4.62 (95% CI 3.75-5.69, p < 0.001). CV events rate, including coronary and cerebrovascular fatal and non-fatal events, was 1.9% in the statin group vs. 0.7% in the control group and a multiple regression analysis demonstrated an association between statin therapy and CV events. A further Cox regression performed only in the statin treated population revealed a significant association of CV events with age, serum creatinine levels, and incident T2D. Of note, the increased rate of new-onset T2D associated with statin use does not modify the class of CV risk of this population. All these findings were confirmed at the propensity score matched analysis. CONCLUSIONS Statin therapy in primary prevention is associated with a higher risk of incident T2D, especially in hypertensive patients. However, since the final CV risk of those who develop T2D during statin treatment was lower than the one required for statin prescription according to the ESC guidelines, indicating that this phenomenon does not impair the benefit in CV prevention associated with the lipid lowering effect of statins.
Collapse
Affiliation(s)
- Maria Lembo
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Valentina Trimarco
- Department of Neuroscience, Reproductive Sciences, and Dentistry, "Federico II" University, Naples, Italy
| | - Raffaele Izzo
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Daniela Pacella
- Department of Public Health, "Federico II" University, Naples, Italy
| | - Stanislovas S Jankauskas
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York City, NY, USA
| | - Paola Gallo
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | | | - Carmine Morisco
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
- Academic Research Unit, International Translational Research and Medical Education (ITME) Consortium, Naples, Italy
- Italian Society for Cardiovascular Prevention (SIPREC), Rome, Italy
| | - Gaetano Piccinocchi
- COMEGEN Primary Care Physician Cooperative, Italian Society of General Medicine (SIMG), Naples, Italy
| | - Luca Bardi
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Stefano Cristiano
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Giuseppe Giugliano
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Fahimeh Varzideh
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York City, NY, USA
| | - Maria Virginia Manzi
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
- Academic Research Unit, International Translational Research and Medical Education (ITME) Consortium, Naples, Italy
- Italian Society for Cardiovascular Prevention (SIPREC), Rome, Italy
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy.
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York City, NY, USA.
- Academic Research Unit, International Translational Research and Medical Education (ITME) Consortium, Naples, Italy.
- Department of Molecular Pharmacology, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York City, NY, USA.
| |
Collapse
|
2
|
Park M, Kim JS, Park YA, Lee DH, Choi SA, Chang Y, Song TJ, Gwak HS, Yee J. Association between insulin-associated gene polymorphisms and new-onset diabetes mellitus in statin-treated patients. Eur J Clin Invest 2025; 55:e14366. [PMID: 39614667 DOI: 10.1111/eci.14366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND While statins are effective at managing lipid levels, there is growing evidence for new-onset diabetes mellitus (NODM). The insulin signalling pathway (ISP) inhibited by statins is one of the potential mechanisms; however, most studies have been limited to in vitro settings. Therefore, this study aimed to identify the genetic associations within the ISP-related genes and NODM. METHODS We performed a retrospective analysis of samples collected prospectively from February 2021 to May 2021. Among ISP-related genes, we selected 11 candidate genes (IGF1, IGF2, IGF1R, INSR, IRS1, IRS2, PIK3CA, PIK3CB, PIK3R1, AKT1 and AKT2). An additional analysis was conducted comparing patients with DM prior to statin therapy and controls to determine whether the single nucleotide polymorphisms (SNPs) are specific to statin. RESULTS A total of 602 patients were analysed, including 71 (11.8%) with statin-induced NODM. After adjustment, IGF1R rs2715439, INSR rs1799817, INSR rs2059807 and PIK3R1 rs3730089 were found to be independently associated with NODM. In an additional analysis, all SNPs that demonstrated an association with statin-induced NODM lost their significance in patients with DM prior to statin therapy. CONCLUSION This study revealed the ISP-related genetic effects, specifically involving genes such as INSR, IGF1R and PIK3R1, in the development of statin-induced NODM. Our findings suggest a potential mechanism of statin-induced NODM related to ISP-related genetic variants.
Collapse
Affiliation(s)
- Minju Park
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Jung Sun Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Yoon-A Park
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Da Hoon Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Seo-A Choi
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Yoonkyung Chang
- Department of Neurology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Tae-Jin Song
- Department of Neurology, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Hye Sun Gwak
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Jeong Yee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
3
|
Song B, Sun L, Qin X, Fei J, Yu Q, Chang X, He Y, Liu Y, Shi M, Guo D, Shen O, Zhu Z. Associations of Lipid-Lowering Drugs With Blood Pressure and Fasting Glucose: A Mendelian Randomization Study. Hypertension 2025; 82:743-751. [PMID: 39902581 DOI: 10.1161/hypertensionaha.124.23829] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Observational studies have linked LDL-C (low-density lipoprotein-cholesterol)-lowering drugs with lower blood pressure (BP) and higher fasting glucose, but the causality remains unclear. We conducted a drug target Mendelian randomization study to assess the causal associations of genetically proxied inhibition of HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase), PCSK9 (proprotein convertase subtilisin/kexin type 9), and NPC1L1 (Niemann-Pick C1-Like 1) with BP and fasting glucose. METHODS Single-nucleotide polymorphisms in HMGCR, NPC1L1, and PCSK9 associated with LDL-C in a genome-wide association study meta-analysis from the Global Lipid Genetics Consortium (173 082 European individuals) were used to proxy LDL-C-lowering drug targets. BP and fasting glucose data were obtained from genome-wide association studies conducted by the International Consortium of Blood Pressure (757 601 European participants) and the Glucose and Insulin-related Traits Consortium (58 074 European participants). We used the inverse-variance weighted method and a series of sensitivity analyses for assessment. RESULTS Genetically proxied inhibition of HMGCR was negatively associated with systolic BP (β, -0.81 [95% CI, -1.26 to -0.37 mm Hg]; P=3.72×10-4) and diastolic BP (β, -1.58 [95% CI, -2.24 to -0.91 mm Hg]; P=3.23×10-6). Conversely, we observed a positive association between genetically proxied inhibition of HMGCR and high fasting glucose (β, 0.13 [95% CI, 0.08-0.17 mmol/L]; P=4.25×10-8). However, there was no association of PCSK9 and NPC1L1 inhibition with BP or fasting glucose. CONCLUSIONS Genetically proxied inhibition of HMGCR was significantly associated with low BP and high fasting glucose, while there was no effect of PCSK9 and NPC1L1 inhibition on BP or fasting glucose.
Collapse
Affiliation(s)
- Beiping Song
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Lulu Sun
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Xiaoli Qin
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Jiawen Fei
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Quan Yu
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Xinyue Chang
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Yu He
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Yi Liu
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Mengyao Shi
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (M.S., Z.Z.)
| | - Daoxia Guo
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Ouxi Shen
- Department of Occupational Health, Suzhou Industrial Park Center for Disease Control and Prevention, China (O.S.)
| | - Zhengbao Zhu
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (M.S., Z.Z.)
| |
Collapse
|
4
|
Ariyanto EF, Wijaya I, Pradian ZA, Bhaskara APM, Lastialno MP, Rahman PHA, Bashari MH, Oktavia N, Putra MIP, Pratiwi YS, Heryaman H, Dhianawaty D. Aaptamine Inhibits Lipid Accumulation and Pparg and Slc2a4 Expression While Maintaining the Methylation of the Pparg Promoter During 3T3-L1 Adipocyte Differentiation. J Exp Pharmacol 2025; 17:159-168. [PMID: 40130029 PMCID: PMC11932118 DOI: 10.2147/jep.s511866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/08/2025] [Indexed: 03/26/2025] Open
Abstract
Purpose Excessive adipogenesis plays a role in the development of obesity and related metabolic disorders. Aaptamine is an alkaloid compound that has been proven to have various effects, however, no studies have yet investigated its effects on adipogenesis. This study aims to examine whether aaptamine inhibits lipid accumulation and Pparg and Slc2a4, two important genes in adipogenesis, mRNA expression, and increases the methylation of the Pparg promoter. This study strengthens the insights regarding these genes regulation, with future research potentially expanding to other adipogenic regulators for a broader perspective. Methods The effects of aaptamine (0 µM, 25 µM and 50 µM) were investigated in 3T3-L1 preadipocytes. The adipocytes were differentiated using a medium containing 3-isobutyl-1-methylxanthine, dexamethasone, and insulin. Cell viability was evaluated by the MTT assay, gene expression was analyzed by RT-qPCR, and lipid accumulation was determined using Oil Red O staining. Pyrosequencing was performed to measure the methylation of the Pparg promoter region. Results Aaptamine treatment significantly dose-dependently decreased lipid accumulation and inhibited Pparg and Slc2a4 mRNA expression. However, there were no significant differences in the methylation level of the Pparg promoter. Conclusion Aaptamine inhibits lipid accumulation and Pparg and Slc2a4 mRNA expression while maintaining the methylation level of the Pparg promoter during 3T3-L1 adipocyte differentiation.
Collapse
Affiliation(s)
- Eko Fuji Ariyanto
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| | - Ibnu Wijaya
- Undergraduate Program of Medical Doctor, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| | - Zaky Alif Pradian
- Undergraduate Program of Medical Doctor, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| | | | | | | | - Muhammad Hasan Bashari
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| | - Nandina Oktavia
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| | - Muhammad Iman Pratama Putra
- Department of Internal Medicine, Faculty of Medicine, Universitas Padjadjaran - Hasan Sadikin General Hospitale, Bandung, Indonesia
| | - Yuni Susanti Pratiwi
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| | - Henhen Heryaman
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| | - Diah Dhianawaty
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
5
|
Bredefeld CL, Choi P, Cullen T, Nicolich-Henkin SJ, Waters L. Statin Use and Hyperglycemia: Do Statins Cause Diabetes? Curr Atheroscler Rep 2024; 27:18. [PMID: 39699704 DOI: 10.1007/s11883-024-01266-8] [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] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
PURPOSE OF REVIEW Atherosclerotic cardiovascular disease (ASCVD) and diabetes are leading causes of morbidity and mortality in the United States and globally. Statin medications, a cornerstone of ASCVD prevention and treatment strategies, have been demonstrated to cause hyperglycemia and new onset diabetes mellitus (NODM). The purpose of this review is to summarize existing and emerging knowledge around the intersection of statins and these two important clinical problems. RECENT FINDINGS Since initial reporting of statin-induced hyperglycemia and NODM, the totality of available data corroborates an association between incident diabetes and statin use. A consensus that high-intensity statin and individuals with obesity or glycemic parameters approximating diabetes thresholds constitute the majority of risk exists. Alterations in insulin signaling, glucose transport and gastrointestinal microbiota are leading hypotheses underlying the mechanisms of statin-induced hyperglycemia. The probability of NODM based on an individual's risk factors and statin specific properties can be anticipated. This risk needs to be contextualized with the risk of ASCVD. In order to effectively adjudicate the risk of NODM, improvement in formulating and ultimately conveying a comprehensive ASCVD risk assessment to patients is necessary.
Collapse
Affiliation(s)
- Cindy L Bredefeld
- Department of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, 11530, USA.
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, 11501, USA.
| | - Paula Choi
- Department of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, 11530, USA
| | - Tiffany Cullen
- Department of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, 11530, USA
| | - Sophie J Nicolich-Henkin
- Department of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, 11530, USA
| | - Lauren Waters
- Department of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, 11530, USA
| |
Collapse
|
6
|
Anwar MY, Highland H, Buchanan VL, Graff M, Young K, Taylor KD, Tracy RP, Durda P, Liu Y, Johnson CW, Aguet F, Ardlie KG, Gerszten RE, Clish CB, Lange LA, Ding J, Goodarzi MO, Chen YDI, Peloso GM, Guo X, Stanislawski MA, Rotter JI, Rich SS, Justice AE, Liu CT, North K. Machine learning-based clustering identifies obesity subgroups with differential multi-omics profiles and metabolic patterns. Obesity (Silver Spring) 2024; 32:2024-2034. [PMID: 39497627 PMCID: PMC11540333 DOI: 10.1002/oby.24137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/18/2024] [Accepted: 07/22/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE Individuals living with obesity are differentially susceptible to cardiometabolic diseases. We hypothesized that an integrative multi-omics approach might improve identification of subgroups of individuals with obesity who have distinct cardiometabolic disease patterns. METHODS We performed machine learning-based, integrative unsupervised clustering to identify proteomics- and metabolomics-defined subpopulations of individuals living with obesity (BMI ≥ 30 kg/m2), leveraging data from 243 individuals in the Multi-Ethnic Study of Atherosclerosis (MESA) cohort. Omics that contributed to the observed clusters were functionally characterized. We performed multivariate regression to assess whether the individuals in each cluster demonstrated differential patterns of cardiometabolic traits. RESULTS We identified two distinct clusters (iCluster1 and 2). iCluster2 had significantly higher average BMI values, fasting blood glucose, and inflammation. iCluster1 was associated with higher levels of total cholesterol and high-density lipoprotein cholesterol. Pathways mediating cell growth, lipogenesis, and energy expenditures were positively associated with iCluster1. Inflammatory response and insulin resistance pathways were positively associated with iCluster2. CONCLUSIONS Although the two identified clusters may represent progressive obesity-related pathologic processes measured at different stages, other mechanisms in combination could also underpin the identified clusters given no significant age difference between the comparative groups. For instance, clusters may reflect differences in dietary/behavioral patterns or differential rates of metabolic damage.
Collapse
Affiliation(s)
- Mohammad Y Anwar
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Heather Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Victoria Lynn Buchanan
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kristin Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Yongmei Liu
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Craig W Johnson
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Francois Aguet
- Program of Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kristin G Ardlie
- Program of Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Robert E Gerszten
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clary B Clish
- Metabolite Profiling Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Leslie A Lange
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jingzhong Ding
- Section of Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Gina M Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston University, Boston, Massachusetts, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Maggie A Stanislawski
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Anne E Justice
- Department of Population Health Sciences, Geisinger Health System, Danville, Pennsylvania, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston University, Boston, Massachusetts, USA
| | - Kari North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
7
|
Wu Z, Chen S, Tao X, Liu H, Sun P, Richards AM, Tan HC, Yu Y, Yang Q, Wu S, Zhou X. Risk and effect modifiers for poor glycemic control among the chinese diabetic adults on statin therapy: the kailuan study. Clin Res Cardiol 2024; 113:1219-1231. [PMID: 38265512 DOI: 10.1007/s00392-024-02381-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024]
Abstract
BACKGROUND Limited studies have investigated the association between statin therapy and poor glycemic control, especially in the Chinese diabetic population. METHODS Two prospective diabetes cohorts were drawn from the Kailuan Cohort. In Cohort 1, linear regression models were used to evaluate the association between statin therapy and glycated hemoglobin (HbA1c) level change. In Cohort 2, new user design and conditional logistic models were used to assess associations between statin initiation and poor glycemic control which was a composite outcome comprised of hypoglycemic agent escalation and new-onset hyperglycemia. RESULTS Among 11,755 diabetic patients with medication information, 1400 statin users and 1767 statin nonusers with repeated HbA1c measurements were included in Cohort 1 (mean age: 64.6 ± 10.0 years). After a median follow-up of 3.02 (1.44, 5.00) years, statin therapy was associated with higher HbA1c levels (β: 0.20%; 95%CI: 0.05% to 0.34%). In Cohort 2, 1319 pairs of matched cases/controls were included (mean age: 61.6 ± 9.75 years). After a median follow-up of 4.87 (2.51, 8.42) years, poor glycemic control occurred in 43.0% of statin new users and 31.8% of statin nonusers (OR: 1.69; 95% CI: 1.32 to 2.17; P < 0.001). The statin-associated poor glycemic control risk was significantly higher among patients with lower body mass index (Pint = 0.089). Furthermore, a nonlinear association was observed between statin therapy duration and poor glycemic control (P = 0.003). CONCLUSIONS Among Chinese diabetic adults, statin therapy was associated with a higher level of HbA1c, and a higher risk of hypoglycemic agent escalation and new-onset hyperglycemia, especially among those who had lower body mass index levels and longer statin therapy duration.
Collapse
Affiliation(s)
- Zhaogui Wu
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan General Hospital, 57 Xinhuadong Street, Lubei District, TangshanHebei, 063001, China
| | - Xixi Tao
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Hangkuan Liu
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Pengfei Sun
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Arthur Mark Richards
- Cardiovascular Research Institute, National University Health System, 14 Medical Drive, Singapore, 117599, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive (MD6), Singapore, 117597, Singapore
| | - Huay Cheem Tan
- Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive (MD6), Singapore, 117597, Singapore
- Department of Cardiology, National University Heart Centre, 5 Lower Kent Ridge Rd, Singapore, 119074, Singapore
| | - Ying Yu
- Department of Pharmacology and Tianjin Key Laboratory of Inflammatory Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Qing Yang
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China.
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, 57 Xinhuadong Street, Lubei District, TangshanHebei, 063001, China.
| | - Xin Zhou
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
8
|
Shah R, Kong A, De Melo S, Boktor M, Henriquez R, Mandalia A, Samant H, Alvarez CA, Mansi IA. Association of statins with nonalcoholic fatty liver disease in patients with diabetes. J Investig Med 2024; 72:497-510. [PMID: 38594224 DOI: 10.1177/10815589241248076] [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] [Indexed: 04/11/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in patients with diabetes; limited data suggested that statins may reduce the risk of NAFLD progression. This study aimed to examine the association between statins and the development or progression of NAFLD in veterans with diabetes. In a new-user negative control design, we conducted a retrospective propensity score (PS)-matched cohort study of patients with diabetes between 2003 and 2015. After excluding patients with other causes of liver disease, we formed PS using 85 characteristics. The primary outcome was a composite NAFLD progression outcome. Primary analysis examined odds of outcome in PS-matched cohort. Post-hoc analysis included a PS-matched cohort of statin users with intensive lowering of low-density lipoprotein-cholesterol (LDL-C) vs low-intensity lowering. We matched 34,102 pairs from 300,739 statin users and 38,038 non-users. The composite outcome occurred in 8.8% of statin users and 8.6% of non-users (odds ratio (OR) 1.02, 95% confidence interval (95% CI) 0.97-1.08). In the post-hoc analysis, intensive lowering of LDL-C compared to low-intensity showed increased NAFLD progression (OR 1.21, 95% CI 1.13-1.30). This study showed that statin use in patients with diabetes was not associated with decreased or increased risk of NAFLD progression. Intensive LDL-C lowering, compared to low-intensity LDL-C lowering, was associated with an increased risk of NAFLD progression.
Collapse
Affiliation(s)
- Raj Shah
- Education Services, Orlando VA Healthcare System, Orlando, FL, USA
- University of Central Florida/HCA Florida Healthcare, Greater Orlando, FL, USA
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Alexander Kong
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Silvio De Melo
- Section of Gastroenterology, Orlando VA Healthcare System, Orlando, FL, USA
| | - Moheb Boktor
- Digestive and Liver Diseases Division. University of Texas Southwestern Medical Center, Dallas, TX
| | - Richard Henriquez
- Education Services, Orlando VA Healthcare System, Orlando, FL, USA
- University of Central Florida/HCA Florida Healthcare, Greater Orlando, FL, USA
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Amar Mandalia
- Section of Gastroenterology, Orlando VA Healthcare System, Orlando, FL, USA
| | - Hrishikesh Samant
- Department of Hepatology, Ochsner Medical Clinic, New Orleans, LA, USA
| | - Carlos A Alvarez
- Department of Pharmacy Practice and Center of Excellence in Real-world Evidence, Texas Tech University Health Sciences Center, Dallas, TX, USA
| | - Ishak A Mansi
- Education Services, Orlando VA Healthcare System, Orlando, FL, USA
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, USA
| |
Collapse
|
9
|
Oh KI, Lim E, Uprety LP, Jeong J, Jeong H, Park E, Jeong SY. Anti-adipogenic and anti-obesity effects of morroniside in vitro and in vivo. Biomed Pharmacother 2024; 176:116762. [PMID: 38788597 DOI: 10.1016/j.biopha.2024.116762] [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: 03/29/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Obesity is a multifaceted medical condition characterized by the pathological accumulation of excessive lipids in the body. We investigated the effects of morroniside, a bioactive compound derived from Cornus officinalis, on adipogenesis. We used a preadipocyte 3T3-L1 stable cell line and primary cultured adipose-derived stem cells (ADSCs) in vitro and ovariectomized (OVX) and a high-fat diet (HFD)-fed obese mouse model in vivo. Preadipocyte 3T3-L1 cells and ADSCs incubated with morroniside during adipocyte differentiation and obese mice subjected to OVX and HFD received oral morroniside treatment for 12 weeks. Morroniside treatment significantly reduced adipocyte differentiation and fatty acid accumulation and downregulated adipogenesis-related gene expression, concomitant with a decrease in triglyceride content and an increase in glycerol release in cells. The results of the in vivo study showed that morroniside ameliorated obesity-related phenotypes by reducing body weight gain, hepatic steatosis, and adipose tissue in obese mice. These findings suggest that morroniside is a promising compound for preventing and treating obesity.
Collapse
Affiliation(s)
- Kang-Il Oh
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, the Republic of Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, the Republic of Korea
| | - Eunguk Lim
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, the Republic of Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, the Republic of Korea
| | - Laxmi Prasad Uprety
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, the Republic of Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, the Republic of Korea
| | - Junhwan Jeong
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, the Republic of Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, the Republic of Korea
| | - Hyesoo Jeong
- Nine B Co., Ltd., Daejeon 34121, the Republic of Korea
| | - Eunkuk Park
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, the Republic of Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, the Republic of Korea.
| | - Seon-Yong Jeong
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, the Republic of Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, the Republic of Korea.
| |
Collapse
|
10
|
Heurtebize MA, Faillie JL. Drug-induced hyperglycemia and diabetes. Therapie 2024; 79:221-238. [PMID: 37985310 DOI: 10.1016/j.therap.2023.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/14/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Drug-induced hyperglycemia and diabetes have negative and potentially serious health consequences but can often be unnoticed. METHODS We reviewed the literature searching Medline database for articles addressing drug-induced hyperglycemia and diabetes up to January 31, 2023. We also selected drugs that could induce hyperglycemia or diabetes according official data from drug information databases Thériaque and Micromedex. For each selected drug or pharmacotherapeutic class, the mechanisms of action potentially involved were investigated. For drugs considered to be at risk of hyperglycemia or diabetes, disproportionality analyses were performed using data from the international pharmacovigilance database VigiBase. In order to detect new pharmacovigilance signals, additional disproportionality analyses were carried out for drug classes with more than 100 cases reported in VigiBase, but not found in the literature or official documents. RESULTS The main drug classes found to cause hyperglycemia are glucocorticoids, HMG-coA reductase inhibitors, thiazide diuretics, beta-blockers, antipsychotics, fluoroquinolones, antiretrovirals, antineoplastic agents and immunosuppressants. The main mechanisms involved are alterations in insulin secretion and sensitivity, direct cytotoxic effects on pancreatic cells and increases in glucose production. Pharmacovigilance signal were found for a majority of drugs or pharmacological classes identified as being at risk of diabetes or hyperglycemia. We identified new pharmacovigilance signals with drugs not known to be at risk according to the literature or official data: phosphodiesterase type 5 inhibitors, endothelin receptor antagonists, sodium oxybate, biphosphonates including alendronic acid, digoxin, sartans, linosipril, diltiazem, verapamil, and darbepoetin alpha. Further studies will be needed to confirm these signals. CONCLUSIONS The risks of induced hyperglycemia vary from one drug to another, and the underlying mechanisms are multiple and potentially complex. Clinicians need to be vigilant when using at-risk drugs in order to detect and manage these adverse drug reactions. However, it is to emphasize that the benefits of appropriately prescribed treatments most often outweigh their metabolic risks.
Collapse
Affiliation(s)
- Marie-Anne Heurtebize
- CHU de Montpellier, Medical Pharmacology and Toxicology Department, 34000 Montpellier, France
| | - Jean-Luc Faillie
- CHU de Montpellier, Medical Pharmacology and Toxicology Department, 34000 Montpellier, France; IDESP, Université de Montpellier, Inserm, 34295 Montpellier, France.
| |
Collapse
|
11
|
Masson W, Lobo M, Barbagelata L, Nogueira JP. Statins and new-onset diabetes in primary prevention setting: an updated meta-analysis stratified by baseline diabetes risk. Acta Diabetol 2024; 61:351-360. [PMID: 37934231 DOI: 10.1007/s00592-023-02205-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/16/2023] [Indexed: 11/08/2023]
Abstract
AIMS The use of statins has been associated with an increased risk of new-onset diabetes. The characteristics of the population could influence this association. The objective of this study was to determine the risk of new-onset diabetes with the use of statins in patients in primary prevention, with an assessment of the results according to the baseline risk of developing diabetes of the included population. METHODS We performed an updated meta-analysis including randomized trials of statin therapy in primary prevention settings that report new-onset diabetes. The rate of new cases of diabetes in the control arms was estimated for each study. The studies were classified into two groups (low rate: < 7.5 events per 1000 patients-year; high rate; ≥ 7.5 events per 1000 patients-year). The fixed-effects model was performed. RESULTS Eight studies (70,453 patients) were included. Globally, statin therapy was associated with an increased risk of new-onset diabetes (OR 1.1; 95% CI 1.0-1.2, I2 35%). When we analyzed the studies according to the baseline diabetes risk in the control groups, the results showed that there was a greater risk only in the studies with a high baseline rate (OR 1.2; 95% CI 1.1-1.3, I2 0%; interaction p value = 0.01). CONCLUSION Globally, the use of statins in patients in primary prevention was associated with an increased risk of new-onset diabetes. In the stratified analysis, this association was observed only in the group of studies with a high baseline rate of events.
Collapse
Affiliation(s)
- Walter Masson
- Cardiology Department, Hospital Italiano de Buenos Aires, Perón 4190. Ciudad Autónoma de Buenos Aires, C1199ABB, Buenos Aires, Argentina.
| | - Martín Lobo
- Cardiology Department, Hospital Militar Campo de Mayo, Buenos Aires, Argentina
| | - Leandro Barbagelata
- Cardiology Department, Hospital Italiano de Buenos Aires, Perón 4190. Ciudad Autónoma de Buenos Aires, C1199ABB, Buenos Aires, Argentina
| | - Juan P Nogueira
- Universidad Internacional de Las Américas, San José, Costa Rica
- Centro de Investigación en Endocrinología, Nutrición y Metabolismo (CIENM), Universidad Nacional de Formosa, Facultad de Ciencias de La Salud, Formosa, Argentina
| |
Collapse
|
12
|
Koponen K, Kambur O, Joseph B, Ruuskanen MO, Jousilahti P, Salido R, Brennan C, Jain M, Meric G, Inouye M, Lahti L, Niiranen T, Havulinna AS, Knight R, Salomaa V. Role of Gut Microbiota in Statin-Associated New-Onset Diabetes-A Cross-Sectional and Prospective Analysis of the FINRISK 2002 Cohort. Arterioscler Thromb Vasc Biol 2024; 44:477-487. [PMID: 37970720 PMCID: PMC10805357 DOI: 10.1161/atvbaha.123.319458] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Dyslipidemia is treated effectively with statins, but treatment has the potential to induce new-onset type-2 diabetes. Gut microbiota may contribute to this outcome variability. We assessed the associations of gut microbiota diversity and composition with statins. Bacterial associations with statin-associated new-onset type-2 diabetes (T2D) risk were also prospectively evaluated. METHODS We examined shallow-shotgun-sequenced fecal samples from 5755 individuals in the FINRISK-2002 population cohort with a 17+-year-long register-based follow-up. Alpha-diversity was quantified using Shannon index and beta-diversity with Aitchison distance. Species-specific differential abundances were analyzed using general multivariate regression. Prospective associations were assessed with Cox regression. Applicable results were validated using gradient boosting. RESULTS Statin use associated with differing taxonomic composition (R2, 0.02%; q=0.02) and 13 differentially abundant species in fully adjusted models (MaAsLin; q<0.05). The strongest positive association was with Clostridium sartagoforme (β=0.37; SE=0.13; q=0.02) and the strongest negative association with Bacteroides cellulosilyticus (β=-0.31; SE=0.11; q=0.02). Twenty-five microbial features had significant associations with incident T2D in statin users, of which only Bacteroides vulgatus (HR, 1.286 [1.136-1.457]; q=0.03) was consistent regardless of model adjustment. Finally, higher statin-associated T2D risk was seen with [Ruminococcus] torques (ΔHRstatins, +0.11; q=0.03), Blautia obeum (ΔHRstatins, +0.06; q=0.01), Blautia sp. KLE 1732 (ΔHRstatins, +0.05; q=0.01), and beta-diversity principal component 1 (ΔHRstatin, +0.07; q=0.03) but only when adjusting for demographic covariates. CONCLUSIONS Statin users have compositionally differing microbiotas from nonusers. The human gut microbiota is associated with incident T2D risk in statin users and possibly has additive effects on statin-associated new-onset T2D risk.
Collapse
Affiliation(s)
- Kari Koponen
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland (K.K., O.K., B.J., P.J., T.N., A.S.H., V.S.)
| | - Oleg Kambur
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland (K.K., O.K., B.J., P.J., T.N., A.S.H., V.S.)
| | - Bijoy Joseph
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland (K.K., O.K., B.J., P.J., T.N., A.S.H., V.S.)
| | | | - Pekka Jousilahti
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland (K.K., O.K., B.J., P.J., T.N., A.S.H., V.S.)
| | - Rodolfo Salido
- Department of Pediatrics (R.S., C.B., R.K.), University of California San Diego, La Jolla
- Department of Bioengineering (R.S., R.K.), University of California San Diego, La Jolla
| | - Caitriona Brennan
- Department of Pediatrics (R.S., C.B., R.K.), University of California San Diego, La Jolla
| | - Mohit Jain
- Department of Medicine and Pharmacology (M.J.), University of California San Diego, La Jolla
| | - Guillaume Meric
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia (G.M., M.I.)
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Australia (G.M.)
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia (G.M., M.I.)
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, United Kingdom (M.I.)
| | - Leo Lahti
- Department of Computing, University of Turku, Finland (M.O.R., L.L.)
| | - Teemu Niiranen
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland (K.K., O.K., B.J., P.J., T.N., A.S.H., V.S.)
- Department of Medicine, Turku University Hospital and University of Turku, Finland (T.N.)
| | - Aki S. Havulinna
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland (K.K., O.K., B.J., P.J., T.N., A.S.H., V.S.)
- Institute for Molecular Medicine Finland, FiMM-HiLIFE, Helsinki, Finland (A.S.H.)
| | - Rob Knight
- Department of Pediatrics (R.S., C.B., R.K.), University of California San Diego, La Jolla
- Department of Bioengineering (R.S., R.K.), University of California San Diego, La Jolla
- Department of Computer Science and Engineering (R.K.), University of California San Diego, La Jolla
- Center for Microbiome Innovation (R.K.), University of California San Diego, La Jolla
| | - Veikko Salomaa
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland (K.K., O.K., B.J., P.J., T.N., A.S.H., V.S.)
| |
Collapse
|
13
|
Chen WH, Chen CH, Hsu MC, Chang RW, Wang CH, Lee TS. Advances in the molecular mechanisms of statins in regulating endothelial nitric oxide bioavailability: Interlocking biology between eNOS activity and L-arginine metabolism. Biomed Pharmacother 2024; 171:116192. [PMID: 38262153 DOI: 10.1016/j.biopha.2024.116192] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/25/2024] Open
Abstract
Statins, inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A, are widely used to treat hypercholesterolemia. In addition, statins have been suggested to reduce the risk of cardiovascular events owing to their pleiotropic effects on the vascular system, including vasodilation, anti-inflammation, anti-coagulation, anti-oxidation, and inhibition of vascular smooth muscle cell proliferation. The major beneficial effect of statins in maintaining vascular homeostasis is the induction of nitric oxide (NO) bioavailability by activating endothelial NO synthase (eNOS) in endothelial cells. The mechanisms underlying the increased NO bioavailability and eNOS activation by statins have been well-established in various fields, including transcriptional and post-transcriptional regulation, kinase-dependent phosphorylation and protein-protein interactions. However, the mechanism by which statins affect the metabolism of L-arginine, a precursor of NO biosynthesis, has rarely been discussed. Autophagy, which is crucial for energy homeostasis, regulates endothelial functions, including NO production and angiogenesis, and is a potential therapeutic target for cardiovascular diseases. In this review, in addition to summarizing the molecular mechanisms underlying increased NO bioavailability and eNOS activation by statins, we also discuss the effects of statins on the metabolism of L-arginine.
Collapse
Affiliation(s)
- Wen-Hua Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Hui Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Man-Chen Hsu
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ru-Wen Chang
- Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Chih-Hsien Wang
- Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
14
|
Song I, Kim M, Choi H, Kim JH, Lim KH, Yoon HS, Rah YC, Park E, Im GJ, Song JJ, Chae SW, Choi J. Hydrophilic and lipophilic statin use and risk of hearing loss in hyperlipidemia using a Common Data Model: multicenter cohort study. Sci Rep 2023; 13:12373. [PMID: 37524760 PMCID: PMC10390480 DOI: 10.1038/s41598-023-39316-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/23/2023] [Indexed: 08/02/2023] Open
Abstract
Hearing impairment, the third largest health burden worldwide, currently lacks definitive treatments or preventive drugs. This study compared the effects of hydrophilic and lipophilic statin on hearing loss using a common database model. This retrospective multicenter study was conducted in three hospitals in South Korea (Anam, Guro, Ansan). We enrolled patients with hyperlipidemia with an initial hearing loss diagnosis. Data were collected during January 1, 2022-December 31, 2021 using the Observational Health Data Science and Informatics open-source software and Common Data Model database. The primary outcome was the occurrence of first-time hearing loss following a hyperlipidemia diagnosis, as documented in the Common Data Model cohort database. The measures of interest were hearing loss risk between hydrophilic and lipophilic statin use. Variables were compared using propensity score matching, Cox proportional regression, and meta-analysis. Among 37,322 patients with hyperlipidemia, 13,751 (7669 men and 6082 women) and 23,631 (11,390 men and 12,241 women) were treated with hydrophilic and lipophilic statins, respectively. After propensity score matching, according to the Kaplan-Meier curve, hearing loss risk did not significantly differ among the hospitals. The hazard ratio (HR) of the male patients from Anam (0.29, [95% confidence interval (CI), 0.05-1.51]), Guro (HR, 0.56, [95% CI 0.18-1.71]), and Ansan (hazard ratio, 0.29, [95% CI 0.05-1.51]) hospitals were analyzed using Cox proportional regression. Overall effect size (HR, 0.40, [95% CI 0.18-0.91]) was estimated using meta-analysis, which indicated that hearing loss risk among hydrophilic statin users was less than that among lipophilic statin users and was statistically significant. Men in the hydrophilic statin group had a lower risk of hearing impairment than those in the lipophilic statin group.
Collapse
Affiliation(s)
- Insik Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea
| | - Minjin Kim
- Department of Biostatistics, Korea University College of Medicine, Seoul, Republic of Korea
- Biomedical Research Center, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Hangseok Choi
- Department of Biostatistics, Korea University College of Medicine, Seoul, Republic of Korea
- Biomedical Research Center, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Jeong Hwan Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea
| | - Kang Hyeon Lim
- Department of Otorhinolaryngology-Head and Neck Surgery, Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea
| | - Hee Soo Yoon
- Department of Otorhinolaryngology-Head and Neck Surgery, Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea
| | - Yoon Chan Rah
- Department of Otorhinolaryngology-Head and Neck Surgery, Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea
| | - Euyhyun Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Gi Jung Im
- Department of Otorhinolaryngology-Head and Neck Surgery, Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sung-Won Chae
- Department of Otorhinolaryngology-Head and Neck Surgery, Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - June Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea.
- Department of Medical Informatics, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
15
|
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
|
16
|
Kwon J, Yeh YS, Kawarasaki S, Minamino H, Fujita Y, Okamatsu-Ogura Y, Takahashi H, Nomura W, Matsumura S, Yu R, Kimura K, Saito M, Inagaki N, Inoue K, Kawada T, Goto T. Mevalonate biosynthesis pathway regulates the development and survival of brown adipocytes. iScience 2023; 26:106161. [PMID: 36895651 PMCID: PMC9988578 DOI: 10.1016/j.isci.2023.106161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/08/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
The high thermogenic activity of brown adipose tissue (BAT) has received considerable attention. Here, we demonstrated the role of the mevalonate (MVA) biosynthesis pathway in the regulation of brown adipocyte development and survival. The inhibition of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), the rate-limiting enzyme in the MVA pathway and the molecular target of statins, suppressed brown adipocyte differentiation by suppressing protein geranylgeranylation-mediated mitotic clonal expansion. The development of BAT in neonatal mice exposed to statins during the fetal period was severely impaired. Moreover, statin-induced geranylgeranyl pyrophosphate (GGPP) deficiency led to the apoptosis of mature brown adipocytes. Brown adipocyte-specific Hmgcr knockout induced BAT atrophy and disrupted thermogenesis. Importantly, both genetic and pharmacological inhibition of HMGCR in adult mice induced morphological changes in BAT accompanied by an increase in apoptosis, and statin-treated diabetic mice showed worsened hyperglycemia. These findings revealed that MVA pathway-generated GGPP is indispensable for BAT development and survival.
Collapse
Affiliation(s)
- Jungin Kwon
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan
| | - Yu-Sheng Yeh
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan
| | - Satoko Kawarasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan
| | - Hiroto Minamino
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yoshihito Fujita
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yuko Okamatsu-Ogura
- Departments of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Haruya Takahashi
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan
| | - Wataru Nomura
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan.,Research Unit for Physiological Chemistry, the Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan
| | - Shigenobu Matsumura
- Division of Clinical Nutrition, Graduate School of Comprehensive Rehabilitation, Osaka Prefecture University, Osaka 583-0872, Japan
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Kazuhiro Kimura
- Departments of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Masayuki Saito
- Departments of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Kazuo Inoue
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan.,Research Unit for Physiological Chemistry, the Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan
| | - Teruo Kawada
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan.,Research Unit for Physiological Chemistry, the Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan.,Research Unit for Physiological Chemistry, the Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
17
|
Akash MSH, Rasheed S, Rehman K, Ibrahim M, Imran M, Assiri MA. Biochemical Activation and Regulatory Functions of Trans-Regulatory KLF14 and Its Association with Genetic Polymorphisms. Metabolites 2023; 13:metabo13020199. [PMID: 36837818 PMCID: PMC9962810 DOI: 10.3390/metabo13020199] [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: 12/27/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Krüpple-Like family of transcription factor-14 (KLF14) is a master trans-regulatory gene that has multiple biological regulatory functions and is involved in many pathological mechanisms. It controls the expressions of several other genes which are involved in multiple regulatory functions. KLF14 plays a significant role in lipid metabolism, glucose regulation and insulin sensitivity. Cell apoptosis, proliferation, and differentiation are regulated by the KLF14 gene, and up-regulation of KLF14 prevents cancer progression. KLF14 has been used as an epigenetic biomarker for the estimation of chronological age due to the presence of different age-related CpG sites on genes that become methylated with age. Different genome-wide association studies have identified several KLF14 variants in adipose tissues. These single nucleotide polymorphisms in KLF14 have been associated with dyslipidemia, insulin resistance, and glucose intolerance. Moreover, the prevalence of genetic polymorphism is different in different populations due to ethnic differences and epigenetic modifications. In addition, environmental and physiological factors such as diet, age, gender, and obesity are also responsible for genetic mutations in KLF14.
Collapse
Affiliation(s)
- Muhammad Sajid Hamid Akash
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad 38000, Pakistan
- Correspondence: (M.S.H.A.); (K.R.)
| | - Sumbal Rasheed
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, The Women University, Multan 60000, Pakistan
- Correspondence: (M.S.H.A.); (K.R.)
| | - Muhammad Ibrahim
- Department of Applied Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Muhammad Imran
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 62413, Saudi Arabia
- Department of Chemistry, Faculty of Science, King Khalid University, Abha 62413, Saudi Arabia
| | - Mohammed A. Assiri
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 62413, Saudi Arabia
- Department of Chemistry, Faculty of Science, King Khalid University, Abha 62413, Saudi Arabia
| |
Collapse
|
18
|
Ruscica M, Ferri N, Banach M, Sirtori CR, Corsini A. Side effects of statins: from pathophysiology and epidemiology to diagnostic and therapeutic implications. Cardiovasc Res 2023; 118:3288-3304. [PMID: 35238338 DOI: 10.1093/cvr/cvac020] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/02/2022] [Indexed: 01/25/2023] Open
Abstract
Treatment with statins, inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, has proven beneficial preventive effects on cardiovascular events. However, discontinuation due to intolerance and non-adherence remain two of the major gaps in both primary and secondary prevention. This leads many patients with high-risk of atherosclerotic cardiovascular disease (ASCVD) to be inadequately treated or not to achieve target lipid level goals, and as consequence they undergo an increased risk of cardiovascular events. The aim of this review is thus to give an overview of the reasons for discontinuation and on the possible mechanisms behind them. Although statins, as a class, are generally safe, they are associated with an increased risk of diabetes mellitus and hepatic transaminase elevations. Incidence of cataracts or cognitive dysfunction and others presented in the literature (e.g. proteinuria and haematuria) have been never confirmed to have a causal link. Conversely, debated remains the effect on myalgia. Muscle side effects are the most commonly reported, although myalgia is still believed by some to be the result of a nocebo/drucebo effect. Concerning mechanisms behind muscular side effects, no clear conclusions have been reached. Thus, if on one side it is important to identify individuals either at higher risk to develop a side effect, or with confirmed risk factors and conditions of statin intolerance, on the other side alternative strategies should be identified to avoid an increased ASCVD risk.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, Universita degli Studi di Padova, Padova, Italy
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland.,Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| |
Collapse
|
19
|
Muacevic A, Adler JR, Ray SD. The Benefits Outweigh the Risks of Treating Hypercholesterolemia: The Statin Dilemma. Cureus 2023; 15:e33648. [PMID: 36788860 PMCID: PMC9912858 DOI: 10.7759/cureus.33648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2023] [Indexed: 01/13/2023] Open
Abstract
Cardiovascular diseases are one of the leading causes of death in the United States; therefore, primary and secondary prevention are of the utmost importance. In this regard, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMG-CoA) reductase inhibitors, also known as statins, have been anointed as the primary treatment method for lowering cholesterol to prevent cardiovascular diseases. Statins decrease the low-density lipoprotein (LDL) cholesterol and triglycerides in the body, thus lowering the total body cholesterol levels. Despite the benefits associated with statins, it is essential to understand the adverse effects of these drugs. Myotoxicity and statin-associated muscle symptoms are the most common adverse effects of statins. The impairment of mitochondrial function is another adverse effect that can lead to hepatic dysfunction, neurocognitive effects, and potentially the new onset of diabetes. The exact pathophysiology of these side effects is still not fully understood. However, several mechanisms have been proposed, although there is significant overlap among the hypothetical propositions. Understanding the overall outcomes of each of these adverse effects can allow a healthcare practitioner to carefully map out whether statin administration should be used to prevent hypercholesterolemia in the body. The adverse effect of statins is dependent on both the dose and the type of statin used. Lipophilic statins tend to possess a more remarkable ability to infiltrate membranes; they have been hypothesized to cause statin-induced myopathies as well as neurocognitive effects by significantly crossing the blood-brain barrier. In summary, this review has focused on the mechanistic and clinical aspects of this statin class of medication. Proposed mechanisms for different adverse effects associated with statins remain a focus of this communication.
Collapse
|
20
|
Ruzieh M, Ahmad TA, Liu G, Foy AJ. Association between statin exposure and diabetes incidence among privately-insured patients before and after applying a novel technique to control for selection bias. Am J Med Sci 2023; 365:26-30. [PMID: 36096188 DOI: 10.1016/j.amjms.2022.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 08/08/2022] [Accepted: 08/30/2022] [Indexed: 01/04/2023]
Abstract
INTRODUCTION The association between statins and incident diabetes mellitus (DM) in observational studies is much larger than that reported from randomized controlled trials. We sought to assess this association using a novel design controlling for selection bias. METHODS Using data from MarketScan, we identified a cohort of non-diabetic patients who initiated a statin and matched them to patients not taking statins. From the statin-user cohort, we identified two subgroups: patients who received statin refills for >6 months (continuers) and patients who received statin refills <6 months (discontinuers). Patients were followed for a minimum of two years to determine incident DM. RESULTS We included 442,526 patients, divided equally between statin users and non-users. Statin use was associated with increased DM (9.9% vs. 4.4%, HR 2.2, p < 0.001). Among the 221,263 statin users, there were 194,357 continuers and 26,906 discontinuers. There was no significant difference in the incidence rate of DM between both groups (10.0% vs. 9.3%, HR 1.03, p = 0.22). CONCLUSIONS Statin use was strongly associated with incident diabetes when users were compared to non-users but not when continuers were compared to discontinuers. Selection bias confounds the association between statin use and incident diabetes in observational studies.
Collapse
Affiliation(s)
- Mohammed Ruzieh
- Division of Cardiovascular Medicine. University of Florida, Gainesville, FL, United States.
| | - Tariq Ali Ahmad
- Main Line Health System. Department of Interventional Cardiology. Wynnewood, PA, United States
| | - Guodong Liu
- Penn State College of Medicine. Department of Public Health Sciences. Hershey, PA, United States
| | - Andrew J Foy
- Penn State College of Medicine. Department of Public Health Sciences. Hershey, PA, United States; Penn State Heart and Vascular Institute. Penn State College of Medicine. Hershey, PA, United States
| |
Collapse
|
21
|
Kuretu A, Arineitwe C, Mothibe M, Ngubane P, Khathi A, Sibiya N. Drug-induced mitochondrial toxicity: Risks of developing glucose handling impairments. Front Endocrinol (Lausanne) 2023; 14:1123928. [PMID: 36860368 PMCID: PMC9969099 DOI: 10.3389/fendo.2023.1123928] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
Mitochondrial impairment has been associated with the development of insulin resistance, the hallmark of type 2 diabetes mellitus (T2DM). However, the relationship between mitochondrial impairment and insulin resistance is not fully elucidated due to insufficient evidence to support the hypothesis. Insulin resistance and insulin deficiency are both characterised by excessive production of reactive oxygen species and mitochondrial coupling. Compelling evidence states that improving the function of the mitochondria may provide a positive therapeutic tool for improving insulin sensitivity. There has been a rapid increase in reports of the toxic effects of drugs and pollutants on the mitochondria in recent decades, interestingly correlating with an increase in insulin resistance prevalence. A variety of drug classes have been reported to potentially induce toxicity in the mitochondria leading to skeletal muscle, liver, central nervous system, and kidney injury. With the increase in diabetes prevalence and mitochondrial toxicity, it is therefore imperative to understand how mitochondrial toxicological agents can potentially compromise insulin sensitivity. This review article aims to explore and summarise the correlation between potential mitochondrial dysfunction caused by selected pharmacological agents and its effect on insulin signalling and glucose handling. Additionally, this review highlights the necessity for further studies aimed to understand drug-induced mitochondrial toxicity and the development of insulin resistance.
Collapse
Affiliation(s)
- Auxiliare Kuretu
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Charles Arineitwe
- 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 Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andile Khathi
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Ntethelelo Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
- *Correspondence: Ntethelelo Sibiya,
| |
Collapse
|
22
|
Does Pitavastatin Therapy for Patients with Type 2 Diabetes and Dyslipidemia Affect Serum Adiponectin Levels and Insulin Sensitivity? J Clin Med 2022; 11:jcm11226756. [PMID: 36431233 PMCID: PMC9692324 DOI: 10.3390/jcm11226756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/07/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022] Open
Abstract
(1) Background: We aimed to demonstrate the effects of pitvastatin therapy on the serum levels of total adiponectin and high-molecular-weight (HMW) adiponectin in type 2 diabetes and the correlation with insulin sensitivity. (2) Methods: This study was designed as an open-labelled randomized trial. Patients with diabetes who were prescribed pitavastatin therapy were enrolled and randomized to either treatment with 2 mg of pitavastatin once daily (n = 44) (PITA group) or diet and exercise only, except their antidiabetic medications (n = 49), for 24 weeks. (3) Results: In lipid profiles, the reduction in total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) was significantly increased in the PITA group (TC; 207.5 ± 20 vs. 195.5 ± 30.0 ng/dL, p < 0.001, LDL-C; 132.0 ± 15.8 vs. 123.1 ± 25.7 mg/dL, p < 0.001). Adiponectin and HMW adiponectin were elevated in the PITA group, compared to the control group without significance. The PITA group showed a lower level of HOMA-IR and HOMA-β levels. However, there was no significance (HOMA-IR; p = 0.5921 -at 12 weeks and p = 0.3645 at 24 weeks; HOMA-β; p = 0.8915 at 12 weeks and p = 0.7313 in 6 months). (4) Conclusions: The present study did not show a significant change in serum adiponectin or HMW adiponectin from baseline in serum adiponectin following pitavastatin therapy. Although statin has been considered as a risk for dysglycemia, pitavastatin did not affect insulin sensitivity.
Collapse
|
23
|
Shu X, Wu J, Zhang T, Ma X, Du Z, Xu J, You J, Wang L, Chen N, Luo M, Wu J. Statin-Induced Geranylgeranyl Pyrophosphate Depletion Promotes Ferroptosis-Related Senescence in Adipose Tissue. Nutrients 2022; 14:nu14204365. [PMID: 36297049 PMCID: PMC9607568 DOI: 10.3390/nu14204365] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Statin treatment is accepted to prevent adverse cardiovascular events. However, atorvastatin, an HMG-CoA reductase inhibitor, has been reported to exhibit distinct effects on senescent phenotypes. Whether atorvastatin can induce adipose tissue senescence and the mechanisms involved are unknown. The effects of atorvastatin-induced senescence were examined in mouse adipose tissue explants. Here, we showed that statin initiated higher levels of mRNA related to cellular senescence markers and senescence-associated secretory phenotype (SASP), as well as increased accumulation of the senescence-associated β-galactosidase (SA-β-gal) stain in adipose tissues. Furthermore, we found that the levels of reactive oxygen species (ROS), malondialdehyde (MDA), and Fe2+ were elevated in adipose tissues treated with atorvastatin, accompanied by a decrease in the expression of glutathione (GSH), and glutathione peroxidase 4 (GPX4), indicating an iron-dependent ferroptosis. Atorvastatin-induced was prevented by a selective ferroptosis inhibitor (Fer-1). Moreover, supplementation with geranylgeranyl pyrophosphate (GGPP), a metabolic intermediate, reversed atorvastatin-induced senescence, SASP, and lipid peroxidation in adipose tissue explants. Atorvastatin depleted GGPP production, but not Fer-1. Atorvastatin was able to induce ferroptosis in adipose tissue, which was due to increased ROS and an increase in cellular senescence. Moreover, this effect could be reversed by the supplement of GGPP. Taken together, our results suggest that the induction of ferroptosis contributed to statin-induced cell senescence in adipose tissue.
Collapse
Affiliation(s)
- Xin Shu
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
| | - Jiaqi Wu
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
| | - Tao Zhang
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
| | - Xiaoyu Ma
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
| | - Zuoqin Du
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
| | - Jin Xu
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
| | - Jingcan You
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
| | - Liqun Wang
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
| | - Ni Chen
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
| | - Mao Luo
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
| | - Jianbo Wu
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, China
- Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Southwest Medical University, Luzhou 646000, China
- Correspondence: ; Tel./Fax: +86-830-3161702
| |
Collapse
|
24
|
Statins and renal disease progression, ophthalmic manifestations, and neurological manifestations in veterans with diabetes: A retrospective cohort study. PLoS One 2022; 17:e0269982. [PMID: 35862466 PMCID: PMC9302779 DOI: 10.1371/journal.pone.0269982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 06/01/2022] [Indexed: 11/19/2022] Open
Abstract
Background Statins increase insulin resistance, which may increase risk of diabetic microvascular complications. Little is known about the impact of statins on renal, ophthalmologic, and neurologic complications of diabetes in practice. The objective of this study was to examine the association of statins with renal disease progression, ophthalmic manifestations, and neurological manifestations in diabetes. Methods This is a retrospective cohort study, new-user active comparator design, that included a national Veterans Health Administration (VA) patients with diabetes from 2003 to 2015. Patients were age 30 years or older and were regular users of the VA with data encompassing clinical encounters, demographics, vital signs, laboratory tests, and medications. Patients were divided into statin users or nonusers (active comparators). Statin users initiated statins and nonusers initiated H2-blockers or proton pump-inhibitors (H2-PPI) as an active comparator. Study outcomes were: 1) Composite renal disease progression outcome; 2) Incident diabetes with ophthalmic manifestations; and 3) Incident diabetes with neurological manifestations. Results Out of 705,774 eligible patients, we propensity score matched 81,146 pairs of statin users and active comparators. Over a mean (standard deviation) of follow up duration of 4.8 (3) years, renal disease progression occurred in 9.5% of statin users vs 8.3% of nonusers (odds ratio [OR]: 1.16; 95% confidence interval [95%CI]: 1.12–1.20), incident ophthalmic manifestations in 2.7% of statin users vs 2.0% of nonusers (OR: 1.35, 95%CI:1.27–1.44), and incident neurological manifestations in 6.7% of statin users vs 5.7% of nonusers (OR: 1.19, 95%CI:1.15–1.25). Secondary, sensitivity, and post-hoc analyses were consistent and demonstrated highest risks among the healthier subgroup and those with intensive lowering of LDL-cholesterol. Conclusions Statin use in patients with diabetes was associated with modestly higher risk of renal disease progression, incident ophthalmic, and neurological manifestations. More research is needed to assess the overall harm/benefit balance for statins in the lower risk populations with diabetes and those who receive intensive statin therapy.
Collapse
|
25
|
García-Fernández-Bravo I, Torres-Do-Rego A, López-Farré A, Galeano-Valle F, Demelo-Rodriguez P, Alvarez-Sala-Walther LA. Undertreatment or Overtreatment With Statins: Where Are We? Front Cardiovasc Med 2022; 9:808712. [PMID: 35571155 PMCID: PMC9105719 DOI: 10.3389/fcvm.2022.808712] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/29/2022] [Indexed: 12/26/2022] Open
Abstract
Statins, in addition to healthy lifestyle interventions, are the cornerstone of lipid-lowering therapy. Other low-density lipoprotein (LDL)-lowering drugs include ezetimibe, bile acid sequestrants, and PCSK9 inhibitors. As new evidence emerges from new clinical trials, therapeutic goals change, leading to renewed clinical guidelines. Nowadays, LDL goals are getting lower, leading to the "lower is better" paradigm in LDL-cholesterol (LDL-C) management. Several observational studies have shown that LDL-C control in real life is suboptimal in both primary and secondary preventions. It is critical to enhance the adherence to guideline recommendations through shared decision-making between clinicians and patients, with patient engagement in selecting interventions based on individual values, preferences, and associated conditions and comorbidities. This narrative review summarizes the evidence regarding the benefits of lipid-lowering drugs in reducing cardiovascular events, the pleiotropic effect of statins, real-world data on overtreatment and undertreatment of lipid-lowering therapies, and the changing LDL-C in targets in the clinical guidelines of dyslipidemias over the years.
Collapse
Affiliation(s)
| | - Ana Torres-Do-Rego
- Internal Medicine, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Grupo (departamento) de investigación Riesgo cardiovascular y lípidos, Instituto de investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Antonio López-Farré
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisco Galeano-Valle
- Internal Medicine, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Grupo (departamento) de investigación Riesgo cardiovascular y lípidos, Instituto de investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Pablo Demelo-Rodriguez
- Internal Medicine, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Grupo (departamento) de investigación Riesgo cardiovascular y lípidos, Instituto de investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis A. Alvarez-Sala-Walther
- Internal Medicine, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Grupo (departamento) de investigación Riesgo cardiovascular y lípidos, Instituto de investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
26
|
Xiao X, Luo Y, Peng D. Updated Understanding of the Crosstalk Between Glucose/Insulin and Cholesterol Metabolism. Front Cardiovasc Med 2022; 9:879355. [PMID: 35571202 PMCID: PMC9098828 DOI: 10.3389/fcvm.2022.879355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/07/2022] [Indexed: 12/19/2022] Open
Abstract
Glucose and cholesterol engage in almost all human physiological activities. As the primary energy substance, glucose can be assimilated and converted into diverse essential substances, including cholesterol. Cholesterol is mainly derived from de novo biosynthesis and the intestinal absorption of diets. It is evidenced that glucose/insulin promotes cholesterol biosynthesis and uptake, which have been targeted by several drugs for lipid-lowering, e.g., bempedoic acid, statins, ezetimibe, and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors. Inversely, these lipid-lowering drugs may also interfere with glucose metabolism. This review would briefly summarize the mechanisms of glucose/insulin-stimulated cholesterol biosynthesis and uptake, and discuss the effect and mechanisms of lipid-lowering drugs and genetic mutations on glucose homeostasis, aiming to help better understand the intricate relationship between glucose and cholesterol metabolism.
Collapse
|
27
|
Fateh ST, Fateh ST, Shekari F, Mahdavi M, Aref AR, Salehi-Najafabadi A. The Effects of Sesquiterpene Lactones on the Differentiation of Human or Animal Cells Cultured In-Vitro: A Critical Systematic Review. Front Pharmacol 2022; 13:862446. [PMID: 35444549 PMCID: PMC9014292 DOI: 10.3389/fphar.2022.862446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/08/2022] [Indexed: 12/03/2022] Open
Abstract
Cellular differentiation is pivotal in health and disease. Interfering with the process of differentiation, such as inhibiting the differentiation of adipocytes and inducing the differentiation of cancer cells, is considered a therapeutic approach. Sesquiterpene lactones, primarily found in plants, have been attracted attention as differentiating/dedifferentiating agents tested on various human or animal cells. However, a consensus on sesquiterpene lactones’ effects and their mechanism of action is required. In this sense, through a systematic review, we have investigated the differentiating/dedifferentiating effects of sesquiterpene lactones on human or animal cells. 13 different cell lines originated from humans, mice, and rats, in addition to the effects of a total of 21 sesquiterpene lactones, were evaluated in the included studies. These components had either inducing, inhibiting, or no effect on the cells, mediating their effects through JAK-STAT, PI3K-Akt, mitogen-activated protein kinases, NFκB, PPARγ pathways. Although nearly all inducing and inhibiting effects were attributed to cancerous and normal cells, respectively, this is likely a result of a biased study design. Few studies reported negative results along with others, and no study was found reporting only negative results. As a result, not only are the effects and mechanism of action of sesquiterpene lactones not vivid but our knowledge and decisions are also misconducted. Moreover, there is a significant knowledge gap regarding the type of evaluated cells, other sesquiterpene lactones, and the involved signaling pathways. In conclusion, sesquiterpene lactones possess significant effects on differentiation status, leading to potentially efficient therapy of obesity, osteoporosis, and cancer. However, reporting negative results and further investigations on other cells, sesquiterpene lactones, and signaling pathways are highly suggested to pave the path of sesquiterpene lactones to the clinic more consciously.
Collapse
Affiliation(s)
- Sepand Tehrani Fateh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Majid Mahdavi
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States.,Translational Sciences, Xsphera Biosciences Inc., Boston, MA, United States
| | - Amir Salehi-Najafabadi
- Department of Microbiology, School of Biology, University College of Science, University of Tehran, Tehran, Iran.,Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran
| |
Collapse
|
28
|
Xu S, Rha SW, Choi BG, Seo HS. The Impact of Age on Statin-Related Glycemia: A Propensity Score-Matched Cohort Study in Korea. Healthcare (Basel) 2022; 10:healthcare10050777. [PMID: 35627914 PMCID: PMC9141400 DOI: 10.3390/healthcare10050777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/17/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to investigate the influence of statin on glycemic control in different age groups. Patients admitted for suspected or confirmed coronary artery disease between January 2005 and December 2013 in Seoul, Korea were initially enrolled. After propensity score matching, 2654 patients (1:1 statin users and non-users) were selected out of total 5041 patients, including 1477 “young” patients (≤60 y) and 1177 elderly patients (>60 y). HbA1c was decreased by 0.04% (±0.86%) in statin non-users. On the contrary, a slight increment of 0.05% (±0.71%) was found in statin users (p < 0.001). The change patterns of HbA1c were constant in both young and elderly patient groups. Furthermore, elderly statin users demonstrated significantly worse glycemic control in serum insulin and homeostatic model assessment—insulin resistance (HOMA-IR) index. In elderly patients, statin users were found to have a 2.61 ± 8.34 μU/mL increment in serum insulin, whereas it was 2.35 ± 6.72 μU/mL for non-users (p = 0.012). Statin users had a 0.78 ± 3.28 increment in HOMA-IR, in contrast to the 0.67 ± 2.51 increment in statin non-users (p = 0.008). In conclusion, statin treatment was associated with adverse glycemic control in the elderly population.
Collapse
Affiliation(s)
- Shaopeng Xu
- Cardiovascular Department, Tianjin Medical University General Hospital, Tianjin 300052, China
- Cardiovascular Center, Division of Cardiology, Korea University Guro Hospital, Seoul 08308, Korea;
- Correspondence: (S.X.); (S.-W.R.)
| | - Seung-Woon Rha
- Cardiovascular Center, Division of Cardiology, Korea University Guro Hospital, Seoul 08308, Korea;
- Correspondence: (S.X.); (S.-W.R.)
| | - Byoung Geol Choi
- Cardiovascular Research Institute, Korea University College of Medicine, Seoul 02841, Korea;
| | - Hong Seog Seo
- Cardiovascular Center, Division of Cardiology, Korea University Guro Hospital, Seoul 08308, Korea;
| |
Collapse
|
29
|
Patel KK, Sehgal VS, Kashfi K. Molecular targets of statins and their potential side effects: Not all the glitter is gold. Eur J Pharmacol 2022; 922:174906. [PMID: 35321818 PMCID: PMC9007885 DOI: 10.1016/j.ejphar.2022.174906] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/12/2022] [Accepted: 03/17/2022] [Indexed: 12/11/2022]
Abstract
Statins are a class of drugs widely used worldwide to manage hypercholesterolemia and the prevention of secondary heart attacks. Currently, available statins vary in terms of their pharmacokinetic and pharmacodynamic profiles. Although the primary target of statins is the inhibition of HMG-CoA reductase (HMGR), the rate-limiting enzyme in cholesterol biosynthesis, statins exhibit many pleiotropic effects downstream of the mevalonate pathway. These pleiotropic effects include the ability to reduce myocardial fibrosis, pathologic cardiac disease states, hypertension, promote bone differentiation, anti-inflammatory, and antitumor effects through multiple mechanisms. Although these pleiotropic effects of statins may be a cause for enthusiasm, there are many adverse effects that, for the most part, are unappreciated and need to be highlighted. These adverse effects include myopathy, new-onset type 2 diabetes, renal and hepatic dysfunction. Although these adverse effects may be relatively uncommon, considering the number of people worldwide who use statins daily, the actual number of people affected becomes quite large. Also, co-administration of statins with several other medications, herbal agents, and foods, which interact through common enzymatic pathways, can have untoward clinical consequences. In this review, we address these concerns.
Collapse
Affiliation(s)
- Kush K Patel
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Viren S Sehgal
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA; Graduate Program in Biology, City University of New York Graduate Center, New York, USA.
| |
Collapse
|
30
|
Tabaei BS, Mousavi SN, Rahimian A, Rostamkhani H, Mellati AA, Jameshorani M. Co-Administration of Vitamin E and Atorvastatin Improves Insulin Sensitivity and Peroxisome Proliferator-Activated Receptor-γ Expression in Type 2 Diabetic Patients: A Randomized Double-Blind Clinical Trial. IRANIAN JOURNAL OF MEDICAL SCIENCES 2022; 47:114-122. [PMID: 35291435 PMCID: PMC8919307 DOI: 10.30476/ijms.2021.89102.1981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/20/2021] [Accepted: 05/26/2021] [Indexed: 11/13/2022]
Abstract
BACKGROUND Negative effects of statins on glucose metabolism have been reported. The present study aimed to investigate the effects of co-administration of vitamin E and atorvastatin on glycemic control in hyperlipidemic patients with type 2 diabetes mellitus (T2DM). METHODS A randomized double-blind clinical trial was conducted at Vali-e-Asr Teaching Hospital (Zanjan, Iran) from July 2017 to March 2018. A total of 30 T2DM female patients were allocated to two groups, namely atorvastatin with placebo (n=15) and atorvastatin with vitamin E (n=15). The patients received daily 20 mg atorvastatin and 400 IU vitamin E or placebo for 12 weeks. Anthropometric and biochemical measures were recorded pre- and post-intervention. Peroxisome proliferator-activated receptor-γ (PPAR-γ) expression was measured in peripheral blood mononuclear cells (PBMCs). Independent sample t test and paired t test were used to analyze between- and within-group variables, respectively. The analysis of covariance (ANCOVA) was used to adjust the effect of baseline variables on the outcomes. P<0.05 was considered statistically significant. RESULTS After baseline adjustment, there was a significant improvement in homeostatic model assessment for insulin resistance (HOMA-IR) (P=0.04) and serum insulin (P<0.001) in the atorvastatin with vitamin E group compared to the atorvastatin with the placebo group. In addition, co-administration of vitamin E with atorvastatin significantly upregulated PPAR-γ expression (OR=5.4, P=0.04) in the PBMCs of T2DM patients. CONCLUSION Co-administration of vitamin E and atorvastatin reduced insulin resistance and improved PPAR-γ mRNA expression. Further studies are required to substantiate our findings. TRIAL REGISTRATION NUMBER IRCT 20170918036256N.
Collapse
Affiliation(s)
- Banafsheh Sadat Tabaei
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyedeh Neda Mousavi
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Nutrition, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Aliasghar Rahimian
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hadi Rostamkhani
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Awsat Mellati
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Maryam Jameshorani
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
31
|
Grunwald SA, Haafke S, Grieben U, Kassner U, Steinhagen-Thiessen E, Spuler S. Statins Aggravate the Risk of Insulin Resistance in Human Muscle. Int J Mol Sci 2022; 23:2398. [PMID: 35216514 PMCID: PMC8876152 DOI: 10.3390/ijms23042398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 01/23/2023] Open
Abstract
Beside their beneficial effects on cardiovascular events, statins are thought to contribute to insulin resistance and type-2 diabetes. It is not known whether these effects are long-term events from statin-treatment or already triggered with the first statin-intake. Skeletal muscle is considered the main site for insulin-stimulated glucose uptake and therefore, a primary target for insulin resistance in the human body. We analyzed localization and expression of proteins related to GLUT4 mediated glucose uptake via AMPKα or AKT in human skeletal muscle tissue from patients with statin-intake >6 months and in primary human myotubes after 96 h statin treatment. The ratio for AMPKα activity significantly increased in human skeletal muscle cells treated with statins for long- and short-term. Furthermore, the insulin-stimulated counterpart, AKT, significantly decreased in activity and protein level, while GSK3ß and mTOR protein expression reduced in statin-treated primary human myotubes, only. However, GLUT4 was normally distributed whereas CAV3 was internalized from plasma membrane around the nucleus in statin-treated primary human myotubes. Statin-treatment activates AMPKα-dependent glucose uptake and remains active after long-term statin treatment. Permanent blocking of its insulin-dependent counterpart AKT activation may lead to metabolic inflexibility and insulin resistance in the long run and may be a direct consequence of statin-treatment.
Collapse
Affiliation(s)
- Stefanie A. Grunwald
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| | - Stefanie Haafke
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| | - Ulrike Grieben
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| | - Ursula Kassner
- Interdisciplinary Lipid Metabolic Center, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (U.K.); (E.S.-T.)
| | - Elisabeth Steinhagen-Thiessen
- Interdisciplinary Lipid Metabolic Center, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (U.K.); (E.S.-T.)
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| |
Collapse
|
32
|
Abdel-Bakky MS, Alqasoumi A, Altowayan WM, Amin E, Darwish MA. Simvastatin mitigates streptozotocin-induced type 1 diabetes in mice through downregulation of ADAM10 and ADAM17. Life Sci 2022; 289:120224. [PMID: 34896343 DOI: 10.1016/j.lfs.2021.120224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND T cell mediates immune response in type 1 diabetes mellitus (T1DM) through its trafficking into pancreatic islets. The role of A Disintigrin And Metalloproteinase 10 (ADAM10) and 17 (ADAM17) in pancreatic T-cells recruitment into the pancreatic islets during T1DM is not known. AIM Explore the role of ADAM10 and ADAM17 in the processing of CXCL16 in T1DM and possible protective effect of simvastatin (SIM) in streptozotocin (STZ)-induced T1DM. MAIN METHODS Balb/c mice were classified into 4 groups, 10 each. Control group received buffer while SIM group received 50 mg/kg, i.p daily for 12 days starting from day 4 of the experiment. Diabetic group; received STZ (55 mg/kg, i.p.) for 5 consecutive days starting from day 1 of the experiment. SIM + STZ group; received SIM (50 mg/kg, i.p.) daily for 12 days and STZ (55 mg/kg, i.p.) for 5 consecutive days. Biochemical, inflammatory and apoptotic markers as well as expression of CXCL16, ADAM10, NF-κB and pancreatic T-cells expression were analyzed. KEY FINDINGS Significant increase in biochemical, inflammatory, apoptotic parameters, expression of ADAM10, ADAM17, CXCL16, NF-κB, and infiltrated T-cells to the pancreatic islets were found in STZ group. SIM treatment in the presence of STZ improved biochemical and inflammatory parameters as well as it reduced the expression of CXCL16, ADAM10, ADAM17, NF-κΒ, T-cells migration and apoptosis in the pancreatic islets. SIGNIFICANCE SIM mitigated pancreatic β-cell death induced by STZ through down regulation of ADAM10, ADAM17and CXCL16. Therefore, ADAM10/ADAM17 and CXCL16 may serve as novel therapeutic targets for T1DM.
Collapse
Affiliation(s)
- Mohamed Sadek Abdel-Bakky
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt.
| | - Abdulmajeed Alqasoumi
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | | | - Elham Amin
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Mostafa Assem Darwish
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt
| |
Collapse
|
33
|
Wu P, Moon JY, Daghlas I, Franco G, Porneala BC, Ahmadizar F, Richardson TG, Isaksen JL, Hindy G, Yao J, Sitlani CM, Raffield LM, Yanek LR, Feitosa MF, Cuadrat RR, Qi Q, Arfan Ikram M, Ellervik C, Ericson U, Goodarzi MO, Brody JA, Lange L, Mercader JM, Vaidya D, An P, Schulze MB, Masana L, Ghanbari M, Olesen MS, Cai J, Guo X, Floyd JS, Jäger S, Province MA, Kalyani RR, Psaty BM, Orho-Melander M, Ridker PM, Kanters JK, Uitterlinden A, Davey Smith G, Gill D, Kaplan RC, Kavousi M, Raghavan S, Chasman DI, Rotter JI, Meigs JB, Florez JC, Dupuis J, Liu CT, Merino J. Obesity Partially Mediates the Diabetogenic Effect of Lowering LDL Cholesterol. Diabetes Care 2022; 45:232-240. [PMID: 34789503 PMCID: PMC8753762 DOI: 10.2337/dc21-1284] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/15/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE LDL cholesterol (LDLc)-lowering drugs modestly increase body weight and type 2 diabetes risk, but the extent to which the diabetogenic effect of lowering LDLc is mediated through increased BMI is unknown. RESEARCH DESIGN AND METHODS We conducted summary-level univariable and multivariable Mendelian randomization (MR) analyses in 921,908 participants to investigate the effect of lowering LDLc on type 2 diabetes risk and the proportion of this effect mediated through BMI. We used data from 92,532 participants from 14 observational studies to replicate findings in individual-level MR analyses. RESULTS A 1-SD decrease in genetically predicted LDLc was associated with increased type 2 diabetes odds (odds ratio [OR] 1.12 [95% CI 1.01, 1.24]) and BMI (β = 0.07 SD units [95% CI 0.02, 0.12]) in univariable MR analyses. The multivariable MR analysis showed evidence of an indirect effect of lowering LDLc on type 2 diabetes through BMI (OR 1.04 [95% CI 1.01, 1.08]) with a proportion mediated of 38% of the total effect (P = 0.03). Total and indirect effect estimates were similar across a number of sensitivity analyses. Individual-level MR analyses confirmed the indirect effect of lowering LDLc on type 2 diabetes through BMI with an estimated proportion mediated of 8% (P = 0.04). CONCLUSIONS These findings suggest that the diabetogenic effect attributed to lowering LDLc is partially mediated through increased BMI. Our results could help advance understanding of adipose tissue and lipids in type 2 diabetes pathophysiology and inform strategies to reduce diabetes risk among individuals taking LDLc-lowering medications.
Collapse
Affiliation(s)
- Peitao Wu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Jee-Young Moon
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Iyas Daghlas
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Giulianini Franco
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Bianca C. Porneala
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA
| | - Fariba Ahmadizar
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tom G. Richardson
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
- Novo Nordisk Research Centre Oxford, Old Road Campus, Oxford, U.K
| | - Jonas L. Isaksen
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Georgy Hindy
- Department of Clinical Sciences, Skåne University Hospital Malmo Clinical Research Center, Lund University, Malmo, Sweden
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Colleen M. Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Laura M. Raffield
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Lisa R. Yanek
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Rafael R.C. Cuadrat
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Christina Ellervik
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Department of Research, Region Zealand, Sorø, Denmark
| | - Ulrika Ericson
- Department of Clinical Sciences, Skåne University Hospital Malmo Clinical Research Center, Lund University, Malmo, Sweden
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Leslie Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Josep M. Mercader
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Dhananjay Vaidya
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ping An
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Matthias B. Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Lluis Masana
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Rovira i Virgil University, IISPV, Reus, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Morten S. Olesen
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jianwen Cai
- Collaborative Studies Coordinating Center, Department of Biostatistics, The University of North Carolina at Chapel Hill, NC
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - James S. Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
| | - Susanne Jäger
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Michael A. Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Rita R. Kalyani
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
- Department of Health Services, University of Washington, Seattle, WA
| | - Marju Orho-Melander
- Department of Clinical Sciences, Skåne University Hospital Malmo Clinical Research Center, Lund University, Malmo, Sweden
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Jørgen K. Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andre Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Dipender Gill
- Novo Nordisk Research Centre Oxford, Old Road Campus, Oxford, U.K
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, U.K
- Clinical Pharmacology and Therapeutics Section, Institute of Medical and Biomedical Education and Institute for Infection and Immunity, St George’s, University of London, London, U.K
- Clinical Pharmacology Group, Pharmacy and Medicines Directorate, St George’s University Hospitals NHS Foundation Trust, London, U.K
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle WA
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sridharan Raghavan
- Department of Veterans Affairs Medical Center, Eastern Colorado Health Care System, Denver, CO
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Denver, CO
| | - Daniel I. Chasman
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - James B. Meigs
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Jose C. Florez
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Jordi Merino
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Rovira i Virgil University, IISPV, Reus, Spain
| |
Collapse
|
34
|
Neves JC, Neves JS, Neves C, Carvalho D. Predictors of the effectiveness of insulin pumps in patients with type 1 diabetes mellitus. Endocrine 2022; 75:119-128. [PMID: 34339007 DOI: 10.1007/s12020-021-02837-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Insulin pump therapy has become the preferential treatment for type 1 diabetes (T1D) as it mimics the physiological secretion of insulin better than multiple daily injections. However, not all patients improve with insulin pump therapy. This study aims to determine the predictors of the effectiveness of insulin pumps in T1D. METHODS We conducted a retrospective observational study of patients who started insulin pumps. Data from four timepoints (before, at 6, 12, and 36 months) were evaluated for outcomes of glycemic control and safety. The association of baseline predictors with outcomes was analyzed using linear and logistic regression models. RESULTS We evaluated 136 patients (57.4% females, age 36 ± 12 years, duration of T1D 14 ± 9 years). During the follow-up, there was a mean decrease of HbA1c of 0.9 ± 1.2%. The improvement in HbA1c was independent of sex, age, and duration of T1D. Higher baseline HbA1c, family history of diabetes, and not being treated with statins were predictors of improvement in HbA1c. Not being treated with statins and higher baseline HbA1c predicted improvement in HbA1c without worsening hypoglycemia. History of hypoglycemia was a predictor of severe hypoglycemia. Family history, higher baseline HbA1c, and psychological/psychiatric disorders were predictors of ketoacidosis. CONCLUSION Benefits of insulin pump were independent of sex, age, and duration of T1D. Baseline HbA1c, family history of diabetes, treatment with statins, history of hypoglycemia, and psychological/psychiatric disorders were predictors of outcomes, and may allow the identification of patients who benefit most from insulin pump therapy or who are at increased risk of complications.
Collapse
Affiliation(s)
- Joana Camões Neves
- Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário de São João, Porto, Portugal.
- Faculty of Medicine, University of Porto, Porto, Portugal.
| | - João Sérgio Neves
- Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário de São João, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Surgery and Physiology, Cardiovascular Research Unit, Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health Sciences, University of Porto, Porto, Portugal
| | - Celestino Neves
- Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário de São João, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health Sciences, University of Porto, Porto, Portugal
| | - Davide Carvalho
- Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário de São João, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health Sciences, University of Porto, Porto, Portugal
| |
Collapse
|
35
|
Missikpode C, Durazo-Arvizu RA, Cooper RS, OʼBrien MJ, Castaneda SF, Talavera GA, Gallo LC, Llabre MM, Perera MJ, Perreira KM, Ricardo AC, Pirzada A, Lash JP, Daviglus M. Cardiovascular disease and risk of incident diabetes mellitus: Findings from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). J Diabetes 2021; 13:1043-1053. [PMID: 34536057 PMCID: PMC8942503 DOI: 10.1111/1753-0407.13224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 07/26/2021] [Accepted: 09/02/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Studies have reported an association between prevalent cardiovascular disease (CVD) and risk of diabetes mellitus (DM). However, factors that may explain the association remain unclear. We examined the association of prevalent CVD with incident DM and assessed whether weight gain and medication use may explain the association. METHODS Data from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL) Visit 1 (2008-2011) and Visit 2 (2014-2017) were used to compare incidence of DM among individuals with and without self-reported CVD at Visit 1. A total of 1899 individuals with self-reported CVD were matched to controls free of self-reported CVD at Visit 1 using 1:1 propensity score matching. Covariates included in the propensity model were sociodemographic characteristics, lifestyle factors, comorbid conditions, and study site. The effect of self-reported CVD on incident DM was examined using a generalized estimating equation. The mediating effects of weight gain and use of cardiovascular medications were evaluated. RESULTS Covariate distributions were similar among individuals with and without self-reported CVD. The incidence of DM among persons with self-reported CVD was 15.3% vs 12.7% among those without self-reported CVD. Compared to individuals without self-reported CVD, individuals with self-reported CVD had a 24% increased risk for incident DM (odds ratio = 1.24, 95% confidence interval = 1.01, 1.51). The association between self-reported CVD and DM was mediated by the use of beta-blockers (proportion explained = 25.4%), statins (proportion explained = 18%), and diuretics (proportion explained = 8%). We found that weight gain did not explain the observed association. CONCLUSIONS Prevalent cardiovascular disease was associated with a significant increased risk of incident diabetes. The observed association was partially explained by some medications used to manage CVD.
Collapse
Affiliation(s)
- Celestin Missikpode
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ramon A. Durazo-Arvizu
- Department of Public Health Sciences, Loyola University Chicago Health Sciences Campus, Maywood, Illinois, USA
| | - Richard S. Cooper
- Department of Public Health Sciences, Loyola University Chicago Health Sciences Campus, Maywood, Illinois, USA
| | | | - Sheila F. Castaneda
- Department of Psychology, San Diego State University, San Diego, California, USA
| | - Gregory A. Talavera
- Department of Psychology, San Diego State University, San Diego, California, USA
| | - Linda C. Gallo
- Department of Psychology, San Diego State University, San Diego, California, USA
| | - Maria M. Llabre
- Department of Psychology, University of Miami, Miami, Florida, USA
| | - Marisa J. Perera
- Department of Psychology, University of Miami, Miami, Florida, USA
| | - Krista M. Perreira
- Collaborative Studies Coordinating Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ana C. Ricardo
- Division of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Amber Pirzada
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, Illinois, USA
| | - James P. Lash
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, Illinois, USA
- Division of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Martha Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
36
|
Bonilha I, Hajduch E, Luchiari B, Nadruz W, Le Goff W, Sposito AC. The Reciprocal Relationship between LDL Metabolism and Type 2 Diabetes Mellitus. Metabolites 2021; 11:metabo11120807. [PMID: 34940565 PMCID: PMC8708656 DOI: 10.3390/metabo11120807] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes mellitus and insulin resistance feature substantial modifications of the lipoprotein profile, including a higher proportion of smaller and denser low-density lipoprotein (LDL) particles. In addition, qualitative changes occur in the composition and structure of LDL, including changes in electrophoretic mobility, enrichment of LDL with triglycerides and ceramides, prolonged retention of modified LDL in plasma, increased uptake by macrophages, and the formation of foam cells. These modifications affect LDL functions and favor an increased risk of cardiovascular disease in diabetic individuals. In this review, we discuss the main findings regarding the structural and functional changes in LDL particles in diabetes pathophysiology and therapeutic strategies targeting LDL in patients with diabetes.
Collapse
Affiliation(s)
- Isabella Bonilha
- Cardiology Division, Atherosclerosis and Vascular Biology Laboratory (AtheroLab), State University of Campinas (Unicamp), Campinas 13083-887, Brazil; (I.B.); (B.L.)
| | - Eric Hajduch
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, F-75006 Paris, France;
| | - Beatriz Luchiari
- Cardiology Division, Atherosclerosis and Vascular Biology Laboratory (AtheroLab), State University of Campinas (Unicamp), Campinas 13083-887, Brazil; (I.B.); (B.L.)
| | - Wilson Nadruz
- Cardiology Division, Cardiovascular Pathophysiology Laboratory, State University of Campinas (Unicamp), Campinas 13083-887, Brazil;
| | - Wilfried Le Goff
- Unité de Recherche sur les Maladies Cardiovasculaires, le Métabolisme et la Nutrition, ICAN, Inserm, Sorbonne Université, F-75013 Paris, France;
| | - Andrei C. Sposito
- Cardiology Division, Atherosclerosis and Vascular Biology Laboratory (AtheroLab), State University of Campinas (Unicamp), Campinas 13083-887, Brazil; (I.B.); (B.L.)
- Correspondence: ; Tel.: +55-19-3521-7098; Fax: +55-19-3289-410
| |
Collapse
|
37
|
Effect of Metformin and Simvastatin in Inhibiting Proadipogenic Transcription Factors. Curr Issues Mol Biol 2021; 43:2082-2097. [PMID: 34940118 PMCID: PMC8929042 DOI: 10.3390/cimb43030144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/13/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is a multifactorial chronic disease characterized by the excessive accumulation of fat in adipose tissue driven by hypertrophy and hyperplasia of adipocytes through adipogenesis. Adipogenesis plays a key role in the development of obesity and related metabolic disorders, which makes it potential target for the therapeutic approach to obesity. An increasing number of studies confirm the pleiotropic action of the combined treatment with metformin and statins, suggesting their anti-hypertensive, anti-inflammatory, and anti-adipogenic effect. The aim of this study was to analyze the effect of different doses of metformin (MET) and simvastatin (SIM) on the expression of key transcription factors of adipogenesis. Mouse 3T3-L1 preadipocytes were induced to differentiation in adipogenic medium with sustained MET and SIM treatment to assess the effect on adipogenesis. Nine days after initiating adipogenesis, the cells were prepared for further experiments, including Oil Red O staining, RT-PCR, Western blotting, and immunocytochemistry. Treating the cells with the combination of MET and SIM slightly reduced the intensity of Oil Red O staining compared with the control group, and down-regulated mRNA and protein expression of PPARγ, C/EBPα, and SREBP-1C. In conclusion, the inhibitory effect of MET and SIM on adipocyte differentiation, as indicated by decreased lipid accumulation, appears to be mediated through the down-regulation of adipogenic transcription factors, peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer binding pro-tein α (C/EBPα), and sterol regulatory element-binding protein 1 (SREBP-1C).
Collapse
|
38
|
de Pádua Borges R, Degobi NAH, Bertoluci MC. Choosing statins: a review to guide clinical practice. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2021; 64:639-653. [PMID: 33166435 PMCID: PMC10528630 DOI: 10.20945/2359-3997000000306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/13/2020] [Indexed: 11/23/2022]
Abstract
Statins are among the most widely prescribed medicines in the world and have proved their value in reducing cardiovascular events and mortality. Many patients report adverse effects that lead to interruption of treatment. This review aims to individualize statin treatment, considering efficacy for reducing cardiovascular risk and safety, in the setting of specific diseases, to minimize the side effects and improve compliance. We gathered evidence that may help clinicians to choose specific statins in different clinical situations, such as the risk of new diabetes, chronic kidney disease, liver disease, human immunodeficiency virus infection, organ transplant, heart failure and elderly people. Efficacy of statins is well established in a large number of clinical conditions. Therefore, main objective is to revise statin in specific clinical settings, based on pharmacokinetics, safety, drug metabolism and interactions to provide the best choice in different clinical scenarios.
Collapse
Affiliation(s)
- Roberta de Pádua Borges
- Programa de Pós-Graduação em Endocrinologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brasil
| | - Nathália Abi Habib Degobi
- Serviço de Endocrinologia, Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brasil
| | - Marcello Casaccia Bertoluci
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brasil
- Serviço de Endocrinologia, Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brasil,
- Departamento de Medicina Interna, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brasil
| |
Collapse
|
39
|
Abbasi F, Lamendola C, Harris CS, Harris V, Tsai MS, Tripathi P, Abbas F, Reaven G, Reaven P, Snyder MP, Kim SH, Knowles JW. Statins Are Associated With Increased Insulin Resistance and Secretion. Arterioscler Thromb Vasc Biol 2021; 41:2786-2797. [PMID: 34433298 PMCID: PMC8551023 DOI: 10.1161/atvbaha.121.316159] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/09/2021] [Indexed: 11/16/2022]
Abstract
Objective Statin treatment reduces the risk of atherosclerotic cardiovascular disease but is associated with a modest increased risk of type 2 diabetes, especially in those with insulin resistance or prediabetes. Our objective was to determine the physiological mechanism for the increased type 2 diabetes risk. Approach and Results We conducted an open-label clinical trial of atorvastatin 40 mg daily in adults without known atherosclerotic cardiovascular disease or type 2 diabetes at baseline. The co-primary outcomes were changes at 10 weeks versus baseline in insulin resistance as assessed by steady-state plasma glucose during the insulin suppression test and insulin secretion as assessed by insulin secretion rate area under the curve (ISRAUC) during the graded-glucose infusion test. Secondary outcomes included glucose and insulin, both fasting and during oral glucose tolerance test. Of 75 participants who enrolled, 71 completed the study (median age 61 years, 37% women, 65% non-Hispanic White, median body mass index, 27.8 kg/m2). Atorvastatin reduced LDL (low-density lipoprotein)-cholesterol (median decrease 53%, P<0.001) but did not change body weight. Compared with baseline, atorvastatin increased insulin resistance (steady-state plasma glucose) by a median of 8% (P=0.01) and insulin secretion (ISRAUC) by a median of 9% (P<0.001). There were small increases in oral glucose tolerance test glucoseAUC (median increase, 0.05%; P=0.03) and fasting insulin (median increase, 7%; P=0.01). Conclusions In individuals without type 2 diabetes, high-intensity atorvastatin for 10 weeks increases insulin resistance and insulin secretion. Over time, the risk of new-onset diabetes with statin use may increase in individuals who become more insulin resistant but are unable to maintain compensatory increases in insulin secretion.
Collapse
Affiliation(s)
- Fahim Abbasi
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, USA
- Cardiovascular Institute, Stanford University, Stanford, California, USA
- Department of Medicine, Stanford University, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
| | - Cindy Lamendola
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, USA
- Cardiovascular Institute, Stanford University, Stanford, California, USA
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Chelsea S. Harris
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, USA
- Cardiovascular Institute, Stanford University, Stanford, California, USA
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Vander Harris
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, USA
- Cardiovascular Institute, Stanford University, Stanford, California, USA
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Ming-Shian Tsai
- Cardiovascular Institute, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
| | - Pragya Tripathi
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, USA
- Cardiovascular Institute, Stanford University, Stanford, California, USA
| | - Fakhar Abbas
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, USA
- Cardiovascular Institute, Stanford University, Stanford, California, USA
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Gerald Reaven
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, USA
- Cardiovascular Institute, Stanford University, Stanford, California, USA
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Peter Reaven
- University of Arizona and Phoenix VA Health Care System, Phoenix, Arizona, USA
| | - Michael P. Snyder
- Cardiovascular Institute, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
| | - Sun H. Kim
- Department of Medicine, Stanford University, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
- Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, California, USA
| | - Joshua W. Knowles
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, USA
- Cardiovascular Institute, Stanford University, Stanford, California, USA
- Department of Medicine, Stanford University, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
- Stanford Prevention Research Center, Stanford University, Stanford, California, USA
| |
Collapse
|
40
|
Guber K, Pemmasani G, Malik A, Aronow WS, Yandrapalli S, Frishman WH. Statins and Higher Diabetes Mellitus Risk: Incidence, Proposed Mechanisms, and Clinical Implications. Cardiol Rev 2021; 29:314-322. [PMID: 32947479 DOI: 10.1097/crd.0000000000000348] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors are ubiquitously prescribed for cardiovascular disease (CVD) prevention and treatment. However, the use of statins has been linked to the development of new-onset diabetes mellitus (NODM), which could possibly increase future CVD risk. This phenomenon necessitates a clear discussion of the possible etiologies of this relationship and its broader clinical consequences. We discuss the reported incidence of NODM in statin users through a rigorous review of data from metaanalyses of randomized control trials examining this association. We also highlight the various possible mechanisms responsible for the development of statin-induced diabetes mellitus. Finally, we examine the clinical implications of this effect on future CVD risk and identify specific patient factors that can be used for risk-stratification strategies. Data from 14 randomized control trials metaanalyses suggest a 9-33% higher risk of NODM with statin use. Several cellular, molecular, and genetic mechanisms, as well as lifestyle habits, have been identified as potential underlying factors responsible for this elevated risk. The principle mode of the diabetogenic action of statins is still unclear, though it is likely the result of a complex interplay of pancreatic and extrapancreatic effects. It is understood that patient populations with a greater predisposition to diabetes mellitus, and those with thicker epicardial adiposity are more at risk for the development of statin-induced NODM. Despite these observations, robust data from a variety of investigations suggest that the CVD preventative benefits of statin treatment significantly outweigh the risks associated with the development of NODM. Nevertheless, further study must better identify the causative mechanisms involved in this process, its natural history, and the unique factors that will help clinicians risk stratify and appropriately monitor patients on statin therapy.
Collapse
Affiliation(s)
- Kenneth Guber
- From the Department of Medicine, New York Presbyterian-Columbia University Medical Center, New York, NY
| | | | | | | | | | - William H Frishman
- Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| |
Collapse
|
41
|
Drugs Interfering with Insulin Resistance and Their Influence on the Associated Hypermetabolic State in Severe Burns: A Narrative Review. Int J Mol Sci 2021; 22:ijms22189782. [PMID: 34575946 PMCID: PMC8466307 DOI: 10.3390/ijms22189782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 12/31/2022] Open
Abstract
It has become widely accepted that insulin resistance and glucose hypermetabolism can be linked to acute pathologies, such as burn injury, severe trauma, or sepsis. Severe burns can determine a significant increase in catabolism, having an important effect on glucose metabolism and on muscle protein metabolism. It is imperative to acknowledge that these alterations can lead to increased mortality through organ failure, even when the patients survive the initial trauma caused by the burn. By limiting the peripheral use of glucose with consequent hyperglycemia, insulin resistance determines compensatory increased levels of insulin in plasma. However, the significant alterations in cellular metabolism lead to a lack of response to insulin's anabolic functions, as well as to a decrease in its cytoprotective role. In the end, via pathological insulin signaling associated with increased liver gluconeogenesis, elevated levels of glucose are detected in the blood. Several cellular mechanisms have been incriminated in the development of insulin resistance in burns. In this context, the main aim of this review article is to summarize some of the drugs that might interfere with insulin resistance in burns, taking into consideration that such an approach can significantly improve the prognosis of the burned patient.
Collapse
|
42
|
Trias F, Pintó X, Corbella E, Suárez-Tembra M, Ruíz-García A, Díaz-Díaz JL, Sánchez-Ruíz-Granado E, Sarasa I, Martínez-Porqueras R, Rodríguez-Sánchez MA, Corbella X. Differences in the diabetogenic effect of statins in patients with prediabetes. The PRELIPID study. Med Clin (Barc) 2021; 158:531-539. [PMID: 34517987 DOI: 10.1016/j.medcli.2021.06.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Statins are used with the understanding that a slightly increased risk of diabetes is outweighed by their cardiovascular benefits. However, it may be necessary to reconsider whether statin therapy really increase this risk mainly in the population with prediabetes. METHODS A multicenter, cross-sectional, observational study was conducted to assess the relationship between statin therapy and glucose metabolism in 407 patients aged 63.1 years (11SD) diagnosed with dyslipidemia and prediabetes treated in specialized lipid clinics in Spain. RESULTS Significant differences were found in HbA1c values among treatment groups (p=0.015). Patients treated with pitavastatin (1-4mg/day) showed the lowest HbA1c levels, with significant differences compared to patients treated with atorvastatin 40-80mg/day (p=0.016) and simvastatin 10-40mg/day (p=0.036). By contrast, patients treated with atorvastatin 40-80mg/day showed the highest HbA1c levels compared to those receiving atorvastatin 10-20mg/day (p=0.003), pitavastatin 1-4mg/day (p=0.016), pravastatin 20-40mg/day (p=0.027), rosuvastatin 5-10mg/day (p=0.043), and no statin treatment (p=0.004). Patients treated with simvastatin 10-40mg/day also had higher values than those treated with atorvastatin 10-20mg/day (p=0.016) and pitavastatin 1-4mg/day (p=0.036) or with no statin treatment (p=0.018). CONCLUSIONS This study suggests that there are differences in the diabetogenic effect of statins. Simvastatin and high doses of atorvastatin may be associated with greater impairment in glucose metabolism than pitavastatin and other statins with less lipid-lowering potency such as pravastatin.
Collapse
Affiliation(s)
- Ferran Trias
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell, Spain; Fundación para la Investigación y Prevención de Enfermedades Cardiovasculares (FIPEC), Spain
| | - Xavier Pintó
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell, Spain; Fundación para la Investigación y Prevención de Enfermedades Cardiovasculares (FIPEC), Spain; Centro de Investigación Biomédica en Red, Fisiopatologia de la Obesidad y Nutrición CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Facultad de Medicina, Universidad de Barcelona, Spain.
| | - Emili Corbella
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell, Spain; Fundación para la Investigación y Prevención de Enfermedades Cardiovasculares (FIPEC), Spain; Centro de Investigación Biomédica en Red, Fisiopatologia de la Obesidad y Nutrición CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Antonio Ruíz-García
- Centro de Salud Universitario Pinto, Unidad de Lípidos y Prevención Cardiovascular, Pinto, Madrid, Spain
| | - José Luis Díaz-Díaz
- Unidad de Lípidos y Riesgo Cardiovascular, Complejo Hospitalario Universitario, A Coruña, Spain
| | | | - Itziar Sarasa
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell, Spain; Fundación para la Investigación y Prevención de Enfermedades Cardiovasculares (FIPEC), Spain
| | - Raul Martínez-Porqueras
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell, Spain; Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Maria Angeles Rodríguez-Sánchez
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell, Spain; Fundación para la Investigación y Prevención de Enfermedades Cardiovasculares (FIPEC), Spain
| | - Xavier Corbella
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell, Spain; Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| |
Collapse
|
43
|
Sarsenbayeva A, Jui BN, Fanni G, Barbosa P, Ahmed F, Kristófi R, Cen J, Chowdhury A, Skrtic S, Bergsten P, Fall T, Eriksson JW, Pereira MJ. Impaired HMG-CoA Reductase Activity Caused by Genetic Variants or Statin Exposure: Impact on Human Adipose Tissue, β-Cells and Metabolome. Metabolites 2021; 11:574. [PMID: 34564389 PMCID: PMC8468287 DOI: 10.3390/metabo11090574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 11/20/2022] Open
Abstract
Inhibition of 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase is associated with an increased risk of new-onset type 2 diabetes. We studied the association of genetic or pharmacological HMG-CoA reductase inhibition with plasma and adipose tissue (AT) metabolome and AT metabolic pathways. We also investigated the effects of statin-mediated pharmacological inhibition of HMG-CoA reductase on systemic insulin sensitivity by measuring the HOMA-IR index in subjects with or without statin therapy. The direct effects of simvastatin (20-250 nM) or its active metabolite simvastatin hydroxy acid (SA) (8-30 nM) were investigated on human adipocyte glucose uptake, lipolysis, and differentiation and pancreatic insulin secretion. We observed that the LDL-lowering HMGCR rs12916-T allele was negatively associated with plasma phosphatidylcholines and sphingomyelins, and HMGCR expression in AT was correlated with various metabolic and mitochondrial pathways. Clinical data showed that statin treatment was associated with HOMA-IR index after adjustment for age, sex, BMI, HbA1c, LDL-c levels, and diabetes status in the subjects. Supra-therapeutic concentrations of simvastatin reduced glucose uptake in adipocytes and normalized fatty acid-induced insulin hypersecretion from β-cells. Our data suggest that inhibition of HMG-CoA reductase is associated with insulin resistance. However, statins have a very mild direct effect on AT and pancreas, hence, other tissues as the liver or muscle appear to be of greater importance.
Collapse
Affiliation(s)
- Assel Sarsenbayeva
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Bipasha Nandi Jui
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Giovanni Fanni
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Pedro Barbosa
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Fozia Ahmed
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Robin Kristófi
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Jing Cen
- Department of Medical Cell Biology, Uppsala University, 751 85 Uppsala, Sweden; (J.C.); (A.C.); (P.B.)
| | - Azazul Chowdhury
- Department of Medical Cell Biology, Uppsala University, 751 85 Uppsala, Sweden; (J.C.); (A.C.); (P.B.)
| | - Stanko Skrtic
- Innovation Strategies & External Liaison, Pharmaceutical Technologies & Development, AstraZeneca, 431 83 Gothenburg, Sweden;
- Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Peter Bergsten
- Department of Medical Cell Biology, Uppsala University, 751 85 Uppsala, Sweden; (J.C.); (A.C.); (P.B.)
| | - Tove Fall
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Jan W. Eriksson
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Maria J. Pereira
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| |
Collapse
|
44
|
Lee D, Kim JY, Kim HW, Yoo JE, Kang KS. Combined Beneficial Effect of Genistein and Atorvastatin on Adipogenesis in 3T3-L1 Adipocytes. Biomolecules 2021; 11:biom11071052. [PMID: 34356676 PMCID: PMC8301876 DOI: 10.3390/biom11071052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
Genistein (4,5,7-trihydroxyisoflavone) is abundant in various dietary vegetables, especially soybeans, and is known to have not only an estrogenic effect but also an antiadipogenic effect. Atorvastatin (dihydroxy monocarboxylic acid) is a statin used to prevent heart disease. Although genistein and atorvastatin have been reported to possess antiadipogenic effects, their combined effects are still unclear. The aim of the current study was to explore whether the combination of genistein and atorvastatin at low concentrations significantly suppresses adipogenesis in a murine preadipocyte cell line (3T3-L1) compared to treatment with genistein or atorvastatin alone. Our results showed that cotreatment with 50 µM genistein and 50 nM atorvastatin significantly suppressed preadipocyte differentiation, whereas when each compound was used alone, there was no inhibitory effect. Additionally, cotreatment with genistein and atorvastatin significantly downregulated adipogenic marker proteins, including mitogen-activated protein kinases (MAPKs), peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer-binding protein alpha (C/EBPα), glucocorticoid receptor (GR), and CCAAT/enhancer-binding protein β (C/EBPβ). This is the first evidence of the combined antiadipogenic effects of genistein and atorvastatin. Although additional experiments are required, combinational treatment with genistein and atorvastatin may be an alternative treatment for menopause-associated lipid metabolic disorders and obesity.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
| | - Ji-Youn Kim
- Department of Obstetrics and Gynecology, College of Korean Medicine, Daejeon University, Daejeon 35235, Korea; (J.-Y.K.); (H.-W.K.)
| | - Hae-Won Kim
- Department of Obstetrics and Gynecology, College of Korean Medicine, Daejeon University, Daejeon 35235, Korea; (J.-Y.K.); (H.-W.K.)
| | - Jeong-Eun Yoo
- Department of Obstetrics and Gynecology, College of Korean Medicine, Daejeon University, Daejeon 35235, Korea; (J.-Y.K.); (H.-W.K.)
- Correspondence: (J.-E.Y.); (K.S.K.); Tel.: +82-42-470-9139 (J.-E.Y.); +82-31-750-5402 (K.S.K.)
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
- Correspondence: (J.-E.Y.); (K.S.K.); Tel.: +82-42-470-9139 (J.-E.Y.); +82-31-750-5402 (K.S.K.)
| |
Collapse
|
45
|
Haxhi J, Thompson PD. Rationale for the use of metformin and exercise to counteract statin-associated side effects. Int J Clin Pract 2021; 75:e13900. [PMID: 33277775 DOI: 10.1111/ijcp.13900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/01/2020] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Statins are the most widely prescribed drugs for lowering low-density lipoprotein cholesterol (LDL-C) and reducing cardiovascular morbidity and mortality. They are usually well-tolerated, but have two main safety concerns: statin-associated muscle symptoms (SAMS) and new-onset type 2 diabetes (NOD). METHODS A PubMed search was carried out using the following key words were used: statins, statin-associated muscle symptoms, statin myalgia, statin-associated diabetes, metformin and statins, exercise and statins. RESULTS Mitochondrial damage and muscle atrophy are likely the central mechanisms producing SAMS, whereas decreased glucose transport, fatty acid oxidation and insulin secretion are likely involved in the development of NOD. Metformin and exercise training share many pathways that could potentially contrast SAMS and NOD. Clinical evidence also supports the combination of statins with metformin and exercise. CONCLUSION This combination appears attractive both from a clinical and an economical viewpoint, since all three therapies are highly cost-effective and their combination could result in diabetes and cardiovascular disease prevention.
Collapse
Affiliation(s)
- Jonida Haxhi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- Division of Cardiology, Hartford Hospital, Hartford, CT, USA
| | - Paul D Thompson
- Division of Cardiology, Hartford Hospital, Hartford, CT, USA
| |
Collapse
|
46
|
Bell DSH, Goncalves E. Diabetogenic effects of cardioprotective drugs. Diabetes Obes Metab 2021; 23:877-885. [PMID: 33319474 DOI: 10.1111/dom.14295] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/22/2020] [Accepted: 12/07/2020] [Indexed: 01/11/2023]
Abstract
Drugs that protect against cardiovascular events in the patient with diabetes may also positively or negatively affect glycaemic control in the patient with established diabetes and may induce the development of diabetes in the predisposed patient. Mainly through increasing insulin resistance, beta-blockers, statins and high-dose diuretics have the potential to worsen glycaemic control. Dihydropyridine calcium channel blockers, low-dose diuretics, vasodilating beta-blockers, alpha-blockers and pitavastatin have little or no effect on glycaemic control. Blockers of the renin-angiotensin-aldosterone system, colesevelam, ranolazine and verapamil, through slowing breakdown of bradykinin, vasodilation, increasing cholecystokinin levels, blocking sodium channels and decreasing beta cell apoptosis, may improve glycaemic control and avoid the development of diabetes.
Collapse
|
47
|
Carcamo-Orive I, Henrion MYR, Zhu K, Beckmann ND, Cundiff P, Moein S, Zhang Z, Alamprese M, D’Souza SL, Wabitsch M, Schadt EE, Quertermous T, Knowles JW, Chang R. Predictive network modeling in human induced pluripotent stem cells identifies key driver genes for insulin responsiveness. PLoS Comput Biol 2020; 16:e1008491. [PMID: 33362275 PMCID: PMC7790417 DOI: 10.1371/journal.pcbi.1008491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/07/2021] [Accepted: 11/03/2020] [Indexed: 12/16/2022] Open
Abstract
Insulin resistance (IR) precedes the development of type 2 diabetes (T2D) and increases cardiovascular disease risk. Although genome wide association studies (GWAS) have uncovered new loci associated with T2D, their contribution to explain the mechanisms leading to decreased insulin sensitivity has been very limited. Thus, new approaches are necessary to explore the genetic architecture of insulin resistance. To that end, we generated an iPSC library across the spectrum of insulin sensitivity in humans. RNA-seq based analysis of 310 induced pluripotent stem cell (iPSC) clones derived from 100 individuals allowed us to identify differentially expressed genes between insulin resistant and sensitive iPSC lines. Analysis of the co-expression architecture uncovered several insulin sensitivity-relevant gene sub-networks, and predictive network modeling identified a set of key driver genes that regulate these co-expression modules. Functional validation in human adipocytes and skeletal muscle cells (SKMCs) confirmed the relevance of the key driver candidate genes for insulin responsiveness. Insulin resistance is characterized by a defective response (“resistance”) to normal insulin concentrations to uptake the glucose present in the blood, and is the underlying condition that leads to type 2 diabetes (T2D) and increases the risk of cardiovascular disease. It is estimated that 25–33% of the US population are insulin resistant enough to be at risk of serious clinical consequences. For more than a decade, large population studies have tried to discover the genes that participate in the development of insulin resistance, but without much success. It is now increasingly clear that the complex genetic nature of insulin resistance requires novel approaches centered in patient specific cellular models. To fill this gap, we have generated an induced pluripotent stem cell (iPSC) library from individuals with accurate measurements of insulin sensitivity, and performed gene expression and key driver analyses. Our work demonstrates that iPSCs can be used as a revolutionary technology to model insulin resistance and to discover key genetic drivers. Moreover, they can develop our basic knowledge of the disease, and are ultimately expected to increase the therapeutic targets to treat insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Ivan Carcamo-Orive
- Stanford University School of Medicine, Division of Cardiovascular Medicine, Cardiovascular Institute, and Diabetes Research Center, Stanford, California, United States of America
- * E-mail: (ICO); (JWK); (RC)
| | - Marc Y. R. Henrion
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
- Malawi—Liverpool—Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Kuixi Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Neurology, University of Arizona, Tucson, Arizona, United States of America
- The Center for Innovations in Brain Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Noam D. Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Paige Cundiff
- Vertex Pharmaceuticals, Boston, Massachusetts, United States of America
| | - Sara Moein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Neurology, University of Arizona, Tucson, Arizona, United States of America
- The Center for Innovations in Brain Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Zenan Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Melissa Alamprese
- Department of Neurology, University of Arizona, Tucson, Arizona, United States of America
- The Center for Innovations in Brain Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Sunita L. D’Souza
- Department of Cellular, Developmental and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Martin Wabitsch
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Endocrinology, Ulm University, Ulm, Germany
| | - Eric E. Schadt
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Thomas Quertermous
- Stanford University School of Medicine, Division of Cardiovascular Medicine, Cardiovascular Institute, and Diabetes Research Center, Stanford, California, United States of America
| | - Joshua W. Knowles
- Stanford University School of Medicine, Division of Cardiovascular Medicine, Cardiovascular Institute, and Diabetes Research Center, Stanford, California, United States of America
- * E-mail: (ICO); (JWK); (RC)
| | - Rui Chang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Neurology, University of Arizona, Tucson, Arizona, United States of America
- The Center for Innovations in Brain Sciences, University of Arizona, Tucson, Arizona, United States of America
- INTelico Therapeutics LLC, Tucson, Arizona, United States of America
- * E-mail: (ICO); (JWK); (RC)
| |
Collapse
|
48
|
Liu WT, Lin C, Tsai MC, Cheng CC, Chen SJ, Liou JT, Lin WS, Cheng SM, Lin CS, Tsao TP. Effects of Pitavastatin, Atorvastatin, and Rosuvastatin on the Risk of New-Onset Diabetes Mellitus: A Single-Center Cohort Study. Biomedicines 2020; 8:biomedicines8110499. [PMID: 33202854 PMCID: PMC7696728 DOI: 10.3390/biomedicines8110499] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 01/20/2023] Open
Abstract
Statins constitute the mainstay treatment for atherosclerotic cardiovascular disease, which is associated with the risk of new-onset diabetes mellitus (NODM). However, the effects of individual statins on the risk of NODM remain unclear. We recruited 48,941 patients taking one of the three interested statins in a tertiary hospital between 2006 and 2018. Among them, 8337 non-diabetic patients taking moderate-intensity statins (2 mg/day pitavastatin, 10 mg/day atorvastatin, and 10 mg/day rosuvastatin) were included. The pitavastatin group had a higher probability of being NODM-free than the atorvastatin and rosuvastatin groups during the 4-year follow-up (log-rank test: p = 0.038). A subgroup analysis revealed that rosuvastatin had a significantly higher risk of NODM than pitavastatin among patients with coronary artery disease (CAD) (adjusted HR [aHR], 1.47, 95% confidence interval [CI], 1.05–2.05, p = 0.025), hypertension (aHR, 1.26, 95% CI, 1.00–1.59, p = 0.047), or chronic obstructive pulmonary disease (COPD) (aHR, 1.74, 95% CI, 1.02–2.94, p = 0.04). We concluded that compared with rosuvastatin, reduced diabetogenic effects of pitavastatin were observed among patients treated with moderate-intensity statin who had hypertension, COPD, or CAD. Additional studies are required to prove the effects of different statins on the risk of NODM.
Collapse
Affiliation(s)
- Wei-Ting Liu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Chin Lin
- School of Public Health, National Defense Medical Center, Taipei 11490, Taiwan;
- School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Cheng-Chung Cheng
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (C.-C.C.); (J.-T.L.); (W.-S.L.); (S.-M.C.)
| | - Sy-Jou Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
- Graduate Institute of Injury Prevention and Control, College of Public Health and Nutrition, Taipei Medical University, Taipei 11031, Taiwan
| | - Jun-Ting Liou
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (C.-C.C.); (J.-T.L.); (W.-S.L.); (S.-M.C.)
| | - Wei-Shiang Lin
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (C.-C.C.); (J.-T.L.); (W.-S.L.); (S.-M.C.)
| | - Shu-Meng Cheng
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (C.-C.C.); (J.-T.L.); (W.-S.L.); (S.-M.C.)
| | - Chin-Sheng Lin
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (C.-C.C.); (J.-T.L.); (W.-S.L.); (S.-M.C.)
- Correspondence: (C.-S.L.); (T.-P.T.); Tel.: +886-2-6601-2656 (C.-S.L.); +886-2-2826-4400 (T.-P.T.)
| | - Tien-Ping Tsao
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (C.-C.C.); (J.-T.L.); (W.-S.L.); (S.-M.C.)
- Division of Cardiology, Cheng Hsin General Hospital, Taipei 11220, Taiwan
- Correspondence: (C.-S.L.); (T.-P.T.); Tel.: +886-2-6601-2656 (C.-S.L.); +886-2-2826-4400 (T.-P.T.)
| |
Collapse
|
49
|
Takei S, Nagashima S, Takei A, Yamamuro D, Wakabayashi T, Murakami A, Isoda M, Yamazaki H, Ebihara C, Takahashi M, Ebihara K, Dezaki K, Takayanagi Y, Onaka T, Fujiwara K, Yashiro T, Ishibashi S. β-Cell-Specific Deletion of HMG-CoA (3-hydroxy-3-methylglutaryl-coenzyme A) Reductase Causes Overt Diabetes due to Reduction of β-Cell Mass and Impaired Insulin Secretion. Diabetes 2020; 69:2352-2363. [PMID: 32796082 DOI: 10.2337/db19-0996] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 08/03/2020] [Indexed: 11/13/2022]
Abstract
Inhibitors of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), statins, which are used to prevent cardiovascular diseases, are associated with a modest increase in the risk of new-onset diabetes. To investigate the role of HMGCR in the development of β-cells and glucose homeostasis, we deleted Hmgcr in a β-cell-specific manner by using the Cre-loxP technique. Mice lacking Hmgcr in β-cells (β-KO) exhibited hypoinsulinemic hyperglycemia as early as postnatal day 9 (P9) due to decreases in both β-cell mass and insulin secretion. Ki67-positive cells were reduced in β-KO mice at P9; thus, β-cell mass reduction was caused by proliferation disorder immediately after birth. The mRNA expression of neurogenin3 (Ngn3), which is transiently expressed in endocrine progenitors of the embryonic pancreas, was maintained despite a striking reduction in the expression of β-cell-associated genes, such as insulin, pancreatic and duodenal homeobox 1 (Pdx1), and MAF BZIP transcription factor A (Mafa) in the islets from β-KO mice. Histological analyses revealed dysmorphic islets with markedly reduced numbers of β-cells, some of which were also positive for glucagon. In conclusion, HMGCR plays critical roles not only in insulin secretion but also in the development of β-cells in mice.
Collapse
Affiliation(s)
- Shoko Takei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Shuichi Nagashima
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Akihito Takei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Daisuke Yamamuro
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Tetsuji Wakabayashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Akiko Murakami
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Masayo Isoda
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Hisataka Yamazaki
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Chihiro Ebihara
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Manabu Takahashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Ken Ebihara
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Katsuya Dezaki
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Yuki Takayanagi
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Ken Fujiwara
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Takashi Yashiro
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| |
Collapse
|
50
|
Statin Treatment-Induced Development of Type 2 Diabetes: From Clinical Evidence to Mechanistic Insights. Int J Mol Sci 2020; 21:ijms21134725. [PMID: 32630698 PMCID: PMC7369709 DOI: 10.3390/ijms21134725] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
Statins are the gold-standard treatment for the prevention of primary and secondary cardiovascular disease, which is the leading cause of mortality worldwide. Despite the safety and relative tolerability of statins, observational studies, clinical trials and meta-analyses indicate an increased risk of developing new-onset type 2 diabetes mellitus (T2DM) after long-term statin treatment. It has been shown that statins can impair insulin sensitivity and secretion by pancreatic β-cells and increase insulin resistance in peripheral tissues. The mechanisms involved in these processes include, among others, impaired Ca2+ signaling in pancreatic β-cells, down-regulation of GLUT-4 in adipocytes and compromised insulin signaling. In addition, it has also been described that statins’ impact on epigenetics may also contribute to statin-induced T2DM via differential expression of microRNAs. This review focuses on the evidence and mechanisms by which statin therapy is associated with the development of T2DM. This review describes the multifactorial combination of effects that most likely contributes to the diabetogenic effects of statins. Clinically, these findings should encourage clinicians to consider diabetes monitoring in patients receiving statin therapy in order to ensure early diagnosis and appropriate management.
Collapse
|