1
|
Geiger M, Gorica E, Mohammed SA, Mongelli A, Mengozi A, Delfine V, Ruschitzka F, Costantino S, Paneni F. Epigenetic Network in Immunometabolic Disease. Adv Biol (Weinh) 2024; 8:e2300211. [PMID: 37794610 DOI: 10.1002/adbi.202300211] [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: 06/08/2023] [Revised: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Although a large amount of data consistently shows that genes affect immunometabolic characteristics and outcomes, epigenetic mechanisms are also heavily implicated. Epigenetic changes, including DNA methylation, histone modification, and noncoding RNA, determine gene activity by altering the accessibility of chromatin to transcription factors. Various factors influence these alterations, including genetics, lifestyle, and environmental cues. Moreover, acquired epigenetic signals can be transmitted across generations, thus contributing to early disease traits in the offspring. A closer investigation is critical in this aspect as it can help to understand the underlying molecular mechanisms further and gain insights into potential therapeutic targets for preventing and treating diseases arising from immuno-metabolic dysregulation. In this review, the role of chromatin alterations in the transcriptional modulation of genes involved in insulin resistance, systemic inflammation, macrophage polarization, endothelial dysfunction, metabolic cardiomyopathy, and nonalcoholic fatty liver disease (NAFLD), is discussed. An overview of emerging chromatin-modifying drugs and the importance of the individual epigenetic profile for personalized therapeutic approaches in patients with immuno-metabolic disorders is also presented.
Collapse
Affiliation(s)
- Martin Geiger
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Shafeeq Ahmed Mohammed
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessandro Mengozi
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Valentina Delfine
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- Department of Research and Education, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| |
Collapse
|
2
|
Marino F, Salerno N, Scalise M, Salerno L, Torella A, Molinaro C, Chiefalo A, Filardo A, Siracusa C, Panuccio G, Ferravante C, Giurato G, Rizzo F, Torella M, Donniacuo M, De Angelis A, Viglietto G, Urbanek K, Weisz A, Torella D, Cianflone E. Streptozotocin-Induced Type 1 and 2 Diabetes Mellitus Mouse Models Show Different Functional, Cellular and Molecular Patterns of Diabetic Cardiomyopathy. Int J Mol Sci 2023; 24:ijms24021132. [PMID: 36674648 PMCID: PMC9860590 DOI: 10.3390/ijms24021132] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
The main cause of morbidity and mortality in diabetes mellitus (DM) is cardiovascular complications. Diabetic cardiomyopathy (DCM) remains incompletely understood. Animal models have been crucial in exploring DCM pathophysiology while identifying potential therapeutic targets. Streptozotocin (STZ) has been widely used to produce experimental models of both type 1 and type 2 DM (T1DM and T2DM). Here, we compared these two models for their effects on cardiac structure, function and transcriptome. Different doses of STZ and diet chows were used to generate T1DM and T2DM in C57BL/6J mice. Normal euglycemic and nonobese sex- and age-matched mice served as controls (CTRL). Immunohistochemistry, RT-PCR and RNA-seq were employed to compare hearts from the three animal groups. STZ-induced T1DM and T2DM affected left ventricular function and myocardial performance differently. T1DM displayed exaggerated apoptotic cardiomyocyte (CM) death and reactive hypertrophy and fibrosis, along with increased cardiac oxidative stress, CM DNA damage and senescence, when compared to T2DM in mice. T1DM and T2DM affected the whole cardiac transcriptome differently. In conclusion, the STZ-induced T1DM and T2DM mouse models showed significant differences in cardiac remodeling, function and the whole transcriptome. These differences could be of key relevance when choosing an animal model to study specific features of DCM.
Collapse
Affiliation(s)
- Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Nadia Salerno
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Annalaura Torella
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Claudia Molinaro
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Andrea Filardo
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Chiara Siracusa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Giuseppe Panuccio
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Carlo Ferravante
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana′, University of Salerno, 84081 Salerno, Italy
| | - Giorgio Giurato
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana′, University of Salerno, 84081 Salerno, Italy
| | - Francesca Rizzo
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana′, University of Salerno, 84081 Salerno, Italy
| | - Michele Torella
- Department of Translational Medical Science, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Maria Donniacuo
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 88121 Naples, Italy
| | - Alessandro Weisz
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana′, University of Salerno, 84081 Salerno, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
- Correspondence: (D.T.); (E.C.); Tel.: +39-0961369-7564 (D.T.); +39-0961369-4185 (E.C.)
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
- Correspondence: (D.T.); (E.C.); Tel.: +39-0961369-7564 (D.T.); +39-0961369-4185 (E.C.)
| |
Collapse
|
3
|
Dhar A, Venkadakrishnan J, Roy U, Vedam S, Lalwani N, Ramos KS, Pandita TK, Bhat A. A comprehensive review of the novel therapeutic targets for the treatment of diabetic cardiomyopathy. Ther Adv Cardiovasc Dis 2023; 17:17539447231210170. [PMID: 38069578 PMCID: PMC10710750 DOI: 10.1177/17539447231210170] [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: 12/08/2022] [Accepted: 10/09/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is characterized by structural and functional abnormalities in the myocardium affecting people with diabetes. Treatment of DCM focuses on glucose control, blood pressure management, lipid-lowering, and lifestyle changes. Due to limited therapeutic options, DCM remains a significant cause of morbidity and mortality in patients with diabetes, thus emphasizing the need to develop new therapeutic strategies. Ongoing research is aimed at understanding the underlying molecular mechanism(s) involved in the development and progression of DCM, including oxidative stress, inflammation, and metabolic dysregulation. The goal is to develope innovative pharmaceutical therapeutics, offering significant improvements in the clinical management of DCM. Some of these approaches include the effective targeting of impaired insulin signaling, cardiac stiffness, glucotoxicity, lipotoxicity, inflammation, oxidative stress, cardiac hypertrophy, and fibrosis. This review focuses on the latest developments in understanding the underlying causes of DCM and the therapeutic landscape of DCM treatment.
Collapse
Affiliation(s)
- Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | | | - Utsa Roy
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | - Sahithi Vedam
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | - Nikita Lalwani
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | - Kenneth S. Ramos
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Tej K. Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir (UT) 184311, India
| |
Collapse
|
4
|
Chen B, Zhang W, Lin C, Zhang L. A Comprehensive Review on Beneficial Effects of Catechins on Secondary Mitochondrial Diseases. Int J Mol Sci 2022; 23:ijms231911569. [PMID: 36232871 PMCID: PMC9569714 DOI: 10.3390/ijms231911569] [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] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/13/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondria are the main sites for oxidative phosphorylation and synthesis of adenosine triphosphate in cells, and are known as cellular power factories. The phrase "secondary mitochondrial diseases" essentially refers to any abnormal mitochondrial function other than primary mitochondrial diseases, i.e., the process caused by the genes encoding the electron transport chain (ETC) proteins directly or impacting the production of the machinery needed for ETC. Mitochondrial diseases can cause adenosine triphosphate (ATP) synthesis disorder, an increase in oxygen free radicals, and intracellular redox imbalance. It can also induce apoptosis and, eventually, multi-system damage, which leads to neurodegenerative disease. The catechin compounds rich in tea have attracted much attention due to their effective antioxidant activity. Catechins, especially acetylated catechins such as epicatechin gallate (ECG) and epigallocatechin gallate (EGCG), are able to protect mitochondria from reactive oxygen species. This review focuses on the role of catechins in regulating cell homeostasis, in which catechins act as a free radical scavenger and metal ion chelator, their protective mechanism on mitochondria, and the protective effect of catechins on mitochondrial deoxyribonucleic acid (DNA). This review highlights catechins and their effects on mitochondrial functional metabolic networks: regulating mitochondrial function and biogenesis, improving insulin resistance, regulating intracellular calcium homeostasis, and regulating epigenetic processes. Finally, the indirect beneficial effects of catechins on mitochondrial diseases are also illustrated by the warburg and the apoptosis effect. Some possible mechanisms are shown graphically. In addition, the bioavailability of catechins and peracetylated-catechins, free radical scavenging activity, mitochondrial activation ability of the high-molecular-weight polyphenol, and the mitochondrial activation factor were also discussed.
Collapse
|
5
|
Purnama U, Castro-Guarda M, Sahoo OS, Carr CA. Modelling Diabetic Cardiomyopathy: Using Human Stem Cell-Derived Cardiomyocytes to Complement Animal Models. Metabolites 2022; 12:metabo12090832. [PMID: 36144236 PMCID: PMC9503602 DOI: 10.3390/metabo12090832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Diabetes is a global epidemic, with cardiovascular disease being the leading cause of death in diabetic patients. There is a pressing need for an in vitro model to aid understanding of the mechanisms driving diabetic heart disease, and to provide an accurate, reliable tool for drug testing. Human induced-pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have potential as a disease modelling tool. There are several factors that drive molecular changes inside cardiomyocytes contributing to diabetic cardiomyopathy, including hyperglycaemia, lipotoxicity and hyperinsulinemia. Here we discuss these factors and how they can be seen in animal models and utilised in cell culture to mimic the diabetic heart. The use of human iPSC-CMs will allow for a greater understanding of disease pathogenesis and open up new avenues for drug testing.
Collapse
Affiliation(s)
- Ujang Purnama
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Marcos Castro-Guarda
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Om Saswat Sahoo
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713216, India
| | - Carolyn A. Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Correspondence: ; Tel.: +44-1865-282247
| |
Collapse
|
6
|
Deng J, Liao Y, Liu J, Liu W, Yan D. Research Progress on Epigenetics of Diabetic Cardiomyopathy in Type 2 Diabetes. Front Cell Dev Biol 2022; 9:777258. [PMID: 35004678 PMCID: PMC8740193 DOI: 10.3389/fcell.2021.777258] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/06/2021] [Indexed: 12/24/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is characterized by diastolic relaxation abnormalities in its initial stages and by clinical heart failure (HF) without dyslipidemia, hypertension, and coronary artery disease in its last stages. DCM contributes to the high mortality and morbidity rates observed in diabetic populations. Diabetes is a polygenic, heritable, and complex condition that is exacerbated by environmental factors. Recent studies have demonstrated that epigenetics directly or indirectly contribute to pathogenesis. While epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNAs, have been recognized as key players in the pathogenesis of DCM, some of their impacts remain not well understood. Furthering our understanding of the roles played by epigenetics in DCM will provide novel avenues for DCM therapeutics and prevention strategies.
Collapse
Affiliation(s)
- Jianxin Deng
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University; Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen, China
| | - Yunxiu Liao
- Health Science Center of Shenzhen University, Shenzhen, China
| | - Jianpin Liu
- Health Science Center of Shenzhen University, Shenzhen, China
| | - Wenjuan Liu
- Health Science Center of Shenzhen University, Shenzhen, China
| | - Dewen Yan
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University; Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen, China
| |
Collapse
|
7
|
Oyinloye BE, Ajiboye BO, Johnson O, Owolabi OV, Ejeje JN, Brai BIC, Omotuyi OI. Ameliorative Effect of Flavonoid-rich Extracts from Gongronema latifolium Against Diabetic Cardiomyopathy via serpin A 3 and socs3-a in Streptozocin Treated Rats. Biomarkers 2021; 27:169-177. [PMID: 34951557 DOI: 10.1080/1354750x.2021.2023220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
INTRODUCTION The present study access the effect of the flavonoid-rich isolate from Gongronema latifolium against cardiomyopathy in a streptozotocin-rich extract. MATERIAL AND METHODS The flavonoid-rich isolate from G. latifolium leaf (FREGL) was prepared using a standard method. A single injection of streptozotocin was induced into the experimental rats. The experimental animals were divided into five groups as non-diabetic rats, diabetic control, diabetic rats administered low and high doses of FREGL (13 and 26 mg/kg), and metformin glibenclamide orally for 21 days. Hence, the experimental animals were sacrificed; blood and heart were harvested to determine diverse biochemical parameters, including the gene expressions of serpin A3 and socs3-a as well as histological examination. RESULTS The results demonstrated that FREGL significantly (p < 0.05) reduced fasting blood glucose, total cholesterol, low density lipoprotein (LDL), triglyceride (TG), lipid peroxidation levels, as well as the activities of lactate dehydrogenase and creatine kinase-MB, including the relative gene expressions of serpin A3 and Socs3-A in diabetic rats. Also, diabetic rats that received different doses of FREGL showed a substantial rise in insulin and high density lipoprotein (HDL) levels, and antioxidant enzyme activities, as well as, normal histoarchitecture of the heart tissues. CONCLUSION Therefore, FREGL may be beneficial in alleviating diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Babatunji Emmanuel Oyinloye
- Phytomedicine, Biochemical Toxicology and Biotechnology Research Laboratories, Department of Biochemistry, College of Sciences, Afe Babalola University, PMB 5454, Ado-Ekiti, 360001, Nigeria.,Institute of Drug Research and Development, SE Bogoro Center, Afe Babalola University, PMB 5454, Ado-Ekiti, 360001, Nigeria.,Biotechnology and Structural Biology (BSB) Group, Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, 3886, South Africa
| | - B O Ajiboye
- Phytomedicine and Molecular Toxicology Research Laboratory, Department of Biochemistry, Federal University Oye-Ekiti, PMB 373, Oye-Ekiti, 371104, Nigeria.,Phytomedicine, Biochemical Toxicology and Biotechnology Research Laboratories, Department of Biochemistry, College of Sciences, Afe Babalola University, PMB 5454, Ado-Ekiti, 360001, Nigeria.,Institute of Drug Research and Development, SE Bogoro Center, Afe Babalola University, PMB 5454, Ado-Ekiti, 360001, Nigeria
| | - Oluwafolakemi Johnson
- Phytomedicine, Biochemical Toxicology and Biotechnology Research Laboratories, Department of Biochemistry, College of Sciences, Afe Babalola University, PMB 5454, Ado-Ekiti, 360001, Nigeria
| | - Olutunmise Victoria Owolabi
- Medical Biochemistry Unit, College of Medicine and Health Sciences, Afe Babalola University, PMB 5454, Ado-Ekiti 360001, Nigeria
| | - Jerius Nkwuda Ejeje
- Phytomedicine, Biochemical Toxicology and Biotechnology Research Laboratories, Department of Biochemistry, College of Sciences, Afe Babalola University, PMB 5454, Ado-Ekiti, 360001, Nigeria.,Department of Chemistry/Biochemistry/Molecular Biology, Alex- Ekwueme Federal University Ndufu-Alike, P.O. Box 1010, Abakaliki 482131, Nigeria
| | - Batholemn I C Brai
- Department of Biochemistry, Federal University Oye-Ekiti, PMB 373, Oye-Ekiti, 371104, Nigeria
| | - Olaposi Idowu Omotuyi
- Institute of Drug Research and Development, SE Bogoro Center, Afe Babalola University, PMB 5454, Ado-Ekiti, 360001, Nigeria.,Department of Pharmacceutical Science, Faculty of Pharmacy, Afe Babalola University Ado-Ekiti, Ekiti State, Nigeria
| |
Collapse
|
8
|
Erukainure OL, Msomi NZ, Beseni BK, Salau VF, Ijomone OM, Koorbanally NA, Islam MS. Cola nitida infusion modulates cardiometabolic activities linked to cardiomyopathy in diabetic rats. Food Chem Toxicol 2021; 154:112335. [PMID: 34129900 DOI: 10.1016/j.fct.2021.112335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 05/29/2021] [Accepted: 06/09/2021] [Indexed: 12/11/2022]
Abstract
This study investigated the therapeutic mechanism of Cola nitida seeds on diabetic cardiomyopathy in hearts of diabetic rats. Type 2 diabetic (T2D) rats were treated with C. nitida infusion at 150 or 300 mg/kg body weight (bw). The rats were sacrificed after 6 weeks of treatment, and their hearts harvested. There was an upsurge in oxidative stress on induction of T2D as depicted by the depleted levels of glutathione, superoxide dismutase and catalase activities, and elevated malondialdehyde level. The activities of acetylcholinesterase, and ATPase were significantly elevated, with suppressed ENTPDase and 5'nucleotodase activities in hearts of T2D rats depicting cholinergic and purinergic dysfunctions. Induction of T2D further led to elevated activity of ACE and altered myocardial morphology. Treatment with C. nitida infusion led to reversal of these biomarkers' activities and levels, while maintaining an intact morphology. The infusion caused decreased lipase activity and depletion of diabetes-generated cardiac lipid metabolites, while concomitantly generating saturated and unsaturated fatty acids, fatty esters and alcohols. There was also an inactivation of plasmalogen synthesis and mitochondrial beta-oxidation of long chain saturated fatty acids pathways in T2D rats treated with C. nitida infusion. These results indicate the therapeutic effect of C. nitida infusion against diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Ochuko L Erukainure
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa; Department of Pharmacology, University of the Free State, Bloemfontein, 9300, South Africa.
| | - Nontokozo Z Msomi
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Brian K Beseni
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Veronica F Salau
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Omamuyovwi M Ijomone
- Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Neil A Koorbanally
- School of Chemistry and Physics, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa.
| |
Collapse
|
9
|
Torre E, Arici M, Lodrini AM, Ferrandi M, Barassi P, Hsu SC, Chang GJ, Boz E, Sala E, Vagni S, Altomare C, Mostacciuolo G, Bussadori C, Ferrari P, Bianchi G, Rocchetti M. SERCA2a stimulation by istaroxime improves intracellular Ca2+ handling and diastolic dysfunction in a model of diabetic cardiomyopathy. Cardiovasc Res 2021; 118:1020-1032. [PMID: 33792692 PMCID: PMC8930067 DOI: 10.1093/cvr/cvab123] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/20/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022] Open
Abstract
Aims Diabetic cardiomyopathy is a multifactorial disease characterized by an early onset of diastolic dysfunction (DD) that precedes the development of systolic impairment. Mechanisms that can restore cardiac relaxation improving intracellular Ca2+ dynamics represent a promising therapeutic approach for cardiovascular diseases associated to DD. Istaroxime has the dual properties to accelerate Ca2+ uptake into sarcoplasmic reticulum (SR) through the SR Ca2+ pump (SERCA2a) stimulation and to inhibit Na+/K+ ATPase (NKA). This project aims to characterize istaroxime effects at a concentration (100 nmol/L) marginally affecting NKA, in order to highlight its effects dependent on the stimulation of SERCA2a in an animal model of mild diabetes. Methods and results Streptozotocin (STZ) treated diabetic rats were studied at 9 weeks after STZ injection in comparison to controls (CTR). Istaroxime effects were evaluated in vivo and in left ventricular (LV) preparations. STZ animals showed (i) marked DD not associated to cardiac fibrosis, (ii) LV mass reduction associated to reduced LV cell dimension and T-tubules loss, (iii) reduced LV SERCA2 protein level and activity and (iv) slower SR Ca2+ uptake rate, (v) LV action potential (AP) prolongation and increased short-term variability (STV) of AP duration, (vi) increased diastolic Ca2+, and (vii) unaltered SR Ca2+ content and stability in intact cells. Acute istaroxime infusion (0.11 mg/kg/min for 15 min) reduced DD in STZ rats. Accordingly, in STZ myocytes istaroxime (100 nmol/L) stimulated SERCA2a activity and blunted STZ-induced abnormalities in LV Ca2+ dynamics. In CTR myocytes, istaroxime increased diastolic Ca2+ level due to NKA blockade albeit minimal, while its effects on SERCA2a were almost absent. Conclusions SERCA2a stimulation by istaroxime improved STZ-induced DD and intracellular Ca2+ handling anomalies. Thus, SERCA2a stimulation can be considered a promising therapeutic approach for DD treatment.
Collapse
Affiliation(s)
- Eleonora Torre
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Martina Arici
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Alessandra Maria Lodrini
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Mara Ferrandi
- Windtree Therapeutics Inc., Warrington, Pennsylvania, USA
| | - Paolo Barassi
- Windtree Therapeutics Inc., Warrington, Pennsylvania, USA
| | | | | | | | - Emanuela Sala
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Sara Vagni
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | | | - Gaspare Mostacciuolo
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | | | | | | | - Marcella Rocchetti
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| |
Collapse
|
10
|
Human induced pluripotent stem cell-derived cardiomyocytes reveal abnormal TGFβ signaling in type 2 diabetes mellitus. J Mol Cell Cardiol 2020; 142:53-64. [PMID: 32251671 DOI: 10.1016/j.yjmcc.2020.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 12/17/2022]
Abstract
Diabetes mellitus is a serious metabolic condition associated with a multitude of cardiovascular complications. Moreover, the prevalence of diabetes in heart failure populations is higher than that in control populations. However, the role of cardiomyocyte alterations in type 2 diabetes mellitus (T2DM) has not been well characterized and the underlying mechanisms remain elusive. In this study, two patients who were diagnosed as T2DM were recruited and patient-specific induced pluripotent stem cells (iPSCs) were generated from urine epithelial cells using nonintegrated Sendai virus. The iPSC lines derived from five healthy subjects were used as controls. All iPSCs were differentiated into cardiomyocytes (iPSC-CMs) using the monolayer-based differentiation protocol. T2DM iPSC-CMs exhibited various disease phenotypes, including cellular hypertrophy and lipid accumulation. Moreover, T2DM iPSC-CMs exhibited higher susceptibility to high-glucose/high-lipid challenge than control iPSC-CMs, manifesting an increase in apoptosis. RNA-Sequencing analysis revealed a differential transcriptome profile and abnormal activation of TGFβ signaling pathway in T2DM iPSC-CMs. We went on to show that inhibition of TGFβ significantly rescued the hypertrophic phenotype in T2DM iPSC-CMs. In conclusion, we demonstrate that the iPSC-CM model is able to recapitulate cellular phenotype of T2DM. Our results indicate that iPSC-CMs can therefore serve as a suitable model for investigating molecular mechanisms underlying diabetic cardiomyopathies and for screening therapeutic drugs.
Collapse
|
11
|
Pathophysiological mechanisms of diabetic cardiomyopathy and the therapeutic potential of epigallocatechin-3-gallate. Biomed Pharmacother 2018; 109:2155-2172. [PMID: 30551473 DOI: 10.1016/j.biopha.2018.11.086] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/14/2018] [Accepted: 11/20/2018] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular complications are considered one of the leading causes of morbidity and mortality among diabetic patients. Diabetic cardiomyopathy (DCM) is a type of cardiovascular damage presents in diabetic patients independent of the coexistence of ischemic heart disease or hypertension. It is characterized by impaired diastolic relaxation time, myocardial dilatation and hypertrophy and reduced systolic and diastolic functions of the left ventricle. Molecular mechanisms underlying these pathological changes in the diabetic heart are most likely multifactorial and include, but not limited to, oxidative/nitrosative stress, increased advanced glycation end products, mitochondrial dysfunction, inflammation and cell death. The aim of this review is to address the major molecular mechanisms implicated in the pathogenesis of DCM. In addition, this review provides studies conducted to determine the pharmacological effects of (-)-epigallocatechin-3-gallate (EGCG), the major polyphenol in green tea, focusing on its therapeutic potential against the processes involved in the pathogenesis and progression of DCM. EGCG has been shown to exert several potential therapeutic properties both in vitro and in vivo. Given its therapeutic potential, EGCG might be a promising drug candidate to decrease the morbidity and mortality associated with DCM and other diabetes complications.
Collapse
|
12
|
Singh R, Moreno P, Hajjar RJ, Lebeche D. A role for calcium in resistin transcriptional activation in diabetic hearts. Sci Rep 2018; 8:15633. [PMID: 30353146 PMCID: PMC6199245 DOI: 10.1038/s41598-018-34112-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/06/2018] [Indexed: 12/12/2022] Open
Abstract
The adipokine resistin has been proposed to link obesity, insulin resistance and diabetes. We have previously reported that diabetic hearts express high levels of resistin while overexpression of resistin in adult rat hearts gives rise to a phenotype resembling diabetic cardiomyopathy. The transcriptional regulation of resistin in diabetic cardiac tissue is currently unknown. This study investigated the mechanism of resistin upregulation and the role of Serca2a in its transcriptional suppression. We demonstrate that restoration of Ca2+ homeostasis in diabetic hearts, through normalization of Serca2a function genetically and pharmacologically, suppressed resistin expression via inhibition of NFATc. H9c2 myocytes stimulated with high-glucose concentration or Ca2+ time-dependently increased NFATc and resistin expression while addition of the Ca2+ chelator BAPTA-AM attenuated this effect. NFATc expression was enhanced in hearts from ob/ob diabetic and from cardiac-specific Serca2a−/− mice. Similarly, NFATc increased resistin expression in myocytes cultured in low glucose while the NFATc inhibitor VIVIT blocked glucose-induced resistin expression, suggesting that hyperglycemia/diabetes induces resistin expression possibly through NFATc activation. Interestingly, overexpression of Serca2a or VIVIT mitigated glucose-stimulated resistin and NFATc expression and enhanced AMPK activity, a downstream target of resistin signaling. NFATc direct activation of resistin was verified by resistin promoter luciferase activity and chromatin-immunoprecipitation analysis. Interestingly, activation of Serca2a by a novel agonist, CDN1163, mirrored the effects of AAV9-Serca2a gene transfer on resistin expression and its promoter activity and AMPK signaling in diabetic mice. These findings parse a role for Ca2+ in resistin transactivation and provide support that manipulation of Serca2a-NFATc-Resistin axis might be useful in hyper-resistinemic conditions.
Collapse
Affiliation(s)
- Rajvir Singh
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Pedro Moreno
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Roger J Hajjar
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Djamel Lebeche
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA. .,Diabetes, Obesity and Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA. .,Graduate School of Biological Sciences, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.
| |
Collapse
|
13
|
Anupama N, Preetha Rani MR, Shyni GL, Raghu KG. Glucotoxicity results in apoptosis in H9c2 cells via alteration in redox homeostasis linked mitochondrial dynamics and polyol pathway and possible reversal with cinnamic acid. Toxicol In Vitro 2018; 53:178-192. [PMID: 30144576 DOI: 10.1016/j.tiv.2018.08.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/02/2018] [Accepted: 08/20/2018] [Indexed: 01/03/2023]
Abstract
Several mechanisms have been proposed for the heart dysfunction during hyperglycemia. The aim of the present in vitro study is to elucidate the role of alterations in redox homeostasis in the induction of apoptosis during hyperglycemia in H9c2 cells via dysfunction in mitochondria and polyol pathway and evaluation of the beneficial effect of cinnamic acid against the same. The H9c2 cells were incubated with 33 mM glucose for 48 h to simulate the diabetic condition. Cell injury was confirmed with a significant increase of atrial natriuretic peptide and lactate dehydrogenase release. Alterations in the innate antioxidant system, polyol pathway, mitochondrial integrity, dynamics and apoptosis were investigated. Hyperglycemic insult has significantly affected redox homeostasis via depletion of superoxide dismutase, glutathione and enhanced reactive oxygen species generation. It also caused dysregulation in mitochondrial dynamics (fusion, fission proteins), dissipation of mitochondrial transmembrane potential and increased sorbitol accumulation. Finally, apoptosis was observed with upregulation of Bax, activation of caspase-3 and downregulation of Bcl-2. Cinnamic acid cotreatment increased the innate antioxidant status, improved mitochondrial function and prevented apoptosis in H9c2 cardiomyoblasts. Moreover, this in vitro model is found to be ideal for the elucidation of mechanisms at the cellular and molecular level of any physiological, pharmacological and toxicological incidents in H9c2 cells.
Collapse
Affiliation(s)
- Nair Anupama
- Biochemistry and Molecular Mechanism Laboratory, Agro-processing and Technology Division, CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram 695019, Kerala, India
| | - M R Preetha Rani
- Biochemistry and Molecular Mechanism Laboratory, Agro-processing and Technology Division, CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram 695019, Kerala, India
| | - G L Shyni
- Biochemistry and Molecular Mechanism Laboratory, Agro-processing and Technology Division, CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram 695019, Kerala, India
| | - K G Raghu
- Biochemistry and Molecular Mechanism Laboratory, Agro-processing and Technology Division, CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram 695019, Kerala, India.
| |
Collapse
|
14
|
Hammoudi N, Jeong D, Singh R, Farhat A, Komajda M, Mayoux E, Hajjar R, Lebeche D. Empagliflozin Improves Left Ventricular Diastolic Dysfunction in a Genetic Model of Type 2 Diabetes. Cardiovasc Drugs Ther 2018. [PMID: 28643218 DOI: 10.1007/s10557-017-6734-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Cardiovascular (CV) diseases in type 2 diabetes (T2DM) represent an enormous burden with high mortality and morbidity. Sodium-glucose cotransporter 2 (SGLT2) inhibitors have recently emerged as a new antidiabetic class that improves glucose control, as well as body weight and blood pressure with no increased risk of hypoglycemia. The first CV outcome study terminated with empagliflozin, a specific SGLT2 inhibitor, has shown a reduction in CV mortality and in heart failure hospitalization, suggesting a beneficial impact on cardiac function which remains to be demonstrated. This study was designed to examine the chronic effect of empagliflozin on left ventricular (LV) systolic and diastolic functions in a genetic model of T2DM, ob/ob mice. METHODS AND RESULTS Cardiac phenotype was characterized by echocardiography, in vivo hemodynamics, histology, and molecular profiling. Our results demonstrate that empagliflozin significantly lowered HbA1c and slightly reduced body weight compared to vehicle treatment with no obvious changes in insulin levels. Empagliflozin also improved LV maximum pressure and in vivo indices of diastolic function. While systolic function was grossly not affected in both groups at steady state, response to dobutamine stimulation was significantly improved in the empagliflozin-treated group, suggesting amelioration of contractile reserve. This was paralleled by an increase in phospholamban (PLN) phosphorylation and increased SERCA2a/PLN ratio, indicative of enhanced SERCA2a function, further supporting improved cardiac relaxation and diastolic function. In addition, empagliflozin reconciled diabetes-associated increase in MAPKs and dysregulated phosphorylation of IRS1 and Akt, leading to improvement in myocardial insulin sensitivity and glucose utilization. CONCLUSION The data show that chronic treatment with empagliflozin improves diastolic function, preserves calcium handling and growth signaling pathways and attenuates myocardial insulin resistance in ob/ob mice, findings suggestive of a potential clinical utility for empagliflozin in the treatment of diastolic dysfunction.
Collapse
Affiliation(s)
- Nadjib Hammoudi
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Sorbonne Universités, UPMC University Paris 06, Institut de Cardiologie (AP-HP), Centre Hospitalier Universitaire Pitié-Salpêtrière, Institute of Cardiometabolism and Nutrition (ICAN), INSERM UMRS 1166, Paris, France
| | - Dongtak Jeong
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rajvir Singh
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ahmed Farhat
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Graduate School of Biological Sciences, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Michel Komajda
- Sorbonne Universités, UPMC University Paris 06, Institut de Cardiologie (AP-HP), Centre Hospitalier Universitaire Pitié-Salpêtrière, Institute of Cardiometabolism and Nutrition (ICAN), INSERM UMRS 1166, Paris, France
| | - Eric Mayoux
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cardio-metabolic Diseases, Binger Straße 173, 55216, Ingelheim am Rhein, Germany
| | - Roger Hajjar
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Djamel Lebeche
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Graduate School of Biological Sciences, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Diabetes, Obesity and Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
15
|
Tang SG, Liu XY, Ye JM, Hu TT, Yang YY, Han T, Tan W. Isosteviol ameliorates diabetic cardiomyopathy in rats by inhibiting ERK and NF-κB signaling pathways. J Endocrinol 2018; 238:47-60. [PMID: 29720537 DOI: 10.1530/joe-17-0681] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 04/16/2018] [Indexed: 12/23/2022]
Abstract
Diabetes-induced injury of myocardium, defined as diabetic cardiomyopathy (DCM), accounts for significant mortality and morbidity in diabetic population. Alleviation of DCM by a potent drug remains considerable interests in experimental and clinical researches because hypoglycemic drugs cannot effectively control this condition. Here, we explored the beneficial effects of isosteviol sodium (STVNa) on type 1 diabetes-induced DCM and the potential mechanisms involved. Male Wistar rats were induced to diabetes by injection of streptozotocin (STZ). One week later, diabetic rats were randomly grouped to receive STVNa (STZ/STVNa) or its vehicle (STZ). After 11 weeks of treatment or 11 weeks treatment following 4 weeks of removal of the treatment, the cardiac function and structure were evaluated and related mechanisms were investigated. In diabetic rats, oxidative stress, inflammation, blood glucose and plasma advanced glycation end products (AGEs) were significantly increased, whereas superoxide dismutase 2 (SOD-2) expression and activity were decreased. STVNa treatment inhibited cardiac hypertrophy, fibrosis and inflammation, showed similar ratio of heart to body weight and antioxidant capacities almost similar to the normal controls, which can be sustained at least 4 weeks. Moreover, STVNa inhibited diabetes-inducted stimulation of both extracellular signal-regulated kinase (ERK) and nuclear factor κB (NF-κB) signal pathways. However, blood glucose, plasma AGE and insulin levels were not altered by STVNa treatment. These results indicate that STVNa may be developed into a potent therapy for DCM. The mechanism underlying this therapeutic effect involves the suppression of oxidative stress and inflammation by inhibiting ERK and NF-κB without changing blood glucose or AGEs.
Collapse
Affiliation(s)
- Sheng-Gao Tang
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Xiao-Yu Liu
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Ji-Ming Ye
- Molecular Pharmacology for DiabetesSchool of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Ting-Ting Hu
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Ying-Ying Yang
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Ting Han
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Wen Tan
- Institute of Biomedical & Pharmaceutical ScienceGuangdong University of Technology, Guangzhou, China
| |
Collapse
|
16
|
Recovery of Cardiac Remodeling and Dysmetabolism by Pancreatic Islet Injury Improvement in Diabetic Rats after Yacon Leaf Extract Treatment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1821359. [PMID: 30057670 PMCID: PMC6051012 DOI: 10.1155/2018/1821359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/12/2018] [Indexed: 02/06/2023]
Abstract
Yacon (Smallanthus sonchifolius) is a native Andean plant rich in phenolic compounds, and its effects on dysmetabolism and cardiomyopathy in diabetic rats was evaluated. The rats (10/group) were allocated as follows: C, controls; C + Y, controls treated with Yacon leaf extract (YLE); DM, diabetic controls; and DM + Y, diabetic rats treated with YLE. Type 1 diabetes (T1DM) was induced by the administration of streptozotocin (STZ; 40 mg−1/kg body weight, single dose, i.p.), and treated groups received 100 mg/kg body weight YLE daily via gavage for 30 d. The YLE group shows an improvement in dysmetabolism and cardiomyopathy in the diabetic condition (DM versus DM + Y) promoting a significant reduction of glycemia by 63.39%, an increase in insulin concentration by 49.30%, and a decrease in serum triacylglycerol and fatty acid contents by 0.39- and 0.43-fold, respectively, by ameliorating the pancreatic islet injury, as well as increasing the activity of the antioxidant enzymes (catalase, superoxide dismutase, and glutathione peroxidase) and decreasing the fibrosis and cellular disorganization in cardiac tissue. The apparent benefits of YLE seem to be mediated by ameliorating dysmetabolism and oxidative stress in pancreatic and cardiac tissues.
Collapse
|
17
|
Li T, Bai B, Tian C, Wang H, Jiang D, Ma F, Shan M. High sucrose/fat diet and isosorbide mononitrate increase insulin resistance, nitric oxide production and myocardial apoptosis in a hypertensive rat model. Mol Med Rep 2018; 17:6789-6795. [PMID: 29488615 DOI: 10.3892/mmr.2018.8651] [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: 05/05/2017] [Accepted: 12/07/2017] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to investigate the association between insulin resistance (IR), nitric oxide (NO) production and myocardial apoptosis in a background of coexisting hypertension in a rodent animal model. A hypertensive rat model was established by feeding Wistar and spontaneously hypertensive rats (SHR) with a high sucrose/fat (HSF) diet for 12 weeks, in conjunction with isosorbide mononitrate (ISMN). Increased IR, NO content, apoptotic gene and protein expression, and morphological alterations within rat myocardium were evaluated. Following a total of 12 weeks of feeding with HSF and ISMN resulted in increased IR and NO content within the myocardial tissue of Wistar and SHR rats. HSF and ISMN activated myocardial apoptosis by downregulating the gene transcription and protein expression levels of the anti‑apoptotic B‑cell lymphoma 2 (Bcl‑2), and increasing the pro‑apoptotic Bcl‑2 associated X protein. Apoptosis was demonstrated by DNA fragmentation in terminal deoxynucleotidyl‑transferase‑mediated dUTP nick end labelling assay. In all experiments, the combination of HSF and ISMN was associated with more pronounced effects, indicating the possible synergistic effects. In addition, the correlation analysis in the Wistar rats fed with HSF only, revealed a positive association between NO production and IR. The results of the present study indicated that HSF and ISMN simultaneously increased IR, NO production and myocardial apoptosis in the hypertensive rat model, and may therefore contribute to investigations into the long‑term clinical use of ISMN in hypertensive patients.
Collapse
Affiliation(s)
- Ting Li
- Department of Endocrinology and Metabolic Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Bing Bai
- Department of Endocrinology and Metabolic Diseases, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chenguang Tian
- Department of Endocrinology and Metabolic Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Huihui Wang
- Department of Endocrinology and Metabolic Diseases, The First Affiliated Hospital of Henan Polytechnic University, Jiaozuo, Henan 454000, P.R. China
| | - Deyue Jiang
- Department of Endocrinology and Metabolic Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Fangfei Ma
- Department of Endocrinology and Metabolic Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Mengting Shan
- Department of Endocrinology and Metabolic Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| |
Collapse
|
18
|
Liu W, Gong W, He M, Liu Y, Yang Y, Wang M, Wu M, Guo S, Yu Y, Wang X, Sun F, Li Y, Zhou L, Qin S, Zhang Z. Spironolactone Protects against Diabetic Cardiomyopathy in Streptozotocin-Induced Diabetic Rats. J Diabetes Res 2018; 2018:9232065. [PMID: 30406151 PMCID: PMC6204188 DOI: 10.1155/2018/9232065] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/16/2018] [Accepted: 08/02/2018] [Indexed: 12/16/2022] Open
Abstract
Spironolactone (SPR) has been shown to protect diabetic cardiomyopathy (DCM), but the specific mechanisms are not fully understood. Here, we determined the cardioprotective role of SPR in diabetic mice and further explored the potential mechanisms in both in vivo and in vitro models. Streptozotocin- (STZ-) induced diabetic rats were used as the in vivo model. After the onset of diabetes, rats were treated with either SPR (STZ + SPR) or saline (STZ + NS) for 12 weeks; nondiabetic rats were used as controls (NDCs). In vitro, H9C2 cells were exposed to aldosterone, with or without SPR. Cardiac structure was investigated with transmission electron microscopy and pathological examination; immunohistochemistry was performed to detect nitrotyrosine, collagen-1, TGF-β1, TNF-α, and F4/80 expression; and gene expression of markers for oxidative stress, inflammation, fibrosis, and energy metabolism was detected. Our results suggested that SPR attenuated mitochondrial morphological abnormalities and sarcoplasmic reticulum enlargement in diabetic rats. Compared to the STZ + NS group, cardiac oxidative stress, fibrosis, inflammation, and mitochondrial dysfunction were improved by SPR treatment. Our study showed that SPR had cardioprotective effects in diabetic rats by ameliorating mitochondrial dysfunction and reducing fibrosis, oxidative stress, and inflammation. This study, for the first time, indicates that SPR might be a potential treatment for DCM.
Collapse
Affiliation(s)
- Wenjuan Liu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Wei Gong
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Min He
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Yemei Liu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Department of Endocrinology, The Second People's Hospital, 4 Duchun Road, Wuhu, Anhui 241001, China
| | - Yeping Yang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Meng Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Meng Wu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, 1055 Sanxiang Rd, Suzhou, Jiangsu 215000, China
| | - Shizhe Guo
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Yifei Yu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Xuanchun Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Fei Sun
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Yiming Li
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Linuo Zhou
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Shengmei Qin
- Department of Cardiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Zhaoyun Zhang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| |
Collapse
|
19
|
Singh RM, Waqar T, Howarth FC, Adeghate E, Bidasee K, Singh J. Hyperglycemia-induced cardiac contractile dysfunction in the diabetic heart. Heart Fail Rev 2017; 23:37-54. [DOI: 10.1007/s10741-017-9663-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
20
|
|
21
|
Federico M, Portiansky EL, Sommese L, Alvarado FJ, Blanco PG, Zanuzzi CN, Dedman J, Kaetzel M, Wehrens XHT, Mattiazzi A, Palomeque J. Calcium-calmodulin-dependent protein kinase mediates the intracellular signalling pathways of cardiac apoptosis in mice with impaired glucose tolerance. J Physiol 2017; 595:4089-4108. [PMID: 28105734 DOI: 10.1113/jp273714] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 01/17/2017] [Indexed: 01/01/2023] Open
Abstract
KEY POINTS Spontaneous sarcoplasmic reticulum (SR) Ca2+ release events increased in fructose-rich diet mouse (FRD) myocytes vs. control diet (CD) mice, in the absence of significant changes in SR Ca2+ load. In HEK293 cells, hyperglycaemia significantly enhanced [3 H]ryanodine binding and Ca2+ /calmodulin-dependent protein kinase II (CaMKII) phosphorylation of RyR2-S2814 residue vs. normoglycaemia. These increases were prevented by CaMKII inhibition. FRD significantly augmented cardiac apoptosis in WT vs. CD-WT mice, which was prevented by co-treatment with the reactive oxygen species scavenger Tempol. Oxidative stress was also increased in FRD-SR-autocamide inhibitory peptide (AIP) mice, expressing the SR-targeted CaMKII inhibitor AIP, without any significant enhancement of apoptosis vs. CD-SR-AIP mice. FRD produced mitochondrial swelling and membrane depolarization in FRD-WT mice but not in FRD-S2814A mice, in which the CaMKII site on ryanodine receptor 2 was ablated. FRD decreased mitochondrial area, mean Feret diameter and the mean distance between SR and the outer mitochondrial membrane vs. CD hearts. This remodelling was prevented in AC3I mice, with cardiac-targeted CaMKII inhibition. ABSTRACT The impact of cardiac apoptosis in pre-diabetic stages of diabetic cardiomyopathy is unknown. We show that myocytes from fructose-rich diet (FRD) animals exhibit arrhythmias produced by exacerbated Ca2+ /calmodulin-protein kinase (CaMKII) activity, ryanodine receptor 2 (RyR2) phosphorylation and sarcoplasmic reticulum (SR) Ca2+ leak. We tested the hypothesis that this mechanism also underlies cardiac apoptosis in pre-diabetes. We generated a pre-diabetic model in FRD mice. FRD mice showed an increase in oxidative stress, hypertrophy and systolic dysfunction. FRD myocytes exhibited enhanced SR Ca2+ spontaneous events in the absence of SR Ca2+ load alterations vs. control-diet (CD) myocytes. In HEK293 cells, hyperglycaemia significantly enhanced [3 H]ryanodine binding and CaMKII phosphorylation of RyR2-S2814 residue vs. normoglycaemia. CaMKII inhibition prevented hyperglycaemia-induced alterations. FRD also evoked cardiac apoptosis in WT mice vs. CD-WT mice. Co-treatment with the reactive oxygen species scavenger Tempol prevented FRD-induced apoptosis in WT mice. In contrast, FRD enhanced oxidative stress but not apoptosis in FRD-SR-AIP mice, in which a CaMKII inhibitor is targeted to the SR. FRD produced mitochondrial membrane depolarization in WT mice but not in S2814A mice, in which the CaMKII phosphorylation site on RyR2 was ablated. Furthermore, FRD decreased mitochondrial area, mean Feret diameter and mean SR-mitochondrial distance vs. CD-WT hearts. This remodelling was prevented in AC3I mice, with cardiac-targeted CaMKII inhibition. CaMKII phosphorylation of RyR2, SR Ca2+ leak and mitochondrial membrane depolarization are critically involved in the apoptotic pathway of the pre-diabetic heart. The FRD-induced decrease in SR-mitochondrial distance is likely to additionally favour Ca2+ transit between the two organelles.
Collapse
Affiliation(s)
- Marilen Federico
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Médicas, UNLP, La Plata, Argentina
| | - Enrique L Portiansky
- Laboratorio de Análisis de Imágenes, Facultad de Cs. Veterinarias, UNLP, La Plata, Argentina
| | - Leandro Sommese
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Médicas, UNLP, La Plata, Argentina
| | - Francisco J Alvarado
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Paula G Blanco
- Servicio de Ecocardiografía, Facultad de Veterinaria, UNLP, La Plata, Argentina
| | - Carolina N Zanuzzi
- Laboratorio de Análisis de Imágenes, Facultad de Cs. Veterinarias, UNLP, La Plata, Argentina
| | - John Dedman
- Department of Genome Science, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Marcia Kaetzel
- Department of Genome Science, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Departments of Molecular Physiology and Biophysics, Medicine (in Cardiology), Pediatrics; and Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Médicas, UNLP, La Plata, Argentina
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Médicas, UNLP, La Plata, Argentina
| |
Collapse
|
22
|
Szostak J, Boué S, Talikka M, Guedj E, Martin F, Phillips B, Ivanov NV, Peitsch MC, Hoeng J. Aerosol from Tobacco Heating System 2.2 has reduced impact on mouse heart gene expression compared with cigarette smoke. Food Chem Toxicol 2017; 101:157-167. [PMID: 28111298 DOI: 10.1016/j.fct.2017.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/10/2017] [Accepted: 01/18/2017] [Indexed: 02/05/2023]
Abstract
Experimental studies clearly demonstrate a causal effect of cigarette smoking on cardiovascular disease. To reduce the individual risk and population harm caused by smoking, alternative products to cigarettes are being developed. We recently reported on an apolipoprotein E-deficient (Apoe-/-) mouse inhalation study that compared the effects of exposure to aerosol from a candidate modified risk tobacco product, Tobacco Heating System 2.2 (THS2.2), and smoke from the reference cigarette (3R4F) on pulmonary and vascular biology. Here, we applied a transcriptomics approach to evaluate the impact of the exposure to 3R4F smoke and THS2.2 aerosol on heart tissues from the same cohort of mice. The systems response profiles demonstrated that 3R4F smoke exposure led to time-dependent transcriptomics changes (False Discovery Rate (FDR) < 0.05; 44 differentially expressed genes at 3-months; 491 at 8-months). Analysis of differentially expressed genes in the heart tissue indicated that 3R4F exposure induced the downregulation of genes involved in cytoskeleton organization and the contractile function of the heart, notably genes that encode beta actin (Actb), actinin alpha 4 (Actn4), and filamin C (Flnc). This was accompanied by the downregulation of genes related to the inflammatory response. None of these effects were observed in the group exposed to THS2.2 aerosol.
Collapse
Affiliation(s)
- Justyna Szostak
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Stéphanie Boué
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Marja Talikka
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Emmanuel Guedj
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Florian Martin
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Blaine Phillips
- Philip Morris International Research Laboratories Pte Ltd, Science Park II, Singapore.
| | - Nikolai V Ivanov
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Manuel C Peitsch
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Julia Hoeng
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| |
Collapse
|
23
|
Abd El-Kader SM, Saiem Al-Dahr MH. Impact of weight loss on oxidative stress and inflammatory cytokines in obese type 2 diabetic patients. Afr Health Sci 2016; 16:725-733. [PMID: 27917205 DOI: 10.4314/ahs.v16i3.12] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Type 2 diabetes mellitus is associated with abnormal markers of inflammatory cytokines and oxidative stress markers. Although, these abnormalities could be modulated with weight reduction; there is limitation in clinical studies that have addressed the beneficial effects of weight reduction in modulating biomarkers of inflammatory cytokines and oxidative stress for obesity associated with type 2 diabetes mellitus. OBJECTIVE This study was designed to detect the effects of weight loss on the inflammatory cytokines, oxidative stress markers in obese type 2 diabetic patients. MATERIAL AND METHODS Eighty obese patients with type 2 diabetes mellitus, their age ranged from 35-57 years and their body mass index ranged from 31-35 kg/m2 were equally assigned into 2 groups: the weight reduction group received aerobic exercises, diet regimen, where as the control group received medical treatment only for 12 weeks. RESULTS The mean values of body mass index (BMI), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), C-reactive protein (sCRP), conjugated dienes (CD) and malondialdehyde (MDA) were significantly decreased, while the mean values of glutathione peroxidase (GPx), superoxide dismutase (SOD) and glutathione (GSH) were significantly increased in patients of group (A), while changes were not significant in group (B). Also, there were significant differences between mean levels of the investigated parameters in group (A) and group (B) at the end of the study. CONCLUSION Weight loss ameliorates inflammatory cytokines and oxidative stress markers in obese type 2 diabetic patients.
Collapse
Affiliation(s)
- Shehab M Abd El-Kader
- Department of Physical Therapy, Faculty of Applied Medical Sciences, King Abdulaziz University
| | - Mohammed H Saiem Al-Dahr
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University
| |
Collapse
|
24
|
Wu HE, Baumgardt SL, Fang J, Paterson M, Liu Y, Du J, Shi Y, Qiao S, Bosnjak ZJ, Warltier DC, Kersten JR, Ge ZD. Cardiomyocyte GTP Cyclohydrolase 1 Protects the Heart Against Diabetic Cardiomyopathy. Sci Rep 2016; 6:27925. [PMID: 27295516 PMCID: PMC4904741 DOI: 10.1038/srep27925] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/26/2016] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy increases the risk of heart failure and death. At present, there are no effective approaches to preventing its development in the clinic. Here we report that reduction of cardiac GTP cyclohydrolase 1 (GCH1) degradation by genetic and pharmacological approaches protects the heart against diabetic cardiomyopathy. Diabetic cardiomyopathy was induced in C57BL/6 wild-type mice and transgenic mice with cardiomyocyte-specific overexpression of GCH1 with streptozotocin, and control animals were given citrate buffer. We found that diabetes-induced degradation of cardiac GCH1 proteins contributed to adverse cardiac remodeling and dysfunction in C57BL/6 mice, concomitant with decreases in tetrahydrobiopterin, dimeric and phosphorylated neuronal nitric oxide synthase, sarcoplasmic reticulum Ca(2+) handling proteins, intracellular [Ca(2+)]i, and sarcoplasmic reticulum Ca(2+) content and increases in phosphorylated p-38 mitogen-activated protein kinase and superoxide production. Interestingly, GCH-1 overexpression abrogated these detrimental effects of diabetes. Furthermore, we found that MG 132, an inhibitor for 26S proteasome, preserved cardiac GCH1 proteins and ameliorated cardiac remodeling and dysfunction during diabetes. This study deepens our understanding of impaired cardiac function in diabetes, identifies GCH1 as a modulator of cardiac remodeling and function, and reveals a new therapeutic target for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Hsiang-En Wu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MA 21224, USA
| | - Shelley L. Baumgardt
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Mark Paterson
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Yanan Liu
- Department of Medicine, Columbia University, 630 W. 168th Street, New York, NY 10032, USA
| | - Jianhai Du
- Department of Biochemistry, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Yang Shi
- Aurora Research Institute, Aurora Health Care, 750 W. Virginia Street, Milwaukee, WI 53234, USA
| | - Shigang Qiao
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Zeljko J. Bosnjak
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - David C. Warltier
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Judy R. Kersten
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Zhi-Dong Ge
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
25
|
Forkhead box transcription factor 1: role in the pathogenesis of diabetic cardiomyopathy. Cardiovasc Diabetol 2016; 15:44. [PMID: 26956801 PMCID: PMC4784400 DOI: 10.1186/s12933-016-0361-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/02/2016] [Indexed: 12/17/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a disorder of the heart muscle in people with diabetes that can occur independent of hypertension or vascular disease. The underlying mechanism of DCM is incompletely understood. Some transcription factors have been suggested to regulate the gene program intricate in the pathogenesis of diabetes prompted cardiac injury. Forkhead box transcription factor 1 is a pleiotropic transcription factor that plays a pivotal role in a variety of physiological processes. Altered FOXO1 expression and function have been associated with cardiovascular diseases, and the important role of FOXO1 in DCM has begun to attract attention. In this review, we focus on the FOXO1 pathway and its role in various processes that have been related to DCM, such as metabolism, oxidative stress, endothelial dysfunction, inflammation and apoptosis.
Collapse
|
26
|
Abstract
Cardiovascular disease, including heart failure (HF), is the major cause of death in patients with diabetes. A contributing factor to the occurrence of HF in such patients is the development of diabetic cardiomyopathy. Recent evidence demonstrates that perturbations associated with adipokines secretion and signaling result in lusitropic and inotropic defects in diabetic cardiomyopathy. This perspective editorial will discuss the central role of resistin, a recently discovered adipokine, in the maladaptive cardiac phenotype seen in diabetic hearts. Given the pleiotropic effects of resistin, strategies targeting the control of resistin levels may constitute a potentially viable therapeutic utility in patients with diabetes and diabetes-induced cardiovascular diseases.
Collapse
Affiliation(s)
- Djamel Lebeche
- Cardiovascular Research Institute, Graduate School of Biological Sciences, Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
27
|
Yilmaz S, Canpolat U, Aydogdu S, Abboud HE. Diabetic Cardiomyopathy; Summary of 41 Years. Korean Circ J 2015; 45:266-72. [PMID: 26240579 PMCID: PMC4521103 DOI: 10.4070/kcj.2015.45.4.266] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/27/2015] [Accepted: 03/05/2015] [Indexed: 01/04/2023] Open
Abstract
Patients with diabetes have an increased risk for development of cardiomyopathy, even in the absence of well known risk factors like coronary artery disease and hypertension. Diabetic cardiomyopathy was first recognized approximately four decades ago. To date, several pathophysiological mechanisms thought to be responsible for this new entity have also been recognized. In the presence of hyperglycemia, non-enzymatic glycosylation of several proteins, reactive oxygen species formation, and fibrosis lead to impairment of cardiac contractile functions. Impaired calcium handling, increased fatty acid oxidation, and increased neurohormonal activation also contribute to this process. Demonstration of left ventricular hypertrophy, early diastolic and late systolic dysfunction by sensitive techniques, help us to diagnose diabetic cardiomyopathy. Traditional treatment of heart failure is beneficial in diabetic cardiomyopathy, but specific strategies for prevention or treatment of cardiac dysfunction in diabetic patients has not been clarified yet. In this review we will discuss clinical and experimental studies focused on pathophysiology of diabetic cardiomyopathy, and summarize diagnostic and therapeutic approaches developed towards this entity.
Collapse
Affiliation(s)
- Samet Yilmaz
- Turkey Yuksek Ihtisas Education and Research Hospital, Cardiology Clinic, Ankara, Turkey
| | - Ugur Canpolat
- Turkey Yuksek Ihtisas Education and Research Hospital, Cardiology Clinic, Ankara, Turkey
| | - Sinan Aydogdu
- Turkey Yuksek Ihtisas Education and Research Hospital, Cardiology Clinic, Ankara, Turkey
| | - Hanna Emily Abboud
- Division of Nephrology, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
28
|
Rochette L, Zeller M, Cottin Y, Vergely C. Diabetes, oxidative stress and therapeutic strategies. Biochim Biophys Acta Gen Subj 2014; 1840:2709-29. [PMID: 24905298 DOI: 10.1016/j.bbagen.2014.05.017] [Citation(s) in RCA: 332] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/12/2014] [Accepted: 05/27/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Diabetes has emerged as a major threat to health worldwide. SCOPE OF REVIEW The exact mechanisms underlying the disease are unknown; however, there is growing evidence that excess generation of reactive oxygen species (ROS), largely due to hyperglycemia, causes oxidative stress in a variety of tissues. Oxidative stress results from either an increase in free radical production, or a decrease in endogenous antioxidant defenses, or both. ROS and reactive nitrogen species (RNS) are products of cellular metabolism and are well recognized for their dual role as both deleterious and beneficial species. In type 2 diabetic patients, oxidative stress is closely associated with chronic inflammation. Multiple signaling pathways contribute to the adverse effects of glucotoxicity on cellular functions. There are many endogenous factors (antioxidants, vitamins, antioxidant enzymes, metal ion chelators) that can serve as endogenous modulators of the production and action of ROS. Clinical trials that investigated the effect of antioxidant vitamins on the progression of diabetic complications gave negative or inconclusive results. This lack of efficacy might also result from the fact that they were administered at a time when irreversible alterations in the redox status are already under way. Another strategy to modulate oxidative stress is to exploit the pleiotropic properties of drugs directed primarily at other targets and thus acting as indirect antioxidants. MAJOR CONCLUSIONS It appears important to develop new compounds that target key vascular ROS producing enzymes and mimic endogenous antioxidants. GENERAL SIGNIFICANCE This strategy might prove clinically relevant in preventing the development and/or retarding the progression of diabetes associated with vascular diseases.
Collapse
Affiliation(s)
- Luc Rochette
- Laboratoire de Physiopathologie et Pharmacologie Cardio-Métaboliques, INSERM UMR866, Université de Bourgogne, Facultés de Médecine et Pharmacie, 7 Boulevard Jeanne d'Arc, 21079 Dijon, France.
| | - Marianne Zeller
- Laboratoire de Physiopathologie et Pharmacologie Cardio-Métaboliques, INSERM UMR866, Université de Bourgogne, Facultés de Médecine et Pharmacie, 7 Boulevard Jeanne d'Arc, 21079 Dijon, France
| | - Yves Cottin
- Laboratoire de Physiopathologie et Pharmacologie Cardio-Métaboliques, INSERM UMR866, Université de Bourgogne, Facultés de Médecine et Pharmacie, 7 Boulevard Jeanne d'Arc, 21079 Dijon, France
| | - Catherine Vergely
- Laboratoire de Physiopathologie et Pharmacologie Cardio-Métaboliques, INSERM UMR866, Université de Bourgogne, Facultés de Médecine et Pharmacie, 7 Boulevard Jeanne d'Arc, 21079 Dijon, France
| |
Collapse
|
29
|
Triposkiadis F, Giamouzis G, Butler J. The importance of managing diabetes correctly to prevent heart failure. Expert Rev Cardiovasc Ther 2014; 9:257-9. [DOI: 10.1586/erc.11.6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
30
|
Crespo MJ, Cruz N, Quidgley J, Torres H, Hernandez C, Casiano H, Rivera K. Daily Administration of Atorvastatin and Simvastatin for One Week Improves Cardiac Function in Type 1 Diabetic Rats. Pharmacology 2014; 93:84-91. [DOI: 10.1159/000358256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 12/24/2013] [Indexed: 12/15/2022]
|
31
|
Joshi M, Kotha SR, Malireddy S, Selvaraju V, Satoskar AR, Palesty A, McFadden DW, Parinandi NL, Maulik N. Conundrum of pathogenesis of diabetic cardiomyopathy: role of vascular endothelial dysfunction, reactive oxygen species, and mitochondria. Mol Cell Biochem 2013; 386:233-49. [PMID: 24307101 DOI: 10.1007/s11010-013-1861-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 10/09/2013] [Indexed: 12/11/2022]
Abstract
Diabetic cardiomyopathy and heart failure have been recognized as the leading causes of mortality among diabetics. Diabetic cardiomyopathy has been characterized primarily by the manifestation of left ventricular dysfunction that is independent of coronary artery disease and hypertension among the patients affected by diabetes mellitus. A complex array of contributing factors including the hypertrophy of left ventricle, alterations of metabolism, microvascular pathology, insulin resistance, fibrosis, apoptotic cell death, and oxidative stress have been implicated in the pathogenesis of diabetic cardiomyopathy. Nevertheless, the exact mechanisms underlying the pathogenesis of diabetic cardiomyopathy are yet to be established. The critical involvement of multifarious factors including the vascular endothelial dysfunction, microangiopathy, reactive oxygen species (ROS), oxidative stress, mitochondrial dysfunction has been identified in the mechanism of pathogenesis of diabetic cardiomyopathy. Although it is difficult to establish how each factor contributes to disease, the involvement of ROS and mitochondrial dysfunction are emerging as front-runners in the mechanism of pathogenesis of diabetic cardiomyopathy. This review highlights the role of vascular endothelial dysfunction, ROS, oxidative stress, and mitochondriopathy in the pathogenesis of diabetic cardiomyopathy. Furthermore, the review emphasizes that the puzzle has to be solved to firmly establish the mitochondrial and/or ROS mechanism(s) by identifying their most critical molecular players involved at both spatial and temporal levels in diabetic cardiomyopathy as targets for specific and effective pharmacological/therapeutic interventions.
Collapse
Affiliation(s)
- Mandip Joshi
- Department of Surgery, University of Connecticut Health Center, Farmington Avenue, Farmington, CT, 06032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Turer AT, Hill JA, Elmquist JK, Scherer PE. Adipose tissue biology and cardiomyopathy: translational implications. Circ Res 2013; 111:1565-77. [PMID: 23223931 DOI: 10.1161/circresaha.111.262493] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
It is epidemiologically established that obesity is frequently associated with the metabolic syndrome and poses an increased risk for the development of type 2 diabetes mellitus and cardiovascular disease. The molecular links that connect the phenomenon of obesity, per se, with insulin resistance and cardiovascular disease are still not fully elucidated. It is increasingly apparent that fully functional adipose tissue can be cardioprotective by reducing lipotoxic effects in other peripheral tissues and by maintaining a healthy balance of critical adipokines, thereby allowing the heart to maintain its full metabolic flexibility. The present review highlights both basic and clinical findings that emphasize the complex interplay of adipose tissue physiology and adipokine-mediated effects on the heart exerted by either direct effects on cardiac myocytes or indirect actions via central mechanisms through sympathetic outflow to the heart.
Collapse
Affiliation(s)
- Aslan T Turer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | |
Collapse
|
33
|
Ilkun O, Boudina S. Cardiac dysfunction and oxidative stress in the metabolic syndrome: an update on antioxidant therapies. Curr Pharm Des 2013; 19:4806-17. [PMID: 23323621 DOI: 10.2174/1381612811319270003] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 01/10/2013] [Indexed: 01/14/2023]
Abstract
The metabolic syndrome (MetS) is a cluster of risk factors including obesity, insulin resistance, dyslipidemia, elevated blood pressure and glucose intolerance. The MetS increases the risk for cardiovascular disease (CVD) and type 2 diabetes. Each component of the MetS causes cardiac dysfunction and their combination carries additional risk. The mechanisms underlying cardiac dysfunction in the MetS are complex and might include lipid accumulation, increased fibrosis and stiffness, altered calcium homeostasis, abnormal autophagy, altered substrate utilization, mitochondrial dysfunction and increased oxidative stress. Mitochondrial and extra-mitochondrial sources of reactive oxygen species (ROS) and reduced antioxidant defense mechanisms characterize the myocardium of humans and animals with the MetS. The mechanisms for increased cardiac oxidative stress in the MetS are not fully understood but include increased fatty acid oxidation, mitochondrial dysfunction and enhanced NADPH oxidase activity. Therapies aimed to reduce oxidative stress and enhance antioxidant defense have been employed to reduce cardiac dysfunction in the MetS in animals. In contrast, large scale clinical trials using antioxidants therapies for the treatment of CVD have been disappointing because of the lack of efficacy and undesired side effects. The focus of this review is to summarize the current knowledge about the mechanisms underlying cardiac dysfunction in the MetS with a special interest in the role of oxidative stress. Finally, we will update the reader on the results obtained with natural antioxidant and mitochondria-targeted antioxidant therapies for the treatment of CVD in the MetS.
Collapse
Affiliation(s)
- Olesya Ilkun
- Division of Endocrinology, Metabolism and Diabetes, Program in Human Molecular Biology & Genetics, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| | | |
Collapse
|
34
|
Tocchetti CG, Caceres V, Stanley BA, Xie C, Shi S, Watson WH, O’Rourke B, Spadari-Bratfisch RC, Cortassa S, Akar FG, Paolocci N, Aon MA. GSH or palmitate preserves mitochondrial energetic/redox balance, preventing mechanical dysfunction in metabolically challenged myocytes/hearts from type 2 diabetic mice. Diabetes 2012; 61:3094-105. [PMID: 22807033 PMCID: PMC3501888 DOI: 10.2337/db12-0072] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In type 2 diabetes, hyperglycemia and increased sympathetic drive may alter mitochondria energetic/redox properties, decreasing the organelle's functionality. These perturbations may prompt or sustain basal low-cardiac performance and limited exercise capacity. Yet the precise steps involved in this mitochondrial failure remain elusive. Here, we have identified dysfunctional mitochondrial respiration with substrates of complex I, II, and IV and lowered thioredoxin-2/glutathione (GSH) pools as the main processes accounting for impaired state 4→3 energetic transition shown by mitochondria from hearts of type 2 diabetic db/db mice upon challenge with high glucose (HG) and the β-agonist isoproterenol (ISO). By mimicking clinically relevant conditions in type 2 diabetic patients, this regimen triggers a major overflow of reactive oxygen species (ROS) from mitochondria that directly perturbs cardiac electro-contraction coupling, ultimately leading to heart dysfunction. Exogenous GSH or, even more so, the fatty acid palmitate rescues basal and β-stimulated function in db/db myocyte/heart preparations exposed to HG/ISO. This occurs because both interventions provide the reducing equivalents necessary to counter mitochondrial ROS outburst and energetic failure. Thus, in the presence of poor glycemic control, the diabetic patient's inability to cope with increased cardiac work demand largely stems from mitochondrial redox/energetic disarrangements that mutually influence each other, leading to myocyte or whole-heart mechanical dysfunction.
Collapse
Affiliation(s)
- Carlo G. Tocchetti
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Viviane Caceres
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brian A. Stanley
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chaoqin Xie
- Cardiovascular Research Center, Division of Cardiology, Mount Sinai School of Medicine, New York, New York
| | - Sa Shi
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Walter H. Watson
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, Kentucky
| | - Brian O’Rourke
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Sonia Cortassa
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Fadi G. Akar
- Cardiovascular Research Center, Division of Cardiology, Mount Sinai School of Medicine, New York, New York
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Dipartimento di Medicina Clinica e Sperimentale, Universita di Perugia, Perugia, Italy
| | - Miguel A. Aon
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Corresponding author: Miguel A. Aon,
| |
Collapse
|
35
|
Abstract
CONTEXT Recent studies on mediators of inflammation, experimental models of atherosclerosis, and acute ischemia have identified novel mechanisms through which insulin may exert cardiovascular protective effects. This review aims to summarize current knowledge regarding the cardiovascular, antiinflammatory, and antiatherogenic effects of insulin, and the effect of intensive glycemic control in acute cardiovascular disease. EVIDENCE ACQUISITION Publications of interest were identified using preselected MeSH terminology and keywords to search online databases such as PubMed and OVID for the period January 1988 to February 2012. Relevant publications were obtained and reviewed by two independent observers, then evaluated a priori against the following criteria: study quality, main clinical outcomes, and applicability to clinical practice. EVIDENCE SYNTHESIS Insulin has been shown to exert vasodilatory, antiinflammatory, and antiatherogenic effects in experimental models, independent of its glucose-lowering effects. Additionally, glucose is known to exert potent proinflammatory, prothrombotic, and proapoptotic effects during myocardial infarct, indicative that hyperglycemia is likely to be injurious to the heart. In this context, through its nonmetabolic and metabolic (glucose-lowering) effects, insulin is likely to be cardioprotective and to improve clinical outcomes in acute myocardial infarction. CONCLUSIONS Despite promising experimental data and evidence of benefit from single-center randomized clinical trials, clinical evidence supporting the cardioprotective effects of insulin from a multicenter randomized clinical trial is still lacking. Future prospective studies with insulin infused at adequate concentrations, individually titrated to achieve and maintain euglycemia (blood glucose < 140 mg/dl) and minimize hypoglycemia, are required to investigate the role of insulin in the management of patients with acute cardiovascular disease.
Collapse
Affiliation(s)
- Ajay Chaudhuri
- Diabetes-Endocrinology Center of Western New York, Division of Endocrinology, Kaleida Health/Millard Fillmore Hospital, Buffalo, New York, USA
| | | | | |
Collapse
|
36
|
Li CJ, Lv L, Li H, Yu DM. Cardiac fibrosis and dysfunction in experimental diabetic cardiomyopathy are ameliorated by alpha-lipoic acid. Cardiovasc Diabetol 2012; 11:73. [PMID: 22713251 PMCID: PMC3472273 DOI: 10.1186/1475-2840-11-73] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 06/08/2012] [Indexed: 12/17/2022] Open
Abstract
Background Alpha-lipoic acid (ALA), a naturally occurring compound, exerts powerful protective effects in various cardiovascular disease models. However, its role in protecting against diabetic cardiomyopathy (DCM) has not been elucidated. In this study, we have investigated the effects of ALA on cardiac dysfunction, mitochondrial oxidative stress (MOS), extracellular matrix (ECM) remodeling and interrelated signaling pathways in a diabetic rat model. Methods Diabetes was induced in rats by I.V. injection of streptozotocin (STZ) at 45 mg/kg. The animals were randomly divided into 4 groups: normal groups with or without ALA treatment, and diabetes groups with or without ALA treatment. All studies were carried out 11 weeks after induction of diabetes. Cardiac catheterization was performed to evaluate cardiac function. Mitochondrial oxidative biochemical parameters were measured by spectophotometeric assays. Extracellular matrix content (total collagen, type I and III collagen) was assessed by staining with Sirius Red. Gelatinolytic activity of Pro- and active matrix metalloproteinase-2 (MMP-2) levels were analyzed by a zymogram. Cardiac fibroblasts differentiation to myofibroblasts was evaluated by Western blot measuring smooth muscle actin (α-SMA) and transforming growth factor–β (TGF-β). Key components of underlying signaling pathways including the phosphorylation of c-Jun N-terminal kinase (JNK), p38 MAPK and ERK were also assayed by Western blot. Results DCM was successfully induced by the injection of STZ as evidenced by abnormal heart mass and cardiac function, as well as the imbalance of ECM homeostasis. After administration of ALA, left ventricular dysfunction greatly improved; interstitial fibrosis also notably ameliorated indicated by decreased collagen deposition, ECM synthesis as well as enhanced ECM degradation. To further assess the underlying mechanism of improved DCM by ALA, redox status and cardiac remodeling associated signaling pathway components were evaluated. It was shown that redox homeostasis was disturbed and MAPK signaling pathway components activated in STZ-induced DCM animals. While ALA treatment favorably shifted redox homeostasis and suppressed JNK and p38 MAPK activation. Conclusions These results, coupled with the excellent safety and tolerability profile of ALA in humans, demonstrate that ALA may have therapeutic potential in the treatment of DCM by attenuating MOS, ECM remodeling and JNK, p38 MAPK activation.
Collapse
Affiliation(s)
- Chun-jun Li
- Key Laboratory of Hormone and Development (Ministry of Health), Metabolic Disease Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | | | | | | |
Collapse
|
37
|
He X, Ma Q. Disruption of Nrf2 Synergizes with High Glucose to Cause Heightened Myocardial Oxidative Stress and Severe Cardiomyopathy in Diabetic Mice. ACTA ACUST UNITED AC 2012; Suppl 7. [PMID: 26691239 DOI: 10.4172/2155-6156.s7-002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
High glucose-induced oxidative stress is a major contributing mechanism to the development of diabetic cardiomyopathy. Nrf2 is an emerging critical regulator of cellular defense against oxidative damage. The role of Nrf2 in diabetic cardiomyopathy was investigated in vivo. Streptozotocin (STZ) induced diabetes in Nrf2 knockout (KO) mice that rapidly progressed to severe conditions with high mortality within two weeks of injection; whereas, in wild type (WT) mice, diabetes was less severe with no death. Severe myocardial lesions were observed in diabetic KO mice that had high, sublethal levels of blood glucose including: (a) irregular myocardial arrangements, myofibrillar discontinuation, and cell death; (b) reduced electron density, discontinuation of myocardial fibers, and mitochondrial damage; and (c) markedly reduced contractility of the cardiomyocytes to β-agonist stimulation. Parallel to severe cardiomyopathy, the diabetic KO hearts showed: (a) increased apoptosis as revealed by TUNEL and PARP1 cleavage assays; (b) infiltration of granulocytes and macrophages as well as fibrosis indicating robust inflammatory response; and (c) heightened oxidative stress as evidenced by increased levels of 8-hydroxydeoxyquanine, free malondialdehyde, and 3-nitrotyrosine. Increased oxidative stress in the KO hearts was attributed to decrease or loss of the basal and induced expression of Nrf2-dependent cytoprotective genes. Our findings demonstrate that loss of Nrf2 function synergizes with high glucose to cause heightened oxidative stress in the heart leading to severe diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Xiaoqing He
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505, USA
| | - Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505, USA
| |
Collapse
|
38
|
Bai SZ, Sun J, Wu H, Zhang N, Li HX, Li GW, Li HZ, He W, Zhang WH, Zhao YJ, Wang LN, Tian Y, Yang BF, Yang GD, Wu LY, Wang R, Xu CQ. Decrease in calcium-sensing receptor in the progress of diabetic cardiomyopathy. Diabetes Res Clin Pract 2012; 95:378-85. [PMID: 22137362 DOI: 10.1016/j.diabres.2011.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 10/31/2011] [Accepted: 11/07/2011] [Indexed: 02/06/2023]
Abstract
To observe the dynamic expression of calcium-sensing receptor (CaSR) in myocardium of diabetic rats and explore its role in diabetic cardiomyopathy (DCM), 40 male Wistar rats were randomly divided into 4 groups including control, diabetic-4 weeks, diabetic-8 weeks and spermine treatment groups (240 μM of spermine in drinking water). The type 2 Diabetes mellitus (DM) models were established by intraperitoneal injection of streptozotocin (STZ, 30 mg/kg) after high-fat and high-sugar diet for one month. The echocardiographic parameters were measured, cardiac morphology was observed by electron microscope and HE staining. The intracellular calcium concentration ([Ca(2+)](i)) was detected by laser-scanning confocal microscope. Western blot analyzed the expression of CaSR, protein kinase C α(PKC-α) and calcium handling regulators, such as phospholamban (PLN), Ca(2+)-ATPase (SERCA), and ryanodine receptor (RyR). Compared with control group, [Ca(2+)](i) and the expression of CaSR, RyR and SERCA/PLN were decreased, while PKC-α and PLN were significantly increased in a time-dependent manner in diabetic groups. Meanwhile diabetic rats displayed abnormal cardiac structure and systolic and diastolic dysfunction, and spermine (CaSR agonist) could prevent or slow its progression. These results indicate that the CaSR expression of myocardium is reduced in the progress of DCM, and its potential mechanism is related to the impaired intracellular calcium homeostasis.
Collapse
Affiliation(s)
- Shu-zhi Bai
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Battiprolu PK, Hojayev B, Jiang N, Wang ZV, Luo X, Iglewski M, Shelton JM, Gerard RD, Rothermel BA, Gillette TG, Lavandero S, Hill JA. Metabolic stress-induced activation of FoxO1 triggers diabetic cardiomyopathy in mice. J Clin Invest 2012; 122:1109-18. [PMID: 22326951 DOI: 10.1172/jci60329] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 01/04/2012] [Indexed: 12/26/2022] Open
Abstract
The leading cause of death in diabetic patients is cardiovascular disease; diabetic cardiomyopathy is typified by alterations in cardiac morphology and function, independent of hypertension or coronary disease. However, the molecular mechanism that links diabetes to cardiomyopathy is incompletely understood. Insulin resistance is a hallmark feature of diabetes, and the FoxO family of transcription factors, which regulate cell size, viability, and metabolism, are established targets of insulin and growth factor signaling. Here, we set out to evaluate a possible role of FoxO proteins in diabetic cardiomyopathy. We found that FoxO proteins were persistently activated in cardiac tissue in mice with diabetes induced either genetically or by high-fat diet (HFD). FoxO activity was critically linked with development of cardiomyopathy: cardiomyocyte-specific deletion of FoxO1 rescued HFD-induced declines in cardiac function and preserved cardiomyocyte insulin responsiveness. FoxO1-depleted cells displayed a shift in their metabolic substrate usage, from free fatty acids to glucose, associated with decreased accumulation of lipids in the heart. Furthermore, we found that FoxO1-dependent downregulation of IRS1 resulted in blunted Akt signaling and insulin resistance. Together, these data suggest that activation of FoxO1 is an important mediator of diabetic cardiomyopathy and is a promising therapeutic target for the disease.
Collapse
Affiliation(s)
- Pavan K Battiprolu
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75390-8573, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
The heart exhibits remarkable adaptive responses to a wide array of genetic and extrinsic factors to maintain contractile function. When compensatory responses are not sustainable, cardiac dysfunction occurs, leading to cardiomyopathy. The many forms of cardiomyopathy exhibit a set of overlapping phenotypes reflecting the limited range of compensatory responses that the heart can use. These include cardiac hypertrophy, induction of genes normally expressed during development, fibrotic deposits that replace necrotic and apoptotic cardiomyocytes, and metabolic disturbances. The compensatory responses are mediated by signaling pathways that initially serve to maintain normal contractility; however, persistent activation of these pathways leads to cardiac dysfunction. Current research focuses on ways to target these specific pathways therapeutically.
Collapse
Affiliation(s)
- Pamela A Harvey
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado at Boulder, USA
| | | |
Collapse
|
41
|
Rodríguez-Penas D, Feijóo-Bandín S, Lear PV, Mosquera-Leal A, García-Rúa V, Otero MF, Rivera M, Gualillo O, González-Juanatey JR, Lago F. Aliskiren affects fatty-acid uptake and lipid-related genes in rodent and human cardiomyocytes. Biochem Pharmacol 2011; 82:491-504. [DOI: 10.1016/j.bcp.2011.05.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 05/13/2011] [Accepted: 05/18/2011] [Indexed: 12/13/2022]
|
42
|
Rajamani U, Joseph D, Roux S, Essop MF. The hexosamine biosynthetic pathway can mediate myocardial apoptosis in a rat model of diet-induced insulin resistance. Acta Physiol (Oxf) 2011; 202:151-7. [PMID: 21385329 DOI: 10.1111/j.1748-1716.2011.02275.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AIMS Type 2 diabetes is characterized by deranged metabolic pathways that may result in cardiovascular complications. For example, hyperglycaemia promotes flux through the hexosamine biosynthetic pathway (HBP) leading to greater O-GlcNAcylation of target proteins, with pathophysiological outcomes. This study investigated mechanisms whereby increased HBP flux elicits myocardial apoptosis in a rat model of diet-induced hyperglycaemia/insulin resistance. METHODS Four-week-old male Wistar rats were fed a high-fat diet (86 days) after which insulin resistance was assessed vs. matched controls. Oxidative stress was evaluated, and apoptotic peptide levels, BAD phosphorylation and overall O-GlcNAcylation assessed by immunoblotting. Protein-specific O-GlcNAcylation and BAD-Bcl-2 dimerization were determined by immunoprecipitation and Western blotting. RESULTS Rats consuming the high-fat diet exhibited a moderate elevation in body weight, higher fasting insulin and glucose levels, and insulin resistance vs. controls. Overall protein O-GlcNAcylation was increased in hyperglycaemic/insulin-resistant hearts. In parallel, myocardial peptide levels of apoptotic markers (caspase-3, cytochrome-c, BAD) were significantly higher with insulin resistance. To gain mechanistic insight into our findings, we evaluated O-GlcNAcylation of BAD, a pro-apoptotic Bcl-2 homolog. Here we found increased BAD O-GlcNAcylation and decreased BAD phosphorylation (Ser136) in hyperglycaemic/insulin-resistant rat hearts. These data are in agreement with competition by phosphorylation and O-GlcNAcylation for the same or neighbouring site(s) on target proteins. Moreover, we observed increased BAD-Bcl-2 dimerization in hyperglycaemic/insulin-resistant hearts. CONCLUSION The main finding of this study is that increased apoptosis in hyperglycaemic/insulin-resistant hearts can also be mediated through HBP-induced BAD O-GlcNAcylation and greater formation of BAD-Bcl-2 dimers (pro-apoptotic).
Collapse
Affiliation(s)
- U Rajamani
- Cardio-Metabolic Research Group, Department of Physiological Sciences, Stellenbosch University, South Africa
| | | | | | | |
Collapse
|
43
|
Chemaly ER, Hadri L, Zhang S, Kim M, Kohlbrenner E, Sheng J, Liang L, Chen J, K-Raman P, Hajjar RJ, Lebeche D. Long-term in vivo resistin overexpression induces myocardial dysfunction and remodeling in rats. J Mol Cell Cardiol 2011; 51:144-55. [PMID: 21549710 DOI: 10.1016/j.yjmcc.2011.04.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 04/05/2011] [Accepted: 04/17/2011] [Indexed: 10/18/2022]
Abstract
We have previously reported that resistin induces hypertrophy and impairs contractility in isolated rat cardiomyocytes. To examine the long-term cardiovascular effects of resistin, we induced in vivo overexpression of resistin using adeno-associated virus serotype 9 injected by tail vein in rats and compared to control animals. Ten weeks after viral injection, overexpression of resistin was associated with increased ratio of left ventricular (LV) weight/body weight, increased end-systolic LV volume and significant decrease in LV contractility, measured by the end-systolic pressure volume relationship slope in LV pressure volume loops, compared to controls. At the molecular level, mRNA expression of ANF and β-MHC, and protein levels of phospholamban were increased in the resistin group without a change in the level of SERCA2a protein expression. Increased fibrosis by histology, associated with increased mRNA levels of collagen, fibronectin and connective tissue growth factor were observed in the resistin-overexpressing hearts. Resistin overexpression was also associated with increased apoptosis in vivo, along with an apoptotic molecular phenotype in vivo and in vitro. Resistin-overexpressing LV tissue had higher levels of TNF-α receptor 1 and iNOS, and reduced levels of eNOS. Cardiomyocytes overexpressing resistin in vitro produced larger amounts of TNFα in the medium, had increased phosphorylation of IκBα and displayed increased intracellular reactive oxygen species (ROS) content with increased expression and activity of ROS-producing NADPH oxidases compared to controls. Long-term resistin overexpression is associated with a complex phenotype of oxidative stress, inflammation, fibrosis, apoptosis and myocardial remodeling and dysfunction in rats. This phenotype recapitulates key features of diabetic cardiomyopathy. This article is part of Special Issue Item Group entitled "Possible Editorial".
Collapse
Affiliation(s)
- Elie R Chemaly
- Cardiovascular Research Institute, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW A significant number of patients hospitalized with heart failure are malnourished. Depletion of micronutrients, which is known to occur in heart failure for a variety of reasons, may contribute to myocardial abnormalities noted in heart failure. In this review, we focus on nutritional supplementation strategies that might improve myocardial performance and, as a consequence, decrease mortality and morbidity in these patients. RECENT FINDINGS The available data suggest that micronutrient and macronutrient supplementation may play a role in improving the myocardial metabolic abnormalities noted in heart failure. A recent trial of omega-3 fatty acid macronutrient supplementation showed a modest decrease in mortality and hospitalizations when used in patients with New York Heart Association class II-IV heart failure. SUMMARY Recommendations for nutritional support in patients with heart failure are difficult to make due to a lack of large randomized trials. Supplementation with omega-3 fatty acids, and micronutrients such as thiamine, coenzyme Q-10 and carnitine has shown promise in several studies. Since the data is not conclusive, large trials are needed to address whether these positive findings are reproducible in a wider subset of patients. In addition, these trials should study the combination of different micronutrients and macronutrients since heart failure patients are rarely deficient in just one micronutrient or macronutrient.
Collapse
|
45
|
Sun D, Shen M, Li J, Li W, Zhang Y, Zhao L, Zhang Z, Yuan Y, Wang H, Cao F. Cardioprotective effects of tanshinone IIA pretreatment via kinin B2 receptor-Akt-GSK-3β dependent pathway in experimental diabetic cardiomyopathy. Cardiovasc Diabetol 2011; 10:4. [PMID: 21232147 PMCID: PMC3033797 DOI: 10.1186/1475-2840-10-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Accepted: 01/13/2011] [Indexed: 11/26/2022] Open
Abstract
Aims Diabetic cardiomyopathy, characterized by myocardial structural and functional changes, is a specific cardiomyopathy develops in patients with diabetes mellitus. The present study was to investigate the role of kinin B2 receptor-Akt-glycogen synthase kinase (GSK)-3β signalling pathway in mediating the protective effects of tanshinone IIA (TSN) on diabetic cardiomyopathy. Methods and results Streptozocin (STZ) induced diabetic rats (n = 60) were randomized to receive TSN, TSN plus HOE140 (a kinin B2 receptor antagonist), or saline. Healthy Sprague-Dawley (SD) rats (n = 20) were used as control. Left ventricular function, myocardial apoptosis, myocardial ultrastructure, Akt, GSK-3β and NF-κB phosphorylation, the expression of TNF-α, IL-6 and myeloperoxidase (MPO) were examined. Cardiac function was well preserved as evidenced by increased left ventricular ejection fraction (LVEF) and ± dp/dt (maximum speed of contraction/relaxation), along with decreased myocardial apoptotic death after TSN administration. TSN pretreatment alleviated mitochondria ultrastructure changes. TSN also enhanced Akt and GSK-3β phosphorylation and inhibited NF-κB phosphorylation, resulting in decreased TNF-α, IL-6 and MPO activities. Moreover, pretreatment with HOE140 abolished the beneficial effects of TSN: a decrease in LVEF and ± dp/dt, an inhibition of cardiomyocyte apoptosis, a destruction of cardiomyocyte mitochondria cristae, a reduction of Akt and GSK-3β phosphorylation, an enhancement of NF-κB phosphorylation and an increase of TNF-α, IL-6 and MPO production. Conclusion These data indicated that TSN is cardioprotective in the context of diabetic cardiomyopathy through kinin B2 receptor-Akt-GSK-3β dependent pathway.
Collapse
Affiliation(s)
- Dongdong Sun
- Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Battiprolu PK, Gillette TG, Wang ZV, Lavandero S, Hill JA. Diabetic Cardiomyopathy: Mechanisms and Therapeutic Targets. ACTA ACUST UNITED AC 2010; 7:e135-e143. [PMID: 21274425 DOI: 10.1016/j.ddmec.2010.08.001] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The incidence and prevalence of diabetes mellitus are each increasing rapidly in our society. The majority of patients with diabetes succumb ultimately to heart disease, much of which stems from atherosclerotic disease and hypertension. However, cardiomyopathy can develop independent of elevated blood pressure or coronary artery disease, a process termed diabetic cardiomyopathy. This disorder is a complex diabetes-associated process characterized by significant changes in the physiology, structure, and mechanical function of the heart. Here, we review recently derived insights into mechanisms and molecular events involved in the pathogenesis of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Pavan K Battiprolu
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | | | | | | | | |
Collapse
|