1
|
Bracamonte JH, Watkins L, Pat B, Dell'Italia LJ, Saucerman JJ, Holmes JW. Contributions of mechanical loading and hormonal changes to eccentric hypertrophy during volume overload: A Bayesian analysis using logic-based network models. PLoS Comput Biol 2025; 21:e1012390. [PMID: 40238825 DOI: 10.1371/journal.pcbi.1012390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/23/2025] [Indexed: 04/18/2025] Open
Abstract
Primary mitral regurgitation (MR) is a pathology that alters mechanical loading on the left ventricle, triggers an array of compensatory neurohormonal responses, and induces a distinctive ventricular remodeling response known as eccentric hypertrophy. Drug therapies may alleviate symptoms, but only mitral valve repair or replacement can provide significant recovery of cardiac function and dimensions. Questions remain about the optimal timing of surgery, with 20% of patients developing systolic dysfunction post-operatively despite being treated according to the current guidelines. Thus, better understanding of the hypertrophic process in the setting of ventricular volume overload (VO) is needed to improve and better personalize the management of MR. To address this knowledge gap, we employ a Bayesian approach to combine data from 70 studies on experimental volume overload in dogs and rats and use it to calibrate a logic-based network model of hypertrophic signaling in myocytes. The calibrated model predicts that growth in experimental VO is mostly driven by the neurohormonal response, with an initial increase in myocardial tissue stretch being compensated by subsequent remodeling fairly early in the time course of VO. This observation contrasts with a common perception that volume-overload hypertrophy is driven primarily by increased myocyte strain. The model reproduces many aspects of 43 studies not used in its calibration, including infusion of individual hypertrophic agonists alone or in combination with various drugs commonly employed to treat heart failure, as well as administration of some of those drugs in the setting of experimental volume overload. We believe this represents a promising approach to using the known structure of an intracellular signaling network to integrate information from multiple studies into quantitative predictions of the range of expected responses to potential interventions in the complex setting of cardiac hypertrophy driven by a combination of hormonal and mechanical factors.
Collapse
Affiliation(s)
- Johane H Bracamonte
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Lionel Watkins
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Betty Pat
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Louis J Dell'Italia
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiothoracic Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
2
|
Bader Eddin L, Nagoor Meeran MF, Kumar Jha N, Goyal SN, Ojha S. Isoproterenol mechanisms in inducing myocardial fibrosis and its application as an experimental model for the evaluation of therapeutic potential of phytochemicals and pharmaceuticals. Animal Model Exp Med 2025; 8:67-91. [PMID: 39690876 PMCID: PMC11798751 DOI: 10.1002/ame2.12496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 08/14/2024] [Indexed: 12/19/2024] Open
Abstract
Cardiac injury initiates repair mechanisms and results in cardiac remodeling and fibrosis, which appears to be a leading cause of cardiovascular diseases. Cardiac fibrosis is characterized by the accumulation of extracellular matrix proteins, mainly collagen in the cardiac interstitium. Many experimental studies have demonstrated that fibrotic injury in the heart is reversible; therefore, it is vital to understand different molecular mechanisms that are involved in the initiation, progression, and resolution of cardiac fibrosis to enable the development of antifibrotic agents. Of the many experimental models, one of the recent models that has gained renewed interest is isoproterenol (ISP)-induced cardiac fibrosis. ISP is a synthetic catecholamine, sympathomimetic, and nonselective β-adrenergic receptor agonist. The overstimulated and sustained activation of β-adrenergic receptors has been reported to induce biochemical and physiological alterations and ultimately result in cardiac remodeling. ISP has been used for decades to induce acute myocardial infarction. However, the use of low doses and chronic administration of ISP have been shown to induce cardiac fibrosis; this practice has increased in recent years. Intraperitoneal or subcutaneous ISP has been widely used in preclinical studies to induce cardiac remodeling manifested by fibrosis and hypertrophy. The induced oxidative stress with subsequent perturbations in cellular signaling cascades through triggering the release of free radicals is considered the initiating mechanism of myocardial fibrosis. ISP is consistently used to induce fibrosis in laboratory animals and in cardiomyocytes isolated from animals. In recent years, numerous phytochemicals and synthetic molecules have been evaluated in ISP-induced cardiac fibrosis. The present review exclusively provides a comprehensive summary of the pathological biochemical, histological, and molecular mechanisms of ISP in inducing cardiac fibrosis and hypertrophy. It also summarizes the application of this experimental model in the therapeutic evaluation of natural as well as synthetic compounds to demonstrate their potential in mitigating myocardial fibrosis and hypertrophy.
Collapse
Affiliation(s)
- Lujain Bader Eddin
- Department of Pharmacology and Therapeutics, College of Medicine and Health SciencesUAE UniversityAl AinUnited Arab Emirates
| | - Mohamed Fizur Nagoor Meeran
- Department of Pharmacology and Therapeutics, College of Medicine and Health SciencesUAE UniversityAl AinUnited Arab Emirates
| | - Niraj Kumar Jha
- School of Bioengineering & BiosciencesLovely Professional UniversityPhagwaraIndia
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha UniversityChennaiIndia
| | - Samer N. Goyal
- Shri Vile Parle Kelvani Mandal's Institute of PharmacyDhuleMaharashtraIndia
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health SciencesUAE UniversityAl AinUnited Arab Emirates
- Zayed Bin Sultan Center for Health SciencesUnited Arab Emirates UniversityAl AinUnited Arab Emirates
| |
Collapse
|
3
|
Erdogan BR, Yesilyurt-Dirican ZE, Karaomerlioglu I, Muderrisoglu AE, Sevim K, Michel MC, Arioglu-Inan E. Sacubitril/Valsartan Combination Partially Improves Cardiac Systolic, but Not Diastolic, Function through β-AR Responsiveness in a Rat Model of Type 2 Diabetes. Int J Mol Sci 2024; 25:10617. [PMID: 39408945 PMCID: PMC11476658 DOI: 10.3390/ijms251910617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/22/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Cardiovascular complications are the major cause of diabetes mellitus-related morbidity and mortality. Increased renin-angiotensin-aldosterone system activity and decreased β-adrenergic receptor (β-AR) responsiveness contribute to diabetic cardiac dysfunction. We evaluated the effect of sacubitril/valsartan (neprilysin inhibitor plus angiotensin receptor antagonist combination) and valsartan treatments on the diabetic cardiac function through β-AR responsiveness and on protein expression of diastolic components. Six-week-old male Sprague Dawley rats were divided into control, diabetic, sacubitril/valsartan (68 mg/kg)-, and valsartan-treated (31 mg/kg) diabetic groups. Diabetes was induced by a high-fat diet plus low-dose streptozotocin (30 mg/kg, intraperitoneal). After 10 weeks of diabetes, rats were treated for 4 weeks. Systolic/diastolic function was assessed by in vivo echocardiography and pressure-volume loop analysis. β-AR-mediated responsiveness was assessed by in vitro papillary muscle and Langendorff heart experiments. Protein expression of sarcoplasmic reticulum calcium ATPase2a, phospholamban, and phosphorylated phospholamban was determined by Western blot. Sacubitril/valsartan improved ejection fraction and fractional shortening to a similar extent as valsartan alone. None of the treatments affected in vivo diastolic parameters or the expression of related proteins. β1-/β2-AR-mediated responsiveness was partially restored in treated animals. β3-AR-mediated cardiac relaxation (an indicator of diastolic function) responses were comparable among groups. The beneficial effect of sacubitril/valsartan on systolic function may be attributed to improved β1-/β2-AR responsiveness.
Collapse
Affiliation(s)
- Betul R. Erdogan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara 06560, Türkiye; (B.R.E.); (Z.E.Y.-D.); (I.K.); (A.E.M.)
| | - Zeynep E. Yesilyurt-Dirican
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara 06560, Türkiye; (B.R.E.); (Z.E.Y.-D.); (I.K.); (A.E.M.)
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Ankara 06330, Türkiye
| | - Irem Karaomerlioglu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara 06560, Türkiye; (B.R.E.); (Z.E.Y.-D.); (I.K.); (A.E.M.)
| | - Ayhanim Elif Muderrisoglu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara 06560, Türkiye; (B.R.E.); (Z.E.Y.-D.); (I.K.); (A.E.M.)
- Department of Medical Pharmacology, Istanbul Medipol University, Istanbul 34815, Türkiye
| | - Kadir Sevim
- Department of Internal Medicine, Faculty of Veterinary Medicine, Ankara University, Ankara 06110, Türkiye;
| | - Martin C. Michel
- Department of Pharmacology, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara 06560, Türkiye; (B.R.E.); (Z.E.Y.-D.); (I.K.); (A.E.M.)
| |
Collapse
|
4
|
Mima A, Gotoda H, Lee S, Lee R, Murakami A, Akai R, Kidooka S, Matsumoto K, Saito Y, Hishida S, Nakamoto T, Kido S, Hamada T. Effect of Sacubitril/Valsartan on Patients Having Heart Failure With Preserved Left Ventricular Ejection Fraction Undergoing Hemodialysis: A Long-term Observational Study. In Vivo 2024; 38:1266-1270. [PMID: 38688596 PMCID: PMC11059904 DOI: 10.21873/invivo.13564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/25/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND/AIM Sacubitril/valsartan (SV), a novel pharmacological class of angiotensin receptor neprilysin inhibitors, is effective in treating heart failure (HF) by inhibiting the degradation of natriuretic peptides and the renin-angiotensin-aldosterone system. However, no studies have observed the long-term effects of SV on patients with HF and preserved left ventricular ejection fraction (LVEF) undergoing hemodialysis (HD) over a long period. PATIENTS AND METHODS This single-center retrospective study of 21 months duration involved consecutive patients with HF and preserved LVEF undergoing HD, who received 50-200 mg/day. All patients were followed up regularly, and clinical, biochemical, and echocardiographic parameters were recorded at baseline and during follow-up. The efficacy and safety of SV were also analyzed. RESULTS This longitudinal study included nine patients, with a median age of 76 years. The median HD duration was 7 years. At baseline, the mean brain natriuretic peptide (BNP) was 133±73.6 pg/ml and that of LVEF was 66%±9%. After SV therapy, the systolic blood pressure, diastolic blood pressure, and heart rate decreased, albeit without statistical significance. BNP levels, LVEF, left atrial anteroposterior dimension, and left ventricular mass index did not change, compared to baseline values. No adverse effects were observed in any of the patients. CONCLUSION SV tended to decrease blood pressure and heart rate in patients with HF and preserved LVEF undergoing HD but did not alter cardiac function assessments, such as BNP or echocardiography.
Collapse
Affiliation(s)
- Akira Mima
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan;
| | - Hidemasa Gotoda
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Shinji Lee
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Rina Lee
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Ami Murakami
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Ryosuke Akai
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Sayumi Kidooka
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Keishi Matsumoto
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Yuta Saito
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Shinji Hishida
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takahiro Nakamoto
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Suguru Kido
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | | |
Collapse
|
5
|
Xiao Y, Zhou ZY, Sun JC, Xing W, Yan J, Xu WJ, Lu YS, Liu T, Jin Y. Protective effect of novel angiotensin receptor neprilysin inhibitor S086 on target organ injury in spontaneously hypertensive rats. Biomed Pharmacother 2024; 170:115968. [PMID: 38039752 DOI: 10.1016/j.biopha.2023.115968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Hypertension is a clinical syndrome characterized by elevated systemic arterial blood pressure associated with injury to the heart, kidney, brain, and other organs. Angiotensin receptor neprilysin inhibitors (ARNi), including angiotensin receptor blockers (ARBs) and neprilysin inhibitors (NEPi), have been shown to be safe and effective at reducing blood pressure and alleviating development of target organ injury. This study was used to develop S086 as a novel ARNi and conducted preclinical studies in animal models to evaluate the protective effects of S086 on target organs. METHODS This study used a 14-month-old spontaneously hypertensive rat (SHR) model to evaluate the protective effects of S086 on the cardiovascular system and organs such as heart and kidney by blood pressure monitoring, urine and blood examination, pathological examination, and immunological index detection. RESULTS After administering S086 orally to the SHR, their blood pressure and levels of renal injury indicators such as serum creatinine and urinary microalbumin were reduced, and myocardial cell necrosis and cardiac fibrosis of the heart were significantly improved. In addition, there were also significantly improvements in the histological lesions of blood vessels and the kidneys. CONCLUSIONS The findings showed that S086 effectively reduced the blood pressure of SHR and had effects on alleviating development of heart, blood vessels and kidney.
Collapse
Affiliation(s)
- Ying Xiao
- Shenzhen Salubris Pharmaceutical Co., Ltd., Shenzhen, Guangdong, China
| | - Zheng-Yang Zhou
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi 712100, China
| | - Jing-Chao Sun
- Shenzhen Salubris Pharmaceutical Co., Ltd., Shenzhen, Guangdong, China.
| | - Wei Xing
- Shenzhen Salubris Pharmaceutical Co., Ltd., Shenzhen, Guangdong, China
| | - Jie Yan
- Shenzhen Salubris Pharmaceutical Co., Ltd., Shenzhen, Guangdong, China
| | - Wen-Jie Xu
- Shenzhen Salubris Pharmaceutical Co., Ltd., Shenzhen, Guangdong, China
| | - Yin-Suo Lu
- Shenzhen Salubris Pharmaceutical Co., Ltd., Shenzhen, Guangdong, China
| | - Tao Liu
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi 712100, China.
| | - Yi Jin
- Shenzhen Institute for Drug Control (Shenzhen Testing Center of Medical Devices), Shenzhen, Guangdong, China.
| |
Collapse
|
6
|
Confalonieri F, Lumi X, Petrovski G. Spontaneous Epiretinal Membrane Resolution and Angiotensin Receptor Blockers: Case Observation, Literature Review and Perspectives. Biomedicines 2023; 11:1976. [PMID: 37509613 PMCID: PMC10377102 DOI: 10.3390/biomedicines11071976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/05/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
INTRODUCTION Epiretinal membrane (ERM) is a relatively common condition affecting the macula. When symptoms become apparent and compromise a patient's quality of vision, the only therapeutic approach available today is surgery with a vitrectomy and peeling of the ERM. Angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACE-Is) reduce the effect of angiotensin II, limit the amount of fibrosis, and demonstrate consequences on fibrinogenesis in the human body. Case Description and Materials and Methods: A rare case of spontaneous ERM resolution with concomitant administration of ARB is reported. The patient was set on ARB treatment for migraines and arterial hypertension, and a posterior vitreous detachment was already present at the first diagnosis of ERM. The scientific literature addressing the systemic relationship between ARB, ACE-Is, and fibrosis in the past 25 years was searched in the PubMed, Medline, and EMBASE databases. RESULTS In total, 38 and 16 original articles have been selected for ARBs and ACE-Is, respectively, in regard to fibrosis modulation. CONCLUSION ARBs and ACE-Is might have antifibrotic activity on ERM formation and resolution. Further clinical studies are necessary to explore this phenomenon.
Collapse
Affiliation(s)
- Filippo Confalonieri
- Department of Ophthalmology, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, University of Oslo, Kirkeveien 166, 0450 Oslo, Norway
| | - Xhevat Lumi
- Department of Ophthalmology, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway
- Eye Hospital, University Medical Centre Ljubljana, Zaloška Cesta 2, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Goran Petrovski
- Department of Ophthalmology, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, University of Oslo, Kirkeveien 166, 0450 Oslo, Norway
- Department of Ophthalmology, University of Split School of Medicine and University Hospital Centre, 21000 Split, Croatia
| |
Collapse
|
7
|
Mustafa NH, Jalil J, Zainalabidin S, Saleh MS, Asmadi AY, Kamisah Y. Molecular mechanisms of sacubitril/valsartan in cardiac remodeling. Front Pharmacol 2022; 13:892460. [PMID: 36003518 PMCID: PMC9393311 DOI: 10.3389/fphar.2022.892460] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/11/2022] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases have become a major clinical burden globally. Heart failure is one of the diseases that commonly emanates from progressive uncontrolled hypertension. This gives rise to the need for a new treatment for the disease. Sacubitril/valsartan is a new drug combination that has been approved for patients with heart failure. This review aims to detail the mechanism of action for sacubitril/valsartan in cardiac remodeling, a cellular and molecular process that occurs during the development of heart failure. Accumulating evidence has unveiled the cardioprotective effects of sacubitril/valsartan on cellular and molecular modulation in cardiac remodeling, with recent large-scale randomized clinical trials confirming its supremacy over other traditional heart failure treatments. However, its molecular mechanism of action in cardiac remodeling remains obscure. Therefore, comprehending the molecular mechanism of action of sacubitril/valsartan could help future research to study the drug's potential therapy to reduce the severity of heart failure.
Collapse
Affiliation(s)
- Nor Hidayah Mustafa
- Centre for Drug and Herbal Research Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Juriyati Jalil
- Centre for Drug and Herbal Research Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Satirah Zainalabidin
- Program of Biomedical Science, Centre of Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohammed S.M. Saleh
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ahmad Yusof Asmadi
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia, Kuala Lumpur, Malaysia
| | - Yusof Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Nikolic M, Srejovic I, Jovic JJ, Sretenovic J, Jeremic J, Cekerevac I, Simovic S, Djokovic D, Muric N, Stojic V, Bolevich S, Bolevich S, Jakovljevic V. Sacubitril/valsartan in Heart Failure and Beyond-From Molecular Mechanisms to Clinical Relevance. Rev Cardiovasc Med 2022; 23:238. [PMID: 39076908 PMCID: PMC11266818 DOI: 10.31083/j.rcm2307238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 07/31/2024] Open
Abstract
As the ultimate pathophysiological event, heart failure (HF) may arise from various cardiovascular (CV) conditions, including sustained pressure/volume overload of the left ventricle, myocardial infarction or ischemia, and cardiomyopathies. Sacubitril/valsartan (S/V; formerly termed as LCZ696), a first-in-class angiotensin receptor/neprilysin inhibitor, brought a significant shift in the management of HF with reduced ejection fraction by modulating both renin-angiotensin-aldosterone system (angiotensin II type I receptor blockage by valsartan) and natriuretic peptide system (neprilysin inhibition by sacubitril) pathways. Besides, the efficacy of S/V has been also investigated in the setting of other CV pathologies which are during their pathophysiological course and progression deeply interrelated with HF. However, its mechanism of action is not entirely clarified, suggesting other off-target benefits contributing to its cardioprotection. In this review article our goal was to highlight up-to-date clinical and experimental evidence on S/V cardioprotective effects, as well as most discussed molecular mechanisms achieved by this dual-acting compound. Although S/V was extensively investigated in HF patients, additional large studies are needed to elucidate its effects in the setting of other CV conditions. Furthermore, with its antiinflamatory potential, this agent should be investigated in animal models of inflammatory heart diseases, such as myocarditis, while it may possibly improve cardiac dysfunction as well as inflammatory response in this pathophysiological setting. Also, discovering other signalling pathways affected by S/V should be of particular interest for basic researches, while it can provide additional understanding of its cardioprotective mechanisms.
Collapse
Affiliation(s)
- Maja Nikolic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivan Srejovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Jovana Joksimovic Jovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Jasmina Sretenovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Jovana Jeremic
- Department of Pharmacology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivan Cekerevac
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Clinic of Pulmology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Stefan Simovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Clinic of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Danijela Djokovic
- Department of Psychiatry, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Clinic of Psychiatry, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Nemanja Muric
- Department of Psychiatry, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Clinic of Psychiatry, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Vladislava Stojic
- Department of Medical Statistics and Informatics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Stefani Bolevich
- Department of Pathophysiology, 1st Moscow State Medical University IM Sechenov, 119991 Moscow, Russia
| | - Sergey Bolevich
- Department of Human Pathology, 1st Moscow State Medical University IM Sechenov, 119991 Moscow, Russia
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Department of Human Pathology, 1st Moscow State Medical University IM Sechenov, 119991 Moscow, Russia
| |
Collapse
|
9
|
Zhang Y, Yuan M, Suo Y, Yang Q, Shao S, Li Y, Wang Y, Bao Q, Liu T, Li G. Angiotensin Receptor-Neprilysin Inhibitor Attenuates Cardiac Hypertrophy and Improves Diastolic Dysfunction in A Mouse Model of Heart Failure with Preserved Ejection Fraction. Clin Exp Pharmacol Physiol 2022; 49:848-857. [PMID: 35596518 DOI: 10.1111/1440-1681.13672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022]
Abstract
LCZ696, an angiotensin receptor-neprilysin inhibitor, has shown promising clinical efficacy in patients with heart failure (HF) with reduced ejection fraction. However, its potential effects on heart failure with preserved ejection fraction (HFpEF) are still not fully understood. We evaluated the effect of LCZ696 on HFpEF in transverse aortic constriction mice and compared it with the effect of the angiotensin receptor blocker valsartan. We found that LCZ696 improved cardiac diastolic function by reducing ventricular hypertrophy and fibrosis in mice with overload-induced diastolic dysfunction. In addition, there was superior inhibition of LCZ696 than stand-alone valsartan. As a potential underlying mechanism, we demonstrated that LCZ696 behaves as a potent suppressor of calcium-mediated calcineurin-NFAT signaling transduction pathways. Hence, we demonstrated the protective effects of LCZ696 in overload-induced HFpEF and provided a pharmaceutical therapeutic strategy for related diseases.
Collapse
Affiliation(s)
- Yue Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Meng Yuan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Ya Suo
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Qian Yang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Shuai Shao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Ying Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Yuanyuan Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Qiankun Bao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
10
|
Hou M, Lu L, Wu X, Liu H. LCZ696 Ameliorates Isoproterenol-Induced Acute Heart Failure in Rats by Activating the Nrf2 Signaling Pathway. Appl Bionics Biomech 2022; 2022:6077429. [PMID: 35528528 PMCID: PMC9076311 DOI: 10.1155/2022/6077429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/25/2022] Open
Abstract
Objective LCZ696 (sacubitril/valsartan) is an angiotensin II (Ang II) type 1 receptor-neprilysin inhibitor, with effects of immunosuppression, anti-inflammation, antiapoptosis, and antioxidation. The present study was aimed at determining whether LCZ696 has a protective effect against isoproterenol-induced acute heart failure (AHF) in rats. Methods SD rats were randomly divided into four groups: control group, HF group, LCZ696 group, and enalapril group. The cardiac function of rats was evaluated using echocardiographic parameters, heart weight (HW), serum levels of cardiac troponin I (cTnI), and lactate dehydrogenase (LDH). HE is staining, which was used to determine the pathological damage of rat myocardial tissue. Also, we measured oxidative stress markers including reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT). Finally, the expression of Nrf2 signaling pathway-related proteins was determined using Western blot. Results Compared with the HF group, LCZ696 could significantly improve cardiac function and myocardial injury in rats and reduce AHF-induced oxidative stress. In addition, the results of Western blot confirmed that LCZ696 could upregulate the expression of Nrf2 and HO-1 while decreasing Keap1 expression. Conclusion LCZ696 ameliorates isoproterenol-induced AHF in rats by alleviating oxidative stress injury and activating the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Min Hou
- Department of Emergency, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Linxin Lu
- Department of Emergency, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaobo Wu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Lymphoma, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Hongxuan Liu
- Department of Emergency, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
11
|
LCZ696 ameliorates doxorubicin-induced cardiomyocyte toxicity in rats. Sci Rep 2022; 12:4930. [PMID: 35322164 PMCID: PMC8943022 DOI: 10.1038/s41598-022-09094-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/17/2022] [Indexed: 12/11/2022] Open
Abstract
Doxorubicin (DOX)-based chemotherapy induces cardiotoxicity, which is considered the main bottleneck for its clinical application. In this study, we investigated the potential benefit of LCZ696, an angiotensin receptor-neprilysin inhibitor against DOX-induced cardiotoxicity in rats and H9c2 cells and determined whether the mechanism underlying any such effects involves its antioxidant activity. Male Sprague-Dawley rats were randomly separated into four groups, each consisting of 15 rats (DOX (1.5 mg/kg/day intraperitoneally for 10 days followed by non-treatment for 8 days); DOX + valsartan (31 mg/kg/day by gavage from day 1 to day 18); DOX + LCZ696 (68 mg/kg/day by gavage from day 1 to day 18); and control (saline intraperitoneally for 10 days). DOX-induced elevation of cardiac troponin T levels on day 18 was significantly reduced by LCZ696, but not valsartan. The DOX-induced increase in myocardial reactive oxygen species (ROS) levels determined using dihydroethidium was significantly ameliorated by LCZ696, but not valsartan, and was accompanied by the suppression of DOX-induced increase in p47phox. LCZ696 recovered the DOX-induced decrease in phosphorylation of adenosine monophosphate-activated protein kinase and increased the ratio of Bax and Bcl-2. In H9c2 cardiomyocytes, LCZ696 reduced DOX-induced mitochondrial ROS generation and improved cell viability more than valsartan. Our findings indicated that LCZ696 ameliorated DOX-induced cardiotoxicity in rat hearts in vivo and in vitro, possibly by mediating a decrease in oxidative stress.
Collapse
|
12
|
Liu L, Ding X, Han Y, Lv J. Effects and Safety of Sacubitril/Valsartan for Patients with Myocardial Infarction: A Systematic Review and Meta-Analysis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7840852. [PMID: 35035857 PMCID: PMC8754592 DOI: 10.1155/2022/7840852] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/19/2021] [Accepted: 11/27/2021] [Indexed: 11/17/2022]
Abstract
Patients who develop heart failure (HF) after an acute myocardial infarction (AMI) are at higher risk of adverse fatal and nonfatal outcomes. Studies have shown sacubitril/valsartan can further reduce the risk of cardiovascular death or hospitalization for heart failure by 20% compared with enalapril. At the same time, its tolerance and safety are better. However, the current evidence regarding the efficacy of sacubitril/valsartan in patients with heart failure after acute myocardial infarction is controversial. To assess the effect of sacubitril/valsartan on heart failure after acute myocardial infarction, we conducted a systematic review of the literature and a meta-analysis of existing randomized clinical trials. Meta-analysis of randomized controlled trails is used where data are collected from PubMed, the Cochrane library, Embase, and Web of Science. Data about sacubitril/valsartan were available from 5 studies. Forest plots showed that the sacubitril/valsartan group had a 299% higher value of sacubitril/valsartan to the control group (MD = 2.99%, 95% CI: 2.01, 3.96, I 2 = 78%, P < 0.00001, Figure 2), and the difference was statistically significant. Forest plots showed that the sacubitril/valsartan group had a 531% lower value of LVEF to the control group (MD = -5.31%, 95% CI: -7.36, -3.26, I 2 = 91%, P < 0.00001, Figure 2), and the difference was statistically significant. Forest plots showed that the sacubitril/valsartan group had a 133% lower value of NT-proBNP to the control group (MD = -1.33%, 95% CI: -1.54, -1.12, I 2 = 96%, P < 0.00001, Figure 3). Forest plots showed that the sacubitril/valsartan group had a 49% lower risk of heart failure to the control group (MD = 0.49, 95% CI: 0.27, 0.89, I 2 = 0%, P=0.02, Figure 3). The patients in experimental showed an obviously lower OR of MACE (OR = 0.47, 95% CI: 0.27, 0.82, P=0.007, Figure 3). The data were statistically significant. We have observed that for patients with heart failure after acute myocardial infarction, early administration of sacubitril/valsartan can significantly reduce the incidence of heart rate, left ventricular ejection fraction, NT-proBNP, and MACE. Our meta-analysis suggests that taking sacubitril/valsartan is relatively safe and effective, especially if started early after acute myocardial infarction.
Collapse
Affiliation(s)
- Lang Liu
- General Hospital of Ningxia Medical University, Yinchuan City 750000, China
| | - Xiaofang Ding
- General Hospital of Ningxia Medical University, Yinchuan City 750000, China
| | - Yaxiang Han
- General Hospital of Ningxia Medical University, Yinchuan City 750000, China
| | - Jianfeng Lv
- Affiliated RenHe Hospital of China, Three Gorges University Second Clinical Medical College of China Three Gorges University, Yichang City 443000, China
| |
Collapse
|
13
|
Syed AM, Kundu S, Ram C, Kulhari U, Kumar A, Mugale MN, Murty US, Sahu BD. Aloin alleviates pathological cardiac hypertrophy via modulation of the oxidative and fibrotic response. Life Sci 2022; 288:120159. [PMID: 34801516 DOI: 10.1016/j.lfs.2021.120159] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 12/28/2022]
Abstract
AIMS Pathological cardiac hypertrophy is a characteristic feature in many cardiovascular diseases (CVDs). Aloin is an anthraquinone glycoside from Aloe species, and the effect of aloin on cardiac hypertrophy and associated fibrotic changes have not been elucidated. This study investigated the effect of aloin against the isoproterenol (ISO)-induced cardiac hypertrophy in rats. MAIN METHODS Cardiac hypertrophy experimental model was induced in rats by subcutaneous injection of ISO for 14 days. Meanwhile, the animals were administered orally with aloin at doses of 25 and 50 mg/kg/day. On the 15th day, cardiac echocardiography was performed, the heart was collected and subjected for histopathological, gene expression, and immunoblot studies. Additionally, the effect of aloin on ISO-induced hypertrophic changes in H9c2 cells was investigated. KEY FINDINGS Aloin markedly alleviated ISO-induced heart injury, reduced cardiac hypertrophy, improved cardiac function, and histological alterations in the heart. Mechanistically, aloin attenuated ISO-induced fibrosis via inhibition of the levels of collagen I, α-smooth muscle actin (α-SMA), fibronectin, transforming growth factor-β (TGF-β) and pSmad2/3 proteins in the heart. Aloin alleviated ISO-induced myocardial oxidative damage and up-regulated the levels of antioxidant transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) proteins. Moreover, aloin treatment attenuated ISO-induced hypertrophic changes and the generation of reactive oxygen species (ROS) in H9c2 cells in vitro. SIGNIFICANCE Our findings demonstrated that aloin alleviated ISO-induced cardiac hypertrophy and fibrosis via inhibiting TGF-β/pSmad2/3 signaling and restoring myocardial antioxidants, and therefore has promising therapeutic potential against cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Abu Mohammad Syed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, PIN-781101, Assam, India
| | - Sourav Kundu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, PIN-781101, Assam, India
| | - Chetan Ram
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, PIN-781101, Assam, India
| | - Uttam Kulhari
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, PIN-781101, Assam, India
| | - Akhilesh Kumar
- Toxicology & Experimental Medicine, CSIR - Central Drug Research Institute (CDRI), Lucknow 226 031, India
| | - Madhav Nilakanth Mugale
- Toxicology & Experimental Medicine, CSIR - Central Drug Research Institute (CDRI), Lucknow 226 031, India
| | - Upadhyayula Suryanarayana Murty
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, PIN-781101, Assam, India
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, PIN-781101, Assam, India.
| |
Collapse
|
14
|
Differential Effects of Sacubitril/Valsartan on Diastolic Function in Mice With Obesity-Related Metabolic Heart Disease. JACC Basic Transl Sci 2020; 5:916-927. [PMID: 33015414 PMCID: PMC7524781 DOI: 10.1016/j.jacbts.2020.07.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/13/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022]
Abstract
MHD associated with obesity, diabetes, and/or metabolic syndrome is an important precursor of HFpEF. Mice fed a HFHS diet develop MHD with myocardial hypertrophy, fibrosis, diastolic dysfunction, and impaired energetics. Mice on HFHS diet were treated with matched doses of VAL or sac SAC/VAL for 16 weeks. Only SAC/VAL prevented diastolic dysfunction and fibrosis, and to a lesser extent oxidative stress, whereas VAL and SAC/VAL had similar effects on hypertrophy and energetics. Neprilysin inhibition exerts beneficial effects on MHD that are complimentary to VAL, suggesting that SAC/VAL has promise to prevent the development of HFpEF in patients with or at risk for MHD.
Mice with obesity and metabolic heart disease (MHD) due to a high-fat, high-sucrose diet were treated with placebo, a clinically relevant dose of sacubitril (SAC)/valsartan (VAL), or an equivalent dose of VAL for 4 months. There were striking differences between SAC/VAL and VAL with regard to: 1) diastolic dysfunction; 2) interstitial fibrosis; and to a lesser degree; 3) oxidative stress—all of which were more favorably affected by SAC/VAL. SAC/VAL and VAL similarly attenuated myocardial hypertrophy and improved myocardial energetics. In mice with obesity-related MHD, neprilysin inhibition exerts favorable effects on diastolic function.
Collapse
Key Words
- ARB, angiotensin receptor blocker
- ATP, adenosine triphosphate
- CD, control diet
- GMP, guanosine monophosphate
- HFHS, high-fat, high-sucrose
- HFpEF, heart failure with a preserved ejection fraction
- LV, left ventricular
- MHD, metabolic heart disease
- MNA, methoxy-2-naphthlamine
- NMR, nuclear magnetic resonance
- PCr, phosphocreatine
- ROS, reactive oxygen species
- RPP, rate × pressure product
- SAC/VAL, sacubitril/valsartan
- VAL, valsartan
- diastolic
- neprilysin
- obesity
Collapse
|
15
|
Pathak MP, Das A, Patowary P, Chattopadhyay P. Contentious role of 'Good Adiponectin' in pulmonary and cardiovascular diseases: Is adiponectin directed therapy a boon or a bane? Biochimie 2020; 175:106-119. [PMID: 32473183 DOI: 10.1016/j.biochi.2020.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/27/2020] [Accepted: 05/12/2020] [Indexed: 11/24/2022]
Abstract
After two decades of its discovery, numerous facts of adiponectin (APN) biology has been uncovered, yet, APN remains an elusive adipokine. Findings from clinical studies and animal models established APN's ameliorative role in cardiovascular disease (CVD) and pulmonary disease (PD) but the same condition is prognostic for mortality in the same set of patients which cornered APN towards a dubious state. A repertoire of mechanisms associated with the positive association of APN in both lean/cachectic or obese CVD and PD patients from past publications are evaluated. Newer pharmacological agent may be explored to regulate elevated blood APN concentration in COPD or CHF patients whereas administration of recombinant APN as well as growth hormone may augment blood APN concentration in obese subjects associated with low blood and intracellular APN concentration. However, some APN directed therapy in clinical as well as in pre-clinical setup has pronounced some contentious effects. After reviewing the mechanisms of the contentious role of APN functioning in pathologic conditions of CVD and PD in both lean and obese conditions, the authors came to conclusion that APN directed therapy may be utilized with caution keeping in mind the different age group, sex and the different CVD as well as pulmonary diseases they are suffering from.
Collapse
Affiliation(s)
- Manash Pratim Pathak
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, India; Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, India
| | - Aparoop Das
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, India
| | - Pompy Patowary
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, India; Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, India
| | | |
Collapse
|
16
|
Hatipoglu OF, Miyoshi T, Yonezawa T, Kondo M, Amioka N, Yoshida M, Akagi S, Nakamura K, Hirohata S, Ito H. Deficiency of CD44 prevents thoracic aortic dissection in a murine model. Sci Rep 2020; 10:6869. [PMID: 32321956 PMCID: PMC7176701 DOI: 10.1038/s41598-020-63824-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/07/2020] [Indexed: 01/16/2023] Open
Abstract
Thoracic aortic dissection (TAD) is a life-threatening vascular disease. We showed that CD44, a widely distributed cell surface adhesion molecule, has an important role in inflammation. In this study, we examined the role of CD44 in the development of TAD. TAD was induced by the continuous infusion of β-aminopropionitrile (BAPN), a lysyl oxidase inhibitor, and angiotensin II (AngII) for 7 days in wild type (WT) mice and CD44 deficient (CD44-/-) mice. The incidence of TAD in CD44-/- mice was significantly reduced compared with WT mice (44% and 6%, p < 0.01). Next, to evaluate the initial changes, aortic tissues at 24 hours after BAPN/AngII infusion were examined. Neutrophil accumulation into thoracic aortic adventitia in CD44-/- mice was significantly decreased compared with that in WT mice (5.7 ± 0.3% and 1.6 ± 0.4%, p < 0.01). In addition, BAPN/AngII induced interleukin-6, interleukin-1β, matrix metalloproteinase-2 and matrix metalloproteinase-9 in WT mice, all of which were significantly reduced in CD44−/− mice (all p < 0.01). In vitro transmigration of neutrophils from CD44−/− mice through an endothelial monolayer was significantly decreased by 18% compared with WT mice (p < 0.01). Our findings indicate that CD44 has a critical role in TAD development in association with neutrophil infiltration into adventitia.
Collapse
Affiliation(s)
- Omer F Hatipoglu
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan.,Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama, Japan
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan.
| | - Tomoko Yonezawa
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Megumi Kondo
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Naofumi Amioka
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Masashi Yoshida
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Satoshi Hirohata
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama, Japan
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| |
Collapse
|
17
|
LCZ696, an Angiotensin Receptor-Neprilysin Inhibitor, Improves Cardiac Hypertrophy and Fibrosis and Cardiac Lymphatic Remodeling in Transverse Aortic Constriction Model Mice. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7256862. [PMID: 32420365 PMCID: PMC7201829 DOI: 10.1155/2020/7256862] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/15/2019] [Accepted: 12/09/2019] [Indexed: 11/20/2022]
Abstract
Cardiac hypertrophy and ventricular remodeling following heart failure are important causes of high mortality in heart disease patients. The cardiac lymphatic system has been associated with limited research, but it plays an important role in the improvement of myocardial edema and the promotion of fluid balance. LCZ696 is a novel combination of angiotensin and neprilysin inhibitors. Here, we studied the role played by LCZ696 during transverse aortic constriction (TAC) induced cardiac hypertrophy and changes in the lymphatic system. Mice undergoing aortic coarctation were constructed to represent a cardiac hypertrophy model and then divided into random groups that either received treatment with LCZ696 (60 mg/kg/d) or no treatment. Cardiac ultrasonography was used to detect cardiac function, and hematoxylin and eosin (H&E) and Masson staining were used to detect myocardial hypertrophy and fibrosis. The proinflammatory factors interleukin-6 (IL-6), IL-1β, and tumor necrosis factor-α (TNF-α) were detected in the blood and heart tissues of mice. The protein expression levels of lymphatic-specific markers, such as vascular endothelial growth factor C (VEGF-C), VEGF receptor 3 (VEGFR3), and lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1) were detected in mouse heart tissues. We also examined the colocalization of lymphatic vessels and macrophages by immunofluorescence. The results showed that LCZ696 significantly improved heart dysfunction, cardiac hypertrophy, and fibrosis and inhibited the expression of proinflammatory factors IL-6, IL-1β, and TNF-α in the circulating blood and heart tissues of mice. LCZ696 also decreased the protein expression levels of VEGF-C, VEGFR3, and LYVE-1 in mouse heart tissues, ameliorated the transport load of lymphatic vessels to macrophages, and improved the remodeling of the lymphatic system in the hypertrophic cardiomyopathy model induced by TAC.
Collapse
|
18
|
Zhang H, Tian L, Shen M, Tu C, Wu H, Gu M, Paik DT, Wu JC. Generation of Quiescent Cardiac Fibroblasts From Human Induced Pluripotent Stem Cells for In Vitro Modeling of Cardiac Fibrosis. Circ Res 2019; 125:552-566. [PMID: 31288631 DOI: 10.1161/circresaha.119.315491] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RATIONALE Activated fibroblasts are the major cell type that secretes excessive extracellular matrix in response to injury, contributing to pathological fibrosis and leading to organ failure. Effective anti-fibrotic therapeutic solutions, however, are not available due to the poorly defined characteristics and unavailability of tissue-specific fibroblasts. Recent advances in single-cell RNA-sequencing fill such gaps of knowledge by enabling delineation of the developmental trajectories and identification of regulatory pathways of tissue-specific fibroblasts among different organs. OBJECTIVE This study aims to define the transcriptome profiles of tissue-specific fibroblasts using recently reported mouse single-cell RNA-sequencing atlas and to develop a robust chemically defined protocol to derive cardiac fibroblasts (CFs) from human induced pluripotent stem cells for in vitro modeling of cardiac fibrosis and drug screening. METHODS AND RESULTS By analyzing the single-cell transcriptome profiles of fibroblasts from 10 selected mouse tissues, we identified distinct tissue-specific signature genes, including transcription factors that define the identities of fibroblasts in the heart, lungs, trachea, and bladder. We also determined that CFs in large are of the epicardial lineage. We thus developed a robust chemically defined protocol that generates CFs from human induced pluripotent stem cells. Functional studies confirmed that iPSC-derived CFs preserved a quiescent phenotype and highly resembled primary CFs at the transcriptional, cellular, and functional levels. We demonstrated that this cell-based platform is sensitive to both pro- and anti-fibrosis drugs. Finally, we showed that crosstalk between human induced pluripotent stem cell-derived cardiomyocytes and CFs via the atrial/brain natriuretic peptide-natriuretic peptide receptor-1 pathway is implicated in suppressing fibrogenesis. CONCLUSIONS This study uncovers unique gene signatures that define tissue-specific identities of fibroblasts. The bona fide quiescent CFs derived from human induced pluripotent stem cells can serve as a faithful in vitro platform to better understand the underlying mechanisms of cardiac fibrosis and to screen anti-fibrotic drugs.
Collapse
Affiliation(s)
- Hao Zhang
- From the Stanford Cardiovascular Institute (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
- Department of Radiology (H.Z., L.T., M.S., C.T., H.W., M.G., D.T.P., J.C.W.), CA
- Department of Medicine, Division of Cardiology (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
| | - Lei Tian
- From the Stanford Cardiovascular Institute (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
- Department of Radiology (H.Z., L.T., M.S., C.T., H.W., M.G., D.T.P., J.C.W.), CA
- Department of Medicine, Division of Cardiology (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
| | - Mengcheng Shen
- From the Stanford Cardiovascular Institute (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
- Department of Radiology (H.Z., L.T., M.S., C.T., H.W., M.G., D.T.P., J.C.W.), CA
- Department of Medicine, Division of Cardiology (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
| | - Chengyi Tu
- From the Stanford Cardiovascular Institute (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
- Department of Radiology (H.Z., L.T., M.S., C.T., H.W., M.G., D.T.P., J.C.W.), CA
- Department of Medicine, Division of Cardiology (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
| | - Haodi Wu
- From the Stanford Cardiovascular Institute (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
- Department of Radiology (H.Z., L.T., M.S., C.T., H.W., M.G., D.T.P., J.C.W.), CA
- Department of Medicine, Division of Cardiology (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
| | - Mingxia Gu
- Department of Radiology (H.Z., L.T., M.S., C.T., H.W., M.G., D.T.P., J.C.W.), CA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases (M.G.), CA
- Department of Pediatrics, Stanford University School of Medicine (M.G.), CA
| | - David T Paik
- From the Stanford Cardiovascular Institute (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
- Department of Radiology (H.Z., L.T., M.S., C.T., H.W., M.G., D.T.P., J.C.W.), CA
- Department of Medicine, Division of Cardiology (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
- Department of Radiology (H.Z., L.T., M.S., C.T., H.W., M.G., D.T.P., J.C.W.), CA
- Department of Medicine, Division of Cardiology (H.Z., L.T., M.S., C.T., H.W., D.T.P., J.C.W.), CA
| |
Collapse
|
19
|
Korshunov VA, Quinn B, Faiyaz A, Ahmed R, Sowden MP, Doyley MM, Berk BC. Strain-selective efficacy of sacubitril/valsartan on carotid fibrosis in response to injury in two inbred mouse strains. Br J Pharmacol 2019; 176:2795-2807. [PMID: 31077344 DOI: 10.1111/bph.14708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Sacubitril/valsartan (Sac/val) is more effective than valsartan in lowering BP and mortality in patients with heart failure. Here, we proposed that Sac/val treatment would be more effective in preventing pathological vascular remodelling in 129X1/SvJ (129X1), than in C57BL/6J (B6) inbred mice. EXPERIMENTAL APPROACH Sac/val (60 mg·kg-1 ·day-1 ) and valsartan (27 mg·kg-1 ·day-1 ) were given as prophylactic or therapeutic treatments, to 129X1 or B6 mice with carotid artery ligation for 14 days. Blood flow was measured by ultrasound. Ex vivo, carotid tissue was analysed with histological and morphometric techniques, together with RNA sequencing and gene ontology. KEY RESULTS Sac/val was more effective than valsartan in lowering BP in 129X1 compared with B6 mice. Liver expression of CYP2C9 and plasma cGMP levels were similar across treatments. A reduction in carotid thickening after prophylactic treatment with valsartan or Sac/val also resulted in significant arterial shrinkage in B6 mice. In 129X1 mice, Sac/val and prophylactic treatment with valsartan had no effect on carotid thickening but preserved carotid size. BP lowering significantly correlated with a decline in carotid stiffness (R2 = .37, P = .0096) in 129X1 but not in B6 mice. The gene expression signature associated with hyalurononglucosaminidase activity was down-regulated in injured arteries after both regimens of Sac/val only in 129X1 mice. Administration of Sac/val but not valsartan significantly reduced deposition of hyaluronic acid and carotid fibrosis in 129X1 mice. CONCLUSION AND IMPLICATIONS These results underscore the importance of the genetic background in the efficacy of the Sac/val on vascular fibrosis.
Collapse
Affiliation(s)
- Vyacheslav A Korshunov
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York
| | - Breandan Quinn
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York
| | - Abrar Faiyaz
- Department of Electrical and Computer Engineering, Hajim School of Engineering and Applied Sciences, University of Rochester, Rochester, New York
| | - Rifat Ahmed
- Department of Electrical and Computer Engineering, Hajim School of Engineering and Applied Sciences, University of Rochester, Rochester, New York
| | - Mark P Sowden
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York
| | - Marvin M Doyley
- Department of Electrical and Computer Engineering, Hajim School of Engineering and Applied Sciences, University of Rochester, Rochester, New York
| | - Bradford C Berk
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York.,Neurorestoration Institute, University of Rochester, Rochester, New York
| |
Collapse
|