1
|
Han YL, Yan TT, Li HX, Chen SS, Zhang ZZ, Wang MY, Chen MJ, Chen YL, Yang XX, Wei LL, Duan YJ, Zhang S. Geniposide alleviates heart failure with preserved ejection fraction in mice by regulating cardiac oxidative stress via MMP2/SIRT1/GSK3β pathway. Acta Pharmacol Sin 2024; 45:2567-2578. [PMID: 39060523 PMCID: PMC11579491 DOI: 10.1038/s41401-024-01341-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex clinical syndrome with cardiac dysfunction, fluid retention and reduced exercise tolerance as the main manifestations. Current treatment of HFpEF is using combined medications of related comorbidities, there is an urgent need for a modest drug to treat HFpEF. Geniposide (GE), an iridoid glycoside extracted from Gardenia Jasminoides, has shown significant efficacy in the treatment of cardiovascular, digestive and central nervous system disorders. In this study we investigated the therapeutic effects of GE on HFpEF experimental models in vivo and in vitro. HFpEF was induced in mice by feeding with HFD and L-NAME (0.5 g/L) in drinking water for 8 weeks, meanwhile the mice were treated with GE (25, 50 mg/kg) every other day. Cardiac echocardiography and exhaustive exercise were performed, blood pressure was measured at the end of treatment, and heart tissue specimens were collected after the mice were euthanized. We showed that GE administration significantly ameliorated cardiac oxidative stress, inflammation, apoptosis, fibrosis and metabolic disturbances in the hearts of HFpEF mice. We demonstrated that GE promoted the transcriptional activation of Nrf2 by targeting MMP2 to affect upstream SIRT1 and downstream GSK3β, which in turn alleviated the oxidative stress in the hearts of HFpEF mice. In H9c2 cells and HL-1 cells, we showed that treatment with GE (1 μM) significantly alleviated H2O2-induced oxidative stress through the MMP2/SIRT1/GSK3β pathway. In summary, GE regulates cardiac oxidative stress via MMP2/SIRT1/GSK3β pathway and reduces cardiac inflammation, apoptosis, fibrosis and metabolic disorders as well as cardiac dysfunction in HFpEF. GE exerts anti-oxidative stress properties by binding to MMP2, inhibiting ROS generation in HFpEF through the SIRT1/Nrf2 signaling pathway. In addition, GE can also affect the inhibition of the downstream MMP2 target GSK3β, thereby suppressing the inflammatory and apoptotic responses in HFpEF. Taken together, GE alleviates oxidative stress/apoptosis/fibrosis and metabolic disorders as well as HFpEF through the MMP2/SIRT1/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Yan-Lu Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Teng-Teng Yan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Hua-Xin Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Sha-Sha Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Zhen-Zhen Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Meng-Yao Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Mei-Jie Chen
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, China
| | - Yuan-Li Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Xiao-Xiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Ling-Ling Wei
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Ya-Jun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China.
| |
Collapse
|
2
|
Koay YC, Liu RP, McIntosh B, Vigder N, Lauren S, Bai AY, Tomita S, Li D, Harney D, Hunter B, Zhang Y, Yang J, Bannon P, Philp A, Philp A, Kaye DM, Larance M, Lal S, O’Sullivan JF. The Efficacy of Risk Factor Modification Compared to NAD + Repletion in Diastolic Heart Failure. JACC Basic Transl Sci 2024; 9:733-750. [PMID: 39070276 PMCID: PMC11282886 DOI: 10.1016/j.jacbts.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 07/30/2024]
Abstract
Heart failure (HF) with left ventricular diastolic dysfunction is a growing global concern. This study evaluated myocardial oxidized nicotinamide adenine dinucleotide (NAD+) levels in human systolic and diastolic HF and in a murine model of HF with preserved ejection fraction, exploring NAD+ repletion as therapy. We quantified myocardial NAD+ and nicotinamide phosphoribosyltransferase levels, assessing restoration with nicotinamide riboside (NR). Findings show significant NAD+ and nicotinamide phosphoribosyltransferase depletion in human diastolic HF myocardium, but NR successfully restored NAD+ levels. In murine HF with preserved ejection fraction, NR as preventive and therapeutic intervention improved metabolic and antioxidant profiles. This study underscores NAD+ repletion's potential in diastolic HF management.
Collapse
Affiliation(s)
- Yen Chin Koay
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Ren Ping Liu
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Bailey McIntosh
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Niv Vigder
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Serlin Lauren
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Angela Yu Bai
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Saki Tomita
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Desmond Li
- BCAL Diagnostics, National Innovation Centre, Eveleigh, New South Wales, Australia
| | - Dylan Harney
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Hunter
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Precision Cardiovascular Laboratory, The University of Sydney, New South Wales, Australia
| | - Yunwei Zhang
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Jean Yang
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul Bannon
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiothoracic Surgery, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Ashleigh Philp
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, New South Wales, Australia
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | - Andrew Philp
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Centre for Healthy Aging, Centenary Institute, Sydney, New South Wales, Australia
- School of Sport, Exercise and Rehabilitation Sciences, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - David M. Kaye
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
- Faculty of Medicine, Monash University, Melbourne, Australia
| | - Mark Larance
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Sean Lal
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Precision Cardiovascular Laboratory, The University of Sydney, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - John F. O’Sullivan
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
3
|
Dye C, Dela Cruz M, Larsen T, Nair G, Marinescu K, Suboc T, Engelstein E, Marsidi J, Patel P, Sharma P, Volgman AS. A review of the impact, pathophysiology, and management of atrial fibrillation in patients with heart failure with preserved ejection fraction. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2023; 33:100309. [PMID: 38510554 PMCID: PMC10946048 DOI: 10.1016/j.ahjo.2023.100309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 03/22/2024]
Abstract
Patients with heart failure with preserved ejection fraction (HFpEF) and atrial fibrillation (AF) have increased mortality and increased risk of stroke. Due to the heterogeneous nature of both disease processes, it is difficult to ascertain whether the diagnosis and progression of AF is the cause of deterioration or if it is a symptom of worsening heart failure. This presents physicians with a clinical conundrum of whether optimizing their heart failure will decrease the overall AF burden or if restoration of sinus rhythm is necessary to optimize patients with HFpEF. In this paper, we will review the impact of AF in patients with HFpEF, the pathophysiology and heterogeneity of HFpEF and AF, and the management of these patients. As HFpEF and AF become more prevalent, managing these disease processes needs standardization to improve outcomes. Further research is needed to understand the complex interplay between AF and HFpEF to help determine the best management strategy.
Collapse
Affiliation(s)
- Cicely Dye
- Division of Cardiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Mark Dela Cruz
- Advocate Heart Institute, Advocate Christ Medical Center, Chicago, IL 60453, USA
| | - Timothy Larsen
- Division of Cardiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Gatha Nair
- Division of Cardiology, University of Washington, Seattle, WA 98105, USA
| | - Karolina Marinescu
- Division of Cardiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Tisha Suboc
- Division of Cardiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Erica Engelstein
- Division of Cardiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jennifer Marsidi
- Division of Cardiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Priya Patel
- Division of Cardiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Parikshit Sharma
- Division of Cardiology, Rush University Medical Center, Chicago, IL 60612, USA
| | | |
Collapse
|
4
|
Masenga SK, Povia JP, Lwiindi PC, Kirabo A. Recent Advances in Microbiota-Associated Metabolites in Heart Failure. Biomedicines 2023; 11:2313. [PMID: 37626809 PMCID: PMC10452327 DOI: 10.3390/biomedicines11082313] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Heart failure is a risk factor for adverse events such as sudden cardiac arrest, liver and kidney failure and death. The gut microbiota and its metabolites are directly linked to the pathogenesis of heart failure. As emerging studies have increased in the literature on the role of specific gut microbiota metabolites in heart failure development, this review highlights and summarizes the current evidence and underlying mechanisms associated with the pathogenesis of heart failure. We found that gut microbiota-derived metabolites such as short chain fatty acids, bile acids, branched-chain amino acids, tryptophan and indole derivatives as well as trimethylamine-derived metabolite, trimethylamine N-oxide, play critical roles in promoting heart failure through various mechanisms. Mainly, they modulate complex signaling pathways such as nuclear factor kappa-light-chain-enhancer of activated B cells, Bcl-2 interacting protein 3, NLR Family Pyrin Domain Containing inflammasome, and Protein kinase RNA-like endoplasmic reticulum kinase. We have also highlighted the beneficial role of other gut metabolites in heart failure and other cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | - Joreen P. Povia
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
| | - Propheria C. Lwiindi
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| |
Collapse
|
5
|
Wójcicka G, Pradiuch A, Fornal E, Stachniuk A, Korolczuk A, Marzec-Kotarska B, Nikolaichuk H, Czechowska G, Kozub A, Trzpil A, Góralczyk A, Bełtowski J. The effect of exenatide (a GLP-1 analogue) and sitagliptin (a DPP-4 inhibitor) on asymmetric dimethylarginine (ADMA) metabolism and selected biomarkers of cardiac fibrosis in rats with fructose-induced metabolic syndrome. Biochem Pharmacol 2023; 214:115637. [PMID: 37290595 DOI: 10.1016/j.bcp.2023.115637] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023]
Abstract
Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide (NO) synthesis, is a risk factor for endothelial dysfunction, a common pathophysiological denominator for both atherogenesis and cardiac fibrosis. We aimed to investigate whether the cardioprotective and antifibrotic effects of incretin drugs, exenatide and sitagliptin, may be associated with their ability to affect circulating and cardiac ADMA metabolism. Normal and fructose-fed rats were treated with sitagliptin (5.0/10 mg/kg) or exenatide (5/10 µg/kg) for 4 weeks. The following methods were used: LC-MS/MS, ELISA, Real-Time-PCR, colorimetry, IHC and H&E staining, PCA and OPLS-DA projections. Eight-week fructose feeding resulted in an increase in plasma ADMA and a decrease in NO concentration. Exenatide administration into fructose-fed rats reduced the plasma ADMA level and increased NO level. In the heart of these animals exenatide administration increased NO and PRMT1 level, reduced TGF-ß1, α-SMA levels and COL1A1 expression. In the exenatide treated rats renal DDAH activity positively correlated with plasma NO level and negatively with plasma ADMA level and cardiac α-SMA concentration. Sitagliptin treatment of fructose-fed rats increased plasma NO concentration, reduced circulating SDMA level, increased renal DDAH activity and reduced myocardial DDAH activity. Both drugs attenuated the myocardial immunoexpression of Smad2/3/P and perivascular fibrosis. In the metabolic syndrome condition both sitagliptin and exenatide positively modulated cardiac fibrotic remodeling and circulating level of endogenous NOS inhibitors but had no effects on ADMA levels in the myocardium.
Collapse
Affiliation(s)
- G Wójcicka
- Department of Pathophysiology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - A Pradiuch
- Department of Pathophysiology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - E Fornal
- Department of Bioanalytic, Medical University of Lublin ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - A Stachniuk
- Department of Bioanalytic, Medical University of Lublin ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - A Korolczuk
- Department of Clinical Pathology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - B Marzec-Kotarska
- Department of Clinical Pathology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - H Nikolaichuk
- Department of Bioanalytic, Medical University of Lublin ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - G Czechowska
- Department of Pharmacology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - A Kozub
- Department of Bioanalytic, Medical University of Lublin ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - A Trzpil
- Department of Bioanalytic, Medical University of Lublin ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - A Góralczyk
- Department of Pathophysiology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - J Bełtowski
- Department of Pathophysiology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| |
Collapse
|
6
|
Yu W, Jiang Y, Xu H, Zhou Y. The Interaction of Gut Microbiota and Heart Failure with Preserved Ejection Fraction: From Mechanism to Potential Therapies. Biomedicines 2023; 11:biomedicines11020442. [PMID: 36830978 PMCID: PMC9953339 DOI: 10.3390/biomedicines11020442] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a disease for which there is no definite and effective treatment, and the number of patients is more than 50% of heart failure (HF) patients. Gut microbiota (GMB) is a general term for a group of microbiota living in humans' intestinal tracts, which has been proved to be related to cardiovascular diseases, including HFpEF. In HFpEF patients, the composition of GMB is significantly changed, and there has been a tendency toward dysbacteriosis. Metabolites of GMB, such as trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs) and bile acids (BAs) mediate various pathophysiological mechanisms of HFpEF. GMB is a crucial influential factor in inflammation, which is considered to be one of the main causes of HFpEF. The role of GMB in its important comorbidity-metabolic syndrome-also mediates HFpEF. Moreover, HF would aggravate intestinal barrier impairment and microbial translocation, further promoting the disease progression. In view of these mechanisms, drugs targeting GMB may be one of the effective ways to treat HFpEF. This review focuses on the interaction of GMB and HFpEF and analyzes potential therapies.
Collapse
Affiliation(s)
- Wei Yu
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Yufeng Jiang
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Hui Xu
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Yafeng Zhou
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
- Correspondence: ; Tel./Fax: 86-512-65955057
| |
Collapse
|
7
|
Gui XY, Rabkin SW. C-Reactive Protein, Interleukin-6, Trimethylamine-N-Oxide, Syndecan-1, Nitric Oxide, and Tumor Necrosis Factor Receptor-1 in Heart Failure with Preserved Versus Reduced Ejection Fraction: a Meta-Analysis. Curr Heart Fail Rep 2023; 20:1-11. [PMID: 36479675 DOI: 10.1007/s11897-022-00584-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review was to synthesize the evidence on non-traditional biomarkers from proteomic and metabolomic studies that may distinguish heart failure (HF) with preserved ejection fraction (HFpEF) from heart failure with reduced ejection fraction (HFrEF) and non-HF. RECENT FINDINGS Understanding the pathophysiology of HFpEF continues to be challenging. A number of inflammatory and metabolic biomarkers that have recently been suggested to be involved include C-reactive protein (CRP), interleukin-6 (IL-6), trimethylamine-N-oxide (TMAO), syndecan-1 (SDC-1), nitric oxide (NO), and tumor necrosis factor receptor-1 (TNFR-1). A systematic search was conducted using Medline, EMBASE, and Web of Science with search terms such as "HFpEF," "metabolomics," and "proteomics," and a meta-analysis was conducted. The results demonstrate significantly higher levels of TMAO, CRP, SDC-1, and IL-6 in HFpEF compared to controls without HF and significantly higher levels of TMAO and CRP in HFrEF compared to controls. The results further suggest that HFpEF might be distinguishable from HFrEF based on higher levels of IL-6 and lower levels of SDC-1 and NO. These data may reflect pathophysiological differences between HFpEF and HFrEF.
Collapse
Affiliation(s)
- Xi Yao Gui
- Department of Medicine, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Simon W Rabkin
- Department of Medicine, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.
- Division of Cardiology, University of British Columbia, 9Th Floor 2775 Laurel St, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
8
|
Teuber JP, Essandoh K, Hummel SL, Madamanchi NR, Brody MJ. NADPH Oxidases in Diastolic Dysfunction and Heart Failure with Preserved Ejection Fraction. Antioxidants (Basel) 2022; 11:1822. [PMID: 36139898 PMCID: PMC9495396 DOI: 10.3390/antiox11091822] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases regulate production of reactive oxygen species (ROS) that cause oxidative damage to cellular components but also regulate redox signaling in many cell types with essential functions in the cardiovascular system. Research over the past couple of decades has uncovered mechanisms by which NADPH oxidase (NOX) enzymes regulate oxidative stress and compartmentalize intracellular signaling in endothelial cells, smooth muscle cells, macrophages, cardiomyocytes, fibroblasts, and other cell types. NOX2 and NOX4, for example, regulate distinct redox signaling mechanisms in cardiac myocytes pertinent to the onset and progression of cardiac hypertrophy and heart failure. Heart failure with preserved ejection fraction (HFpEF), which accounts for at least half of all heart failure cases and has few effective treatments to date, is classically associated with ventricular diastolic dysfunction, i.e., defects in ventricular relaxation and/or filling. However, HFpEF afflicts multiple organ systems and is associated with systemic pathologies including inflammation, oxidative stress, arterial stiffening, cardiac fibrosis, and renal, adipose tissue, and skeletal muscle dysfunction. Basic science studies and clinical data suggest a role for systemic and myocardial oxidative stress in HFpEF, and evidence from animal models demonstrates the critical functions of NOX enzymes in diastolic function and several HFpEF-associated comorbidities. Here, we discuss the roles of NOX enzymes in cardiovascular cells that are pertinent to the development and progression of diastolic dysfunction and HFpEF and outline potential clinical implications.
Collapse
Affiliation(s)
- James P. Teuber
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kobina Essandoh
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Scott L. Hummel
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Ann Arbor Veterans Affairs Health System, Ann Arbor, MI 48105, USA
| | | | - Matthew J. Brody
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
9
|
Nashawi M, Ahmed MS, Amin T, Abualfoul M, Chilton R. Cardiovascular benefits from SGLT2 inhibition in type 2 diabetes mellitus patients is not impaired with phosphate flux related to pharmacotherapy. World J Cardiol 2021; 13:676-694. [PMID: 35070111 PMCID: PMC8716977 DOI: 10.4330/wjc.v13.i12.676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/02/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
The beneficial cardiorenal outcomes of sodium-glucose cotransporter 2 inhibitors (SGLT2i) in patients with type 2 diabetes mellitus (T2DM) have been substantiated by multiple clinical trials, resulting in increased interest in the multifarious pathways by which their mechanisms act. The principal effect of SGLT2i (-flozin drugs) can be appreciated in their ability to block the SGLT2 protein within the kidneys, inhibiting glucose reabsorption, and causing an associated osmotic diuresis. This ameliorates plasma glucose elevations and the negative cardiorenal sequelae associated with the latter. These include aberrant mitochondrial metabolism and oxidative stress burden, endothelial cell dysfunction, pernicious neurohormonal activation, and the development of inimical hemodynamics. Positive outcomes within these domains have been validated with SGLT2i administration. However, by modulating the sodium-glucose cotransporter in the proximal tubule (PT), SGLT2i consequently promotes sodium-phosphate cotransporter activity with phosphate retention. Phosphatemia, even at physiologic levels, poses a risk in cardiovascular disease burden, more so in patients with type 2 diabetes mellitus (T2DM). There also exists an association between phosphatemia and renal impairment, the latter hampering cardiovascular function through an array of physiologic roles, such as fluid regulation, hormonal tone, and neuromodulation. Moreover, increased phosphate flux is associated with an associated increase in fibroblast growth factor 23 levels, also detrimental to homeostatic cardiometabolic function. A contemporary commentary concerning this notion unifying cardiovascular outcome trial data with the translational biology of phosphate is scant within the literature. Given the apparent beneficial outcomes associated with SGLT2i administration notwithstanding negative effects of phosphatemia, we discuss in this review the effects of phosphate on the cardiometabolic status in patients with T2DM and cardiorenal disease, as well as the mechanisms by which SGLT2i counteract or overcome them to achieve their net effects. Content drawn to develop this conversation begins with proceedings in the basic sciences and works towards clinical trial data.
Collapse
Affiliation(s)
- Mouhamed Nashawi
- Department of Internal Medicine, Baylor Scott and White All Saints Medical Center, Fort Worth, TX 76132, United States.
| | - Mahmoud S Ahmed
- Division of Medicine-Cardiology, UT Health San Antonio, San Antonio, TX 78229, United States
| | - Toka Amin
- Division of Medicine-Cardiology, UT Health San Antonio, San Antonio, TX 78229, United States
| | - Mujahed Abualfoul
- Department of Internal Medicine, Faculty of Medicine, Cairo University, Dallas, TX 75203, United States
| | - Robert Chilton
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, TX 75203, United States
| |
Collapse
|
10
|
Pleiotropic Properties of Valsartan: Do They Result from the Antiglycooxidant Activity? Literature Review and In Vitro Study. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5575545. [PMID: 33763167 PMCID: PMC7946482 DOI: 10.1155/2021/5575545] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/13/2021] [Accepted: 02/16/2021] [Indexed: 12/15/2022]
Abstract
Valsartan belongs to angiotensin II type 1 (AT1) receptor blockers (ARB) used in cardiovascular diseases like heart failure and hypertension. Except for its AT1-antagonism, another mechanism of drug action has been suggested in recent research. One of the supposed actions refers to the positive impact on redox balance and reducing protein glycation. Our study is aimed at assessing the antiglycooxidant properties of valsartan in an in vitro model of oxidized bovine serum albumin (BSA). Glucose, fructose, ribose, glyoxal (GO), methylglyoxal (MGO), and chloramine T were used as glycation or oxidation agents. Protein oxidation products (total thiols, protein carbonyls (PC), and advanced oxidation protein products (AOPP)), glycooxidation products (tryptophan, kynurenine, N-formylkynurenine, and dityrosine), glycation products (amyloid-β structure, fructosamine, and advanced glycation end products (AGE)), and albumin antioxidant activity (total antioxidant capacity (TAC), DPPH assay, and ferric reducing antioxidant power (FRAP)) were measured in each sample. In the presence of valsartan, concentrations of protein oxidation and glycation products were significantly lower comparing to control. Moreover, albumin antioxidant activity was significantly higher in those samples. The drug's action was comparable to renowned antiglycation agents and antioxidants, e.g., aminoguanidine, metformin, Trolox, N-acetylcysteine, or alpha-lipoic acid. The conducted experiment proves that valsartan can ameliorate protein glycation and oxidation in vitro in various conditions. Available animal and clinical studies uphold this statement, but further research is needed to confirm it, as reduction of protein oxidation and glycation may prevent cardiovascular disease development.
Collapse
|
11
|
Toth PP, Gauthier D. Heart failure with preserved ejection fraction: strategies for disease management and emerging therapeutic approaches. Postgrad Med 2020; 133:125-139. [PMID: 33283589 DOI: 10.1080/00325481.2020.1842620] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Approximately 50% of patients with heart failure (HF) have a preserved ejection fraction (HFpEF), and the incidence of HFpEF is increasing relative to HF with reduced ejection fraction (HFrEF). Both types of HF are associated with reduced survival and increased risk for hospitalization. However, in contrast to HFrEF, there are no approved treatments specifically indicated for HFpEF, and current therapy is largely focused on management of symptoms and comorbidities. Diagnosis of HFpEF in the outpatient setting also presents unique challenges compared with HFrEF because of factors including a high burden of comorbidities in HFpEF and difficulties in distinguishing HFpEF from normal aging. Primary care providers (PCPs) play a pivotal role in the delivery of holistic, patient-centric care from diagnosis to management and palliative care. As the prevalence of HF continues to rise in an aging population, PCPs will need to play a greater role in HFpEF care. This article will review HFpEF etiology and pathophysiology, diagnostic workup, and management of symptoms and comorbidities, with a focus on the critical role of PCPs throughout the clinical course of HFpEF.
Collapse
Affiliation(s)
- Peter P Toth
- Preventive Cardiology, CGH Medical Center, Rock Falls, IL, USA.,Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Diane Gauthier
- Section of Cardiology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
12
|
Nashawi M, Sheikh O, Battisha A, Ghali A, Chilton R. Neural tone and cardio-renal outcomes in patients with type 2 diabetes mellitus: a review of the literature with a focus on SGLT2 inhibitors. Heart Fail Rev 2020; 26:643-652. [PMID: 33169337 DOI: 10.1007/s10741-020-10046-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 12/18/2022]
Abstract
Recent clinical trials involving the systemic effects of sodium-glucose cotransporter 2 inhibitors (SGLT2i) have revealed beneficial outcomes pertaining to the microvascular sequelae of type 2 diabetes mellitus (T2DM) such as nephropathy, as well as macrovascular effects such as major adverse cardiovascular effects (MACE). Such findings have spurred the elevation of these agents to level A-tiers of recommendation within clinical guidelines addressing the management of complicated T2DM. While the mechanisms of SGLTi (-flozin drugs) are still being elucidated, a paucity of data exists within the literature appraising the role of neuromodulation and associated mechanisms in the aforementioned outcome studies. Given the role of the nervous system in orchestrating the pathologic processes that hamper cardio-renal status, insight into this topic offers an expanded perspective on T2DM. In this review we investigate the mechanisms by which SGLTi improve cardio-renal function in T2DM patients with emphases on neural tone and nervous system physiology.
Collapse
Affiliation(s)
- Mouhamed Nashawi
- Division of Medicine-Cardiology, UT Health San Antonio, 7872, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| | - Omar Sheikh
- Division of Medicine-Cardiology, UT Health San Antonio, 7872, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Ayman Battisha
- Department of Internal Medicine, University of Massachusetts Medical School-Bay State, 759 Chestnut Street, Springfield, MA, 01199, USA
| | - Abdullah Ghali
- Division of Medicine-Cardiology, UT Health San Antonio, 7872, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Robert Chilton
- Division of Medicine-Cardiology, UT Health San Antonio, 7872, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| |
Collapse
|
13
|
Beyond the myocardium? SGLT2 inhibitors target peripheral components of reduced oxygen flux in the diabetic patient with heart failure with preserved ejection fraction. Heart Fail Rev 2020; 27:219-234. [PMID: 32583230 DOI: 10.1007/s10741-020-09996-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent cardiovascular outcome trials have highlighted the propensity of the antidiabetic agents, SGLT2 inhibitors (SGLT2is or -flozin drugs), to exert positive clinical outcomes in patients with cardiovascular disease at risk for major adverse cardiovascular events (MACEs). Of interest in cardiac diabetology is the physiological status of the patient with T2DM and heart failure with preserved ejection fraction (HFpEF), a well-examined association. Underlying this pathologic tandem are the effects that long-standing hyperglycemia has on the ability of the HFpEF heart to adequately deliver oxygen. It is believed that shortcomings in oxygen diffusion or utilization and the resulting hypoxia thereafter may play a role in underlying the clinical sequelae of patients with T2DM and HFpEF, with implications in the long-term decline of extra-cardiac tissue. Oxygen consumption is one of the most critical factors in indexing heart failure disease burden, warranting a probe into the role of SGLT2i on oxygen utility in HFpEF and T2DM. We investigated the role of oxygen flux in the patient with T2DM and HFpEF extending beyond the heart with focuses on cellular metabolism, perivascular fibrosis with endothelial dysfunction, hematologic changes, and renal effects with neurohormonal considerations in the patient with HFpEF and T2DM. Moreover, we give a commentary on potential therapeutic targets of these components with SGLT2i to gain insight into disease burden amelioration in patients with HFpEF and T2DM.
Collapse
|
14
|
Membrane cholesterol oxidation downregulates atrial β-adrenergic responses in ROS-dependent manner. Cell Signal 2020; 67:109503. [DOI: 10.1016/j.cellsig.2019.109503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/15/2019] [Accepted: 12/15/2019] [Indexed: 01/06/2023]
|
15
|
Henning RJ. Diagnosis and treatment of heart failure with preserved left ventricular ejection fraction. World J Cardiol 2020; 12:7-25. [PMID: 31984124 PMCID: PMC6952725 DOI: 10.4330/wjc.v12.i1.7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/17/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023] Open
Abstract
Nearly six million people in United States have heart failure. Fifty percent of these people have normal left ventricular (LV) systolic heart function but abnormal diastolic function due to increased LV myocardial stiffness. Most commonly, these patients are elderly women with hypertension, ischemic heart disease, atrial fibrillation, obesity, diabetes mellitus, renal disease, or obstructive lung disease. The annual mortality rate of these patients is 8%-12% per year. The diagnosis is based on the history, physical examination, laboratory data, echocardiography, and, when necessary, by cardiac catheterization. Patients with obesity, hypertension, atrial fibrillation, and volume overload require weight reduction, an exercise program, aggressive control of blood pressure and heart rate, and diuretics. Miniature devices inserted into patients for pulmonary artery pressure monitoring provide early warning of increased pulmonary pressure and congestion. If significant coronary heart disease is present, coronary revascularization should be considered.
Collapse
Affiliation(s)
- Robert J Henning
- College of Public Health, University of South Florida, Tampa, FL33612, United States
| |
Collapse
|
16
|
Correale M, Paolillo S, Mercurio V, Limongelli G, Barillà F, Ruocco G, Palazzuoli A, Scrutinio D, Lagioia R, Lombardi C, Lupi L, Magrì D, Masarone D, Pacileo G, Scicchitano P, Matteo Ciccone M, Parati G, Tocchetti CG, Nodari S. Comorbidities in chronic heart failure: An update from Italian Society of Cardiology (SIC) Working Group on Heart Failure. Eur J Intern Med 2020; 71:23-31. [PMID: 31708358 DOI: 10.1016/j.ejim.2019.10.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 07/27/2019] [Accepted: 10/05/2019] [Indexed: 12/25/2022]
Abstract
The increasing number of patients with heart failure HF and comorbidities is due to aging population and increase of life expectancy of patients with cardiovascular disease. Encouraging results derived by recent trials may suggest some comorbidities as new targets for new drugs, highlighting the need for a better understanding of the comorbidities' effects in HF patients and the need of a multidisciplinary approach for the management of chronic HF with comorbidities. We report a brief review about main cardiovascular and non-cardiovascular comorbidities in HF patients in order to update physicians and researchers engaged in the HF research or in "fight against heart failure."
Collapse
Affiliation(s)
| | - Stefania Paolillo
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Giuseppe Limongelli
- Heart Failure Unit, AORN dei Colli, Monaldi Hospital, Naples, Italy; Department of Translational Medical Sciences, Luigi Vanvitelli University, Naples, Italy; Institute of Cardiovascular Sciences, University College of London, London, United Kingdom
| | - Francesco Barillà
- Department of Cardiovascular, Respiratory, Nephrologic, Anesthesiologic and Geriatric Sciences, Sapienza" University of Rome, Italy
| | - Gaetano Ruocco
- Cardiovascular Diseases Unit Department of Internal Medicine, University of Siena, Siena, Italy
| | - Alberto Palazzuoli
- Cardiovascular Diseases Unit Department of Internal Medicine, University of Siena, Siena, Italy
| | | | - Rocco Lagioia
- Cardiology Department, IRCCS "S. Maugeri" Cassano (BA), Bari, Italy
| | - Carolina Lombardi
- Istituto Auxologico Italiano, IRCCS, Sleep Disorders Center & Department of Cardiovascular, Neural and Metabolic Sciences. San Luca Hospital, Milan, Italy
| | - Laura Lupi
- Section of Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Italy
| | - Damiano Magrì
- Department of Clinical and Molecular Medicine, University "La Sapienza", Rome, Italy
| | - Daniele Masarone
- Heart Failure Unit, AORN dei Colli, Monaldi Hospital, Naples, Italy
| | - Giuseppe Pacileo
- Heart Failure Unit, AORN dei Colli, Monaldi Hospital, Naples, Italy
| | - Pietro Scicchitano
- Section of Cardiovascular Diseases, Department of Emergency and Organ Transplantation, University of Bari "A. Moro", Bari, Italy
| | - Marco Matteo Ciccone
- Section of Cardiovascular Diseases, Department of Emergency and Organ Transplantation, University of Bari "A. Moro", Bari, Italy
| | - Gianfranco Parati
- Istituto Auxologico Italiano, IRCCS, Sleep Disorders Center & Department of Cardiovascular, Neural and Metabolic Sciences. San Luca Hospital, Milan, Italy
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Savina Nodari
- Section of Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Italy
| |
Collapse
|
17
|
Abstract
The microcirculation maintains tissue homeostasis through local regulation of blood flow and oxygen delivery. Perturbations in microvascular function are characteristic of several diseases and may be early indicators of pathological changes in the cardiovascular system and in parenchymal tissue function. These changes are often mediated by various reactive oxygen species and linked to disruptions in pathways such as vasodilation or angiogenesis. This overview compiles recent advances relating to redox regulation of the microcirculation by adopting both cellular and functional perspectives. Findings from a variety of vascular beds and models are integrated to describe common effects of different reactive species on microvascular function. Gaps in understanding and areas for further research are outlined. © 2020 American Physiological Society. Compr Physiol 10:229-260, 2020.
Collapse
Affiliation(s)
- Andrew O Kadlec
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - David D Gutterman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Medicine-Division of Cardiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
18
|
Dziubak A, Wójcicka G, Wojtak A, Bełtowski J. Metabolic Effects of Metformin in the Failing Heart. Int J Mol Sci 2018; 19:2869. [PMID: 30248910 PMCID: PMC6213955 DOI: 10.3390/ijms19102869] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/11/2018] [Accepted: 09/17/2018] [Indexed: 01/03/2023] Open
Abstract
Accumulating evidence shows that metformin is an insulin-sensitizing antidiabetic drug widely used in the treatment of type 2 diabetes mellitus (T2DM), which can exert favorable effects on cardiovascular risk and may be safely used in patients with heart failure (HF), and even able to reduce the incidence of HF and to reduce HF mortality. In failing hearts, metformin improves myocardial energy metabolic status through the activation of AMP (adenosine monophosphate)-activated protein kinase (AMPK) and the regulation of lipid and glucose metabolism. By increasing nitric oxide (NO) bioavailability, limiting interstitial fibrosis, reducing the deposition of advanced glycation end-products (AGEs), and inhibiting myocardial cell apoptosis metformin reduces cardiac remodeling and hypertrophy, and thereby preserves left ventricular systolic and diastolic functions. While a lot of preclinical and clinical studies showed the cardiovascular safety of metformin therapy in diabetic patients and HF, to confirm observed benefits, the specific large-scale trials configured for HF development in diabetic patients as a primary endpoints are necessary.
Collapse
Affiliation(s)
- Aleksandra Dziubak
- Department of Pathophysiology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - Grażyna Wójcicka
- Department of Pathophysiology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - Andrzej Wojtak
- Department of Vascular Surgery, Medical University of Lubin, 20-090 Lublin, Poland.
| | - Jerzy Bełtowski
- Department of Pathophysiology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| |
Collapse
|
19
|
Improved heart failure by Rhein lysinate is associated with p38MAPK pathway. Exp Ther Med 2018; 16:2046-2051. [PMID: 30186438 DOI: 10.3892/etm.2018.6423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 03/26/2018] [Indexed: 01/07/2023] Open
Abstract
The present study aimed to explore the role of Rhein lysinate (RHL) in neonatal rat ventricular myocytes (NRVMs) and congestive heart failure induced by co-arctation of the abdominal aorta. Male Sprague-Dawley rats were divided into 3 groups randomly: co-arctation of abdominal aorta group (A group, n=10), sham operation group (SH group, n=10) and RHL treatment rats (A+RHL group, n=10). To establish an in vitro oxidative stressed cardiomyocyte model, NRVMs were treated with 10 µM H2O2 for 24 h. MTT assay indicated that H2O2 treatment reduced primary cardiomyocyte viability in a time- and dose- dependent manner, whereas RHL abolished the detrimental effects of H2O2, indicating a protective role of RHL. Further study demonstrated that H2O2-induced reactive oxygen species (ROS) production was reversed by RHL. Then, TUNEL staining was carried out and the results revealed that H2O2 markedly enhanced primary cardiomyocyte apoptosis. Conversely, RHL incubation decreased H2O2-induced cell apoptosis, indicating the protective role of RHL in primary cardiomyocytes. Furthermore, abnormal p38 activation was identified in the failed heart. Notably, treatment with RHL reduced p38 activation. In addition, RHL significantly enhanced the expression of anti-apoptotic protein, B cell lymphoma (Bcl)-2, however markedly reduced the protein level of Bcl-2 associated X, apoptosis regulator in primary cardiomyocytes, indicating its anti-apoptotic role in the cardiac setting. Overall, RHL protects heart failure primarily by reducing ROS production and cardiomyocyte apoptosis via suppressing p38 mitogen activated protein kinase activation.
Collapse
|
20
|
|
21
|
Krylatov AV, Maslov LN, Voronkov NS, Boshchenko AA, Popov SV, Gomez L, Wang H, Jaggi AS, Downey JM. Reactive Oxygen Species as Intracellular Signaling Molecules in the Cardiovascular System. Curr Cardiol Rev 2018; 14:290-300. [PMID: 29962348 PMCID: PMC6300799 DOI: 10.2174/1573403x14666180702152436] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Redox signaling plays an important role in the lives of cells. This signaling not only becomes apparent in pathologies but is also thought to be involved in maintaining physiological homeostasis. Reactive Oxygen Species (ROS) can activate protein kinases: CaMKII, PKG, PKA, ERK, PI3K, Akt, PKC, PDK, JNK, p38. It is unclear whether it is a direct interaction of ROS with these kinases or whether their activation is a consequence of inhibition of phosphatases. ROS have a biphasic effect on the transport of Ca2+ in the cell: on one hand, they activate the sarcoplasmic reticulum Ca2+-ATPase, which can reduce the level of Ca2+ in the cell, and on the other hand, they can inactivate Ca2+-ATPase of the plasma membrane and open the cation channels TRPM2, which promote Ca2+-loading and subsequent apoptosis. ROS inhibit the enzyme PHD2, which leads to the stabilization of HIF-α and the formation of the active transcription factor HIF. CONCLUSION Activation of STAT3 and STAT5, induced by cytokines or growth factors, may include activation of NADPH oxidase and enhancement of ROS production. Normal physiological production of ROS under the action of cytokines activates the JAK/STAT while excessive ROS production leads to their inhibition. ROS cause the activation of the transcription factor NF-κB. Physiological levels of ROS control cell proliferation and angiogenesis. ROS signaling is also involved in beneficial adaptations to survive ischemia and hypoxia, while further increases in ROS can trigger programmed cell death by the mechanism of apoptosis or autophagy. ROS formation in the myocardium can be reduced by moderate exercise.
Collapse
Affiliation(s)
| | - Leonid N. Maslov
- Address correspondence to this author at the Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of
Science, Tomsk, Russia; Tel: 3822 262174; Fax: 3822 555057;
E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Biolo G, Di Girolamo FG, Fiotti N, Mearelli F, Sarto P. Exercise-mediated reactive oxygen species generation in athletes and in patients with chronic disease. Intern Emerg Med 2017; 12:741-744. [PMID: 28567558 DOI: 10.1007/s11739-017-1689-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/24/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Gianni Biolo
- Department of Medical, Surgical and Health Sciences, Clinica Medica, ASUITs, University of Trieste, Cattinara University Hospital, Strada di Fiume 447, 34149, Trieste, Italy.
| | - Filippo Giorgio Di Girolamo
- Department of Medical, Surgical and Health Sciences, Clinica Medica, ASUITs, University of Trieste, Cattinara University Hospital, Strada di Fiume 447, 34149, Trieste, Italy
| | - Nicola Fiotti
- Department of Medical, Surgical and Health Sciences, Clinica Medica, ASUITs, University of Trieste, Cattinara University Hospital, Strada di Fiume 447, 34149, Trieste, Italy
| | - Filippo Mearelli
- Department of Medical, Surgical and Health Sciences, Clinica Medica, ASUITs, University of Trieste, Cattinara University Hospital, Strada di Fiume 447, 34149, Trieste, Italy
| | | |
Collapse
|
23
|
Norvik JV, Schirmer H, Ytrehus K, Storhaug HM, Jenssen TG, Eriksen BO, Mathiesen EB, Løchen M, Wilsgaard T, Solbu MD. Uric acid predicts mortality and ischaemic stroke in subjects with diastolic dysfunction: the Tromsø Study 1994-2013. ESC Heart Fail 2017; 4:154-161. [PMID: 28451452 PMCID: PMC5396037 DOI: 10.1002/ehf2.12134] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/08/2016] [Indexed: 01/19/2023] Open
Abstract
AIMS To investigate whether serum uric acid predicts adverse outcomes in persons with indices of diastolic dysfunction in a general population. METHODS AND RESULTS We performed a prospective cohort study among 1460 women and 1480 men from 1994 to 2013. Endpoints were all-cause mortality, incident myocardial infarction, and incident ischaemic stroke. We stratified the analyses by echocardiographic markers of diastolic dysfunction, and uric acid was the independent variable of interest. Hazard ratios (HR) were estimated per 59 μmol/L increase in baseline uric acid. Multivariable adjusted Cox proportional hazards models showed that uric acid predicted all-cause mortality in subjects with E/A ratio <0.75 (HR 1.12, 95% confidence interval [CI] 1.00-1.25) or E/A ratio >1.5 (HR 1.51, 95% CI 1.09-2.09, P for interaction between E/A ratio category and uric acid = 0.02). Elevated uric acid increased mortality risk in persons with E-wave deceleration time <140 ms or >220 ms (HR 1.46, 95% CI 1.01-2.12 and HR 1.13, 95% CI 1.02-1.26, respectively; P for interaction = 0.04). Furthermore, in participants with isovolumetric relaxation time ≤60 ms, mortality risk was higher with increasing uric acid (HR 4.98, 95% CI 2.02-12.26, P for interaction = 0.004). Finally, elevated uric acid predicted ischaemic stroke in subjects with severely enlarged left atria (HR 1.62, 95% CI 1.03-2.53, P for interaction = 0.047). CONCLUSIONS Increased uric acid was associated with higher all-cause mortality risk in subjects with echocardiographic indices of diastolic dysfunction, and with higher ischaemic stroke risk in persons with severely enlarged left atria.
Collapse
Affiliation(s)
- Jon V. Norvik
- Metabolic and Renal Research GroupUiT The Arctic University of NorwayN‐9037TromsøNorway
- Cardiovascular Research Group IMBUiT The Arctic University of NorwayN‐9037TromsøNorway
| | - Henrik Schirmer
- Department of CardiologyUniversity Hospital of North NorwayN‐9038TromsøNorway
- Cardiovascular Research Group IKMUiT The Arctic University of NorwayN‐9037TromsøNorway
| | - Kirsti Ytrehus
- Metabolic and Renal Research GroupUiT The Arctic University of NorwayN‐9037TromsøNorway
- Cardiovascular Research Group IMBUiT The Arctic University of NorwayN‐9037TromsøNorway
| | - Hilde M. Storhaug
- Metabolic and Renal Research GroupUiT The Arctic University of NorwayN‐9037TromsøNorway
| | - Trond G. Jenssen
- Metabolic and Renal Research GroupUiT The Arctic University of NorwayN‐9037TromsøNorway
- Department of Transplant MedicineOslo University Hospital RikshospitaletN‐0424OsloNorway
| | - Bjørn O. Eriksen
- Metabolic and Renal Research GroupUiT The Arctic University of NorwayN‐9037TromsøNorway
- Section of NephrologyUniversity Hospital of North NorwayN‐9038TromsøNorway
| | - Ellisiv B. Mathiesen
- Department of NeurologyUniversity Hospital of North NorwayN‐9038TromsøNorway
- Brain and Circulation Research GroupUiT The Arctic University of NorwayN‐9037TromsøNorway
| | - Maja‐Lisa Løchen
- Epidemiology of Chronic Diseases Research GroupUiT The Arctic University of NorwayN‐9037TromsøNorway
| | - Tom Wilsgaard
- Epidemiology of Chronic Diseases Research GroupUiT The Arctic University of NorwayN‐9037TromsøNorway
| | - Marit D. Solbu
- Metabolic and Renal Research GroupUiT The Arctic University of NorwayN‐9037TromsøNorway
- Section of NephrologyUniversity Hospital of North NorwayN‐9038TromsøNorway
| |
Collapse
|
24
|
Wu J, Yu J, Xie P, Maimaitili Y, Wang J, Yang L, Ma H, Zhang X, Yang Y, Zheng H. Sevoflurane postconditioning protects the myocardium against ischemia/reperfusion injury via activation of the JAK2-STAT3 pathway. PeerJ 2017; 5:e3196. [PMID: 28392989 PMCID: PMC5382923 DOI: 10.7717/peerj.3196] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/17/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Sevoflurane postconditioning (S-post) has similar cardioprotective effects as ischemic preconditioning. However, the underlying mechanism of S-post has not been fully elucidated. Janus kinase signaling transduction/transcription activator (JAK2-STAT3) plays an important role in cardioprotection. The purpose of this study was to determine whether the cardioprotective effects of S-post are associated with activation of the JAK2-STAT3 signal pathway. METHODS An adult male Sprague-Dawley (SD) rat model of myocardial ischemia/reperfusion (I/R) injury was established using the Langendorff isolated heart perfusion apparatus. At the beginning of reperfusion, 2.4% sevoflurane alone or in combination with AG490 (a JAK2 selective inhibitor) was used as a postconditioning treatment. The cardiac function indicators, myocardial infarct size, lactic dehydrogenase (LDH) release, mitochondrial ultrastructure, mitochondrial reactive oxygen species (ROS) generation rates, ATP content, protein expression of p-JAK, p-STAT3, Bcl-2 and Bax were measured. RESULTS Compared with the I/R group, S-post significantly increased the expression of p-JAK, p-STAT3 and Bcl-2 and reduced the protein expression of Bax, which markedly decreased the myocardial infarction areas, improved the cardiac function indicators and the mitochondrial ultrastructure, decreased the mitochondrial ROS and increased the ATP content. However, the cardioprotective effects of S-post were abolished by treatment with a JAK2 selective inhibitor (p < 0.05). CONCLUSION This study demonstrates that the cardioprotective effects of S-post are associated with the activation of JAK2-STAT3. The mechanism may be related to an increased expression of p-JAK2 and p-STAT3 after S-post, which reduced mitochondrial ROS generation and increased mitochondrial ATP content, thereby reducing apoptosis and myocardial infarct size.
Collapse
Affiliation(s)
- Jianjiang Wu
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Jin Yu
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Peng Xie
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Yiliyaer Maimaitili
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Jiang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Long Yang
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Haiping Ma
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Xing Zhang
- Department of Aerospace Medicine, Fourth Military Medical University , Xi'an, Shanxi , China
| | - Yining Yang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Hong Zheng
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| |
Collapse
|
25
|
Francisco C, Neves JS, Falcão-Pires I, Leite-Moreira A. Can Adiponectin Help us to Target Diastolic Dysfunction? Cardiovasc Drugs Ther 2017; 30:635-644. [PMID: 27757724 DOI: 10.1007/s10557-016-6694-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adiponectin is the most abundant adipokine and exhibits anti-inflammatory, antiatherogenic and antidiabetic properties. Unlike other adipokines, it inversely correlates with body weight and obesity-linked cardiovascular complications. Diastolic dysfunction is the main mechanism responsible for approximately half of all heart failure cases, the so-called heart failure with preserved ejection fraction (HFpEF), but therapeutic strategies specifically directed towards these patients are still lacking. In the last years, a link between adiponectin and diastolic dysfunction has been suggested. There are several mechanisms through which adiponectin may prevent most of the pathophysiologic mechanisms underlying diastolic dysfunction and HFpEF, including the prevention of myocardial hypertrophy, cardiac fibrosis, nitrative and oxidative stress, atherosclerosis and inflammation, while promoting angiogenesis. Thus, understanding the mechanisms underlying adiponectin-mediated improvement of diastolic function has become an exciting field of research, making adiponectin a promising therapeutic target. In this review, we explore the relevance of adiponectin signaling for the prevention of diastolic dysfunction and identify prospective therapeutic targets aiming at the treatment of this clinical condition.
Collapse
Affiliation(s)
- Catarina Francisco
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Alameda Hernâni Monteiro, 4200-319, Porto, Portugal
| | - João Sérgio Neves
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Alameda Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar São João, Alameda Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Inês Falcão-Pires
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Alameda Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Adelino Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Alameda Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
26
|
Blaustein MP, Chen L, Hamlyn JM, Leenen FHH, Lingrel JB, Wier WG, Zhang J. Pivotal role of α2 Na + pumps and their high affinity ouabain binding site in cardiovascular health and disease. J Physiol 2016; 594:6079-6103. [PMID: 27350568 DOI: 10.1113/jp272419] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/18/2016] [Indexed: 12/13/2022] Open
Abstract
Reduced smooth muscle (SM)-specific α2 Na+ pump expression elevates basal blood pressure (BP) and increases BP sensitivity to angiotensin II (Ang II) and dietary NaCl, whilst SM-α2 overexpression lowers basal BP and decreases Ang II/salt sensitivity. Prolonged ouabain infusion induces hypertension in rodents, and ouabain-resistant mutation of the α2 ouabain binding site (α2R/R mice) confers resistance to several forms of hypertension. Pressure overload-induced heart hypertrophy and failure are attenuated in cardio-specific α2 knockout, cardio-specific α2 overexpression and α2R/R mice. We propose a unifying hypothesis that reconciles these apparently disparate findings: brain mechanisms, activated by Ang II and high NaCl, regulate sympathetic drive and a novel neurohumoral pathway mediated by both brain and circulating endogenous ouabain (EO). Circulating EO modulates ouabain-sensitive α2 Na+ pump activity and Ca2+ transporter expression and, via Na+ /Ca2+ exchange, Ca2+ homeostasis. This regulates sensitivity to sympathetic activity, Ca2+ signalling and arterial and cardiac contraction.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Ling Chen
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Frans H H Leenen
- Hypertension Unit, University of Ottawa Heart Institute, Ottawa, ON, Canada, K1Y 4W7
| | - Jerry B Lingrel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0524, USA
| | - W Gil Wier
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jin Zhang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
27
|
Hiebert JB, Shen Q, Thimmesch A, Pierce J. Impaired Myocardial Bioenergetics in HFpEF and the Role of Antioxidants. Open Cardiovasc Med J 2016; 10:158-62. [PMID: 27583040 PMCID: PMC4974825 DOI: 10.2174/1874192401610010158] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 06/10/2016] [Accepted: 06/10/2016] [Indexed: 12/16/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a significant cardiovascular condition for more than 50% of patients with heart failure. Currently, there is no effective treatment to decrease morbidity and mortality rates associated with HFpEF because of its pathophysiological heterogeneity. Recent evidence shows that deficiency in myocardial bioenergetics is one of the key pathophysiological factors contributing to diastolic dysfunction in HFpEF. Another known mechanism for HFpEF is an overproduction of free radicals, specifically reactive oxygen species. To reduce free radical formation, antioxidants are often used. This article is a summative review of the recent relevant literature that addresses cardiac bioenergetics, deficiency in myocardial bioenergetics, and increased reactive oxygen species associated with HFpEF and the promising potential use of antioxidants in managing this condition.
Collapse
Affiliation(s)
| | - Qiuhua Shen
- University of Kansas, School of Nursing, Kansas, USA
| | | | - Janet Pierce
- University of Kansas, School of Nursing, Kansas, USA
| |
Collapse
|
28
|
Fu B, Guo S, Yu X, Chang H, Wang W. Astragalus Salvia Granules to Benefit the Qi (Qishen Yiqi Keli) protects H9C2 cardiomyocytes by suppressing oxidative stress. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2016. [DOI: 10.1016/j.jtcms.2016.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
29
|
Liu MH, Lin XL, Yuan C, He J, Tan TP, Wu SJ, Yu S, Chen L, Liu J, Tian W, Chen YD, Fu HY, Li J, Zhang Y. Hydrogen sulfide attenuates doxorubicin-induced cardiotoxicity by inhibiting the expression of peroxiredoxin III in H9c2 cells. Mol Med Rep 2015; 13:367-72. [PMID: 26573464 DOI: 10.3892/mmr.2015.4544] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 10/20/2015] [Indexed: 11/06/2022] Open
Abstract
Doxorubicin (DOX) is a widely used chemotherapeutic agent, which can give rise to severe cardiotoxicity, limiting its clinical use. Preliminary evidence suggests that hydrogen sulfide (H2S) may exert protective effects on DOX‑induced cardiotoxicity. Therefore, the aim of the present study was to investigate whether peroxiredoxin III is involved in the cardioprotection of H2S against DOX‑induced cardiotoxicity. The results demonstrated that DOX not only markedly induced injuries, including cytotoxicity and apoptosis, it also increased the expression levels of peroxiredoxin III. Notably, pretreatment with sodium hydrosulfide significantly attenuated the DOX‑induced decrease in cell viability and increase in apoptosis, and also reversed the increased expression levels of peroxiredoxin III in H9c2 cardiomyocytes. In addition, pretreatment of the H9c2 cells with N‑acetyl‑L‑cysteine, a scavenger of reactive oxygen species, prior to exposure to DOX markedly decreased the expression levels of peroxiredoxin III. In conclusion, the results of the present study suggested that exogenous H2S attenuates DOX‑induced cardiotoxicity by inhibiting the expression of peroxiredoxin III in H9c2 cells. In the present study, the apoptosis of H9c2 cardiomyocytes was assessed using an methyl thiazolyl tetrazolium assay and Hoechst staining. The levels of Prx III and cystathionine-γ-lyase were examined by western blotting.
Collapse
Affiliation(s)
- Mi-Hua Liu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiao-Long Lin
- Department of Pathology, Affiliated Huizhou Hospital of Guangzhou Medical University, The Third People's Hospital of Huizhou, Huizhou, Guangdong 516002, P.R. China
| | - Cong Yuan
- Department of Cardiology, The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Jun He
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Tian-Ping Tan
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shao-Jian Wu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shan Yu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Li Chen
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jun Liu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wei Tian
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yu-Dan Chen
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong-Yun Fu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jian Li
- Department of Ultrasonic Diagnosis, Bo'ai Hospital of Zhongshan, Zhongshan, Guangdong 528403, P.R. China
| | - Yuan Zhang
- Department of Pathology, Mawangdui Hospital, Changsha, Hunan 410016, P.R. China
| |
Collapse
|
30
|
Shimizu T, Yoshihisa A, Kanno Y, Takiguchi M, Sato A, Miura S, Nakamura Y, Yamauchi H, Owada T, Abe S, Sato T, Suzuki S, Oikawa M, Yamaki T, Sugimoto K, Kunii H, Nakazato K, Suzuki H, Saitoh SI, Takeishi Y. Relationship of hyperuricemia with mortality in heart failure patients with preserved ejection fraction. Am J Physiol Heart Circ Physiol 2015; 309:H1123-9. [PMID: 26297226 DOI: 10.1152/ajpheart.00533.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/17/2015] [Indexed: 01/08/2023]
Abstract
Serum uric acid is a predictor of cardiovascular mortality in heart failure with reduced ejection fraction. However, the impact of uric acid on heart failure with preserved ejection fraction (HFpEF) remains unclear. Here, we investigated the association between hyperuricemia and mortality in HFpEF patients. Consecutive 424 patients, who were admitted to our hospital for decompensated heart failure and diagnosed as having HFpEF, were divided into two groups based on presence of hyperuricemia (serum uric acid ≥7 mg/dl or taking antihyperuricemic agents). We compared patient characteristics, echocardiographic data, cardio-ankle vascular index, and cardiopulmonary exercise test findings between the two groups and prospectively followed cardiac and all-cause mortality. Compared with the non-hyperuricemia group (n = 170), the hyperuricemia group (n = 254) had a higher prevalence of hypertension (P = 0.013), diabetes mellitus (P = 0.01), dyslipidemia (P = 0.038), atrial fibrillation (P = 0.001), and use of diuretics (P < 0.001). Cardio-ankle vascular index (8.7 vs. 7.5, P < 0.001) and V̇e/V̇co2 slope (34.9 vs. 31.9, P = 0.02) were also higher. In addition, peak V̇o2 (14.9 vs. 17.9 ml·kg(-1)·min(-1), P < 0.001) was lower. In the follow-up period (mean 897 days), cardiac and all-cause mortalities were significantly higher in those with hyperuricemia (P = 0.006 and P = 0.004, respectively). In the multivariable Cox proportional hazard analyses after adjustment for several confounding factors including chronic kidney disease and use of diuretics, hyperuricemia was an independent predictor of all-cause mortality (hazard ratio 1.98, 95% confidence interval 1.036-3.793, P = 0.039). Hyperuricemia is associated with arterial stiffness, impaired exercise capacity, and high mortality in HFpEF.
Collapse
Affiliation(s)
- Takeshi Shimizu
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Akiomi Yoshihisa
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and Department of Advanced Cardiac Therapeutics, Fukushima Medical University, Fukushima, Japan
| | - Yuki Kanno
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Mai Takiguchi
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Akihiko Sato
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Shunsuke Miura
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Yuichi Nakamura
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Hiroyuki Yamauchi
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Takashi Owada
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Satoshi Abe
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Takamasa Sato
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Satoshi Suzuki
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and Department of Advanced Cardiac Therapeutics, Fukushima Medical University, Fukushima, Japan
| | - Masayoshi Oikawa
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Takayoshi Yamaki
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Koichi Sugimoto
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Hiroyuki Kunii
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Kazuhiko Nakazato
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Hitoshi Suzuki
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Shu-ichi Saitoh
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and
| | - Yasuchika Takeishi
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan; and Department of Advanced Cardiac Therapeutics, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
31
|
McDonald KS, Emter CA. Exploring new concepts in the management of heart failure with preserved ejection fraction: is exercise the key for improving treatment? J Appl Physiol (1985) 2015; 119:724-5. [PMID: 26229001 DOI: 10.1152/japplphysiol.00570.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Kerry S McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri; and
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
32
|
Skyttä T, Tuohinen S, Boman E, Virtanen V, Raatikainen P, Kellokumpu-Lehtinen PL. Troponin T-release associates with cardiac radiation doses during adjuvant left-sided breast cancer radiotherapy. Radiat Oncol 2015; 10:141. [PMID: 26159409 PMCID: PMC4496940 DOI: 10.1186/s13014-015-0436-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/05/2015] [Indexed: 12/13/2022] Open
Abstract
Background Adjuvant radiotherapy (RT) for left-sided breast cancer increases cardiac morbidity and mortality. For the heart, no safe radiation threshold has been established. Troponin T is a sensitive marker of myocardial damage. Our aim was to evaluate the effect of left-sided breast cancer RT on serum high sensitivity troponin T (hscTnT) levels and its association with cardiac radiation doses and echocardiographic parameters. Methods A total of 58 patients with an early stage, left-sided breast cancer or ductal carcinoma in situ (DCIS) who received adjuvant breast RT without prior chemotherapy were included in this prospective, non-randomized study. Serum samples were taken before, during and after RT. An increase of hscTnT >30 % was predefined as significant. A comprehensive 2D echocardiograph and electrocardiogram (ECG) were performed before and after RT. Dose-volume histograms (DVHs) were generated for different cardiac structures. Results The hscTnT increased during RT from baseline in 12/58 patients (21 %). Patients with increased hscTnT values (group A, N = 12) had significantly higher radiation doses for the whole heart (p = 0.02) and left ventricle (p = 0.03) than patients without hscTnT increase (group B, N = 46). For the left anterior descending artery (LAD), differences between groups A and B were found in volumes receiving 15 Gy (p = 0.03) and 20 Gy (p = 0.03) Furthermore, after RT, the interventricular septum thickened (p = 0.01), and the deceleration time was prolonged (p = 0.008) more in group A than in group B. Conclusions The increase in hscTnT level during adjuvant RT was positively associated with the cardiac radiation doses for the whole heart and LV in chemotherapy-naive breast cancer patients. Whether these acute subclinical changes increase the risk of excessive long-term cardiovascular morbidity or mortality, will be addressed in the follow-up of our patients.
Collapse
Affiliation(s)
- Tanja Skyttä
- Department of Oncology and School of Medicine, Tampere University Hospital, University of Tampere, 33521, Tampere, Finland.
| | - Suvi Tuohinen
- Heart Center Co and School of Medicine, University of Tampere, Tampere University Hospital, Tampere, Finland
| | - Eeva Boman
- Department of Oncology, Tampere University Hospital and Department of Medical Physics, Medical Imaging Center and Hospital Pharmacy, Tampere, Finland
| | - Vesa Virtanen
- Heart Center Co and School of Medicine, University of Tampere, Tampere University Hospital, Tampere, Finland
| | - Pekka Raatikainen
- Department of Medicine, Central Finland Health Care District and University of Eastern Finland, Jyväskylä, Finland
| | | |
Collapse
|