1
|
Mollace R, Scarano F, Bava I, Carresi C, Maiuolo J, Tavernese A, Gliozzi M, Musolino V, Muscoli S, Palma E, Muscoli C, Salvemini D, Federici M, Macrì R, Mollace V. Modulation of the nitric oxide/cGMP pathway in cardiac contraction and relaxation: Potential role in heart failure treatment. Pharmacol Res 2023; 196:106931. [PMID: 37722519 DOI: 10.1016/j.phrs.2023.106931] [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: 05/31/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Evidence exists that heart failure (HF) has an overall impact of 1-2 % in the global population being often associated with comorbidities that contribute to increased disease prevalence, hospitalization, and mortality. Recent advances in pharmacological approaches have significantly improved clinical outcomes for patients with vascular injury and HF. Nevertheless, there remains an unmet need to clarify the crucial role of nitric oxide/cyclic guanosine 3',5'-monophosphate (NO/cGMP) signalling in cardiac contraction and relaxation, to better identify the key mechanisms involved in the pathophysiology of myocardial dysfunction both with reduced (HFrEF) as well as preserved ejection fraction (HFpEF). Indeed, NO signalling plays a crucial role in cardiovascular homeostasis and its dysregulation induces a significant increase in oxidative and nitrosative stress, producing anatomical and physiological cardiac alterations that can lead to heart failure. The present review aims to examine the molecular mechanisms involved in the bioavailability of NO and its modulation of downstream pathways. In particular, we focus on the main therapeutic targets and emphasize the recent evidence of preclinical and clinical studies, describing the different emerging therapeutic strategies developed to counteract NO impaired signalling and cardiovascular disease (CVD) development.
Collapse
Affiliation(s)
- Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Federica Scarano
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Irene Bava
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Cristina Carresi
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Jessica Maiuolo
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Annamaria Tavernese
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Micaela Gliozzi
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Vincenzo Musolino
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Saverio Muscoli
- Division of Cardiology, Foundation PTV Polyclinic Tor Vergata, Rome 00133, Italy
| | - Ernesto Palma
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Carolina Muscoli
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy.
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy; Renato Dulbecco Institute, Lamezia Terme, Catanzaro 88046, Italy.
| |
Collapse
|
2
|
Rodkin S, Nwosu C, Sannikov A, Tyurin A, Chulkov VS, Raevskaya M, Ermakov A, Kirichenko E, Gasanov M. The Role of Gasotransmitter-Dependent Signaling Mechanisms in Apoptotic Cell Death in Cardiovascular, Rheumatic, Kidney, and Neurodegenerative Diseases and Mental Disorders. Int J Mol Sci 2023; 24:ijms24076014. [PMID: 37046987 PMCID: PMC10094524 DOI: 10.3390/ijms24076014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 04/14/2023] Open
Abstract
Cardiovascular, rheumatic, kidney, and neurodegenerative diseases and mental disorders are a common cause of deterioration in the quality of life up to severe disability and death worldwide. Many pathological conditions, including this group of diseases, are based on increased cell death through apoptosis. It is known that this process is associated with signaling pathways controlled by a group of gaseous signaling molecules called gasotransmitters. They are unique messengers that can control the process of apoptosis at different stages of its implementation. However, their role in the regulation of apoptotic signaling in these pathological conditions is often controversial and not completely clear. This review analyzes the role of nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H2S), and sulfur dioxide (SO2) in apoptotic cell death in cardiovascular, rheumatic, kidney, and neurodegenerative diseases. The signaling processes involved in apoptosis in schizophrenia, bipolar, depressive, and anxiety disorders are also considered. The role of gasotransmitters in apoptosis in these diseases is largely determined by cell specificity and concentration. NO has the greatest dualism; scales are more prone to apoptosis. At the same time, CO, H2S, and SO2 are more involved in cytoprotective processes.
Collapse
Affiliation(s)
- Stanislav Rodkin
- Faculty of Bioengineering and Veterinary Medicine, Department of Bioengineering, Don State Technical University, Rostov-on-Don 344000, Russia
| | - Chizaram Nwosu
- Faculty of Bioengineering and Veterinary Medicine, Department of Bioengineering, Don State Technical University, Rostov-on-Don 344000, Russia
| | - Alexander Sannikov
- Department of Psychiatry, Rostov State Medical University, Rostov-on-Don 344022, Russia
| | - Anton Tyurin
- Internal Medicine Department, Bashkir State Medical University, Ufa 450008, Russia
| | | | - Margarita Raevskaya
- Faculty of Bioengineering and Veterinary Medicine, Department of Bioengineering, Don State Technical University, Rostov-on-Don 344000, Russia
| | - Alexey Ermakov
- Faculty of Bioengineering and Veterinary Medicine, Department of Bioengineering, Don State Technical University, Rostov-on-Don 344000, Russia
| | - Evgeniya Kirichenko
- Faculty of Bioengineering and Veterinary Medicine, Department of Bioengineering, Don State Technical University, Rostov-on-Don 344000, Russia
| | - Mitkhat Gasanov
- Department of Internal Diseases #1, Rostov State Medical University, Rostov-on-Don 344022, Russia
| |
Collapse
|
3
|
LaPenna KB, Li Z, Doiron JE, Sharp TE, Xia H, Moles K, Koul K, Wang JS, Polhemus DJ, Goodchild TT, Patel RB, Shah SJ, Lefer DJ. Combination Sodium Nitrite and Hydralazine Therapy Attenuates Heart Failure With Preserved Ejection Fraction Severity in a "2-Hit" Murine Model. J Am Heart Assoc 2023; 12:e028480. [PMID: 36752224 PMCID: PMC10111505 DOI: 10.1161/jaha.122.028480] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/04/2023] [Indexed: 02/09/2023]
Abstract
Background Recent studies have suggested that cardiac nitrosative stress mediated by pathological overproduction of nitric oxide (NO) via inducible NO synthase (iNOS) contributes to the pathogenesis of heart failure with preserved ejection fraction (HFpEF). Other studies have suggested that endothelial NO synthase (eNOS) dysfunction and attenuated NO bioavailability contribute to HFpEF morbidity and mortality. We sought to further investigate dysregulated NO signaling and to examine the effects of a NO-based dual therapy (sodium nitrite+hydralazine) following the onset of HFpEF using a "2-hit" murine model. Methods and Results Nine-week-old male C57BL/6 N mice (n=15 per group) were treated concurrently with high-fat diet and N(ω)-nitro-L-arginine methyl ester (L-NAME) (0.5 g/L per day) via drinking water for 10 weeks. At week 5, mice were randomized into either vehicle (normal saline) or combination treatment with sodium nitrite (75 mg/L in the drinking water) and hydralazine (2.0 mg/kg IP, BID). Cardiac structure and function were monitored with echocardiography and invasive hemodynamic measurements. Cardiac mitochondrial respiration, aortic vascular function, and exercise performance were also evaluated. Circulating and myocardial nitrite were measured to determine the bioavailability of NO. Circulating markers of oxidative or nitrosative stress as well as systemic inflammation were also determined. Severe HFpEF was evident by significantly elevated E/E', LVEDP, and Tau in mice treated with L-NAME and HFD, which was associated with impaired NO bioavailability, mitochondrial respiration, aortic vascular function, and exercise capacity. Treatment with sodium nitrite and hydralazine restored NO bioavailability, reduced oxidative and nitrosative stress, preserved endothelial function and mitochondrial respiration, limited the fibrotic response, and improved exercise capacity, ultimately attenuating the severity of "two-hit" HFpEF. Conclusions Our data demonstrate that nitrite, a well-established biomarker of NO bioavailability and a physiological source of NO, is significantly reduced in the heart and circulation in the "2-hit" mouse HFpEF model. Furthermore, sodium nitrite+hydralazine combined therapy significantly attenuated the severity of HFpEF in the "2-hit" cardiometabolic HFpEF. These data suggest that supplementing NO-based therapeutics with a potent antioxidant and vasodilator agent may result in synergistic benefits for the treatment of HFpEF.
Collapse
Affiliation(s)
- Kyle B. LaPenna
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA
| | - Zhen Li
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCA
| | - Jake E. Doiron
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA
| | - Thomas E. Sharp
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
- Department of Medicine, Section of CardiologyLouisiana State University Health Sciences CenterNew OrleansLA
| | - Huijing Xia
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | - Karl Moles
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | - Kashyap Koul
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | - John S. Wang
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | | | - Traci T. Goodchild
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCA
| | - Ravi B. Patel
- Division of Cardiology, Department of Medicine and Bluhm Cardiovascular InstituteNorthwestern University Feinberg School of MedicineChicagoIL
| | - Sanjiv J. Shah
- Division of Cardiology, Department of Medicine and Bluhm Cardiovascular InstituteNorthwestern University Feinberg School of MedicineChicagoIL
| | - David J. Lefer
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCA
| |
Collapse
|
4
|
Islam RK, Donnelly E, Donnarumma E, Hossain F, Gardner JD, Islam KN. H 2S Prodrug, SG-1002, Protects against Myocardial Oxidative Damage and Hypertrophy In Vitro via Induction of Cystathionine β-Synthase and Antioxidant Proteins. Biomedicines 2023; 11:biomedicines11020612. [PMID: 36831146 PMCID: PMC9953594 DOI: 10.3390/biomedicines11020612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Endogenously produced hydrogen sulfide (H2S) is critical for cardiovascular homeostasis. Therapeutic strategies aimed at increasing H2S levels have proven cardioprotective in models of acute myocardial infarction (MI) and heart failure (HF). The present study was undertaken to investigate the effects of a novel H2S prodrug, SG-1002, on stress induced hypertrophic signaling in murine HL-1 cardiac muscle cells. Treatment of HL-1 cells with SG-1002 under serum starvation without or with H2O2 increased the levels of H2S, H2S producing enzyme, and cystathionine β-synthase (CBS), as well as antioxidant protein levels, such as super oxide dismutase1 (SOD1) and catalase, and additionally decreased oxidative stress. SG-1002 also decreased the expression of hypertrophic/HF protein markers such as atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), galectin-3, TIMP1, collagen type III, and TGF-β1 in stressed HL-1 cells. Treatment with SG-1002 caused a significant induction of cell viability and a marked reduction of cellular cytotoxicity in HL-1 cells under serum starvation incubated without or with H2O2. Experimental results of this study suggest that SG-1002 attenuates myocardial cellular oxidative damage and/or hypertrophic signaling via increasing H2S levels or H2S producing enzymes, CBS, and antioxidant proteins.
Collapse
Affiliation(s)
- Rahib K. Islam
- Departments of Pharmacology and Experimental Medicine, Genetics, and Physiology, Louisiana State University Health Sciences Center, 1901 Perdido St., New Orleans, LA 70112, USA
| | - Erinn Donnelly
- Departments of Pharmacology and Experimental Medicine, Genetics, and Physiology, Louisiana State University Health Sciences Center, 1901 Perdido St., New Orleans, LA 70112, USA
| | - Erminia Donnarumma
- Mitochondrial Biology Group, Institute Pasteur, CNRS UMR 3691, 75015 Paris, France
| | - Fokhrul Hossain
- Departments of Pharmacology and Experimental Medicine, Genetics, and Physiology, Louisiana State University Health Sciences Center, 1901 Perdido St., New Orleans, LA 70112, USA
| | - Jason D. Gardner
- Departments of Pharmacology and Experimental Medicine, Genetics, and Physiology, Louisiana State University Health Sciences Center, 1901 Perdido St., New Orleans, LA 70112, USA
| | - Kazi N. Islam
- Agricultural Research Development Program, College of Engineering, Science, Technology and Agriculture, Central State University, 1400 Brush Row Road, Wilberforce, OH 45384, USA
- Correspondence: ; Tel.: +1-937-376-6635
| |
Collapse
|
5
|
Cryptotanshinone Attenuated Pathological Cardiac Remodeling In Vivo and In Vitro Experiments. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:4015199. [PMID: 36743695 PMCID: PMC9897919 DOI: 10.1155/2023/4015199] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 12/03/2022] [Accepted: 12/29/2022] [Indexed: 01/30/2023]
Abstract
Objective Cardiac remodeling has been demonstrated to be the early stage and common pathway for various types of cardiomyopathy, but no specific treatment has been suggested to prevent its development and progress. This study was aimed at assessing whether Cryptotanshinone (CTS) treatment could effectively attenuate cardiac remodeling in vivo and in vitro. Methods Aortic banding (AB) surgery was performed to establish a pressure-overload-induced mouse cardiac remodeling model. Echocardiography and pressure-volume proof were used to examine mouse cardiac function. Hematoxylin and eosin (HE) and Picro-Sirius Red (PSR) staining were used to assess cardiac remodeling in vivo. Mouse hearts were collected to analysis signaling pathway and cardiac remodeling markers, respectively. Furthermore, neonatal rat cardiomyocyte (NRCMs) and cardiac fibroblast (CF) were isolated to investigate the roles and mechanisms of CTS treatment in vitro. Results CTS administration significantly alleviated pressure-overload-induced mouse cardiac dysfunction, inhibited cardiac hypertrophy, and reduced cardiac fibrosis. Mechanically, CTS treatment significantly inhibited the STAT3 and TGF-β/SMAD3 signaling pathways. In vitro experiments, CTS treatment markedly inhibited AngII-induced cardiomyocyte hypertrophy and TGF-β-induced myofibroblast activation via inhibiting STAT3 phosphorylation and its nuclear translocation. Finally, CTS treatment could not protect against pressure overload-induced mouse cardiac remodeling after adenovirus-associated virus (AAV)9-mediated STAT3 overexpression in mouse heart. Conclusion CTS treatment might attenuate pathological cardiac remodeling via inhibiting STAT3-dependent pathway.
Collapse
|
6
|
Beetroot Juice Produces Changes in Heart Rate Variability and Reduces Internal Load during Resistance Training in Men: A Randomized Double-Blind Crossover. Nutrients 2022; 14:nu14235119. [PMID: 36501148 PMCID: PMC9738238 DOI: 10.3390/nu14235119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/23/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
Beetroot juice (BJ) has been used as a sport supplement, improving performance in resistance training (RT). However, its effect on the modulation of the autonomic nervous system has not yet been widely studied. Therefore, the objective of this randomized double-blind crossover study was to assess the effect of acute BJ supplementation compared to placebo in blood pressure (BP), heart rate (HR), heart rate variability (HRV) and internal load during RT measure as Root Mean Square of the Successive Differences between adjacent RR intervals Slope (RMSSD and RMSSD-Slope, respectively). Eleven men performed an incremental RT test (three sets at 60%, 70% and 80% of their repetition maximum) composed by back squat and bench press with. HR, HRV and RMSSD-Slope were measured during and post exercise. As the main results, RMSSD during exercise decrease in the BJ group compared to placebo (p = 0.023; ES = 0.999), there were no differences in RMSSD post-exercise, and there were differences in RMSSD-Slope between groups in favor of the BJ group (p = 0.025; ES = 1.104) with a lower internal load. In conclusion, BJ supplementation seems to be a valuable tool for the reduction in the internal load of exercise during RT measured as RMSSD-Slope while enhancing performance.
Collapse
|
7
|
Chen Y, Li L, Hu C, Zhao X, Zhang P, Chang Y, Shang Y, Pang Y, Qian W, Qiu X, Zhang H, Zhang D, Zhang S, Li Y. Lingguizhugan decoction dynamically regulates MAPKs and AKT signaling pathways to retrogress the pathological progression of cardiac hypertrophy to heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153951. [PMID: 35131606 DOI: 10.1016/j.phymed.2022.153951] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/08/2022] [Accepted: 01/15/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Heart failure (HF) is a grave health concern, with high morbidity and mortality, calling for the urgent need for new and alternative pharmacotherapies. Lingguizhugan decoction (LD) is a classic Chinese formula clinically used to treat HF. However, the underlying mechanisms involved are not fully elucidated. PURPOSE Based on that, this study aims to investigate the effects and underlying mechanisms of LD on HF. METHODS After confirming the therapeutic benefits of LD in transverse aortic constriction (TAC)-induced HF mice, network pharmacology and transcriptomic analyzes were utilized to predict the potential molecular targets and pathways of LD treatment in failing hearts, which were evaluated at 3 and 9 w after TAC. UHPLC-QE-MS analysis was utilized to detect bioactive ingredients from LD and plasma of LD-treated rats. RESULTS Our results showed that LD markedly alleviated cardiac dysfunction via down-regulating CH-related genes and proteins expression in TAC mice. Significantly, cardiac hypertrophy signaling, including AKT and MAPKs signaling pathways, were identified, suggesting the pathways as likely regulatory targets for LD treatment. LD inhibited p38 and ERK phosphorylated expression levels, with the latter effect likely dependent on regulation of AMPK. Interestingly, LD exerted a dual modulatory role in the AKT-GSK3β/mTOR/P70S6K signaling pathway's regulation, which was characterized by stimulatory activity at 3 w and inhibitory effects at 9 w. Finally, 15 bioactive compounds detected from plasma were predicted as the potential regulators of the AKT-GSK3β/mTOR and MAPKs signaling pathways. CONCLUSION Our study shows LD's therapeutic efficacy in failing hearts, signifies LD as HF medication that acts dynamically by balancing AKT-GSK3β/mTOR/P70S6K and MAPKs pathways, and reveals possible bioactive compounds responsible for LD effects on HF.
Collapse
Affiliation(s)
- Yao Chen
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Cunyu Hu
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Xin Zhao
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Peng Zhang
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Yanxu Chang
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Ye Shang
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Yafen Pang
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Weiqiang Qian
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Xianzhe Qiu
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Hongxia Zhang
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, 69 Zeng Chan Road, He Bei, Tianjin 300250, China
| | - Deqin Zhang
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Shukun Zhang
- Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, 6 Chang Jiang Road, Nan Kai, Tianjin 300100, China.
| | - Yuhong Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China.
| |
Collapse
|
8
|
Gee LC, Massimo G, Lau C, Primus C, Fernandes D, Chen J, Rathod KS, Hamers AJP, Filomena F, Nuredini G, Ibrahim AS, Khambata RS, Gupta AK, Moon JC, Kapil V, Ahluwalia A. Inorganic nitrate attenuates cardiac dysfunction: role for xanthine oxidoreductase and nitric oxide. Br J Pharmacol 2021; 179:4757-4777. [PMID: 34309015 DOI: 10.1111/bph.15636] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 11/28/2022] Open
Abstract
Nitric oxide (NO) is a vasodilator and independent modulator of cardiac remodelling. Commonly, in cardiac disease (e.g. heart failure) endothelial dysfunction (synonymous with NO-deficiency) has been implicated in increased blood pressure (BP), cardiac hypertrophy and fibrosis. Currently no effective therapies replacing NO have succeeded in the clinic. Inorganic nitrate (NO3 - ), through chemical reduction to nitrite and then NO, exerts potent BP-lowering but whether it might be useful in treating undesirable cardiac remodelling is unknown. In a nested age- and sex-matched case-control study of hypertensive patients +/- left ventricular hypertrophy (NCT03088514) we show that lower plasma nitrite concentration and vascular dysfunction accompany cardiac hypertrophy and fibrosis in patients. In mouse models of cardiac remodelling, we also show that restoration of circulating nitrite levels using dietary nitrate improves endothelial dysfunction through targeting of xanthine oxidoreductase (XOR)-driven H2 O2 and superoxide, and reduces cardiac fibrosis through NO-mediated block of SMAD-phosphorylation leading to improvements in cardiac structure and function. We show that via these mechanisms dietary nitrate offers easily translatable therapeutic options for treatment of cardiac dysfunction.
Collapse
Affiliation(s)
- Lorna C Gee
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Gianmichele Massimo
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Clement Lau
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Christopher Primus
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Daniel Fernandes
- Departamento de Farmacologia, Federal University of Santa Catarina, Florianópolis, Santa Catarina,, Brazil
| | - Jianmin Chen
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Krishnaraj S Rathod
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Alexander Jozua Pedro Hamers
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Federica Filomena
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Gani Nuredini
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Abdiwahab Shidane Ibrahim
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Rayomand S Khambata
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Ajay K Gupta
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - James C Moon
- UCL Institute of Cardiovascular Science, University College London, London, UK
| | - Vikas Kapil
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
9
|
Attenuating senescence and dead cells accumulation as heart failure therapy: Break the communication networks. Int J Cardiol 2021; 334:72-85. [PMID: 33794236 DOI: 10.1016/j.ijcard.2021.03.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 02/03/2023]
|
10
|
Pansani AP, Ghazale PP, Dos Santos EG, Dos Santos Borges K, Gomes KP, Lacerda IS, Castro CH, Mendes EP, Dos Santos FCA, Biancardi MF, Nejm MB, Dogini DB, Rabelo LA, Nunes-Souza V, Scorza FA, Colugnati DB. The number and periodicity of seizures induce cardiac remodeling and changes in micro-RNA expression in rats submitted to electric amygdala kindling model of epilepsy. Epilepsy Behav 2021; 116:107784. [PMID: 33548915 DOI: 10.1016/j.yebeh.2021.107784] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/03/2021] [Accepted: 01/03/2021] [Indexed: 12/20/2022]
Abstract
Generalized tonic-clonic seizures (GTCS) are the main risk factor for sudden unexpected death in epilepsy (SUDEP). Also, among the several mechanisms underlying SUDEP there is the cardiac dysfunction. So, we aimed to evaluate the impact of the number of seizures on heart function and morphology in rats with epilepsy. Rats were randomized into three groups: Sham (without epilepsy), 5 S, and 10 S groups, referred as rats with epilepsy with a total of 5 or 10 GTCS, respectively. Epilepsy was induced by electrical amygdala kindling. The ventricular function was analyzed by the Langendorff technique and challenged by ischemia/reperfusion protocol. Cardiac fibrosis and hypertrophy were analyzed by histology. We also analyzed cardiac metalloproteinases (MMP2 and MMP9), ERK 1/2 and phosphorylated ERK1/2 (P-ERK) by western blot; microRNA-21 and -320 by RT-PCR; and oxidative stress (TBARS, catalase activity and nitrite) by biochemical analysis. Only the 5S group presented decreased values of ventricular function at before ischemia/reperfusion (baseline): intraventricular systolic pressure, developed intraventricular pressure, positive and negative dP/dt. During ischemia/reperfusion protocol, the variation of the ventricular function did not differ among groups. Both 5S and 10S groups had increased cardiomyocyte hypertrophy and fibrosis compared to Sham, but in the 5S group, these alterations were higher than in the 10S group. The 5S group increased in microRNA-21 and decreased in microRNA-320 expression compared to Sham and the 10S group. The 10S group increased in MMP9 and decreased in P-ERK/ERK expression, and increased in nitrite content compared to both Sham and the 5S group. Therefore, seizures impair cardiac function and morphology, probably through microRNA modulation. The continuation of seizures seems to exert a preconditioning-like stimulus that fails to compensate the cardiac tissue alteration.
Collapse
Affiliation(s)
- Aline Priscila Pansani
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil.
| | - Poliana Peres Ghazale
- Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, Brazil
| | - Emilly Gomes Dos Santos
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Kiscilla Dos Santos Borges
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Karina Pereira Gomes
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Ismaley Santos Lacerda
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Carlos Henrique Castro
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Elizabeth Pereira Mendes
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | | | - Mariana Bocca Nejm
- Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, Brazil
| | - Danyella Barbosa Dogini
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Luiza Antas Rabelo
- Department of Physiology, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, Alagoas, Brazil
| | - Valéria Nunes-Souza
- Department of Physiological and Pharmacology Sciences, Institute of Biological Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Fulvio Alexandre Scorza
- Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, Brazil
| | - Diego Basile Colugnati
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
11
|
Rahman MA, Glasgow JN, Nadeem S, Reddy VP, Sevalkar RR, Lancaster JR, Steyn AJC. The Role of Host-Generated H 2S in Microbial Pathogenesis: New Perspectives on Tuberculosis. Front Cell Infect Microbiol 2020; 10:586923. [PMID: 33330130 PMCID: PMC7711268 DOI: 10.3389/fcimb.2020.586923] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
For centuries, hydrogen sulfide (H2S) was considered primarily as a poisonous gas and environmental hazard. However, with the discovery of prokaryotic and eukaryotic enzymes for H2S production, breakdown, and utilization, H2S has emerged as an important signaling molecule in a wide range of physiological and pathological processes. Hence, H2S is considered a gasotransmitter along with nitric oxide (•NO) and carbon monoxide (CO). Surprisingly, despite having overlapping functions with •NO and CO, the role of host H2S in microbial pathogenesis is understudied and represents a gap in our knowledge. Given the numerous reports that followed the discovery of •NO and CO and their respective roles in microbial pathogenesis, we anticipate a rapid increase in studies that further define the importance of H2S in microbial pathogenesis, which may lead to new virulence paradigms. Therefore, this review provides an overview of sulfide chemistry, enzymatic production of H2S, and the importance of H2S in metabolism and immunity in response to microbial pathogens. We then describe our current understanding of the role of host-derived H2S in tuberculosis (TB) disease, including its influences on host immunity and bioenergetics, and on Mycobacterium tuberculosis (Mtb) growth and survival. Finally, this review discusses the utility of H2S-donor compounds, inhibitors of H2S-producing enzymes, and their potential clinical significance.
Collapse
Affiliation(s)
| | - Joel N Glasgow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sajid Nadeem
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vineel P Reddy
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ritesh R Sevalkar
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jack R Lancaster
- Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Adrie J C Steyn
- Africa Health Research Institute, Durban, South Africa.,Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States.,Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
12
|
Pulmonary Hypertension and Heart Failure With Preserved Ejection Fraction: Treating Resistance, Impedance, and Compliance. J Card Fail 2020; 26:662-663. [DOI: 10.1016/j.cardfail.2020.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 11/21/2022]
|
13
|
Kapil V, Khambata RS, Jones DA, Rathod K, Primus C, Massimo G, Fukuto JM, Ahluwalia A. The Noncanonical Pathway for In Vivo Nitric Oxide Generation: The Nitrate-Nitrite-Nitric Oxide Pathway. Pharmacol Rev 2020; 72:692-766. [PMID: 32576603 DOI: 10.1124/pr.120.019240] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
In contrast to nitric oxide, which has well established and important roles in the regulation of blood flow and thrombosis, neurotransmission, the normal functioning of the genitourinary system, and the inflammation response and host defense, its oxidized metabolites nitrite and nitrate have, until recently, been considered to be relatively inactive. However, this view has been radically revised over the past decade and more. Much evidence has now accumulated demonstrating that nitrite serves as a storage form of nitric oxide, releasing nitric oxide preferentially under acidic and/or hypoxic conditions but also occurring under physiologic conditions: a phenomenon that is catalyzed by a number of distinct mammalian nitrite reductases. Importantly, preclinical studies demonstrate that reduction of nitrite to nitric oxide results in a number of beneficial effects, including vasodilatation of blood vessels and lowering of blood pressure, as well as cytoprotective effects that limit the extent of damage caused by an ischemia/reperfusion insult, with this latter issue having been translated more recently to the clinical setting. In addition, research has demonstrated that the other main metabolite of the oxidation of nitric oxide (i.e., nitrate) can also be sequentially reduced through processing in vivo to nitrite and then nitrite to nitric oxide to exert a range of beneficial effects-most notably lowering of blood pressure, a phenomenon that has also been confirmed recently to be an effective method for blood pressure lowering in patients with hypertension. This review will provide a detailed description of the pathways involved in the bioactivation of both nitrate and nitrite in vivo, their functional effects in preclinical models, and their mechanisms of action, as well as a discussion of translational exploration of this pathway in diverse disease states characterized by deficiencies in bioavailable nitric oxide. SIGNIFICANCE STATEMENT: The past 15 years has seen a major revision in our understanding of the pathways for nitric oxide synthesis in the body with the discovery of the noncanonical pathway for nitric oxide generation known as the nitrate-nitrite-nitric oxide pathway. This review describes the molecular components of this pathway, its role in physiology, potential therapeutics of targeting this pathway, and their impact in experimental models, as well as the clinical translation (past and future) and potential side effects.
Collapse
Affiliation(s)
- V Kapil
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - R S Khambata
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - D A Jones
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - K Rathod
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - C Primus
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - G Massimo
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - J M Fukuto
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - A Ahluwalia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| |
Collapse
|
14
|
Vitturi DA, Maynard C, Olsufka M, Straub AC, Krehel N, Kudenchuk PJ, Nichol G, Sayre M, Kim F, Dezfulian C. Nitrite elicits divergent NO-dependent signaling that associates with outcome in out of hospital cardiac arrest. Redox Biol 2020; 32:101463. [PMID: 32087553 PMCID: PMC7033352 DOI: 10.1016/j.redox.2020.101463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/01/2020] [Accepted: 02/11/2020] [Indexed: 01/16/2023] Open
Abstract
Brain and heart injury cause most out-of-hospital cardiac arrest deaths but limited pharmacotherapy exists to protect these tissues. Nitrite is a nitric oxide precursor that is protective in pre-clinical models of ischemic injury and safe in Phase I testing. Protection may occur by cGMP generation via the sGC pathway or through S-nitrosothiol and nitrated conjugated linoleic acid (NO2-CLA) formation. We hypothesized that nitrite provided during CPR signals through multiple pathways and that activation of signals is associated with OHCA outcome. To this end, we performed a secondary analysis of a phase 1 study of intravenous nitrite administration during resuscitation in adult out-of-hospital cardiac arrest. Associations between whole blood nitrite and derived plasma signals (cGMP and NO2-CLA) with patient characteristics and outcomes were defined using Chi-square or t-tests and multiple logistic regression. Whole blood nitrite levels correlated inversely with plasma NO2-CLA (p = 0.039) but not with cGMP. Patients with shockable rhythms had higher cGMP (p = 0.027), NO2-CLA (p < 0.0001) and trended towards lower nitrite (p = 0.077). Importantly, plasma cGMP and NO2-CLA levels were higher in survivors (p = 0.033 and 0.019) and in those with good neurological outcome (p = 0.046 and 0.021). Nitrite was lower in patients with good neurologic outcome (p = 0.029). cGMP (OR 4.02; 95% CI 1.04–15.54; p = 0.044) and NO2-CLA (OR 3.74; 95% CI 1.11–12.65; p = 0.034) were associated with survival. Nitrite (OR 0.20; 95% CI 0.05–0.08; p = 0.026) and NO2-CLA (OR 3.96; 95% CI 1.01–15.60; p = 0.049) were associated with favorable neurologic outcome. In summary, nitrite administration was associated with increased plasma cGMP and NO2-CLA formation in selected OHCA patients. Furthermore, patients with the highest levels of cGMP and NO2-CLA were more likely to survive and experience better neurological outcomes.
Collapse
Affiliation(s)
- Dario A Vitturi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, USA; Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, USA
| | - Charles Maynard
- Department of Health Services, University of Washington, USA
| | - Michele Olsufka
- Department of Health Services, University of Washington, USA; Department of Medicine, Harborview Medical Center, University of Washington, USA
| | - Adam C Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, USA; Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, USA
| | - Nick Krehel
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh, USA
| | - Peter J Kudenchuk
- Department of Medicine, Harborview Medical Center, University of Washington, USA
| | - Graham Nichol
- Department of Medicine, Harborview Medical Center, University of Washington, USA
| | - Michael Sayre
- Department of Medicine, Harborview Medical Center, University of Washington, USA
| | - Francis Kim
- Department of Medicine, Harborview Medical Center, University of Washington, USA
| | - Cameron Dezfulian
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, USA; Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh, USA.
| |
Collapse
|
15
|
Huang J, Wang D, Huang LH, Huang H. Roles of Reconstituted High-Density Lipoprotein Nanoparticles in Cardiovascular Disease: A New Paradigm for Drug Discovery. Int J Mol Sci 2020; 21:ijms21030739. [PMID: 31979310 PMCID: PMC7037452 DOI: 10.3390/ijms21030739] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 02/08/2023] Open
Abstract
Epidemiological results revealed that there is an inverse correlation between high-density lipoprotein (HDL) cholesterol levels and risks of atherosclerotic cardiovascular disease (ASCVD). Mounting evidence supports that HDLs are atheroprotective, therefore, many therapeutic approaches have been developed to increase HDL cholesterol (HDL-C) levels. Nevertheless, HDL-raising therapies, such as cholesteryl ester transfer protein (CETP) inhibitors, failed to ameliorate cardiovascular outcomes in clinical trials, thereby casting doubt on the treatment of cardiovascular disease (CVD) by increasing HDL-C levels. Therefore, HDL-targeted interventional studies were shifted to increasing the number of HDL particles capable of promoting ATP-binding cassette transporter A1 (ABCA1)-mediated cholesterol efflux. One such approach was the development of reconstituted HDL (rHDL) particles that promote ABCA1-mediated cholesterol efflux from lipid-enriched macrophages. Here, we explore the manipulation of rHDL nanoparticles as a strategy for the treatment of CVD. In addition, we discuss technological capabilities and the challenge of relating preclinical in vivo mice research to clinical studies. Finally, by drawing lessons from developing rHDL nanoparticles, we also incorporate the viabilities and advantages of the development of a molecular imaging probe with HDL nanoparticles when applied to ASCVD, as well as gaps in technology and knowledge required for putting the HDL-targeted therapeutics into full gear.
Collapse
Affiliation(s)
- Jiansheng Huang
- Department of Medicine, Vanderbilt University Medical Center, 318 Preston Research Building, 2200 Pierce Avenue, Nashville, TN 37232, USA
- Correspondence:
| | - Dongdong Wang
- Institute of Clinical Chemistry, University Hospital Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland;
| | - Li-Hao Huang
- Pathology and Immunology Department, Washington University School of Medicine, St. Louis, MO 63110-1093, USA;
| | - Hui Huang
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA;
| |
Collapse
|
16
|
Abstract
Hydrogen sulfide (H2S)-a potent gaseous signaling molecule-has emerged as a critical regulator of cardiovascular homeostasis. H2S is produced enzymatically by 3 constitutively active endogenous enzymes in all mammalian species. Within the past 2 decades, studies administering H2S-donating agents and the genetic manipulation of H2S-producing enzymes have revealed multiple beneficial effects of H2S, including vasodilation, activation of antiapoptotic and antioxidant pathways, and anti-inflammatory effects. More recently, the heightened enthusiasm in this field has shifted to the development of novel H2S-donating agents that exert favorable pharmacological profiles. This has led to the discovery of novel H2S-mediated signaling pathways. This review will discuss recently developed H2S therapeutics, introduce signaling pathways that are influenced by H2S-dependent sulfhydration, and explore the dual-protective effect of H2S in cardiorenal syndrome.
Collapse
Affiliation(s)
- Zhen Li
- From the Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Science Center, New Orleans, LA
| | - David J Polhemus
- From the Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Science Center, New Orleans, LA
| | - David J Lefer
- From the Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Science Center, New Orleans, LA
| |
Collapse
|
17
|
Si-Miao-Yong-An decoction ameliorates cardiac function through restoring the equilibrium of SOD and NOX2 in heart failure mice. Pharmacol Res 2019; 146:104318. [DOI: 10.1016/j.phrs.2019.104318] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/29/2019] [Accepted: 06/16/2019] [Indexed: 11/19/2022]
|
18
|
Abstract
OBJECTIVE This research was aimed to investigate the correct dose of nitrite that would act as a protection against the ischemic effects induced by acute myocardial infarction (AMI). METHODS Mice were randomly divided into a sham-operation group (sham), an AMI operation group (AMI), and a nitrite pretreatment+AMI operation group (N+AMI). Seven days before the AMI operation, mice in the N+AMI group were pretreated with sodium nitrite in drinking water. RESULTS One week after the AMI operation, serum lactate dehydrogenase (LDH) and creatine kinase (CK) activities in both AMI and N+AMI group were significantly higher than those in the sham group, but there were no significant differences between AMI and N+AMI mice. Contents of inducible nitric oxide synthase (iNOS) in the noninfarct area of the left ventricle in the N+AMI mice were significantly higher than those in the AMI mice, with no difference in the infarct area. Coagulation necrosis in the cardiomyocytes was observed in both AMI and N+AMI mice; however, it was less severe in the N+AMI mice. Western blot analyses showed that nitrite pretreatment resulted in up-regulation of antiapoptotic factors Bcl-2 and p21waf1/cip1 signal proteins, but down-regulation of the proapoptotic factor Bax signal protein. Furthermore, nitrite pretreatment also showed significant alleviation of AMI-induced signal protein expressions of inflammatory factors of NF-K B and oxidative factors of Hsp 70 and HO-1. CONCLUSION These results suggest that nitrite show certain protective effects against the ischemic effects induced by AMI in mice, which might be attributed to the synthesis of NO induced by iNOS through up-regulation of antiapoptotic factors and down-regulation of proapoptotic and inflammatory factors.
Collapse
|
19
|
Mulkareddy V, Racette SB, Coggan AR, Peterson LR. Dietary nitrate's effects on exercise performance in heart failure with reduced ejection fraction (HFrEF). Biochim Biophys Acta Mol Basis Dis 2019; 1865:735-740. [PMID: 30261290 PMCID: PMC6401215 DOI: 10.1016/j.bbadis.2018.09.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/13/2018] [Accepted: 09/18/2018] [Indexed: 01/09/2023]
Abstract
Heart failure with reduced ejection fraction (HFrEF) is a deadly and disabling disease. A key derangement contributing to impaired exercise performance in HFrEF is decreased nitric oxide (NO) bioavailability. Scientists recently discovered the inorganic nitrate pathway for increasing NO. This has advantages over organic nitrates and NO synthase production of NO. Small studies using beetroot juice as a source of inorganic nitrate demonstrate its power to improve exercise performance in HFrEF. A larger-scale trial is now underway to determine if inorganic nitrate may be a new arrow for physicians' quiver of HFrEF treatments.
Collapse
Affiliation(s)
- Vinaya Mulkareddy
- The Department of Medicine, 4960 Children's Place, Campus Box 8066, St. Louis, MO 63110, USA.
| | - Susan B Racette
- The Department of Medicine, 4960 Children's Place, Campus Box 8066, St. Louis, MO 63110, USA; Program in Physical Therapy, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park Ave., St. Louis, MO 63108-2212, USA.
| | - Andrew R Coggan
- Department of Kinesiology, Indiana University Purdue University Indianapolis, 901 West New York Street, Indianapolis, IN 46202, USA; Department of Cellular and Integrative Physiology, Indiana University Purdue University Indianapolis, 901 West New York Street, Indianapolis, IN 46202, USA.
| | - Linda R Peterson
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
20
|
Yan X, Zhang YL, Zhang L, Zou LX, Chen C, Liu Y, Xia YL, Li HH. Gallic Acid Suppresses Cardiac Hypertrophic Remodeling and Heart Failure. Mol Nutr Food Res 2018; 63:e1800807. [PMID: 30521107 DOI: 10.1002/mnfr.201800807] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/21/2018] [Indexed: 12/14/2022]
Abstract
SCOPE Gallic acid (GA) is a dietary phenolic acid found in tea, red wine, and some plants. It exhibits anti-oxidative and anti-inflammatory activities. Recent studies have revealed that GA has beneficial effects against several cardiovascular diseases; however, whether GA attenuates pressure-overload-induced cardiac hypertrophy and the underlying mechanism remains unclear. METHODS AND RESULTS Primary cardiomyocyte hypertrophy is stimulated with angiotensin II (Ang II). Cardiac hypertrophic remodeling is induced in mice by transverse aortic constriction (TAC). Myocardial function is evaluated by echocardiographic and hemodynamic analyses, while cardiac tissues are analyzed by histological staining. It is observed that GA significantly decreases Ang II-induced increases in cardiomyocyte size in vitro. Administration of GA in mice markedly improves TAC-induced cardiac dysfunction and attenuates pathological changes, including cardiac myocyte hypertrophy, fibrosis, inflammation, and oxidative stress. Mechanistically, GA inhibits ULK1 and activates autophagy, which induces the degradation of EGFR, gp130, and calcineurin A, thereby inhibiting the downstream signaling cascades (AKT, ERK1/2, JAK2/STAT3, and NFATc1). CONCLUSIONS The results demonstrate for the first time that GA prevents myocardial hypertrophy and dysfunction via an autophagy-dependent mechanism. Thus, GA represents a promising therapeutic candidate for treating cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Xiao Yan
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, 116044, China.,Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yun-Long Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, 116044, China.,Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Liang Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, 116044, China.,Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Lei-Xin Zou
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, 116044, China.,Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Chen Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, 116044, China.,Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Ying Liu
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yun-Long Xia
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Hui-Hua Li
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, 116044, China.,Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| |
Collapse
|
21
|
Faconti L, Mills CE, Govoni V, Gu H, Morant S, Jiang B, Cruickshank JK, Webb AJ. Cardiac effects of 6 months' dietary nitrate and spironolactone in patients with hypertension and with/at risk of type 2 diabetes, in the factorial design, double-blind, randomized controlled VaSera trial. Br J Clin Pharmacol 2018; 85:169-180. [PMID: 30294825 DOI: 10.1111/bcp.13783] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/13/2018] [Accepted: 09/09/2018] [Indexed: 12/12/2022] Open
Abstract
AIMS The aims of the present study were to explore whether a long-term intervention with dietary nitrate [(NO3 - ), a potential tolerance-free source of beneficial vasoactive nitric oxide] and spironolactone (to oppose aldosterone's potential deleterious cardiovascular effects) improve cardiac structure/function, independently of blood pressure (BP), in patients with/at risk of type 2 diabetes (a population at risk of heart failure). METHODS A subsample of participants in our double-blind, randomized, factorial-design intervention (VaSera) trial of active beetroot juice as a nitrate source (≤11.2 mmol) or placebo (nitrate depleted) beetroot juice, and either ≤50 mg spironolactone or ≤16 mg doxazosin (control), had transthoracic cardiac ultrasounds at baseline (n = 105), and at 3 months and 6 months (n = 87) after the start of the intervention. Analysis was by modified intent-to-treat. RESULTS Nitrate-containing juice (n = 40) decreased left ventricular (LV) end-diastolic volume {-6.3 [95% confidence interval (CI) -11.1, -1.6] ml} and end-systolic volume [-3.2 (95% CI -5.9, -0.5) ml], and increased end-diastolic mass/volume ratio [+0.04 (95% CI 0.00, 0.07)], relative to placebo juice (n = 47). Spironolactone (n = 44) reduced relative wall thickness compared with doxazosin (n = 43) [-0.01 (95% CI -0.02, -0.00)]. Although spironolactone reduced LV mass index relative to baseline [-1.48 (95% CI -2.08, -0.88) g m-2.7 ], there was no difference vs. doxazosin [-0.85 (95% CI -1.76, 0.05) g m-2.7 ]. Spironolactone also decreased the E/A ratio [-0.12 (95% CI -0.19, -0.04)] and increased S' (a tissue-Doppler systolic function index) by 0.52 (95% CI 0.05, 1.0) cm s-1 . BP did not differ between the juices, or between the drugs. CONCLUSIONS Six months' dietary nitrate decreased LV volumes ~5%, representing new, sustained, BP-independent benefits on cardiac structure, extending mechanisms characterized in preclinical models of heart failure. Spironolactone's effects on cardiac remodelling and systolic-diastolic function, although confirmatory, were independent of BP.
Collapse
Affiliation(s)
- Luca Faconti
- Department of Clinical Pharmacology, School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, London, UK.,Department of Nutritional Sciences, School of Life Course Sciences, King's College London, London, UK.,Biomedical Research Centre, Clinical Research Facility, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Charlotte Elizabeth Mills
- Department of Nutritional Sciences, School of Life Course Sciences, King's College London, London, UK.,Biomedical Research Centre, Clinical Research Facility, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Virginia Govoni
- Department of Nutritional Sciences, School of Life Course Sciences, King's College London, London, UK.,Biomedical Research Centre, Clinical Research Facility, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Haotian Gu
- Department of Clinical Pharmacology, School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, London, UK.,Biomedical Research Centre, Clinical Research Facility, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Steven Morant
- Medicines Monitoring Unit (MEMO), University of Dundee, Dundee, UK
| | - Benju Jiang
- Department of Clinical Pharmacology, School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, London, UK.,Biomedical Research Centre, Clinical Research Facility, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - J Kennedy Cruickshank
- Department of Nutritional Sciences, School of Life Course Sciences, King's College London, London, UK.,Biomedical Research Centre, Clinical Research Facility, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Andrew James Webb
- Department of Clinical Pharmacology, School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, London, UK.,Biomedical Research Centre, Clinical Research Facility, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
22
|
An Update on Hydrogen Sulfide and Nitric Oxide Interactions in the Cardiovascular System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4579140. [PMID: 30271527 PMCID: PMC6151216 DOI: 10.1155/2018/4579140] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/25/2018] [Indexed: 01/19/2023]
Abstract
Hydrogen sulfide (H2S) and nitric oxide (NO) are now recognized as important regulators in the cardiovascular system, although they were historically considered as toxic gases. As gaseous transmitters, H2S and NO share a wide range of physical properties and physiological functions: they penetrate into the membrane freely; they are endogenously produced by special enzymes, they stimulate endothelial cell angiogenesis, they regulate vascular tone, they protect against heart injury, and they regulate target protein activity via posttranslational modification. Growing evidence has determined that these two gases are not independent regulators but have substantial overlapping pathophysiological functions and signaling transduction pathways. H2S and NO not only affect each other's biosynthesis but also produce novel species through chemical interaction. They play a regulatory role in the cardiovascular system involving similar signaling mechanisms or molecular targets. However, the natural precise mechanism of the interactions between H2S and NO remains unclear. In this review, we discuss the current understanding of individual and interactive regulatory functions of H2S and NO in biosynthesis, angiogenesis, vascular one, cardioprotection, and posttranslational modification, indicating the importance of their cross-talk in the cardiovascular system.
Collapse
|
23
|
Wallace JL, Vaughan D, Dicay M, MacNaughton WK, de Nucci G. Hydrogen Sulfide-Releasing Therapeutics: Translation to the Clinic. Antioxid Redox Signal 2018; 28:1533-1540. [PMID: 28388861 DOI: 10.1089/ars.2017.7068] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
SIGNIFICANCE Shortly after the discovery of the role of hydrogen sulfide (H2S) in many physiological and pathological processes, attempts were made to develop novel pharmaceuticals that may be of benefit for treatment or prevention of a wide range of disorders. The promise of H2S-based therapeutics is now being demonstrated in clinical trials. Recent Advances: H2S-releasing drugs, such as SG1002 for cardiovascular disorders, and ATB-346 for arthritis, have progressed into clinical trials and have shown considerable promise. Some older drugs, such as zofenopril, have now been recognized to produce at least some of the beneficial effects through release of H2S. CRITICAL ISSUES There remains a need to better understand the underlying mechanisms for some of the observed effects of H2S-releasing drugs in a clinical setting, such as the marked increase in analgesic potency that has been observed with ATB-346. FUTURE DIRECTIONS The proof-of-concept clinical studies reviewed herein pave the way for examination, in a clinical setting, of several other potential applications of H2S-based drugs in a wide range of disorders, including diabetes, hypertension, and cancer chemoprevention. Antioxid. Redox Signal. 28, 1533-1540.
Collapse
Affiliation(s)
- John L Wallace
- 1 Department of Physiology and Pharmacology, University of Calgary , Calgary, Canada .,2 Antibe Therapeutics, Inc. , Toronto, Canada .,3 Universidade Camilo Castelo Branco , Department of Medicine, Fernandopolis, Brazil
| | | | - Michael Dicay
- 1 Department of Physiology and Pharmacology, University of Calgary , Calgary, Canada
| | - Wallace K MacNaughton
- 1 Department of Physiology and Pharmacology, University of Calgary , Calgary, Canada
| | - Gilberto de Nucci
- 3 Universidade Camilo Castelo Branco , Department of Medicine, Fernandopolis, Brazil
| |
Collapse
|
24
|
Abstract
Metabolomics is the study of small, organic molecules within biochemical pathways. With advancement of technology, nuclear magnetic resonance, gas chromatography, and mass spectrometry have allowed for the discovery and analysis of large databases of metabolites implicated in heart failure. Metabolomics also explores the patient and environment interactions and unlocks the link between environmental exposures and the development of cardiovascular disease. Although a relatively new field, metabolomics is poised to become a clinically impactful field that develops novel biomarkers and explores new therapeutic interventions in heart failure.
Collapse
|
25
|
Trivedi RK, Polhemus DJ, Li Z, Yoo D, Koiwaya H, Scarborough A, Goodchild TT, Lefer DJ. Combined Angiotensin Receptor-Neprilysin Inhibitors Improve Cardiac and Vascular Function Via Increased NO Bioavailability in Heart Failure. J Am Heart Assoc 2018; 7:JAHA.117.008268. [PMID: 29502102 PMCID: PMC5866338 DOI: 10.1161/jaha.117.008268] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background There is a paucity of data about the mechanisms by which sacubitril/valsartan (also known as LCZ696) improves outcomes in patients with heart failure. Specifically, the effects of sacubitril/valsartan on vascular function and NO bioavailability have not been investigated. We hypothesized that sacubitril/valsartan therapy increases circulating NO levels and improves vascular function in the setting of heart failure. Methods and Results Male spontaneously hypertensive rats underwent myocardial ischemia/reperfusion surgery to induce heart failure and were followed for up to 12 weeks with serial echocardiography. Rats received sacubitril/valsartan (68 mg/kg), valsartan (31 mg/kg), or vehicle starting at 4 weeks after reperfusion. At 8 or 12 weeks of reperfusion, animals were euthanized and tissues were collected for ex vivo analyses of NO bioavailability, aortic vascular reactivity, myocardial and vascular histology, and cardiac molecular assays. Left ventricular structure and function were improved by both valsartan and sacubitril/valsartan compared with vehicle. Sacubitril/valsartan resulted in superior cardiovascular benefits, as evidenced by sustained improvements in left ventricular ejection fraction and end‐diastolic pressure. Ex vivo vascular function, as measured by aortic vasorelaxation responses to acetylcholine and sodium nitroprusside, was significantly improved by valsartan and sacubitril/valsartan, with more sustained improvements afforded by sacubitril/valsartan. Furthermore, myocardial NO bioavailability was significantly enhanced in animals receiving sacubitril/valsartan therapy. Conclusions Sacubitril/valsartan offers superior cardiovascular protection in heart failure and improves vascular function to a greater extent than valsartan alone. Sacubitril/valsartan‐mediated improvements in cardiac and vascular function are likely related to increases in NO bioavailability and explain, in part, the benefits beyond angiotensin receptor blockade.
Collapse
Affiliation(s)
- Rishi K Trivedi
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA.,Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - David J Polhemus
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA.,Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Zhen Li
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA.,Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Daniel Yoo
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Hiroshi Koiwaya
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Amy Scarborough
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Traci T Goodchild
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA.,Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - David J Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA .,Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA
| |
Collapse
|
26
|
Abstract
Nitric oxide (NO) signalling has pleiotropic roles in biology and a crucial function in cardiovascular homeostasis. Tremendous knowledge has been accumulated on the mechanisms of the nitric oxide synthase (NOS)-NO pathway, but how this highly reactive, free radical gas signals to specific targets for precise regulation of cardiovascular function remains the focus of much intense research. In this Review, we summarize the updated paradigms on NOS regulation, NO interaction with reactive oxidant species in specific subcellular compartments, and downstream effects of NO in target cardiovascular tissues, while emphasizing the latest developments of molecular tools and biomarkers to modulate and monitor NO production and bioavailability.
Collapse
Affiliation(s)
- Charlotte Farah
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Lauriane Y M Michel
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| |
Collapse
|
27
|
An injectable conductive hydrogel encapsulating plasmid DNA-eNOs and ADSCs for treating myocardial infarction. Biomaterials 2018; 160:69-81. [PMID: 29396380 DOI: 10.1016/j.biomaterials.2018.01.021] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/14/2018] [Indexed: 12/15/2022]
Abstract
Myocardial infarction (MI) leads to the mass death of cardiomyocytes accompanying with the unfavorable alternation of microenvironment, a fibrosis scar deprived of electrical communications, and the lack of blood supply in the infarcted myocardium. The three factors are inextricably intertwined and thus result in a conservative MI therapy efficacy in clinic. A holistic approach pertinently targeted to these three key points would be favorable to rebuild the heart functions. Here, an injectable conductive hydrogel was constructed via in situ Michael addition reaction between multi-armed conductive crosslinker tetraaniline-polyethylene glycol diacrylate (TA-PEG) and thiolated hyaluronic acid (HA-SH). The resultant soft conductive hydrogel with equivalent myocardial conductivity and anti-fatigue performance was loaded with plasmid DNA encoding eNOs (endothelial nitric oxide synthase) nanocomplexes and adipose derived stem cells (ADSCs) for treating MI. The TA-PEG/HA-SH/ADSCs/Gene hydrogel-based holistic system was injected into the infarcted myocardium of SD rats. We demonstrated an increased expression of eNOs in myocardial tissue the heightening of nitrite concentration, accompanied with upregulation of proangiogenic growth factors and myocardium related mRNA. The results of electrocardiography, cardiogram, and histological analysis convincingly revealed a distinct increase of ejection fraction (EF), shortened QRS interval, smaller infarction size, less fibrosis area, and higher vessel density, indicating a significant improvement of heart functions. This conception of combination approach by a conductive injectable hydrogel loaded with stem cells and gene-encoding eNOs nanoparticles will become a robust therapeutic strategy for the treatment of MI.
Collapse
|
28
|
NO Signaling in the Cardiovascular System and Exercise. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1000:211-245. [DOI: 10.1007/978-981-10-4304-8_13] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Bradley JM, Li Z, Organ CL, Polhemus DJ, Otsuka H, Islam KN, Bhushan S, Gorodnya OM, Ruchko MV, Gillespie MN, Wilson GL, Lefer DJ. A novel mtDNA repair fusion protein attenuates maladaptive remodeling and preserves cardiac function in heart failure. Am J Physiol Heart Circ Physiol 2017; 314:H311-H321. [PMID: 29101177 DOI: 10.1152/ajpheart.00515.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Oxidative stress results in mtDNA damage and contributes to myocardial cell death. mtDNA repair enzymes are crucial for mtDNA repair and cell survival. We investigated a novel, mitochondria-targeted fusion protein (Exscien1-III) containing endonuclease III in myocardial ischemia-reperfusion injury and transverse aortic constriction (TAC)-induced heart failure. Male C57/BL6J mice (10-12 wk) were subjected to 45 min of myocardial ischemia and either 24 h or 4 wk of reperfusion. Exscien1-III (4 mg/kg ip) or vehicle was administered at the time of reperfusion. Male C57/BL6J mice were subjected to TAC, and Exscien1-III (4 mg/kg i.p) or vehicle was administered daily starting at 3 wk post-TAC and continued for 12 wk. Echocardiography was performed to assess left ventricular (LV) structure and function. Exscien1-III reduced myocardial infarct size ( P < 0.01) at 24 h of reperfusion and preserved LV ejection fraction at 4 wk postmyocardial ischemia. Exscien1-III attenuated TAC-induced LV dilation and dysfunction at 6-12 wk post-TAC ( P < 0.05). Exscien1-III reduced ( P < 0.05) cardiac hypertrophy and maladaptive remodeling after TAC. Assessment of cardiac mitochondria showed that Exscien1-III localized to mitochondria and increased mitochondrial antioxidant and reduced apoptotic markers. In conclusion, our results indicate that administration of Exscien1-III provides significant protection against myocardial ischemia and preserves myocardial structure and LV performance in the setting of heart failure. NEW & NOTEWORTHY Oxidative stress-induced mitochondrial DNA damage is a prominent feature in the pathogenesis of cardiovascular diseases. In the present study, we demonstrate the efficacy of a novel, mitochondria-targeted fusion protein that traffics endonuclease III specifically for mitochondrial DNA repair in two well-characterized murine models of cardiac injury and failure.
Collapse
Affiliation(s)
- Jessica M Bradley
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Zhen Li
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Chelsea L Organ
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - David J Polhemus
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Hiroyuki Otsuka
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Kazi N Islam
- Department of Surgery, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Shashi Bhushan
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Olena M Gorodnya
- College of Medicine, University of South Alabama , Mobile, Alabama
| | | | - Mark N Gillespie
- College of Medicine, University of South Alabama , Mobile, Alabama
| | - Glenn L Wilson
- College of Medicine, University of South Alabama , Mobile, Alabama.,Exscien Corporation , Mobile, Alabama
| | - David J Lefer
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| |
Collapse
|
30
|
Bienvenu LA, Morgan J, Reichelt ME, Delbridge LM, Young MJ. Chronic in vivo nitric oxide deficiency impairs cardiac functional recovery after ischemia in female (but not male) mice. J Mol Cell Cardiol 2017; 112:8-15. [DOI: 10.1016/j.yjmcc.2017.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/24/2017] [Accepted: 08/26/2017] [Indexed: 12/29/2022]
|
31
|
Reddy YNV, Lewis GD, Shah SJ, LeWinter M, Semigran M, Davila-Roman VG, Anstrom K, Hernandez A, Braunwald E, Redfield MM, Borlaug BA. INDIE-HFpEF (Inorganic Nitrite Delivery to Improve Exercise Capacity in Heart Failure With Preserved Ejection Fraction): Rationale and Design. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.117.003862. [PMID: 28476756 DOI: 10.1161/circheartfailure.117.003862] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/29/2017] [Indexed: 02/06/2023]
Abstract
Approximately half of patients with heart failure have preserved ejection fraction. There is no proven treatment that improves outcome. The pathophysiology of heart failure with preserved ejection fraction is complex and includes left ventricular systolic and diastolic dysfunction, pulmonary vascular disease, endothelial dysfunction, and peripheral abnormalities. Multiple lines of evidence point to impaired nitric oxide (NO)-cGMP bioavailability as playing a central role in each of these abnormalities. In contrast to traditional organic nitrate therapies, an alternative strategy to restore NO-cGMP signaling is via inorganic nitrite. Inorganic nitrite, previously considered to be an inert byproduct of NO metabolism, functions as an important in vivo reservoir for NO generation, particularly under hypoxic and acidosis conditions. As such, inorganic nitrite becomes most active at times of greater need for NO signaling, as during exercise when left ventricular filling pressures and pulmonary artery pressures increase. Herein, we present the rationale and design for the INDIE-HFpEF trial (Inorganic Nitrite Delivery to Improve Exercise Capacity in Heart Failure with Preserved Ejection Fraction), which is a multicenter, randomized, double-blind, placebo-controlled cross-over study assessing the effect of inhaled inorganic nitrite on peak exercise capacity, conducted in the National Heart, Lung, and Blood Institute-sponsored Heart Failure Clinical Research Network. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT02742129.
Collapse
Affiliation(s)
- Yogesh N V Reddy
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Gregory D Lewis
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Sanjiv J Shah
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Martin LeWinter
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Marc Semigran
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Victor G Davila-Roman
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Kevin Anstrom
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Adrian Hernandez
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Eugene Braunwald
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Margaret M Redfield
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Barry A Borlaug
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.).
| |
Collapse
|
32
|
Koch CD, Gladwin MT, Freeman BA, Lundberg JO, Weitzberg E, Morris A. Enterosalivary nitrate metabolism and the microbiome: Intersection of microbial metabolism, nitric oxide and diet in cardiac and pulmonary vascular health. Free Radic Biol Med 2017; 105:48-67. [PMID: 27989792 PMCID: PMC5401802 DOI: 10.1016/j.freeradbiomed.2016.12.015] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/18/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023]
Abstract
Recent insights into the bioactivation and signaling actions of inorganic, dietary nitrate and nitrite now suggest a critical role for the microbiome in the development of cardiac and pulmonary vascular diseases. Once thought to be the inert, end-products of endothelial-derived nitric oxide (NO) heme-oxidation, nitrate and nitrite are now considered major sources of exogenous NO that exhibit enhanced vasoactive signaling activity under conditions of hypoxia and stress. The bioavailability of nitrate and nitrite depend on the enzymatic reduction of nitrate to nitrite by a unique set of bacterial nitrate reductase enzymes possessed by specific bacterial populations in the mammalian mouth and gut. The pathogenesis of pulmonary hypertension (PH), obesity, hypertension and CVD are linked to defects in NO signaling, suggesting a role for commensal oral bacteria to shape the development of PH through the formation of nitrite, NO and other bioactive nitrogen oxides. Oral supplementation with inorganic nitrate or nitrate-containing foods exert pleiotropic, beneficial vascular effects in the setting of inflammation, endothelial dysfunction, ischemia-reperfusion injury and in pre-clinical models of PH, while traditional high-nitrate dietary patterns are associated with beneficial outcomes in hypertension, obesity and CVD. These observations highlight the potential of the microbiome in the development of novel nitrate- and nitrite-based therapeutics for PH, CVD and their risk factors.
Collapse
Affiliation(s)
- Carl D Koch
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA.
| | - Mark T Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA; Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA
| | - Bruce A Freeman
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA; Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| |
Collapse
|
33
|
Li X, Hu Y, Zhang F, Chen Y, Zhou H, Guo D, Zhao Q. Unbalanced Oxidant-Antioxidant Status: A Potential Therapeutic Target for Coronary Chronic Total Occlusion in Very Old Patients. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:4910829. [PMID: 28044093 PMCID: PMC5156810 DOI: 10.1155/2016/4910829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/06/2016] [Accepted: 11/10/2016] [Indexed: 01/21/2023]
Abstract
Unbalanced oxidant and antioxidant status played an important role in myocardial infarction. The present study was a clinical trial combined preclinically with targeted agent against cardiovascular injuries and ischemia in vivo model. We tried to confirm the association of unbalanced oxidant and antioxidant status with coronary chronic total occlusion (CTO) in 399 very old patients (80~89 years) and investigated the potential therapeutic value of purified polysaccharide from endothelium corneum gigeriae galli (PECGGp). We analyzed levels of circulating superoxide dismutase 3 (SOD3), nitric oxide (NO), endothelial nitric oxide synthase (eNOS), and malondialdehyde (MDA) in very old patients with coronary CTO. Levels of SOD3, NO, eNOS, and MDA in the cardiac tissue were measured in myocardial infarction rats. Levels of SOD3, eNOS, and NO were lowered (p < 0.001) and levels of MDA were increased (p < 0.001). PECGGp treatment increased levels of SOD3, eNOS, and NO (p < 0.01) in cardiac tissue, while decreasing levels of MDA (p < 0.01). PECGGp may suppress unbalanced oxidant and antioxidant status in infarcted myocardium by inhibiting levels of MDA and elevating NO, eNOS, and SOD3 levels. PECGGp could be considered as a potential therapeutic agent for coronary CTO in very old patients.
Collapse
Affiliation(s)
- Xia Li
- Department of Geriatrics, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, China
| | - Youdong Hu
- Department of Geriatrics, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, China
| | - Fenglin Zhang
- Department of Geriatrics, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, China
| | - Ying Chen
- Department of Geriatrics, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, China
| | - Hualan Zhou
- Department of Geriatrics, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, China
| | - Dianxuan Guo
- Department of Geriatrics, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, China
| | - Qingna Zhao
- Department of Geriatrics, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, China
| |
Collapse
|
34
|
Borlaug BA, Melenovsky V, Koepp KE. Inhaled Sodium Nitrite Improves Rest and Exercise Hemodynamics in Heart Failure With Preserved Ejection Fraction. Circ Res 2016; 119:880-6. [PMID: 27458234 DOI: 10.1161/circresaha.116.309184] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/25/2016] [Indexed: 12/14/2022]
Abstract
RATIONALE Abnormalities in nitric oxide signaling play a pivotal role in heart failure with preserved ejection fraction (HFpEF). Intravenous sodium nitrite, which is converted to nitric oxide in vivo, improves hemodynamics in HFpEF, but its use is limited by the need for parenteral administration. Nitrite can also be administered using a novel, portable micronebulizer system suitable for chronic use. OBJECTIVE Determine whether inhaled nitrite improves hemodynamics in HFpEF. METHODS AND RESULTS In a double-blind, randomized, placebo-controlled, parallel-group trial, subjects with HFpEF (n=26) underwent cardiac catheterization with simultaneous expired gas analysis at rest and during exercise before and after treatment with inhaled sodium nitrite (90 mg) or placebo. The primary end point was the pulmonary capillary wedge pressure during exercise. Before study drug administration, HFpEF subjects displayed an increase in pulmonary capillary wedge pressure with exercise from 20±6 to 34±7 mm Hg (P<0.0001). After study drug administration, exercise pulmonary capillary wedge pressure was substantially improved by nitrite as compared with placebo (baseline-adjusted mean 25±5 versus 31±6 mm Hg; analysis of covariance P=0.022). Inhaled nitrite reduced resting pulmonary capillary wedge pressure (-4±3 versus -1±2 mm Hg; P=0.002), improved pulmonary artery compliance (+1.5±1.1 versus +0.6±0.9 mL/mm Hg), and decreased mean pulmonary artery pressures at rest (-7±4 versus -3±4 mm Hg; P=0.007) and with exercise (-10±6 versus -5±6 mm Hg; P=0.05). Nitrite reduced right atrial pressures, with no effect on cardiac output or stroke volume. CONCLUSIONS Acute administration of inhaled sodium nitrite reduces biventricular filling pressures and pulmonary artery pressures at rest and during exercise in HFpEF. Further study is warranted to evaluate chronic effects of inhaled nitrite in HFpEF. CLINICAL TRIAL REGISTRATION This single center randomized clinical trial is registered at clinicaltrials.gov (NCT02262078).
Collapse
Affiliation(s)
- Barry A Borlaug
- From the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic Rochester, MN.
| | - Vojtech Melenovsky
- From the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic Rochester, MN
| | - Katlyn E Koepp
- From the Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic Rochester, MN
| |
Collapse
|
35
|
Ahluwalia A, Gladwin M, Coleman GD, Hord N, Howard G, Kim-Shapiro DB, Lajous M, Larsen FJ, Lefer DJ, McClure LA, Nolan BT, Pluta R, Schechter A, Wang CY, Ward MH, Harman JL. Dietary Nitrate and the Epidemiology of Cardiovascular Disease: Report From a National Heart, Lung, and Blood Institute Workshop. J Am Heart Assoc 2016; 5:e003402. [PMID: 27385425 PMCID: PMC5015377 DOI: 10.1161/jaha.116.003402] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Amrita Ahluwalia
- The William Harvey Research Institute, Barts & The London Medical School, Queen Mary University of London, UK
| | - Mark Gladwin
- Vascular Medicine Institute, Pittsburgh University, Pittsburgh, PA
| | | | | | | | | | - Martin Lajous
- Nacional de Salud Pública de Mexico, Mexico, Albania
| | | | - David J Lefer
- Louisiana State University Health Sciences Center, New Orleans, LA
| | - Leslie A McClure
- Dornsife School of Public Health at Drexel University, Philadelphia, PA
| | | | - Ryszard Pluta
- National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - Alan Schechter
- National Institute for Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Chia-Yih Wang
- National Center for Health Statistics, CDC, Hyattsville, MD
| | | | - Jane L Harman
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, MD
| |
Collapse
|
36
|
Donnarumma E, Ali MJ, Rushing AM, Scarborough AL, Bradley JM, Organ CL, Islam KN, Polhemus DJ, Evangelista S, Cirino G, Jenkins JS, Patel RAG, Lefer DJ, Goodchild TT. Zofenopril Protects Against Myocardial Ischemia-Reperfusion Injury by Increasing Nitric Oxide and Hydrogen Sulfide Bioavailability. J Am Heart Assoc 2016; 5:JAHA.116.003531. [PMID: 27381758 PMCID: PMC5015391 DOI: 10.1161/jaha.116.003531] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Zofenopril, a sulfhydrylated angiotensin‐converting enzyme inhibitor (ACEI), reduces mortality and morbidity in infarcted patients to a greater extent than do other ACEIs. Zofenopril is a unique ACEI that has been shown to increase hydrogen sulfide (H2S) bioavailability and nitric oxide (NO) levels via bradykinin‐dependent signaling. Both H2S and NO exert cytoprotective and antioxidant effects. We examined zofenopril effects on H2S and NO bioavailability and cardiac damage in murine and swine models of myocardial ischemia/reperfusion (I/R) injury. Methods and Results Zofenopril (10 mg/kg PO) was administered for 1, 8, and 24 hours to establish optimal dosing in mice. Myocardial and plasma H2S and NO levels were measured along with the levels of H2S and NO enzymes (cystathionine β‐synthase, cystathionine γ‐lyase, 3‐mercaptopyruvate sulfur transferase, and endothelial nitric oxide synthase). Mice received 8 hours of zofenopril or vehicle pretreatment followed by 45 minutes of ischemia and 24 hours of reperfusion. Pigs received placebo or zofenopril (30 mg/daily orally) 7 days before 75 minutes of ischemia and 48 hours of reperfusion. Zofenopril significantly augmented both plasma and myocardial H2S and NO levels in mice and plasma H2S (sulfane sulfur) in pigs. Cystathionine β‐synthase, cystathionine γ‐lyase, 3‐mercaptopyruvate sulfur transferase, and total endothelial nitric oxide synthase levels were unaltered, while phospho‐endothelial nitric oxide synthase1177 was significantly increased in mice. Pretreatment with zofenopril significantly reduced myocardial infarct size and cardiac troponin I levels after I/R injury in both mice and swine. Zofenopril also significantly preserved ischemic zone endocardial blood flow at reperfusion in pigs after I/R. Conclusions Zofenopril‐mediated cardioprotection during I/R is associated with an increase in H2S and NO signaling.
Collapse
Affiliation(s)
- Erminia Donnarumma
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Murtuza J Ali
- Department of Cardiology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Amanda M Rushing
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Amy L Scarborough
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Jessica M Bradley
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Chelsea L Organ
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Kazi N Islam
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - David J Polhemus
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | | | - Giuseppe Cirino
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | | | | | - David J Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Traci T Goodchild
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| |
Collapse
|
37
|
Lionetti V. Simultaneous exposure to nitric oxide inhibition and angiotensin II overload: is it a murine model of mitochondrial dysfunction in nonischemic heart failure? Am J Physiol Heart Circ Physiol 2016; 310:H1385-7. [DOI: 10.1152/ajpheart.00127.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Vincenzo Lionetti
- Laboratory of Medical Science, Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; and Fondazione Toscana “G. Monasterio,” Pisa, Italy
| |
Collapse
|
38
|
Ou D, Wang Q, Huang Y, Zeng D, Wei T, Ding L, Li X, Zheng Q, Jin Y. Co-culture with neonatal cardiomyocytes enhances the proliferation of iPSC-derived cardiomyocytes via FAK/JNK signaling. BMC DEVELOPMENTAL BIOLOGY 2016; 16:11. [PMID: 27141946 PMCID: PMC4855360 DOI: 10.1186/s12861-016-0112-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/26/2016] [Indexed: 01/26/2023]
Abstract
Background We previously reported that the pluripotent stem cells can differentiate into cardiomyocytes (CMs) by co-culture with neonatal CMs (NCMs) in vitro. However, the involving mechanism is not clear. Methods Mouse induced pluripotent stem cells (iPSCs) were cultured in hanging drops to form embryoid bodies (EBs) and to induce myocardial differentiation. Co-culture of EBs and NCMs was established in a transwell insert system, while EBs grown alone in the wells were used as controls. Results Co-culture with NCMs markedly increased the generation of functional CMs from iPSCs. The focal adhesion kinase (FAK) phosphorylation, and c-Jun N-terminal kinase (JNK) phosphorylation in co-culture were higher than that in EBs grown alone. Treating FAK small interfering RNA (FAK siRNA) or specific inhibitor for JNK (SP600125) to iPSCs significantly reduced the phosphorylation of JNK and the expressions of Mef2c and Bcl-2. The expressions of cTnT and MLC-2V were also decreased. Our results revealed that co-culture with NCMs significantly enhance the differentiation ability of iPSCs by increasing Mef2c and Bcl-2 expressions concomitantly with a marked augment on cell proliferation through JNK signaling pathways. Conclusions These findings indicated that co-culture of EBs with NCMs induces genes expressed in a mature pattern and stimulates the proliferation of iPSC-derived CMs (iPS-CMs) by activating FAK/JNK signaling. Electronic supplementary material The online version of this article (doi:10.1186/s12861-016-0112-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dongbo Ou
- State Key Laboratory of Military Stomatology, Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, 1st Kang-fu Road, Xi'an, 710032, Shaanxi, China.,Department of Cardiology, NO. 422 Hospital of PLA, Zhanjiang, 524005, Guangdong, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Qi Wang
- Department of Cardiology, NO. 422 Hospital of PLA, Zhanjiang, 524005, Guangdong, China
| | - Yanjin Huang
- Department of Cardiology, NO. 422 Hospital of PLA, Zhanjiang, 524005, Guangdong, China
| | - Di Zeng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Ting Wei
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Lu Ding
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Xiaoli Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Qiangsun Zheng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology, Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, 1st Kang-fu Road, Xi'an, 710032, Shaanxi, China. .,Research and Development Center for Tissue Engineering, Fourth Military Medical University, 1st Kang-fu Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
39
|
Polhemus DJ, Li Z, Pattillo CB, Gojon G, Gojon G, Giordano T, Krum H. A novel hydrogen sulfide prodrug, SG1002, promotes hydrogen sulfide and nitric oxide bioavailability in heart failure patients. Cardiovasc Ther 2016; 33:216-26. [PMID: 25930144 PMCID: PMC5034803 DOI: 10.1111/1755-5922.12128] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recent studies demonstrate robust molecular cross talk and signaling between hydrogen sulfide (H2 S) and nitric oxide (NO). Heart failure (HF) patients are deficient in both H2 S and NO, two molecules that are critical for cardiovascular homeostasis. A phase I clinical trial of a novel H2 S prodrug (SG1002) was designed to assess safety and changes in H2 S and NO bioavailability in healthy and HF subjects. Healthy subjects (n = 7) and heart failure patients (n = 8) received oral SG1002 treatment in escalating dosages of 200, 400, and 800 mg twice daily for 7 days for each dose. Safety and tolerability were assessed by physical examination, vital signs, and ECG analysis. Plasma samples were collected during a 24-h period each week for H2 S and NO analysis. BNP and glutathione levels were analyzed as markers of cardiac health and redox status. Administration of SG1002 resulted in increased H2 S levels in healthy subjects. We also observed increased H2 S levels in HF subjects following 400 mg SG1002. Nitrite, a metabolite of NO, was increased in both healthy and HF patients receiving 400 mg and 800 mg SG1002. HF subjects treated with SG1002 displayed stable drug levels over the course of the trial. SG1002 was safe and well tolerated at all doses in both healthy and HF subjects. These data suggest that SG1002 increases blood H2 S levels and circulating NO bioavailability. The finding that SG1002 attenuates increases in BNP in HF patients suggests that this novel agent warrants further study in a larger clinical study.
Collapse
Affiliation(s)
- David J Polhemus
- Cardiovascular Center of Excellence, LSU Health Sciences Center, New Orleans, LA, USA
| | - Zhen Li
- Cardiovascular Center of Excellence, LSU Health Sciences Center, New Orleans, LA, USA
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Gabriel Gojon
- Medicinal Chemistry Division, NAN Laboratorios, Monterrey, Mexico
| | - Gabriel Gojon
- Medicinal Chemistry Division, NAN Laboratorios, Monterrey, Mexico
| | - Tony Giordano
- Sulfagenix Australia Pty Ltd., Melbourne, Vic., Australia
| | - Henry Krum
- Monash University, Melbourne, Vic., Australia
| |
Collapse
|
40
|
Gee LC, Ahluwalia A. Dietary Nitrate Lowers Blood Pressure: Epidemiological, Pre-clinical Experimental and Clinical Trial Evidence. Curr Hypertens Rep 2016; 18:17. [PMID: 26815004 PMCID: PMC4729801 DOI: 10.1007/s11906-015-0623-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO), a potent vasodilator critical in maintaining vascular homeostasis, can reduce blood pressure in vivo. Loss of constitutive NO generation, for example as a result of endothelial dysfunction, occurs in many pathological conditions, including hypertension, and contributes to disease pathology. Attempts to therapeutically deliver NO via organic nitrates (e.g. glyceryl trinitrate, GTN) to reduce blood pressure in hypertensives have been largely unsuccessful. However, in recent years inorganic (or 'dietary') nitrate has been identified as a potential solution for NO delivery through its sequential chemical reduction via the enterosalivary circuit. With dietary nitrate found in abundance in vegetables this review discusses epidemiological, pre-clinical and clinical data supporting the idea that dietary nitrate could represent a cheap and effective dietary intervention capable of reducing blood pressure and thereby improving cardiovascular health.
Collapse
Affiliation(s)
- Lorna C Gee
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
41
|
Organ CL, Otsuka H, Bhushan S, Wang Z, Bradley J, Trivedi R, Polhemus DJ, Tang WHW, Wu Y, Hazen SL, Lefer DJ. Choline Diet and Its Gut Microbe-Derived Metabolite, Trimethylamine N-Oxide, Exacerbate Pressure Overload-Induced Heart Failure. Circ Heart Fail 2015; 9:e002314. [PMID: 26699388 DOI: 10.1161/circheartfailure.115.002314] [Citation(s) in RCA: 261] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 11/12/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Trimethylamine N-oxide (TMAO), a gut microbe-dependent metabolite of dietary choline and other trimethylamine-containing nutrients, is both elevated in the circulation of patients having heart failure and heralds worse overall prognosis. In animal studies, dietary choline or TMAO significantly accelerates atherosclerotic lesion development in ApoE-deficient mice, and reduction in TMAO levels inhibits atherosclerosis development in the low-density lipoprotein receptor knockout mouse. METHODS AND RESULTS C57BL6/J mice were fed either a control diet, a diet containing choline (1.2%) or a diet containing TMAO (0.12%) starting 3 weeks before surgical transverse aortic constriction. Mice were studied for 12 weeks after transverse aortic constriction. Cardiac function and left ventricular structure were monitored at 3-week intervals using echocardiography. Twelve weeks post transverse aortic constriction, myocardial tissues were collected to evaluate cardiac and vascular fibrosis, and blood samples were evaluated for cardiac brain natriuretic peptide, choline, and TMAO levels. Pulmonary edema, cardiac enlargement, and left ventricular ejection fraction were significantly (P<0.05, each) worse in mice fed either TMAO- or choline-supplemented diets when compared with the control diet. In addition, myocardial fibrosis was also significantly greater (P<0.01, each) in the TMAO and choline groups relative to controls. CONCLUSIONS Heart failure severity is significantly enhanced in mice fed diets supplemented with either choline or the gut microbe-dependent metabolite TMAO. The present results suggest that additional studies are warranted examining whether gut microbiota and the dietary choline → TMAO pathway contribute to increased heart failure susceptibility.
Collapse
Affiliation(s)
- Chelsea L Organ
- From the Department of Pharmacology, Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans (C.L.O., H.O., S.B., J.B., R.T., D.J.P., D.J.L.); Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, OH (Z.W., W.H.W.T., S.L.H.); and Department of Mathematics, Cleveland State University, OH (Y.W.)
| | - Hiroyuki Otsuka
- From the Department of Pharmacology, Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans (C.L.O., H.O., S.B., J.B., R.T., D.J.P., D.J.L.); Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, OH (Z.W., W.H.W.T., S.L.H.); and Department of Mathematics, Cleveland State University, OH (Y.W.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Halls ML, Bathgate RAD, Sutton SW, Dschietzig TB, Summers RJ. International Union of Basic and Clinical Pharmacology. XCV. Recent advances in the understanding of the pharmacology and biological roles of relaxin family peptide receptors 1-4, the receptors for relaxin family peptides. Pharmacol Rev 2015; 67:389-440. [PMID: 25761609 DOI: 10.1124/pr.114.009472] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Relaxin, insulin-like peptide 3 (INSL3), relaxin-3, and INSL5 are the cognate ligands for the relaxin family peptide (RXFP) receptors 1-4, respectively. RXFP1 activates pleiotropic signaling pathways including the signalosome protein complex that facilitates high-sensitivity signaling; coupling to Gα(s), Gα(i), and Gα(o) proteins; interaction with glucocorticoid receptors; and the formation of hetero-oligomers with distinctive pharmacological properties. In addition to relaxin-related ligands, RXFP1 is activated by Clq-tumor necrosis factor-related protein 8 and by small-molecular-weight agonists, such as ML290 [2-isopropoxy-N-(2-(3-(trifluoromethylsulfonyl)phenylcarbamoyl)phenyl)benzamide], that act allosterically. RXFP2 activates only the Gα(s)- and Gα(o)-coupled pathways. Relaxin-3 is primarily a neuropeptide, and its cognate receptor RXFP3 is a target for the treatment of depression, anxiety, and autism. A variety of peptide agonists, antagonists, biased agonists, and an allosteric modulator target RXFP3. Both RXFP3 and the related RXFP4 couple to Gα(i)/Gα(o) proteins. INSL5 has the properties of an incretin; it is secreted from the gut and is orexigenic. The expression of RXFP4 in gut, adipose tissue, and β-islets together with compromised glucose tolerance in INSL5 or RXFP4 knockout mice suggests a metabolic role. This review focuses on the many advances in our understanding of RXFP receptors in the last 5 years, their signal transduction mechanisms, the development of novel compounds that target RXFP1-4, the challenges facing the field, and current prospects for new therapeutics.
Collapse
Affiliation(s)
- Michelle L Halls
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (M.L.H., R.J.S.); Neuropeptides Division, Florey Institute of Neuroscience and Mental Health and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia (R.A.D.B.); Neuroscience Drug Discovery, Janssen Research & Development, LLC, San Diego, California (S.W.S.); Immundiagnostik AG, Bensheim, Germany (T.B.D.); and Charité-University Medicine Berlin, Campus Mitte, Medical Clinic for Cardiology and Angiology, Berlin, Germany (T.B.D.)
| | - Ross A D Bathgate
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (M.L.H., R.J.S.); Neuropeptides Division, Florey Institute of Neuroscience and Mental Health and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia (R.A.D.B.); Neuroscience Drug Discovery, Janssen Research & Development, LLC, San Diego, California (S.W.S.); Immundiagnostik AG, Bensheim, Germany (T.B.D.); and Charité-University Medicine Berlin, Campus Mitte, Medical Clinic for Cardiology and Angiology, Berlin, Germany (T.B.D.)
| | - Steve W Sutton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (M.L.H., R.J.S.); Neuropeptides Division, Florey Institute of Neuroscience and Mental Health and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia (R.A.D.B.); Neuroscience Drug Discovery, Janssen Research & Development, LLC, San Diego, California (S.W.S.); Immundiagnostik AG, Bensheim, Germany (T.B.D.); and Charité-University Medicine Berlin, Campus Mitte, Medical Clinic for Cardiology and Angiology, Berlin, Germany (T.B.D.)
| | - Thomas B Dschietzig
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (M.L.H., R.J.S.); Neuropeptides Division, Florey Institute of Neuroscience and Mental Health and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia (R.A.D.B.); Neuroscience Drug Discovery, Janssen Research & Development, LLC, San Diego, California (S.W.S.); Immundiagnostik AG, Bensheim, Germany (T.B.D.); and Charité-University Medicine Berlin, Campus Mitte, Medical Clinic for Cardiology and Angiology, Berlin, Germany (T.B.D.)
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (M.L.H., R.J.S.); Neuropeptides Division, Florey Institute of Neuroscience and Mental Health and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia (R.A.D.B.); Neuroscience Drug Discovery, Janssen Research & Development, LLC, San Diego, California (S.W.S.); Immundiagnostik AG, Bensheim, Germany (T.B.D.); and Charité-University Medicine Berlin, Campus Mitte, Medical Clinic for Cardiology and Angiology, Berlin, Germany (T.B.D.)
| |
Collapse
|
43
|
Kobayashi J, Ohtake K, Uchida H. NO-Rich Diet for Lifestyle-Related Diseases. Nutrients 2015; 7:4911-37. [PMID: 26091235 PMCID: PMC4488823 DOI: 10.3390/nu7064911] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/08/2015] [Accepted: 06/09/2015] [Indexed: 02/07/2023] Open
Abstract
Decreased nitric oxide (NO) availability due to obesity and endothelial dysfunction might be causally related to the development of lifestyle-related diseases such as insulin resistance, ischemic heart disease, and hypertension. In such situations, instead of impaired NO synthase (NOS)-dependent NO generation, the entero-salivary nitrate-nitrite-NO pathway might serve as a backup system for NO generation by transmitting NO activities in the various molecular forms including NO and protein S-nitrosothiols. Recently accumulated evidence has demonstrated that dietary intake of fruits and vegetables rich in nitrate/nitrite is an inexpensive and easily-practicable way to prevent insulin resistance and vascular endothelial dysfunction by increasing the NO availability; a NO-rich diet may also prevent other lifestyle-related diseases, including osteoporosis, chronic obstructive pulmonary disease (COPD), and cancer. This review provides an overview of our current knowledge of NO generation through the entero-salivary pathway and discusses its safety and preventive effects on lifestyle-related diseases.
Collapse
Affiliation(s)
- Jun Kobayashi
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Saitama 350-0295, Japan.
| | - Kazuo Ohtake
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Saitama 350-0295, Japan.
| | - Hiroyuki Uchida
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Saitama 350-0295, Japan.
| |
Collapse
|
44
|
Bradley JM, Islam KN, Polhemus DJ, Donnarumma E, Brewster LP, Tao YX, Goodchild TT, Lefer DJ. Sustained release nitrite therapy results in myocardial protection in a porcine model of metabolic syndrome with peripheral vascular disease. Am J Physiol Heart Circ Physiol 2015; 309:H305-17. [PMID: 25957218 DOI: 10.1152/ajpheart.00163.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/06/2015] [Indexed: 11/22/2022]
Abstract
Metabolic syndrome (MetS) reduces endothelial nitric oxide (NO) bioavailability and exacerbates vascular dysfunction in patients with preexisting vascular diseases. Nitrite, a storage form of NO, can mediate vascular function during pathological conditions when endogenous NO is reduced. The aims of the present study were to characterize the effects of severe MetS and obesity on dyslipidemia, myocardial oxidative stress, and endothelial NO synthase (eNOS) regulation in the obese Ossabaw swine (OS) model and to examine the effects of a novel, sustained-release formulation of sodium nitrite (SR-nitrite) on coronary vascular reactivity and myocardial redox status in obese OS subjected to critical limb ischemia (CLI). After 6 mo of an atherogenic diet, obese OS displayed a MetS phenotype. Obese OS had decreased eNOS functionality and NO bioavailability. In addition, obese OS exhibited increased oxidative stress and a significant reduction in antioxidant enzymes. The efficacy of SR-nitrite therapy was examined in obese OS subjected to CLI. After 3 wk of treatment, SR-nitrite (80 mg · kg(-1) · day(-1) bid po) increased myocardial nitrite levels and eNOS function. Treatment with SR-nitrite reduced myocardial oxidative stress while increasing myocardial antioxidant capacity. Ex vivo assessment of vascular reactivity of left anterior descending coronary artery segments demonstrated marked improvement in vasoreactivity to sodium nitroprusside but not to substance P and bradykinin in SR-nitrite-treated animals compared with placebo-treated animals. In conclusion, in a clinically relevant, large-animal model of MetS and CLI, treatment with SR-nitrite enhanced myocardial NO bioavailability, attenuated oxidative stress, and improved ex vivo coronary artery vasorelaxation.
Collapse
Affiliation(s)
- Jessica M Bradley
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Kazi N Islam
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - David J Polhemus
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Erminia Donnarumma
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Luke P Brewster
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia; Surgery and Research Services, Atlanta Veterans Affairs Medical Center, Decatur, Georgia; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia; and
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, Alabama
| | - Traci T Goodchild
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - David J Lefer
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana;
| |
Collapse
|
45
|
Polhemus DJ, Bradley JM, Islam KN, Brewster LP, Calvert JW, Tao YX, Chang CC, Pipinos II, Goodchild TT, Lefer DJ. Therapeutic potential of sustained-release sodium nitrite for critical limb ischemia in the setting of metabolic syndrome. Am J Physiol Heart Circ Physiol 2015; 309:H82-92. [PMID: 25910804 DOI: 10.1152/ajpheart.00115.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/13/2015] [Indexed: 01/12/2023]
Abstract
Nitrite is a storage reservoir of nitric oxide that is readily reduced to nitric oxide under pathological conditions. Previous studies have demonstrated that nitrite levels are significantly reduced in cardiovascular disease states, including peripheral vascular disease. We investigated the cytoprotective and proangiogenic actions of a novel, sustained-release formulation of nitrite (SR-nitrite) in a clinically relevant in vivo swine model of critical limb ischemia (CLI) involving central obesity and metabolic syndrome. CLI was induced in obese Ossabaw swine (n = 18) by unilateral external iliac artery deployment of a full cross-sectional vessel occlusion device positioned within an endovascular expanded polytetrafluoroethylene-lined nitinol stent-graft. At post-CLI day 14, pigs were randomized to placebo (n = 9) or SR-nitrite (80 mg, n = 9) twice daily by mouth for 21 days. SR-nitrite therapy increased nitrite, nitrate, and S-nitrosothiol in plasma and ischemic skeletal muscle. Oxidative stress was reduced in ischemic limb tissue of SR-nitrite- compared with placebo-treated pigs. Ischemic limb tissue levels of proangiogenic growth factors were increased following SR-nitrite therapy compared with placebo. Despite the increases in cytoprotective and angiogenic signals with SR-nitrite therapy, new arterial vessel formation and enhancement of blood flow to the ischemic limb were not different from placebo. Our data clearly demonstrate cytoprotective and proangiogenic signaling in ischemic tissues following SR-nitrite therapy in a very severe model of CLI. Further studies evaluating longer-duration nitrite therapy and/or additional nitrite dosing strategies are warranted to more fully evaluate the therapeutic potential of nitrite therapy in peripheral vascular disease.
Collapse
Affiliation(s)
- David J Polhemus
- Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Jessica M Bradley
- Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Kazi N Islam
- Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Luke P Brewster
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia; Surgery and Research Services, Atlanta Veterans Affairs Medical Center, Decatur, Georgia; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - John W Calvert
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, Alabama
| | | | - Iraklis I Pipinos
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Traci T Goodchild
- Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Sciences Center, New Orleans, Louisiana
| | - David J Lefer
- Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Sciences Center, New Orleans, Louisiana;
| |
Collapse
|
46
|
Ormerod JOM, Arif S, Mukadam M, Evans JDW, Beadle R, Fernandez BO, Bonser RS, Feelisch M, Madhani M, Frenneaux MP. Short-term intravenous sodium nitrite infusion improves cardiac and pulmonary hemodynamics in heart failure patients. Circ Heart Fail 2015; 8:565-71. [PMID: 25838311 PMCID: PMC4435579 DOI: 10.1161/circheartfailure.114.001716] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 03/25/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Nitrite exhibits hypoxia-dependent vasodilator properties, selectively dilating capacitance vessels in healthy subjects. Unlike organic nitrates, it seems not to be subject to the development of tolerance. Currently, therapeutic options for decompensated heart failure (HF) are limited. We hypothesized that by preferentially dilating systemic capacitance and pulmonary resistance vessels although only marginally dilating resistance vessels, sodium nitrite (NaNO2) infusion would increase cardiac output but reduce systemic arterial blood pressure only modestly. We therefore undertook a first-in-human HF proof of concept/safety study, evaluating the hemodynamic effects of short-term NaNO2 infusion. METHODS AND RESULTS Twenty-five patients with severe chronic HF were recruited. Eight received short-term (5 minutes) intravenous NaNO2 at 10 μg/kg/min and 17 received 50 μg/kg/min with measurement of cardiac hemodynamics. During infusion of 50 μg/kg/min, left ventricular stroke volume increased (from 43.22±21.5 to 51.84±23.6 mL; P=0.003), with marked falls in pulmonary vascular resistance (by 29%; P=0.03) and right atrial pressure (by 40%; P=0.007), but with only modest falls in mean arterial blood pressure (by 4 mm Hg; P=0.004). The increase in stroke volume correlated with the increase in estimated trans-septal gradient (=pulmonary capillary wedge pressure-right atrial pressure; r=0.67; P=0.003), suggesting relief of diastolic ventricular interaction as a contributory mechanism. Directionally similar effects were observed for the above hemodynamic parameters with 10 μg/kg/min; this was significant only for stroke volume, not for other parameters. CONCLUSIONS This first-in-human HF efficacy/safety study demonstrates an attractive profile during short-term systemic NaNO2 infusion that may be beneficial in decompensated HF and warrants further evaluation with longer infusion regimens.
Collapse
Affiliation(s)
- Julian O M Ormerod
- From the Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (J.O.M.O., S.A., J.D.W.E., R.B., M.M.); Department of Cardiothoracic Surgery, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, United Kingdom (M.M., R.S.B.); Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom (B.O.F., M.F.); and Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom (M.P.F.)
| | - Sayqa Arif
- From the Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (J.O.M.O., S.A., J.D.W.E., R.B., M.M.); Department of Cardiothoracic Surgery, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, United Kingdom (M.M., R.S.B.); Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom (B.O.F., M.F.); and Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom (M.P.F.)
| | - Majid Mukadam
- From the Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (J.O.M.O., S.A., J.D.W.E., R.B., M.M.); Department of Cardiothoracic Surgery, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, United Kingdom (M.M., R.S.B.); Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom (B.O.F., M.F.); and Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom (M.P.F.)
| | - Jonathan D W Evans
- From the Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (J.O.M.O., S.A., J.D.W.E., R.B., M.M.); Department of Cardiothoracic Surgery, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, United Kingdom (M.M., R.S.B.); Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom (B.O.F., M.F.); and Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom (M.P.F.)
| | - Roger Beadle
- From the Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (J.O.M.O., S.A., J.D.W.E., R.B., M.M.); Department of Cardiothoracic Surgery, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, United Kingdom (M.M., R.S.B.); Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom (B.O.F., M.F.); and Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom (M.P.F.)
| | - Bernadette O Fernandez
- From the Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (J.O.M.O., S.A., J.D.W.E., R.B., M.M.); Department of Cardiothoracic Surgery, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, United Kingdom (M.M., R.S.B.); Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom (B.O.F., M.F.); and Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom (M.P.F.)
| | - Robert S Bonser
- From the Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (J.O.M.O., S.A., J.D.W.E., R.B., M.M.); Department of Cardiothoracic Surgery, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, United Kingdom (M.M., R.S.B.); Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom (B.O.F., M.F.); and Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom (M.P.F.)
| | - Martin Feelisch
- From the Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (J.O.M.O., S.A., J.D.W.E., R.B., M.M.); Department of Cardiothoracic Surgery, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, United Kingdom (M.M., R.S.B.); Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom (B.O.F., M.F.); and Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom (M.P.F.)
| | - Melanie Madhani
- From the Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (J.O.M.O., S.A., J.D.W.E., R.B., M.M.); Department of Cardiothoracic Surgery, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, United Kingdom (M.M., R.S.B.); Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom (B.O.F., M.F.); and Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom (M.P.F.)
| | - Michael P Frenneaux
- From the Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (J.O.M.O., S.A., J.D.W.E., R.B., M.M.); Department of Cardiothoracic Surgery, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, United Kingdom (M.M., R.S.B.); Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom (B.O.F., M.F.); and Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom (M.P.F.).
| |
Collapse
|
47
|
Farah C, Reboul C. NO Better Way to Protect the Heart during Ischemia-Reperfusion: To be in the Right Place at the Right Time. Front Pediatr 2015; 3:6. [PMID: 25705614 PMCID: PMC4319379 DOI: 10.3389/fped.2015.00006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/26/2015] [Indexed: 11/24/2022] Open
Affiliation(s)
- Charlotte Farah
- EA4278, LaPEC, Université d'Avignon , Avignon , France ; UMR-CNRS 9214, INSERM U1046, Université de Montpellier , Montpellier , France
| | - Cyril Reboul
- EA4278, LaPEC, Université d'Avignon , Avignon , France
| |
Collapse
|
48
|
Buglioni A, Burnett JC. Pathophysiology and the cardiorenal connection in heart failure. Circulating hormones: biomarkers or mediators. Clin Chim Acta 2014; 443:3-8. [PMID: 25445413 DOI: 10.1016/j.cca.2014.10.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/17/2014] [Accepted: 10/19/2014] [Indexed: 12/11/2022]
Abstract
Heart failure (HF) is a syndrome characterized by a complex pathophysiology which involves multiple organ systems, with the kidney playing a major role. HF can present with reduced ejection fraction (EF), HFrEF, or with preserved EF (HFpEF). The interplay between diverse organ systems contributing to HF is mediated by the activation of counteracting neurohormonal pathways focused to re-establishing hemodynamic homeostasis. During early stages of HF, these biochemical signals, consisting mostly of hormones and neurotransmitters secreted by a variety of cell types, are compensatory and the patient is asymptomatic. However, with disease progression, the attempt to reverse or delay cardiac dysfunction is deleterious, leading to multi-organ congestion, fibrosis and decompensation and finally symptomatic HF. In conclusion, these neurohormonal pathways mediate the evolution of HF and have become a way to monitor HF. Specifically, these mediators have become important in the diagnosis and prognosis of this highly fatal cardiovascular disease. Finally, while these multiple neurohumoral factors serve as important HF biomarkers, they can also be targeted for more effective and curative HF treatments.
Collapse
Affiliation(s)
- Alessia Buglioni
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Department of Medicine and Health Sciences Research, College of Medicine, Mayo Clinic, Rochester, MN, USA.
| | - John C Burnett
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Department of Medicine and Health Sciences Research, College of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
49
|
Polhemus DJ, Calvert JW, Butler J, Lefer DJ. The cardioprotective actions of hydrogen sulfide in acute myocardial infarction and heart failure. SCIENTIFICA 2014; 2014:768607. [PMID: 25045576 PMCID: PMC4090472 DOI: 10.1155/2014/768607] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/30/2014] [Accepted: 05/16/2014] [Indexed: 06/03/2023]
Abstract
It has now become universally accepted that hydrogen sulfide (H2S), previously considered only as a lethal toxin, has robust cytoprotective actions in multiple organ systems. The diverse signaling profile of H2S impacts multiple pathways to exert cytoprotective actions in a number of pathological states. This paper will review the recently described cardioprotective actions of hydrogen sulfide in both myocardial ischemia/reperfusion injury and congestive heart failure.
Collapse
Affiliation(s)
- David J. Polhemus
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | - John W. Calvert
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Javed Butler
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - David J. Lefer
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, LSU Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|