1
|
Stone C, Sabe SA, Harris DD, Broadwin M, Kant RJ, Kanuparthy M, Abid MR, Sellke FW. Metformin Preconditioning Augments Cardiac Perfusion and Performance in a Large Animal Model of Chronic Coronary Artery Disease. Ann Surg 2024; 280:547-556. [PMID: 39041226 DOI: 10.1097/sla.0000000000006437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
OBJECTIVE To test the efficacy of metformin (MET) during the induction of coronary ischemia on myocardial performance in a large animal model of coronary artery disease (CAD) and metabolic syndrome (MS), with or without concomitant extracellular vesicular (EV) therapy. BACKGROUND Although surgical and endovascular revascularization are durably efficacious for many patients with CAD, up to one-third are poor candidates for standard therapies. For these patients, many of whom have comorbid MS, adjunctive strategies are needed. EV therapy has shown promise in this context, but its efficacy is attenuated by MS. We investigated whether MET pretreatment could ameliorate therapeutic decrements associated with MS. METHODS Yorkshire swine (n = 29) were provided a high-fat diet to induce MS, whereupon an ameroid constrictor was placed to induce CAD. Animals were initiated on 1000 mg oral MET or placebo; all then underwent repeat thoracotomy for intramyocardial injection of EVs or saline. Swine were maintained for 5 weeks before the acquisition of functional and perfusion data immediately before terminal myocardial harvest. Immunoblotting and immunofluorescence were performed on the most ischemic tissue from all groups. RESULTS Regardless of EV administration, animals that received MET exhibited significantly improved ejection fraction, cardiac index, and contractility at rest and during rapid myocardial pacing, improved perfusion to the most ischemic myocardial region at rest and during pacing, and markedly reduced apoptosis. CONCLUSIONS MET administration reduced apoptotic cell death, improved perfusion, and augmented both intrinsic and load-dependent myocardial performance in a highly translatable large animal model of chronic myocardial ischemia and MS.
Collapse
Affiliation(s)
- Christopher Stone
- Department of Surgery, Division of Cardiothoracic Surgery, The Warren Alpert Medical School, Brown University, Providence, RI
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Vanni E, Beauloye C, Horman S, Bertrand L. AMPK and O-GlcNAcylation: interplay in cardiac pathologies and heart failure. Essays Biochem 2024:EBC20240003. [PMID: 39319471 DOI: 10.1042/ebc20240003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024]
Abstract
Heart failure (HF) represents a multifaceted clinical syndrome characterized by the heart's inability to pump blood efficiently to meet the body's metabolic demands. Despite advances in medical management, HF remains a major cause of morbidity and mortality worldwide. In recent years, considerable attention has been directed toward understanding the molecular mechanisms underlying HF pathogenesis, with a particular focus on the role of AMP-activated protein kinase (AMPK) and protein O-GlcNAcylation. This review comprehensively examines the current understanding of AMPK and O-GlcNAcylation signalling pathways in HF, emphasizing their interplay and dysregulation. We delve into the intricate molecular mechanisms by which AMPK and O-GlcNAcylation contribute to cardiac energetics, metabolism, and remodelling, highlighting recent preclinical and clinical studies that have explored novel therapeutic interventions targeting these pathways.
Collapse
Affiliation(s)
- Ettore Vanni
- Pole of Cardiovascular Research, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Christophe Beauloye
- Pole of Cardiovascular Research, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
- Division of Cardiology, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Sandrine Horman
- Pole of Cardiovascular Research, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Luc Bertrand
- Pole of Cardiovascular Research, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| |
Collapse
|
3
|
Caturano A, Vetrano E, Galiero R, Sardu C, Rinaldi L, Russo V, Monda M, Marfella R, Sasso FC. Advances in the Insulin-Heart Axis: Current Therapies and Future Directions. Int J Mol Sci 2024; 25:10173. [PMID: 39337658 PMCID: PMC11432093 DOI: 10.3390/ijms251810173] [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: 09/03/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
The insulin-heart axis plays a pivotal role in the pathophysiology of cardiovascular disease (CVD) in insulin-resistant states, including type 2 diabetes mellitus. Insulin resistance disrupts glucose and lipid metabolism, leading to systemic inflammation, oxidative stress, and atherogenesis, which contribute to heart failure (HF) and other CVDs. This review was conducted by systematically searching PubMed, Scopus, and Web of Science databases for peer-reviewed studies published in the past decade, focusing on therapeutic interventions targeting the insulin-heart axis. Studies were selected based on their relevance to insulin resistance, cardiovascular outcomes, and the efficacy of pharmacologic treatments. Key findings from the review highlight the efficacy of lifestyle modifications, such as dietary changes and physical activity, which remain the cornerstone of managing insulin resistance and improving cardiovascular outcomes. Moreover, pharmacologic interventions, such as metformin, sodium-glucose cotransporter 2 inhibitors, glucagon-like peptide-1 receptor agonists, and dipeptidyl peptidase-4 inhibitors, have shown efficacy in reducing cardiovascular risk by addressing metabolic dysfunction, reducing inflammation, and improving endothelial function. Furthermore, emerging treatments, such as angiotensin receptor-neprilysin inhibitors, and mechanical interventions like ventricular assist devices offer new avenues for managing HF in insulin-resistant patients. The potential of these therapies to improve left ventricular ejection fraction and reverse pathological cardiac remodeling highlights the importance of early intervention. However, challenges remain in optimizing treatment regimens and understanding the long-term cardiovascular effects of these agents. Future research should focus on personalized approaches that integrate lifestyle and pharmacologic therapies to effectively target the insulin-heart axis and mitigate the burden of cardiovascular complications in insulin-resistant populations.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Luca Rinaldi
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86100 Campobasso, Italy
| | - Vincenzo Russo
- Department of Biology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| |
Collapse
|
4
|
Turkistani A, Al‐Kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, Bahaa MM, Al‐Windy S, Batiha GE. Pharmacological characterization of the antidiabetic drug metformin in atherosclerosis inhibition: A comprehensive insight. Immun Inflamm Dis 2024; 12:e1346. [PMID: 39092773 PMCID: PMC11295104 DOI: 10.1002/iid3.1346] [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: 11/16/2023] [Revised: 05/05/2024] [Accepted: 07/06/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Atherosclerosis (AS) is a progressive disease that interferes with blood flow, leading to cardiovascular complications such as hypertension, ischemic heart disease, ischemic stroke, and vascular ischemia. The progression of AS is correlated with inflammation, oxidative stress, and endothelial dysfunction. Various signaling pathways, like nuclear erythroid-related factor 2 (Nrf2) and Kruppel-like factor 2 (KLF2), are involved in the pathogenesis of AS. Nrf2 and KLF2 have anti-inflammatory and antioxidant properties. Thus, activation of these pathways may reduce the development of AS. Metformin, an insulin-sensitizing drug used in the management of type 2 diabetes mellitus (T2DM), increases the expression of Nrf2 and KLF2. AS is a common long-term macrovascular complication of T2DM. Thus, metformin, through its pleiotropic anti-inflammatory effect, may attenuate the development and progression of AS. AIMS Therefore, this review aims to investigate the possible role of metformin in AS concerning its effect on Nrf2 and KLF2 and inhibition of reactive oxygen species (ROS) formation. In addition to its antidiabetic effect, metformin can reduce cardiovascular morbidities and mortalities compared to other antidiabetic agents, even with similar blood glucose control by the Nrf2/KLF2 pathway activation. CONCLUSION In conclusion, metformin is an effective therapeutic strategy against the development and progression of AS, mainly through activation of the KLF2/Nrf2 axis.
Collapse
Affiliation(s)
- Areej Turkistani
- Department of Pharmacology and Toxicology, College of MedicineTaif UniversityTaifSaudi Arabia
| | - Haydar M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
- Department of Clinical Pharmacology and MedicineJabir ibn Hayyan Medical UniversityKufaIraq
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP MedWienAustria
- Department of Research & DevelopmentFunogenAthensGreece
- University Centre for Research & DevelopmentChandigarh UniversityPunjabIndia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐HerdeckeUniversity of Witten‐HerdeckeWuppertalGermany
| | - Mostafa M. Bahaa
- Pharmacy Practice Department, Faculty of PharmacyHorus UniversityNew DamiettaEgypt
| | - Salah Al‐Windy
- Department of Biology, College of ScienceBaghdad UniversityBaghdadIraq
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
5
|
Bailey CJ. Metformin: Therapeutic profile in the treatment of type 2 diabetes. Diabetes Obes Metab 2024; 26 Suppl 3:3-19. [PMID: 38784991 DOI: 10.1111/dom.15663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/02/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
Metformin (dimethyl-biguanide) can claim its origins in the use of Galega officinalis as a plant treatment for symptoms ascribed to diabetes. Since the first clinical use of metformin as a glucose-lowering agent in 1957, this medicine has emerged as a first-line pharmacological option to support lifestyle interventions in the management of type 2 diabetes (T2D). It acts through multiple cellular pathways, principally in the gut, liver and muscle, to counter insulin resistance and lower blood glucose without weight gain or risk of overt hypoglycaemia. Other effects include improvements in lipid metabolism, decreased inflammation and lower long-term cardiovascular risk. Metformin is conveniently combined with other diabetes medications, can be prescribed in prediabetes to reduce the risk of progression to T2D, and is used in some regions to assist glycaemic control in pregnancy. Consistent with its diversity of actions, established safety profile and cost-effectiveness, metformin is being assessed for further possible clinical applications. The use of metformin requires adequate renal function for drug elimination, and may cause initial gastrointestinal side effects, which can be moderated by taking with meals or using an extended-release formulation. Thus, metformin serves as a valuable therapeutic resource for use throughout the natural history of T2D.
Collapse
|
6
|
Bakhashab S, Barber R, O’Neill J, Arden C, Weaver JU. Overexpression of miR-199b-5p in Colony Forming Unit-Hill's Colonies Positively Mediates the Inflammatory Response in Subclinical Cardiovascular Disease Model: Metformin Therapy Attenuates Its Expression. Int J Mol Sci 2024; 25:8087. [PMID: 39125657 PMCID: PMC11311364 DOI: 10.3390/ijms25158087] [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: 06/25/2024] [Revised: 07/20/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Well-controlled type 1 diabetes (T1DM) is characterized by inflammation and endothelial dysfunction, thus constituting a suitable model of subclinical cardiovascular disease (CVD). miR-199b-5p overexpression in murine CVD has shown proatherosclerotic effects. We hypothesized that miR-199b-5p would be overexpressed in subclinical CVD yet downregulated following metformin therapy. Inflammatory and vascular markers were measured in 29 individuals with T1DM and 20 matched healthy controls (HCs). miR-199b-5p expression in CFU-Hill's colonies was analyzed from each study group, and correlations with inflammatory/vascular health indices were evaluated. Significant upregulation of miR-199b-5p was observed in T1DM, which was significantly downregulated by metformin. miR-199b-5p correlated positively with vascular endothelial growth factor-D and c-reactive protein (CRP: nonsignificant). ROC analysis determined miR-199b-5p to define subclinical CVD by discriminating between HCs and T1DM individuals. ROC analyses of HbA1c and CRP showed that the upregulation of miR-199b-5p in T1DM individuals defined subclinical CVD at HbA1c > 44.25 mmol and CRP > 4.35 × 106 pg/mL. Ingenuity pathway analysis predicted miR-199b-5p to inhibit the target genes SIRT1, ETS1, and JAG1. Metformin was predicted to downregulate miR-199b-5p via NFATC2 and STAT3 and reverse its downstream effects. This study validated the antiangiogenic properties of miR-199b-5p and substantiated miR-199b-5p overexpression as a biomarker of subclinical CVD. The downregulation of miR-199b-5p by metformin confirmed its cardio-protective effect.
Collapse
Affiliation(s)
- Sherin Bakhashab
- Biochemistry Department, King Abdulaziz University, P.O. Box 80218, Jeddah 21589, Saudi Arabia;
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK (J.O.)
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| | - Rosie Barber
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK (J.O.)
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Josie O’Neill
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK (J.O.)
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Catherine Arden
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Jolanta U. Weaver
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK (J.O.)
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle upon Tyne NE9 6SH, UK
- Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| |
Collapse
|
7
|
Wong C, Junqueira E, Poldiak NP, Crossley N, Jenkins S. Influence of Metformin Discontinuation on Readmission Rate in Patients With Acute Heart Failure. J Community Hosp Intern Med Perspect 2024; 14:12-17. [PMID: 39391108 PMCID: PMC11464055 DOI: 10.55729/2000-9666.1366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 10/12/2024] Open
Abstract
Background The consequences of discontinuing metformin in patients with heart failure have not been determined. Knowing that acute exacerbation of chronic heart failure contributes to substantial increases in major adverse cardiovascular events (MACE), we proposed a retrospective study to examine whether discontinuing metformin in patients hospitalized with heart failure impacts mortality and readmission rates. Methods We conducted a retrospective analysis of patients admitted with a diagnosis of acute heart failure to hospitals in the HCA Healthcare System from 2020 to 2022. Included patients had a prior diagnosis of diabetes mellitus, acute heart failure, and were taking metformin prior to admission. After applying our exclusion criteria, a total of 7740 patients remained. The primary outcomes were 30-, 60-, and 90-day readmission rates and secondary outcomes were mortality and length of stay. Results Patients who were discharged without a prescription for metformin (NONDIS-MET) were 4.489 (95% CI 3.673-5.488, p < 0.0001) times more likely to have a MACE outcome in 30 days compared to patients who received a discharge order for metformin (DIS-MET). The findings were similar for 60-day and 90-day readmission rates, with NONDIS-MET patients 3.457 (95% CI 2.893-4.131, p < 0.0001) and 2.992 (95% CI 2.534-3.533 p < 0.0001) times more likely to have a MACE outcome than MET patients, respectively. However, when metformin was continued during the patients' hospital stay (CONT-MET) there was no significant association with MACE outcomes, readmission, or mortality rates. Conclusion We found that diabetic patients admitted with acute heart failure exacerbations had a higher incidence of major adverse cardiac events and were more likely to be readmitted when they were not prescribed metformin after discharge. Our findings agree with prior work showing the cardioprotective effects of metformin; however, continuing metformin during hospital admission did not affect our patients adverse outcomes.
Collapse
Affiliation(s)
- Curtis Wong
- Trident Health, Internal Medicine Residency Program,
United States
| | | | | | - Nancy Crossley
- Trident Health, Internal Medicine Residency Program,
United States
| | - Shantae Jenkins
- Trident Health, Internal Medicine Residency Program,
United States
| |
Collapse
|
8
|
Zhong C, Bai J, Qu X, Liu Y, Li Z, Hao H, Qiao S, Zhang Z, Xu X, Si J, Xu W, Xu B, Kang L. Metformin reduces new-onset atrial fibrillation risk rather than atrial fibrillation burden in type 2 diabetes patients: A case-control study. Heliyon 2024; 10:e30992. [PMID: 38818187 PMCID: PMC11137400 DOI: 10.1016/j.heliyon.2024.e30992] [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: 09/05/2023] [Revised: 04/19/2024] [Accepted: 05/09/2024] [Indexed: 06/01/2024] Open
Abstract
Background The effects of metformin on atrial fibrillation (AF) in type 2 diabetes patients remain unclear. We aimed to explore the effects of metformin on AF, including new-onset AF and AF burden, in type 2 diabetes patients with pacemakers. Methods and results This retrospective study included a total of 227 patients. Based on the presence of paroxysmal AF, the patients were divided into a paroxysmal AF group (n = 80) and a non-AF group (n = 147). In the non-AF group, a significant association was observed between metformin use and a lower risk of new-onset AF in multivariable Cox hazards models (hazard ratio [HR]: 0.36; 95 % confidence interval [CI]: 0.14-0.91; p = 0.0311*) when adjusted for age, sex, body mass index (BMI), drinking, smoking, left atrial dimension, creatinine, complications, and drugs. In the paroxysmal AF group, univariable analysis indicated no association between the AF burden and metformin use (p = 0.817). Furthermore, when adjusted for metformin use, age, sex, BMI, drinking, smoking, cardiovascular disease, myocardial infarction, heart failure, stroke, and ejection fraction in multivariable Cox hazards models, we found a lower proportion of major adverse cardiovascular events (MACEs) both in the total (HR: 0.28; 95 % CI: 0.1-0.82; p = 0.0202*) and the non-AF group (HR: 0.19; 95 % CI: 0.05-0.79; p = 0.0223*) compared to that in the AF group (HR: 0.31; 95 % CI: 0.02-4.41; p = 0.3879). Conclusion In type 2 diabetes patients with pacemakers, metformin reduced the probability of new-onset AF instead of addressing the AF burden. Furthermore, metformin therapy decreased the incidence of MACEs in type 2 diabetes patients without AF.
Collapse
Affiliation(s)
- Chongxia Zhong
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Jian Bai
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xinhong Qu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yihai Liu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Zhu Li
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Han Hao
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Shiyang Qiao
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Zhe Zhang
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical School of Nanjing Medical University, Nanjing, China
| | - Xiaoying Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical School of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Jiayi Si
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical School of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Wei Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, China
| | - Lina Kang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, China
| |
Collapse
|
9
|
Yu Y, Ge X, Cao L, Li F. Diagnostic and Prognostic Value of Plasma lncRNA SRA1 in Chronic Heart Failure. Rev Cardiovasc Med 2024; 25:178. [PMID: 39076490 PMCID: PMC11267213 DOI: 10.31083/j.rcm2505178] [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: 11/03/2023] [Revised: 12/17/2023] [Accepted: 12/22/2023] [Indexed: 07/31/2024] Open
Abstract
Background The pathogenesis and development of chronic heart failure (CHF) may involve long non-coding ribonucleic acid (lncRNA) steroid receptor RNA activator 1 (SRA1), a known cardiomyopathy risk factor and regulator of cardiac myofibroblast activation. This study aimed to investigate the application of SRA1 in the early detection and prediction of CHF. Methods SRA1 plasma expression was determined in CHF patients and healthy individuals/using real time-quantitative polymerase chain reaction (RT-qPCR). The diagnostic and prognostic value of SRA1 was assessed using receiver operating curve (ROC) and Cox regression analyses. Results Compared with the healthy controls, the patients with CHF had increased brain natriuretic peptide (BNP) levels, left atrial end-systolic diameter (LAD), left ventricular end-diastolic diameter (LVDd), and decreased left ventricular ejection fraction (LVEF). SRA1 was significantly upregulated in CHF patients as well as positively correlated with BNP level, LAD, and LVDd, and negatively correlated with LVEF. SRA1 could sensitively discriminate CHF patients from healthy individuals and was an independent predictor of adverse event-free survival in CHF patients. Conclusions Upregulated plasma SRA1 can discriminate patients with CHF from healthy individuals and predict adverse outcomes in CHF patients. Thus, SRA1 is a potential molecular indicator for monitoring chronic heart failure development.
Collapse
Affiliation(s)
- Yiming Yu
- Department of General Practice, The First Affiliated Hospital of Weifang Medical University, Weifang People's Hospital, 261041 Weifang, Shandong, China
| | - Xiao Ge
- Department of General Practice, The First Affiliated Hospital of Weifang Medical University, Weifang People's Hospital, 261041 Weifang, Shandong, China
| | - Lifang Cao
- Department of General Practice, The First Affiliated Hospital of Weifang Medical University, Weifang People's Hospital, 261041 Weifang, Shandong, China
| | - Feng Li
- Department of General Practice, The First Affiliated Hospital of Weifang Medical University, Weifang People's Hospital, 261041 Weifang, Shandong, China
| |
Collapse
|
10
|
García-Díez E, Pérez-Jiménez J, Martín MÁ, Ramos S. (-)-Epicatechin and colonic metabolite 2,3-dihydroxybenzoic acid, alone or in combination with metformin, protect cardiomyocytes from high glucose/high palmitic acid-induced damage by regulating redox status, apoptosis and autophagy. Food Funct 2024; 15:2536-2549. [PMID: 38347828 DOI: 10.1039/d3fo04039a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
(-)-Epicatechin (EC) and a main colonic phenolic acid derived from flavonoid intake, 2,3-dihydroxybenzoic acid (DHBA), display antioxidant and antidiabetic activities. Diabetic cardiomyopathy (DCM) is one of the main causes of mortality in patients with diabetes, lacking a suitable treatment. Hyperglycaemia and dyslipidaemia are mainly responsible for oxidative stress and altered apoptosis and autophagy in cardiomyocytes during DCM. In this context, phenolic compounds could be suitable candidates for alleviating DCM, but have scarcely been investigated or their use in combination with antidiabetic drugs. This study evaluates the effects of EC, DHBA and antidiabetic drug metformin (MET), alone or all combined (MIX), on redox status, autophagy and apoptosis in H9c2 cardiomyocytes challenged with high concentrations of glucose (HG) and palmitic acid (PA). Under HG + PA conditions, EC, DHBA, MET and MIX equally improved redox status, reduced apoptosis induction and ameliorated autophagy inhibition. Mechanistically, all treatments alleviated HG + PA-induced oxidative stress by reinforcing antioxidant defences (∼40% increase in glutathione, ∼30% diminution in GPx activity and ∼15% increase in SOD activity) and reducing ROS generation (∼20%), protein oxidation (∼35%) and JNK phosphorylation (∼200%). Additionally, all treatments mitigated HG + PA-induced apoptosis and activated autophagy by decreasing Bax (∼15-25%), caspase-3 (∼20-40%) and p62 (∼20-40%), and increasing Bcl-2, beclin-1 and LC3-II/LC3-I (∼40-60%, ∼15-20%, and ∼25-30%, respectively). JNK inhibition improved protective changes to redox status, apoptosis and autophagy that were observed in EC-, DHBA- and MIX-mediated protection. Despite no additive or synergistic effects being detected when phenolic compounds and MET were combined, these results provide the first evidence for the benefits of EC and DHBA, comparable to those of MET alone, to ameliorate cardiomyocyte damage, that involve an improvement in antioxidant competence, autophagy and apoptosis, these effects being mediated at least by targeting JNK.
Collapse
Affiliation(s)
- Esther García-Díez
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), José Antonio Novais 10, Ciudad Universitaria, 28040, Madrid, Spain.
| | - Jara Pérez-Jiménez
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), José Antonio Novais 10, Ciudad Universitaria, 28040, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - María Ángeles Martín
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), José Antonio Novais 10, Ciudad Universitaria, 28040, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Sonia Ramos
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), José Antonio Novais 10, Ciudad Universitaria, 28040, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| |
Collapse
|
11
|
Xu J, Qin G. Nomogram for Predicting the Risk of Short Sleep Duration in Myocardial Infarction Survivors. Rev Cardiovasc Med 2024; 25:77. [PMID: 39076948 PMCID: PMC11263857 DOI: 10.31083/j.rcm2503077] [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: 07/20/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 07/31/2024] Open
Abstract
Background Research on post-infarction insomnia, particularly short sleep duration following myocardial infarction (MI), remains limited. Currently, there are no existing guidelines or risk prediction models to assist physicians in managing or preventing short sleep duration or insomnia following MI. This study aims to develop a nomogram for predicting the risk of short sleep duration after MI. Methods We conducted a retrospective study on 1434 MI survivors aged 20 and above, utilizing data from the National Health and Nutrition Examination Survey (NHANES) database spanning from 2007 to 2018. Among them, 710 patients were assigned to the training group, while 707 patients were allocated to the testing group. We utilized logistic regression, least absolute shrinkage and selection operator (LASSO) regression, and the elastic network for variable selection. The stability and accuracy of the prediction model were assessed using receiver operator characteristics (ROCs) and calibration curves. Results We included five variables in the nomogram: age, poverty income ratio (PIR), body mass index (BMI), race, and depression. The ROC curves yielded values of 0.636 for the training group and 0.657 for the testing group, demonstrating the model's good prediction accuracy and robustness through a calibration curve test. Conclusions Our nomogram can effectively predict the likelihood of short sleep duration in MI survivors, providing valuable support for clinicians in preventing and managing post-MI short sleep duration.
Collapse
Affiliation(s)
- Jun Xu
- First School of Clinical Medicine, Shanxi Medical University, 030000 Taiyuan, Shanxi, China
| | - Gang Qin
- Department of Cardiology, First Hospital of Shanxi Medical University, 030000 Taiyuan, Shanxi, China
| |
Collapse
|
12
|
Zhang T, Xu L, Guo X, Tao H, Liu Y, Liu X, Zhang Y, Meng X. The potential of herbal drugs to treat heart failure: The roles of Sirt1/AMPK. J Pharm Anal 2024; 14:157-176. [PMID: 38464786 PMCID: PMC10921247 DOI: 10.1016/j.jpha.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/09/2023] [Accepted: 09/05/2023] [Indexed: 03/12/2024] Open
Abstract
Heart failure (HF) is a highly morbid syndrome that seriously affects the physical and mental health of patients and generates an enormous socio-economic burden. In addition to cardiac myocyte oxidative stress and apoptosis, which are considered mechanisms for the development of HF, alterations in cardiac energy metabolism and pathological autophagy also contribute to cardiac abnormalities and ultimately HF. Silent information regulator 1 (Sirt1) and adenosine monophosphate-activated protein kinase (AMPK) are nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases and phosphorylated kinases, respectively. They play similar roles in regulating some pathological processes of the heart through regulating targets such as peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), protein 38 mitogen-activated protein kinase (p38 MAPK), peroxisome proliferator-activated receptors (PPARs), and mammalian target of rapamycin (mTOR). We summarized the synergistic effects of Sirt1 and AMPK in the heart, and listed the traditional Chinese medicine (TCM) that exhibit cardioprotective properties by modulating the Sirt1/AMPK pathway, to provide a basis for the development of Sirt1/AMPK activators or inhibitors for the treatment of HF and other cardiovascular diseases (CVDs).
Collapse
Affiliation(s)
- Tao Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lei Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiaowei Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Honglin Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yue Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianfeng Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, Sichuan, 620032, China
| |
Collapse
|
13
|
Wen J, Chen C. From Energy Metabolic Change to Precision Therapy: a Holistic View of Energy Metabolism in Heart Failure. J Cardiovasc Transl Res 2024; 17:56-70. [PMID: 37450209 DOI: 10.1007/s12265-023-10412-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Heart failure (HF) is a complex and multifactorial disease that affects millions of people worldwide. It is characterized by metabolic disturbances of substrates such as glucose, fatty acids (FAs), ketone bodies, and amino acids, which lead to changes in cardiac energy metabolism pathways. These metabolic alterations can directly or indirectly promote myocardial remodeling, thereby accelerating the progression of HF, resulting in a vicious cycle of worsening symptoms, and contributing to the increased hospitalization and mortality among patients with HF. In this review, we summarized the latest researches on energy metabolic profiling in HF and provided the related translational therapeutic strategies for this devastating disease. By taking a holistic approach to understanding energy metabolism changes in HF, we hope to provide comprehensive insights into the pathophysiology of this challenging condition and identify novel precise targets for the development of more effective treatments.
Collapse
Affiliation(s)
- Jianpei Wen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| |
Collapse
|
14
|
Chun KH, Oh J, Lee CJ, Park JJ, Lee SE, Kim MS, Cho HJ, Choi JO, Lee HY, Hwang KK, Kim KH, Yoo BS, Choi DJ, Baek SH, Jeon ES, Kim JJ, Cho MC, Chae SC, Oh BH, Kang SM. Metformin treatment is associated with improved survival in diabetic patients hospitalized with acute heart failure: A prospective observational study using the Korean acute heart failure registry data. DIABETES & METABOLISM 2024; 50:101504. [PMID: 38097010 DOI: 10.1016/j.diabet.2023.101504] [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: 09/10/2023] [Revised: 11/24/2023] [Accepted: 12/10/2023] [Indexed: 12/18/2023]
Abstract
AIMS Although the hypothesis that metformin is beneficial for patients with diabetes and heart failure (HF) has been steadily raised, there is limited data on metformin use in patients with acute HF. We analyzed the association of metformin on all-cause mortality in hospitalized patients with type 2 diabetes and acute HF. METHODS The Korean Acute Heart Failure registry prospectively enrolled patients hospitalized for acute HF from 2011 to 2014. Among this cohort, we analyzed patients with diabetes with baseline estimated glomerular filtration rate (eGFR) of 30 ml/min/1.73 m2 or more. We analyzed the all-cause mortality and re-hospitalization for HF within 1 year after discharge. Inverse probability treatment weighting method was used to adjust baseline differences on metformin treatment. RESULTS The study analyzed data from 1,309 patients with HF and diabetes (mean age 69 years, 56 % male). Among them, 613 (47 %) patients were on metformin at admission. During the median follow-up period of 11 months, 132 (19 %) and 74 (12 %) patients not receiving and receiving metformin treatment died, respectively. The mortality rate was lower in metformin users than in non-users (hazard ratio 0.616 [0.464-0.819] P<0.001). After adjustment, metformin was significantly associated with a lower risk for the mortality (hazard ratio 0.677 [0.495-0.928] P=0.015). In subgroup analyses, this association remains significant irrespective of baseline kidney function (eGFR <60 or ≥60 ml/min/1.73 m2, P-for-interaction=0.176) or left ventricular ejection fraction (<40 %, 40-49 %, or ≥50 %, P-for-interaction=0.224). CONCLUSIONS Metformin treatment at the time of admission was associated with a lower risk for 1-year mortality in patients with diabetes, hospitalized for acute HF.
Collapse
Affiliation(s)
- Kyeong-Hyeon Chun
- Division of Cardiology, National Health Insurance Service Ilsan Hospital, Goyang, Republic of Korea
| | - Jaewon Oh
- Cardiology Division, Severance Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chan Joo Lee
- Cardiology Division, Severance Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Joo Park
- Division of Cardiology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sang Eun Lee
- Division of Cardiology, Asan Medical Center, Seoul, Republic of Korea
| | - Min-Seok Kim
- Division of Cardiology, Asan Medical Center, Seoul, Republic of Korea
| | - Hyun-Jai Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jin-Oh Choi
- Department of Internal Medicine, Sungkyunkwan University College of Medicine, Seoul, Republic of Korea
| | - Hae-Young Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyung-Kuk Hwang
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Kye Hun Kim
- Department of Internal Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Byung-Su Yoo
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Dong-Ju Choi
- Division of Cardiology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sang Hong Baek
- Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun-Seok Jeon
- Department of Internal Medicine, Sungkyunkwan University College of Medicine, Seoul, Republic of Korea
| | - Jae-Joong Kim
- Division of Cardiology, Asan Medical Center, Seoul, Republic of Korea
| | - Myeong-Chan Cho
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Shung Chull Chae
- Department of Internal Medicine, Kyungpook National University College of Medicine, Daegu, Republic of Korea
| | - Byung-Hee Oh
- Department of Internal Medicine, Mediplex Sejong Hospital, Incheon, Republic of Korea
| | - Seok-Min Kang
- Cardiology Division, Severance Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Sabbar R, Kadhim SAA, Fawzi HA, Flayih A, Mohammad B, Swadi A. The impact of empagliflozin and metformin on cardiac parameters in patients with mid-range ejection fraction heart failure without diabetes. J Med Life 2024; 17:57-62. [PMID: 38737651 PMCID: PMC11080507 DOI: 10.25122/jml-2023-0340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/17/2023] [Indexed: 05/14/2024] Open
Abstract
Heart failure (HF) remains a significant problem for healthcare systems, requiring the use of intervention and multimodal management strategies. We aimed to assess the short-term effect of empagliflozin (EMPA) and metformin on cardiac function parameters, including ventricular dimension-hypertrophy, septal thickness, ejection fraction (EF), and N-terminal pro-brain natriuretic peptide (NT-proBNP) levels in patients with HF and mildly reduced EF. A case-control study included 60 newly diagnosed patients with HF. Patients were divided into two groups: Group E received standard HF treatment (carvedilol, bumetanide, sacubitril-valsartan, spironolactone) plus EMPA 10 mg daily, and Group M received standard HF treatment plus metformin 500 mg daily. After three months of treatment, Group E had a significantly higher EF than Group M compared to initial measurements (a change of 9.2% versus 6.1%, respectively). We found similar results in the left ventricular end-systolic dimension (LVESD), with mean reductions of 0.72 mm for Group E and 0.23 mm for Group M. Regarding cardiac indicators, the level of NT-proBNP was considerably decreased in both groups. However, the reduction was significantly greater in group E than in group M compared to the initial level (mean reduction: 719.9 vs. 973.6, respectively). When combined with quadruple anti-heart failure therapy, metformin enhanced several echocardiographic parameters, showing effects similar to those of EMPA when used in the same treatment regimen. However, the benefits of EMPA were more pronounced, particularly regarding improvements in EF and LVESD.
Collapse
Affiliation(s)
- Reeman Sabbar
- Department of Pharmacology, College of Medicine, University of Al-Qadisiyah, Al-Qadisiyah, Iraq
| | - Sinaa Abdul Amir Kadhim
- Department of Pharmacology, College of Medicine, University of Al-Qadisiyah, Al-Qadisiyah, Iraq
| | | | - Ali Flayih
- Department of Pharmacology, College of Medicine, University of Al-Qadisiyah, Al-Qadisiyah, Iraq
| | - Bassim Mohammad
- Department of Pharmacology, College of Medicine, University of Al-Qadisiyah, Al-Qadisiyah, Iraq
| | - Asma Swadi
- Department of Pharmacology, College of Medicine, University of Al-Qadisiyah, Al-Qadisiyah, Iraq
| |
Collapse
|
16
|
Chakraborty P, Po SS, Scherlag BJ, Dasari TW. The neurometabolic axis: A novel therapeutic target in heart failure. Life Sci 2023; 333:122122. [PMID: 37774940 DOI: 10.1016/j.lfs.2023.122122] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
Abnormal cardiac metabolism or cardiac metabolic remodeling is reported before the onset of heart failure with reduced ejection fraction (HFrEF) and is known to trigger and maintain the mechanical dysfunction and electrical, and structural abnormalities of the ventricle. A dysregulated cardiac autonomic tone characterized by sympathetic overdrive with blunted parasympathetic activation is another pathophysiological hallmark of HF. Emerging evidence suggests a link between autonomic nervous system activity and cardiac metabolism. Chronic β-adrenergic activation promotes maladaptive metabolic remodeling whereas cholinergic activation attenuates the metabolic aberrations through favorable modulation of key metabolic regulatory molecules. Restoration of sympathovagal balance by neuromodulation strategies is emerging as a novel nonpharmacological treatment strategy in HF. The current review attempts to evaluate the 'neuro-metabolic axis' in HFrEF and whether neuromodulation can mitigate the adverse metabolic remodeling in HFrEF.
Collapse
Affiliation(s)
- Praloy Chakraborty
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sunny S Po
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Benjamin J Scherlag
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tarun W Dasari
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
17
|
Povar-Echeverría M, Méndez-Bailón M, Martín-Sánchez FJ, Montero-Pérez-Barquero M, Trullàs JC, Miró Ò. Prognostic impact of metformin in patients with type 2 diabetes mellitus and acute heart failure: Combined analysis of the EAHFE and RICA registries. Rev Clin Esp 2023; 223:542-551. [PMID: 37717921 DOI: 10.1016/j.rceng.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/29/2023] [Indexed: 09/19/2023]
Abstract
INTRODUCTION Patients with diabetes mellitus (DM) and heart failure (HF) have a worse prognosis despite therapeutic advances in both diseases. Sodium-glucose co-transporter type 2 and GLP-1 receptor agonists have shown cardiovascular benefits and they have been positioned as the first step in the treatment of DM in patients with HF or high cardiovascular risk. However, in the pivotal trials the majority of patients receive concomitant treatment with metformin. Randomized clinical trials have not yet been developed to assess the prognostic impact of metformin at the cardiovascular level. Our objective has been centered in analyzing whether patients with DM and acute HF who receive treatment with metformin at the time of discharge may have a better prognosis at one year of follow-up. METHODS Prospective cohort trial using the combined analysis of the two main Spanish HF registries, the EAHFE Registry (Epidemiology of Acute Heart Failure in Emergency Departments) and the RICA (National Registry of Patients with Heart Failure). RESULTS 33% (1453) of a total of 4403 patients with DM type 2 received treatment with metformin. This group presents significantly lower mortality after one year of treatment (22 versus 32%; Log Rank test P < 0.001). In the adjusted analysis of mortality, patients receiving treatment with metformin have lower mortality at one year of follow-up regardless of the rest of the variables (RR 0,814; 95%IC 0,712-0,930; P < 0.01). CONCLUSIONS Patients with DM type 2 and acute HF who receive metformin have a better prognosis after one year of follow-up, so we believe that this drug should continue to be a fundamental pillar in the treatment of these patients.
Collapse
Affiliation(s)
- M Povar-Echeverría
- Internal Medicine Service, Hospital Comarcal de Barbastro, Barbastro, Huesca, Spain.
| | - M Méndez-Bailón
- Internal Medicine Service, Hospital Universitario Clínico San Carlos, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IDISS), Madrid, Spain
| | - F J Martín-Sánchez
- Internal Medicine Service, Hospital Universitario Clínico San Carlos, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IDISS), Madrid, Spain
| | - M Montero-Pérez-Barquero
- Internal Medicine Service, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Córdoba, Spain
| | - J C Trullàs
- Internal Medicine Service, Hospital d'Olot i Comarcal de la Garrotxa, Olot, Girona, Spain; Grupo de Investigación en Reparación y Regeneración Q2 Tisular (TR2Lab), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central, Barcelona, Spain
| | - Ò Miró
- Emergencies Service, Hospital Clínic de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
18
|
Fazio S, Mercurio V, Affuso F, Bellavite P. The Negative Impact of Insulin Resistance/Hyperinsulinemia on Chronic Heart Failure and the Potential Benefits of Its Screening and Treatment. Biomedicines 2023; 11:2928. [PMID: 38001929 PMCID: PMC10669553 DOI: 10.3390/biomedicines11112928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
This opinion article highlights the potential alterations caused by insulin resistance and hyperinsulinemia on the cardiovascular system and their negative impact on heart failure (HF), and describes the potential benefits of an early screening with consequent prompt treatment. HF is the final event of several different cardiovascular diseases. Its incidence has been increasing over the last decades because of increased survival from ischemic heart disease thanks to improvements in its treatment (including myocardial revascularization interventions) and the increase in life span. In particular, incidence of HF with preserved ejection fraction (HFpEF) is significantly increasing, and patients with HFpEF often are also affected by diabetes mellitus and insulin resistance (IR), with a prevalence > 45%. Concentric left ventricular (LV) remodeling and diastolic dysfunction are the main structural abnormalities that characterize HFpEF. It is well documented in the literature that IR with chronic hyperinsulinemia, besides causing type 2 diabetes mellitus, can cause numerous cardiovascular alterations, including endothelial dysfunction and increased wall thicknesses of the left ventricle with concentric remodeling and diastolic dysfunction. Therefore, it is conceivable that IR might play a major role in the pathophysiology and the progressive worsening of HF. To date, several substances have been shown to reduce IR/hyperinsulinemia and have beneficial clinical effects in patients with HF, including SGLT2 inhibitors, metformin, and berberine. For this reason, an early screening of IR could be advisable in subjects at risk and in patients with heart failure, to promptly intervene with appropriate therapy. Future studies aimed at comparing the efficacy of the substances used both alone and in association are needed.
Collapse
Affiliation(s)
- Serafino Fazio
- Department of Internal Medicine, University of Naples Federico II, 80138 Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy;
| | | | | |
Collapse
|
19
|
Yoon JP, Park SJ, Kim DH, Chung SW. Metformin increases the expression of proinflammatory cytokines and inhibits supraspinatus fatty infiltration. J Orthop Surg Res 2023; 18:674. [PMID: 37700364 PMCID: PMC10496168 DOI: 10.1186/s13018-023-04163-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND After a rotator cuff (RC) tendon tear, the supraspinatus (SS) inflammatory response induces fatty infiltration (FI). Metformin has the effect of regulating the initial inflammatory response of atrophic muscles. Therefore, this study aimed to investigate the effect of metformin use on modulating the expression of proinflammatory cytokines and SS FI in an acute RC tear rat model. METHODS This study used 26 male Sprague-Dawley rats. Animals were randomly divided into two groups: The metformin group received metformin for 5 days after cutting the RC tendon, and the control group was administered only with saline after cutting the tendon. Metformin 50 mg/kg was intraperitoneally injected for 5 days. Three rats in each group were sacrificed 5 days after SS tendon rupture surgery, and 10 rats in each group were sacrificed 14 days after surgery. The SS was sampled 5 days after SS tendon tear surgery, and the expression of proinflammatory cytokines was measured by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). On day 14 after sampling, histological analysis of the SS was performed using hematoxylin and eosin, Masson's trichrome, and picrosirius red staining. RESULTS On day 5 of surgery, the expression values of interferon gamma (increased 7.2-fold, P < .01), tumor necrosis factor alpha (increased 13-fold, P < .05), interleukin-1β (increased 4.7-fold, P < .001), and interleukin-6 (increased 4.6-fold, P < .01) increased significantly in the metformin group compared with those in the control group. As a result of Oil Red O staining, SS FI was significantly suppressed in the metformin group compared with that in the control group (metformin group, 305 ± 50.3 µm2, P < .001; control group, 3136 ± 662.8 µm2, P < .001). In addition, the SS volume of the metformin group was not reduced compared with those of the control group, and the morphology and structure of the SS were better preserved. CONCLUSIONS The results of this study revealed that metformin can increase the expression of proinflammatory cytokines and suppress SS fat infiltration in delayed sutures.
Collapse
Affiliation(s)
- Jong Pil Yoon
- Department of Orthopaedic Surgery, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Sung-Jin Park
- Department of Orthopaedic Surgery, School of Medicine, Kyungpook National University, Daegu, Korea.
| | - Dong-Hyun Kim
- Department of Orthopaedic Surgery, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Seok Won Chung
- Department of Orthopaedic Surgery, School of Medicine, Konkuk University Medical Center, Seoul, Korea
| |
Collapse
|
20
|
Conza D, Mirra P, Fiory F, Insabato L, Nicolò A, Beguinot F, Ulianich L. Metformin: A New Inhibitor of the Wnt Signaling Pathway in Cancer. Cells 2023; 12:2182. [PMID: 37681914 PMCID: PMC10486775 DOI: 10.3390/cells12172182] [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: 07/19/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 09/09/2023] Open
Abstract
The biguanide drug metformin is widely used in type 2 diabetes mellitus therapy, due to its ability to decrease serum glucose levels, mainly by reducing hepatic gluconeogenesis and glycogenolysis. A considerable number of studies have shown that metformin, besides its antidiabetic action, can improve other disease states, such as polycystic ovary disease, acute kidney injury, neurological disorders, cognitive impairment and renal damage. In addition, metformin is well known to suppress the growth and progression of different types of cancer cells both in vitro and in vivo. Accordingly, several epidemiological studies suggest that metformin is capable of lowering cancer risk and reducing the rate of cancer deaths among diabetic patients. The antitumoral effects of metformin have been proposed to be mainly mediated by the activation of the AMP-activated protein kinase (AMPK). However, a number of signaling pathways, both dependent and independent of AMPK activation, have been reported to be involved in metformin antitumoral action. Among these, the Wingless and Int signaling pathway have recently been included. Here, we will focus our attention on the main molecular mechanisms involved.
Collapse
Affiliation(s)
- Domenico Conza
- URT Genomics of Diabetes, Institute of Endocrinology and Experimental Oncology, National Research Council & Department of Translational Medicine, University of Naples “Federico II”, 80131 Naples, Italy; (D.C.); (P.M.); (F.F.); (A.N.); (F.B.)
| | - Paola Mirra
- URT Genomics of Diabetes, Institute of Endocrinology and Experimental Oncology, National Research Council & Department of Translational Medicine, University of Naples “Federico II”, 80131 Naples, Italy; (D.C.); (P.M.); (F.F.); (A.N.); (F.B.)
| | - Francesca Fiory
- URT Genomics of Diabetes, Institute of Endocrinology and Experimental Oncology, National Research Council & Department of Translational Medicine, University of Naples “Federico II”, 80131 Naples, Italy; (D.C.); (P.M.); (F.F.); (A.N.); (F.B.)
| | - Luigi Insabato
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Antonella Nicolò
- URT Genomics of Diabetes, Institute of Endocrinology and Experimental Oncology, National Research Council & Department of Translational Medicine, University of Naples “Federico II”, 80131 Naples, Italy; (D.C.); (P.M.); (F.F.); (A.N.); (F.B.)
| | - Francesco Beguinot
- URT Genomics of Diabetes, Institute of Endocrinology and Experimental Oncology, National Research Council & Department of Translational Medicine, University of Naples “Federico II”, 80131 Naples, Italy; (D.C.); (P.M.); (F.F.); (A.N.); (F.B.)
| | - Luca Ulianich
- URT Genomics of Diabetes, Institute of Endocrinology and Experimental Oncology, National Research Council & Department of Translational Medicine, University of Naples “Federico II”, 80131 Naples, Italy; (D.C.); (P.M.); (F.F.); (A.N.); (F.B.)
| |
Collapse
|
21
|
Cimmino G, Natale F, Alfieri R, Cante L, Covino S, Franzese R, Limatola M, Marotta L, Molinari R, Mollo N, Loffredo FS, Golino P. Non-Conventional Risk Factors: "Fact" or "Fake" in Cardiovascular Disease Prevention? Biomedicines 2023; 11:2353. [PMID: 37760794 PMCID: PMC10525401 DOI: 10.3390/biomedicines11092353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiovascular diseases (CVDs), such as arterial hypertension, myocardial infarction, stroke, heart failure, atrial fibrillation, etc., still represent the main cause of morbidity and mortality worldwide. They significantly modify the patients' quality of life with a tremendous economic impact. It is well established that cardiovascular risk factors increase the probability of fatal and non-fatal cardiac events. These risk factors are classified into modifiable (smoking, arterial hypertension, hypercholesterolemia, low HDL cholesterol, diabetes, excessive alcohol consumption, high-fat and high-calorie diet, reduced physical activity) and non-modifiable (sex, age, family history, of previous cardiovascular disease). Hence, CVD prevention is based on early identification and management of modifiable risk factors whose impact on the CV outcome is now performed by the use of CV risk assessment models, such as the Framingham Risk Score, Pooled Cohort Equations, or the SCORE2. However, in recent years, emerging, non-traditional factors (metabolic and non-metabolic) seem to significantly affect this assessment. In this article, we aim at defining these emerging factors and describe the potential mechanisms by which they might contribute to the development of CVD.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Cardiology Unit, Azienda Ospedaliera Universitaria Luigi Vanvitelli, 80138 Naples, Italy
| | - Francesco Natale
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Roberta Alfieri
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Luigi Cante
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Simona Covino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Rosa Franzese
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Mirella Limatola
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Luigi Marotta
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Riccardo Molinari
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Noemi Mollo
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Francesco S Loffredo
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Paolo Golino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| |
Collapse
|
22
|
Caturano A, D’Angelo M, Mormone A, Russo V, Mollica MP, Salvatore T, Galiero R, Rinaldi L, Vetrano E, Marfella R, Monda M, Giordano A, Sasso FC. Oxidative Stress in Type 2 Diabetes: Impacts from Pathogenesis to Lifestyle Modifications. Curr Issues Mol Biol 2023; 45:6651-6666. [PMID: 37623239 PMCID: PMC10453126 DOI: 10.3390/cimb45080420] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
Oxidative stress is a critical factor in the pathogenesis and progression of diabetes and its associated complications. The imbalance between reactive oxygen species (ROS) production and the body's antioxidant defence mechanisms leads to cellular damage and dysfunction. In diabetes, chronic hyperglycaemia and mitochondrial dysfunction contribute to increased ROS production, further exacerbating oxidative stress. This oxidative burden adversely affects various aspects of diabetes, including impaired beta-cell function and insulin resistance, leading to disrupted glucose regulation. Additionally, oxidative stress-induced damage to blood vessels and impaired endothelial function contribute to the development of diabetic vascular complications such as retinopathy, nephropathy, and cardiovascular diseases. Moreover, organs and tissues throughout the body, including the kidneys, nerves, and eyes, are vulnerable to oxidative stress, resulting in diabetic nephropathy, neuropathy, and retinopathy. Strategies to mitigate oxidative stress in diabetes include antioxidant therapy, lifestyle modifications, and effective management of hyperglycaemia. However, further research is necessary to comprehensively understand the underlying mechanisms of oxidative stress in diabetes and to evaluate the efficacy of antioxidant interventions in preventing and treating diabetic complications. By addressing oxidative stress, it might be possible to alleviate the burden of diabetes and improve patient outcomes.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy; (M.D.)
| | - Margherita D’Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy; (M.D.)
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Andrea Mormone
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
| | - Vincenzo Russo
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, I-80134 Naples, Italy
| | - Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy; (M.D.)
| | - Antonio Giordano
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
| |
Collapse
|
23
|
Gorący A, Rosik J, Szostak J, Szostak B, Retfiński S, Machaj F, Pawlik A. Improving mitochondrial function in preclinical models of heart failure: therapeutic targets for future clinical therapies? Expert Opin Ther Targets 2023; 27:593-608. [PMID: 37477241 DOI: 10.1080/14728222.2023.2240021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/19/2023] [Indexed: 07/22/2023]
Abstract
INTRODUCTION Heart failure is a complex clinical syndrome resulting from the unsuccessful compensation of symptoms of myocardial damage. Mitochondrial dysfunction is a process that occurs because of an attempt to adapt to the disruption of metabolic and energetic pathways occurring in the myocardium. This, in turn, leads to further dysfunction in cardiomyocyte processes. Currently, many therapeutic strategies have been implemented to improve mitochondrial function, but their effectiveness varies widely. AREAS COVERED This review focuses on new models of therapeutic strategies targeting mitochondrial function in the treatment of heart failure. EXPERT OPINION Therapeutic strategies targeting mitochondria appear to be a valuable option for treating heart failure. Currently, the greatest challenge is to develop new research models that could restore the disrupted metabolic processes in mitochondria as comprehensively as possible. Only the development of therapies that focus on improving as many dysregulated mitochondrial processes as possible in patients with heart failure will be able to bring the expected clinical improvement, along with inhibition of disease progression. Combined strategies involving the reduction of the effects of oxidative stress and mitochondrial dysfunction, appear to be a promising possibility for developing new therapies for a complex and multifactorial disease such as heart failure.
Collapse
Affiliation(s)
- Anna Gorący
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin, Poland
| | - Jakub Rosik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Joanna Szostak
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Szymon Retfiński
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Filip Machaj
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
- Department of Medical Biology, Medical University of Warsaw, Warsaw, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
24
|
Chen Y, Zhu S, Lin Z, Zhang Y, Jin C, He S, Chen X, Zhou X. Metformin alleviates ethanol-induced cardiomyocyte injury by activating AKT/Nrf2 signaling in an ErbB2-dependent manner. Mol Biol Rep 2023; 50:3469-3478. [PMID: 36765018 DOI: 10.1007/s11033-023-08310-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/26/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND Metformin, a first-line oral anti-diabetic drug, has recently been reported to exert protective effect on various cardiovascular diseases. However, the potential role of metformin in ethanol-induced cardiomyocyte injury is still unknown. Therefore, this study was aimed to investigate the effect of metformin on ethanol-induced cardiomyocyte injury and its underlying mechanism. METHODS AND RESULTS H9c2 cardiomyocytes were exposed to ethanol for 24 h to establish an ethanol-induced cardiomyocyte injury model, and followed by treatment with metformin in the presence or absence of Lapatinib (an ErbB2 inhibition). CCK8 and LDH assays demonstrated that metformin improved cell viability in cardiomyocytes exposed to ethanol. Furthermore, metformin suppressed cardiomyocyte apoptosis and reduced the expressions of apoptosis-related proteins (Bax and C-CAS-3). In addition, our results showed that metformin activated the AKT/Nrf2 pathway, and then promoted Nrf2 nuclear translocation and the transcription of its downstream antioxidant genes (HO-1, CAT and SOD2), thereby inhibiting oxidative stress. Interestingly, we found that ErbB2 protein expression was significantly inhibited in ethanol-treated cardiomyocytes, which was markedly reversed by metformin. In contrast, Lapatinib largely abrogated the activation of AKT/Nrf2 signaling by metformin, accompanied by the increases in oxidative stress and cardiomyocyte apoptosis, indicating that metformin prevented ethanol-induced cardiomyocyte injury in an ErbB2-dependent manner. CONCLUSION In summary, our study provides the first evidence that metformin protects cardiomyocyte against ethanol-induced oxidative stress and apoptosis by activating ErbB2-mediated AKT/Nrf2 signaling. Thus, metformin may be a potential novel treatment approach for alcoholic cardiomyopathy.
Collapse
Affiliation(s)
- Yunjie Chen
- Department of Pharmacy, Ningbo first Hospital, 315010, Ningbo, People's Republic of China.,Central Laboratory of the Medical Research Center, Ningbo First Hospital, 315010, Ningbo, People's Republic of China
| | - Suyan Zhu
- Department of Pharmacy, Ningbo first Hospital, 315010, Ningbo, People's Republic of China
| | - Zhu Lin
- Department of Pharmacy, Ningbo first Hospital, 315010, Ningbo, People's Republic of China
| | - Yuanbin Zhang
- Department of Pharmacy, Ningbo first Hospital, 315010, Ningbo, People's Republic of China.,Central Laboratory of the Medical Research Center, Ningbo First Hospital, 315010, Ningbo, People's Republic of China
| | - Cheng Jin
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Shengqu He
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Xueqin Chen
- Department of Traditional Chinese Medicine, Ningbo First Hospital, 315010, Ningbo, People's Republic of China.
| | - Xuan Zhou
- Department of Pharmacy, Ningbo first Hospital, 315010, Ningbo, People's Republic of China.
| |
Collapse
|
25
|
Metformin Directly Binds to MMP-9 to Improve Plaque Stability. J Cardiovasc Dev Dis 2023; 10:jcdd10020054. [PMID: 36826550 PMCID: PMC9962015 DOI: 10.3390/jcdd10020054] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/19/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Vulnerable atherosclerotic plaque rupture is the principal mechanism that accounts for myocardial infarction and stroke. High matrix metalloproteinase-9 (MMP-9) expression and activity have been proven to lead to plaque instability. Metformin, a first-line treatment for type 2 diabetes, is beneficial to plaque vulnerability. However, the mechanism underlying its anti-atherogenic effect remains unclear. Molecular docking and surface plasmon resonance experiments showed that metformin directly interacts with MMP-9, and incubated MMP-9 overexpressing HEK293A cells with metformin (1 μmol·L-1) significantly attenuates MMP-9's activity using zymography and MMP activity assays. Moreover, metformin treatment drives MMP-9 degradation. Next, we constructed a carotid artery atherosclerotic plaque model and administered consecutive 14-day metformin (200 mg·kg-1·d-1) treatment by intragastric gavage. Immunofluorescence staining of the right carotid common artery and serum MMP activity assay results showed that metformin treatment decreased local plaque MMP-9 protein level and circulating MMP-9 activity, respectively. Histochemical staining revealed that after metformin treatment, the collagen content in plaque was significantly preserved, and the plaque vulnerability index decreased. These findings suggested that metformin improved atherosclerotic plaque stability by directly binding to MMP-9 and driving its degradation.
Collapse
|
26
|
Shati AA, Maarouf A, Dawood AF, Bayoumy NM, Alqahtani YA, A. Eid R, Alqahtani SM, Abd Ellatif M, Al-Ani B, Albawardi A. Lower Extremity Arterial Disease in Type 2 Diabetes Mellitus: Metformin Inhibits Femoral Artery Ultrastructural Alterations as well as Vascular Tissue Levels of AGEs/ET-1 Axis-Mediated Inflammation and Modulation of Vascular iNOS and eNOS Expression. Biomedicines 2023; 11:biomedicines11020361. [PMID: 36830898 PMCID: PMC9953164 DOI: 10.3390/biomedicines11020361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 01/28/2023] Open
Abstract
Lower extremity arterial disease (LEAD) is a major risk factor for amputation in diabetic patients. The advanced glycation end products (AGEs)/endothelin-1 (ET-1)/nitric oxide synthase (NOS) axis-mediated femoral artery injury with and without metformin has not been previously investigated. Type 2 diabetes mellitus (T2DM) was established in rats, with another group of rats treated for two weeks with 200 mg/kg metformin, before being induced with T2DM. The latter cohort were continued on metformin until they were sacrificed at week 12. Femoral artery injury was established in the diabetic group as demonstrated by substantial alterations to the femoral artery ultrastructure, which importantly were ameliorated by metformin. In addition, diabetes caused a significant (p < 0.0001) upregulation of vascular tissue levels of AGEs, ET-1, and iNOS, as well as high blood levels of glycated haemoglobin, TNF-α, and dyslipidemia. All of these parameters were also significantly inhibited by metformin. Moreover, metformin treatment augmented arterial eNOS expression which had been inhibited by diabetes progression. Furthermore, a significant correlation was observed between femoral artery endothelial tissue damage and glycemia, AGEs, ET-1, TNF-α, and dyslipidemia. Thus, in a rat model of T2DM-induced LEAD, an association between femoral artery tissue damage and the AGEs/ET-1/inflammation/NOS/dyslipidemia axis was demonstrated, with metformin treatment demonstrating beneficial vascular protective effects.
Collapse
Affiliation(s)
- Ayed A. Shati
- Department of Child Health, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Amro Maarouf
- Department of Clinical Biochemistry, Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B9 5SS, UK
| | - Amal F. Dawood
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Nervana M. Bayoumy
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Youssef A. Alqahtani
- Department of Child Health, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Refaat A. Eid
- Department of Pathology, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Saeed M. Alqahtani
- Department of Surgery, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Mohamed Abd Ellatif
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
- Department of Medical Biochemistry, College of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Bahjat Al-Ani
- Department of Physiology, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Alia Albawardi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates
- Correspondence:
| |
Collapse
|
27
|
Ziqubu K, Mazibuko-Mbeje SE, Mthembu SXH, Mabhida SE, Jack BU, Nyambuya TM, Nkambule BB, Basson AK, Tiano L, Dludla PV. Anti-Obesity Effects of Metformin: A Scoping Review Evaluating the Feasibility of Brown Adipose Tissue as a Therapeutic Target. Int J Mol Sci 2023; 24:ijms24032227. [PMID: 36768561 PMCID: PMC9917329 DOI: 10.3390/ijms24032227] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Brown adipose tissue (BAT) is increasingly recognized as the major therapeutic target to promote energy expenditure and ameliorate diverse metabolic complications. There is a general interest in understanding the pleiotropic effects of metformin against metabolic complications. Major electronic databases and search engines such as PubMed/MEDLINE, Google Scholar, and the Cochrane library were used to retrieve and critically discuss evidence reporting on the impact of metformin on regulating BAT thermogenic activity to ameliorate complications linked with obesity. The summarized evidence suggests that metformin can reduce body weight, enhance insulin sensitivity, and improve glucose metabolism by promoting BAT thermogenic activity in preclinical models of obesity. Notably, this anti-diabetic agent can affect the expression of major thermogenic transcriptional factors such as uncoupling protein 1 (UCP1), nuclear respiratory factor 1 (NRF1), and peroxisome-proliferator-activated receptor gamma coactivator 1-alpha (PGC1-α) to improve BAT mitochondrial function and promote energy expenditure. Interestingly, vital molecular markers involved in glucose metabolism and energy regulation such as AMP-activated protein kinase (AMPK) and fibroblast growth factor 21 (FGF21) are similarly upregulated by metformin treatment in preclinical models of obesity. The current review also discusses the clinical relevance of BAT and thermogenesis as therapeutic targets. This review explored critical components including effective dosage and appropriate intervention period, consistent with the beneficial effects of metformin against obesity-associated complications.
Collapse
Affiliation(s)
- Khanyisani Ziqubu
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
| | - Sithandiwe E. Mazibuko-Mbeje
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
- Correspondence: (S.E.M.-M.); (P.V.D.); Tel.: +27-21-938-0333 (P.V.D.)
| | - Sinenhlanhla X. H. Mthembu
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
| | - Sihle E. Mabhida
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Babalwa U. Jack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Tawanda M. Nyambuya
- Department of Health Sciences, Namibia University of Science and Technology, Windhoek 9000, Namibia
| | - Bongani B. Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Albertus K. Basson
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3880, South Africa
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Phiwayinkosi V. Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3880, South Africa
- Correspondence: (S.E.M.-M.); (P.V.D.); Tel.: +27-21-938-0333 (P.V.D.)
| |
Collapse
|
28
|
Abstract
BACKGROUND Metformin has good anti-hyperglycemic effectiveness, but does not induce hypoglycemia,is very safe, and has become the preferred drug for the treatment of type 2 diabetes. Recently, the other effects of metformin, such as being anti-inflammatory and delaying aging, have also attracted increased attention. METHODS AND RESULTS The relevant literatures on pubmed and other websites for reading, classification and sorting, and did not involve any animal experiments. CONCLUSION Metformin has anti-inflammatory effects through multiple routes, which provides potential therapeutic targets for certain inflammatory diseases, such as neuroinflammation and rheumatoid arthritis. In addition, inflammation is a key component of tumor occurrence and development ; thus, targeted inflammatory intervention is a significant benefit for both cancer prevention and treatment. Therefore, metformin may have further potential for inflammation-related disease prevention and treatmen. However, the inflammatory mechanism is complex; various molecules are connected and influence each other. For example, metformin significantly inhibits p65 nuclear translocation, but pretreatment with compound C, an AMPK inhibitor, abolishes this effect, and silencing of HMGB1 inhibits NF-κB activation . SIRT1 deacetylates FoxO, increasing its transcriptional activity . mTOR in dendritic cells regulates FoxO1 via AKT. The interactions among various molecules should be further explored to clarify their specific mechanisms and provide more direction for the treatment of inflammatory diseases, as well as cancer.
Collapse
|
29
|
Hou Y, Hou X, Nie Q, Xia Q, Hu R, Yang X, Song G, Ren L. Association of Bone Turnover Markers with Type 2 Diabetes Mellitus and Microvascular Complications: A Matched Case-Control Study. Diabetes Metab Syndr Obes 2023; 16:1177-1192. [PMID: 37139349 PMCID: PMC10149773 DOI: 10.2147/dmso.s400285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/12/2023] [Indexed: 05/05/2023] Open
Abstract
Purpose The aim of this study was to evaluate the association of bone turnover markers (BTMs) with type 2 diabetes mellitus (T2DM) and microvascular complications. Methods A total of 166 T2DM patients and 166 non-diabetic controls matched by gender and age were enrolled. T2DM patients were sub-classified into groups based on whether they had diabetic peripheral neuropathy (DPN), diabetic retinopathy (DR), and diabetic kidney disease (DKD). Clinical data including demographic characteristics and blood test results [serum levels of osteocalcin (OC), N-terminal propeptide of type 1 procollagen (P1NP), and β-crosslaps (β-CTX)] were collected. Logistic regression and restrictive cubic spline curves were performed to examine the association of BTMs with the risk of T2DM and microvascular complications. Results After adjusting for family history of diabetes, sex and age, an inverse association was observed between elevated serum OC levels [O, p < 0.001] and increased serum P1NP levels , p < 0.001] with the risk of T2DM. Moreover, there was an inverse linear association of serum OC and P1NP levels with the risk of T2DM. However, β-CTX was not associated with T2DM. Further analysis showed a nonlinear association between OC and the risk of DR, while P1NP and β-CTX were not correlated with DR. Serum concentrations of BTMs were not associated with the risks of DPN and DKD. Conclusion Serum OC and P1NP levels were negatively correlated with T2DM risk. Particularly, serum OC levels were associated with DR risk. Given that BTMs are widely used as markers of bone remodeling, the present finding provides a new perspective for estimating the risk of diabetic microvascular complications.
Collapse
Affiliation(s)
- Yilin Hou
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Xiaoyu Hou
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Qian Nie
- Health Examination Center, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Qiuyang Xia
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Rui Hu
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Xiaoyue Yang
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Guangyao Song
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
- Correspondence: Guangyao Song; Luping Ren, Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China, Email ;
| | - Luping Ren
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
| |
Collapse
|
30
|
Kiss A, Nadasy GL, Fees A, Arnold Z, Aykac I, Dostal C, Szabó GT, Szabó PL, Szekeres M, Pokreisz P, Hunyady L, Podesser BK. Alterations in Coronary Resistance Artery Network Geometry in Diabetes and the Role of Tenascin C. Rev Cardiovasc Med 2023; 24:6. [PMID: 39076867 PMCID: PMC11270457 DOI: 10.31083/j.rcm2401006] [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: 07/18/2022] [Revised: 11/01/2022] [Accepted: 11/11/2022] [Indexed: 07/31/2024] Open
Abstract
Background Geometrical alterations in the coronary resistance artery network and the potential involvement of Tenascin C (TNC) extracellular matrix protein were investigated in diabetic and control mice. Methods Diabetes was induced by streptozotocin (STZ) injections (n = 7-11 animals in each group) in Tenascin C KO (TNC KO) mice and their Wild type (A/J) littermates. After 16-18 weeks the heart was removed and the whole subsurface network of the left coronary artery was prepared (down to branches of 40 μ m outer diameter), in situ pressure-perfused and studied using video-microscopy. Outer and inner diameters, wall thicknesses and bifurcation angles were measured on whole network pictures reconstructed into collages at 1.7 μ m pixel resolutions. Results Diabetes induced abnormal morphological alterations including trifurcations, sharp bends of larger branches, and branches directed retrogradely (p < 0.001 by the χ 2 test). Networks of TNC KO mice tended to form early divisions producing parallelly running larger branches (p < 0.001 by the χ 2 probe). Networks of coronary resistance arteries were substantially more abundant in 100-180 μ m components, appearing in 2-5 mm flow distance from orifice in diabetes. This was accompanied by thickening of the wall of larger arterioles ( > 220 μ m) and thinning of the wall of smaller (100-140 μ m) arterioles (p < 0.001). Blood flow should cover larger distances in diabetic networks, but interestingly STZ-induced diabetes did not generate further geometrical changes in TNC KO mice. Conclusions Diabetes promotes hypertrophic and hypotrophic vascular remodeling and induces vasculogenesis at well defined, specific positions of the coronary vasculature. TNC plays a pivotal role in the formation of coronary network geometry, and TNC deletion causes parallel fragmentation preventing diabetes-induced abnormal vascular morphologies.
Collapse
Affiliation(s)
- Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Gyorgy L Nadasy
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1094 Budapest, Hungary
| | | | - Zsuzsanna Arnold
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Ibrahim Aykac
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Christopher Dostal
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Gábor T Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Petra Lujza Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Maria Szekeres
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 1088 Budapest, Hungary
| | - Peter Pokreisz
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Laszlo Hunyady
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1094 Budapest, Hungary
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
31
|
Metformin Acutely Mitigates Oxidative Stress in Human Atrial Tissue: A Pilot Study in Overweight Non-Diabetic Cardiac Patients. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122058. [PMID: 36556423 PMCID: PMC9785172 DOI: 10.3390/life12122058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Metformin, the first-line drug in type 2 diabetes mellitus, elicits cardiovascular protection also in obese patients via pleiotropic effects, among which the anti-oxidant is one of the most investigated. The aim of the present study was to assess whether metformin can acutely mitigate oxidative stress in atrial tissue harvested from overweight non-diabetic patients. Right atrial appendage samples were harvested during open-heart surgery and used for the evaluation of reactive oxygen species (ROS) production by means of confocal microscopy (superoxide anion) and spectrophotometry (hydrogen peroxide). Experiments were performed after acute incubation with metformin (10 µM) in the presence vs. absence of angiotensin II (AII, 100 nM), lipopolysaccharide (LPS, 1 μg/mL), and high glucose (Gluc, 400 mg/dL). Stimulation with AII, LPS, and high Gluc increased ROS production. The magnitude of oxidative stress correlated with several echocardiographic parameters. Metformin applied in the lowest therapeutic concentration (10 µM) was able to decrease ROS generation in stimulated but also non-stimulated atrial samples. In conclusion, in a pilot group of overweight non-diabetic cardiac patients, acute incubation with metformin at a clinically relevant dose alleviated oxidative stress both in basal conditions and conditions that mimicked the activation of the renin-angiotensin-aldosterone system, acute inflammation, and uncontrolled hyperglycemia.
Collapse
|
32
|
Song BW, Kim S, Kim R, Jeong S, Moon H, Kim H, Vasileva EA, Mishchenko NP, Fedoreyev SA, Stonik VA, Lee MY, Kim J, Kim HK, Han J, Chang W. Regulation of Inflammation-Mediated Endothelial to Mesenchymal Transition with Echinochrome a for Improving Myocardial Dysfunction. Mar Drugs 2022; 20:756. [PMID: 36547903 PMCID: PMC9781361 DOI: 10.3390/md20120756] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is a process by which endothelial cells (ECs) transition into mesenchymal cells (e.g., myofibroblasts and smooth muscle cells) and induce fibrosis of cells/tissues, due to ischemic conditions in the heart. Previously, we reported that echinochrome A (EchA) derived from sea urchin shells can modulate cardiovascular disease by promoting anti-inflammatory and antioxidant activity; however, the mechanism underlying these effects was unclear. We investigated the role of EchA in the EndMT process by treating human umbilical vein ECs (HUVECs) with TGF-β2 and IL-1β, and confirmed the regulation of cell migration, inflammatory, oxidative responses and mitochondrial dysfunction. Moreover, we developed an EndMT-induced myocardial infarction (MI) model to investigate the effect of EchA in vivo. After EchA was administered once a day for a total of 3 days, the histological and functional improvement of the myocardium was investigated to confirm the control of the EndMT. We concluded that EchA negatively regulates early or inflammation-related EndMT and reduces the myofibroblast proportion and fibrosis area, meaning that it may be a potential therapy for cardiac regeneration or cardioprotection from scar formation and cardiac fibrosis due to tissue granulation. Our findings encourage the study of marine bioactive compounds for the discovery of new therapeutics for recovering ischemic cardiac injuries.
Collapse
Affiliation(s)
- Byeong-Wook Song
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea
| | - Sejin Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea
| | - Ran Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea
| | - Seongtae Jeong
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea
| | - Hanbyeol Moon
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea
| | - Hojin Kim
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea
| | - Elena A. Vasileva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia
| | - Natalia P. Mishchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia
| | - Sergey A. Fedoreyev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia
| | - Valentin A. Stonik
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women’s University, Seoul 04310, Republic of Korea
| | - Hyoung Kyu Kim
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutic Center, Inje University, Busan 47392, Republic of Korea
| | - Jin Han
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutic Center, Inje University, Busan 47392, Republic of Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
33
|
Katsiki N, Kazakos K, Triposkiadis F. Contemporary choice of glucose lowering agents in heart failure patients with type 2 diabetes. Expert Opin Pharmacother 2022; 23:1957-1974. [DOI: 10.1080/14656566.2022.2143263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - Kyriakos Kazakos
- Nursing Department, International Hellenic University, Thessaloniki, Greece
| | | |
Collapse
|
34
|
Sun B, Gao Y, He F, Liu Z, Zhou J, Wang X, Zhang W. Association of visit-to-visit HbA1c variability with cardiovascular diseases in type 2 diabetes within or outside the target range of HbA1c. Front Public Health 2022; 10:1052485. [PMID: 36438253 PMCID: PMC9686379 DOI: 10.3389/fpubh.2022.1052485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Background Although a growing attention has been recently paid to the role of HbA1c variability in the risk of diabetic complications, the impact of HbA1c variability on cardiovascular diseases (CVD) in type 2 diabetes is still debated. The aim of the study is to investigate the association of HbA1c variability with CVD in individuals within or outside the target range of HbA1c. Methods Using data from Action in Diabetes and Vascular disease: preterAx and diamicroN-MR Controlled Evaluation (ADVANCE), we enrolled 855 patients with type 2 diabetes in China. The primary outcomes included major macrovascular events and major microvascular events. Visit-to-visit HbA1c variability was expressed as the coefficient of variation (CV) of five measurements of HbA1c taken 3-24 months after treatment. Cox proportional hazard models were used to estimate adjusted hazard ratios (aHR). Results Among 855 patients in the intensive glucose treatment group, 563 and 292 patients were assigned to the group of "within the target range of HbA1c" (WTH) (updated mean HbA1c ≤ 7.0%) and "outside the target range of HbA1c" (OTH) (updated mean HbA1c > 7.0%), respectively. HbA1c variability was positively associated with the risk of major microvascular events in all patients and both the subgroups during a median follow-up period of 4.8 years. Particularly, the risk related to HbA1c variability was higher in patients in WTH group for the new or worsening nephropathy [aHR: 3.35; 95% confidence interval (CI): 1.05-10.74; P = 0.042]. Conclusions This retrospective cohort study confirmed the positive correlation between HbA1c variability and major microvascular events, especially in subjects in WTH or OTH.
Collapse
Affiliation(s)
- Bao Sun
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China,Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Fazhong He
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Jiecan Zhou
- The First Affiliated Hospital, Clinical Medical Research Center, Hengyang Medical School, University of South China, Hengyang, China,The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, China,Jiecan Zhou
| | - Xingyu Wang
- Beijing Hypertension League Institute, Beijing, China,Xingyu Wang
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China,National Clinical Research Center for Geriatric Disorders, Changsha, China,*Correspondence: Wei Zhang
| |
Collapse
|
35
|
Brand KMG, Schlachter J, Foch C, Boutmy E. Quality and Characteristics of 4241 Case Reports of Lactic Acidosis in Metformin Users Reported to a Large Pharmacovigilance Database. Ther Clin Risk Manag 2022; 18:1037-1047. [PMID: 36389204 PMCID: PMC9642855 DOI: 10.2147/tcrm.s372430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/11/2022] [Indexed: 11/05/2022] Open
Abstract
Objective Metformin-associated lactic acidosis (MaLA) occurs rarely and is thus difficult to study. We analysed 4241 individual case safety reports of lactic acidosis (LA) that implicated metformin as a suspected drug reported to the pharmacovigilance database of Merck KGaA, Darmstadt, Germany. The primary objective was to review reports for quality and completeness of data to support diagnoses of MaLA. We also explored the correlations between reported biomarkers, and associations between biomarkers and outcomes. Research Design and Methods Records were analysed for completeness in supporting diagnoses of LA or metformin-associated LA (MaLA), against commonly used diagnostic criteria. Correlations between indices of exposure to metformin and biomarkers of LA and mortality were investigated. Results Missing data was common, especially for plasma metformin. Clinical/biomarker evidence supported a diagnosis of LA in only 33% of cases (LA subpopulation) and of MaLA in only 9% (MaLA subpopulation). The metformin plasma level correlated weakly with plasma lactate (positive) and pH (negative). About one-fifth (21.9%) of cases reported a fatal outcome. Metformin exposure (plasma level or dose) was not associated with increased mortality risk (there was a suggestion of decreased risk at higher levels of exposure to metformin). Plasma lactate was the only variable associated with increased risk of mortality. Examination of concomitant risk factors for MaLA identified renal dysfunction (including of iatrogenic origin) as a potential driver of mortality in this population. Conclusion Despite the high frequency of missing data, this is the largest analysis of cases of MaLA supported by measurements of circulating metformin, and lactate, and pH, to date. Plasma lactate, and not metformin dose or plasma level, appeared to be the main driver of mortality in the setting of LA or MaLA. Further research with more complete case reports is required.
Collapse
Affiliation(s)
- Kerstin M G Brand
- Global Medical Affairs, Merck Healthcare KGaA, Darmstadt, Germany
- Correspondence: Kerstin MG Brand, Global Medical Affairs, Merck Healthcare KGaA, F135/00_N1, Frankfurter Str. 250, Darmstadt, 64293, Germany, Tel +49 6151 72 2301, Email
| | | | - Caroline Foch
- Global Epidemiology, Merck Healthcare KGaA, Darmstadt, Germany
| | | |
Collapse
|
36
|
Association of Diet-Related Systemic Inflammation with Periodontitis and Tooth Loss: The Interaction Effect of Diabetes. Nutrients 2022; 14:nu14194118. [PMID: 36235769 PMCID: PMC9572370 DOI: 10.3390/nu14194118] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Diet is an important factor that can affect inflammatory processes. Diet-related systemic inflammation is closely linked to periodontitis and tooth loss. However, the role that systemic conditions play in influencing this association remains unclear. A cross-sectional analysis was conducted using the National Health and Nutrition Examination Survey (NHANES) from 2009 to 2014. Diet-related systemic inflammation was assessed by the Dietary Inflammatory Index (DII). Multivariate Cox regression models were used to investigate the association between DII and periodontal results, including total periodontitis, tooth loss, severe tooth loss, and the number of teeth lost. The interaction effects between DII and established covariates were tested. Higher DII scores, corresponding to a higher pro-inflammatory potential of the diet, were associated with an increased risk of periodontitis and tooth loss among the 10,096 eligible participants. There was an interaction between diabetes and DII on total periodontitis (p = 0.0136). No significant interaction effect was detected between DII and other established covariates. Participants who consumed an anti-inflammatory diet, and did not have diabetes, experienced the lowest risks of periodontitis and tooth loss. However, in the context of diabetes, the efficacy of such a diet may be weakened or even eliminated. Dietary interventions to manage oral health problems may need to take the individual's metabolic condition into account.
Collapse
|
37
|
Kamel AM, Sabry N, Farid S. Effect of metformin on left ventricular mass and functional parameters in non-diabetic patients: a meta-analysis of randomized clinical trials. BMC Cardiovasc Disord 2022; 22:405. [PMID: 36088302 PMCID: PMC9464374 DOI: 10.1186/s12872-022-02845-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Left ventricular hypertrophy is a common finding in patients with ischemic heart disease and is associated with mortality in patients with cardiovascular disease (CVD). Metformin, an antidiabetic drug, has been shown to reduce oxidative stress and left ventricular mass index (LVMI) in animal hypertrophy models. We summarized evidence regarding the effect of metformin on LVMI and LVEF. METHODS Electronic databases were searched for randomized clinical trials (RCTs) that used metformin in non-diabetic patients with or without pre-existing CVD. The standardized mean change using change score standardization (SMCC) was calculated for each study. The random-effects model was used to pool the SMCC across studies. Meta-regression analysis was used to assess the association of heart failure (HF), metformin dose, and duration with the SMCC. RESULTS Data synthesis from nine RCTs (754 patients) showed that metformin use resulted in higher reduction in LVMI after 12 months (SMCC = -0.63, 95% CI - 1.23; - 0.04, p = 0.04) and an overall higher reduction in LVMI (SMCC = -0.5, 95% CI - 0.84; - 0.16, p < 0.01). These values equate to absolute values of 11.3 (95% CI 22.1-0.72) and 8.97 (95% CI 15.06-2.87) g/m2, respectively. The overall improvement in LVEF was also higher in metformin users after excluding one outlier (SMCC = 0.26, 95% CI 0.03-0.49, P = 0.03) which translates to a higher absolute improvement of 2.99% (95% CI 0.34; 5.63). Subgroup analysis revealed a favorable effect for metformin on LVEF in patients who received > 1000 mg/day (SMCC = 0.28, 95% CI 0.04; 0.52, P = 0.04), and patients with HF (SMCC = 0.23; 95% CI 0.1; 0.36; P = 0.004). These values translate to a higher increase of 2.64% and 3.21%, respectively. CONCLUSION Results suggest a favorable effect for metformin on LVMI and LVEF in patients with or without pre-existing CVD. Additional trials are needed to address the long-term effect of metformin. Registration The study was registered on the PROSPERO database with the registration number CRD42021239368 ( https://www.crd.york.ac.uk/prospero ).
Collapse
Affiliation(s)
- Ahmed M Kamel
- Clinical Pharmacy Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Nirmeen Sabry
- Clinical Pharmacy Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Samar Farid
- Clinical Pharmacy Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
38
|
Phowira J, Ahmed FW, Bakhashab S, Weaver JU. Upregulated miR-18a-5p in Colony Forming Unit-Hill’s in Subclinical Cardiovascular Disease and Metformin Therapy; MERIT Study. Biomedicines 2022; 10:biomedicines10092136. [PMID: 36140236 PMCID: PMC9496122 DOI: 10.3390/biomedicines10092136] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Colony forming unit-Hill’s (CFU-Hill’s) colonies are hematopoietic-derived cells that participate in neovasculogenesis and serve as a biomarker for vascular health. In animals, overexpression of miR-18a-5p was shown to be pro-atherogenic. We had shown that well-controlled type 1 diabetes mellitus (T1DM) is characterized by an inflammatory state, endothelial dysfunction, and reduced number of CFU-Hill’s, a model of subclinical cardiovascular disease (CVD). MERIT study explored the role of miR-18a-5p expression in CFU-Hill’s colonies in T1DM, and the cardioprotective effect of metformin in subclinical CVD. In T1DM, miR-18a-5p was significantly upregulated whereas metformin reduced it to HC levels. MiR-18a-5p was inversely correlated with CFU-Hill’s colonies, CD34+, CD34+CD133+ cells, and positively with IL-10, C-reactive protein, vascular endothelial growth factor-D (VEGF-D), and thrombomodulin. The receiver operating characteristic curve demonstrated, miR-18a-5p as a biomarker of T1DM, and upregulated miR-18a-5p defining subclinical CVD at HbA1c of 44.5 mmol/mol (pre-diabetes). Ingenuity pathway analysis documented miR-18a-5p inhibiting mRNA expression of insulin-like growth factor-1, estrogen receptor-1, hypoxia-inducible factor-1α cellular communication network factor-2, and protein inhibitor of activated STAT 3, whilst metformin upregulated these mRNAs via transforming growth factor beta-1 and VEGF. We confirmed the pro-atherogenic effect of miR-18a-5p in subclinical CVD and identified several target genes for future CVD therapies.
Collapse
Affiliation(s)
- Jason Phowira
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Fahad W. Ahmed
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle upon Tyne NE9 6SH, UK
- Department of Medical Oncology, King Faisal Specialist Hospital and Research Centre, Madinah 42522, Saudi Arabia
| | - Sherin Bakhashab
- Biochemistry Department, King Abdulaziz University, P.O. Box 80218, Jeddah 21589, Saudi Arabia
| | - Jolanta U. Weaver
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle upon Tyne NE9 6SH, UK
- Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Correspondence: ; Tel.: +44-191-445-2181
| |
Collapse
|
39
|
Long C, Tang Y, Huang J, Liu S, Xing Z. Association of long-term visit-to-visit variability of HbA1c and fasting glycemia with hypoglycemia in type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2022; 13:975468. [PMID: 36034445 PMCID: PMC9402888 DOI: 10.3389/fendo.2022.975468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/21/2022] [Indexed: 11/14/2022] Open
Abstract
Background Self-management of blood glucose levels to avoid hypoglycemia is vital for patients with type 2 diabetes mellitus (T2DM). The association between specific metrics of glycemic variability (glycosylated hemoglobin A1c [HbA1c] and fasting plasma glucose [FPG]) and severe hypoglycemia has not been fully studied in patients with T2DM. Methods In this post hoc analysis, patients with established T2DM with a high risk of cardiovascular disease were included in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) study. The Cox proportional hazards model was used to investigate the relationship between glycemic variability and hypoglycemia requiring medical assistance (HMA) and hypoglycemia requiring any third-party assistance (HAA). The prognostic value of HbA1c/FPG variability for our predefined outcomes was compared using Harrell's C method. Results After adjusting for confounders, each increase in HbA1c variability of 1 standard deviation (SD) indicated a higher risk of HAA (hazard ratio [HR]: 1.10; 95% confidence interval [CI]: 1.03-1.16; P < 0.01) and HMA events (HR: 1.11; 95% CI: 1.03-1.20; P < 0.01). Meanwhile, each increase in FPG variability of 1 SD increased the risk of HAA (HR: 1.40; 95% CI: 1.31-1.49; P < 0.01) and HMA events (HR: 1.46; 95% CI: 1.35-1.57; P < 0.01). Meanwhile, models, including FPG variability, had better prognostic value for our predefined outcomes than HbA1c variability (P < 0.01). Conclusions Increased visit-to-visit variability in HbA1c and fasting glycemia is associated with a greater risk of severe hypoglycemic events in T2DM patients. FPG variability is a more sensitive indicator than HbA1c variability. Trial registration http://www.clinicaltrials.gov. Unique identifier: NCT00000620.
Collapse
Affiliation(s)
- Chen Long
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yaling Tang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiangsheng Huang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Suo Liu
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhenhua Xing
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
40
|
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: 104] [Impact Index Per Article: 52.0] [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
|
41
|
Metformin use and the risk of total knee replacement among diabetic patients: a propensity-score-matched retrospective cohort study. Sci Rep 2022; 12:11571. [PMID: 35798867 PMCID: PMC9262887 DOI: 10.1038/s41598-022-15871-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 06/30/2022] [Indexed: 12/30/2022] Open
Abstract
Metformin has been shown to modulate meta-inflammation, an important pathogenesis in knee osteoarthritis (OA). The study aimed to test the association between regular metformin use with total knee replacement (TKR) in patients with diabetes. This is a retrospective study with electronic records retrieved in Hong Kong public primary care. Patients with diabetes aged ≥ 45 who visited during 2007 to 2010, were followed up for a four-year period from 2011 to 2014 to determine the incidence of TKR. Propensity score matching based on age, sex, co-medications and chronic conditions was conducted to adjust for confounding. Cox regression was implemented to examine the association between metformin use and TKR. In total, 196,930 patients were eligible and 93,330 regular metformin users (defined as ≥ 4 prescriptions over the previous year) and non-users were matched. Among 46,665 regular users, 184 TKRs were conducted, 17.1% fewer than that among non-users. Cox regression showed that regular metformin users had a 19%-lower hazard of TKR [hazard ratio (HR) = 0.81, 95% confidence interval: 0.67 to 0.98, P = 0.033], with a dose–response relationship. Findings suggest a potential protective effect of metformin on knee OA progression and later TKR incidence among diabetic patients.
Collapse
|
42
|
Di Magno L, Di Pastena F, Bordone R, Coni S, Canettieri G. The Mechanism of Action of Biguanides: New Answers to a Complex Question. Cancers (Basel) 2022; 14:cancers14133220. [PMID: 35804992 PMCID: PMC9265089 DOI: 10.3390/cancers14133220] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 01/27/2023] Open
Abstract
Biguanides are a family of antidiabetic drugs with documented anticancer properties in preclinical and clinical settings. Despite intensive investigation, how they exert their therapeutic effects is still debated. Many studies support the hypothesis that biguanides inhibit mitochondrial complex I, inducing energy stress and activating compensatory responses mediated by energy sensors. However, a major concern related to this “complex” model is that the therapeutic concentrations of biguanides found in the blood and tissues are much lower than the doses required to inhibit complex I, suggesting the involvement of additional mechanisms. This comprehensive review illustrates the current knowledge of pharmacokinetics, receptors, sensors, intracellular alterations, and the mechanism of action of biguanides in diabetes and cancer. The conditions of usage and variables affecting the response to these drugs, the effect on the immune system and microbiota, as well as the results from the most relevant clinical trials in cancer are also discussed.
Collapse
Affiliation(s)
- Laura Di Magno
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (L.D.M.); (F.D.P.); (R.B.); (S.C.)
| | - Fiorella Di Pastena
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (L.D.M.); (F.D.P.); (R.B.); (S.C.)
| | - Rosa Bordone
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (L.D.M.); (F.D.P.); (R.B.); (S.C.)
| | - Sonia Coni
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (L.D.M.); (F.D.P.); (R.B.); (S.C.)
| | - Gianluca Canettieri
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (L.D.M.); (F.D.P.); (R.B.); (S.C.)
- Istituto Pasteur—Fondazione Cenci—Bolognetti, 00161 Rome, Italy
- Correspondence:
| |
Collapse
|
43
|
The Regulatory Mechanism and Effect of RIPK3 on PE-induced Cardiomyocyte Hypertrophy. J Cardiovasc Pharmacol 2022; 80:236-250. [PMID: 35561290 DOI: 10.1097/fjc.0000000000001293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/26/2022] [Indexed: 12/07/2022]
Abstract
ABSTRACT As a critical regulatory molecule, receptor-interacting protein kinase 3 (RIPK3) can mediate the signaling pathway of programmed necrosis. Calcium/calmodulin-dependent protein kinase II (CaMKII) has been proved as a new substrate for RIPK3-induced necroptosis. In the present study, we aimed to investigate the regulatory mechanism of RIPK3 on phenylephrine (PE)-induced cardiomyocyte hypertrophy. Cardiomyocyte hypertrophy was induced by exposure to PE (100 μM) for 48 h. Primary cardiomyocytes were pretreated with RIPK3 inhibitor GSK'872 (10 μM), and RIPK3 siRNA was used to deplete the intracellular expression of RIPK3. The indexes related to myocardial hypertrophy, cell injury, necroptosis, CaMKII activation, gene expression, oxidative stress, and mitochondrial membrane potential were measured. We found that after cardiomyocytes were stimulated by PE, the expressions of hypertrophy markers, atrial and brain natriuretic peptides (ANP and BNP), were increased, the release of lactate dehydrogenase (LDH) was increased, the level of adenosine triphosphate (ATP)was decreased, the oxidation and phosphorylation levels of CaMKII were increased, and CaMKIIδ alternative splicing was disturbed. However, both GSK'872 and depletion of RIPK3 could reduce myocardial dysfunction, inhibit CaMKII activation and necroptosis, and finally alleviate myocardial hypertrophy. In addition, the pretreatment of RIPK3 could also lessen the accumulation of reactive oxygen species (ROS) induced by PE and stabilize the membrane potential of mitochondria. These results indicated that targeted inhibition of RIPK3 could suppress the activation of CaMKII and reduce necroptosis and oxidative stress, leading to alleviated myocardial hypertrophy. Collectively, our findings provided valuable insights into the clinical treatment of hypertrophic cardiomyopathy.
Collapse
|
44
|
Dia M, Leon C, Chanon S, Bendridi N, Gomez L, Rieusset J, Thibault H, Paillard M. Effect of Metformin on T2D-Induced MAM Ca 2+ Uncoupling and Contractile Dysfunction in an Early Mouse Model of Diabetic HFpEF. Int J Mol Sci 2022; 23:ijms23073569. [PMID: 35408928 PMCID: PMC8998623 DOI: 10.3390/ijms23073569] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 01/27/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a leading complication in type 2 diabetes patients. Recently, we have shown that the reticulum-mitochondria Ca2+ uncoupling is an early and reversible trigger of the cardiac dysfunction in a diet-induced mouse model of DCM. Metformin is a first-line antidiabetic drug with recognized cardioprotective effect in myocardial infarction. Whether metformin could prevent the progression of DCM remains not well understood. We therefore investigated the effect of a chronic 6-week metformin treatment on the reticulum-mitochondria Ca2+ coupling and the cardiac function in our high-fat high-sucrose diet (HFHSD) mouse model of DCM. Although metformin rescued the glycemic regulation in the HFHSD mice, it did not preserve the reticulum-mitochondria Ca2+ coupling either structurally or functionally. Metformin also did not prevent the progression towards cardiac dysfunction, i.e., cardiac hypertrophy and strain dysfunction. In summary, despite its cardioprotective role, metformin is not sufficient to delay the progression to early DCM.
Collapse
Affiliation(s)
- Maya Dia
- Laboratoire CarMeN—IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500 Bron, France; (M.D.); (C.L.); (L.G.); (H.T.)
| | - Christelle Leon
- Laboratoire CarMeN—IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500 Bron, France; (M.D.); (C.L.); (L.G.); (H.T.)
| | - Stephanie Chanon
- Laboratoire CarMeN—MERISM Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69921 Oullins, France; (S.C.); (N.B.); (J.R.)
| | - Nadia Bendridi
- Laboratoire CarMeN—MERISM Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69921 Oullins, France; (S.C.); (N.B.); (J.R.)
| | - Ludovic Gomez
- Laboratoire CarMeN—IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500 Bron, France; (M.D.); (C.L.); (L.G.); (H.T.)
| | - Jennifer Rieusset
- Laboratoire CarMeN—MERISM Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69921 Oullins, France; (S.C.); (N.B.); (J.R.)
| | - Helene Thibault
- Laboratoire CarMeN—IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500 Bron, France; (M.D.); (C.L.); (L.G.); (H.T.)
- Hospices Civils de Lyon, 69500 Bron, France
| | - Melanie Paillard
- Laboratoire CarMeN—IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500 Bron, France; (M.D.); (C.L.); (L.G.); (H.T.)
- Correspondence: ; Tel.: +33-(0)4-78-78-56-10
| |
Collapse
|
45
|
Buczyńska A, Sidorkiewicz I, Krętowski AJ, Zbucka-Krętowska M, Adamska A. Metformin Intervention—A Panacea for Cancer Treatment? Cancers (Basel) 2022; 14:cancers14051336. [PMID: 35267644 PMCID: PMC8909770 DOI: 10.3390/cancers14051336] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 02/07/2023] Open
Abstract
The molecular mechanism of action and the individual influence of various metabolic pathways related to metformin intervention are under current investigation. The available data suggest that metformin provides many advantages, exhibiting anti-inflammatory, anti-cancer, hepatoprotective, cardioprotective, otoprotective, radioprotective, and radio-sensitizing properties depending on cellular context. This literature review was undertaken to provide novel evidence concerning metformin intervention, with a particular emphasis on cancer treatment and prevention. Undoubtedly, the pleiotropic actions associated with metformin include inhibiting inflammatory processes, increasing antioxidant capacity, and improving glycemic and lipid metabolism. Consequently, these characteristics make metformin an attractive medicament to translate to human trials, the promising results of which were also summarized in this review.
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (I.S.); (A.J.K.)
- Correspondence: (A.B.); (A.A.); Tel.: +48-85-746-8513 (A.B.); +48-85-746-8660 (A.A.)
| | - Iwona Sidorkiewicz
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (I.S.); (A.J.K.)
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (I.S.); (A.J.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Monika Zbucka-Krętowska
- Department of Gynecological Endocrinology and Adolescent Gynecology, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Agnieszka Adamska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
- Correspondence: (A.B.); (A.A.); Tel.: +48-85-746-8513 (A.B.); +48-85-746-8660 (A.A.)
| |
Collapse
|
46
|
Metformin and Breast Cancer: Where Are We Now? Int J Mol Sci 2022; 23:ijms23052705. [PMID: 35269852 PMCID: PMC8910543 DOI: 10.3390/ijms23052705] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is the most prevalent cancer and the leading cause of cancer-related death among women worldwide. Type 2 diabetes–associated metabolic traits such as hyperglycemia, hyperinsulinemia, inflammation, oxidative stress, and obesity are well-known risk factors for breast cancer. The insulin sensitizer metformin, one of the most prescribed oral antidiabetic drugs, has been suggested to function as an antitumoral agent, based on epidemiological and retrospective clinical data as well as preclinical studies showing an antiproliferative effect in cultured breast cancer cells and animal models. These benefits provided a strong rationale to study the effects of metformin in routine clinical care of breast cancer patients. However, the initial enthusiasm was tempered after disappointing results in randomized controlled trials, particularly in the metastatic setting. Here, we revisit the current state of the art of metformin mechanisms of action, critically review past and current metformin-based clinical trials, and briefly discuss future perspectives on how to incorporate metformin into the oncologist’s armamentarium for the prevention and treatment of breast cancer.
Collapse
|
47
|
Metformin: Expanding the Scope of Application-Starting Earlier than Yesterday, Canceling Later. Int J Mol Sci 2022; 23:ijms23042363. [PMID: 35216477 PMCID: PMC8875586 DOI: 10.3390/ijms23042363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/29/2022] [Accepted: 02/18/2022] [Indexed: 12/15/2022] Open
Abstract
Today the area of application of metformin is expanding, and a wealth of data point to its benefits in people without carbohydrate metabolism disorders. Already in the population of people leading an unhealthy lifestyle, before the formation of obesity and prediabetes metformin smooths out the adverse effects of a high-fat diet. Being prescribed at this stage, metformin will probably be able to, if not prevent, then significantly reduce the progression of all subsequent metabolic changes. To a large extent, this review will discuss the proofs of the evidence for this. Another recent important change is a removal of a number of restrictions on its use in patients with heart failure, acute coronary syndrome and chronic kidney disease. We will discuss the reasons for these changes and present a new perspective on the role of increasing lactate in metformin therapy.
Collapse
|
48
|
Dysregulated Epicardial Adipose Tissue as a Risk Factor and Potential Therapeutic Target of Heart Failure with Preserved Ejection Fraction in Diabetes. Biomolecules 2022; 12:biom12020176. [PMID: 35204677 PMCID: PMC8961672 DOI: 10.3390/biom12020176] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
Cardiovascular (CV) disease and heart failure (HF) are the leading cause of mortality in type 2 diabetes (T2DM), a metabolic disease which represents a fast-growing health challenge worldwide. Specifically, T2DM induces a cluster of systemic metabolic and non-metabolic signaling which may promote myocardium derangements such as inflammation, fibrosis, and myocyte stiffness, which represent the hallmarks of heart failure with preserved ejection fraction (HFpEF). On the other hand, several observational studies have reported that patients with T2DM have an abnormally enlarged and biologically transformed epicardial adipose tissue (EAT) compared with non-diabetic controls. This expanded EAT not only causes a mechanical constriction of the diastolic filling but is also a source of pro-inflammatory mediators capable of causing inflammation, microcirculatory dysfunction and fibrosis of the underlying myocardium, thus impairing the relaxability of the left ventricle and increasing its filling pressure. In addition to representing a potential CV risk factor, emerging evidence shows that EAT may guide the therapeutic decision in diabetic patients as drugs such as metformin, glucagon-like peptide‑1 (GLP-1) receptor agonists and sodium-glucose cotransporter 2 inhibitors (SGLT2-Is), have been associated with attenuation of EAT enlargement.
Collapse
|
49
|
Tseng CH. Metformin and risk of gingival/periodontal diseases in diabetes patients: A retrospective cohort study. Front Endocrinol (Lausanne) 2022; 13:1036885. [PMID: 36277720 PMCID: PMC9583654 DOI: 10.3389/fendo.2022.1036885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 09/20/2022] [Indexed: 12/03/2022] Open
Abstract
AIM To compare the risk of gingival and periodontal diseases (GPD) between ever users and never users of metformin in patients with type 2 diabetes mellitus. METHODS The Taiwan's National Health Insurance database was used to enroll 423,949 patients with new onset diabetes mellitus from 1999 to 2005. After excluding ineligible patients, 60,309 ever users and 5578 never users were followed up for the incidence of GPD from January 1, 2006 until December 31, 2011. Propensity score-weighted hazard ratios were estimated by Cox regression. RESULTS GPD was newly diagnosed in 18,528 ever users (incidence: 7746.51 per 100,000 person-years) and 2283 never users (incidence: 12158.59 per 100,000 person-years). The hazard ratio that compared ever users to never users was 0.627 (95% confidence interval: 0.600-0.655). When metformin use was categorized by tertiles of cumulative duration and cumulative dose, the risk significantly reduced in a dose-response pattern when the cumulative duration reached approximately 2 years or the cumulative dose reached 670 grams. Analyses on the tertiles of defined daily dose of metformin showed that the reduction of GPD risk could be seen in all three subgroups but the benefit would be greater when the daily dose increased. CONCLUSION Long-term use of metformin is associated with a significantly reduced risk of GPD.
Collapse
Affiliation(s)
- Chin-Hsiao Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
50
|
Fan X, Xu M, Chen X, Ren Q, Fan Y, Wang R, Chen J, Cui L, Wang Z, Sun X, Guo N. Proteomic profiling and correlations with clinical features reveal biomarkers indicative of diabetic retinopathy with diabetic kidney disease. Front Endocrinol (Lausanne) 2022; 13:1001391. [PMID: 36277688 PMCID: PMC9581084 DOI: 10.3389/fendo.2022.1001391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/20/2022] [Indexed: 11/20/2022] Open
Abstract
Diabetic retinopathy (DR) and diabetic kidney disease (DKD) are complications of diabetes and place serious health and economic burdens on society. However, the identification and characterization of early biomarkers for DKD, especially for nonproliferative DR (NPDR) patients with DKD, are still needed. This study aimed to demonstrate the plasma proteomic profiles of NPDR+DKD and NPDR patients and identify potential biomarkers for early diagnosis of DKD. Fifteen plasma samples from the NPDR group and nine from the NPDR+DKD group were analyzed by LC-MS/MS to identify the differentially expressed proteins between the two groups. Functional enrichment, protein-protein interaction and clinical feature correlation analyses revealed the target protein candidates, which were verified using ELISA and receiver operating characteristic (ROC) analysis. In total, 410 proteins were detected in plasma; 15 were significantly upregulated and 7 were downregulated in the NPDR+DKD group. Bioinformatics analysis suggested that DKD is closely related to cell adhesion and immunity pathways. β-2-Microglobulin (B2M) and vimentin (VIM) were upregulated in NPDR+DKD, enriched as hub proteins and strongly correlated with clinical features. ELISA showed that B2M (p<0.001) and VIM (p<0.0001) were significantly upregulated in NPDR+DKD compared with NPDR. In ROC analysis, B2M and VIM could distinguish DKD from NPDR with area under the curve values of 0.9000 (p < 0.0001) and 0.9950. Our proteomic study revealed alterations in the proteomic profile and identified VIM and B2M as early biomarkers of DKD, laying the foundation for the prevention, diagnosis and treatment of DKD.
Collapse
Affiliation(s)
- Xiao’e Fan
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
- *Correspondence: Xiao’e Fan,
| | - Manhong Xu
- Department of Vitreoretinal and Ocular Trauma, Tianjin Medical University Eye Hospital, Eye Institute and School of Optometry, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin International Joint Research Center of Ophthalmology and Visual Science, Tianjin, China
| | - Xin Chen
- Department of Vitreoretinal and Ocular Trauma, Tianjin Medical University Eye Hospital, Eye Institute and School of Optometry, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin International Joint Research Center of Ophthalmology and Visual Science, Tianjin, China
| | - Qianfeng Ren
- Department of Pathology, Jincheng People’s Hospital, Jincheng, China
| | - Yan Fan
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
| | - Ranran Wang
- Department of Laboratory, Jincheng People’s Hospital, Jincheng, China
| | - Jiaqi Chen
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
| | - Li Cui
- Department of Nephrology, Jincheng People’s Hospital, Jincheng, China
| | - Zhengmin Wang
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
| | - Xiaoyan Sun
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
| | - Nannan Guo
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
| |
Collapse
|