1
|
Özel HF, Alpay Ş, Asker E, Gültekin ES, Kazdağlı H. SGLT-2 inhibitors on cardiac autonomic function in individuals with and without type 2 diabetes mellitus. J Diabetes Complications 2025; 39:109021. [PMID: 40158451 DOI: 10.1016/j.jdiacomp.2025.109021] [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: 02/23/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Sodium-glucose cotransporter-2 (SGLT-2) inhibitors have emerged as key therapeutic agents in managing type 2 diabetes mellitus (T2DM) and obesity, offering benefits that extend beyond glycemic control. This review examines the role of SGLT-2 inhibitors in modulating cardiac autonomic function, with a particular focus on heart rate variability (HRV) as a biomarker of autonomic balance. These agents improve metabolic profiles through enhanced glucosuria, natriuresis, and weight loss, while concurrently reducing blood pressure. Importantly, they also attenuate sympathetic nervous system overactivity and promote parasympathetic modulation, which may lower the risk of adverse cardiovascular events. The underlying mechanisms include not only the metabolic effects but also anti-inflammatory and antioxidative actions, which together contribute to improved endothelial function and vascular health. Advanced HRV analyses, encompassing traditional time and frequency domain methods as well as nonlinear approaches, have proven valuable in detecting early autonomic dysfunction in high-risk populations. Some studies suggest that SGLT-2 inhibitors may be associated with improvements in HRV parameters, such as increased SDNN and RMSSD and a reduced LF/HF ratio. However, findings are inconsistent across studies, and further research is needed to determine the extent and mechanisms of these potential effects. Although these findings are promising, further standardized, long-term studies are essential to clarify the mechanisms and optimal therapeutic strategies involving SGLT-2 inhibitors in the management of autonomic dysfunction. Future research should also explore the synergistic potential of combining SGLT-2 inhibitors with other cardiometabolic therapies to enhance cardiovascular outcomes in individuals with and without T2DM.
Collapse
Affiliation(s)
- Hasan Fehmi Özel
- Vocational School of Health Services, Manisa Celal Bayar University, Manisa, Türkiye
| | - Şüheda Alpay
- Physiology Dept., Faculty of Medicine, Manisa Celal Bayar University, Manisa, Türkiye
| | - Emre Asker
- Physiology Dept., Faculty of Medicine, Manisa Celal Bayar University, Manisa, Türkiye; Pathology Dept., Faculty of Medicine, Trakya University, Edirne, Türkiye
| | - Elif Sıdal Gültekin
- Family Medicine Dept., Faculty of Medicine, Manisa Celal Bayar University, Manisa, Türkiye
| | - Hasan Kazdağlı
- Vocational School of Health Services, Izmir University of Economics, Izmir, Türkiye.
| |
Collapse
|
2
|
Sawalha K, Gautam N, Sivakumar K, Paydak H, Mehta JL. Metformin: Its salutary effects beyond diabetes mellitus. J Investig Med 2025:10815589251327511. [PMID: 40033492 DOI: 10.1177/10815589251327511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Metformin, an oral hypoglycemic agent, is commonly used in patients with type II diabetes mellitus. Studies have shown its use is associated with a reduction in major cardiovascular events (MACE) in patients with type 2 diabetes such as hospitalization for acute myocardial infarction, stroke, transient ischemic attack, or cardiovascular death. There is also a suggestion that metformin may have effects beyond those relating to lowering of blood sugar. The goal of this review is to assess the effects of metformin in coronary artery disease (CAD), but more importantly, its effects on disease states other than CAD.
Collapse
Affiliation(s)
- Khalid Sawalha
- Division of Cardiovascular Disease, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nitesh Gautam
- Division of Cardiovascular Disease, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kalaivani Sivakumar
- Division of Cardiovascular Disease, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Hakan Paydak
- Division of Cardiovascular Disease, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jawaher L Mehta
- Division of Cardiovascular Disease, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
3
|
Siam NH, Snigdha NN, Tabasumma N, Parvin I. Diabetes Mellitus and Cardiovascular Disease: Exploring Epidemiology, Pathophysiology, and Treatment Strategies. Rev Cardiovasc Med 2024; 25:436. [PMID: 39742220 PMCID: PMC11683709 DOI: 10.31083/j.rcm2512436] [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: 07/14/2024] [Revised: 08/27/2024] [Accepted: 09/02/2024] [Indexed: 01/03/2025] Open
Abstract
Diabetes mellitus (DM) affects 537 million people as of 2021, and is projected to rise to 783 million by 2045. This positions DM as the ninth leading cause of death globally. Among DM patients, cardiovascular disease (CVD) is the primary cause of morbidity and mortality. Notably, the prevalence rates of CVD is alarmingly high among diabetic individuals, particularly in North America and the Caribbean (46.0%), and Southeast Asia (42.5%). The predominant form of CVD among diabetic patients is coronary artery disease (CAD), accounting for 29.4% of cases. The pathophysiology of DM is complex, involving insulin resistance, β-cell dysfunction, and associated cardiovascular complications including diabetic cardiomyopathy (DCM) and cardiovascular autonomic neuropathy (CAN). These conditions exacerbate CVD risks underscoring the importance of managing key risk factors including hypertension, dyslipidemia, obesity, and genetic predisposition. Understanding the genetic networks and molecular processes that link diabetes and cardiovascular disease can lead to new diagnostics and therapeutic interventions. Imeglimin, a novel mitochondrial bioenergetic enhancer, represents a promising medication for diabetes with the potential to address both insulin resistance and secretion difficulties. Effective diabetes management through oral hypoglycemic agents (OHAs) can protect the cardiovascular system. Additionally, certain antihypertensive medications can significantly reduce the risk of diabetes-related CVD. Additionally, lifestyle changes, including diet and exercise are vital in managing diabesity and reducing CVD risks. These interventions, along with emerging therapeutic agents and ongoing clinical trials, offer hope for improved patient outcomes and long-term DM remission. This study highlights the urgent need for management strategies to address the overlapping epidemics of DM and CVD. By elucidating the underlying mechanisms and risk factors, this study aims to guide future perspectives and enhance understanding of the pathogenesis of CVD complications in patients with DM, thereby guiding more effective treatment strategies.
Collapse
Affiliation(s)
- Nawfal Hasan Siam
- Department of Pharmacy, School of Pharmacy and Public Health, Independent University, Bangladesh (IUB), 1229 Dhaka, Bangladesh
| | - Nayla Nuren Snigdha
- Department of Pharmacy, School of Pharmacy and Public Health, Independent University, Bangladesh (IUB), 1229 Dhaka, Bangladesh
| | - Noushin Tabasumma
- Department of Pharmacy, School of Pharmacy and Public Health, Independent University, Bangladesh (IUB), 1229 Dhaka, Bangladesh
| | - Irin Parvin
- Department of Biomedical Science, School of Health and Life Sciences, Teesside University, TS1 3BX Middlesbrough, UK
| |
Collapse
|
4
|
Sharrack N, Knott KD, Gulsin GS, Kotecha T, Brown LAE, Yeo JL, Porcari A, Adam RD, Thirunavukarasu S, Chowdhary A, Levelt E, Moon JC, McCann GP, Fontana M, Kellman P, Munyombwe T, Gale CP, Buckley DL, Greenwood JP, Swoboda PP, Plein S. Metformin associates with higher myocardial perfusion reserve and survival in type 2 diabetes mellitus patients. Sci Rep 2024; 14:27280. [PMID: 39516499 PMCID: PMC11549305 DOI: 10.1038/s41598-024-77280-2] [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] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Metformin is an antihyperglycemic used to treat type 2 diabetes mellitus (T2DM). Patients with T2DM are at increased risk of cardiovascular disease. We explored the association between metformin use and cardiovascular magnetic resonance (CMR) derived stress myocardial blood flow (MBF), myocardial perfusion reserve (MPR) and major adverse cardiovascular events (MACE; all cause death, MI, stroke, heart failure hospitalisation and coronary revascularisation) in patients with T2DM. Multi-centre study of patients with T2DM, and healthy controls, underwent quantitative myocardial perfusion CMR using an artificial intelligence supported process. Multivariable regression analysis, and cox proportional hazard models of propensity score weighted patients quantified associations between metformin use, MBF, MPR, all cause death and MACE. Analysis included 572 patients with T2DM (68% prescribed metformin) with median follow-up 851 days (IQR 935 - 765). Metformin use was associated with an increase of MPR of 0.12 [0.08-0.40], p = 0.004. There were 82 MACE events (14.3%) including 25 (4.4%) deaths of which 16 were in those not prescribed metformin (8.7%), compared to 9 in patients prescribed metformin (2.3%): adjusted hazard ratio 0.24 (95% CI 0.08-0.70, p = 0.009). MACE events were similar between groups. This multicentre, inverse probability weighting propensity score analysis study showed that in patients with T2DM, metformin use is associated with higher MPR and improved all cause survival.
Collapse
Affiliation(s)
- Noor Sharrack
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK.
| | - Kristopher D Knott
- Barts Heart Centre, St Bartholomew's Hospital, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
| | - Gaurav S Gulsin
- Department of Cardiovascular Sciences, Cardiovascular Biomedical Research Centre, University of Leicester and the NIHR Leicester, University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester, UK
| | - Tushar Kotecha
- Institute of Cardiovascular Science, University College London, London, UK
- Division of Medicine, National Amyloidosis Centre, University College London, Royal Free Campus, London, UK
| | - Louise A E Brown
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Jian L Yeo
- Department of Cardiovascular Sciences, Cardiovascular Biomedical Research Centre, University of Leicester and the NIHR Leicester, University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester, UK
| | - Aldostefano Porcari
- Division of Medicine, National Amyloidosis Centre, University College London, Royal Free Campus, London, UK
- Centre for Diagnosis and Treatment of Cardiomyopathies, Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - Robert D Adam
- Barts Heart Centre, St Bartholomew's Hospital, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Sharmaine Thirunavukarasu
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Amrit Chowdhary
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Eylem Levelt
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - James C Moon
- Barts Heart Centre, St Bartholomew's Hospital, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
| | - Gerry P McCann
- Department of Cardiovascular Sciences, Cardiovascular Biomedical Research Centre, University of Leicester and the NIHR Leicester, University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester, UK
| | - Marianna Fontana
- Institute of Cardiovascular Science, University College London, London, UK
- Division of Medicine, National Amyloidosis Centre, University College London, Royal Free Campus, London, UK
| | - Peter Kellman
- Department of Health and Human Services, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Theresa Munyombwe
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
| | - Chris P Gale
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
- Department of Cardiology, Leeds Teaching Hosptislas NHS Trust, Leeds, UK
| | - David L Buckley
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - John P Greenwood
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Peter P Swoboda
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Sven Plein
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
5
|
Tran J, Parekh S, Rockcole J, Wilson D, Parmar MS. Repurposing antidiabetic drugs for Alzheimer's disease: A review of preclinical and clinical evidence and overcoming challenges. Life Sci 2024; 355:123001. [PMID: 39173996 DOI: 10.1016/j.lfs.2024.123001] [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: 07/05/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Repurposing antidiabetic drugs for the treatment of Alzheimer's disease (AD) has emerged as a promising therapeutic strategy. This review examines the potential of repurposing antidiabetic drugs for AD treatment, focusing on preclinical evidence, clinical trials, and observational studies. In addition, the review aims to explore challenges and opportunities in repurposing antidiabetic drugs for AD, emphasizing the importance of well-designed clinical trials that consider patient selection criteria, refined outcome measures, adverse effects, and combination therapies to enhance therapeutic efficacy. Preclinical evidence suggests that glucagon-like peptide-1 (GLP-1) analogs, dipeptidyl peptidase-4 (DPP4) inhibitors, metformin, thiazolidinediones, and sodium-glucose co-transporter-2 (SGLT2) inhibitors exhibit neuroprotective effects in AD preclinical models. In preclinical studies, antidiabetic drugs have demonstrated neuroprotective effects by reducing amyloid beta (Aβ) plaques, tau hyperphosphorylation, neuroinflammation, and cognitive impairment. Antidiabetic drug classes, notably GLP-1 analogs and SGLT2 inhibitors, and a reduced risk of dementia in patients with diabetes mellitus. While the evidence for DPP4 inhibitors is mixed, some studies suggest a potential protective effect. On the other hand, alpha-glucosidase inhibitors (AGIs) and sulfonylureas may potentially increase the risk, especially in those experiencing recurrent hypoglycemic events. Repurposing antidiabetic drugs for AD is a promising therapeutic strategy, but challenges such as disease heterogeneity, limited biomarkers, and benefits versus risk evaluation need to be addressed. Ongoing clinical trials in mild cognitive impairment (MCI) and early AD patients without diabetes will be crucial in determining the clinical efficacy and safety of the antidiabetic drugs, paving the way for potential treatments for AD.
Collapse
Affiliation(s)
- Jacky Tran
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Sneh Parekh
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Julia Rockcole
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Danielle Wilson
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Mayur S Parmar
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA.
| |
Collapse
|
6
|
Cimellaro A, Cavallo M, Mungo M, Suraci E, Spagnolo F, Addesi D, Pintaudi M, Pintaudi C. Cardiovascular Effectiveness and Safety of Antidiabetic Drugs in Patients with Type 2 Diabetes and Peripheral Artery Disease: Systematic Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1542. [PMID: 39336583 PMCID: PMC11434261 DOI: 10.3390/medicina60091542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Peripheral artery disease (PAD) is an atherosclerotic condition commonly complicating type 2 diabetes (T2D), leading to poor quality of life and increased risk of major adverse lower-limb (MALE) and cardiovascular (CV) events (MACE). Therapeutic management of PAD in T2D patients is much more arduous, often due to bilateral, multi-vessel, and distal vascular involvement, in addition to increased systemic polyvascular atherosclerotic burden. On the other hand, the pathophysiological link between PAD and T2D is very complex, involving mechanisms such as endothelial dysfunction and increased subclinical inflammation in addition to chronic hyperglycemia. Therefore, the clinical approach should not ignore vascular protection with the aim of reducing limb and overall CV events besides a mere glucose-lowering effect. However, the choice of the best medications in this setting is challenging due to low-grade evidence or lacking targeted studies in PAD patients. The present review highlighted the strong relationship between T2D and PAD, focusing on the best treatment strategy to reduce CV risk and prevent PAD occurrence and worsening in patients with T2D. The Medline databases were searched for studies including T2D and PAD up to June 2024 and reporting the CV effectiveness and safety of the most used glucose-lowering agents, with no restriction on PAD definition, study design, or country. The main outcomes considered were MACE-including nonfatal acute myocardial infarction, nonfatal stroke, and CV death-and MALE-defined as lower-limb complications, amputations, or need for revascularization. To the best of our current knowledge, GLP-1 receptor agonists and SGLT2 inhibitors represent the best choice to reduce CV risk in T2D and PAD settings, but a personalized approach should be considered. GLP-1 receptor agonists should be preferred in subjects with prevalent atherosclerotic burden and a history of previous MALE, while SGLT2 inhibitors should be used in those with heart failure if overall CV benefits outweigh the risk of lower-limb complications.
Collapse
Affiliation(s)
- Antonio Cimellaro
- Internal Medicine Unit, Department of Medicine Specialties, “Pugliese-Ciaccio” Hospital of Catanzaro, Azienda Ospedaliero-Universitaria Renato Dulbecco, Via Pio X n.83, 88100 Catanzaro, Italy; (M.C.); (E.S.); (F.S.); (D.A.); (C.P.)
| | - Michela Cavallo
- Internal Medicine Unit, Department of Medicine Specialties, “Pugliese-Ciaccio” Hospital of Catanzaro, Azienda Ospedaliero-Universitaria Renato Dulbecco, Via Pio X n.83, 88100 Catanzaro, Italy; (M.C.); (E.S.); (F.S.); (D.A.); (C.P.)
| | - Marialaura Mungo
- Internal Medicine Unit, Department of Medical and Surgical Sciences, ‘Magna Græcia’ University of Catanzaro, Viale Europa, Località Germaneto, 88100 Catanzaro, Italy;
| | - Edoardo Suraci
- Internal Medicine Unit, Department of Medicine Specialties, “Pugliese-Ciaccio” Hospital of Catanzaro, Azienda Ospedaliero-Universitaria Renato Dulbecco, Via Pio X n.83, 88100 Catanzaro, Italy; (M.C.); (E.S.); (F.S.); (D.A.); (C.P.)
| | - Francesco Spagnolo
- Internal Medicine Unit, Department of Medicine Specialties, “Pugliese-Ciaccio” Hospital of Catanzaro, Azienda Ospedaliero-Universitaria Renato Dulbecco, Via Pio X n.83, 88100 Catanzaro, Italy; (M.C.); (E.S.); (F.S.); (D.A.); (C.P.)
| | - Desirée Addesi
- Internal Medicine Unit, Department of Medicine Specialties, “Pugliese-Ciaccio” Hospital of Catanzaro, Azienda Ospedaliero-Universitaria Renato Dulbecco, Via Pio X n.83, 88100 Catanzaro, Italy; (M.C.); (E.S.); (F.S.); (D.A.); (C.P.)
| | - Medea Pintaudi
- Unit of Plastic Surgery, Department of Surgery, Azienda Ospedaliero-Universitaria “Gaetano Martino”, 98124 Messina, Italy;
| | - Carmelo Pintaudi
- Internal Medicine Unit, Department of Medicine Specialties, “Pugliese-Ciaccio” Hospital of Catanzaro, Azienda Ospedaliero-Universitaria Renato Dulbecco, Via Pio X n.83, 88100 Catanzaro, Italy; (M.C.); (E.S.); (F.S.); (D.A.); (C.P.)
| |
Collapse
|
7
|
Sirtori CR, Castiglione S, Pavanello C. METFORMIN: FROM DIABETES TO CANCER TO PROLONGATION OF LIFE. Pharmacol Res 2024; 208:107367. [PMID: 39191336 DOI: 10.1016/j.phrs.2024.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
The metformin molecule dates back to over a century, but its clinical use started in the '50s. Since then, its use in diabetics has grown constantly, with over 150 million users today. The therapeutic profile also expanded, with improved understanding of novel mechanisms. Metformin has a major activity on insulin resistance, by acting on the insulin receptors and mitochondria, most likely by activation of the adenosine monophosphate-activated kinase. These and associated mechanisms lead to significant lipid lowering and body weight loss. An anti-cancer action has come up in recent years, with mechanisms partly dependent on the mitochondrial activity and also on phosphatidylinositol 3-kinase resistance occurring in some malignant tumors. The potential of metformin to raise life-length is the object of large ongoing studies and of several basic and clinical investigations. The present review article will attempt to investigate the basic mechanisms behind these diverse activities and the potential clinical benefits. Metformin may act on transcriptional activity by histone modification, DNA methylation and miRNAs. An activity on age-associated inflammation (inflammaging) may occur via activation of the nuclear factor erythroid 2 related factor and changes in gut microbiota. A senolytic activity, leading to reduction of cells with the senescent associated secretory phenotype, may be crucial in lifespan prolongation as well as in ancillary properties in age-associated diseases, such as Parkinson's disease. Telomere prolongation may be related to the activity on mitochondrial respiratory factor 1 and on peroxisome gamma proliferator coactivator 1-alpha. Very recent observations on the potential to act on the most severe neurological disorders, such as amyotrophic lateral sclerosis and frontotemporal dementia, have raised considerable hope.
Collapse
Affiliation(s)
- Cesare R Sirtori
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Sofia Castiglione
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Pavanello
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
8
|
Stroope C, Nettersheim FS, Coon B, Finney AC, Schwartz MA, Ley K, Rom O, Yurdagul A. Dysregulated cellular metabolism in atherosclerosis: mediators and therapeutic opportunities. Nat Metab 2024; 6:617-638. [PMID: 38532071 PMCID: PMC11055680 DOI: 10.1038/s42255-024-01015-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
Accumulating evidence over the past decades has revealed an intricate relationship between dysregulation of cellular metabolism and the progression of atherosclerotic cardiovascular disease. However, an integrated understanding of dysregulated cellular metabolism in atherosclerotic cardiovascular disease and its potential value as a therapeutic target is missing. In this Review, we (1) summarize recent advances concerning the role of metabolic dysregulation during atherosclerosis progression in lesional cells, including endothelial cells, vascular smooth muscle cells, macrophages and T cells; (2) explore the complexity of metabolic cross-talk between these lesional cells; (3) highlight emerging technologies that promise to illuminate unknown aspects of metabolism in atherosclerosis; and (4) suggest strategies for targeting these underexplored metabolic alterations to mitigate atherosclerosis progression and stabilize rupture-prone atheromas with a potential new generation of cardiovascular therapeutics.
Collapse
Affiliation(s)
- Chad Stroope
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Felix Sebastian Nettersheim
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Brian Coon
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Cardiovascular Biology Research Program, OMRF, Oklahoma City, OK, USA
- Department of Cell Biology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
- Immunology Center of Georgia (IMMCG), Augusta University Immunology Center of Georgia, Augusta, GA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
9
|
Scurt FG, Ganz MJ, Herzog C, Bose K, Mertens PR, Chatzikyrkou C. Association of metabolic syndrome and chronic kidney disease. Obes Rev 2024; 25:e13649. [PMID: 37783465 DOI: 10.1111/obr.13649] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/15/2023] [Indexed: 10/04/2023]
Abstract
The prevalence of kidney disease is increasing rapidly worldwide, reflecting rising rates of obesity, diabetes, and associated metabolic syndrome (MetS). Chronic kidney disease and related comorbidities such as obesity, diabetes, and hypertension place a significant financial burden on healthcare systems. Despite the widespread use of RAAS inhibitors, intensive blood pressure and glycemic control, and newer therapeutic options consisting of sodium/glucose cotransporter-2 (SGLT-2) inhibitors or glucagon-like peptide-1 (GLP-1) receptor agonists, a significant risk of progression to end-stage renal disease remains in the high-risk obese and diabetic population. The MetS is a cluster of cardiovascular risk factors that adversely affect the development and progression of chronic kidney failure. According to the criteria of the World Health Organization, it is defined by visceral adiposity, impaired glucose tolerance or insulin resistance, atherogenic dyslipidemia, raised blood pressure, and microalbuminuria with a albumin-to-creatinine ratio ≥30 mg/g. At molecular level MetS is marked by a proinflammatory state and increased oxidative stress leading to various pathophysiological changes causing endothelial dysfunction and a hypercoagulable state. Because the kidney is a highly vascularized organ, it is especially susceptible for those microvascular changes. Therefore, the MetS and its individual components are associated with the premature development, acceleration, and progression of chronic kidney disease. Therefore, it is becoming increasingly important to elucidate the underlying mechanisms of MetS-associated chronic kidney disease in order to develop new strategies for preventing and slowing the progression of renal disease. In this review, we will elucidate (i) the renal structural, hemodynamic, and metabolic changes that occur in obesity and obesity-related kidney injury; (ii) the clinicopathological characteristics of obesity-related kidney injury, primarily focusing on obesity-associated glomerulopathy; (iii) the potential additional factors or predisposing factors that may turn patients more susceptible to renal structural or functional compensatory failure and subsequent injury.
Collapse
Affiliation(s)
- Florian G Scurt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, Medical Faculty, Otto-von Guericke University Magdeburg, Magdeburg, Germany
| | - Maximilian J Ganz
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, Medical Faculty, Otto-von Guericke University Magdeburg, Magdeburg, Germany
| | - Carolin Herzog
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, Medical Faculty, Otto-von Guericke University Magdeburg, Magdeburg, Germany
| | - Katrin Bose
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Magdeburg, Germany
| | - Peter R Mertens
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, Medical Faculty, Otto-von Guericke University Magdeburg, Magdeburg, Germany
| | | |
Collapse
|
10
|
Cannarella R, Condorelli RA, Leanza C, Garofalo V, Aversa A, Papa G, Calogero AE, La Vignera S. Dapagliflozin improves erectile dysfunction in patients with type 2 diabetes mellitus: An open-label, non-randomized pilot study. Diabet Med 2024; 41:e15217. [PMID: 37669131 DOI: 10.1111/dme.15217] [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: 06/15/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/07/2023]
Abstract
INTRODUCTION The role of dapagliflozin on erectile dysfunction (ED), a condition widely affecting patients with type 2 diabetes mellitus (T2DM), has not yet been studied. AIM The aim of the study was to evaluate the effects of dapagliflozin alone or in combination with tadalafil on ED in patients with T2DM. METHODS This was an open-label, non-randomized pilot study involving 30 Caucasian male patients with T2DM and severe ED. They were equally divided into three groups, assigned to treatment with tadalafil 5 mg/day (Group 1), tadalafil 5 mg/day plus dapagliflozin 10 mg/day (Group 2) and dapagliflozin 10 mg/day (Group 3) for 3 months. The presence and the severity of ED were evaluated at enrolment and after treatment, by the International Index of Erectile Function 5-item (IIEF-5) questionnaire and the dynamic penile echo colour Doppler ultrasound (PCDU) examination. RESULTS At the end of treatment, the three groups showed a significant improvement in IIEF-5 score, by 294%, 375% and 197%, in Groups 1, 2 and 3, respectively. PCDU evaluation showed a significant increase in peak systolic velocity by 178.9%, 339% and 153%; acceleration time was significantly shortened in Group 2 (-26.2%) and was significantly lower than in Group 1 and 3 (-7.2% and -6.6%), while no significant difference was found in end-diastolic velocity after treatment. The greatest rates of improvement were observed in Group 2 for all the end points. CONCLUSIONS Dapagliflozin improves ED in patients with T2DM and enhances the efficacy of tadalafil. Further studies are needed to confirm our results explain the mechanism(s) by which dapagliflozin exerts its effects on ED.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Claudia Leanza
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Vincenzo Garofalo
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Antonio Aversa
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Giuseppe Papa
- Unit of Metabolic and Endocrine Disease, Centro Catanese di Medicina e Chirurgia Clinic, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
11
|
Xu CM, Karbasiafshar C, Brinck‐Teixeira R, Broadwin M, Sellke FW, Abid MR. Diabetic state of human coronary artery endothelial cells results in altered effects of bone mesenchymal stem cell-derived extracellular vesicles. Physiol Rep 2023; 11:e15866. [PMID: 38114067 PMCID: PMC10730301 DOI: 10.14814/phy2.15866] [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: 07/10/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 12/21/2023] Open
Abstract
Human bone mesenchymal stem cell-derived extracellular vesicles (HBMSC-EV) have been used successfully in animal models of myocardial ischemia, yet have dampened effects in metabolic syndrome through unknown mechanisms. This study demonstrates the basal differences between non-diabetic human coronary artery endothelial cells (HCAEC) and diabetic HCAEC (DM-HCAEC), and how these cells respond to the treatment of HBMSC-EV. HCAEC and DM-HCAEC were treated with HBMSC-EV for 6 h. Proteomics, western blot analysis, and tube formation assays were performed. Key metabolic, growth, and stress/starvation cellular responses were significantly altered in DM-HCAEC in comparison to that of HCAEC at baseline. Proteomics demonstrated increased phosphorus metabolic process and immune pathways and decreased RNA processing and biosynthetic pathways in DM-HCAEC. Similar to previous in vivo findings, HCAEC responded to the HBMSC-EV with regenerative and anti-inflammatory effects through the upregulation of multiple RNA pathways and downregulation of immune cell activation pathways. In contrast, DM-HCAEC had a significantly diminished response to HBMSC-EV, likely due to the baseline abnormalities in DM-HCAEC. To achieve the full benefits of HBMSC-EV and for a successful transition of this potential therapeutic agent to clinical studies, the abnormalities found in DM-HCAEC will need to be further studied.
Collapse
Affiliation(s)
- Cynthia M. Xu
- Cardiovascular Research Center, Rhode Island HospitalProvidenceRhode IslandUSA
- Division of Cardiothoracic SurgeryAlpert Medical School of Brown University and Rhode Island HospitalProvidenceRhode IslandUSA
| | | | - Rayane Brinck‐Teixeira
- Cardiovascular Research Center, Rhode Island HospitalProvidenceRhode IslandUSA
- Division of Cardiothoracic SurgeryAlpert Medical School of Brown University and Rhode Island HospitalProvidenceRhode IslandUSA
| | - Mark Broadwin
- Cardiovascular Research Center, Rhode Island HospitalProvidenceRhode IslandUSA
- Division of Cardiothoracic SurgeryAlpert Medical School of Brown University and Rhode Island HospitalProvidenceRhode IslandUSA
| | - Frank W. Sellke
- Cardiovascular Research Center, Rhode Island HospitalProvidenceRhode IslandUSA
- Division of Cardiothoracic SurgeryAlpert Medical School of Brown University and Rhode Island HospitalProvidenceRhode IslandUSA
| | - M. Ruhul Abid
- Cardiovascular Research Center, Rhode Island HospitalProvidenceRhode IslandUSA
- Division of Cardiothoracic SurgeryAlpert Medical School of Brown University and Rhode Island HospitalProvidenceRhode IslandUSA
| |
Collapse
|
12
|
David SR, Lai PPN, Chellian J, Chakravarthi S, Rajabalaya R. Influence of rutin and its combination with metformin on vascular functions in type 1 diabetes. Sci Rep 2023; 13:12423. [PMID: 37528147 PMCID: PMC10394083 DOI: 10.1038/s41598-023-39442-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023] Open
Abstract
The present work examined the effect of oral administration of rutin and its combination with metformin, an antidiabetic drug on blood glucose, total cholesterol and triglycerides level and vascular function in streptozotocin (STZ) -induced diabetic rats. Male Sprague Dawley rats were rendered diabetic by a single intraperitoneal injection of STZ (50 mg/kg). Rutin and metformin were orally administered to diabetic rats at a dose of 100 mg/kg and 300 mg/kg body weight/day, respectively, for 4 weeks. Plasma analysis was conducted to determine changes in the plasma glucose and lipid levels. Rat aortic ring reactivity in response to endothelium-dependent (acetylcholine, ACh) and endothelium-independent (sodium nitroprusside, SNP) relaxants, and to the α1-adrenergic agonist phenylephrine (PE) were recorded. Histology of pancreas, liver and kidney were evaluated. In results, rutin and metformin alone and in combination has led to significant improvements in blood glucose, cholesterol and triglyceride levels compared to diabetic group. Diabetic aortic rings showed significantly greater contraction in response to PE, and less relaxation in response to ACh and SNP. Treatment with rutin and metformin in combination significantly reduced PE-induced contraction and increased ACh-induced and SNP-induced relaxation in diabetes when compared to rutin or metformin alone. Significant histological improvements were seen with combination therapy. In conclusion, rutin and metformin combination therapy has the most potentiality for restoring blood glucose and lipid level as well as vascular function.
Collapse
Affiliation(s)
- Sheba R David
- School of Pharmacy, University of Wyoming, Laramie, WY, 82071, USA.
| | - Penny Pei Ni Lai
- School of Medicine, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Jestin Chellian
- School of Pharmacy, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Srikumar Chakravarthi
- SEGi University and Colleges, No. 9, Jalan Teknologi, Taman Sains Selangor, 47810, Kota Damansara, Selangor, Malaysia
| | - Rajan Rajabalaya
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Bandar Seri Begawan, BE1410, Brunei Darussalam.
| |
Collapse
|
13
|
Volino-Souza M, Oliveira GVD, Pinheiro VDS, Conte-Junior CA, Alvares TDS. The effect of dietary nitrate on macro- and microvascular function: A systematic review. Crit Rev Food Sci Nutr 2022; 64:1225-1236. [PMID: 36062809 DOI: 10.1080/10408398.2022.2113989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Previous studies have investigated the impact of dietary nitrate on vascular function due to the association between dietary nitrate ingestion and improvement in nitric oxide (NO) bioavailability. Considering that NO can present different effects through vascular beds (macro- vs. microvasculature) due to the specific characteristic (function and morphology) that each vessel exhibits, it is crucial to investigate the effect of dietary nitrate ingestion on the macro- and microvascular function to understand the effect of nitrate on vascular function. For this reason, this review aimed to evaluate the impact of dietary nitrate on macro- and microvascular function in humans. A total of 29 studies were included in the systematic review, of which 19 studies evaluated the effect of nitrate supplementation on macrovascular function, eight studies evaluated the effect on microvascular function, and two studies evaluated the impact on both macro- and microvascular function. The literature suggests that dietary nitrate ingestion seems to improve the vascular function in macrovasculature, whereas microvascular function appears to be modest. Future studies investigating the effect of nitrate ingestion on vascular function should focus on measuring macro- and microvascular function whenever possible so that the impact of nitrate-rich foods on vascular segments could be better understood.
Collapse
Affiliation(s)
- Mônica Volino-Souza
- Nutrition and Exercise Metabolism Research Group, Multidisciplinary Center UFRJ-Macaé, Federal University of Rio de Janeiro, Macaé, Rio de Janeiro, Brazil
- Postgraduate Program in Food Science, Chemistry Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | - Gustavo Vieira de Oliveira
- Nutrition and Exercise Metabolism Research Group, Multidisciplinary Center UFRJ-Macaé, Federal University of Rio de Janeiro, Macaé, Rio de Janeiro, Brazil
- Nanotechnology Network, Carlos Chagas Filho Research Support Foundation of the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, RJ, Brazil
| | - Vivian Dos Santos Pinheiro
- Nutrition and Exercise Metabolism Research Group, Multidisciplinary Center UFRJ-Macaé, Federal University of Rio de Janeiro, Macaé, Rio de Janeiro, Brazil
- Postgraduate Program in Food Science, Chemistry Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | - Carlos Adam Conte-Junior
- Postgraduate Program in Food Science, Chemistry Institute, Federal University of Rio de Janeiro, RJ, Brazil
- Nanotechnology Network, Carlos Chagas Filho Research Support Foundation of the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, RJ, Brazil
| | - Thiago da Silveira Alvares
- Nutrition and Exercise Metabolism Research Group, Multidisciplinary Center UFRJ-Macaé, Federal University of Rio de Janeiro, Macaé, Rio de Janeiro, Brazil
- Food and Nutrition Institute, Multidisciplinary Center UFRJ-Macaé, Federal University of Rio de Janeiro, Macaé, Brazil
| |
Collapse
|
14
|
Triggle CR, Mohammed I, Bshesh K, Marei I, Ye K, Ding H, MacDonald R, Hollenberg MD, Hill MA. Metformin: Is it a drug for all reasons and diseases? Metabolism 2022; 133:155223. [PMID: 35640743 DOI: 10.1016/j.metabol.2022.155223] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
Metformin was first used to treat type 2 diabetes in the late 1950s and in 2022 remains the first-choice drug used daily by approximately 150 million people. An accumulation of positive pre-clinical and clinical data has stimulated interest in re-purposing metformin to treat a variety of diseases including COVID-19. In polycystic ovary syndrome metformin improves insulin sensitivity. In type 1 diabetes metformin may help reduce the insulin dose. Meta-analysis and data from pre-clinical and clinical studies link metformin to a reduction in the incidence of cancer. Clinical trials, including MILES (Metformin In Longevity Study), and TAME (Targeting Aging with Metformin), have been designed to determine if metformin can offset aging and extend lifespan. Pre-clinical and clinical data suggest that metformin, via suppression of pro-inflammatory pathways, protection of mitochondria and vascular function, and direct actions on neuronal stem cells, may protect against neurodegenerative diseases. Metformin has also been studied for its anti-bacterial, -viral, -malaria efficacy. Collectively, these data raise the question: Is metformin a drug for all diseases? It remains unclear as to whether all of these putative beneficial effects are secondary to its actions as an anti-hyperglycemic and insulin-sensitizing drug, or result from other cellular actions, including inhibition of mTOR (mammalian target for rapamycin), or direct anti-viral actions. Clarification is also sought as to whether data from ex vivo studies based on the use of high concentrations of metformin can be translated into clinical benefits, or whether they reflect a 'Paracelsus' effect. The environmental impact of metformin, a drug with no known metabolites, is another emerging issue that has been linked to endocrine disruption in fish, and extensive use in T2D has also raised concerns over effects on human reproduction. The objectives for this review are to: 1) evaluate the putative mechanism(s) of action of metformin; 2) analyze the controversial evidence for metformin's effectiveness in the treatment of diseases other than type 2 diabetes; 3) assess the reproducibility of the data, and finally 4) reach an informed conclusion as to whether metformin is a drug for all diseases and reasons. We conclude that the primary clinical benefits of metformin result from its insulin-sensitizing and antihyperglycaemic effects that secondarily contribute to a reduced risk of a number of diseases and thereby enhancing healthspan. However, benefits like improving vascular endothelial function that are independent of effects on glucose homeostasis add to metformin's therapeutic actions.
Collapse
Affiliation(s)
- Chris R Triggle
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar; Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar.
| | - Ibrahim Mohammed
- Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Khalifa Bshesh
- Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Isra Marei
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Kevin Ye
- Department of Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Hong Ding
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar; Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Ross MacDonald
- Distribution eLibrary, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, a Cumming School of Medicine, University of Calgary, T2N 4N1, Canada
| | - Michael A Hill
- Dalton Cardiovascular Research Center, Department of Medical Pharmacology & Physiology, School of Medicine, University of Missouri, Columbia 65211, MO, USA
| |
Collapse
|
15
|
Zhang Y, Zhang H, Li S, Huang K, Jiang L, Wang Y. Metformin Alleviates LPS-Induced Acute Lung Injury by Regulating the SIRT1/NF-κB/NLRP3 Pathway and Inhibiting Endothelial Cell Pyroptosis. Front Pharmacol 2022; 13:801337. [PMID: 35910360 PMCID: PMC9334876 DOI: 10.3389/fphar.2022.801337] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS), a devastating complication of numerous conditions, is often associated with high mortality. It is well known that endothelial cell (EC) damage and inflammation are vital processes in the pathogenesis of ARDS. Nevertheless, the mechanisms of EC damage are largely unknown. In the present study, we investigated the role of pyroptosis in the initiation of ARDS and demonstrated that endothelial pyroptosis might play a pivotal role in the pathophysiology of ARDS. Metformin, an antidiabetic drug, exhibited a protective effect in lipopolysaccharide (LPS)-induced lung injury, and we hypothesized that metformin alleviated LPS-induced lung injury via inhibiting ECs pyroptosis. In vivo, male ICR mice were intratracheally injected with LPS, and metformin was previously administered intraperitoneally. Morphological properties of lung tissues were detected. We showed that metformin inhibited NLRP3 inflammasome activation and NLRP3-stimulated pyroptosis induction, as shown by decreased levels of cleaved caspase-1, N-terminal fragment of GSDMD, and protein contents of IL-1β in lung tissues of mice exposed to LPS. LPS-induced expression of vascular adhesion molecules was also reduced after the treatment with metformin. In vitro, exposure of pulmonary ECs to LPS resulted in increased expression of NLRP3 and pyroptosis-associated indicators. By inhibiting the expression of NLRP3 with NLRP3 inhibitor MCC950, pyroptosis-related markers and vascular adhesion molecules were ameliorated. Moreover, metformin treatment significantly inhibited the NF-κB signaling pathway and increased the expression of sirtuin 1 (SIRT1) both in LPS-stimulated lung tissues and pulmonary ECs. Administration of the selective SIRT1 inhibitor nicotinamide significantly reversed the protective effect of metformin against endothelial pyroptosis and lung injury in LPS-treated ECs and LPS-induced acute lung injury (ALI). Thus, these findings demonstrated that metformin alleviated LPS-induced ALI by inhibiting NF-κB-NLRP3–mediated ECs pyroptosis, possibly by upregulating the expression of SIRT1.
Collapse
Affiliation(s)
| | | | | | | | - Lai Jiang
- *Correspondence: Yan Wang, ; Lai Jiang,
| | - Yan Wang
- *Correspondence: Yan Wang, ; Lai Jiang,
| |
Collapse
|
16
|
Jahn LA, Hartline L, Liu Z, Barrett EJ. Metformin improves skeletal muscle microvascular insulin resistance in metabolic syndrome. Am J Physiol Endocrinol Metab 2022; 322:E173-E180. [PMID: 34957859 PMCID: PMC8858665 DOI: 10.1152/ajpendo.00287.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 11/22/2022]
Abstract
Microvascular insulin resistance is present in metabolic syndrome and may contribute to increased cardiovascular disease risk and the impaired metabolic response to insulin observed. Metformin improves metabolic insulin resistance in humans. Its effects on macro and microvascular insulin resistance have not been defined. Eleven subjects with nondiabetic metabolic syndrome were studied four times (before and after 12 wk of treatment with placebo or metformin) using a crossover design, with an 8-wk washout interval between treatments. On each occasion, we measured three indices of large artery function [pulse wave velocity (PWV), radial pulse wave separation analysis (PWSA), brachial artery endothelial function (flow-mediated dilation-FMD)] as well as muscle microvascular perfusion [contrast-enhanced ultrasound (CEU)] before and at 120 min into a 150 min, 1 mU/min/kg euglycemic insulin clamp. Metformin decreased body mass index (BMI), fat weight, and % body fat (P < 0.05, each), however, placebo had no effect. Metformin (not placebo) improved metabolic insulin sensitivity, (clamp glucose infusion rate, P < 0.01), PWV, and FMD after insulin were unaffected by metformin treatment. PWSA improved with insulin only after metformin P < 0.01). Insulin decreased muscle microvascular blood volume measured by contrast ultrasound both before and after placebo and before metformin (P < 0.02 for each) but not after metformin. Short-term metformin treatment improves both metabolic and muscle microvascular response to insulin. Metformin's effect on microvascular insulin responsiveness may contribute to its beneficial metabolic effects. Metformin did not improve aortic stiffness or brachial artery endothelial function, but enhanced radial pulse wave properties consistent with relaxation of smaller arterioles.NEW & NOTEWORTHY Metformin, a first-line treatment for type 2 diabetes, is often used in patients with insulin resistance and metabolic syndrome. Here, we provide the first evidence for metformin improving muscle microvascular insulin sensitivity in insulin-resistant humans. Simultaneously, metformin improved muscle glucose disposal, supporting a close relationship between insulin's microvascular and its metabolic actions in muscle. Whether enhanced microvascular insulin sensitivity contributes to metformin's ability to decrease microvascular complications in diabetes remains to be resolved.
Collapse
Affiliation(s)
- Linda A Jahn
- Division of Endocrinology, Department of Medicine, University of Virginia, School of Medicine, Charlottesville, Virginia
| | - Lee Hartline
- Division of Endocrinology, Department of Medicine, University of Virginia, School of Medicine, Charlottesville, Virginia
| | - Zhenqi Liu
- Division of Endocrinology, Department of Medicine, University of Virginia, School of Medicine, Charlottesville, Virginia
| | - Eugene J Barrett
- Division of Endocrinology, Department of Medicine, University of Virginia, School of Medicine, Charlottesville, Virginia
- Department of Pharmacology, University of Virginia, School of Medicine, Charlottesville, Virginia
| |
Collapse
|
17
|
Defeudis G, Mazzilli R, Tenuta M, Rossini G, Zamponi V, Olana S, Faggiano A, Pozzilli P, Isidori AM, Gianfrilli D. Erectile dysfunction and diabetes: A melting pot of circumstances and treatments. Diabetes Metab Res Rev 2022; 38:e3494. [PMID: 34514697 PMCID: PMC9286480 DOI: 10.1002/dmrr.3494] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 09/03/2021] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus (DM), a chronic metabolic disease characterised by elevated levels of blood glucose, is among the most common chronic diseases. The incidence and prevalence of DM have been increasing over the years. The complications of DM represent a serious health problem. The long-term complications include macroangiopathy, microangiopathy and neuropathy as well as sexual dysfunction (SD) in both men and women. Erectile dysfunction (ED) has been considered the most important SD in men with DM. The prevalence of ED is approximately 3.5-fold higher in men with DM than in those without DM. Common risk factors for the development of DM and its complications include sedentary lifestyle, overweight/obesity and increased caloric consumption. Although lifestyle changes may help improve sexual function, specific treatments are often needed. This study aims to review the definition and prevalence of ED in DM, the impact of DM complications and DM treatment on ED and, finally, the current and emerging therapies for ED in patients with DM.
Collapse
Affiliation(s)
- Giuseppe Defeudis
- Unit of Endocrinology and DiabetesDepartment of MedicineUniversity Campus Bio‐Medico di RomaRomeItaly
| | - Rossella Mazzilli
- Department of Clinical and Molecular MedicineSapienza University of RomeRomeItaly
| | - Marta Tenuta
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| | - Giovanni Rossini
- Unit of Endocrinology and DiabetesDepartment of MedicineUniversity Campus Bio‐Medico di RomaRomeItaly
| | - Virginia Zamponi
- Department of Clinical and Molecular MedicineSapienza University of RomeRomeItaly
| | - Soraya Olana
- Department of Clinical and Molecular MedicineSapienza University of RomeRomeItaly
| | - Antongiulio Faggiano
- Department of Clinical and Molecular MedicineSapienza University of RomeRomeItaly
| | - Paolo Pozzilli
- Unit of Endocrinology and DiabetesDepartment of MedicineUniversity Campus Bio‐Medico di RomaRomeItaly
| | - Andrea M. Isidori
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| | | |
Collapse
|
18
|
Hemorheological Parameters in Diabetic Patients: Role of Glucose Lowering Therapies. Metabolites 2021; 11:metabo11120806. [PMID: 34940563 PMCID: PMC8708076 DOI: 10.3390/metabo11120806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/26/2022] Open
Abstract
Diabetes mellitus influences several important hemorheological parameters including blood viscosity, erythrocyte aggregation and deformability. In the present study, 159 type-2 diabetic patients and 25 healthy controls were involved. Patient's age, body weight, body mass index (BMI), smoking habits, physical activity, history of cardiovascular diseases, current antidiabetic therapy and concomitant medication were recorded. Patients were grouped according to their antidiabetic treatment with insulin, or with one or more of the following antidiabetic drugs: metformin, sulfonylureas, acarbose, or no antidiabetic therapy. Hemorheological measurements (hematocrit, erythrocyte aggregation, plasma fibrinogen, whole blood and plasma viscosity), von Willebrand factor activity, and platelet aggregation measurements were performed. Platelet aggregation was investigated with the method of Born. Plasma viscosity and red blood cell aggregation were significatly higher in diabetes. No significant difference was found in hemorheological parameters between different antidiabetic regimens. Whole blood and plasma viscosity and red blood cell aggregation correlated with glucose levels but not with HbA1C levels. In conclusion, plasma and whole blood viscosity, as well as red blood cell aggregation appear to be associated with concurrent hyperglycemia, but not with the quality of glycemic control or the applied antidiabetic treatment. Platelet aggregation induced by ADP or epinephrine does not seem to be associated with diabetes even at subthreshold doses.
Collapse
|
19
|
Maruhashi T, Higashi Y. Pathophysiological Association between Diabetes Mellitus and Endothelial Dysfunction. Antioxidants (Basel) 2021; 10:antiox10081306. [PMID: 34439553 PMCID: PMC8389282 DOI: 10.3390/antiox10081306] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Endothelial dysfunction plays a critical role in atherosclerosis progression, leading to cardiovascular complications. There are significant associations between diabetes mellitus, oxidative stress, and endothelial dysfunction. Oxidative stress is increased by chronic hyperglycemia and acute glucose fluctuations induced by postprandial hyperglycemia in patients with diabetes mellitus. In addition, selective insulin resistance in the phosphoinositide 3-kinase/Akt/endothelial nitric oxide (NO) synthase pathway in endothelial cells is involved in decreased NO production and increased endothelin-1 production from the endothelium, resulting in endothelial dysfunction. In a clinical setting, selecting an appropriate therapeutic intervention that improves or augments endothelial function is important for preventing diabetic vascular complications. Hypoglycemic drugs that reduce glucose fluctuations by decreasing the postprandial rise in blood glucose levels, such as glinides, α-glucosidase inhibitors and dipeptidyl peptidase 4 inhibitors, and hypoglycemic drugs that ameliorate insulin sensitivity, such as thiazolidinediones and metformin, are expected to improve or augment endothelial function in patients with diabetes. Glucagon-like peptide 1 receptor agonists, metformin, and sodium-glucose cotransporter 2 inhibitors may improve endothelial function through multiple mechanisms, some of which are independent of glucose control or insulin signaling. Oral administration of antioxidants is not recommended in patients with diabetes due to the lack of evidence for the efficacy against diabetic complications.
Collapse
Affiliation(s)
- Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan;
| | - Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan;
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 734-8551, Japan
- Correspondence: ; Tel.: +81-82-257-5831
| |
Collapse
|
20
|
de Miranda FS, Guimarães JPT, Menikdiwela KR, Mabry B, Dhakal R, Rahman RL, Moussa H, Moustaid-Moussa N. Breast cancer and the renin-angiotensin system (RAS): Therapeutic approaches and related metabolic diseases. Mol Cell Endocrinol 2021; 528:111245. [PMID: 33753205 DOI: 10.1016/j.mce.2021.111245] [Citation(s) in RCA: 2] [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: 11/14/2020] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022]
Abstract
The Renin-Angiotensin System (RAS) is classically recognized for regulating blood pressure and fluid balance. Recently, this role has extended to other areas including inflammation, obesity, diabetes, as well as breast cancer. RAS components are expressed in normal and cancerous breast tissues, and downregulation of RAS inhibits metastasis, proliferation, angiogenesis, and desmoplasia in the tumor microenvironment. Therefore, RAS inhibitors (Angiotensin receptor blockers, ARBs, or angiotensin converting enzyme inhibitors, ACE-I) may be beneficial as preventive adjuvant therapies to thwart breast cancer development and improve outcomes, respectively. Given the beneficial effects of RAS inhibitors in metabolic diseases, which often co-exist in breast cancer patients, combining RAS inhibitors with other breast cancer therapies may enhance the effectiveness of current treatments. This review scrutinizes above associations, to advance our understanding of the role of RAS in breast cancer and its potential for repurposing of RAS inhibitors to improve the therapeutic approach for breast cancer patients.
Collapse
Affiliation(s)
- Flávia Sardela de Miranda
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - João Pedro Tôrres Guimarães
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA; Laboratory of Immunopharmacology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo (ICB/USP), São Paulo, SP, Brazil; Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo (FCF/USP), São Paulo, SP, Brazil
| | - Kalhara R Menikdiwela
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Brennan Mabry
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA
| | - Rabin Dhakal
- Department of Mechanical Engineering, Texas Tech University (TTU), Lubbock, TX, USA
| | - Rakhshanda Layeequr Rahman
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Hanna Moussa
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA; Department of Mechanical Engineering, Texas Tech University (TTU), Lubbock, TX, USA
| | - Naima Moustaid-Moussa
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA.
| |
Collapse
|
21
|
Chen S, Shen Y, Liu YH, Dai Y, Wu ZM, Wang XQ, Yang CD, Li LY, Liu JM, Zhang LP, Shen WF, Ji R, Lu L, Ding FH. Impact of glycemic control on the association of endothelial dysfunction and coronary artery disease in patients with type 2 diabetes mellitus. Cardiovasc Diabetol 2021; 20:64. [PMID: 33714276 PMCID: PMC7956110 DOI: 10.1186/s12933-021-01257-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
Background We investigated whether glycemic control affects the relation between endothelial dysfunction and coronary artery disease in patients with type 2 diabetes mellitus (T2DM). Methods In 102 type 2 diabetic patients with stable angina, endothelial function was evaluated using brachial artery flow-mediated dilation (FMD) with high-resolution ultrasound, and significant stenosis of major epicardial coronary arteries (≥ 50% diameter narrowing) and degree of coronary atherosclerosis (Gensini score and SYNTAX score) were determined. The status of glycemic control was assessed by blood concentration of glycated hemoglobin (HbA1c). Results The prevalence of significant coronary artery stenosis (67.9% vs. 37.0%, P = 0.002) and degree of coronary atherosclerosis (Gensini score: 48.99 ± 48.88 vs. 15.07 ± 21.03, P < 0.001; SYNTAX score: 15.88 ± 16.36 vs. 7.28 ± 10.54, P = 0.003) were higher and FMD was lower (6.03 ± 2.08% vs. 6.94 ± 2.20%, P = 0.036) in diabetic patients with poor glycemic control (HbA1c ≥ 7.0%; n = 56) compared to those with good glycemic control (HbA1c < 7.0%; n = 46). Multivariate regression analysis revealed that tertile of FMD was an independent determinant of presence of significant coronary artery stenosis (OR = 0.227 95% CI 0.056–0.915, P = 0.037), Gensini score (β = − 0.470, P < 0.001) and SYNTAX score (β = − 0.349, P = 0.004) in diabetic patients with poor glycemic control but not for those with good glycemic control (P > 0.05). Conclusion Poor glycemic control negatively influences the association of endothelial dysfunction and coronary artery disease in T2DM patients.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Ying Shen
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Yong-Hua Liu
- Department of Cardiology, Bao Shan People's Hospital, Baoshan, Yunnan Province, China
| | - Yang Dai
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Zhi-Ming Wu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Xiao-Qun Wang
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Chen-Die Yang
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Le-Ying Li
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Jing-Meng Liu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Li-Ping Zhang
- Department of Cardiology, Bao Shan People's Hospital, Baoshan, Yunnan Province, China
| | - Wei-Feng Shen
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Ri Ji
- Department of Ultrasound, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lin Lu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China.
| | - Feng-Hua Ding
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
22
|
Salvatore T, Pafundi PC, Galiero R, Rinaldi L, Caturano A, Vetrano E, Aprea C, Albanese G, Di Martino A, Ricozzi C, Imbriani S, Sasso FC. Can Metformin Exert as an Active Drug on Endothelial Dysfunction in Diabetic Subjects? Biomedicines 2020; 9:biomedicines9010003. [PMID: 33375185 PMCID: PMC7822116 DOI: 10.3390/biomedicines9010003] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular mortality is a major cause of death among in type 2 diabetes (T2DM). Endothelial dysfunction (ED) is a well-known important risk factor for the development of diabetes cardiovascular complications. Therefore, the prevention of diabetic macroangiopathies by preserving endothelial function represents a major therapeutic concern for all National Health Systems. Several complex mechanisms support ED in diabetic patients, frequently cross-talking each other: uncoupling of eNOS with impaired endothelium-dependent vascular response, increased ROS production, mitochondrial dysfunction, activation of polyol pathway, generation of advanced glycation end-products (AGEs), activation of protein kinase C (PKC), endothelial inflammation, endothelial apoptosis and senescence, and dysregulation of microRNAs (miRNAs). Metformin is a milestone in T2DM treatment. To date, according to most recent EASD/ADA guidelines, it still represents the first-choice drug in these patients. Intriguingly, several extraglycemic effects of metformin have been recently observed, among which large preclinical and clinical evidence support metformin’s efficacy against ED in T2DM. Metformin seems effective thanks to its favorable action on all the aforementioned pathophysiological ED mechanisms. AMPK pharmacological activation plays a key role, with metformin inhibiting inflammation and improving ED. Therefore, aim of this review is to assess metformin’s beneficial effects on endothelial dysfunction in T2DM, which could preempt development of atherosclerosis.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio 7, I-80138 Naples, Italy;
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Concetta Aprea
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Carmen Ricozzi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Simona Imbriani
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
- Correspondence: ; Tel.: +39-081-566-5010
| |
Collapse
|
23
|
Hsu JY, Lin HH, Wang ZH, Chen JH. Aqueous extract from Pepino (Solanum muricatum Ait.) leaves ameliorated insulin resistance, hyperlipidemia, and hyperglycemia in mice with metabolic syndrome. J Food Biochem 2020; 44:e13518. [PMID: 33047354 DOI: 10.1111/jfbc.13518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/30/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022]
Abstract
Solanum muricatum Ait. (Pepino) is a plant food commonly cultivated in the Penghu Island, Taiwan. This present study aimed to investigate the protective effects of aqueous extract of Pepino leaves (AEPL) in mice with metabolic syndrome. Metabolic syndrome animal model was induced by continuous high-fat diet feeding and low-dose streptozotocin (40 mg/ml) for 5 days. A 1% AEPL or metformin were given for 6 weeks after streptozotocin injection. The results revealed that 1% AEPL effectively reduced fasting blood glucose, insulin resistance, and hyperlipidemia in metabolic syndrome mice. Histologic examination revealed lipid accumulation in liver decreased by 1% AEPL treatment. Further, western blot analysis revealed 1% AEPL treatment managed enzymes related to lipid synthesis and oxidation pathways and hepatic glucose production. Besides, 1% AEPL treatment increased liver antioxidant activities to against oxidative stress. These results concluded that AEPL treatment attenuated insulin resistance, hyperlipidemia, and hyperglycemia of metabolic syndrome. PRACTICAL APPLICATIONS: Metabolic syndrome (MS) is a multifactorial chronic disease which is characterized by dyslipidemia, insulin resistance, and hyperglycemia. However, there is no single drug or defined medication for MS so far. The present study revealed that AEPL treatment was able to regulate lipid metabolism and glycemic control at the molecular level to alleviate MS. AEPL has the potential to be a novo complementary medication for metabolic syndrome.
Collapse
Affiliation(s)
- Jen-Ying Hsu
- Department of Nutrition, Chung Shan Medical University, Taichung City, Taiwan
| | - Hui-Hsuan Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, Taiwan
| | - Zhi-Hong Wang
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung City, Taiwan
| | - Jing-Hsien Chen
- Department of Nutrition, Chung Shan Medical University, Taichung City, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung City, Taiwan
| |
Collapse
|
24
|
Role of metformin in various pathologies: state-of-the-art microcapsules for improving its pharmacokinetics. Ther Deliv 2020; 11:733-753. [PMID: 32967584 DOI: 10.4155/tde-2020-0102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Metformin was originally derived from a botanical ancestry and became the most prescribed, first-line therapy for Type 2 diabetes in most countries. In the last century, metformin was discovered twice for its antiglycemic properties in addition to its antimalarial and anti-influenza effects. Metformin exhibits flip-flop pharmacokinetics with limited oral bioavailability. This review outlines metformin pharmacokinetics, pharmacodynamics and recent advances in polymeric particulate delivery systems as a potential tool to target metformin delivery to specific tissues/organs. This interesting biguanide is being rediscovered this century for multiple clinical indications as anticancer, anti-aging, anti-inflammatory, anti-Alzheimer's and much more. Microparticulate delivery systems of metformin may improve its oral bioavailability and optimize the therapeutic goals expected.
Collapse
|
25
|
Effect of N-Acetylcysteine on Metabolic Profile in Metabolic Syndrome Patients. Metab Syndr Relat Disord 2020; 18:341-346. [DOI: 10.1089/met.2020.0017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
26
|
Metformin as a Potential Agent in the Treatment of Multiple Sclerosis. Int J Mol Sci 2020; 21:ijms21175957. [PMID: 32825027 PMCID: PMC7503488 DOI: 10.3390/ijms21175957] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
Metformin, a synthetic derivative of guanidine, is commonly used as an oral antidiabetic agent and is considered a multi-vector application agent in the treatment of other inflammatory diseases. Recent studies have confirmed the beneficial effect of metformin on immune cells, with special emphasis on immunological mechanisms. Multiple Sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) characterized by various clinical courses. Although the pathophysiology of MS remains unknown, it is most likely a combination of disturbances of the immune system and biochemical pathways with a disruption of blood-brain barrier (BBB), and it is strictly related to injury of intracerebral blood vessels. Metformin has properties which are greatly desirable for MS therapy, including antioxidant, anti-inflammatory or antiplatelet functions. The latest reports relating to the cardiovascular disease confirm an increased risk of ischemic events in MS patients, which are directly associated with a coagulation cascade and an elevated pro-thrombotic platelet function. Hence, this review examines the potential favourable effects of metformin in the course of MS, its role in preventing inflammation and endothelial dysfunction, as well as its potential antiplatelet role.
Collapse
|
27
|
Kamenova P. Therapeutic potential of metformin in normal glucose tolerant persons with metabolic syndrome. BIOTECHNOL BIOTEC EQ 2020. [DOI: 10.1080/13102818.2019.1711184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Petya Kamenova
- Division of Diabetology, Department of Endocrinology, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
28
|
Markowicz-Piasecka M, Huttunen KM, Sadkowska A, Sikora J. Pleiotropic Activity of Metformin and Its Sulfonamide Derivatives on Vascular and Platelet Haemostasis. Molecules 2019; 25:E125. [PMID: 31905674 PMCID: PMC6982810 DOI: 10.3390/molecules25010125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 12/20/2022] Open
Abstract
As type 2 diabetes mellitus (T2DM) predisposes patients to endothelial cell injury and dysfunction, improvement of vascular function should be an important target for therapy. The aim of this study was to evaluate the effects of metformin, its sulfenamide and sulfonamide derivatives on selected parameters of endothelial and smooth muscle cell function, and platelet activity. Metformin was not found to significantly affect the viability of human umbilical vein endothelial cells (HUVECs) or aortal smooth muscle cells (AoSMC); however, it decreased cell migration by approximately 21.8% in wound healing assays after 24 h stimulation (wound closure 32.5 µm versus 41.5 µm for control). Metformin reduced platelet aggregation manifested by 19.0% decrease in maximum of aggregation (Amax), and 20% reduction in initial platelet aggregation velocity (v0). Furthermore, metformin decreased spontaneous platelet adhesion by 27.7% and ADP-induced adhesion to fibrinogen by 29.6% in comparison to control. Metformin sulfenamide with an n-butyl alkyl chain (compound 1) appeared to exert the most unfavourable effects on AoSMC cell viability (IC50 = 0.902 ± 0.015 μmol/mL), while 4-nitrobenzenesulfonamide (compound 3) and 2-nitrobenzenesulfonamide (compound 4) derivatives of metformin did not affect AoSMC and HUVEC viability at concentrations up to 2.0 μmol/mL. These compounds were also found to significantly reduce migration of smooth muscle cells by approximately 81.0%. Furthermore, sulfonamides 3 and 4 decreased the initial velocity of platelet aggregation by 11.8% and 20.6%, respectively, and ADP-induced platelet adhesion to fibrinogen by 76.3% and 65.6%. Metformin and its p- and o-nitro-benzenesulfonamide derivatives 3, 4 appear to exert beneficial effects on some parameters of vascular and platelet haemostasis.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland;
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland;
| | - Adrianna Sadkowska
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland;
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland;
| |
Collapse
|
29
|
Chen H, Vanhoutte PM, Leung SWS. Vascular adenosine monophosphate-activated protein kinase: Enhancer, brake or both? Basic Clin Pharmacol Toxicol 2019; 127:81-91. [PMID: 31671245 DOI: 10.1111/bcpt.13357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/24/2019] [Indexed: 12/25/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK), expressed/present ubiquitously in the body, contributes to metabolic regulation. In the vasculature, activation of AMPK is associated with several beneficial biological effects including enhancement of vasodilatation, reduction of oxidative stress and inhibition of inflammatory reactions. The vascular protective effects of certain anti-diabetic (metformin and sitagliptin) or lipid-lowering (simvastatin and fenofibrate) therapeutic agents, of active components of Chinese medicinal herbs (resveratrol and berberine) and of pharmacological agents (AICAR, A769662 and PT1) have been attributed to the activation of AMPK (in endothelial cells, vascular smooth muscle cells and/or perivascular adipocytes), independently of changes in the metabolic profile (eg glucose tolerance and/or plasma lipoprotein levels), leading to improved endothelium-derived nitric oxide-mediated vasodilatation and attenuated endothelium-derived cyclooxygenase-dependent vasoconstriction. By contrast, endothelial AMPK activation with pharmacological agents or by genetic modification is associated with reduced endothelium-dependent relaxations in small blood vessels and elevated systolic blood pressure. Indeed, AMPK activators inhibit endothelium-dependent hyperpolarization (EDH)-type relaxations in superior mesenteric arteries, partly by inhibiting endothelial calcium-activated potassium channel signalling. Therefore, AMPK activation is not necessarily beneficial in terms of endothelial function. The contribution of endothelial AMPK in the regulation of vascular tone, in particular in the microvasculature where EDH plays a more important role, remains to be characterized.
Collapse
Affiliation(s)
- Hui Chen
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Paul Michel Vanhoutte
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Susan Wai Sum Leung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
30
|
Zainordin NA, Hatta SFWM, Mohamed Shah FZ, Rahman TA, Ismail N, Ismail Z, Abdul Ghani R. Effects of Dapagliflozin on Endothelial Dysfunction in Type 2 Diabetes With Established Ischemic Heart Disease (EDIFIED). J Endocr Soc 2019; 4:bvz017. [PMID: 31993550 PMCID: PMC6977943 DOI: 10.1210/jendso/bvz017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023] Open
Abstract
Objectives To evaluate the effect of the sodium-glucose cotransporter 2 inhibitor (SGLT2-I) dapagliflozin on endothelial function in patients with high-risk type 2 diabetes mellitus (T2DM). Methods This was a prospective, double-blind, randomized, placebo-controlled, clinical trial of patients with T2DM with underlying ischemic heart disease who were receiving metformin and insulin therapy (n = 81). After 12-weeks of additional therapy with either dapagliflozin (n = 40) or placebo (n = 41), systemic endothelial function was evaluated by change in flow-mediated dilation (ΔFMD), change in nitroglycerin-mediated dilation (ΔNMD) and surrogate markers including intercellular adhesion molecule 1 (ICAM-1), endothelial nitric oxide synthase (eNOS), high-sensitivity C-reactive protein (hs-CRP), and lipoprotein(a) (Lp[a]). Glycemic and lipid profiles were also measured. Results The dapagliflozin group demonstrated significant reductions of hemoglobin A1c (HbA1c) and fasting blood glucose (FBG) compared to the placebo group (ΔHbA1c –0.83 ± 1.47% vs –0.16 ± 1.25%, P = 0.042 and ΔFBG vs –0.73 ± 4.55 mmol/L vs –1.90 ± 4.40 mmol/L, P = 0.015, respectively). The placebo group showed worsening of ΔFMD while the dapagliflozin group maintained similar measurements pre- and posttherapy (P = not significant). There was a reduction in ICAM-1 levels in the dapagliflozin group (–83.9 ± 205.9 ng/mL, P < 0.02), which remained unchanged in the placebo group (–11.0 ± 169.1 ng/mL, P = 0.699). Univariate correlation analysis revealed a significant negative correlation between HbA1c and ΔFMD within the active group. Conclusion A 12-week therapy with dapagliflozin, in addition to insulin and metformin therapies, in high-risk patients resulted in significant reductions in HbA1c, FBG, and surrogate markers of the endothelial function. Although the dapagliflozin group demonstrated a significant association between reduction in HbA1c and improvement in FMD, there was no significant difference in FMD between the 2 groups.
Collapse
Affiliation(s)
- Nur Aisyah Zainordin
- Endocrine Unit, Department of Internal Medicine, Faculty of Medicine, University Teknologi MARA (UiTM), Selangor, Malaysia
| | | | - Fatimah Zaherah Mohamed Shah
- Endocrine Unit, Department of Internal Medicine, Faculty of Medicine, University Teknologi MARA (UiTM), Selangor, Malaysia
| | - Thuhairah Abdul Rahman
- Pathology Diagnostic and Research Laboratories (CPDRL) Faculty of Medicine University Teknologi MARA (UiTM), Sg Buloh Campus, Selangor, Malaysia
| | - Nurhuda Ismail
- Department of Population Health & Preventive Medicine, Faculty of Medicine, University Teknologi MARA (UiTM), Selangor, Malaysia
| | - Zaliha Ismail
- Department of Population Health & Preventive Medicine, Faculty of Medicine, University Teknologi MARA (UiTM), Selangor, Malaysia
| | - Rohana Abdul Ghani
- Endocrine Unit, Department of Internal Medicine, Faculty of Medicine, University Teknologi MARA (UiTM), Selangor, Malaysia
| |
Collapse
|
31
|
Kibrik P, Alsheekh A, Izakovich T, Chait J, Goldstein MA, Monteleone CM, Hingorani A, Ascher E. Does Metformin Have an Effect on Stent Patency Rates. Vasc Endovascular Surg 2019; 53:452-457. [PMID: 31170884 DOI: 10.1177/1538574419849999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Metformin is the most commonly used drug for type 2 diabetes. Research has shown that metformin also has a protective effect on endothelium by decreasing endothelial vascular reactivity. We hypothesize that metformin will decrease restenosis/reintervention rates in patients receiving lower extremity non-drug-eluting stents (nDESs) in the superficial femoral artery(SFA) and/or popliteal artery. MATERIALS/METHODS Retrospective study was performed on 187 patients from October 2012 to December 2015 who received an nDES in the SFA and/or popliteal artery. Patients were divided into 3 groups (Table 1) and compared against for duplex based restenosis (>60%) rates, limb loss rates, and reintervention rates. Each patient's Trans-Atlantic-Inter-Society-Consensus II (TASC-II) class was collected. Postoperative duplex was performed 1 week after the procedure, then every 3 months for the first year, then, every 6 months to check for patency. IBM-SPSS-22 was used for all analyses. RESULTS Average age of the patients was 64.65 ± 73.4 years. 101 patients had 101 procedures performed on the left lower extremity; 86 patients had 86 procedures performed on the right lower extremity; 123 patients were male and 64 were female. Average length of follow-up was 13.1±9.7 months. Most common indication for intervention was claudication, followed by critical limb threatening ischemia. Restenosis and reintervention by groups can be seen in Table 1. No patients experienced limb loss. There were no statistically significant differences between any of the 3 groups and their limb loss, restenosis, or reintervention rates. CONCLUSIONS Despite having multiple proven effects in improving certain clinical outcomes and a proven protective effect on endothelium by decreasing endothelial vascular reactivity, metformin does not appear to reduce restenosis or reintervention rates in patients receiving lower extremity nDESs in the SFA and/or popliteal artery.
Collapse
Affiliation(s)
- Pavel Kibrik
- 1 Vascular Institute of New York, Brooklyn, NY, USA
| | | | | | - Jesse Chait
- 1 Vascular Institute of New York, Brooklyn, NY, USA
| | | | | | | | | |
Collapse
|
32
|
Markowicz-Piasecka M, Huttunen KM, Broncel M, Sikora J. Sulfenamide and Sulfonamide Derivatives of Metformin - A New Option to Improve Endothelial Function and Plasma Haemostasis. Sci Rep 2019; 9:6573. [PMID: 31024058 PMCID: PMC6484023 DOI: 10.1038/s41598-019-43083-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/16/2019] [Indexed: 12/30/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a multi-factorial disease which can cause multiple organ dysfunction, including that of the vascular endothelium. The aim of the present study was to evaluate the effects of metformin, and its sulfenamide and sulfonamide derivatives (compounds 1–8) on the selected markers of endothelial function and blood coagulation. The integrity of endothelial cells(ECs) was examined using the real-time cell electric impedance system. Tissue Factor(TF) production, the release of von Willebrand Factor (vWF) and tissue plasminogen activator(t-PA) from ECs were determined using immunoenzymatic assays, while the process of platelet thrombus formation using the Total Thrombus-Formation Analysis System. Sulfenamide with n-butyl alkyl chain(3) does not interfere with ECs integrity, and viability (nCI(24h) = 1.03 ± 0.03 vs. 1.06 ± 0.11 for control), but possesses anticoagulation properties manifested by prolonged platelet-dependent thrombus formation (Occlusion Time 370.3 ± 77.0 s vs. 286.7 ± 65.5 s for control) in semi-physiological conditions. Both p- and o-nitro-benzenesulfonamides (compounds7,8) exhibit anti-coagulant properties demonstrated by decreased vWF release and prolonged parameters of platelet thrombus formation and total blood thrombogenicity. In conclusion, chemical modification of metformin scaffold into sulfenamides or sulfonamides might be regarded as a good starting point for the design and synthesis of novel biguanide-based compounds with anticoagulant properties and valuable features regarding endothelial function.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151, Lodz, Poland.
| | - Kristiina M Huttunen
- School Of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211, Kuopio, Finland
| | - Marlena Broncel
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Kniaziewicza 1/5, 91-347, Lodz, Poland
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151, Lodz, Poland
| |
Collapse
|
33
|
Schiapaccassa A, Maranhão PA, de Souza MDGC, Panazzolo DG, Nogueira Neto JF, Bouskela E, Kraemer-Aguiar LG. 30-days effects of vildagliptin on vascular function, plasma viscosity, inflammation, oxidative stress, and intestinal peptides on drug-naïve women with diabetes and obesity: a randomized head-to-head metformin-controlled study. Diabetol Metab Syndr 2019; 11:70. [PMID: 31462933 PMCID: PMC6708186 DOI: 10.1186/s13098-019-0466-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Obesity is the main risk factor for diabetes and excessive visceral fat triggers low-grade inflammatory process, mediated by activation and release of cytokines and high flow of free fatty acids that contribute to insulin resistance, increased oxidative stress, and impaired endothelial function. Metformin and vildagliptin have known vasculoprotective actions, but the value of these drugs on drug-naïve diabetic patients during 30 days use warrants investigation. Our purpose was to observe their effects on endothelial function, oxidative stress, inflammatory biomarkers, and plasma viscosity. METHODS 38 women with obesity and type 2 diabetes drug-naïve, aged between 19 and 50 years, BMI ≥ 30 kg/m2, were recruited and subjected to measurements of endothelial function, nutritive skin microvascular reactivity, plasma viscosity, inflammatory and oxidative stress biomarkers at baseline and randomized 1:1 to ingest metformin (850 mg twice/day) or vildagliptin (50 mg twice/day) during 30 days, and then, re-evaluated. RESULTS No differences between groups were noticed at baseline. After treatment, vildagliptin promoted an improvement on endothelial-dependent and -independent vasodilatations, at arteriole level, while metformin resulted in improved nutritive microvascular reactivity, at the capillary level. Intragroup analysis showed that vildagliptin reduced insulin, C-peptide and oxidized LDL, and increased adiponectin and glucagon-like peptide-1 while metformin reduced weight, plasma glucose, total cholesterol, HDL-c, LDL-c, and dipeptidyl peptidase-4 activity, with an unexpected increase on tumor necrosis factor-α. No significant difference in plasma viscosity was noted. CONCLUSIONS In the vascular beds investigated, both drugs used for only 30 days improved endothelial function, through distinct, and possibly, complementary mechanisms on drug-naïve diabetic women.Trial Registration ClinicalTrials.gov: NCT01827280.
Collapse
Affiliation(s)
- Alessandra Schiapaccassa
- Postgraduate Program in Clinical and Experimental Physiopathology (FISCLINEX), Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, RJ 20550-013 Brazil
| | - Priscila A. Maranhão
- Laboratory of Clinical and Experimental Research on Vascular Biology (BioVasc), Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, RJ 20550-013 Brazil
| | - Maria das Graças Coelho de Souza
- Laboratory of Clinical and Experimental Research on Vascular Biology (BioVasc), Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, RJ 20550-013 Brazil
| | - Diogo G. Panazzolo
- Postgraduate Program in Clinical and Experimental Physiopathology (FISCLINEX), Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, RJ 20550-013 Brazil
| | - José Firmino Nogueira Neto
- Lipids Laboratory (Lablip), Policlínica Piquet Carneiro, State University of Rio de Janeiro, Rio de Janeiro, RJ 20550-003 Brazil
| | - Eliete Bouskela
- Laboratory of Clinical and Experimental Research on Vascular Biology (BioVasc), Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, RJ 20550-013 Brazil
| | - Luiz Guilherme Kraemer-Aguiar
- Laboratory of Clinical and Experimental Research on Vascular Biology (BioVasc), Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, RJ 20550-013 Brazil
- Obesity Unit, Policlínica Piquet Carneiro, Department of Internal Medicine, Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, RJ 20550-030 Brazil
| |
Collapse
|
34
|
Mi Q, Li Y, Wang M, Yang G, Zhao X, Liu H, Zheng H, Li L. Circulating C1q/TNF-related protein isoform 15 is a marker for the presence of metabolic syndrome. Diabetes Metab Res Rev 2019; 35:e3085. [PMID: 30303269 DOI: 10.1002/dmrr.3085] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 08/31/2018] [Accepted: 09/21/2018] [Indexed: 11/06/2022]
Abstract
BACKGROUND C1q/TNF-related protein isoform 15 (CTRP15) has been reported to be related to glucose and lipid metabolism, but the results are inconsistent. Metabolic syndrome (MetS) is a cluster of metabolic disorders. The aim of this study is to determine circulating CTRP15 levels in individuals with MetS and investigate the association among circulating CTRP15 and markers for MetS as well as insulin resistance. METHODS A total of 341 individuals were recruited for this cross-sectional study. These subjects were screened for MetS. Serum CTRP15 concentrations were measured by ELISA. RESULTS Serum CTRP15 levels were significantly higher in MetS individuals relative to those of the healthy individuals. Circulating CTRP15 correlated positively with WHR, BMI, SBP, FAT %, 2 h-BG, FIns, 2 h-Ins, TG, FFA, HbA1c, HOMA-IR, and AUCglucose , while negatively with HDL-C and ISI. Multiple linear regression showed that HOMA-IR and HDL-C are independently related factors influencing serum CTRP15 concentrations. In addition, binary logistic regression analysis showed that serum CTRP15 concentrations were significantly related to MetS. When the mean concentrations of circulating CTRP15 in MetS subjects were stratified by the number of components of the MetS, circulating CTRP15 was found to increase progressively with increasing number of the MetS components. Finally, ROC curve analysis showed that the best cutoff values for circulating CTRP15 to predict MetS and insulin resistance were 63.6 and 64.0 μg/L. CONCLUSIONS Serum CTRP15 concentrations were associated with the key components of MetS and insulin resistance.
Collapse
Affiliation(s)
- Qiao Mi
- Key Laboratory of Diagnostic Medicine (Ministry of Education) and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yanxin Li
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Miao Wang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xili Zhao
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Internal Medicine, Shizhu People's Hospital in Chongqing, Chongqing, China
| | - Hua Liu
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Ling Li
- Key Laboratory of Diagnostic Medicine (Ministry of Education) and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
35
|
Anholm C, Kumarathurai P, Jürs A, Pedersen LR, Nielsen OW, Kristiansen OP, Fenger M, Holst JJ, Madsbad S, Sajadieh A, Haugaard SB. Liraglutide improves the beta-cell function without increasing insulin secretion during a mixed meal in patients, who exhibit well-controlled type 2 diabetes and coronary artery disease. Diabetol Metab Syndr 2019; 11:42. [PMID: 31164926 PMCID: PMC6543623 DOI: 10.1186/s13098-019-0438-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/17/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Hyperinsulinemia aggravates insulin resistance and cardio-vascular disease. How the insulinotropic glucagon-like peptide-1 receptor agonist liraglutide in a physiologic post-prandial setting may act on pancreatic alpha and beta-cell function in patients with coronary artery disease (CAD) and type 2 diabetes (T2DM) is less clear. METHODS Insulin resistant patients with established CAD and newly diagnosed well-controlled T2DM were recruited to a placebo-controlled, cross-over trial with two treatment periods of 12 weeks and a 2 weeks wash-out period before and in-between. Treatment was liraglutide or placebo titrated from 0.6 mg q.d. to 1.8 mg q.d. within 4 weeks and metformin titrated from 500 mg b.i.d to 1000 mg b.i.d. within 4 weeks. Before and after intervention in both 12 weeks periods insulin, C-peptide, glucose, and glucagon were measured during a meal test. Beta-cell function derived from the oral glucose tolerance setting was calculated as changes in insulin secretion per unit changes in glucose concentration (Btotal) and whole-body insulin resistance using ISIcomposite. RESULTS Liraglutide increased the disposition index [Btotal × ISIcomposite, by 40% (n = 24, p < 0.001)] compared to placebo. Post-prandial insulin and glucose was reduced by metformin in combination with liraglutide and differed, but not significantly different from placebo, moreover, glucagon concentration was unaffected. Additionally, insulin clearance tended to increase during liraglutide therapy (n = 26, p = 0.06). CONCLUSIONS The insulinotropic drug liraglutide may without increasing the insulin concentration reduce postprandial glucose but not glucagon excursions and improve beta-cell function in newly diagnosed and well-controlled T2DM.Trial registration Clinicaltrials.gov ID: NCT01595789.
Collapse
Affiliation(s)
- Christian Anholm
- Department of Internal Medicine, Copenhagen University Hospital Glostrup, Nordre Ringvej 57, 2600 Glostrup, Denmark
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Preman Kumarathurai
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Anders Jürs
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Lene Rørholm Pedersen
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Olav Wendelboe Nielsen
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Ole Peter Kristiansen
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Mogens Fenger
- Department of Clinical Biochemistry, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
| | - Jens Juul Holst
- NovoNordisk Foundation Center for Metabolic Research and Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
| | - Ahmad Sajadieh
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Steen Bendix Haugaard
- Department of Internal Medicine, Copenhagen University Hospital Glostrup, Nordre Ringvej 57, 2600 Glostrup, Denmark
- Department of Endocrinology I, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| |
Collapse
|
36
|
Nafisa A, Gray SG, Cao Y, Wang T, Xu S, Wattoo FH, Barras M, Cohen N, Kamato D, Little PJ. Endothelial function and dysfunction: Impact of metformin. Pharmacol Ther 2018; 192:150-162. [PMID: 30056057 DOI: 10.1016/j.pharmthera.2018.07.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular and metabolic diseases remain the leading cause of morbidity and mortality worldwide. Endothelial dysfunction is a key player in the initiation and progression of cardiovascular and metabolic diseases. Current evidence suggests that the anti-diabetic drug metformin improves insulin resistance and protects against endothelial dysfunction in the vasculature. Hereby, we provide a timely review on the protective effects and molecular mechanisms of metformin in preventing endothelial dysfunction and cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Asma Nafisa
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.
| | - Susan G Gray
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.
| | - Yingnan Cao
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China
| | - Tinghuai Wang
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - Feroza H Wattoo
- Department of Biochemistry, PMAS Arid Agriculture University, Shamasabad, Muree Road, Rawalpindi 4600, Pakistan..
| | - Michael Barras
- Dept. of Pharmacy, Princess Alexandra Hospital, 199 Ipswich Rd, Woolloongabba, QLD 4102, Australia.
| | - Neale Cohen
- Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia.
| | - Danielle Kamato
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| |
Collapse
|
37
|
Moon KH, Park SY, Kim YW. Obesity and Erectile Dysfunction: From Bench to Clinical Implication. World J Mens Health 2018; 37:138-147. [PMID: 30079640 PMCID: PMC6479091 DOI: 10.5534/wjmh.180026] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/17/2018] [Accepted: 05/23/2018] [Indexed: 12/25/2022] Open
Abstract
Obesity is a major public health issue worldwide and is frequently associated with erectile dysfunction (ED). Both conditions may share an internal pathologic environment, also known as common soil. Their main pathophysiologic processes are oxidative stress, inflammation, and resultant insulin and leptin resistance. Moreover, the severity of ED is correlated with comorbid medical conditions, including obesity. Therefore, amelioration of these comorbidities may increase the efficacy of ED treatment with phosphodiesterase 5 inhibitors, the first-line medication for patients with ED. Although metformin was originally developed as an insulin sensitizer six decades ago, it has also been shown to improve leptin resistance. In addition, metformin has been reported to reduce oxidative stress, inflammatory response, and body weight, as well as improve ED, in animal and human studies. Moreover, administration of a combination of metformin and phosphodiesterase 5 inhibitors improves erectile function in patients with ED who have a poor response to sildenafil and are insulin resistant. Thus, concomitant treatment of metabolic derangements associated with obesity in patients with ED who are obese would improve the efficacy and reduce the refractory response to penile vasodilators. In this review, we discuss the connecting factors between obesity and ED and the possible combined treatment modalities.
Collapse
Affiliation(s)
- Ki Hak Moon
- Department of Urology, Yeungnam University College of Medicine, Daegu, Korea
| | - So Young Park
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Korea
| | - Yong Woon Kim
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Korea.
| |
Collapse
|
38
|
Guzik TJ, Cosentino F. Epigenetics and Immunometabolism in Diabetes and Aging. Antioxid Redox Signal 2018; 29:257-274. [PMID: 28891325 PMCID: PMC6012980 DOI: 10.1089/ars.2017.7299] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 09/04/2017] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE A strong relationship between hyperglycemia, impaired insulin pathway, and cardiovascular disease in type 2 diabetes (T2D) is linked to oxidative stress and inflammation. Immunometabolic pathways link these pathogenic processes and pose important potential therapeutic targets. Recent Advances: The link between immunity and metabolism is bidirectional and includes the role of inflammation in the pathogenesis of metabolic disorders such as T2D, obesity, metabolic syndrome, and hypertension and the role of metabolic factors in regulation of immune cell functions. Low-grade inflammation, oxidative stress, balance between superoxide and nitric oxide, and the infiltration of macrophages, T cells, and B cells in insulin-sensitive tissues lead to metabolic impairment and accelerated aging. CRITICAL ISSUES Inflammatory infiltrate and altered immune cell phenotype precede development of metabolic disorders. Inflammatory changes are tightly linked to alterations in metabolic status and energy expenditure and are controlled by epigenetic mechanisms. FUTURE DIRECTIONS A better comprehension of these mechanistic insights is of utmost importance to identify novel molecular targets. In this study, we describe a complex scenario of epigenetic changes and immunometabolism linking to diabetes and aging-associated vascular disease. Antioxid. Redox Signal. 29, 257-274.
Collapse
Affiliation(s)
- Tomasz J. Guzik
- BHF Centre for Research Excellence, Institute of Cardiovascular and Medical Research (ICAMS), University of Glasgow, Glasgow, United Kingdom
- Department of Internal and Agricultural Medicine, Laboratory of Translational Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
39
|
Abstract
The frequency of prediabetes is increasing as the prevalence of obesity rises worldwide. In prediabetes, hyperglycemia, insulin resistance, and inflammation and metabolic derangements associated with concomitant obesity cause endothelial vasodilator and fibrinolytic dysfunction, leading to increased risk of cardiovascular and renal disease. Importantly, the microvasculature affects insulin sensitivity by affecting the delivery of insulin and glucose to skeletal muscle; thus, endothelial dysfunction and extracellular matrix remodeling promote the progression from prediabetes to diabetes mellitus. Weight loss is the mainstay of treatment in prediabetes, but therapies that improved endothelial function and vasodilation may not only prevent cardiovascular disease but also slow progression to diabetes mellitus.
Collapse
Affiliation(s)
- David H Wasserman
- From the Departments of Molecular Physiology and Biophysics (D.H.W.) and Medicine (T.J.W., N.J.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Thomas J Wang
- From the Departments of Molecular Physiology and Biophysics (D.H.W.) and Medicine (T.J.W., N.J.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Nancy J Brown
- From the Departments of Molecular Physiology and Biophysics (D.H.W.) and Medicine (T.J.W., N.J.B.), Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
40
|
Taheri A, Lavasani H, Kasirzadeh S, Sheikholeslami B, Ardakani YH, Rouini MR. Changes in CYP2D enzyme activity following induction of type 2 diabetes, and administration of cinnamon and metformin: an experimental animal study. Xenobiotica 2017; 48:984-989. [PMID: 29092654 DOI: 10.1080/00498254.2017.1390626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
1. Alterations in the activity of hepatic cytochrome P-450 isoenzymes result in changes in the pharmacokinetic behavior of drugs. This study was designed to explore the impact of type II diabetes, metformin and cinnamon on the activity of CYP2D isoenzyme. 2. Streptozotocin-nicotinamide-induced diabetic and normal rats were gavaged by cinnamon and/or metformin for 14 days. Using isolated perfusion of rat livers, the metabolic activity of CYP2D in the study groups was evaluated based on the oxidative biotransformation of tramadol hydrochloride. 3. The metabolic ratios of O-desmethyltramadol, the product of CYP2D-mediated metabolism of tramadol, in normal and diabetic control rats were found to be 0.33 ± 0.12 and 0.29 ± 0.07, respectively. Cinnamon significantly reduced the mentioned ratio in both normal and diabetic rats (0.13 ± 0.05 and 0.15 ± 0.04) and metformin increased the reduced activity in diabetic rats (0.37 ± 0.09 versus 0.29 ± 0.07). 4. In conclusion, it is evident that this study has shown the significant inhibitory effect of cinnamon on CYP2D. This finding suggests that it should be taken into consideration the possible metabolism-related pharmacokinetic drug-cinnamon interactions. 5. Additionally, type 2 diabetes condition reduced the enzyme activity and metformin consumption reversed this reduction; however, the significance of the latest is not clear.
Collapse
Affiliation(s)
- Ali Taheri
- a Department of Pharmaceutics , Faculty of Pharmacy, Biopharmaceutics and Pharmacokinetics Division, Tehran University of Medical Sciences , Tehran , Iran
| | - Hoda Lavasani
- a Department of Pharmaceutics , Faculty of Pharmacy, Biopharmaceutics and Pharmacokinetics Division, Tehran University of Medical Sciences , Tehran , Iran
| | - Sara Kasirzadeh
- b Department of Toxicology and Pharmacology , Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran
| | | | - Yalda H Ardakani
- a Department of Pharmaceutics , Faculty of Pharmacy, Biopharmaceutics and Pharmacokinetics Division, Tehran University of Medical Sciences , Tehran , Iran
| | - Mohammad-Reza Rouini
- a Department of Pharmaceutics , Faculty of Pharmacy, Biopharmaceutics and Pharmacokinetics Division, Tehran University of Medical Sciences , Tehran , Iran.,d Faculty of Pharmacy, Pharmaceutical Sciences Research Centre , Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
41
|
Valencia WM, Palacio A, Tamariz L, Florez H. Metformin and ageing: improving ageing outcomes beyond glycaemic control. Diabetologia 2017; 60:1630-1638. [PMID: 28770328 PMCID: PMC5709209 DOI: 10.1007/s00125-017-4349-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/17/2017] [Indexed: 12/12/2022]
Abstract
In a world where the population is ageing, there is growing interest and demand for research evaluating strategies that address the ageing process. After 60 years of successful use of metformin in our pharmaceutical armamentarium, we are learning that, beyond improving glycaemic control, metformin may have additional mechanisms and pathways of action that need further study. Although, metformin's effect on clinical ageing outcomes may still be considered speculative, the findings from studies into cellular and animal models and from observational and pilot human studies support the existence of beneficial effects on ageing. At present, progress for human research, using randomised clinical trials to evaluate metformin's clinical impact, has just started. Here, we present a review on the ageing process and the mechanisms involved, and the role that metformin may have to counter these. We go on to discuss the upcoming large randomised clinical trials that may provide insight on the use of metformin for ageing outcomes beyond glycaemic control.
Collapse
Affiliation(s)
- Willy Marcos Valencia
- Geriatric Research Education and Clinical Center (GRECC), Miami VA Healthcare System, 1201 N.W. 16th Street, (11 GRC) CLC 207 A2, Miami, FL, 33125, USA.
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Ana Palacio
- Geriatric Research Education and Clinical Center (GRECC), Miami VA Healthcare System, 1201 N.W. 16th Street, (11 GRC) CLC 207 A2, Miami, FL, 33125, USA
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Leonardo Tamariz
- Geriatric Research Education and Clinical Center (GRECC), Miami VA Healthcare System, 1201 N.W. 16th Street, (11 GRC) CLC 207 A2, Miami, FL, 33125, USA
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hermes Florez
- Geriatric Research Education and Clinical Center (GRECC), Miami VA Healthcare System, 1201 N.W. 16th Street, (11 GRC) CLC 207 A2, Miami, FL, 33125, USA
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
42
|
Patel JP, Lee EH, Mena CI, Walker CN. Effects of metformin on endothelial health and erectile dysfunction. Transl Androl Urol 2017; 6:556-565. [PMID: 28725599 PMCID: PMC5503973 DOI: 10.21037/tau.2017.03.52] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Erectile dysfunction (ED) affects approximately 18 million American men. ED may be attributed to several etiologies, including arteriogenic, psychogenic, neurogenic, hormonal, drug-induced, and systemic disease or aging related factors. Specific to arteriogenic ED, three major mechanisms have been identified: (I) endothelium-dependent vasodilatory impairment; (II) sympathetic nerve activity elevation; (III) atherosclerotic luminal narrowing. Additionally, these insults have been linked to the insulin resistant state, which in turn is comorbid with obesity, dyslipidemia, diabetes, and hypertension. In this review, we summarize the evidence regarding the impact of metformin—an insulin sensitizer—on the three mechanisms of arteriogenic ED. We report that metformin treatment positively affects two of three pathways, specifically through enhanced endothelium-dependent vasodilation and sympathetic nerve activity attenuation, but does not seem to have a significant impact on hypertension regulation. Given the encouraging data found in both animal and clinical studies, we advocate for further studies on metformin use in ED.
Collapse
Affiliation(s)
- Jay Pravin Patel
- Yale School of Medicine & Yale School of Management, New Haven, CT, USA
| | - Eric Hweegeun Lee
- Yale School of Medicine & Yale School of Management, New Haven, CT, USA
| | - Carlos Ignacio Mena
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Charles N Walker
- Department of Urology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
43
|
Alfaras I, Di Germanio C, Bernier M, Csiszar A, Ungvari Z, Lakatta EG, de Cabo R. Pharmacological Strategies to Retard Cardiovascular Aging. Circ Res 2017; 118:1626-42. [PMID: 27174954 DOI: 10.1161/circresaha.116.307475] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 04/08/2016] [Indexed: 01/10/2023]
Abstract
Aging is the major risk factor for cardiovascular diseases, which are the leading cause of death in the United States. Traditionally, the effort to prevent cardiovascular disease has been focused on addressing the conventional risk factors, including hypertension, hyperglycemia, hypercholesterolemia, and high circulating levels of triglycerides. However, recent preclinical studies have identified new approaches to combat cardiovascular disease. Calorie restriction has been reproducibly shown to prolong lifespan in various experimental model animals. This has led to the development of calorie restriction mimetics and other pharmacological interventions capable to delay age-related diseases. In this review, we will address the mechanistic effects of aging per se on the cardiovascular system and focus on the prolongevity benefits of various therapeutic strategies that support cardiovascular health.
Collapse
Affiliation(s)
- Irene Alfaras
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Clara Di Germanio
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Michel Bernier
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Anna Csiszar
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Zoltan Ungvari
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Edward G Lakatta
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Rafael de Cabo
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.).
| |
Collapse
|
44
|
van Haare J, Kooi ME, van Teeffelen JWGE, Vink H, Slenter J, Cobelens H, Strijkers GJ, Koehn D, Post MJ, van Bilsen M. Metformin and sulodexide restore cardiac microvascular perfusion capacity in diet-induced obese rats. Cardiovasc Diabetol 2017; 16:47. [PMID: 28399917 PMCID: PMC5387275 DOI: 10.1186/s12933-017-0525-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/25/2017] [Indexed: 12/26/2022] Open
Abstract
Background Disturbances in coronary microcirculatory function, such as the endothelial glycocalyx, are early hallmarks in the development of obesity and insulin resistance. Accordingly, in the present study myocardial microcirculatory perfusion during rest and stress was assessed following metformin or sulodexide therapy in a rat model of diet-induced obesity. Additionally, the effect of degradation of the glycocalyx on myocardial perfusion was assessed in chow-fed rats. Methods Rats were fed a high fat diet (HFD) for 8 weeks and were divided into a group without therapy, and groups that received the anti-diabetic drug metformin or the glycocalyx-stabilizing drug sulodexide in their drinking water during the last 4 weeks of the feeding period. Myocardial microvascular perfusion was determined using first-pass perfusion MRI before and after adenosine infusion. The effect of HFD on microcirculatory properties was also assessed by sidestream darkfield (SDF) imaging of the gastrocnemius muscle. In an acute experimental setting, hyaluronidase was administered to chow-fed control rats to determine the effect of enzymatical degradation of the glycocalyx on myocardial perfusion. Results HFD-rats developed central obesity and insulin sensitivity was reduced as evidenced by the marked reduction in insulin-induced phosphorylation of Akt in both cardiac and gastrocnemius muscle. We confirmed our earlier findings that the robust increase in myocardial perfusion in chow-fed rats after an adenosine challenge (+56%, p = 0.002) is blunted in HFD rats (+8%, p = 0.68). In contrast, 4-weeks treatment with metformin or sulodexide partly restored the increase in myocardial perfusion during adenosine infusion in HFD rats (+81%, p = 0.002 and +37%, p = 0.02, respectively). Treating chow-fed rats acutely with hyaluronidase, to enzymatically degrade the glyocalyx, completely blunted the increase in myocardial perfusion during stress. Conclusions In early stages of HFD-induced insulin resistance myocardial perfusion becomes compromised, a process that can be countered by treatment with both metformin and sulodexide. The adverse effect of acute glycocalyx degradation and protective effect of long-term sulodexide administration on myocardial perfusion provides indirect evidence, suggesting a role for the glycocalyx in preserving coronary microvascular function in pre-diabetic animals.
Collapse
Affiliation(s)
- Judith van Haare
- Department of Physiology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - M Eline Kooi
- Department of Radiology and Nuclear Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | | | - Hans Vink
- Department of Physiology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Jos Slenter
- Department of Radiology and Nuclear Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Hanneke Cobelens
- Department of Physiology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Gustav J Strijkers
- Biomedical Engineering and Physics, Academic Medical Center, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands
| | - Dennis Koehn
- Pie Medical Imaging, P.O. Box 1132, 6201 BC, Maastricht, The Netherlands
| | - Mark J Post
- Department of Physiology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Marc van Bilsen
- Department of Physiology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands. .,Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|
45
|
Abdelgadir E, Ali R, Rashid F, Bashier A. Effect of Metformin on Different Non-Diabetes Related Conditions, a Special Focus on Malignant Conditions: Review of Literature. J Clin Med Res 2017; 9:388-395. [PMID: 28392858 PMCID: PMC5380171 DOI: 10.14740/jocmr2922e] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2017] [Indexed: 12/16/2022] Open
Abstract
Metformin has been proven to be one of the most safe and effective antihyperglycemic agents. Through more than six decades of metformin use, it became the most studied hypoglycemic agent; through these studies, it showed a marvelous non-glycemic related effect. These effects include modulation of different points of cancer timeline, weight reduction, cardiovascular health, thyroid diseases, polycystic ovaries disease and many other medical conditions. The aim of this review was to assess the effect of metformin on non-diabetes related medical diseases. We have examined the studies published in PubMed and summarized different randomized controlled trials, observational trials and review articles. This review has summarized most of the non-glycemic effects of metformin. Metformin has been solidly shown to be effective in weight control with certain medications, effective in neuroprotection, in endothelial health, in control of anti-HIV agent side effects and many other crucial health jeopardies. The effects in cancer timeline modulation have taken the biggest part, since it was the most studied area outside the diabetes field. Having mentioned all the above privileges, and in addition to the robust evidence in glycemic control, this consolidates the position of metformin as a first line agent in treatment of diabetes and pre-diabetes. Perhaps in the near future, we may see other indications to use metformin in non-diabetes patients.
Collapse
Affiliation(s)
| | - Razan Ali
- Dubai Hospital, Dubai Health Authority, Dubai, UAE
| | | | | |
Collapse
|
46
|
Triggle CR, Ding H. Metformin is not just an antihyperglycaemic drug but also has protective effects on the vascular endothelium. Acta Physiol (Oxf) 2017; 219:138-151. [PMID: 26680745 DOI: 10.1111/apha.12644] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/18/2015] [Accepted: 12/11/2015] [Indexed: 12/28/2022]
Abstract
Metformin, a synthetic dimethyl biguanide, has been in clinical use for over 55 years, and today is considered the first-choice drug for the treatment of type 2 diabetes used by an estimated 125 million people worldwide. Metformin is orally effective, not metabolized, excreted unchanged by the kidney, relatively free of side effects and well tolerated by the majority of patients. Of importance is that the United Kingdom Prospective Diabetes Study 20-year study of type 2 diabetics, completed in 1998, compared patients treated with insulin, sulfonylureas and metformin and concluded that metformin provided vascular protective actions. Cardiovascular disease is the primary basis for the high morbidity and mortality that is associated with diabetes and that metformin proved to be protective resulted in a dramatic increase in its use. The vascular protective actions of metformin are thought to be secondary to the antihyperglycaemic effects of metformin that are mediated via activation of AMP kinase and subsequent inhibition of hepatic gluconeogenesis, fatty acid oxidation as well as an insulin sensitizing action in striated muscle and adipose tissue. As reflected by a number of clinical studies, patients treated with metformin also have improvement in endothelial function as measured by the use of plethysmography and measurement of flow-mediated vasodilatation. These data as well as data from animal studies are supportive that metformin has a direct protective action on the vascular endothelium. In this review article, we discuss the pharmacology of metformin and critique the literature as to its cellular sites and mechanism(s) of action.
Collapse
Affiliation(s)
- C. R. Triggle
- Departments of Pharmacology and Medical Education; Weill Cornell Medicine in Qatar; Qatar Foundation, Education City; Doha Qatar
| | - H. Ding
- Departments of Pharmacology and Medical Education; Weill Cornell Medicine in Qatar; Qatar Foundation, Education City; Doha Qatar
| |
Collapse
|
47
|
Clemmer JS, Xiang L, Lu S, Mittwede PN, Hester RL. Hyperglycemia-Mediated Oxidative Stress Increases Pulmonary Vascular Permeability. Microcirculation 2016; 23:221-9. [PMID: 26749564 DOI: 10.1111/micc.12267] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 01/01/2016] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Hyperglycemia in diabetes mellitus is associated with endothelial dysfunction as evidenced by increased oxidative stress and vascular permeability. Whether impaired glucose control in metabolic syndrome impacts pulmonary vascular permeability is unknown. We hypothesized that in metabolic syndrome, hyperglycemia increases lung vascular permeability through superoxide. METHODS Lung capillary Kf and vascular superoxide were measured in the isolated lungs of LZ and OZ rats. OZ were subjected to 4 weeks of metformin treatment (300 mg/kg/day orally) to improve insulin sensitivity. In a separate experiment, lung vascular permeability and vascular superoxide were measured in LZ exposed to acute hyperglycemia (30 mM). RESULTS As compared to LZ, OZ had impaired glucose and insulin tolerance and elevated vascular superoxide which was associated with an elevated lung Kf. Chronic metformin treatment in OZ improved glucose control and insulin sensitivity which was associated with decreased vascular oxidative stress and lung Kf. Acute hyperglycemia in isolated lungs from LZ increased lung Kf, which was blocked with the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor, apocynin (3 mM). Apocynin also decreased baseline Kf in OZ. CONCLUSIONS These data suggest that hyperglycemia in metabolic syndrome exacerbates lung vascular permeability through increases in vascular superoxide, possibly through NADPH oxidase.
Collapse
Affiliation(s)
- John S Clemmer
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lusha Xiang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Silu Lu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Peter N Mittwede
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Robert L Hester
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
48
|
Abstract
The clustering of metabolic risk factors, overweight and hypertension is of particular importance in postmenopausal women due to the negative effect of menopause on bodyweight, glucose metabolism and the development of hypertension. Menopause acts directly as a risk factor by reducing the direct beneficial effect of ovarian hormones on cardiovascular functions, and indirectly by negatively influencing traditional risk factors for coronary artery disease. All changes occurring after the menopause must be regarded under a unifying mechanism that induces unfavorable changes in cardiovascular risk factors and vascular functions, which interact with each other, amplifying the effect of ovarian hormone deficiency and aging.
Collapse
Affiliation(s)
- Giuseppe MC Rosano
- Centre for Clinical and Basic Research, Department of Internal Medicine, IRCCS San Raffaele Hospital, Via Della Pisana 235, Roma 00163, Italy, Tel.: +39 065 225 2309; Fax: +39 065 225 2344
| | - Cristiana Vitale
- Centre for Clinical and Basic Research, Department of Internal Medicine, IRCCS San Raffaele Hospital, Via Della Pisana 235, Roma 00163, Italy, Tel.: +39 065 225 2309; Fax: +39 065 225 2344
| | | |
Collapse
|
49
|
Abstract
INTRODUCTION Cardiovascular disease remains the major contributor to morbidity and mortality in diabetes. From the need to reduce cardiovascular risk in diabetes and to ensure that such risk is not exacerbated by drug treatments, governmental regulators and drug manufacturers have focused on clinical trials evaluating cardiovascular outcomes. AREAS COVERED Findings from mechanistic and clinical trials of biguanides, sulfonylureas, thiazolidinediones, dipeptidyl peptidase-4 (DPP-4) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, and sodium-glucose co-transporter 2 (SGLT-2) inhibitors will be reviewed. These drug classes will be compared within the context of available cardiovascular outcomes data. Clinical implications of new study regulations will be examined. EXPERT OPINION Recent cardiovascular studies provide a more comprehensive evaluation of specific anti-diabetes therapy in individuals with high cardiovascular risk. Long-term effects of anti-hyperglycemic agents in patients with lower cardiovascular risk are still speculative. Historical data supports continued use of metformin as a first-line agent. DPP-4 inhibitors and GLP-1 receptor agonists appear to have neutral effects on cardiovascular outcomes. The significantly decreased cardiovascular risk associated with empagliflozin SGLT-2 inhibitor therapy is impressive and may change how practitioners prescribe add-on therapy to metformin.
Collapse
Affiliation(s)
- Lisa M Younk
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Elizabeth M Lamos
- b Division of Endocrinology, Diabetes and Nutrition , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Stephen N Davis
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| |
Collapse
|
50
|
Sapkota B, Subramanian A, Priamvada G, Finely H, Blackett PR, Aston CE, Sanghera DK. Association of APOE polymorphisms with diabetes and cardiometabolic risk factors and the role of APOE genotypes in response to anti-diabetic therapy: results from the AIDHS/SDS on a South Asian population. J Diabetes Complications 2015; 29:1191-7. [PMID: 26318958 PMCID: PMC4656127 DOI: 10.1016/j.jdiacomp.2015.07.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/24/2015] [Accepted: 07/29/2015] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND OBJECTIVES Apolipoprotein E (APOE) gene polymorphisms have been examined extensively in multiple global populations particularly due to their crucial role in lipid metabolism and cardiovascular disease. However, the overall contribution of APOE polymorphisms in type 2 diabetes (T2D) and coronary artery disease (CAD) in South Asians is still under-investigated. The objectives of this investigation were: 1) to evaluate the distribution of APOE polymorphisms in a large diabetic case-control sample from South Asia, 2) to examine the impact of APOE polymorphisms on quantitative risk factors of T2D and CAD, and 3) to explore the contribution of APOE genotypes in the response to anti-diabetic therapy. SUBJECTS AND METHODS A total of 3564 individuals (1956 T2D cases and 1608 controls) used in this study were part of the Asian Indian Diabetic Heart Study/Sikh Diabetes Study (AIDHS/SDS). We assessed the association of APOE polymorphisms with T2D, CAD and cardiometabolic traits using logistic and linear regression analysis. RESULTS AND CONCLUSIONS No significant differences in the distribution of APOE genotypes were observed between T2D and CAD cases and controls. The APOE4 genotype carriers had moderately higher diastolic blood pressure (BP) (p=0.022), and lower HDL-cholesterol (p=0.026) compared to E4 non-carriers. Overall, the APOE genotype was not a significant predictor of cardiometabolic disease in this population. Further stratification of data from diabetic patients by APOE genotypes and anti-hyperglycemic agents revealed a significant (~23%) decrease in 2-hour glucose (p=0.004) and ~7% decrease in systolic BP (p<0.001) among APOE4 carriers compared to non-carriers on metformin and sulphonylurea (SU) combination therapy, and no such differences were seen in patients on other agents. Our preliminary findings point to the need for evaluating population-specific genetic variation and its interactions with therapeutic effects.
Collapse
Affiliation(s)
- Bishwa Sapkota
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anuradha Subramanian
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gargi Priamvada
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Hadley Finely
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Piers R Blackett
- Department of Pediatrics, Section of Endocrinology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Christopher E Aston
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Dharambir K Sanghera
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|