1
|
Veedu AP, Kunhipurayil D, Beegum F, George KT, Kanwal A, Shenoy RR, Nandakumar K. Biochanin‑A as SIRT‑1 modulator in preventing statin‑associated diabetogenesis: An in vitro study. Biomed Rep 2025; 22:91. [PMID: 40171401 PMCID: PMC11959223 DOI: 10.3892/br.2025.1969] [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: 07/02/2024] [Accepted: 01/22/2025] [Indexed: 04/03/2025] Open
Abstract
The widespread use of statin therapy for hypercholesterolemia has raised concerns due to its associated risk of inducing diabetes. Biochanin-A (BA), an isoflavone, exhibits potential in preventing diabetes and hyperlipidemia, yet its efficacy in mitigating statin-induced diabetes remains unexplored. This gap prompts a crucial inquiry: Can BA reduce the risk of diabetes associated with statin therapy? The present study investigated the molecular mechanisms behind atorvastatin's diabetogenic nature and evaluated the potential of BA to counteract these effects. Insulin resistance was assessed using L6 skeletal muscle cells and pancreatic beta cell apoptosis in MIN-6 cells. Our hypothesis posits that atorvastatin exacerbates free fatty acid accumulation, leading to the downregulation of sirtuin-1 (SIRT-1) and decreased uncoupling protein (UCP) 3 expression, culminating in insulin resistance. Conversely, BA is assumed to positively modulate SIRT-1 and downregulate UCP2, thus offering a protective effect. In vitro studies using L6 and MIN-6 cells revealed that BA has increased cell viability and shown optimal protection against the toxicity induced by atorvastatin in both cell lines at different concentrations. BA effectively inhibited the reduction in glucose uptake caused by atorvastatin. Pre-treatment with BA upregulated proteins that are involved in the insulin-signaling pathway and reversed the expression levels of UCPs induced by atorvastatin. BA also enhanced insulin release, preserved mitochondrial function, and prevented atorvastatin-induced apoptosis. Furthermore, BA improved SIRT-1 expression, potentially through the nicotinamide phospho-ribosyl-transferase-nicotinamide adenine dinucleotide + SIRT1-pathway, revealing that BA may play a role in modulating cellular processes in statin-associated SIRT-1 downregulation. BA can be considered a promising molecule to counteract statin-induced diabetes, suggesting a prospective therapeutic role in enhancing the safety profile of statin therapy. This research lays the groundwork for future clinical evaluations of BA as an adjunctive treatment for patients at risk of statin-induced diabetes.
Collapse
Affiliation(s)
- Anuranjana Putiya Veedu
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Divya Kunhipurayil
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Krupa Thankam George
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Abhinav Kanwal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, Punjab 151005, India
| | - Rekha Raghuveer Shenoy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Co-ordinator, Center for Animal Research, Ethics and Training (CARET), Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
2
|
Pidd K, Breeze P, Ahern A, Griffin SJ, Brennan A. Effects of weight loss and weight gain on HbA 1c, systolic blood pressure and total cholesterol in three subgroups defined by blood glucose: a pooled analysis of two behavioural weight management trials in England. BMJ Open 2025; 15:e095046. [PMID: 40233948 PMCID: PMC12004498 DOI: 10.1136/bmjopen-2024-095046] [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: 10/14/2024] [Accepted: 03/27/2025] [Indexed: 04/17/2025] Open
Abstract
OBJECTIVES To estimate the association between weight and cardiometabolic risk factors across subgroups of individuals with normoglycaemia, non-diabetic hyperglycaemia and type 2 diabetes (T2D) and to explore whether the association differs between weight loss and weight gain. DESIGN Observational analysis using mixed-effects regression models of pooled trial data. PARTICIPANTS The Weight loss Referral for Adults in Primary care (n=1267) and Glucose Lowering through Weight management (n=577) trials recruited individuals with overweight or obesity (body mass index, BMI >25 kg/m2) from primary care practices across England. PRIMARY AND SECONDARY OUTCOME MEASURES The primary outcome measures were the relationships between a change in (BMI; kg/m2) and a change in glycated haemoglobin (HbA1c; mmol/mol), total cholesterol (mmol/L) or systolic blood pressure (SBP; mm Hg) across three subgroups of individuals with: normoglycaemia, non-diabetic hyperglycaemia and T2D. Secondary outcomes included the influence of weight loss versus weight gain on these relationships. RESULTS HbA1c is positively related to a change in BMI, and a 1 kg/m2 change was related to a 1.5 mmol/mol (95% CI: 1.1 to 1.9) change in HbA1c in individuals with T2D, 0.6 mmol/mol (95% CI: 0.4 to 0.8) change in those with non-diabetic hyperglycaemia and 0.3 mmol/mol (95% CI: 0.2 to 0.4) change in those with normoglycaemia. In individuals with normoglycaemia, weight gain has a larger impact on HbA1c than weight loss, with a 0.5 mmol/mol (95% CI: 0.3 to 0.7) increase per 1 kg/m2 gained, compared with a relationship that is 0.3 mmol/mol smaller (95% CI: -0.6 to -0.1) per 1 kg/m2 of weight loss. BMI reduction improved SBP and total cholesterol significantly; however, effects did not differ between the three subgroups. CONCLUSIONS Cardiometabolic risk factors are associated with changes in weight. The association with HbA1c varies by diabetes status, with increasing magnitude in those with non-diabetic hyperglycaemia and T2D. Weight gain has a larger impact on HbA1c than weight loss in individuals with normoglycaemia, implying an asymmetric relationship.
Collapse
Affiliation(s)
- Katharine Pidd
- School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Penny Breeze
- School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Amy Ahern
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Simon J Griffin
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Alan Brennan
- School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| |
Collapse
|
3
|
Tammineni ER, Manno C, Oza G, Figueroa L. Skeletal muscle disorders as risk factors for type 2 diabetes. Mol Cell Endocrinol 2025; 599:112466. [PMID: 39848431 PMCID: PMC11886953 DOI: 10.1016/j.mce.2025.112466] [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: 11/12/2024] [Revised: 12/27/2024] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
The incidence and prevalence of muscular disorders and of type 2 diabetes (T2D) is increasing and both represent highly significant healthcare problems, both economically and compromising quality of life. Interestingly, skeletal muscle dysfunction and T2D share some commonalities including dysregulated glucose homeostasis, increased oxidative stress, dyslipidemia, and cytokine alterations. Several lines of evidence have hinted to a relationship between skeletal muscle dysfunction and T2D. For instance, T2D affects skeletal muscle morphology, functionality, and overall health through altered protein metabolism, impaired mitochondrial function, and ultimately cell viability. Conversely, humans suffering from myopathies and their experimental models demonstrated increased incidence of T2D through altered muscle glucose disposal function due to abnormal calcium homeostasis, compromised mitochondrial function, dyslipidemia, increased inflammatory cytokines and fiber size alterations and disproportions. Lifestyle modifications are essential for improving and maintaining mobility and metabolic health in individuals suffering from myopathies along with T2D. In this review, we updated current literature evidence on clinical incidence of T2D in inflammatory, mitochondrial, metabolic myopathies, and muscular dystrophies and further discussed the molecular basis of these skeletal muscle disorders leading to T2D.
Collapse
Affiliation(s)
| | - Carlo Manno
- Department of Physiology and Biophysics, Rush University, Chicago, USA
| | - Goldie Oza
- Centro de Investigación y Desarrollo Tecnológico en Electroquímica S. C., Queretaro, Mexico
| | - Lourdes Figueroa
- Department of Physiology and Biophysics, Rush University, Chicago, USA
| |
Collapse
|
4
|
Moura FA, Kamanu FK, Wiviott SD, Giugliano RP, Udler MS, Florez JC, Ellinor PT, Sabatine MS, Ruff CT, Marston NA. Type 2 diabetes genetic risk and incident diabetes across diabetes risk enhancers. Diabetes Obes Metab 2025; 27:1287-1295. [PMID: 39696834 DOI: 10.1111/dom.16123] [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: 09/02/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
AIMS To evaluate the predictive value of a contemporary type 2 diabetes (T2D) polygenic score (PGS) in detecting incident diabetes across a range of diabetes risk factors. MATERIALS AND METHODS We analysed participants in the Further Cardiovascular Outcomes Research With PCSK9 Inhibition in Subjects With Elevated Risk (FOURIER) trial (ClinicalTrials.gov, number NCT0176463), which compared the efficacy of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor evolocumab versus placebo in lowering cardiovascular outcomes in participants with stable atherosclerotic cardiovascular disease and LDL cholesterol levels of 70 mg/dL (1.8 mmol/L) or higher who were receiving statin therapy. Genetic risk was characterized using a previously validated T2D PGS based on ~1.2 million single-nucleotide polymorphisms. PGS was analysed continuously and categorically as high (top 20% of the PGS) and low to intermediate (lower 80% of the PGS). The effect of evolocumab on incident diabetes in patients without diabetes at baseline was also assessed. HbA1c was measured at baseline and every 24 weeks thereafter, while FPG was measured at baseline, week 12, week 24 and every 24 weeks thereafter. Potential cases of incident diabetes were adjudicated centrally. Hazards ratios (HRs) for incident diabetes were adjusted for baseline characteristics and ancestry. RESULTS Among 9388 participants, the mean age was 63 ± 9 years and 22.7% were women, with median HbA1c 39 mmol/mol (36 mmol/mol - 41 mmol/mol; 5.7% [5.4%-5.9%]) and mean body mass index (BMI) 28.7 ± 5 kg/m2. Diabetes developed in 690 participants (7.3%) during 2.3 years of median follow-up. T2D PGS predicted incident T2D (HR per 1-SD 1.22, 95% CI 1.14-1.32, p < 0.001). The rates of incident T2D in the high and low to intermediate genetic risk categories were 12.1% versus 6.8%, respectively (HR 1.43 95% CI 1.20-1.70, p < 0.001). Notably, high T2D genetic risk had greater predictive strength among individuals with lower HbA1c (P-int = 0.0499) and lower BMI (P-int = 0.004). CONCLUSIONS The T2D polygenic score serves as an independent predictor of incident diabetes, particularly among individuals with lower distribution of traditional risk factors.
Collapse
Affiliation(s)
- Filipe A Moura
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- VA Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Frederick K Kamanu
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen D Wiviott
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert P Giugliano
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Miriam S Udler
- Center for Genomic Medicine and Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jose C Florez
- Center for Genomic Medicine and Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marc S Sabatine
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian T Ruff
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicholas A Marston
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Gigante B, Chen Q, Björkbacka H, Björnson E, Brinck J, Chorell E, Djekic D, Edsfeldt A, Engström G, Eriksson JW, Gottsäter A, Gummesson A, Hagström E, Hedin U, Jernberg T, Johnston N, Nilsson L, Nyström F, Otten J, Rosengren A, Söderberg S, Haglöw JT, Östgren CJ. Lipoproteins and lipoprotein lipid composition are associated with stages of dysglycemia and subclinical coronary atherosclerosis. Int J Cardiol 2025; 419:132698. [PMID: 39500476 DOI: 10.1016/j.ijcard.2024.132698] [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: 06/08/2024] [Revised: 09/25/2024] [Accepted: 10/31/2024] [Indexed: 11/10/2024]
Abstract
BACKGROUND Dyslipidaemia in patients with diabetes contributes to the risk of atherosclerotic cardiovascular disease. We aimed to identify a dyslipidemic profile associated with both dysglycemia and subclinical coronary atherosclerosis. METHODS Study participants (n = 5050) were classified in three groups: normoglycemia, pre-diabetes, and diabetes. A coronary artery calcium score (CACS) > 0 defined subclinical coronary atherosclerosis. Two independent methods were used to identify, among 225 lipid biomarkers, those that were associated with pre-diabetes and diabetes and were further tested for association by zero inflated Poisson regression with CACS and with CACS burden in study participants with CACS>0. Estimates were adjusted for cardiovascular risk factors with an interaction term for dispensed lipid lowering drugs. RESULTS Thirty-two biomarkers associated with prediabetes and diabetes were further investigated for association with CACS. HDL diameter [multi-adjusted OR of 0.85 and 95 %CI (0.78-0.92)] as well as free cholesterol, phospholipids and total lipids in extra large HDL were inversely associated with CACS. There was a borderline significant interaction between small HDL and dispensed lipid lowering drugs on the presence of CACS, with and multi-adjusted OR of 0.53 and 95 %CI (0.36-0.77). None of the 32 glycemic profile-related lipid biomarkers associated with the relative increase of CACS in those with CACS>0. No consistent association was observed between non-HDL lipoproteins and CACS. CONCLUSIONS Changes in composition and relative concentration of HDL associated with both dysglycemia and subclinical coronary atherosclerosis. Treatment with lipid lowering drugs may contribute to reduce the risk associated with high circulating levels of small HDL.
Collapse
Affiliation(s)
- Bruna Gigante
- Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Cardiology, Danderyds Hospital, Stockholm, Sweden.
| | - Qiaosen Chen
- Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Harry Björkbacka
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Elias Björnson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jonas Brinck
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elin Chorell
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Demir Djekic
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andreas Edsfeldt
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetology and metabolism, Uppsala University, Uppsala, Sweden
| | - Anders Gottsäter
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden; Department of Medicine, Skåne University Hospital, Malmö, Sweden
| | - Anders Gummesson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Genetics and Genomics, Gothenburg, Sweden
| | - Emil Hagström
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden; Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Ulf Hedin
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Tomas Jernberg
- Department of Clinical Sciences, Danderyds University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Nina Johnston
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Lennart Nilsson
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Fredrik Nyström
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Julia Otten
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Annika Rosengren
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Medicine, Geriatrics and Emergency Medicine, Sahlgrenska university Hospital, Östra Hospital, Gothenburg, Sweden
| | - Stefan Söderberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Jenny Theorell Haglöw
- Department of Medical Sciences, Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Carl Johan Östgren
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden; CMIV Centre of Medical Image Science and Visualization, Linköping University, Linköping, Sweden
| |
Collapse
|
6
|
ElSayed NA, McCoy RG, Aleppo G, Balapattabi K, Beverly EA, Briggs Early K, Bruemmer D, Ebekozien O, Echouffo-Tcheugui JB, Ekhlaspour L, Gaglia JL, Garg R, Khunti K, Lal R, Lingvay I, Matfin G, Pandya N, Pekas EJ, Pilla SJ, Polsky S, Segal AR, Seley JJ, Stanton RC, Bannuru RR. 3. Prevention or Delay of Diabetes and Associated Comorbidities: Standards of Care in Diabetes-2025. Diabetes Care 2025; 48:S50-S58. [PMID: 39651971 PMCID: PMC11635039 DOI: 10.2337/dc25-s003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
The American Diabetes Association (ADA) "Standards of Care in Diabetes" includes the ADA's current clinical practice recommendations and is intended to provide the components of diabetes care, general treatment goals and guidelines, and tools to evaluate quality of care. Members of the ADA Professional Practice Committee, an interprofessional expert committee, are responsible for updating the Standards of Care annually, or more frequently as warranted. For a detailed description of ADA standards, statements, and reports, as well as the evidence-grading system for ADA's clinical practice recommendations and a full list of Professional Practice Committee members, please refer to Introduction and Methodology. Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.
Collapse
|
7
|
Arefanian H, Sindhu S, Al-Rashed F, Alzaid F, Al Madhoun A, Qaddoumi M, Bahman F, Williams MR, Albeloushi S, Almansour N, Ahmad R, Al-Mulla F. Comparative efficacy, toxicity, and insulin-suppressive effects of simvastatin and pravastatin in fatty acid-challenged mouse insulinoma MIN6 β-cell model. Front Endocrinol (Lausanne) 2024; 15:1383448. [PMID: 39544235 PMCID: PMC11560436 DOI: 10.3389/fendo.2024.1383448] [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: 02/07/2024] [Accepted: 09/30/2024] [Indexed: 11/17/2024] Open
Abstract
INTRODUCTION Familial hypercholesterolemia, the highly prevalent form of dyslipidemia, is a well-known risk factor for premature heart disease and stroke worldwide. Statins, which inhibit 3-hydroxy 3-methylglutaryl coenzyme A (HMG-CoA) reductase, are the first-choice treatment for dyslipidemias, and have been effective in reducing the risk of stroke and myocardial infarction. However, emerging evidence indicates that statins may increase the incidence of new-onset type 2 diabetes by reducing β-cell mass and function. Notably, past in vitro reports studying the effects of statins on β-cells were performed without including free fatty acids in the model. This factor should have been addressed since these agents are used to treat individuals with hyperlipidemia. METHODS Here, we used a mouse insulinoma MIN6 β-cell culture model to assess the efficacy, cytotoxicity, and insulin-suppressive effects of simvastatin and pravastatin in the presence of palmitic, linoleic, and oleic acids cocktail to mimic mixed lipids challenge in a biologically relevant setting. RESULTS AND DISCUSSION Our findings indicate that simvastatin was more effective in lowering intracellular cholesterol but was more cytotoxic as compared to pravastatin. Similarly, simvastatin exhibited a higher suppression of total insulin content and insulin secretion. Both drugs suppressed insulin secretion in phases 1 and 2, dose-dependently. No significant effect was observed on mitochondrial respiration. More importantly, elution experiments showed that insulin content diminution by simvastatin treatment was reversible, while exogenous mevalonate did not improve total insulin content. This suggests that simvastatin's influence on insulin content is independent of its specific inhibitory action on HMG-CoA reductase. In conclusion, our study identified that simvastatin was more effective in lowering intracellular cholesterol, albeit it was more toxic and suppressive of β-cells function. Notably, this suppression was found to be reversible.
Collapse
Affiliation(s)
- Hossein Arefanian
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Sardar Sindhu
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
- Animal and Imaging Core Facility, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fatema Al-Rashed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fawaz Alzaid
- Department of Bioenergetics and Neurometabolism, Dasman Diabetes Institute, Kuwait City, Kuwait
- Institut Necker Enfants Malades (INEM), French Institute of Health and Medical Research (INSERM), Immunity and Metabolism of Diabetes (IMMEDIAB), Université de Paris Cité, Paris, France
| | - Ashraf Al Madhoun
- Animal and Imaging Core Facility, Dasman Diabetes Institute, Kuwait City, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Mohammed Qaddoumi
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait
- Department of Pharmacology and Therapeutics, College of Pharmacy, Kuwait University, Kuwait City, Kuwait
| | - Fatemah Bahman
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Michayla R. Williams
- Department of Bioenergetics and Neurometabolism, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Shaima Albeloushi
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Nourah Almansour
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait
| |
Collapse
|
8
|
Galis P, Bartosova L, Farkasova V, Bartekova M, Ferenczyova K, Rajtik T. Update on clinical and experimental management of diabetic cardiomyopathy: addressing current and future therapy. Front Endocrinol (Lausanne) 2024; 15:1451100. [PMID: 39140033 PMCID: PMC11319149 DOI: 10.3389/fendo.2024.1451100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a severe secondary complication of type 2 diabetes mellitus (T2DM) that is diagnosed as a heart disease occurring in the absence of any previous cardiovascular pathology in diabetic patients. Although it is still lacking an exact definition as it combines aspects of both pathologies - T2DM and heart failure, more evidence comes forward that declares DCM as one complex disease that should be treated separately. It is the ambiguous pathological phenotype, symptoms or biomarkers that makes DCM hard to diagnose and screen for its early onset. This re-view provides an updated look on the novel advances in DCM diagnosis and treatment in the experimental and clinical settings. Management of patients with DCM proposes a challenge by itself and we aim to help navigate and advice clinicians with early screening and pharmacotherapy of DCM.
Collapse
Affiliation(s)
- Peter Galis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
| | - Linda Bartosova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
| | - Veronika Farkasova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| | - Kristina Ferenczyova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomas Rajtik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
9
|
Bai L, Kwong JC, Kaufman JS, Benmarhnia T, Chen C, van Donkelaar A, Martin RV, Kim J, Lu H, Burnett RT, Chen H. Effect modification by statin use status on the association between fine particulate matter (PM2.5) and cardiovascular mortality. Int J Epidemiol 2024; 53:dyae084. [PMID: 38961644 PMCID: PMC11222296 DOI: 10.1093/ije/dyae084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 06/19/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Numerous studies have linked fine particulate matter (PM2.5) to increased cardiovascular mortality. Less is known how the PM2.5-cardiovascular mortality association varies by use of cardiovascular medications. This study sought to quantify effect modification by statin use status on the associations between long-term exposure to PM2.5 and mortality from any cardiovascular cause, coronary heart disease (CHD), and stroke. METHODS In this nested case-control study, we followed 1.2 million community-dwelling adults aged ≥66 years who lived in Ontario, Canada from 2000 through 2018. Cases were patients who died from the three causes. Each case was individually matched to up to 30 randomly selected controls using incidence density sampling. Conditional logistic regression models were used to estimate odds ratios (ORs) for the associations between PM2.5 and mortality. We evaluated the presence of effect modification considering both multiplicative (ratio of ORs) and additive scales (the relative excess risk due to interaction, RERI). RESULTS Exposure to PM2.5 increased the risks for cardiovascular, CHD, and stroke mortality. For all three causes of death, compared with statin users, stronger PM2.5-mortality associations were observed among non-users [e.g. for cardiovascular mortality corresponding to each interquartile range increase in PM2.5, OR = 1.042 (95% CI, 1.032-1.053) vs OR = 1.009 (95% CI, 0.996-1.022) in users, ratio of ORs = 1.033 (95% CI, 1.019-1.047), RERI = 0.039 (95% CI, 0.025-0.050)]. Among users, partially adherent users exhibited a higher risk of PM2.5-associated mortality than fully adherent users. CONCLUSIONS The associations of chronic exposure to PM2.5 with cardiovascular and CHD mortality were stronger among statin non-users compared to users.
Collapse
Affiliation(s)
- Li Bai
- Primary Care & Population Health Research Program, ICES, Toronto, ON, Canada
| | - Jeffrey C Kwong
- Primary Care & Population Health Research Program, ICES, Toronto, ON, Canada
- Public Health Ontario, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| | - Jay S Kaufman
- Department of Epidemiology and Biostatistics, McGill University, Montreal, QC, Canada
| | - Tarik Benmarhnia
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA, USA
| | - Chen Chen
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA, USA
| | - Aaron van Donkelaar
- Department of Energy, Environment and Chemical Engineering, Washington University, St Louis, MO, USA
| | - Randall V Martin
- Department of Energy, Environment and Chemical Engineering, Washington University, St Louis, MO, USA
| | - JinHee Kim
- Public Health Ontario, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Hong Lu
- Primary Care & Population Health Research Program, ICES, Toronto, ON, Canada
| | - Richard T Burnett
- Population Studies Division, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | - Hong Chen
- Primary Care & Population Health Research Program, ICES, Toronto, ON, Canada
- Public Health Ontario, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Population Studies Division, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| |
Collapse
|
10
|
Kim J, Kim M, Kim M, You YH, Song Y, Lee BW. Dysregulation of autophagy activation induced by atorvastatin contributes to new-onset diabetes mellitus in western diet-fed mice. Metabolism 2024; 153:155795. [PMID: 38253121 DOI: 10.1016/j.metabol.2024.155795] [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: 10/16/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024]
Abstract
BACKGROUND AND AIMS The incidence of statin-induced new-onset diabetes (NOD) is increasing but its underlying mechanisms remain unclear. We aimed to investigate the effects of various doses of atorvastatin (ATO)-induced autophagy on the development of NOD. METHODS AND RESULTS The isolated rat islets and MIN6 cells-treated with ATO, exhibited impaired glucose-stimulated insulin secretion, reduced insulin content, and induced apoptosis. Additionally, autophagy was induced at all doses (in vitro: 5, 10, 20 μM; in vivo: 10, 15, 20 mg/kg) in ATO-treated MIN6 cells or western diet (WD)-fed mice. In contrast to normal glucose-tolerant mice administered a low-dose (10 mg/kg) ATO, those treated with high-doses (15 or 20 mg/kg) exhibited impaired glucose tolerance. Furthermore, high-dose ATO-treated mice showed decreased β-cell mass and increased apoptosis compared to that of vehicle-treated mice. We also observed that the number of vesicophagous cells in the pancreas of 20 mg/kg ATO-treated WD-fed mice was higher than in vehicle-treated WD-fed mice. Inhibiting autophagy using 3-methyladenine (3-MA) and siAtg5 improved glucose tolerance in vivo and in vitro by preventing apoptotic β-cell death and restoring insulin granules. CONCLUSION These results indicate that high doses of ATO induced hyperactivated autophagy in pancreatic cells, leading to impaired insulin storage, decreased cell viability, and reduced functional cell mass, ultimately resulting in NOD development.
Collapse
Affiliation(s)
- Juhee Kim
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea
| | - Minjune Kim
- Department of gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Minjeong Kim
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea
| | - Young-Hye You
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Youngmi Song
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea.
| | - Byung-Wan Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
11
|
Kip KE, Diamond D, Mulukutla S, Marroquin OC. Is LDL cholesterol associated with long-term mortality among primary prevention adults? A retrospective cohort study from a large healthcare system. BMJ Open 2024; 14:e077949. [PMID: 38548371 PMCID: PMC10982736 DOI: 10.1136/bmjopen-2023-077949] [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: 07/20/2023] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
OBJECTIVES Among primary prevention-type adults not on lipid-lowering therapy, conflicting results exist on the relationship between low-density lipoprotein cholesterol (LDL-C) and long-term mortality. We evaluated this relationship in a real-world evidence population of adults. DESIGN Retrospective cohort study. SETTING Electronic medical record data for adults, from 4 January 2000 through 31 December 2022, were extracted from the University of Pittsburgh Medical Center healthcare system. PARTICIPANTS Adults without diabetes aged 50-89 years not on statin therapy at baseline or within 1 year and classified as primary prevention-type patients. To mitigate potential reverse causation, patients who died within 1 year or had baseline total cholesterol (T-C) ≤120 mg/dL or LDL-C <30 mg/dL were excluded. MAIN EXPOSURE MEASURE Baseline LDL-C categories of 30-79, 80-99, 100-129, 130-159, 160-189 or ≥190 mg/dL. MAIN OUTCOME MEASURE All-cause mortality with follow-up starting 365 days after baseline cholesterol measurement. RESULTS 177 860 patients with a mean (SD) age of 61.1 (8.8) years and mean (SD) LDL-C of 119 (31) mg/dL were evaluated over a mean of 6.1 years of follow-up. A U-shaped relationship was observed between the six LDL-C categories and mortality with crude 10-year mortality rates of 19.8%, 14.7%, 11.7%, 10.7%, 10.1% and 14.0%, respectively. Adjusted mortality HRs as compared with the referent group of LDL-C 80-99 mg/dL were: 30-79 mg/dL (HR 1.23, 95% CI 1.17 to 1.30), 100-129 mg/dL (0.87, 0.83-0.91), 130-159 mg/dL (0.88, 0.84-0.93), 160-189 mg/dL (0.91, 0.84-0.98) and ≥190 mg/dL (1.19, 1.06-1.34), respectively. Unlike LDL-C, both T-C/HDL cholesterol (high-density lipoprotein cholesterol) and triglycerides/HDL cholesterol ratios were independently associated with long-term mortality. CONCLUSIONS Among primary prevention-type patients aged 50-89 years without diabetes and not on statin therapy, the lowest risk for long-term mortality appears to exist in the wide LDL-C range of 100-189 mg/dL, which is much higher than current recommendations. For counselling these patients, minimal consideration should be given to LDL-C concentration.
Collapse
Affiliation(s)
- Kevin E Kip
- Clinical Analytics, University of Pittsburgh Medical Center Health System, Pittsburgh, Pennsylvania, USA
| | - David Diamond
- Department of Psychology, University of South Florida, Tampa, Florida, USA
| | - Suresh Mulukutla
- Clinical Analytics, University of Pittsburgh Medical Center Health System, Pittsburgh, Pennsylvania, USA
| | - Oscar C Marroquin
- Physician Services Division, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
12
|
Duspara K, Sikora R, Petrovic A, Kuna Roguljic L, Matic A, Kralik K, Roguljic H, Kizivat T, Duspara M, Igrec D, Bojanic K, Smolic R, Vcev A, Wyszyńska M, Wu GY, Smolic M. Changes in Dickkopf-1, but Not Sclerostin, in Gingival Crevicular Fluid Are Associated with Peroral Statin Treatment in Patients with Periodontitis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:508. [PMID: 38541234 PMCID: PMC10972349 DOI: 10.3390/medicina60030508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/17/2025]
Abstract
Background and Objectives: Periodontitis is marked by the destruction of alveolar bone. Sclerostin (SOST) and dickkopf-1 (DKK-1) act as inhibitors of the Wingless-type (Wnt) signaling pathway, a key regulator of bone metabolism. Recent studies have suggested that statins play a role in bone resorption and formation by influencing Wnt signaling. The aim of this study was to determine the levels of SOST and DKK-1 in periodontal patients with and without peroral statins treatment in their therapy. Materials and Methods: A total of 79 patients with diagnosed periodontitis were divided into two groups: 39 patients on statin therapy (SP group) and 40 patients without statin therapy as a control group (P group). The periodontal clinical examination probing (pocket) depth (PD) and gingival recession (GR) were measured, and approximal plaque was detected, while vertical and horizontal bone resorption was measured using a panoramic radiograph image. Clinical attachment loss (CAL) values were calculated using PD and GR values. Gingival crevicular fluid (GCF) was collected and used for measuring SOST and DKK-1 levels. A questionnaire was used to assess lifestyle habits and statin intake. Patients' medical records were used to obtain biochemical parameters. Results: There was no significant difference in sclerostin concentration between the SP and P group. DKK-1 values were significantly higher in the SP group compared to the control group (p = 0.04). Also, PD (p = 0.001) and GR (p = 0.03) were significantly higher in the SP group. The level of DKK-1 had a positive relationship with the PD, the greater the PD, the higher the level of DKK-1 (Rho = 0.350), while there was no significant association with other parameters. Conclusions: Peroral statins in periodontal patients are associated with GCF levels of DKK-1 but not with sclerostin levels.
Collapse
Affiliation(s)
- Kristina Duspara
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.D.); (R.S.); (A.P.); (L.K.R.); (A.M.); (D.I.); (K.B.); (R.S.); (A.V.)
- Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.K.); (H.R.); (T.K.)
- Public Health Scientific Institution Medical Center “Dr. Mustafa Sehovic”, 75000 Tuzla, Bosnia and Herzegovina;
| | - Renata Sikora
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.D.); (R.S.); (A.P.); (L.K.R.); (A.M.); (D.I.); (K.B.); (R.S.); (A.V.)
- Health Center Osijek-Baranja County, 31000 Osijek, Croatia
| | - Ana Petrovic
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.D.); (R.S.); (A.P.); (L.K.R.); (A.M.); (D.I.); (K.B.); (R.S.); (A.V.)
| | - Lucija Kuna Roguljic
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.D.); (R.S.); (A.P.); (L.K.R.); (A.M.); (D.I.); (K.B.); (R.S.); (A.V.)
| | - Anita Matic
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.D.); (R.S.); (A.P.); (L.K.R.); (A.M.); (D.I.); (K.B.); (R.S.); (A.V.)
| | - Kristina Kralik
- Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.K.); (H.R.); (T.K.)
| | - Hrvoje Roguljic
- Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.K.); (H.R.); (T.K.)
| | - Tomislav Kizivat
- Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.K.); (H.R.); (T.K.)
- University Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Mirjana Duspara
- Public Health Scientific Institution Medical Center “Dr. Mustafa Sehovic”, 75000 Tuzla, Bosnia and Herzegovina;
| | - Dunja Igrec
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.D.); (R.S.); (A.P.); (L.K.R.); (A.M.); (D.I.); (K.B.); (R.S.); (A.V.)
| | - Kristina Bojanic
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.D.); (R.S.); (A.P.); (L.K.R.); (A.M.); (D.I.); (K.B.); (R.S.); (A.V.)
- Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.K.); (H.R.); (T.K.)
- Health Center Osijek-Baranja County, 31000 Osijek, Croatia
| | - Robert Smolic
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.D.); (R.S.); (A.P.); (L.K.R.); (A.M.); (D.I.); (K.B.); (R.S.); (A.V.)
| | - Aleksandar Vcev
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.D.); (R.S.); (A.P.); (L.K.R.); (A.M.); (D.I.); (K.B.); (R.S.); (A.V.)
| | - Magdalena Wyszyńska
- Division of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 15 Poniatowskiego Street, 40-055 Katowice, Poland;
| | - George Y. Wu
- University of Connecticut Health Center, Farmington, CT 06030, USA;
| | - Martina Smolic
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia; (K.D.); (R.S.); (A.P.); (L.K.R.); (A.M.); (D.I.); (K.B.); (R.S.); (A.V.)
| |
Collapse
|
13
|
Sawhney JPS, Gupta R. Indian dyslipidaemia guidelines: Need of the hour. Indian Heart J 2024; 76 Suppl 1:S2-S5. [PMID: 38219904 PMCID: PMC11019334 DOI: 10.1016/j.ihj.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 01/08/2024] [Indexed: 01/16/2024] Open
Affiliation(s)
| | - Rajeev Gupta
- Department of Preventive Cardiology & Medicine, Eternal Heart Care Centre & Research Institute, Jaipur, India.
| |
Collapse
|
14
|
Guo L, Xiao X. Guideline for the Management of Diabetes Mellitus in the Elderly in China (2024 Edition). Aging Med (Milton) 2024; 7:5-51. [PMID: 38571669 PMCID: PMC10985780 DOI: 10.1002/agm2.12294] [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: 01/30/2024] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 04/05/2024] Open
Abstract
With the deepening of aging in China, the prevalence of diabetes in older people has increased noticeably, and standardized diabetes management is critical for improving clinical outcomes of diabetes in older people. In 2021, the National Center of Gerontology, Chinese Society of Geriatrics, and Diabetes Professional Committee of Chinese Aging Well Association organized experts to write the first guideline for diabetes diagnosis and treatment in older people in China, the Guideline for the Management of Diabetes Mellitus in the Elderly in China (2021 Edition). The guideline emphasizes that older patients with diabetes are a highly heterogeneous group requiring comprehensive assessment and stratified and individualized management strategies. The guideline proposes simple treatments and de-intensified treatment strategies for older patients with diabetes. This edition of the guideline provides clinicians with practical and operable clinical guidance, thus greatly contributing to the comprehensive and full-cycle standardized management of older patients with diabetes in China and promoting the extensive development of clinical and basic research on diabetes in older people and related fields. In the past 3 years, evidence-based medicine for older patients with diabetes and related fields has further advanced, and new treatment concepts, drugs, and technologies have been developed. The guideline editorial committee promptly updated the first edition of the guideline and compiled the Guideline for the Management of Diabetes Mellitus in the Elderly in China (2024 Edition). More precise management paths for older patients with diabetes are proposed, for achieving continued standardization of the management of older Chinese patients with diabetes and improving their clinical outcomes.
Collapse
Affiliation(s)
- Lixin Guo
- National Center of Gerontology, Chinese Society of Geriatrics, Diabetes Professional Committee of Chinese Aging Well AssociationBeijingChina
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Xinhua Xiao
- National Center of Gerontology, Chinese Society of Geriatrics, Diabetes Professional Committee of Chinese Aging Well AssociationBeijingChina
- Department of EndocrinologyPeking Union Medical College Hospital, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
15
|
Kwon J, Kim MS, Blagojevic C, Mailloux J, Medwid S, Tirona RG, Wang R, Schwarz UI. Differential effects of OATP2B1 on statin accumulation and toxicity in a beta cell model. Toxicol Mech Methods 2024; 34:130-147. [PMID: 37771097 DOI: 10.1080/15376516.2023.2262568] [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: 08/18/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023]
Abstract
An increased risk of new-onset diabetes mellitus has been recently reported for statin therapy, and experimental studies have shown reduced glucose-stimulated insulin secretion (GSIS) and mitochondrial dysfunction in beta cells with effects differing among agents. Organic anion transporting polypeptide (OATP) 2B1 contributes to hepatic uptake of rosuvastatin, atorvastatin and pravastatin, three known substrates. Since OATP2B1 is present in beta cells of the human pancreas, we investigated if OATP2B1 facilitates the local accumulation of statins in a rat beta cell model INS-1 832/13 (INS-1) thereby amplifying statin-induced toxicity. OATP2B1 overexpression in INS-1 cells via adenoviral transduction showed 2.5-, 1.8- and 1.4-fold higher cellular retention of rosuvastatin, atorvastatin and pravastatin, respectively, relative to LacZ control, while absolute intracellular concentration was about twice as high for the lipophilic atorvastatin compared to the more hydrophilic rosuvastatin and pravastatin. After 24 h statin treatment at high concentrations, OATP2B1 enhanced statin toxicity involving activation of intrinsic apoptosis (caspase 3/7 activation) and mitochondrial dysfunction (NADH dehydrogenase activity) following rosuvastatin and atorvastatin, which was partly reversed by isoprenoids. OATP2B1 had no effect on statin-induced reduction in GSIS, mitochondrial electron transport chain complex expression or caspase 9 activation. We confirmed a dose-dependent reduction in insulin secretion by rosuvastatin and atorvastatin in native INS-1 with a modest change in cellular ATP. Collectively, our results indicate a role of OATP2B1, which is abundant in human beta cells, in statin accumulation and statin-induced toxicity but not insulin secretion of rosuvastatin and atorvastatin in INS-1 cells.
Collapse
Affiliation(s)
- Jihoon Kwon
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Michelle S Kim
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Christina Blagojevic
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jaymie Mailloux
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Samantha Medwid
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Rommel G Tirona
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Rennian Wang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Ute I Schwarz
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
16
|
ElSayed NA, Aleppo G, Bannuru RR, Bruemmer D, Collins BS, Ekhlaspour L, Gaglia JL, Hilliard ME, Johnson EL, Khunti K, Lingvay I, Matfin G, McCoy RG, Perry ML, Pilla SJ, Polsky S, Prahalad P, Pratley RE, Segal AR, Seley JJ, Selvin E, Stanton RC, Gabbay RA. 3. Prevention or Delay of Diabetes and Associated Comorbidities: Standards of Care in Diabetes-2024. Diabetes Care 2024; 47:S43-S51. [PMID: 38078581 PMCID: PMC10725807 DOI: 10.2337/dc24-s003] [Citation(s) in RCA: 59] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
The American Diabetes Association (ADA) "Standards of Care in Diabetes" includes the ADA's current clinical practice recommendations and is intended to provide the components of diabetes care, general treatment goals and guidelines, and tools to evaluate quality of care. Members of the ADA Professional Practice Committee, an interprofessional expert committee, are responsible for updating the Standards of Care annually, or more frequently as warranted. For a detailed description of ADA standards, statements, and reports, as well as the evidence-grading system for ADA's clinical practice recommendations and a full list of Professional Practice Committee members, please refer to Introduction and Methodology. Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.
Collapse
|
17
|
Agarwal H, Tinsley B, Sarecha AK, Ozcan L. Rap1 in the Context of PCSK9, Atherosclerosis, and Diabetes. Curr Atheroscler Rep 2023; 25:931-937. [PMID: 37979063 DOI: 10.1007/s11883-023-01162-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE OF REVIEW The focus of this article is to highlight the importance of the small GTPase, Ras-associated protein 1 (Rap1), in proprotein convertase subtilisin/kexin type 9 (PCSK9) regulation and atherosclerosis and type 2 diabetes etiology and discuss the potential therapeutic implications of targeting Rap1 in these disease areas. REVIEW FINDINGS Cardiometabolic disease characterized by obesity, glucose intolerance, dyslipidemia, and atherosclerotic cardiovascular disease remain an important cause of mortality. Evidence using mouse models of obesity and insulin resistance indicates that Rap1 deficiency increases proatherogenic PCSK9 and low-density lipoprotein cholesterol levels and predisposes these mice to develop obesity- and statin-induced hyperglycemia, which highlights Rap1's role in cardiometabolic dysfunction. Rap1 may also contribute to cardiovascular disease through its effects on vascular wall cells involved in the atherosclerosis progression. Rap1 activation, specifically in the liver, could be beneficial in the prevention of cardiometabolic perturbations, including type 2 diabetes, hypercholesterolemia, and atherosclerosis.
Collapse
Affiliation(s)
- Heena Agarwal
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Brea Tinsley
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Amesh K Sarecha
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
18
|
De Filippo O, D'Ascenzo F, Iannaccone M, Bertaina M, Leone A, Borzillo I, Ravetti E, Solano A, Pagliassotto I, Nebiolo M, Bruno F, Giacobbe F, Muscoli S, Monticone S, Brizzi MF, Biondi Zoccai G, De Ferrari GM. Safety and efficacy of bempedoic acid: a systematic review and meta-analysis of randomised controlled trials. Cardiovasc Diabetol 2023; 22:324. [PMID: 38017541 PMCID: PMC10685600 DOI: 10.1186/s12933-023-02022-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/10/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND AND AIMS Bempedoic Acid (BA) is a novel Lipid-Lowering Therapy (LLT). We performed a systematic review and meta-analysis to assess the efficacy and safety of BA in patients with hypercholesterolemia. METHODS PubMed, Scopus, and Cochrane library databases were searched for randomised controlled trials evaluating the efficacy and/or safety of BA compared with placebo. Trials investigating dosages other than 180 mg/die were excluded. Major adverse cardiovascular events (MACE) were the primary efficacy endpoint. LDL-cholesterol reduction was the primary laboratory endpoint. Pre-specified safety endpoints included muscle-related adverse events, new-onset diabetes, and gout. The protocol was registered on PROSPERO (temporary ID:399,867). RESULTS Study search identified 275 deduplicated results. 11 studies, encompassing 18,315 patients (9854 on BA vs 8461 on placebo/no treatment) were included. BA was associated with a reduced risk of MACE (OR 0.86, 95% CI 0.79-0.95), myocardial infarction (OR 0.76, 95% CI 0.64-0.88) and unstable angina (OR 0.69, 95% CI 0.54-0.88) compared to control, over a median follow up of 87 (15-162) weeks. BA was associated with a reduction of LDL-Cholesterol (mean difference [MD]-22.42,95% CI - 24.02% to - 20.82%), total cholesterol (- 16.50%,95% - 19.21% to - 13.79%), Apo-B lipoprotein (- 19.55%, - 22.68% to - 16.42%) and high-sensitivity CRP (- 27.83%, - 31.71% to - 23.96%) at 12 weeks. BA was associated with a higher risk of gout (OR 1.55, 95% CI 1.27-1.90) as compared with placebo. Efficacy on laboratory endpoints was confirmed, with a variable extent, across patients on statin or ezetimibe background therapy. CONCLUSIONS The improved cholesterol control achieved with BA translates into a reduced risk of MACE, including myocardial infarction and coronary revascularisation. The drug has a satisfactory safety profile except for an increased risk of gout.
Collapse
Affiliation(s)
- Ovidio De Filippo
- Division of Cardiology, Cardiovascular and Thoracic Department, "Città Della Salute e Della Scienza" Hospital, Turin, Italy
| | - Fabrizio D'Ascenzo
- Division of Cardiology, Cardiovascular and Thoracic Department, "Città Della Salute e Della Scienza" Hospital, Turin, Italy.
- Department of Medical Sciences, University of Turin, Corso Bramante 88, 10126, Turin, Italy.
| | - Mario Iannaccone
- Division of Cardiology, San Giovanni Bosco Hospital, ASL Città Di Torino, Turin, Italy
| | - Maurizio Bertaina
- Division of Cardiology, San Giovanni Bosco Hospital, ASL Città Di Torino, Turin, Italy
| | - Attilio Leone
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Irene Borzillo
- Division of Cardiology, Cardiovascular and Thoracic Department, "Città Della Salute e Della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Corso Bramante 88, 10126, Turin, Italy
| | - Emanuele Ravetti
- Division of Cardiology, Cardiovascular and Thoracic Department, "Città Della Salute e Della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Corso Bramante 88, 10126, Turin, Italy
| | - Andrea Solano
- Division of Cardiology, Cardiovascular and Thoracic Department, "Città Della Salute e Della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Corso Bramante 88, 10126, Turin, Italy
| | - Ilaria Pagliassotto
- Division of Cardiology, Cardiovascular and Thoracic Department, "Città Della Salute e Della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Corso Bramante 88, 10126, Turin, Italy
| | - Marco Nebiolo
- Division of Cardiology, Cardiovascular and Thoracic Department, "Città Della Salute e Della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Corso Bramante 88, 10126, Turin, Italy
| | - Francesco Bruno
- Division of Cardiology, Cardiovascular and Thoracic Department, "Città Della Salute e Della Scienza" Hospital, Turin, Italy
| | - Federico Giacobbe
- Division of Cardiology, Cardiovascular and Thoracic Department, "Città Della Salute e Della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Corso Bramante 88, 10126, Turin, Italy
| | - Saverio Muscoli
- Division of Cardiology, Fondazione Policlinico "Tor Vergata", Rome, Italy
| | - Silvia Monticone
- Department of Medical Sciences, University of Turin, Corso Bramante 88, 10126, Turin, Italy
| | - Maria Felice Brizzi
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giuseppe Biondi Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Gaetano Maria De Ferrari
- Division of Cardiology, Cardiovascular and Thoracic Department, "Città Della Salute e Della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Corso Bramante 88, 10126, Turin, Italy
| |
Collapse
|
19
|
Marx N, Federici M, Schütt K, Müller-Wieland D, Ajjan RA, Antunes MJ, Christodorescu RM, Crawford C, Di Angelantonio E, Eliasson B, Espinola-Klein C, Fauchier L, Halle M, Herrington WG, Kautzky-Willer A, Lambrinou E, Lesiak M, Lettino M, McGuire DK, Mullens W, Rocca B, Sattar N. 2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes. Eur Heart J 2023; 44:4043-4140. [PMID: 37622663 DOI: 10.1093/eurheartj/ehad192] [Citation(s) in RCA: 532] [Impact Index Per Article: 266.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/26/2023] Open
|
20
|
Lee SJ, Kang WC, Lee JY, Lee JB, Yang TH, Yoon J, Lee YJ, Hong SJ, Ahn CM, Kim JS, Kim BK, Ko YG, Choi D, Hong BK, Jang Y, Hong MK. Treat-to-target versus high-intensity statin treatment in patients with or without diabetes mellitus: a pre-specified analysis from the LODESTAR trial. EClinicalMedicine 2023; 64:102227. [PMID: 37767195 PMCID: PMC10520306 DOI: 10.1016/j.eclinm.2023.102227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Background The impact of titrated versus fixed intensity statin therapy in patients with coronary artery disease (CAD) and diabetes mellitus (DM) remains to be elucidated. Methods This was a pre-specified analysis of patients with and without DM from the LODESTAR trial. Patients with CAD were randomly assigned to receive either a treat-to-target strategy with a target LDL-C level of 50-70 mg/dL or a high-intensity statin treatment. Primary outcome was the 3-year composite of all-cause death, myocardial infarction, stroke, or coronary revascularization. Secondary outcomes were safety endpoints. This trial is registered with ClinicalTrials.gov, NCT02579499. Findings Between September 9, 2016 and November 27, 2019, 4400 patients with CAD were enrolled in the LODESTAR trial. The median age was 65 years (interquartile range, 59-73 years), 3172 (72%) were male, and 1468 (33%) had DM at baseline. There was no significant difference in the occurrence of the primary outcome between the treat-to-target group and high-intensity statin group among patients with DM (10.5% versus 11.1%, hazard ratio [HR] 0.94, 95% confidence interval [CI] 0.69-1.29, p = 0.70) and those without DM (6.9% versus 7.5%, HR 0.93, 95% CI 0.71-1.21, p = 0.58). Among patients without DM, there was a trend towards a lower risk of new-onset DM in the treat-to-target group (8.4% versus 10.4% in the high-intensity statin group, HR 0.79, 95% CI 0.62-1.01; p = 0.06). Interpretation In patients with CAD, a treat-to-target LDL-C strategy of 50-70 mg/dL as the goal was comparable to high-intensity statin therapy in terms of 3-year clinical efficacy and safety outcomes regardless of the presence of DM. Funding Sam Jin Pharmaceutical, Seoul, Korea and Chong Kun Dang Pharmaceutical, Seoul, Korea.
Collapse
Affiliation(s)
- Seung-Jun Lee
- Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | | | - Jong-Young Lee
- Kangbuk Samsung Hospital, Sungkyunkwan University Scholl of Medicine, Seoul, Korea
| | - Jin-Bae Lee
- Daegu Catholic University Medical Center, Daegu, Korea
| | | | - Junghan Yoon
- Wonju Severance Christian Hospital, Wonju, Korea
| | - Yong-Joon Lee
- Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sung-Jin Hong
- Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Chul-Min Ahn
- Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Sun Kim
- Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Byeong-Keuk Kim
- Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young-Guk Ko
- Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Donghoon Choi
- Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | | | - Yangsoo Jang
- CHA University College of Medicine, Seongnam, Korea
| | - Myeong-Ki Hong
- Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Laakso M, Fernandes Silva L. Statins and risk of type 2 diabetes: mechanism and clinical implications. Front Endocrinol (Lausanne) 2023; 14:1239335. [PMID: 37795366 PMCID: PMC10546337 DOI: 10.3389/fendo.2023.1239335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/29/2023] [Indexed: 10/06/2023] Open
Abstract
Statins are widely used to prevent cardiovascular disease events. Cardiovascular diseases and type 2 diabetes are tightly connected since type 2 diabetes is a major risk factor for cardiovascular diseases. Additionally, cardiovascular diseases often precede the development of type 2 diabetes. These two diseases have common genetic and environmental antecedents. Statins are effective in the lowering of cardiovascular disease events. However, they have also important side effects, including an increased risk of type 2 diabetes. The first study reporting an association of statin treatment with the risk of type 2 diabetes was the WOSCOPS trial (West of Scotland Coronary Prevention Study) in 2001. Other primary and secondary cardiovascular disease prevention studies as well as population-based studies have confirmed original findings. The purpose of our review is to examine and summarize the most important findings of these studies as well as to describe the mechanisms how statins increase the risk of type 2 diabetes.
Collapse
Affiliation(s)
- Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Kuopio University Hospital, Kuopio, Finland
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
22
|
Lörchner H, Cañes Esteve L, Góes ME, Harzenetter R, Brachmann N, Gajawada P, Günther S, Doll N, Pöling J, Braun T. Neutrophils for Revascularization Require Activation of CCR6 and CCL20 by TNFα. Circ Res 2023; 133:592-610. [PMID: 37641931 DOI: 10.1161/circresaha.123.323071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Activation of immune-inflammatory pathways involving TNFα (tumor necrosis factor alpha) signaling is critical for revascularization and peripheral muscle tissue repair after ischemic injury. However, mechanisms of TNFα-driven inflammatory cascades directing recruitment of proangiogenic immune cells to sites of ischemia are unknown. METHODS Muscle tissue revascularization after permanent femoral artery ligation was monitored in mutant mice by laser Doppler imaging and light sheet fluorescence microscopy. TNFα-mediated signaling and the role of the CCL20 (C-C motif chemokine ligand 20)-CCR6 (C-C chemokine receptor 6) axis for formation of new vessels was studied in vitro and in vivo using bone marrow transplantation, flow cytometry, as well as biochemical and molecular biological techniques. RESULTS TNFα-mediated activation of TNFR (tumor necrosis factor receptor) 1 but not TNFR2 was found to be required for postischemic muscle tissue revascularization. Bone marrow-derived CCR6+ neutrophil granulocytes were identified as a previously undescribed TNFα-induced population of proangiogenic neutrophils, characterized by increased expression of VEGFA (vascular endothelial growth factor A). Mechanistically, postischemic activation of TNFR1 induced expression of the CCL20 in vascular cells and promoted translocation of the CCL20 receptor CCR6 to the cell surface of neutrophils, essentially conditioning VEGFA-expressing proangiogenic neutrophils for CCL20-dependent recruitment to sites of ischemia. Moreover, impaired revascularization of ischemic peripheral muscle tissue in diabetic mice was associated with reduced numbers of proangiogenic neutrophils and diminished CCL20 expression. Administration of recombinant CCL20 enhanced recruitment of proangiogenic neutrophils and improved revascularization of diabetic ischemic skeletal muscles, which was sustained by sequential treatment with fluvastatin. CONCLUSIONS We demonstrate that site-specific activation of the CCL20-CCR6 axis via TNFα recruits proangiogenic VEGFA-expressing neutrophils to sites of ischemic injury for initiation of muscle tissue revascularization. The findings provide an attractive option for tissue revascularization, particularly under diabetic conditions.
Collapse
Affiliation(s)
- Holger Lörchner
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
- German Centre for Cardiovascular Research (DZHK), Frankfurt am Main, Germany (H.L., J.P.)
| | - Laia Cañes Esteve
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| | - Maria Elisa Góes
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| | - Roxanne Harzenetter
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| | - Nathalie Brachmann
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| | - Praveen Gajawada
- Department of Cardiac Surgery, Kerckhoff Heart Center, Bad Nauheim, Germany (P.G.)
| | - Stefan Günther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| | - Nicolas Doll
- Schüchtermann-Klinik, Bad Rothenfelde, Germany (N.D., J.P.)
| | - Jochen Pöling
- Schüchtermann-Klinik, Bad Rothenfelde, Germany (N.D., J.P.)
- German Centre for Cardiovascular Research (DZHK), Frankfurt am Main, Germany (H.L., J.P.)
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| |
Collapse
|
23
|
Chen WH, Guo BC, Chen CH, Hsu MC, Wang CH, Lee TS. Autophagy-urea cycle pathway is essential for the statin-mediated nitric oxide bioavailability in endothelial cells. J Food Drug Anal 2023; 31:519-533. [PMID: 39666279 PMCID: PMC10629920 DOI: 10.38212/2224-6614.3472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 07/11/2023] [Indexed: 04/17/2024] Open
Abstract
Statins induce nitric oxide (NO) bioavailability by activating endothelial nitric oxide synthase via kinase- and calcium-dependent pathways in endothelial cells (ECs). However, their effect on the metabolism of L-arginine, the precursor for NO biosynthesis, and regulatory mechanism have not yet been investigated. In this study, we investigated the role of the autophagy-urea cycle-L-arginine pathway in simvastatin-mediated NO bioavailability in ECs. Griess's assay was used to determine the NO bioavailability. Protein expression was assessed using Western blot analysis. Further, immunocytochemistry was performed to observe autophagosome formation, while conventional assay kits were used to quantify the levels of different intermediate substrates of the urea cycle. In ECs, treatment with simvastatin induced the activation of autophagy flux, as evidenced by the increased levels of microtubule-associated protein 1A/1B-light chain 3 II and autophagolysosome formation and decreased levels of p62. Inhibition of autophagy by ATG7 small interfering RNA (siRNA), chloroquine and bafilomycin A1 abolished simvastatin-induced NO bioavailability, EC proliferation, migration, and tube formation. Additionally, simvastatin increased the intermediate substrates levels of the urea cycle, including glutamate, acetyl-CoA, urea, and L-arginine, all of which were abrogated by chloroquine or bafilomycin A1. Genetic knockdown of argininosuccinate lyase using siRNA abrogated simvastatin-induced increase in NO bioavailability and EC-related functions. Moreover, inhibition of AMP-activated protein kinase (AMPK) and transient receptor potential vanilloid 1 (TRPV1) prevented simvastatin-induced activation of the autophagy-urea cycle pathway and NO production. Our findings suggest that simvastatin activates the autophagy-urea cycle pathway via TRPV1-AMPK signaling, which increases L-arginine bioavailability and ultimately promotes NO production in ECs.
Collapse
Affiliation(s)
- Wen-Hua Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei,
Taiwan
| | - Bei-Chia Guo
- 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
| | - 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
|
24
|
Kononova Y, Abramyan L, Derevitskii I, Babenko A. Predictors of Carbohydrate Metabolism Disorders and Lethal Outcome in Patients after Myocardial Infarction: A Place of Glucose Level. J Pers Med 2023; 13:997. [PMID: 37373986 PMCID: PMC10305089 DOI: 10.3390/jpm13060997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/29/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND AND AIM The aim of this study was to reveal statistical patterns in patients with acute myocardial infarction (AMI) that cause the development of carbohydrate metabolism disorders (CMD) (type 2 diabetes mellitus and prediabetes) and death within 5 years after AMI. METHODS 1079 patients who were treated with AMI in the Almazov National Medical Research Center were retrospectively selected for the study. For each patient, all data from electronic medical records were downloaded. Statistical patterns that determine the development of CMDs and death within 5 years after AMI were identified. To create and train the models used in this study, the classic methods of Data Mining, Data Exploratory Analysis, and Machine Learning were used. RESULTS The main predictors of mortality within 5 years after AMI were advanced age, low relative level of lymphocytes, circumflex artery lesion, and glucose level. Main predictors of CMDs were low basophils, high neutrophils, high platelet distribution width, and high blood glucose level. High values of age and glucose together were relatively independent predictors. With glucose level >11 mmol/L and age >70 years, the 5-year risk of death is about 40% and it rises with increasing glucose levels. CONCLUSION The obtained results make it possible to predict the development of CMDs and death based on simple parameters that are easily available in clinical practice. Glucose level measured on the 1st day of AMI was among the most important predictors of CMDs and death.
Collapse
Affiliation(s)
- Yulia Kononova
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia
| | | | | | | |
Collapse
|
25
|
Rebeiro PF, Emond B, Rossi C, Bookhart BK, Shah A, Caron‐Lapointe G, Lafeuille M, Donga P. Incidence of cardiometabolic outcomes among people living with HIV-1 initiated on integrase strand transfer inhibitor versus non-integrase strand transfer inhibitor antiretroviral therapies: a retrospective analysis of insurance claims in the United States. J Int AIDS Soc 2023; 26:e26123. [PMID: 37306118 PMCID: PMC10258864 DOI: 10.1002/jia2.26123] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 05/17/2023] [Indexed: 06/13/2023] Open
Abstract
INTRODUCTION Integrase strand transfer inhibitor (INSTI)-containing antiretroviral therapy (ART) has been associated with weight gain, though there is limited information on associations between ART-related weight gain and cardiometabolic outcomes among people living with HIV-1 (PLWH). We, therefore, evaluated risks of incident cardiometabolic outcomes following INSTI versus non-INSTI-based ART initiation in the United States. METHODS We conducted a retrospective study using IBM MarketScan Research Databases (12 August 2012-31 January 2021). Treatment-naïve PLWH initiating ART (index date) on/after 12 August 2013 (approval date of the first second-generation INSTI, dolutegravir) were included and censored at regimen switch/discontinuation, end of insurance eligibility or end of data availability. We used inverse probability of treatment weights constructed with baseline (12 months pre-index) characteristics to account for differences between INSTI- and non-INSTI-initiating cohorts. Doubly robust hazard ratios (HRs) obtained from weighted multivariable Cox regression were used to compare time to incident cardiometabolic outcomes (congestive heart failure [CHF], coronary artery disease, myocardial infarction, stroke/transient ischemic attack, hypertension, type II diabetes, lipid disorders, lipodystrophy and metabolic syndrome) by INSTI-initiation status. RESULTS Weighted INSTI (mean age = 39 years, 23% female, 70% commercially insured, 30% Medicaid insured) and non-INSTI (mean age = 39 years, 24% female, 71% commercially insured, 29% Medicaid insured) cohorts included 7059 and 7017 PLWH, respectively. The most common INSTI-containing regimens were elvitegravir-based (43.4%), dolutegravir-based (33.3%) and bictegravir-based (18.4%); the most common non-INSTI-containing regimens were darunavir-based (31.5%), rilpivirine-based (30.4%) and efavirenz-based (28.3%). Mean±standard deviation follow-up periods were 1.5±1.5 and 1.1±1.2 years in INSTI- and non-INSTI-initiating cohorts, respectively. INSTI initiators were at a clinically and significantly increased risk of experiencing incident CHF (HR = 2.12, 95% confidence interval [CI] = 1.08-4.05; p = 0.036), myocardial infarction (HR = 1.79, 95% CI = 1.03-5.65; p = 0.036) and lipid disorders (HR = 1.26, 95% CI = 1.04-1.58; p = 0.020); there was no evidence of an increased risk for other individual or composite outcomes. CONCLUSIONS Over a short average follow-up period of <2 years, INSTI use among treatment-naïve PLWH was associated with an increased risk of several cardiometabolic outcomes, such as CHF, myocardial infarction and lipid disorders, compared to non-INSTI use. Further research accounting for additional potential confounders and with longer follow-up is warranted to more accurately and precisely quantify the impact of INSTI-containing ART on long-term cardiometabolic outcomes.
Collapse
Affiliation(s)
- Peter F. Rebeiro
- Divisions of Infectious Diseases & EpidemiologyDepartment of MedicineDepartment of BiostatisticsVanderbilt UniversityNashvilleTennesseeUSA
| | | | | | | | - Aditi Shah
- Analysis Group, Inc.MontréalQuébecCanada
| | | | | | - Prina Donga
- Janssen Scientific AffairsLLCTitusvilleNew JerseyUSA
| |
Collapse
|
26
|
Smith ML, Bull CJ, Holmes MV, Davey Smith G, Sanderson E, Anderson EL, Bell JA. Distinct metabolic features of genetic liability to type 2 diabetes and coronary artery disease: a reverse Mendelian randomization study. EBioMedicine 2023; 90:104503. [PMID: 36870196 PMCID: PMC10009453 DOI: 10.1016/j.ebiom.2023.104503] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) and coronary artery disease (CAD) both have known genetic determinants, but the mechanisms through which their associated genetic variants lead to disease onset remain poorly understood. METHODS We used large-scale metabolomics data in a two-sample reverse Mendelian randomization (MR) framework to estimate effects of genetic liability to T2D and CAD on 249 circulating metabolites in the UK Biobank (N = 118,466). We examined the potential for medication use to distort effect estimates by conducting age-stratified metabolite analyses. FINDINGS Using inverse variance weighted (IVW) models, higher genetic liability to T2D was estimated to decrease high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C) (e.g. , HDL-C -0.05 SD; 95% CI -0.07 to -0.03, per doubling of liability), whilst increasing all triglyceride groups and branched chain amino acids (BCAAs). IVW estimates for CAD liability suggested an effect on reducing HDL-C as well as raising very-low density lipoprotein cholesterol (VLDL-C) and LDL-C. In pleiotropy-robust models, T2D liability was still estimated to increase BCAAs, but several estimates for higher CAD liability reversed and supported decreased LDL-C and apolipoprotein-B. Estimated effects of CAD liability differed substantially by age for non-HDL-C traits, with higher CAD liability lowering LDL-C only at older ages when statin use was common. INTERPRETATION Overall, our results support largely distinct metabolic features of genetic liability to T2D and CAD, illustrating both challenges and opportunities for preventing these commonly co-occurring diseases. FUNDING Wellcome Trust [218495/Z/19/Z], UK MRC [MC_UU_00011/1; MC_UU_00011/4], the University of Bristol, Diabetes UK [17/0005587], World Cancer Research Fund [IIG_2019_2009].
Collapse
Affiliation(s)
- Madeleine L Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Caroline J Bull
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK; School of Translational Health Sciences, University of Bristol, Bristol, UK
| | - Michael V Holmes
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Eleanor Sanderson
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Emma L Anderson
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Joshua A Bell
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
27
|
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
|
28
|
ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes Association. 3. Prevention or Delay of Type 2 Diabetes and Associated Comorbidities: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S41-S48. [PMID: 36507633 PMCID: PMC9810464 DOI: 10.2337/dc23-s003] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The American Diabetes Association (ADA) "Standards of Care in Diabetes" includes the ADA's current clinical practice recommendations and is intended to provide the components of diabetes care, general treatment goals and guidelines, and tools to evaluate quality of care. Members of the ADA Professional Practice Committee, a multidisciplinary expert committee, are responsible for updating the Standards of Care annually, or more frequently as warranted. For a detailed description of ADA standards, statements, and reports, as well as the evidence-grading system for ADA's clinical practice recommendations and a full list of Professional Practice Committee members, please refer to Introduction and Methodology. Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.
Collapse
|
29
|
Omolaoye TS, Halabi MO, Mubarak M, Cyril AC, Duvuru R, Radhakrishnan R, Du Plessis SS. Statins and Male Fertility: Is There a Cause for Concern? TOXICS 2022; 10:627. [PMID: 36287907 PMCID: PMC9607778 DOI: 10.3390/toxics10100627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 05/06/2023]
Abstract
The well-known 3-hydroxyl 3-methyl glutaryl-Coenzyme A reductase inhibitors, called statins, have been the main medication used in the treatment of hypercholesterolemia and some cases of cardiovascular diseases. The effectiveness of this drug in controlling cholesterol production is impeccable, however, patients often complain of a variety of side effects, such as myalgia, muscle atrophy, and in some cases, rhabdomyolysis. Not only has the use of statins caused the aforementioned side effects, but they are also shown to cause testicular discomfort, erectile dysfunction, altered semen parameters, and modified steroid hormone production. These reported adverse effects on male fertility are not generally agreed upon, as some have shown the use to be beneficial. Hence, this makes the aftermath effect of statin use on male fertility debatable and controversial. The negative effects have been associated with imbalanced or reduced steroid hormones, which are necessary for proper spermatogenesis and other sexual functions. Meanwhile, the beneficial effects are related to statin's anti-inflammatory and cardioprotective properties. These contradictory findings are in part due to the different age of users, concentrations of statins, the type and duration of treatment, and the underlying disease and/or comorbidities. Therefore, the current study aims to analyze the literature and gather evidence as to the effects of statin on male sexual health and reproductive parameters, and subsequently give recommendations for the direction of future studies.
Collapse
Affiliation(s)
- Temidayo S. Omolaoye
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Mouhammad Omar Halabi
- School of Medicine, Royal College of Surgeons Ireland-Bahrain, Busaiteen 15503, Bahrain
| | - Maitha Mubarak
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Asha Caroline Cyril
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Ruthwik Duvuru
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Rajan Radhakrishnan
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Stefan S. Du Plessis
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town P.O. Box 7505, South Africa
| |
Collapse
|
30
|
Wang Y, Spolitu S, Zadroga JA, Sarecha AK, Ozcan L. Hepatocyte Rap1a contributes to obesity- and statin-associated hyperglycemia. Cell Rep 2022; 40:111259. [PMID: 36001955 PMCID: PMC9446800 DOI: 10.1016/j.celrep.2022.111259] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/26/2022] [Accepted: 08/03/2022] [Indexed: 12/28/2022] Open
Abstract
Excessive hepatic glucose production contributes to the development of hyperglycemia and is a key feature of type 2 diabetes. Here, we report that activation of hepatocyte Rap1a suppresses gluconeogenic gene expression and glucose production, whereas Rap1a silencing stimulates them. Rap1a activation is suppressed in obese mouse liver, and restoring its activity improves glucose intolerance. As Rap1a′s membrane localization and activation depends on its geranylgeranylation, which is inhibited by statins, we show that statin-treated hepatocytes and the human liver have lower active-Rap1a levels. Similar to Rap1a inhibition, statins stimulate hepatic gluconeogenesis and increase fasting blood glucose in obese mice. Geranylgeraniol treatment, which acts as the precursor for geranylgeranyl isoprenoids, restores Rap1a activity and improves statin-mediated glucose intolerance. Mechanistically, Rap1a activation induces actin polymerization, which suppresses gluconeogenesis by Akt-mediated FoxO1 inhibition. Thus, Rap1a regulates hepatic glucose homeostasis, and blocking its activity, via lowering geranylgeranyl isoprenoids, contributes to statin-induced glucose intolerance. Wang et al. show that activation of hepatic Rap1a suppresses gluconeogenic gene expression and improves glucose intolerance via Akt-mediated FoxO1 inhibition. Statins lower intracellular isoprenoid levels and inhibit Rap1a activation, which contributes to their hyperglycemic effect. These findings identify a role of hepatic Rap1a in obesity- and statin-associated glucose homeostasis.
Collapse
Affiliation(s)
- Yating Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Stefano Spolitu
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - John A Zadroga
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Amesh K Sarecha
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
31
|
Needamangalam Balaji J, Prakash S, Joshi A, Surapaneni KM. A Scoping Review on the Reported Evidence and Gaps of the Risk of Diabetes in Dyslipidemic Patients under Statin Therapy. Clin Pract 2022; 12:565-578. [PMID: 35892446 PMCID: PMC9326747 DOI: 10.3390/clinpract12040060] [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: 05/20/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
With the increasing global burden of dyslipidemia over the past 30 years, it is estimated that more than 200 million people worldwide are under statin therapy. In India, roughly 25-30% of urban populations and 15-20% of rural populations have abnormal lipid levels. Statin, which is deemed to be the gold standard lipid-lowering agent, is the first treatment of choice for these patients. Although statins at one end are highly effective against dyslipidemiaand cardiovascular diseases, at the other end, they cause adverse effects including an increased risk of diabetes mellitus. The objective of this study was to understand the coexistence of diabetes and dyslipidemia in patients undergoing statin therapy. A scoping review was conducted with published articles selected from PubMed and Google Scholar. The obtained results were filtered based on inclusion/exclusion criteria. Our database search provided a total of 822 articles, of which 48 were selected for this review, with results concluding that statin users are potentially at a greater risk of developing diabetes mellitus compared with patients who are not using statins. Although many studies have been conducted to ascertain the onset of diabetes mellitus amongst statin users, the exact mechanism is not yet precisely established. Future studies are essential for identifying the exact cause of diabetes mellitus in statin users.
Collapse
Affiliation(s)
- Jyotsna Needamangalam Balaji
- Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Chennai 600-123, Tamil Nadu, India; (J.N.B.); (S.P.)
| | - Sreenidhi Prakash
- Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Chennai 600-123, Tamil Nadu, India; (J.N.B.); (S.P.)
| | - Ashish Joshi
- CUNY Graduate School of Public Health & Health Policy, New York, NY 10027, USA;
- SMAART Population Health Informatics Intervention Center, Foundation of Healthcare Technologies Society, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Chennai 600-123, Tamil Nadu, India
| | - Krishna Mohan Surapaneni
- SMAART Population Health Informatics Intervention Center, Foundation of Healthcare Technologies Society, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Chennai 600-123, Tamil Nadu, India
- Departments of Biochemistry, Medical Education, Molecular Virology, Research, Clinical Skills & Simulation, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Chennai 600-123, Tamil Nadu, India
| |
Collapse
|
32
|
Nsaibia MJ, Devendran A, Goubaa E, Bouitbir J, Capoulade R, Bouchareb R. Implication of Lipids in Calcified Aortic Valve Pathogenesis: Why Did Statins Fail? J Clin Med 2022; 11:jcm11123331. [PMID: 35743402 PMCID: PMC9225514 DOI: 10.3390/jcm11123331] [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: 03/29/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022] Open
Abstract
Calcific Aortic Valve Disease (CAVD) is a fibrocalcific disease. Lipoproteins and oxidized phospholipids play a substantial role in CAVD; the level of Lp(a) has been shown to accelerate the progression of valve calcification. Indeed, oxidized phospholipids carried by Lp(a) into the aortic valve stimulate endothelial dysfunction and promote inflammation. Inflammation and growth factors actively promote the synthesis of the extracellular matrix (ECM) and trigger an osteogenic program. The accumulation of ECM proteins promotes lipid adhesion to valve tissue, which could initiate the osteogenic program in interstitial valve cells. Statin treatment has been shown to have the ability to diminish the death rate in subjects with atherosclerotic impediments by decreasing the serum LDL cholesterol levels. However, the use of HMG-CoA inhibitors (statins) as cholesterol-lowering therapy did not significantly reduce the progression or the severity of aortic valve calcification. However, new clinical trials targeting Lp(a) or PCSK9 are showing promising results in reducing the severity of aortic stenosis. In this review, we discuss the implication of lipids in aortic valve calcification and the current findings on the effect of lipid-lowering therapy in aortic stenosis.
Collapse
Affiliation(s)
- Mohamed J. Nsaibia
- Department of Cell Biology and Molecular Medicine, Rutgers University, Newark, NJ 07103, USA;
| | - Anichavezhi Devendran
- Department of Medicine, Cardiovascular Research Institute, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Eshak Goubaa
- Thomas Jefferson University East Falls, Philadelphia, PA 19144, USA;
| | - Jamal Bouitbir
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, 4056 Basel, Switzerland;
| | - Romain Capoulade
- L’institut Du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France;
| | - Rihab Bouchareb
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence: or ; Tel.: +1-(212)-241-8471
| |
Collapse
|
33
|
Alvarez-Jimenez L, Morales-Palomo F, Moreno-Cabañas A, Ortega JF, Mora-Rodriguez R. Statins effect on insulin resistance after a meal and exercise in hypercholesterolemic pre-diabetic individuals. Scand J Med Sci Sports 2022; 32:1346-1355. [PMID: 35612762 PMCID: PMC9541393 DOI: 10.1111/sms.14193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/28/2022]
Abstract
Aim To study if statins, a widely prescribed, inexpensive medication to prevent coronary artery diseases may cause insulin resistance (IR). Methods Fasted (HOMA‐IR) and post‐meal insulin resistance were assessed in 21 pre‐diabetic hypercholesterolemic individuals treated with statins (STA trial). Measurements were compared to another trial conducted 96 h after statin withdrawal using placebo pills (PLAC trial). Trials were duplicated 16–18 h after a bout of moderate‐intensity exercise (500 kcal of energy expenditure) to reduce IR and better appreciate statin effects (EXER+STA and EXER+PLAC trials). Results Statin withdrawal did not affect fasting (HOMA‐IR; 2.35 ± 1.05 vs. 2.18 ± 0.87 for STA vs. PLAC trials; p = 0.150) or post‐meal insulin resistance (i.e., Matsuda‐index, STA 6.23 ± 2.83 vs. PLAC 6.49 ± 3.74; p = 0.536). A bout of aerobic exercise lowered post‐meal IR (p = 0.043), but statin withdrawal did not add to the exercise actions (p = 0.564). Statin withdrawal increased post‐meal plasma free glycerol concentrations (0.136 ± 0.073 vs. 0.185 ± 0.090 mmol·L−1 for STA vs. PLAC trials; p < 0.001) but not plasma free fatty acids or fat oxidation (p = 0.981, and p = 0.621, respectively). Post‐meal fat oxidation was higher in the exercise trials (p = 0.002). Conclusions Withdrawal of statin medication does not affect fasting or post‐meal insulin resistance in pre‐diabetic hypercholesterolemic individuals. Furthermore, statin use does not interfere with the beneficial effects of exercise on lowering IR.
Collapse
Affiliation(s)
| | - Felix Morales-Palomo
- Exercise Physiology Lab at Toledo, University of Castilla-La Mancha, Toledo, Spain
| | | | - Juan Fernando Ortega
- Exercise Physiology Lab at Toledo, University of Castilla-La Mancha, Toledo, Spain
| | | |
Collapse
|
34
|
Doerfler AM, Han J, Jarrett KE, Tang L, Jain A, Saltzman A, De Giorgi M, Chuecos M, Hurley AE, Li A, Morand P, Ayala C, Goodlett DR, Malovannaya A, Martin JF, de Aguiar Vallim TQ, Shroyer N, Lagor WR. Intestinal Deletion of 3-Hydroxy-3-Methylglutaryl-Coenzyme A Reductase Promotes Expansion of the Resident Stem Cell Compartment. Arterioscler Thromb Vasc Biol 2022; 42:381-394. [PMID: 35172604 PMCID: PMC8957608 DOI: 10.1161/atvbaha.122.317320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/21/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The intestine occupies the critical interface between cholesterol absorption and excretion. Surprisingly little is known about the role of de novo cholesterol synthesis in this organ, and its relationship to whole body cholesterol homeostasis. Here, we investigate the physiological importance of this pathway through genetic deletion of the rate-limiting enzyme. METHODS Mice lacking 3-hydroxy-3-methylglutaryl-coenzyme A reductase (Hmgcr) in intestinal villus and crypt epithelial cells were generated using a Villin-Cre transgene. Plasma lipids, intestinal morphology, mevalonate pathway metabolites, and gene expression were analyzed. RESULTS Mice with intestine-specific loss of Hmgcr were markedly smaller at birth, but gain weight at a rate similar to wild-type littermates, and are viable and fertile into adulthood. Intestine lengths and weights were greater relative to body weight in both male and female Hmgcr intestinal knockout mice. Male intestinal knockout had decreased plasma cholesterol levels, whereas fasting triglycerides were lower in both sexes. Lipidomics revealed substantial reductions in numerous nonsterol isoprenoids and sterol intermediates within the epithelial layer, but cholesterol levels were preserved. Hmgcr intestinal knockout mice also showed robust activation of SREBP-2 (sterol-regulatory element binding protein-2) target genes in the epithelium, including the LDLR (low-density lipoprotein receptor). At the cellular level, loss of Hmgcr is compensated for quickly after birth through a dramatic expansion of the stem cell compartment, which persists into adulthood. CONCLUSIONS Loss of Hmgcr in the intestine is compatible with life through compensatory increases in intestinal absorptive surface area, LDLR expression, and expansion of the resident stem cell compartment.
Collapse
Affiliation(s)
- Alexandria M. Doerfler
- Molecular Physiology and Biophysics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Jun Han
- University of Victoria - Genome British Columbia Proteomics Centre, Victoria, British Columbia, Canada
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Kelsey E. Jarrett
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Division of Cardiology, University of California Los Angeles, Los Angeles, USA
| | - Li Tang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
| | - Alexander Saltzman
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
| | - Marco De Giorgi
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Marcel Chuecos
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas, USA
| | - Ayrea E. Hurley
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Ang Li
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Pauline Morand
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, USA
| | - Claudia Ayala
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - David R. Goodlett
- University of Victoria - Genome British Columbia Proteomics Centre, Victoria, British Columbia, Canada
- Department of Biochemistry & Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Anna Malovannaya
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - James F. Martin
- Molecular Physiology and Biophysics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, Texas, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas USA
| | - Thomas Q. de Aguiar Vallim
- Department of Medicine, Division of Cardiology, University of California Los Angeles, Los Angeles, USA
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, USA
- Johnsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, USA
| | - Noah Shroyer
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - William R. Lagor
- Molecular Physiology and Biophysics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas USA
| |
Collapse
|
35
|
Wood AC, Arora A, Newell M, Bland VL, Zhou J, Pirastu N, Ordovas JM, Klimentidis YC. Identification of genetic loci simultaneously associated with multiple cardiometabolic traits. Nutr Metab Cardiovasc Dis 2022; 32:1027-1034. [PMID: 35168826 PMCID: PMC9275655 DOI: 10.1016/j.numecd.2022.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/09/2021] [Accepted: 01/04/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND AIMS Cardiometabolic disorders (CMD) arise from a constellation of features such as increased adiposity, hyperlipidemia, hypertension and compromised glucose control. Many genetic loci have shown associations with individual CMD-related traits, but no investigations have focused on simultaneously identifying loci showing associations across all domains. We therefore sought to identify loci associated with risk across seven continuous CMD-related traits. METHODS AND RESULTS We conducted separate genome-wide association studies (GWAS) for systolic and diastolic blood pressure (SBP/DBP), hemoglobin A1c (HbA1c), low- and high- density lipoprotein cholesterol (LDL-C/HDL-C), waist-to-hip-ratio (WHR), and triglycerides (TGs) in the UK Biobank (N = 356,574-456,823). Multiple loci reached genome-wide levels of significance (N = 145-333) for each trait, but only four loci (in/near VEGFA, GRB14-COBLL1, KLF14, and RGS19-OPRL1) were associated with risk across all seven traits (P < 5 × 10-8). We sought replication of these four loci in an independent set of seven trait-specific GWAS meta-analyses. GRB14-COBLL1 showed the most consistent replication, revealing nominally significant associations (P < 0.05) with all traits except DBP. CONCLUSIONS Our analyses suggest that very few loci are associated in the same direction of risk with traits representing the full spectrum of CMD features. We identified four such loci, and an understanding of the pathways between these loci and CMD risk may eventually identify factors that can be used to identify pathologic disturbances that represent broadly beneficial therapeutic targets.
Collapse
Affiliation(s)
- Alexis C Wood
- USDA/ARS Children's Nutrition Research Center, 1100 Bates Avenue, Houston, TX, USA.
| | - Amit Arora
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Michelle Newell
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Victoria L Bland
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jin Zhou
- Department of Biostatistics, University of California, Los Angeles, CA, USA
| | - Nicola Pirastu
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland, UK
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA; IMDEA-Food, Madrid, Spain
| | - Yann C Klimentidis
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA; BIO5 Institute, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
36
|
Wang L, Zhu L, Zheng Z, Meng L, Liu H, Wang K, Chen J, Li P, Yang H. Mevalonate pathway orchestrates insulin signaling via RAB14 geranylgeranylation-mediated phosphorylation of AKT to regulate hepatic glucose metabolism. Metabolism 2022; 128:155120. [PMID: 34995578 DOI: 10.1016/j.metabol.2021.155120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022]
Abstract
Statin use accompanies with increased risk of new onset of type 2 diabetes, however, the underlying mechanisms remain not be fully understood and effective prevention strategies are still lacking. Herein, we find that both pharmacological and genetic inhibition of GGTase II mimic the disruption of simvastatin on hepatic insulin signaling and glucose metabolism in vitro. AAV8-mediated knockdown of liver RABGGTA, the specific subunit of GGTase II, triggers systemic glucose metabolism disorders in vivo. By adopting a small-scale siRNA screening, we identify RAB14 as a regulator of hepatic insulin signaling and glucose metabolism. Geranylgeranylation deficiency of RAB14 inhibits the phosphorylation of AKT (Ser473) and disrupts hepatic insulin signaling and glucose metabolism possibly via impeding mTORC2 complex assembly. Finally, geranylgeranyl pyrophosphate (GGPP) supplementation is sufficient to prevent simvastatin-caused disruption of hepatic insulin signaling and glucose metabolism in vitro. Geranylgeraniol (GGOH), a precursor of GGPP, is able to ameliorate simvastatin-induced systemic glucose metabolism disorders in vivo. In conclusion, our data indicate that statins-targeted mevalonate pathway regulates hepatic insulin signaling and glucose metabolism via geranylgeranylation of RAB14. GGPP/GGOH supplementation might be an effective strategy for the prevention of the diabetic effects of statins.
Collapse
Affiliation(s)
- Lai Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lijun Zhu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zuguo Zheng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lingchang Meng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hanling Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Keke Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Chen
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Hua Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
37
|
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
|
38
|
Fine-Tuning Fuzzy KNN Classifier Based on Uncertainty Membership for the Medical Diagnosis of Diabetes. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12030950] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Diabetes, a metabolic disease in which the blood glucose level rises over time, is one of the most common chronic diseases at present. It is critical to accurately predict and classify diabetes to reduce the severity of the disease and treat it early. One of the difficulties that researchers face is that diabetes datasets are limited and contain outliers and missing data. Additionally, there is a trade-off between classification accuracy and operational law for detecting diabetes. In this paper, an algorithm for diabetes classification is proposed for pregnant women using the Pima Indians Diabetes Dataset (PIDD). First, a preprocessing step in the proposed algorithm includes outlier rejection, imputing missing values, the standardization process, and feature selection of the attributes, which enhance the dataset’s quality. Second, the classifier uses the fuzzy KNN method and modifies the membership function based on the uncertainty theory. Third, a grid search method is applied to achieve the best values for tuning the fuzzy KNN method based on uncertainty membership, as there are hyperparameters that affect the performance of the proposed classifier. In turn, the proposed tuned fuzzy KNN based on uncertainty classifiers (TFKNN) deals with the belief degree, handles membership functions and operation law, and avoids making the wrong categorization. The proposed algorithm performs better than other classifiers that have been trained and evaluated, including KNN, fuzzy KNN, naïve Bayes (NB), and decision tree (DT). The results of different classifiers in an ensemble could significantly improve classification precision. The TFKNN has time complexity O(kn2d), and space complexity O(n2d). The TFKNN model has high performance and outperformed the others in all tests in terms of accuracy, specificity, precision, and average AUC, with values of 90.63, 85.00, 93.18, and 94.13, respectively. Additionally, results of empirical analysis of TFKNN compared to fuzzy KNN, KNN, NB, and DT demonstrate the global superiority of TFKNN in precision, accuracy, and specificity.
Collapse
|
39
|
Kuhlman AB, Mikkelsen LB, Regnersgaard S, Heinrichsen S, Nielsen FH, Frandsen J, Orlando P, Silvestri S, Larsen S, Helge JW, Dela F. The effect of 8 weeks of physical training on muscle performance and maximal fat oxidation rates in patients treated with simvastatin and coenzyme Q10 supplementation. J Physiol 2021; 600:569-581. [PMID: 34891216 DOI: 10.1113/jp281475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 11/25/2021] [Indexed: 11/08/2022] Open
Abstract
Statins are prescribed for the treatment of elevated cholesterol, but they may negatively affect metabolism, muscle performance, and the response to training. Coenzyme Q10 (CoQ10) supplementation may alleviate these effects. Combined simvastatin and CoQ10 treatment during physical training has never been tested. We studied the response to 8 weeks training (maximal oxygen uptake ( V ̇ O 2 max ), fat oxidation (MFO), the workload at which MFO occurred, and muscle strength) in statin naive dyslipidaemic patients who received simvastatin (40 mg/day) with (S + Q, n = 9) or without (S + Pl, n = 10) CoQ10 supplementation (2 × 200 mg/day) or placebo (Pl + Pl, n = 7) in a randomized, double-blind placebo-controlled study. V ̇ O 2 max and maximal workload increased with training (main effect of time, P < 0.05). MFO increased from 0.29 ± 0.10, 0.26 ± 0.10, and 0.38 ± 0.09 to 0.42 ± 0.09, 0.38 ± 0.10 and 0.48 ± 0.16 g/min in S + Q, S + Pl, and Pl + Pl, respectively (main effect of time, P = 0.0013). The workload at MFO increased from 75 ± 25, 56 ± 23, and 72 ± 17 to 106 ± 25, 84 ± 13 and 102 ± 31 W in S + Q, S + Pl, and Pl + Pl, respectively (main effect of time, P < 0.0001). Maximal voluntary contraction and rate of force development were unchanged. Exercise improved aerobic physical capacity and simvastatin with or without CoQ10 supplementation did not inhibit this adaptation. The similar increases in MFO and in the workload at which MFO occurred in response to training shows that the ability to adapt substrate selection and oxidation rates is preserved with simvastatin treatment, despite the potential negative impact of simvastatin at the mitochondrial level. CoQ10 supplementation does not augment this adaptation. KEY POINTS: Simvastatins are prescribed for treatment of elevated cholesterol, but they may negatively affect metabolism, muscle performance and the response to training. Coenzyme Q10 (CoQ10) supplementation may alleviate some of these effects. We found that simvastatin treatment does not negatively affect training-induced adaptations of substrate oxidation during exercise. Likewise, maximal oxygen uptake increases with physical training also in patients in treatment with simvastatin. CoQ10 supplementation in simvastatin-treated patients presents no advantage in the adaptations to physical training Simvastatin treatment decreases plasma concentrations of total CoQ10, but this can be alleviated by simultaneous supplementation with CoQ10.
Collapse
Affiliation(s)
- Anja Birk Kuhlman
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Bluhme Mikkelsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Signe Regnersgaard
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Heinrichsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Frederikke Hyldahl Nielsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Frandsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Patrick Orlando
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Steen Larsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Jørn Wulff Helge
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Dela
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Geriatrics, Bispebjerg University Hospital, Copenhagen, Denmark
| |
Collapse
|
40
|
Jack A, Mohd MA, Kamaruddin NN, Mohd Din LH, Hajri NA, Tengku Muhammad TS. Acaudina molpadioides mediates lipid uptake by suppressing PCSK9 transcription and increasing LDL receptor in human liver cells. Saudi J Biol Sci 2021; 28:7105-7116. [PMID: 34867013 PMCID: PMC8626262 DOI: 10.1016/j.sjbs.2021.08.003] [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: 12/22/2020] [Revised: 07/30/2021] [Accepted: 08/01/2021] [Indexed: 11/09/2022] Open
Abstract
Acaudina molpadioides has been long used as traditional medicinal resources and reported to demonstrate various important bioactivities such as anticoagulation, antithrombosis, anti-hyperglycemia and anticancer. However, its lipid lowering activity is yet to be fully explored. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is an enzyme that enhances the lysosomal degradation of hepatic low density lipoprotein receptor (LDLR) resulting in excessive accumulation of the plasma levels of LDL-cholesterols (LDL-C) which subsequently accelerate atherosclerosis. In the present study, A. molpadioides fractions were subjected to promoter-reporter luciferase assay to determine its role as PCSK9 inhibitors. It was found both fractions (EFA and EFB) reduced the transcriptional activity of PCSK9 promoter. Among the seven 5′end deletion constructs of PCSK9 promoter, fragments D1 (−1,711/−94), D3 (−709/−94) and D4 (−440/−94), were suppressed in the presence of both fractions whereas D2 (−1,214/−94), and, D6 (−351/−94) as well as D7 (−335/−94) were inhibited only by EFA and EFB, respectively. Further transcription factor binding sites prediction using MatInspector software discovered various potential cis-regulatory elements namely, PPAR, KLFs, RBPJ-kappa and SREBP that may potentially be involved in ameliorating the transcriptional activity of PCSK9. Immunofluorescence staining was used to evaluate the effects of both fractions on LDL-C and LDLR. Results showed that levels of LDL-C uptake in EFA-treated cells were 69.1% followed by EFB at 32.6%, as compared to untreated control after 24 h treatment. The LDLR protein distribution was induced by 62.41% and 32.2%, which corresponded to an increase in LDL-C uptake in both EFA and EFB treatment, respectively. Hence, the inhibition of PCSK9 by bioactive compounds in EFA and EFB could be another promising therapeutic agent in reducing the cholesterol levels and atherosclerosis by targeting PCSK9.
Collapse
Affiliation(s)
- Allicia Jack
- Nutrition & Food Safety Programme, Food Science & Technology Research Centre, Malaysian Agricultural Research & Development Institute (MARDI), Persiaran MARDI-UPM, 43400 Serdang, Selangor, Malaysia.,Institute of Marine Biotechnology, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Muzaida Aminah Mohd
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | | | - Lukman Hakim Mohd Din
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Nor Azwin Hajri
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | | |
Collapse
|
41
|
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
|
42
|
Zhou Z, Chowdhury EK, Breslin M, Curtis AJ, Reid CM, Nelson M. Antihypertensive drug class in combination with lipid lowering treatment for primary prevention of cardiovascular disease in the elderly. Heart Lung 2021; 51:40-45. [PMID: 34731696 DOI: 10.1016/j.hrtlng.2021.09.005] [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: 06/29/2021] [Revised: 08/20/2021] [Accepted: 09/05/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Antihypertensives and lipid-lowering therapy (LLT) are often used concurrently. OBJECTIVES To determine whether there was a difference in clinical outcomes when older patients with LLT were prescribed angiotensin-converting-enzyme-inhibitors (ACE-Is) compared with diuretics. METHODS This analysis included 648 LLT older users free of cardiovascular disease (CVD) from a trial comparing ACE-I versus diuretic-based therapy. Comparisons were made between LLT+ACE-I (n = 335) and LLT+diuretic groups (n = 313) using multivariable Cox proportional-hazard models. Primary endpoints were all-cause and CVD mortality (in-trial [4.1-year]+post-trial [6.9-year]) and secondary endpoints (in-trial) were the composite of all-cause mortality and first CVD events and its components, CVD mortality and incident diabetes. RESULTS There were no significant differences between the two groups for the primary endpoints over the in-trial plus post-trial follow-up, nor was there a difference for any secondary outcomes over the in-trial follow-up. CONCLUSIONS The LLT+ACE-I and LLT+diuretic combinations showed similar effects in CVD-free older individuals. Randomised trials are needed to provide conclusive evidence.
Collapse
Affiliation(s)
- Zhen Zhou
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool street, Hobart TAS 7000, Australia.
| | | | - Monique Breslin
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool street, Hobart TAS 7000, Australia
| | - Andrea J Curtis
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | | | - Mark Nelson
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool street, Hobart TAS 7000, Australia
| |
Collapse
|
43
|
Sanvee GM, Hitzfeld L, Bouitbir J, Krähenbühl S. mTORC2 is an important target for simvastatin-associated toxicity in C2C12 cells and mouse skeletal muscle - Roles of Rap1 geranylgeranylation and mitochondrial dysfunction. Biochem Pharmacol 2021; 192:114750. [PMID: 34461118 DOI: 10.1016/j.bcp.2021.114750] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/25/2022]
Abstract
Statins decrease the serum LDL-cholesterol concentration and reduce the risk for cardiovascular diseases but can cause myopathy, which may be related to mTORC inhibition. In the current study, we investigated which mTORC is inhibited by simvastatin and by which mechanisms. In C2C12 myoblasts and myotubes and mouse gastrocnemius, simvastatin was cytotoxic and inhibited S6rp and Akt Ser473 phosphorylation, indicating inhibition of mTORC1 and mTORC2, respectively. In contrast to simvastatin, the mTORC1 inhibitor rapamycin did not inhibit mTORC2 activity and was not cytotoxic. Like simvastatin, knock-down of Rictor, an essential component of mTORC2, impaired Akt Ser473 and S6rp phosphorylation and was cytotoxic for C2C12 myoblasts, suggesting that mTORC2 inhibition is an important myotoxic mechanism. The investigation of the mechanism of mTORC2 inhibition showed that simvastatin impaired Ras farnesylation, which was prevented by farnesol but without restoring mTORC2 activity. In comparison, Rap1 knock-down reduced mTORC2 activity and was cytotoxic for C2C12 myoblasts. Simvastatin impaired Rap1 geranylgeranylation and function, which was prevented by geranylgeraniol. In addition, simvastatin and the complex III inhibitor antimycin A caused mitochondrial superoxide accumulation and impaired the activity of mTORC2, which could partially be prevented by the antioxidant MitoTEMPO. In conclusion, mTORC2 inhibition is an important mechanism of simvastatin-induced myotoxicity. Simvastatin inhibits mTORC2 by impairing geranylgeranylation of Rap1 and by inducing mitochondrial dysfunction.
Collapse
Affiliation(s)
- Gerda M Sanvee
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland
| | - Leonie Hitzfeld
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland
| | - Jamal Bouitbir
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland; Division of Molecular and Systemic Toxicology, Department of Pharmaceutical Sciences, University of Basel, Switzerland; Swiss Centre for Applied Human Research (SCAHT), Switzerland
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland; Swiss Centre for Applied Human Research (SCAHT), Switzerland.
| |
Collapse
|
44
|
Yeoh SG, Sum JS, Lai JY, W Isa WYH, Lim TS. Potential of Phage Display Antibody Technology for Cardiovascular Disease Immunotherapy. J Cardiovasc Transl Res 2021; 15:360-380. [PMID: 34467463 DOI: 10.1007/s12265-021-10169-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/22/2021] [Indexed: 11/26/2022]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. CVD includes coronary artery diseases such as angina, myocardial infarction, and stroke. "Lipid hypothesis" which is also known as the cholesterol hypothesis proposes the linkage of plasma cholesterol level with the risk of developing CVD. Conventional management involves the use of statins to reduce the serum cholesterol levels as means for CVD prevention or treatment. The regulation of serum cholesterol levels can potentially be regulated with biological interventions like monoclonal antibodies. Phage display is a powerful tool for the development of therapeutic antibodies with successes over the recent decade. Although mainly for oncology, the application of monoclonal antibodies as immunotherapeutic agents could potentially be expanded to CVD. This review focuses on the concept of phage display for antibody development and discusses the potential target antigens that could potentially be beneficial for serum cholesterol management.
Collapse
Affiliation(s)
- Soo Ghee Yeoh
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Jia Siang Sum
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Jing Yi Lai
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - W Y Haniff W Isa
- School of Medical Sciences, Department of Medicine, Universiti Sains Malaysia, Kubang Kerian, 16150, Kelantan, Malaysia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia.
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800, Penang, Malaysia.
| |
Collapse
|
45
|
Veluthakal R, Thurmond DC. Emerging Roles of Small GTPases in Islet β-Cell Function. Cells 2021; 10:1503. [PMID: 34203728 PMCID: PMC8232272 DOI: 10.3390/cells10061503] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/08/2021] [Accepted: 06/08/2021] [Indexed: 12/16/2022] Open
Abstract
Several small guanosine triphosphatases (GTPases) from the Ras protein superfamily regulate glucose-stimulated insulin secretion in the pancreatic islet β-cell. The Rho family GTPases Cdc42 and Rac1 are primarily involved in relaying key signals in several cellular functions, including vesicle trafficking, plasma membrane homeostasis, and cytoskeletal dynamics. They orchestrate specific changes at each spatiotemporal region within the β-cell by coordinating with signal transducers, guanine nucleotide exchange factors (GEFs), GTPase-activating factors (GAPs), and their effectors. The Arf family of small GTPases is involved in vesicular trafficking (exocytosis and endocytosis) and actin cytoskeletal dynamics. Rab-GTPases regulate pre-exocytotic and late endocytic membrane trafficking events in β-cells. Several additional functions for small GTPases include regulating transcription factor activity and mitochondrial dynamics. Importantly, defects in several of these GTPases have been found associated with type 2 diabetes (T2D) etiology. The purpose of this review is to systematically denote the identities and molecular mechanistic steps in the glucose-stimulated insulin secretion pathway that leads to the normal release of insulin. We will also note newly identified defects in these GTPases and their corresponding regulatory factors (e.g., GDP dissociation inhibitors (GDIs), GEFs, and GAPs) in the pancreatic β-cells, which contribute to the dysregulation of metabolism and the development of T2D.
Collapse
Affiliation(s)
- Rajakrishnan Veluthakal
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| |
Collapse
|
46
|
Accelerated DNA methylation age and medication use among African Americans. Aging (Albany NY) 2021; 13:14604-14629. [PMID: 34083497 PMCID: PMC8221348 DOI: 10.18632/aging.203115] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 05/14/2021] [Indexed: 12/16/2022]
Abstract
DNA methylation age acceleration, the discrepancy between epigenetic age and chronological age, is associated with mortality and chronic diseases, including diabetes, hypertension, and hyperlipidemia. In this study, we investigate whether medications commonly used to treat these diseases in 15 drug categories are associated with four epigenetic age acceleration measures: HorvathAge acceleration (HorvathAA), HannumAge acceleration (HannumAA), PhenoAge acceleration, and GrimAge acceleration (GrimAA) using cross-sectional (Phase 1, N=1,100) and longitudinal (Phases 1 and 2, N=266) data from African Americans in the Genetic Epidemiology Network of Arteriopathy (GENOA) study. In cross-sectional analyses, the use of calcium channel blockers was associated with 1.27 years lower HannumAA after adjusting for covariates including hypertension (p=0.001). Longitudinal analyses showed that, compared to those who never used antihypertensives, those who started to take antihypertensives after Phase 1 had a 0.97-year decrease in GrimAA (p=0.007). In addition, compared to those who never used NSAID analgesics, those who started to take them after Phase 1 had a 2.61-year increase in HorvathAA (p=0.0005). Our study demonstrates that three commonly used medications are associated with DNAm age acceleration in African Americans and sheds light on the potential epigenetic effects of pharmaceuticals on aging at the cellular level.
Collapse
|
47
|
Effects of Simvastatin on Lipid Metabolism in Wild-Type Mice and Mice with Muscle PGC-1α Overexpression. Int J Mol Sci 2021; 22:ijms22094950. [PMID: 34066911 PMCID: PMC8125015 DOI: 10.3390/ijms22094950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 12/05/2022] Open
Abstract
Previous studies suggest that statins may disturb skeletal muscle lipid metabolism potentially causing lipotoxicity with insulin resistance. We investigated this possibility in wild-type mice (WT) and mice with skeletal muscle PGC-1α overexpression (PGC-1α OE mice). In WT mice, simvastatin had only minor effects on skeletal muscle lipid metabolism but reduced glucose uptake, indicating impaired insulin sensitivity. Muscle PGC-1α overexpression caused lipid droplet accumulation in skeletal muscle with increased expression of the fatty acid transporter CD36, fatty acid binding protein 4, perilipin 5 and CPT1b but without significant impairment of muscle glucose uptake. Simvastatin further increased the lipid droplet accumulation in PGC-1α OE mice and stimulated muscle glucose uptake. In conclusion, the impaired muscle glucose uptake in WT mice treated with simvastatin cannot be explained by lipotoxicity. PGC-1α OE mice are protected from lipotoxicity of fatty acids and triglycerides by increased the expression of FABP4, formation of lipid droplets and increased expression of CPT1b.
Collapse
|
48
|
O'Flaherty M, Lloyd-Williams F, Capewell S, Boland A, Maden M, Collins B, Bandosz P, Hyseni L, Kypridemos C. Modelling tool to support decision-making in the NHS Health Check programme: workshops, systematic review and co-production with users. Health Technol Assess 2021; 25:1-234. [PMID: 34076574 PMCID: PMC8201571 DOI: 10.3310/hta25350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Local authorities in England commission the NHS Health Check programme to invite everyone aged 40-74 years without pre-existing conditions for risk assessment and eventual intervention, if needed. However, the programme's effectiveness, cost-effectiveness and equity impact remain uncertain. AIM To develop a validated open-access flexible web-based model that enables local commissioners to quantify the cost-effectiveness and potential for equitable population health gain of the NHS Health Check programme. OBJECTIVES The objectives were as follows: (1) co-produce with stakeholders the desirable features of the user-friendly model; (2) update the evidence base to support model and scenario development; (3) further develop our computational model to allow for developments and changes to the NHS Health Check programme and the diseases it addresses; (4) assess the effectiveness, cost-effectiveness and equity of alternative strategies for implementation to illustrate the use of the tool; and (5) propose a sustainability and implementation plan to deploy our user-friendly computational model at the local level. DESIGN Co-production workshops surveying the best-performing local authorities and a systematic literature review of strategies to increase uptake of screening programmes informed model use and development. We then co-produced the workHORSE (working Health Outcomes Research Simulation Environment) model to estimate the health, economic and equity impact of different NHS Health Check programme implementations, using illustrative-use cases. SETTING Local authorities in England. PARTICIPANTS Stakeholders from local authorities, Public Health England, the NHS, the British Heart Foundation, academia and other organisations participated in the workshops. For the local authorities survey, we invited 16 of the best-performing local authorities in England. INTERVENTIONS The user interface allows users to vary key parameters that represent programme activities (i.e. invitation, uptake, prescriptions and referrals). Scenarios can be compared with each other. MAIN OUTCOME MEASURES Disease cases and case-years prevented or postponed, incremental cost-effectiveness ratios, net monetary benefit and change in slope index of inequality. RESULTS The survey of best-performing local authorities revealed a diversity of effective approaches to maximise the coverage and uptake of NHS Health Check programme, with no distinct 'best buy'. The umbrella literature review identified a range of effective single interventions. However, these generally need to be combined to maximally improve uptake and health gains. A validated dynamic, stochastic microsimulation model, built on robust epidemiology, enabled service options analysis. Analyses of three contrasting illustrative cases estimated the health, economic and equity impact of optimising the Health Checks, and the added value of obtaining detailed local data. Optimising the programme in Liverpool can become cost-effective and equitable, but simply changing the invitation method will require other programme changes to improve its performance. Detailed data inputs can benefit local analysis. LIMITATIONS Although the approach is extremely flexible, it is complex and requires substantial amounts of data, alongside expertise to both maintain and run. CONCLUSIONS Our project showed that the workHORSE model could be used to estimate the health, economic and equity impact comprehensively at local authority level. It has the potential for further development as a commissioning tool and to stimulate broader discussions on the role of these tools in real-world decision-making. FUTURE WORK Future work should focus on improving user interactions with the model, modelling simulation standards, and adapting workHORSE for evaluation, design and implementation support. STUDY REGISTRATION This study is registered as PROSPERO CRD42019132087. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 25, No. 35. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Martin O'Flaherty
- Department of Public Health and Policy, University of Liverpool, Liverpool, UK
| | | | - Simon Capewell
- Department of Public Health and Policy, University of Liverpool, Liverpool, UK
| | - Angela Boland
- Liverpool Reviews and Implementation Group, University of Liverpool, Liverpool, UK
| | - Michelle Maden
- Liverpool Reviews and Implementation Group, University of Liverpool, Liverpool, UK
| | - Brendan Collins
- Department of Public Health and Policy, University of Liverpool, Liverpool, UK
| | - Piotr Bandosz
- Department of Public Health and Policy, University of Liverpool, Liverpool, UK
| | - Lirije Hyseni
- Department of Public Health and Policy, University of Liverpool, Liverpool, UK
| | - Chris Kypridemos
- Department of Public Health and Policy, University of Liverpool, Liverpool, UK
| |
Collapse
|
49
|
Pulipati VP, Davidson MH. How I treat statin-associated side effects in an outpatient setting. Future Cardiol 2021; 17:1249-1260. [PMID: 33464124 DOI: 10.2217/fca-2020-0153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Dyslipidemia promotes atherosclerosis and causes cardiovascular diseases. Statins are potent lipid-lowering medications with a cardiovascular mortality benefit. They are generally safe and well tolerated but sometimes can be associated with side effects of variable severity. The most common side effect is statin-associated muscle symptoms. Uncommon side effects include new-onset diabetes mellitus and elevation in liver enzymes. These effects can lead to noncompliance and premature discontinuation of the medication. Hence, it is crucial to identify patients with true statin-associated side effects (SASE) to ensure optimal statin use. The appropriate evaluation of the patient before starting statins and proactive utilization of available diagnostic tests to rule out alternate etiologies mimicking adverse effects are essential for accurate diagnosis of SASE. In patients with true SASE, timely intervention with modified statin or non-statins is beneficial. Herein, we discuss key clinical trial data on statins and non-statins, and describe our center's approach toward patients with SASE.
Collapse
Affiliation(s)
- Vishnu Priya Pulipati
- Preventive Cardiology, Section of Cardiology, The University of Chicago, 5841 S Maryland Avenue, MC 6080 B-608A, Chicago, IL 60637, USA
| | - Michael H Davidson
- Preventive Cardiology, Section of Cardiology, The University of Chicago, 5841 S Maryland Avenue, MC 6080 B-608A, Chicago, IL 60637, USA
| |
Collapse
|
50
|
Cosentino F, Grant PJ, Aboyans V, Bailey CJ, Ceriello A, Delgado V, Federici M, Filippatos G, Grobbee DE, Hansen TB, Huikuri HV, Johansson I, Jüni P, Lettino M, Marx N, Mellbin LG, Östgren CJ, Rocca B, Roffi M, Sattar N, Seferović PM, Sousa-Uva M, Valensi P, Wheeler DC. 2019 ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD. Eur Heart J 2021; 41:255-323. [PMID: 31497854 DOI: 10.1093/eurheartj/ehz486] [Citation(s) in RCA: 2582] [Impact Index Per Article: 645.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|