1
|
Sen P, Hamers J, Sittig T, Shashikadze B, d'Ambrosio L, Stöckl JB, Bierschenk S, Zhang H, d'Alessio C, Zandbergen LM, Pauly V, Clauss S, Wolf E, Dendorfer A, Fröhlich T, Merkus D. Oxidative stress initiates hemodynamic change in CKD-induced heart disease. Basic Res Cardiol 2024; 119:957-971. [PMID: 39404904 PMCID: PMC11628585 DOI: 10.1007/s00395-024-01085-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 12/10/2024]
Abstract
Chronic kidney disease (CKD) predisposes to cardiac remodeling and coronary microvascular dysfunction. Studies in swine identified changes in microvascular structure and function, as well as changes in mitochondrial structure and oxidative stress. However, CKD was combined with metabolic derangement, thereby obscuring the contribution of CKD alone. Therefore, we studied the impact of CKD on the heart and combined proteome studies with measurement of cardiac function and perfusion to identify processes involved in cardiac remodeling in CKD. CKD was induced in swine at 10-12 weeks of age while sham-operated swine served as controls. 5-6 months later, left ventricular (LV) function and coronary flow reserve were measured. LC-MS-MS-based proteomic analysis of LV tissue was performed. LV myocardium and kidneys were histologically examined for interstitial fibrosis and oxidative stress. Renal embolization resulted in mild chronic kidney injury (increased fibrosis and urinary NGAL). PV loops showed LV dilation and increased wall stress, while preload recruitable stroke work was impaired in CKD. Quantitative proteomic analysis of LV myocardium and STRING pre-ranked functional analysis showed enrichments in pathways related to contractile function, reactive oxygen species, and extracellular matrix (ECM) remodeling, which were confirmed histologically and associated with impaired total anti-oxidant capacity. H2O2 exposure of myocardial slices from CKD, but not normal swine, impaired contractile function. Furthermore, in CKD, mitochondrial proteins were downregulated suggesting mitochondrial dysfunction which was associated with higher basal coronary blood flow. Thus, mild CKD induces alterations in mitochondrial proteins along with contractile proteins, oxidative stress and ECM remodeling, that were associated with changes in cardiac function and perfusion.
Collapse
Affiliation(s)
- Payel Sen
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Jules Hamers
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Theresa Sittig
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | | | - Laura d'Ambrosio
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | | | - Susanne Bierschenk
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Hengliang Zhang
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
| | - Chiara d'Alessio
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
| | - Lotte M Zandbergen
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Valerie Pauly
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Department of Medicine I, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Sebastian Clauss
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Department of Medicine I, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | | | - Andreas Dendorfer
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
| | | | - Daphne Merkus
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany.
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany.
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany.
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Akopova O, Korkach Y, Sagach V. The effects of ecdysterone and enalapril on nitric oxide synthesis and the markers of oxidative stress in streptozotocin-induced diabetes in rats: a comparative study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8089-8099. [PMID: 38789633 DOI: 10.1007/s00210-024-03154-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024]
Abstract
Cardiovascular functions in diabetes greatly depend on constitutive NOS (cNOS) activity. A comparative study of the effects of a steroid hormone ecdysterone and enalapril, an ACE inhibitor widely used to treat cardiac disorders on cNOS, inducible NOS (iNOS), xanthine oxidoreductase (XOR) activity, RNS, ROS, and lipid peroxidation in heart tissue in experimental diabetes was conducted. The rat model of diabetes was established by streptozotocin injection. NOS activity, NO2-, NO3-, uric acid, nitrosothiols, hydroperoxide, superoxide, and diene conjugate formation were studied spectrophotomerically. In diabetes, cNOS downregulation correlated with a dramatic fall of NO2- production and ~4.5-fold elevation of nitrosothiols, which agreed with a steep rise of iNOS activity, while NO3- remained close to control. Dramatic activation of XOR was observed, which correlated with the elevation of both superoxide production and nitrate reductase activity and resulted in strong lipid peroxidation. Ecdysterone and enalapril differently affected RNS metabolism. Ecdysterone moderately restored cNOS but strongly suppressed iNOS, which resulted in the reduction of NO3-, but full restoration of NO2- production. Enalapril better restored cNOS but less effectively suppressed iNOS, which promoted NO3- formation. Both drugs similarly inhibited XOR, which equally alleviated oxidative stress and lipid peroxidation. The synergistic action of iNOS and XOR was a plausible explanation for strong lipid peroxidation, abolished by the inhibition of iNOS and XOR by ecdysterone or enalapril. Complementary effects of ecdysterone and enalapril on cNOS, iNOS, and RNS are a promising basis for their combined use in the treatment of cardiovascular disorders caused by cNOS dysfunction in diabetes.
Collapse
Affiliation(s)
- Olga Akopova
- Stem cell laboratory, Bogomoletz Institute of Physiology, NAS of Ukraine, Kiev, Ukraine.
| | - Yulia Korkach
- Circulation department, Bogomoletz Institute of Physiology, NAS of Ukraine, Kiev, Ukraine
| | - Vadim Sagach
- Circulation department, Bogomoletz Institute of Physiology, NAS of Ukraine, Kiev, Ukraine
| |
Collapse
|
3
|
Chen J, Zhang J, Chen Y, Zeng H. Knockout of Sirtuin 3 in endothelial cells impairs endothelial-dependent relaxation and myogenic response in mice. Physiol Rep 2024; 12:e70060. [PMID: 39425510 PMCID: PMC11489619 DOI: 10.14814/phy2.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/21/2024] Open
Abstract
Sirtuin 3 has been shown to regulate endothelial function and coronary flow reserve in mice. Knockout of SIRT3 reduced endothelial nitric oxide synthase expression in the mouse hearts. In this study, we investigate whether endothelial SIRT3 regulates vascular function and myogenic responses in distal intramural branches of the left anterior descending coronary artery (CA) and middle cerebral artery (MCA) of mice. Both male and female endothelial SIRT3 knockout (SIRT3ECKO) mice and control SIRT3LoxP mice were used and CA and MCA were dissected and mounted in a myograph system. The myogenic response was evaluated by measuring changes in inner diameter in response to 20 mmHg stepwise increases in intraluminal pressure in PSS (active diameter) and Ca2+-free PSS (passive diameter). Acetylcholine (Ach)-induced endothelial-dependent relaxation (EDR) and sodium nitroprusside (SNP)-induced endothelial-independent relaxation (EIR) were examined. Our results showed that the myogenic responses were significantly impaired in both the CA and MCA of SIRT3ECKO mice. Furthermore, female mice had worsened myogenic response in MCA. In CA, EDR was abolished in both male and female SIRT3ECKO mice. Intriguingly, EIR was only reduced in the female mice. In MCA, EDR was reduced in male SIRT3ECKO mice, whereas EIR was decreased in both male and female mice. Female SIRT3ECKO mice had profound dysfunction in CA, whereas male mice exhibited more dysfunction in MCA. These data revealed a sex and organ-specific role of endothelial SIRT3 in vascular function and myogenic responses. Our study suggests that endothelial SIRT3 is necessary for maintaining vascular function and blood flow autoregulation.
Collapse
Affiliation(s)
- Jian‐Xiong Chen
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Jin Zhang
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Yingjie Chen
- Department of Physiology and Biophysics, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Heng Zeng
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
4
|
Rao G, Peng B, Zhang G, Fu X, Tian J, Tian Y. MicroRNAs in diabetic macroangiopathy. Cardiovasc Diabetol 2024; 23:344. [PMID: 39285459 PMCID: PMC11406791 DOI: 10.1186/s12933-024-02405-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Diabetic macroangiopathy is a leading cause of diabetes-related mortality worldwide. Both genetic and environmental factors, through a multitude of underlying molecular mechanisms, contribute to the pathogenesis of diabetic macroangiopathy. MicroRNAs (miRNAs), a class of non-coding RNAs known for their functional diversity and expression specificity, are increasingly recognized for their roles in the initiation and progression of diabetes and diabetic macroangiopathy. In this review, we will describe the biogenesis of miRNAs, and summarize their functions in diabetic macroangiopathy, including atherosclerosis, peripheral artery disease, coronary artery disease, and cerebrovascular disease, which are anticipated to provide new insights into future perspectives of miRNAs in basic, translational and clinical research, ultimately advancing the diagnosis, prevention, and treatment of diabetic macroangiopathy.
Collapse
Affiliation(s)
- Guocheng Rao
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China
| | - Boqiang Peng
- Department of General Surgery and Gastric Cancer Center and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Guixiang Zhang
- Department of General Surgery and Gastric Cancer Center and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xianghui Fu
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China.
| | - Jingyan Tian
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Tian
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
5
|
McCallinhart PE, Chade AR, Bender SB, Trask AJ. Expanding landscape of coronary microvascular disease in co-morbid conditions: Metabolic disease and beyond. J Mol Cell Cardiol 2024; 192:26-35. [PMID: 38734061 PMCID: PMC11340124 DOI: 10.1016/j.yjmcc.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Coronary microvascular disease (CMD) and impaired coronary blood flow control are defects that occur early in the pathogenesis of heart failure in cardiometabolic conditions, prior to the onset of atherosclerosis. In fact, recent studies have shown that CMD is an independent predictor of cardiac morbidity and mortality in patients with obesity and metabolic disease. CMD is comprised of functional, structural, and mechanical impairments that synergize and ultimately reduce coronary blood flow in metabolic disease and in other co-morbid conditions, including transplant, autoimmune disorders, chemotherapy-induced cardiotoxicity, and remote injury-induced CMD. This review summarizes the contemporary state-of-the-field related to CMD in metabolic and these other co-morbid conditions based on mechanistic data derived mostly from preclinical small- and large-animal models in light of available clinical evidence and given the limitations of studying these mechanisms in humans. In addition, we also discuss gaps in current understanding, emerging areas of interest, and opportunities for future investigations in this field.
Collapse
Affiliation(s)
- Patricia E McCallinhart
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America
| | - Alejandro R Chade
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, United States of America; Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States of America
| | - Shawn B Bender
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, United States of America.
| | - Aaron J Trask
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States of America.
| |
Collapse
|
6
|
Shi L, Gao W, Ma T, Xu X, Wang H, Lu Y. Preparation of copper nanoparticles fluorescent probes and detection of hydrogen peroxide and glucose. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 311:123980. [PMID: 38335589 DOI: 10.1016/j.saa.2024.123980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/07/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
Fluorescent copper nanoparticles (CuNPs) was synthesized by one-step chemical reduction method using ascorbic acid (AA) and copper sulfate (CuSO4⋅5H2O) as raw materials, which had good water solubility and fluorescence properties. A green, simple and safe CuNPs@Fe2+ fluorescence probe was developed for the detection of hydrogen peroxide and glucose using Fe2+ as a bridge. The prepared CuNPs could obtain the maximum fluorescence emission wavelength at 440 nm when the excitation wavelength was 360 nm. The average particle size of CuNPs was 10 nm, which had good photobleach resistance, stability and salt tolerance. The fluorescence intensity was quenched due to electron transfer (ET) process when hydrogen peroxide was added to CuNPs@Fe2+ system. This result was mainly because Fenton reaction occured between hydrogen peroxide and Fe2+, producing hydroxyl free radicals (OH) and Fe3+. Since glucose could be catalyzed by specific glucose oxidase (GOX) to produce H2O2 and corresponding oxidation products, the quantitative analysis of glucose was realized when glucose oxidase was introduced into the CuNPs@Fe2+ sensor system. Therefore, a novel CuNPs@Fe2+ fluorescent probe sensor study was constructed to further achieve quantitative detection of H2O2 and glucose. Under the optimized experimental conditions, the linear ranges for H2O2 and glucose were 28.219-171.562 μM and 1.237-75.771 μM, respectively. And the detection limits for H2O2 and glucose were 7.169 μM and 0.540 μM, respectively. In addition, the mechanism of fluorescence probe quenching caused by the interaction between H2O2 and CuNPs@Fe2+ was also discussed. The proposed sensing system had been applied successfully to the detection of glucose in human serum samples.
Collapse
Affiliation(s)
- Lin Shi
- Phytochemistry Key Laboratory of Tibetan Plateau of Qinghai Province, China; Modern Tibetan Medicine Creation Engineering Technology Research Center of Qinghai Province, China; College of Pharmacy, Qinghai Minzu University, China
| | - Wuyang Gao
- Phytochemistry Key Laboratory of Tibetan Plateau of Qinghai Province, China; Modern Tibetan Medicine Creation Engineering Technology Research Center of Qinghai Province, China; College of Pharmacy, Qinghai Minzu University, China
| | - Tianfeng Ma
- Phytochemistry Key Laboratory of Tibetan Plateau of Qinghai Province, China; Modern Tibetan Medicine Creation Engineering Technology Research Center of Qinghai Province, China; College of Pharmacy, Qinghai Minzu University, China
| | - Xiaohua Xu
- Phytochemistry Key Laboratory of Tibetan Plateau of Qinghai Province, China; Modern Tibetan Medicine Creation Engineering Technology Research Center of Qinghai Province, China; College of Pharmacy, Qinghai Minzu University, China
| | - Huan Wang
- Phytochemistry Key Laboratory of Tibetan Plateau of Qinghai Province, China; Modern Tibetan Medicine Creation Engineering Technology Research Center of Qinghai Province, China; College of Pharmacy, Qinghai Minzu University, China.
| | - Yongchang Lu
- Phytochemistry Key Laboratory of Tibetan Plateau of Qinghai Province, China; Modern Tibetan Medicine Creation Engineering Technology Research Center of Qinghai Province, China; College of Pharmacy, Qinghai Minzu University, China.
| |
Collapse
|
7
|
Yao W, Tao R, Wang K, Ding X. Icariin attenuates vascular endothelial dysfunction by inhibiting inflammation through GPER/Sirt1/HMGB1 signaling pathway in type 1 diabetic rats. Chin J Nat Med 2024; 22:293-306. [PMID: 38658093 DOI: 10.1016/s1875-5364(24)60618-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Icariin, a flavonoid glycoside, is extracted from Epimedium. This study aimed to investigate the vascular protective effects of icariin in type 1 diabetic rats by inhibiting high-mobility group box 1 (HMGB1)-related inflammation and exploring its potential mechanisms. The impact of icariin on vascular dysfunction was assessed in streptozotocin (STZ)-induced diabetic rats through vascular reactivity studies. Western blotting and immunofluorescence assays were performed to measure the expressions of target proteins. The release of HMGB1 and pro-inflammation cytokines were measured by enzyme-linked immunosorbent assay (ELISA). The results revealed that icariin administration enhanced acetylcholine-induced vasodilation in the aortas of diabetic rats. It also notably reduced the release of pro-inflammatory cytokines, including interleukin-8 (IL-8), IL-6, IL-1β, and tumor necrosis factor-alpha (TNF-α) in diabetic rats and high glucose (HG)-induced human umbilical vein endothelial cells (HUVECs). The results also unveiled that the pro-inflammatory cytokines in the culture medium of HUVECs could be increased by rHMGB1. The increased release of HMGB1 and upregulated expressions of HMGB1-related inflammatory factors, including advanced glycation end products (RAGE), Toll-like receptor 4 (TLR4), and phosphorylated p65 (p-p65) in diabetic rats and HG-induced HUVECs, were remarkably suppressed by icariin. Notably, HMGB1 translocation from the nucleus to the cytoplasm in HUVECs under HG was inhibited by icariin. Meanwhile, icariin could activate G protein-coupled estrogen receptor (GPER) and sirt1. To explore the role of GPER and Sirt1 in the inhibitory effect of icariin on HMGB1 release and HMGB-induced inflammation, GPER inhibitor and Sirt1 inhibitor were used in this study. These inhibitors diminished the effects of icariin on HMGB1 release and HMGB1-induced inflammation. Specifically, the GPER inhibitor also negated the activation of Sirt1 by icariin. These findings suggest that icariin activates GPER and increases the expression of Sirt1, which in turn reduces HMGB1 translocation and release, thereby improving vascular endothelial function in type 1 diabetic rats by inhibiting inflammation.
Collapse
Affiliation(s)
- Wenhui Yao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Rongpin Tao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Kai Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xuansheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
8
|
Lav Madsen P, Sejersen C, Nyberg M, Sørensen MH, Hellsten Y, Gaede P, Bojer AS. The cardiovascular changes underlying a low cardiac output with exercise in patients with type 2 diabetes mellitus. Front Physiol 2024; 15:1294369. [PMID: 38571722 PMCID: PMC10987967 DOI: 10.3389/fphys.2024.1294369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/19/2024] [Indexed: 04/05/2024] Open
Abstract
The significant morbidity and premature mortality of type 2 diabetes mellitus (T2DM) is largely associated with its cardiovascular consequences. Focus has long been on the arterial atheromatosis of DM giving rise to early stroke and myocardial infarctions, whereas less attention has been given to its non-ischemic cardiovascular consequences. Irrespective of ischemic changes, T2DM is associated with heart failure (HF) most commonly with preserved ejection fraction (HFpEF). Largely due to increasing population ages, hypertension, obesity and T2DM, HFpEF is becoming the most prevalent form of heart failure. Unfortunately, randomized controlled trials of HFpEF have largely been futile, and it now seems logical to address the important different phenotypes of HFpEF to understand their underlying pathophysiology. In the early phases, HFpEF is associated with a significantly impaired ability to increase cardiac output with exercise. The lowered cardiac output with exercise results from both cardiac and peripheral causes. T2DM is associated with left ventricular (LV) diastolic dysfunction based on LV hypertrophy with myocardial disperse fibrosis and significantly impaired ability for myocardial blood flow increments with exercise. T2DM is also associated with impaired ability for skeletal muscle vasodilation during exercise, and as is the case in the myocardium, such changes may be related to vascular rarefaction. The present review discusses the underlying phenotypical changes of the heart and peripheral vascular system and their importance for an adequate increase in cardiac output. Since many of the described cardiovascular changes with T2DM must be considered difficult to change if fully developed, it is suggested that patients with T2DM are early evaluated with respect to their cardiovascular compromise.
Collapse
Affiliation(s)
- Per Lav Madsen
- Department Cardiology, Herlev-Gentofte Hospital, Copenhagen University, Copenhagen, Denmark
- Department Clinical Medicine, Copenhagen University, Copenhagen, Denmark
- The August Krogh Section for Human Physiology, Department Nutrition, Exercise and Sports, Copenhagen University, Copenhagen, Denmark
| | - Casper Sejersen
- The August Krogh Section for Human Physiology, Department Nutrition, Exercise and Sports, Copenhagen University, Copenhagen, Denmark
- Department of Anaesthesia, Rigshospitalet, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael Nyberg
- Department Kidney and Vascular Biology, Global Drug Discovery, Novo Nordisk, Copenhagen, Denmark
| | | | - Ylva Hellsten
- The August Krogh Section for Human Physiology, Department Nutrition, Exercise and Sports, Copenhagen University, Copenhagen, Denmark
| | - Peter Gaede
- Department Endocrinology, Slagelse-Næstved Hospital, Copenhagen, Denmark
| | - Annemie Stege Bojer
- Department Cardiology, Herlev-Gentofte Hospital, Copenhagen University, Copenhagen, Denmark
- Department Endocrinology, Slagelse-Næstved Hospital, Copenhagen, Denmark
| |
Collapse
|
9
|
Awata WMC, Alves JV, Costa RM, Bruder-Nascimento A, Singh S, Barbosa GS, Tirapelli CR, Bruder-Nascimento T. Vascular injury associated with ethanol intake is driven by AT1 receptor and mitochondrial dysfunction. Biomed Pharmacother 2023; 169:115845. [PMID: 37951022 DOI: 10.1016/j.biopha.2023.115845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/13/2023] Open
Abstract
BACKGROUND Renin-angiotensin (Ang II)-aldosterone system (RAAS) is crucial for the cardiovascular risk associated with excessive ethanol consumption. Disturbs in mitochondria have been implicated in multiple cardiovascular diseases. However, if mitochondria dysfunction contributes to ethanol-induced vascular dysfunction is still unknown. We investigated whether ethanol leads to vascular dysfunction via RAAS activation, mitochondria dysfunction, and mitochondrial reactive oxygen species (mtROS). METHODS Male C57/BL6J or mt-keima mice (6-8-weeks old) were treated with ethanol (20% vol./vol.) for 12 weeks with or without Losartan (10 mg/kg/day). RESULTS Ethanol induced aortic hypercontractility in an endothelium-dependent manner. PGC1α (a marker of biogenesis), Mfn2, (an essential protein for mitochondria fusion), as well as Pink-1 and Parkin (markers of mitophagy), were reduced in aortas from ethanol-treated mice. Disturb in mitophagy flux was further confirmed in arteries from mt-keima mice. Additionally, ethanol increased mtROS and reduced SOD2 expression. Strikingly, losartan prevented vascular hypercontractility, mitochondrial dysfunction, mtROS, and restored SOD2 expression. Both MnTMPyP (SOD2 mimetic) and CCCP (a mitochondrial uncoupler) reverted ethanol-induced vascular dysfunction. Moreover, L-NAME (NOS inhibitor) and EUK 134 (superoxide dismutase/catalase mimetic) did not affect vascular response in ethanol group, suggesting that ethanol reduces aortic nitric oxide (NO) and H2O2 bioavailability. These responses were prevented by losartan. CONCLUSION AT1 receptor modulates ethanol-induced vascular hypercontractility by promoting mitochondrial dysfunction, mtROS, and reduction of NO and H2O2 bioavailability. Our findings shed a new light in our understanding of ethanol-induced vascular toxicity and open perspectives of new therapeutic approaches for patients with disorder associated with abusive ethanol drinking.
Collapse
Affiliation(s)
- Wanessa M C Awata
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Juliano V Alves
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Rafael M Costa
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Ariane Bruder-Nascimento
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA
| | - Shubhnita Singh
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA
| | - Gabriela S Barbosa
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA; UNIPEX, Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
| | | | - Thiago Bruder-Nascimento
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Pediatrics Research in Obesity and Metabolism (CPROM), University of Pittsburgh, Pittsburgh, PA, USA; Endocrinology Division at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; Vascular Medicine, Institute (VMI), University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Feng W, Guo L, Liu Y, Ren M. Unraveling the role of VLDL in the relationship between type 2 diabetes and coronary atherosclerosis: a Mendelian randomization analysis. Front Cardiovasc Med 2023; 10:1234271. [PMID: 37965087 PMCID: PMC10642525 DOI: 10.3389/fcvm.2023.1234271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/04/2023] [Indexed: 11/16/2023] Open
Abstract
Background The causal link between Type 2 diabetes (T2D) and coronary atherosclerosis has been established through wet lab experiments; however, its analysis with Genome-wide association studies (GWAS) data remains unexplored. This study aims to validate this relationship using Mendelian randomization analysis and explore the potential mediation of VLDL in this mechanism. Methods Employing Mendelian randomization analysis, we investigated the causal connection between T2D and coronary atherosclerosis. We utilized GWAS summary statistics from European ancestry cohorts, comprising 23,363 coronary atherosclerosis patients and 195,429 controls, along with 32,469 T2D patients and 183,185 controls. VLDL levels, linked to SNPs, were considered as a potential mediating causal factor that might contribute to coronary atherosclerosis in the presence of T2D. We employed the inverse variance weighted (IVW), Egger regression (MR-Egger), weighted median, and weighted model methods for causal effect estimation. A leave-one-out sensitivity analysis was conducted to ensure robustness. Results Our Mendelian randomization analysis demonstrated a genetic association between T2D and an increased coronary atherosclerosis risk, with the IVW estimate at 1.13 [95% confidence interval (CI): 1.07-1.20]. Additionally, we observed a suggestive causal link between T2D and VLDL levels, as evidenced by the IVW estimate of 1.02 (95% CI: 0.98-1.07). Further supporting lipid involvement in coronary atherosclerosis pathogenesis, the IVW-Egger estimate was 1.30 (95% CI: 1.06-1.58). Conclusion In conclusion, this study highlights the autonomous contributions of T2D and VLDL levels to coronary atherosclerosis development. T2D is linked to a 13.35% elevated risk of coronary atherosclerosis, and within T2D patients, VLDL concentration rises by 2.49%. Notably, each standard deviation increase in VLDL raises the likelihood of heart disease by 29.6%. This underscores the significant role of lipid regulation, particularly VLDL, as a mediating pathway in coronary atherosclerosis progression.
Collapse
Affiliation(s)
- Wenshuai Feng
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Liuli Guo
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiman Liu
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ming Ren
- Baokang Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
11
|
Liu J, Xiang Y, Chen Y, Zhang H, Ye B, Ren L, Tan W, Kappler A, Hou J. Quantitative Contribution of Oxygen Vacancy Defects to Arsenate Immobilization on Hematite. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:12453-12464. [PMID: 37561149 DOI: 10.1021/acs.est.3c03441] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Hematite is a common iron oxide in natural environments, which has been observed to influence the transport and fate of arsenate by its association with hematite. Although oxygen vacancies were demonstrated to exist in hematite, their contributions to the arsenate immobilization have not been quantified. In this study, hematite samples with tunable oxygen vacancy defect (OVD) concentrations were synthesized by treating defect-free hematite using different NaBH4 solutions. The vacancy defects were characterized by positron annihilation lifetime spectroscopy, Doppler broadening of annihilation radiation, extended X-ray absorption fine structure (EXAFS), thermogravimetric mass spectrometry (TG-MS), electron paramagnetic resonance (EPR), and X-ray photoelectron spectroscopy (XPS). The results revealed that oxygen vacancy was the primary defect type existing on the hematite surface. TG-MS combined with EPR analysis allowed quantification of OVD concentrations in hematite. Batch experiments revealed that OVDs had a positive effect on arsenate adsorption, which could be quantitatively described by a linear relationship between the OVD concentration (Cdef, mmol m-2) and the enhanced arsenate adsorption amount caused by defects (ΔQm, μmol m-2) (ΔQm = 20.94 Cdef, R2 = 0.9813). NH3-diffuse reflectance infrared Fourier transform (NH3-DRIFT) analysis and density functional theory (DFT) calculations demonstrated that OVDs in hematite were beneficial to the improvement in adsorption strength of surface-active sites, thus considerably promoting the immobilization of arsenate.
Collapse
Affiliation(s)
- Juan Liu
- State Environmental Protection Key Laboratory of Soil Health and Green Remediation, Hubei Key Laboratory of Soil Environment and Pollution Remediation, College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
| | - Yongjin Xiang
- State Environmental Protection Key Laboratory of Soil Health and Green Remediation, Hubei Key Laboratory of Soil Environment and Pollution Remediation, College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
| | - Yiwen Chen
- State Key Laboratory of Particle Detection and Electronics, University of Science and Technology of China, Hefei 230026, China
| | - Hongjun Zhang
- State Key Laboratory of Particle Detection and Electronics, University of Science and Technology of China, Hefei 230026, China
| | - Bangjiao Ye
- State Key Laboratory of Particle Detection and Electronics, University of Science and Technology of China, Hefei 230026, China
| | - Lu Ren
- School of Civil Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Wenfeng Tan
- State Environmental Protection Key Laboratory of Soil Health and Green Remediation, Hubei Key Laboratory of Soil Environment and Pollution Remediation, College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
| | - Andreas Kappler
- Geomicrobiology, Department of Geosciences, University of Tuebingen, Tuebingen 72076, Germany
| | - Jingtao Hou
- State Environmental Protection Key Laboratory of Soil Health and Green Remediation, Hubei Key Laboratory of Soil Environment and Pollution Remediation, College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
12
|
Holland A, Enrick M, Diaz A, Yin L. Is miR-21 A Therapeutic Target in Cardiovascular Disease? INTERNATIONAL JOURNAL OF DRUG DISCOVERY AND PHARMACOLOGY 2023; 2:26-36. [PMID: 37799562 PMCID: PMC10552863 DOI: 10.53941/ijddp.0201003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
microRNA-21 (miR-21) serves a multitude of functions at the molecular level through its regulation of messenger RNA. Previous research has sparked interest in the role of miR-21 as a potential therapeutic target in cardiovascular diseases. miR-21 expression contributes to the differentiation, proliferation, and maturation of many cell types, such as fibroblasts, endothelial cells, cardiomyocytes, and endothelial progenitor cells. The function of miR-21 depends upon its expression level in the specific cell types and downstream targets, which determine cell fate. Under pathological conditions, the expression level of miR-21 is altered, leading to abnormal gene regulation of downstream signaling and cardiovascular diseases such as hypertension, cardiac hypertrophy and fibrosis, atherosclerosis, and heart failure. Agomirs or antagomirs can be introduced into the respective tissue type to reverse or stop the progression of the disease. Exosomes in the extracellular vesicles, which mediate many cellular events with high biocompatibility, have a high potential of efficiently delivering miR-21 to their targeted cells. The critical role of miR-21 in cardiovascular disease (CVD) is indisputable, but there are controversial reports on the function of miR-21 in the same disease. This discrepancy sparks interest in better understanding the role of miR-21 in different tissues under different stages of various diseases and the mechanism of how miR-21 inhibitors work.
Collapse
Affiliation(s)
- Antoinette Holland
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Ohio 44272, USA
| | - Molly Enrick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Ohio 44272, USA
| | - Arianna Diaz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Ohio 44272, USA
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Ohio 44272, USA
| |
Collapse
|
13
|
Winnicki A, Gadd J, Ohanyan V, Hernandez G, Wang Y, Enrick M, McKillen H, Kiedrowski M, Kundu D, Kegecik K, Penn M, Chilian WM, Yin L, Dong F. Role of endothelial CXCR4 in the development of aortic valve stenosis. Front Cardiovasc Med 2022; 9:971321. [PMID: 36148060 PMCID: PMC9488705 DOI: 10.3389/fcvm.2022.971321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022] Open
Abstract
Background CXCL12/CXCR4 signaling is essential in cardiac development and repair, however, its contribution to aortic valve stenosis (AVS) remains unclear. In this study, we tested the role of endothelial CXCR4 on the development of AVS. Materials and methods We generated CXCR4 endothelial cell-specific knockout mice (EC CXCR4 KO) by crossing CXCR4fl/fl mice with Tie2-Cre mice to study the role of endothelial cell CXCR4 in AVS. CXCR4fl/fl mice were used as controls. Echocardiography was used to assess the aortic valve and cardiac function. Heart samples containing the aortic valve were stained using Alizarin Red for detection of calcification. Masson’s trichrome staining was used for the detection of fibrosis. The apex of the heart samples was stained with wheat germ agglutinin (WGA) to visualize ventricular hypertrophy. Results Compared with the control group, the deletion of CXCR4 in endothelial cells led to significantly increased aortic valve peak velocity and aortic valve peak pressure gradient, with decreased aortic valve area and ejection fraction. EC CXCR4 KO mice also developed cardiac hypertrophy as evidenced by increased diastolic and systolic left ventricle posterior wall thickness (LVPW), cardiac myocyte size, and heart weight (HW) to body weight (BW) ratio. Our data also confirmed increased microcalcifications, interstitial fibrosis, and thickened valvular leaflets of the EC CXCR4 KO mice. Conclusion The data collected throughout this study suggest the deletion of CXCR4 in endothelial cells is linked to the development of aortic valve stenosis and left ventricular hypertrophy. The statistically significant parameters measured indicate that endothelial cell CXCR4 plays an important role in aortic valve development and function. We have compiled compelling evidence that EC CXCR4 KO mice can be used as a novel model for AVS.
Collapse
Affiliation(s)
- Anna Winnicki
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - James Gadd
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - Gilbert Hernandez
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - Yang Wang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - Molly Enrick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - Hannah McKillen
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - Matthew Kiedrowski
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - Dipan Kundu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - Karlina Kegecik
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - Marc Penn
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
- Summa Cardiovascular Institute, Summa Health, Akron, OH, United States
| | - William M. Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
- Liya Yin,
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown Township, OH, United States
- *Correspondence: Feng Dong,
| |
Collapse
|
14
|
Smith AN, Altara R, Amin G, Habeichi NJ, Thomas DG, Jun S, Kaplan A, Booz GW, Zouein FA. Genomic, Proteomic, and Metabolic Comparisons of Small Animal Models of Heart Failure With Preserved Ejection Fraction: A Tale of Mice, Rats, and Cats. J Am Heart Assoc 2022; 11:e026071. [PMID: 35904190 PMCID: PMC9375492 DOI: 10.1161/jaha.122.026071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) remains a medical anomaly that baffles researchers and physicians alike. The overall phenotypical changes of diastolic function and left ventricular hypertrophy observed in HFpEF are definable; however, the metabolic and molecular alterations that ultimately produce these changes are not well established. Comorbidities such as obesity, hypertension, and diabetes, as well as general aging, play crucial roles in its development and progression. Various animal models have recently been developed to better understand the pathophysiological and metabolic developments in HFpEF and to illuminate novel avenues for pharmacotherapy. These models include multi‐hit rodents and feline aortic constriction animals. Recently, genomic, proteomic, and metabolomic approaches have been used to define altered signaling pathways in the heart associated with HFpEF, including those involved in inflammation, cGMP‐related, Ca2+ handling, mitochondrial respiration, and the unfolded protein response in endoplasmic reticulum stress. This article aims to present an overview of what has been learnt by these studies, focusing mainly on the findings in common while highlighting unresolved issues. The knowledge gained from these research models will not simply be of benefit for treating HFpEF but will undoubtedly provide new insights into the mechanisms by which the heart deals with external stresses and how the processes involved can fail.
Collapse
Affiliation(s)
- Alex N Smith
- Department of Pharmacology and Toxicology, School of Medicine University of Mississippi Medical Center Jackson MS
| | - Raffaele Altara
- Department of Pathology, School of Medicine University of Mississippi Medical Center Jackson MS
| | - Ghadir Amin
- Department of Pharmacology and Toxicology, Faculty of Medicine American University of Beirut Medical Center Beirut Lebanon
| | - Nada J Habeichi
- Department of Pharmacology and Toxicology, Faculty of Medicine American University of Beirut Medical Center Beirut Lebanon.,Laboratory of Signaling and Cardiovascular Pathophysiology, Inserm Unit UMR-S 1180, Faculty of Pharmacy Paris-Saclay University Châtenay-Malabry France
| | - Daniel G Thomas
- Department of Pharmacology and Toxicology, School of Medicine University of Mississippi Medical Center Jackson MS
| | - Seungho Jun
- Division of Cardiology The Johns Hopkins Medical Institutions Baltimore MD
| | - Abdullah Kaplan
- Department of Pharmacology and Toxicology, Faculty of Medicine American University of Beirut Medical Center Beirut Lebanon.,Cardiology Clinic Rumeli Hospital Istanbul Turkey
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine University of Mississippi Medical Center Jackson MS
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, School of Medicine University of Mississippi Medical Center Jackson MS.,Department of Pharmacology and Toxicology, Faculty of Medicine American University of Beirut Medical Center Beirut Lebanon.,Laboratory of Signaling and Cardiovascular Pathophysiology, Inserm Unit UMR-S 1180, Faculty of Pharmacy Paris-Saclay University Châtenay-Malabry France.,The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence American University of Beirut Medical Center Beirut Lebanon
| |
Collapse
|
15
|
Razan MR, Akther F, Islam RA, Graham JL, Stanhope KL, Havel PJ, Rahimian R. 17β-Estradiol Treatment Improves Acetylcholine-Induced Relaxation of Mesenteric Arteries in Ovariectomized UC Davis Type 2 Diabetes Mellitus Rats in Prediabetic State. Front Physiol 2022; 13:900813. [PMID: 35784863 PMCID: PMC9248973 DOI: 10.3389/fphys.2022.900813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
We recently reported sex differences in mesenteric arterial function of the UC Davis type-2 diabetes mellitus (UCD-T2DM) rats as early as the prediabetic state. We reported that mesenteric arteries (MA) from prediabetic male rats exhibited a greater impairment compared to that in prediabetic females. However, when females became diabetic, they exhibited a greater vascular dysfunction than males. Thus, the aim of this study was to investigate whether the female sex hormone, estrogen preserves mesenteric arterial vasorelaxation in UCD-T2DM female rats at an early prediabetic state. Age-matched female Sprague Dawley and prediabetic (PD) UCD-T2DM rats were ovariectomized (OVX) and subcutaneously implanted with either placebo or 17β-estradiol (E2, 1.5 mg) pellets for 45 days. We assessed the contribution of endothelium-derived relaxing factors (EDRF) to acetylcholine (ACh)-induced vasorelaxation, using pharmacological inhibitors. Responses to sodium nitroprusside (SNP) and phenylephrine (PE) were also measured. Additionally, metabolic parameters and expression of some targets associated with vascular and insulin signaling were determined. We demonstrated that the responses to ACh and SNP were severely impaired in the prediabetic state (PD OVX) rats, while E2 treatment restored vasorelaxation in the PD OVX + E2. Moreover, the responses to PE was significantly enhanced in MA of PD OVX groups, regardless of placebo or E2 treatment. Overall, our data suggest that 1) the impairment of ACh responses in PD OVX rats may, in part, result from the elevated contractile responses to PE, loss of contribution of endothelium-dependent hyperpolarization (EDH) to vasorelaxation, and a decreased sensitivity of MA to nitric oxide (NO), and 2) the basis for the protective effects of E2 may be partly attributed to the elevation of the NO contribution to vasorelaxation and its interaction with MA as well as potential improvement of insulin signaling. Here, we provide the first evidence of the role of E2 in protecting MA from early vascular dysfunction in prediabetic female rats.
Collapse
Affiliation(s)
- Md Rahatullah Razan
- Department of Physiology and Pharmacology, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA, United States
| | - Farjana Akther
- Department of Physiology and Pharmacology, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA, United States
| | - Rifat A. Islam
- Department of Physiology and Pharmacology, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA, United States
| | - James L. Graham
- Department of Molecular Biosciences, School of Veterinary Medicine, Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Kimber L. Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Peter J. Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Roshanak Rahimian
- Department of Physiology and Pharmacology, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA, United States
- *Correspondence: Roshanak Rahimian,
| |
Collapse
|
16
|
Tombor LS, Dimmeler S. Why is endothelial resilience key to maintain cardiac health? Basic Res Cardiol 2022; 117:35. [PMID: 35834003 PMCID: PMC9283358 DOI: 10.1007/s00395-022-00941-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/01/2023]
Abstract
Myocardial injury as induced by myocardial infarction results in tissue ischemia, which critically incepts cardiomyocyte death. Endothelial cells play a crucial role in restoring oxygen and nutrient supply to the heart. Latest advances in single-cell multi-omics, together with genetic lineage tracing, reveal a transcriptional and phenotypical adaptation to the injured microenvironment, which includes alterations in metabolic, mesenchymal, hematopoietic and pro-inflammatory signatures. The extent of transition in mesenchymal or hematopoietic cell lineages is still debated, but it is clear that several of the adaptive phenotypical changes are transient and endothelial cells revert back to a naïve cell state after resolution of injury responses. This resilience of endothelial cells to acute stress responses is important for preventing chronic dysfunction. Here, we summarize how endothelial cells adjust to injury and how this dynamic response contributes to repair and regeneration. We will highlight intrinsic and microenvironmental factors that contribute to endothelial cell resilience and may be targetable to maintain a functionally active, healthy microcirculation.
Collapse
Affiliation(s)
- Lukas S. Tombor
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany ,Faculty for Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany ,Faculty for Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|