1
|
Di Pietro G, Improta R, Severino P, D'Amato A, Birtolo LI, De Filippo O, Lattanzio A, De Cristofaro R, Galardo G, D'Ascenzo F, Badagliacca R, Sardella G, Volterrani M, Fedele F, Vizza CD, Mancone M. The in-hospital administration of sacubitril/valsartan in acute myocardial infarction: A meta-analysis. ESC Heart Fail 2025; 12:998-1012. [PMID: 39473218 PMCID: PMC11911573 DOI: 10.1002/ehf2.15082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 03/18/2025] Open
Abstract
There is a need to address the evidence gap regarding the in-hospital administration of sacubitril/valsartan in acute myocardial infarction patients. After searching MEDLINE, Google Scholars and Scopus, a random-effects meta-analysis of randomized controlled trials comparing the in-hospital administration of the angiotensin receptor-neprilysin inhibitors (ARNis) versus the standard therapy in patients with reduced heart failure due to myocardial infarction was performed. The primary outcome was major adverse cardiovascular events. All-cause mortality, cardiac death, rehospitalization for heart failure, non-fatal myocardial infarction (MI), changes in left ventricular ejection fraction, left ventricular volumes, N terminal pro brain natriuretic peptide and adverse events were the secondary endpoints. Nine studies (eight randomized controlled trials and one echo-substudy) with a total 6597 individuals (angiotensin-converting enzyme inhibitor/angiotensin receptor blocker: 3300 patients vs. ARNis: 3297 patients) were included for quantitative analysis. Median follow-up was 6 months. Patients receiving an in-hospital coadministration of ARNi had a lower risk of major cardiovascular event [odds ratio (OR) 0.45, 95% confidence interval (CI) 0.32-0.63, P < 0.0001] and lower rate of repeat rehospitalization for heart failure (OR 0.40, 95% CI 0.26-0.62, P < 0.0001), compared with a standard regimen. Additionally, left ventricle volumes were significantly lower in the ARNi group [left ventricular end-diastolic volume, mean difference (MD) 11.48 mL, 95% CI 6.10-16.85, P < 0.0001; left ventricular end-systolic volume, MD 7.09 mL, 95% CI 2.89-11.29, P = 0.0009] with a significant change in left ventricular ejection fraction (MD 3.07, 95% CI 1.61-4.53, P < 0.0001), compared with standard therapy. No significant differences were observed in terms of cardiac death, all cause of mortality, non-fatal myocardial infarction and N terminal pro brain natriuretic peptide. Higher rates of iatrogenic hypotensive events were observed in the ARNi group compared with the standard therapy (OR 1.42, 95% CI 1.26-1.60, P value < 0.00001). In patients with acute myocardial infarction related heart failure, the in-hospital administration of ARNis was associated with a reduced risk of major cardiovascular events and re-hospitalization for heart failure, as well as cardiac remodelling, but higher rates of hypotensive events compared with standard therapy.
Collapse
Affiliation(s)
- Gianluca Di Pietro
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| | - Riccardo Improta
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| | - Paolo Severino
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| | - Andrea D'Amato
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| | - Lucia Ilaria Birtolo
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| | - Ovidio De Filippo
- Department of Cardiovascular and Thoracic, Division of CardiologyCittà della Salute e della Scienza HospitalTurinItaly
| | - Antonio Lattanzio
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| | - Raffaele De Cristofaro
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| | | | - Fabrizio D'Ascenzo
- Department of Cardiovascular and Thoracic, Division of CardiologyCittà della Salute e della Scienza HospitalTurinItaly
| | - Roberto Badagliacca
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| | - Gennaro Sardella
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| | | | - Francesco Fedele
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
- Casa di Cura San Raffaele MontecompatriRomeItaly
| | - Carmine Dario Vizza
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| | - Massimo Mancone
- Department of Internal Clinical, Anesthesiological, Cardiovascular SciencesLa Sapienza University of RomeRomeItaly
| |
Collapse
|
2
|
Bussmann B, Ayagama T, Liu K, Li D, Herring N. Bayliss Starling Prize Lecture 2023: Neuropeptide-Y being 'unsympathetic' to the broken hearted. J Physiol 2025; 603:1841-1864. [PMID: 38847435 PMCID: PMC11955873 DOI: 10.1113/jp285370] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/01/2024] [Indexed: 04/01/2025] Open
Abstract
William Bayliss and Ernest Starling are not only famous as pioneers in cardiovascular physiology, but also responsible for the discovery of the first hormone (from the Greek 'excite or arouse'), the intestinal signalling molecule and neuropeptide secretin in 1902. Our research group focuses on neuropeptides and neuromodulators that influence cardiovascular autonomic control as potential biomarkers in disease and tractable targets for therapeutic intervention. Acute myocardial infarction (AMI) and chronic heart failure (CHF) result in high levels of cardiac sympathetic stimulation, which is a poor prognostic indicator. Although beta-blockers improve mortality in these conditions by preventing the action of the neurotransmitter noradrenaline, a substantial residual risk remains. Recently, we have identified the sympathetic co-transmitter neuropeptide-Y (NPY) as being released during AMI, leading to larger infarcts and life-threatening arrhythmia in both animal models and patients. Here, we discuss recently published data demonstrating that peripheral venous NPY levels are associated with heart failure hospitalisation and mortality after AMI, and all cause cardiovascular mortality in CHF, even when adjusting for known risk factors (including brain natriuretic peptide). We have investigated the mechanistic basis for these observations in human and rat stellate ganglia and cardiac tissue, manipulating NPY neurochemistry at the same time as using state-of-the-art imaging techniques, to establish the receptor pathways responsible for NPY signalling. We propose NPY as a new mechanistic biomarker in AMI and CHF patients and aim to determine whether specific NPY receptor blockers can prevent arrhythmia and attenuate the development of heart failure.
Collapse
Affiliation(s)
- Benjamin Bussmann
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Thamali Ayagama
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Kun Liu
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Dan Li
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
3
|
Chennappan S, Kontaridis MI. RASopathies in Cardiac Disease. Annu Rev Med 2025; 76:301-314. [PMID: 39576684 DOI: 10.1146/annurev-med-042823-013552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
RASopathies are a group of clinically overlapping autosomal dominant disorders caused primarily by mutations in genes that reside along the canonical Ras-mitogen-activated protein kinase signaling cascade. Though individually rare, collectively, these disorders constitute one of the largest families of congenital disorders worldwide, particularly for infantile hypertrophic cardiomyopathy. Significantly, despite almost five decades of RASopathy research, therapeutic options remain limited and focused primarily on treating symptoms rather than disease etiology. Targeting the genes causal to these disorders, and the nodal pathways critical for their regulation, however, has been challenging. In this review, we highlight these challenges, particularly with respect to congenital heart defects and cardiac diseases and discuss limitations and future directions for approaches to new therapeutic strategies.
Collapse
Affiliation(s)
- Saravanakkumar Chennappan
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, New York, USA;
| | - Maria Irene Kontaridis
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, New York, USA;
| |
Collapse
|
4
|
Jangid MK, Doshi GM. Cross talk on therapeutic strategies: natriuretic peptides and inhibiting neprilysin in hypertension management. Hypertens Res 2025; 48:284-300. [PMID: 39543415 DOI: 10.1038/s41440-024-01989-w] [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: 06/23/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024]
Abstract
Hypertension, a prevalent cardiovascular condition globally, remains a significant public health concern due to its association with increased cardiovascular morbidity and mortality. Despite the availability of various antihypertensive therapies, achieving optimal blood pressure control in patients remains a challenge. Valsartan/sacubitril (ARNi), marketed as Entresto by Novartis, combines valsartan, an angiotensin receptor blocker, with sacubitril, an inhibitor of neprilysin. Neprilysin is responsible for breaking down natriuretic peptides and other vasoactive substances. Inhibiting neprilysin prevents the degradation of natriuretic peptides, enhancing their beneficial effects on blood pressure regulation. Natriuretic Peptides, including atrial natriuretic peptide (ANP) and brain natriuretic peptides (BNP), play pivotal roles in regulating blood pressure and cardiovascular homeostasis by promoting vasodilation, natriuresis, and antagonizing the renin-angiotensin-aldosterone system. Therefore, this combo drug lessens sensitivity to natriuretic peptides and tackles the processes in hypertension that activate the renin-angiotensin-aldosterone system. This review provides an overview of how natriuretic peptides (NPs) contribute to blood pressure regulation for the treatment of hypertension through inhibiting neprilysin. It highlights the ARNi's dual action that works synergistically by blocking the harmful effects of angiotensin II on blood vessels while simultaneously increasing the levels of beneficial natriuretic peptides. Schematic representation of the mechanism of action of ARNi. Abbreviation: -Renin angiotensin aldosterone system (RAAS), Natriuretic peptides (NP), Atrial Natriuretic peptide (ANP), Brain natriuretic peptide (BNP), C-type natriuretic peptide (CNP), Angiotensin II (Ang II), Angiotensin receptor neprilysin inhibitor (ARNI).
Collapse
Affiliation(s)
- Maya K Jangid
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V. M. Road, Vile Parle (W), Mumbai, 400056, Maharashtra, India
| | - Gaurav M Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V. M. Road, Vile Parle (W), Mumbai, 400056, Maharashtra, India.
| |
Collapse
|
5
|
Perez-Ternero C, Li W, Aubdool AA, Goldin RD, Loy J, Devalia K, Alazawi W, Hobbs AJ. Endogenous C-type natriuretic peptide offsets the pathogenesis of steatohepatitis, hepatic fibrosis, and portal hypertension. PNAS NEXUS 2025; 4:pgae579. [PMID: 39816244 PMCID: PMC11734523 DOI: 10.1093/pnasnexus/pgae579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), hepatic fibrosis, and portal hypertension constitute an increasing public health problem due to the growing prevalence of obesity and diabetes. C-type natriuretic peptide (CNP) is an endogenous regulator of cardiovascular homeostasis, immune cell reactivity, and fibrotic disease. Thus, we investigated a role for CNP in the pathogenesis of MASLD. Wild-type (WT), global CNP (gbCNP-/-), and natriuretic peptide receptor-C (NPR-C-/-) knockout mice were fed a choline-deficient defined amino acid diet or administered CCl4. Liver damage was assessed by histological and biochemical analyses, with steatosis and portal vein size determined by ultrasound. Portal vein pressure and reactivity were measured in vivo and ex vivo, respectively. Pharmacological CNP delivery was used to evaluate prospective therapeutic benefit, and plasma CNP concentration was compared in controls and patients with cirrhosis. Circulating CNP concentration was lower in patients with cirrhosis compared with controls. gbCNP-/- mice were more susceptible, versus WT, to advanced steatohepatitis and hepatic fibrosis, characterized by increased immune cell infiltration, fibrosis, ballooning, plasma alanine aminotransferase concentration, and up-regulation of markers driving these processes. gbCNP-/- mice had increased portal vein diameter and pressure, underpinned by CNP insensitivity. NPR-C-/- animals recapitulated, comparatively, the exaggerated pathogenic phenotype in gbCNP-/- mice, whereas CNP reduced hepatic stellate cell proliferation via NPR-B-dependent inhibition of extracellular signal-related kinase 1/2. Administration of CNP reversed many aspects of disease severity. These data define a new intrinsic role for CNP in offsetting the pathogenesis of MASLD, hepatic fibrosis, and portal hypertension and the potential for targeting CNP signaling for treating these disorders.
Collapse
Affiliation(s)
- Cristina Perez-Ternero
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Wenhao Li
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Aisah A Aubdool
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Robert D Goldin
- Centre for Pathology, St Mary’s Hospital, Imperial College, London W2 1NY, United Kingdom
| | - John Loy
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - Kalpana Devalia
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - William Alazawi
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Adrian J Hobbs
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
6
|
Lowe VJ, Aubdool AA, Moyes AJ, Dignam JP, Perez-Ternero C, Baliga RS, Smart N, Hobbs AJ. Cardiomyocyte-derived C-type natriuretic peptide diminishes myocardial ischaemic injury by promoting revascularisation and limiting fibrotic burden. Pharmacol Res 2024; 209:107447. [PMID: 39374886 DOI: 10.1016/j.phrs.2024.107447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND C-type natriuretic peptide (CNP) is a significant player in the maintenance of cardiac and vascular homeostasis regulating local blood flow, platelet and leukocyte activation, heart structure and function, angiogenesis and metabolic balance. Since such processes are perturbed in myocardial infarction (MI), we explored the role of cardiomyocyte-derived CNP, and pharmacological administration of the peptide, in offsetting the pathological consequences of MI. METHODS Wild type (WT) and cardiomyocyte-restricted CNP null (cmCNP-/-) mice were subjected to left anterior descending coronary artery (LADCA) ligation and acute effects on infarct size and longer-term outcomes of cardiac repair explored. Heart structure and function were assessed by combined echocardiographic and molecular analyses. Pharmacological administration of CNP (0.2 mg/kg/day; s.c.) was utilized to assess therapeutic potential. RESULTS Compared to WT littermates, cmCNP-/- mice had a modestly increased infarct size following LADCA ligation but without significant deterioration of cardiac structural and functional indices. However, cmCNP-/- animals exhibited overtly worse heart morphology and contractility 6 weeks following MI, with particularly deleterious reductions in left ventricular ejection fraction, dilatation, fibrosis and revascularization. This phenotype was largely recapitulated in animals with global deletion of natriuretic peptide receptor (NPR)-C (NPR-C-/-). Pharmacological administration of CNP rescued the deleterious pathology in WT and cmCNP-/-, but not NPR-C-/-, animals. CONCLUSIONS AND IMPLICATIONS Cardiomyocytes synthesize and release CNP as an intrinsic protective mechanism in response to MI that reduces cardiac structural and functional deficits; these salutary actions are primarily NPR-C-dependent. Pharmacological targeting of CNP may represent a new therapeutic option for MI.
Collapse
Affiliation(s)
- Vanessa J Lowe
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Aisah A Aubdool
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Amie J Moyes
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Joshua P Dignam
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - C Perez-Ternero
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Reshma S Baliga
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Nicola Smart
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX3 7TY, UK
| | - Adrian J Hobbs
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
7
|
Dickinson YA, Moyes AJ, Hobbs AJ. C-type natriuretic peptide (CNP): The cardiovascular system and beyond. Pharmacol Ther 2024; 262:108708. [PMID: 39154787 DOI: 10.1016/j.pharmthera.2024.108708] [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: 03/28/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
C-type natriuretic peptide (CNP) represents the 'local' member of the natriuretic peptide family, functioning in an autocrine or paracrine capacity to modulate a hugely diverse portfolio of physiological processes. Whilst the best-characterised of these regulatory roles are in the cardiovascular system, akin to its predominantly endocrine siblings atrial (ANP) and brain (BNP) natriuretic peptides, CNP governs many additional, unrelated mechanisms including bone growth, gamete maturation, auditory processing, and neuronal integrity. Furthermore, there is currently great interest in mimicking the biological activity of CNP for therapeutic gain in many of these disparate organ systems. Herein, we provide an overview of the physiology, pathophysiology and pharmacology of CNP in both cardiovascular and non-cardiovascular settings.
Collapse
Affiliation(s)
- Yasmin A Dickinson
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Amie J Moyes
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Adrian J Hobbs
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
8
|
Chiew MY, Wang E, Lan KC, Lin YR, Hsueh YH, Tu YK, Liu CF, Chen PC, Lu HE, Chen WL. Improving iPSC Differentiation Using a Nanodot Platform. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36030-36046. [PMID: 38951110 PMCID: PMC11261571 DOI: 10.1021/acsami.4c04451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
Differentiation of induced pluripotent stem cells (iPSCs) is an extremely complex process that has proven difficult to study. In this research, we utilized nanotopography to elucidate details regarding iPSC differentiation by developing a nanodot platform consisting of nanodot arrays of increasing diameter. Subjecting iPSCs cultured on the nanodot platform to a cardiomyocyte (CM) differentiation protocol revealed several significant gene expression profiles that were associated with poor differentiation. The observed expression trends were used to select existing small-molecule drugs capable of modulating differentiation efficiency. BRD K98 was repurposed to inhibit CM differentiation, while iPSCs treated with NSC-663284, carmofur, and KPT-330 all exhibited significant increases in not only CM marker expression but also spontaneous beating, suggesting improved CM differentiation. In addition, quantitative polymerase chain reaction was performed to determine the gene regulation responsible for modulating differentiation efficiency. Multiple genes involved in extracellular matrix remodeling were correlated with a CM differentiation efficiency, while genes involved in the cell cycle exhibited contrasting expression trends that warrant further studies. The results suggest that expression profiles determined via short time-series expression miner analysis of nanodot-cultured iPSC differentiation can not only reveal drugs capable of enhancing differentiation efficiency but also highlight crucial sets of genes related to processes such as extracellular matrix remodeling and the cell cycle that can be targeted for further investigation. Our findings confirm that the nanodot platform can be used to reveal complex mechanisms behind iPSC differentiation and could be an indispensable tool for optimizing iPSC technology for clinical applications.
Collapse
Affiliation(s)
- Men Yee Chiew
- Center
for Regenerative Medicine and Cellular Therapy, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan, ROC
- Department
of Biological Science and Technology, National
Yang Ming Chiao Tung University, Hsinchu 300, Taiwan, ROC
| | - Erick Wang
- Department
of Biological Science and Technology, National
Yang Ming Chiao Tung University, Hsinchu 300, Taiwan, ROC
- College
of Biological Science and Technology Industrial Ph. D. Program, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan, ROC
| | - Kuan-Chun Lan
- Center
for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8397, Japan
| | - Yan-Ren Lin
- Department
of Emergency and Critical Care Medicine, Changhua Christian Hospital, Changhua 500, Taiwan, ROC
- Department
of Post Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan, ROC
- School
of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, ROC
- School
of Medicine, Chung Shan Medical University, Taichung 402, Taiwan, ROC
| | - Yu-Huan Hsueh
- College
of Biological Science and Technology, National
Yang Ming Chiao Tung University, Hsinchu 300, Taiwan, ROC
- Department
of Orthopedic Surgery, E-Da Hospital, I-Shou
University, Kaohsiung 824, Taiwan
| | - Yuan-Kun Tu
- Department
of Orthopedic Surgery, E-Da Hospital, I-Shou
University, Kaohsiung 824, Taiwan
| | - Chu-Feng Liu
- Emergency Medicine Department, Kaohsiung
Chang Gung Memorial Hospital, Kaohsiung, 833, Taiwan,
ROC
- Ph. D. Degree Program of Biomedical Science
and Engineering, National Yang Ming Chiao
Tung University, Hsinchu 300, Taiwan, ROC
| | - Po-Chun Chen
- Institute of Materials Science and Engineering, National Taipei University of Technology, Taipei 106, Taiwan, ROC
| | - Huai-En Lu
- Center
for Regenerative Medicine and Cellular Therapy, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan, ROC
- Institute of Biochemistry and Molecular
Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan, ROC
- Bioresource
Collection and Research Center, Food Industry Research
and Development Institute, Hsinchu
City 300, Taiwan, ROC
| | - Wen Liang Chen
- Center
for Regenerative Medicine and Cellular Therapy, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan, ROC
- Department
of Biological Science and Technology, National
Yang Ming Chiao Tung University, Hsinchu 300, Taiwan, ROC
- College
of Biological Science and Technology Industrial Ph. D. Program, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan, ROC
- Bioresource
Collection and Research Center, Food Industry Research
and Development Institute, Hsinchu
City 300, Taiwan, ROC
| |
Collapse
|
9
|
Ma X, Peddibhotla S, Zheng Y, Pan S, Mehta A, Moroni DG, Chen QY, Ma X, Burnett JC, Malany S, Sangaralingham SJ. Discovery of small molecule guanylyl cyclase B receptor positive allosteric modulators. PNAS NEXUS 2024; 3:pgae225. [PMID: 38894878 PMCID: PMC11185183 DOI: 10.1093/pnasnexus/pgae225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
Myocardial fibrosis is a pathological hallmark of cardiovascular disease (CVD), and excessive fibrosis can lead to new-onset heart failure and increased mortality. Currently, pharmacological therapies for myocardial fibrosis are limited, highlighting the need for novel therapeutic approaches. The particulate guanylyl cyclase B (GC-B) receptor possesses beneficial antifibrotic actions through the binding of its natural ligand C-type natriuretic peptide (CNP) and the generation of the intracellular second messenger, cyclic guanosine 3',5'-monophosphate (cGMP). These actions include the suppression of fibroblast proliferation and reduction in collagen synthesis. With its abundant expression on fibroblasts, the GC-B receptor has emerged as a key molecular target for innovative CVD therapeutics. However, small molecules that can bind and potentiate the GC-B/cGMP pathway have yet to be discovered. From a cell-based high-throughput screening initiative of the NIH Molecular Libraries Small Molecule Repository and hit-to-lead evolution based on a series of structure-activity relationships, we report the successful discovery of MCUF-42, a GC-B-targeted small molecule that acts as a positive allosteric modulator (PAM). Studies herein support MCUF-42's ability to enhance the binding affinity between GC-B and CNP. Moreover, MCUF-42 potentiated cGMP levels induced by CNP in human cardiac fibroblasts (HCFs) and notably also enhanced the inhibitory effect of CNP on HCF proliferation. Together, our findings highlight that MCUF-42 is a small molecule that can modulate the GC-B/cGMP signaling pathway, potentially enhancing the antifibrotic actions of CNP. Thus, these data underscore the continued development of GC-B small molecule PAMs as a novel therapeutic strategy for targeting cardiac fibrosis and CVD.
Collapse
Affiliation(s)
- Xiao Ma
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Ye Zheng
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Shuchong Pan
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Alka Mehta
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Dante G Moroni
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Qi-Yin Chen
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32610, USA
| | - Xiaoyu Ma
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Siobhan Malany
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
10
|
Yao J, Chen Y, Huang Y, Sun X, Shi X. The role of cardiac microenvironment in cardiovascular diseases: implications for therapy. Hum Cell 2024; 37:607-624. [PMID: 38498133 DOI: 10.1007/s13577-024-01052-3] [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: 11/29/2023] [Accepted: 02/25/2024] [Indexed: 03/20/2024]
Abstract
Due to aging populations and changes in lifestyle, cardiovascular diseases including cardiomyopathy, hypertension, and atherosclerosis, are the leading causes of death worldwide. The heart is a complicated organ composed of multicellular types, including cardiomyocytes, fibroblasts, endothelial cells, vascular smooth muscle cells, and immune cells. Cellular specialization and complex interplay between different cell types are crucial for the cardiac tissue homeostasis and coordinated function of the heart. Mounting studies have demonstrated that dysfunctional cells and disordered cardiac microenvironment are closely associated with the pathogenesis of various cardiovascular diseases. In this paper, we discuss the composition and the homeostasis of cardiac tissues, and focus on the role of cardiac environment and underlying molecular mechanisms in various cardiovascular diseases. Besides, we elucidate the novel treatment for cardiovascular diseases, including stem cell therapy and targeted therapy. Clarification of these issues may provide novel insights into the prevention and potential targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuejun Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuqing Huang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| |
Collapse
|
11
|
Yan T, Song S, Sun W, Ge Y. HAPLN1 knockdown inhibits heart failure development via activating the PKA signaling pathway. BMC Cardiovasc Disord 2024; 24:197. [PMID: 38580957 PMCID: PMC10996236 DOI: 10.1186/s12872-024-03861-8] [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: 10/08/2023] [Accepted: 03/26/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a heterogeneous syndrome that affects millions worldwide, resulting in substantial health and economic burdens. However, the molecular mechanism of HF pathogenesis remains unclear. METHODS HF-related key genes were screened by a bioinformatics approach.The impacts of HAPLN1 knockdown on Angiotensin II (Ang II)-induced AC16 cells were assessed through a series of cell function experiments. Enzyme-linked immunosorbent assay (ELISA) was used to measure levels of oxidative stress and apoptosis-related factors. The HF rat model was induced by subcutaneous injection isoprenaline and histopathologic changes in the cardiac tissue were assessed by hematoxylin and eosin (HE) staining and echocardiographic index. Downstream pathways regulated by HAPLN1 was predicted through bioinformatics and then confirmed in vivo and in vitro by western blot. RESULTS Six hub genes were screened, of which HAPLN1, FMOD, NPPB, NPPA, and COMP were overexpressed, whereas NPPC was downregulated in HF. Further research found that silencing HAPLN1 promoted cell viability and reduced apoptosis in Ang II-induced AC16 cells. HAPLN1 knockdown promoted left ventricular ejection fraction (LVEF) and left ventricular fraction shortening (LVFS), while decreasing left ventricular end-systolic volume (LVESV) in the HF rat model. HAPLN1 knockdown promoted the levels of GSH and suppressed the levels of MDA, LDH, TNF-α, and IL-6. Mechanistically, silencing HAPLN1 activated the PKA pathway, which were confirmed both in vivo and in vitro. CONCLUSION HAPLN1 knockdown inhibited the progression of HF by activating the PKA pathway, which may provide novel perspectives on the management of HF.
Collapse
Affiliation(s)
- Tao Yan
- Department of Cardiology, Zibo Municipal Hospital, Ward 1, No. 139 Huangong Road, Linzi District, Zibo City, Shandong Province, 255400, China
| | - Shushuai Song
- Department of Cardiology, Qingdao Fuwai Cardiovascular Hospital, No. 201 Nanjing Road, Shibei District, Qingdao City, Shandong Province, 266034, China
| | - Wendong Sun
- Department of Cardiology, Zibo Municipal Hospital, No. 139 Huangong Road, Linzi District, Zibo City, Shandong Province, 255400, China
| | - Yiping Ge
- Department of Cardiology, Qingdao Fuwai Cardiovascular Hospital, No. 201 Nanjing Road, Shibei District, Qingdao City, Shandong Province, 266034, China.
| |
Collapse
|
12
|
Yang Q, Yang Q, Wu X, Zheng R, Lin H, Wang S, Joseph J, Sun YV, Li M, Wang T, Zhao Z, Xu M, Lu J, Chen Y, Ning G, Wang W, Bi Y, Zheng J, Xu Y. Sex-stratified genome-wide association and transcriptome-wide Mendelian randomization studies reveal drug targets of heart failure. Cell Rep Med 2024; 5:101382. [PMID: 38237596 PMCID: PMC10897518 DOI: 10.1016/j.xcrm.2023.101382] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 02/23/2024]
Abstract
The prevalence of heart failure (HF) subtypes, which are classified by left ventricular ejection fraction (LVEF), demonstrate significant sex differences. Here, we perform sex-stratified genome-wide association studies (GWASs) on LVEF and transcriptome-wide Mendelian randomization (MR) on LVEF, all-cause HF, HF with reduced ejection fraction (HFrEF), and HF with preserved ejection fraction (HFpEF). The sex-stratified GWASs of LVEF identified three sex-specific loci that were exclusively detected in the sex-stratified GWASs. Three drug target genes show sex-differential effects on HF/HFrEF via influencing LVEF, with NPR2 as the target gene for the HF drug Cenderitide under phase 2 clinical trial. Our study highlights the importance of considering sex-differential genetic effects in sex-balanced diseases such as HF and emphasizes the value of sex-stratified GWASs and MR in identifying putative genetic variants, causal genes, and candidate drug targets for HF, which is not identifiable using a sex-combined strategy.
Collapse
Affiliation(s)
- Qianqian Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qian Yang
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - Xueyan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jacob Joseph
- Cardiology Section, VA Providence Healthcare System, 830 Chalkstone Avenue, Providence, RI 02908, USA; Department of Medicine, Warren Alpert Medical School of Brown University, 222 Richmond Street, Providence, RI 02903, USA
| | - Yan V Sun
- Emory University Rollins School of Public Health, Atlanta, GA, USA; Atlanta VA Health Care System, Decatur, GA, USA
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Ensho T, Hino J, Ueda Y, Miyazato M, Iwakura H. Vascular endothelial cell-specific overexpression of CNP did not improve liver fibrosis in HFFCD-induced NASH, but did improve renal lesions. Peptides 2024; 172:171146. [PMID: 38157939 DOI: 10.1016/j.peptides.2023.171146] [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: 11/09/2023] [Revised: 12/15/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Mice with endothelial-cell-specific overexpression of C-type natriuretic peptide (E-CNP Tg mice) were shown to be protected against hepatic fibrosis and inflammation induced by high fat diet (HFD) feeding, with improved insulin sensitivity and attenuated weight gain. A recently developed high-fat, high-fructose, high-cholesterol diet (HFFCD) is considered to be a superior model to HFD, owing to the resemblance to human non-alcoholic steatohepatitis (NASH). In this study, we therefore aimed to reveal whether these previous findings with E-CNP Tg mice on HFD can be observed in a newly developed NASH model. Patients with NASH have been suggested to be at higher risk of developing chronic kidney disease, so we also assessed the kidney histology of these mice. After 8 months of HFFCD feeding, the livers of E-CNP Tg mice and controls showed progressive fibrosis, which resembled the features of human NASH. However, no significant differences were observed in NAFLD activity scores between E-CNP Tg mice and controls, although there was a tendency for improvement in E-CNP Tg mice. The reduced levels of GCB, a receptor for CNP, may have weakened the action of CNP in the current model. In the kidneys, HFFCD showed glomerular hypertrophy and tubular atrophy in the cortical region, which were suppressed in E-CNP Tg mice. The present study did not prove the therapeutic effect of CNP on NASH in the HFFCD model, but provided evidence of its potential beneficial effects on NASH-associated renal damage.
Collapse
Affiliation(s)
- Takuya Ensho
- Department of Pharmacotherapeutics, School of Pharmaceutical Science, Wakayama Medical University, Wakayama, Japan
| | - Jun Hino
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Yoko Ueda
- Department of Pharmacotherapeutics, School of Pharmaceutical Science, Wakayama Medical University, Wakayama, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Hiroshi Iwakura
- Department of Pharmacotherapeutics, School of Pharmaceutical Science, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
14
|
Dhanusu Sivakalai S, Sowndhar Rajan B, Vellaichamy E. C-type natriuretic peptide (CNP) inhibits 7,12-Dimethylbenz[a]anthracene (DMBA)/Croton oil-induced skin tumor growth by modulating inflammation in Swiss albino mice. J Biochem Mol Toxicol 2023; 37:e23423. [PMID: 37352108 DOI: 10.1002/jbt.23423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 04/14/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023]
Abstract
C-type natriuretic peptide (CNP) exhibits anti-inflammatory activity besides its natriuretic and diuretic functions. The present study aimed to determine the anticancer and synergistic therapeutic activity of CNP against a 7,12-Dimethylbenz[a]anthracene (DMBA)/Croton oil-induced skin tumor mouse model. CNP (2.5 µg/kg body weight) was injected either alone and/or in combination with Cisplatin (CDDP) (2 mg/kg body weight) for 4 weeks. The dorsal skin tumor incidences/growth and mortality rate were recorded during the experimental period of 16 weeks. The serum C-reactive protein (CRP), and lactate dehydrogenase (LDH) levels, infiltrating mast cells, and AgNORs proliferating cells count were analyzed in control and experimental mice. Further, the expression profile of marker genes of proliferation, inflammation, and progression molecules were analyzed using Reverse transcriptase-polymerase chain reaction (RT-PCR)/quantitative PCR (qPCR), western blot, and immunohistochemistry. The DMBA/Croton oil-induced mice exhibited 100% tumor incidence. Whereas, CNP alone, CDDP alone, and CNP+CDDP combination-treated mice exhibited 58%, 46%, and 24% tumor incidence, respectively. Also, a marked reduction in the levels of serum CRP and LDH, the number of infiltrating mast cells count and AgNORs proliferating cells count were noticed in the mice skin sections. Further, a significant reduction in both mRNA and protein expression levels of proliferation, inflammation, and progression markers were noticed in CNP (p < 0.01), CDDP (p < 0.01), and CNP+CDDP combination (p < 0.001) treated mice, respectively. The results of the present study suggest that CNP has anticancer activity. Further, the CNP+CDDP treatment has more promising anticancer activity as compared with CNP or CDDP alone treatment, probably due to the synergistic antiproliferative and anti-inflammatory activities of CNP and CDDP.
Collapse
Affiliation(s)
- Suresh Dhanusu Sivakalai
- Peptide Research and Molecular Cardiology Laboratory, Department of Biochemistry, Guindy Campus, University of Madras, Chennai, Tamil Nadu, India
| | - Boopathi Sowndhar Rajan
- Peptide Research and Molecular Cardiology Laboratory, Department of Biochemistry, Guindy Campus, University of Madras, Chennai, Tamil Nadu, India
| | - Elangovan Vellaichamy
- Peptide Research and Molecular Cardiology Laboratory, Department of Biochemistry, Guindy Campus, University of Madras, Chennai, Tamil Nadu, India
| |
Collapse
|
15
|
Bachmann JC, Kirchhoff JE, Napolitano JE, Sorota S, Gordon WM, Feric N, Aschar‐Sobbi R, Lv J, Cao Z, Coppieters K, Borghetti G, Nyberg M. C-type natriuretic peptide induces inotropic and lusitropic effects in human 3D-engineered cardiac tissue: Implications for the regulation of cardiac function in humans. Exp Physiol 2023; 108:1172-1188. [PMID: 37493451 PMCID: PMC10988518 DOI: 10.1113/ep091303] [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: 05/17/2023] [Accepted: 07/06/2023] [Indexed: 07/27/2023]
Abstract
The role of C-type natriuretic peptide (CNP) in the regulation of cardiac function in humans remains to be established as previous investigations have been confined to animal model systems. Here, we used well-characterized engineered cardiac tissues (ECTs) generated from human stem cell-derived cardiomyocytes and fibroblasts to study the acute effects of CNP on contractility. Application of CNP elicited a positive inotropic response as evidenced by increases in maximum twitch amplitude, maximum contraction slope and maximum calcium amplitude. This inotropic response was accompanied by a positive lusitropic response as demonstrated by reductions in time from peak contraction to 90% of relaxation and time from peak calcium transient to 90% of decay that paralleled increases in maximum contraction decay slope and maximum calcium decay slope. To establish translatability, CNP-induced changes in contractility were also assessed in rat ex vivo (isolated heart) and in vivo models. Here, the effects on force kinetics observed in ECTs mirrored those observed in both the ex vivo and in vivo model systems, whereas the increase in maximal force generation with CNP application was only detected in ECTs. In conclusion, CNP induces a positive inotropic and lusitropic response in ECTs, thus supporting an important role for CNP in the regulation of human cardiac function. The high degree of translatability between ECTs, ex vivo and in vivo models further supports a regulatory role for CNP and expands the current understanding of the translational value of human ECTs. NEW FINDINGS: What is the central question of this study? What are the acute responses to C-type natriuretic peptide (CNP) in human-engineered cardiac tissues (ECTs) on cardiac function and how well do they translate to matched concentrations in animal ex vivo and in vivo models? What is the main finding and its importance? Acute stimulation of ECTs with CNP induced positive lusitropic and inotropic effects on cardiac contractility, which closely reflected the changes observed in rat ex vivo and in vivo cardiac models. These findings support an important role for CNP in the regulation of human cardiac function and highlight the translational value of ECTs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Juan Lv
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| | - Zhiyou Cao
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| | - Ken Coppieters
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| | | | - Michael Nyberg
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| |
Collapse
|
16
|
Sarzani R, Allevi M, Di Pentima C, Schiavi P, Spannella F, Giulietti F. Role of Cardiac Natriuretic Peptides in Heart Structure and Function. Int J Mol Sci 2022; 23:ijms232214415. [PMID: 36430893 PMCID: PMC9697447 DOI: 10.3390/ijms232214415] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Cardiac natriuretic peptides (NPs), atrial NP (ANP) and B-type NP (BNP) are true hormones produced and released by cardiomyocytes, exerting several systemic effects. Together with C-type NP (CNP), mainly expressed by endothelial cells, they also exert several paracrine and autocrine activities on the heart itself, contributing to cardiovascular (CV) health. In addition to their natriuretic, vasorelaxant, metabolic and antiproliferative systemic properties, NPs prevent cardiac hypertrophy, fibrosis, arrhythmias and cardiomyopathies, counteracting the development and progression of heart failure (HF). Moreover, recent studies revealed that a protein structurally similar to NPs mainly produced by skeletal muscles and osteoblasts called musclin/osteocrin is able to interact with the NPs clearance receptor, attenuating cardiac dysfunction and myocardial fibrosis and promoting heart protection during pathological overload. This narrative review is focused on the direct activities of this molecule family on the heart, reporting both experimental and human studies that are clinically relevant for physicians.
Collapse
Affiliation(s)
- Riccardo Sarzani
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
- Correspondence: (R.S.); Tel.: +39-071-5964696
| | - Massimiliano Allevi
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Chiara Di Pentima
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
| | - Paola Schiavi
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Francesco Spannella
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Federico Giulietti
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
| |
Collapse
|
17
|
Tokudome T, Otani K. Molecular Mechanism of Blood Pressure Regulation through the Atrial Natriuretic Peptide. BIOLOGY 2022; 11:biology11091351. [PMID: 36138830 PMCID: PMC9495342 DOI: 10.3390/biology11091351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/09/2022] [Accepted: 09/10/2022] [Indexed: 11/24/2022]
Abstract
Simple Summary Atrial natriuretic peptide (ANP) is a cardiac peptide hormone that was identified by Kangawa and Matsuo in 1984. In Japan, ANP has been used as an intravenous drug for the treatment of acute heart failure since 1995. Because ANP has a hypotensive effect, it is important to avoid excessive lowering of blood pressure when ANP is used. Recently, a compound that inhibits neutral endopeptidase, the enzyme that degrades ANP, has been developed (angiotensin receptor-neprilysin inhibitor (ARNI)). ARNI has been approved worldwide for the treatment of chronic heart failure and has been authorized in Japan as an antihypertensive drug. However, it is not understood exactly how ANP exerts its hypotensive effect. In this review, we discuss the molecular mechanism of the blood pressure-regulating effects of ANP, focusing on our recent findings. Abstract Natriuretic peptides, including atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), have cardioprotective effects and regulate blood pressure in mammals. ANP and BNP are hormones secreted from the heart into the bloodstream in response to increased preload and afterload. Both hormones act through natriuretic peptide receptor 1 (NPR1). In contrast, CNP acts through natriuretic peptide receptor 2 (NPR2) and was found to be produced by the vascular endothelium, chondrocytes, and cardiac fibroblasts. Based on its relatively low plasma concentration compared with ANP and BNP, CNP is thought to function as both an autocrine and a paracrine factor in the vasculature, bone, and heart. The cytoplasmic domains of both NPR1 and NPR2 display a guanylate cyclase activity that catalyzes the formation of cyclic GMP. NPR3 lacks this guanylate cyclase activity and is reportedly coupled to Gi-dependent signaling. Recently, we reported that the continuous infusion of the peptide osteocrin, an endogenous ligand of NPR3 secreted by bone and muscle cells, lowered blood pressure in wild-type mice, suggesting that endogenous natriuretic peptides play major roles in the regulation of blood pressure. Neprilysin is a neutral endopeptidase that degrades several vasoactive peptides, including natriuretic peptides. The increased worldwide clinical use of the angiotensin receptor-neprilysin inhibitor for the treatment of chronic heart failure has brought renewed attention to the physiological effects of natriuretic peptides. In this review, we provide an overview of the discovery of ANP and its translational research. We also highlight our recent findings on the blood pressure regulatory effects of ANP, focusing on its molecular mechanisms.
Collapse
Affiliation(s)
- Takeshi Tokudome
- Department of Pathophysiology of Heart Failure and Therapeutics, National Cerebral and Cardiovascular Center Research Institute, Suita 564-8565, Japan
- Correspondence: ; Tel.: +81-6-6170-1069
| | - Kentaro Otani
- Center for Regenerative Medicine, National Cerebral and Cardiovascular Center Research Institute, Suita 564-8565, Japan
| |
Collapse
|
18
|
Leancă SA, Crișu D, Petriș AO, Afrăsânie I, Genes A, Costache AD, Tesloianu DN, Costache II. Left Ventricular Remodeling after Myocardial Infarction: From Physiopathology to Treatment. Life (Basel) 2022; 12:1111. [PMID: 35892913 PMCID: PMC9332014 DOI: 10.3390/life12081111] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/11/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of death and morbidity worldwide, with an incidence relatively high in developed countries and rapidly growing in developing countries. The most common cause of MI is the rupture of an atherosclerotic plaque with subsequent thrombotic occlusion in the coronary circulation. This causes cardiomyocyte death and myocardial necrosis, with subsequent inflammation and fibrosis. Current therapies aim to restore coronary flow by thrombus dissolution with pharmaceutical treatment and/or intravascular stent implantation and to counteract neurohormonal activation. Despite these therapies, the injury caused by myocardial ischemia leads to left ventricular remodeling; this process involves changes in cardiac geometry, dimension and function and eventually progression to heart failure (HF). This review describes the pathophysiological mechanism that leads to cardiac remodeling and the therapeutic strategies with a role in slowing the progression of remodeling and improving cardiac structure and function.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Daniela Crișu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antoniu Octavian Petriș
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| | - Irina Afrăsânie
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antonia Genes
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Dan Nicolae Tesloianu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Irina Iuliana Costache
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| |
Collapse
|
19
|
CNP, the Third Natriuretic Peptide: Its Biology and Significance to the Cardiovascular System. BIOLOGY 2022; 11:biology11070986. [PMID: 36101368 PMCID: PMC9312265 DOI: 10.3390/biology11070986] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 11/19/2022]
Abstract
Simple Summary CNP is the third natriuretic peptide to be isolated and is widely expressed in the central nervous system, osteochondral system, and vascular system. The receptor that is mainly targeted by CNP is GC-B, which differs from GC-A, the receptor targeted by the other two natriuretic peptides, ANP and BNP. Consequently, the actions of CNP differ somewhat from those of ANP and BNP. Research into the actions of CNP has shown that CNP attenuates cardiac remodeling in animal models of cardiac hypertrophy, myocardial infarction, and myocarditis. Studies examining CNP/GC-B signaling showed that it contributes to the prevention of cardiac stiffness. Endogenous CNP, perhaps acting in part through CNP/NPR-C signaling, contributes to the regulation of vascular function and blood pressure. CNP regulates vascular remodeling and angiogenesis via CNP/GC-B/CGK signaling. CNP attenuates interstitial fibrosis and fibrosis-related gene expression in pressure overload and myocardial infarction models. The clinical application of CNP as a therapeutic agent for cardiovascular diseases is anticipated. Abstract The natriuretic peptide family consists of three biologically active peptides: ANP, BNP, and CNP. CNP is more widely expressed than the other two peptides, with significant levels in the central nervous system, osteochondral system, and vascular system. The receptor that is mainly targeted by CNP is GC-B, which differs from GC-A, the receptor targeted by ANP and BNP. Consequently, the actions of CNP differ somewhat from those of ANP and BNP. CNP knockout leads to severe dwarfism, and there has been important research into the role of CNP in the osteochondral system. As a result, a CNP analog is now available for clinical use in patients with achondroplasia. In the cardiovascular system, CNP and its downstream signaling are involved in the regulatory mechanisms underlying myocardial remodeling, cardiac function, vascular tone, angiogenesis, and fibrosis, among others. This review focuses on the roles of CNP in the cardiovascular system and considers its potential for clinical application in the treatment of cardiovascular diseases.
Collapse
|
20
|
Physiological and Pathophysiological Effects of C-Type Natriuretic Peptide on the Heart. BIOLOGY 2022; 11:biology11060911. [PMID: 35741432 PMCID: PMC9219612 DOI: 10.3390/biology11060911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 01/06/2023]
Abstract
Simple Summary C-type natriuretic peptide (CNP) is the third member of the natriuretic peptide family. Unlike atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), CNP was not previously regarded as an important cardiac modulator. However, recent studies have revealed the physiological and pathophysiological importance of CNP in the heart; in concert with its cognate natriuretic peptide receptor-B (NPR-B), CNP has come to be regarded as the major heart-protective natriuretic peptide in the failed heart. In this review, I introduce the history of research on CNP in the cardiac field. Abstract C-type natriuretic peptide (CNP) is the third member of the natriuretic peptide family. Unlike other members, i.e., atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), which are cardiac hormones secreted from the atrium and ventricle of the heart, respectively, CNP is regarded as an autocrine/paracrine regulator with broad expression in the body. Because of its low expression levels compared to ANP and BNP, early studies failed to show its existence and role in the heart. However, recent studies have revealed the physiological and pathophysiological importance of CNP in the heart; in concert with the distribution of its specific natriuretic peptide receptor-B (NPR-B), CNP has come to be regarded as the major heart-protective natriuretic peptide in the failed heart. NPR-B generates intracellular cyclic guanosine 3′,5′-monophosphate (cGMP) upon CNP binding, followed by various molecular effects including the activation of cGMP-dependent protein kinases, which generates diverse cytoprotective actions in cardiomyocytes, as well as in cardiac fibroblasts. CNP exerts negative inotropic and positive lusitropic responses in both normal and failing heart models. Furthermore, osteocrin, the intrinsic and specific ligand for the clearance receptor for natriuretic peptides, can augment the effects of CNP and may supply a novel therapeutic strategy for cardiac protection.
Collapse
|
21
|
C-Type Natriuretic Peptide (CNP) Induces Cell Death and Sensitizes the Effect of Cisplatin in Human Non-small Cell Lung Cancer Cells (A549). Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10420-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
22
|
Hu P, Chen H, Wang LH, Jiang JB, Li JM, Tang MY, Guo YC, Zhu QF, Pu ZX, Lin XP, Ng S, Liu XB, Wang JA. Elevated N-terminal pro C-type natriuretic peptide is associated with mortality in patients undergoing transcatheter aortic valve replacement. BMC Cardiovasc Disord 2022; 22:164. [PMID: 35413789 PMCID: PMC9004019 DOI: 10.1186/s12872-022-02615-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/01/2022] [Indexed: 12/02/2022] Open
Abstract
Background Unlike N-terminal pro-B-type natriuretic peptide (NT-proBNP), which have been extensively studied, little is known about the role of N-terminal pro-C-type natriuretic peptide (NT-proCNP) for predicting survival post transcatheter aortic valve replacement (TAVR). Methods A total of 309 patients were included in the analysis. Patients were grouped into quartiles (Q1–4) according to the baseline NT-proCNP value. Blood for NT-proCNP analysis was obtained prior to TAVR procedure. The primary endpoint was mortality after a median follow-up of 32 months. Multivariable Cox proportional hazards regression models analyzed prognostic factors. The predictive capability was compared between NT-proBNP and NT-proCNP using receiver operator curve (ROC) analysis. Results A total of 309 subjects with the mean age of 76.8 ± 6.3 years, among whom 58.6% were male, were included in the analysis. A total of 58 (18.8%) patients died during follow-up. Cox multivariable analyses indicated society of thoracic surgeons (STS)-score was a strong independent predictor for mortality (hazard ratio (HR) 1.08, 95% confidential interval (CI) 1.05–1.12, P < 0.001). Elevated NT-proCNP was associated with a higher risk of cardiovascular mortality (HR 1.02, 95% CI 1.00–1.03, P = 0.025) and All-cause mortality (HR 1.01, 95% CI 1.00–1.03, P = 0.027), whereas NT-proBNP showed a small effect size on mortality. ROC analysis indicated that NT-proCNP was superior to NT-proBNP for TAVR risk evaluation in patients with left ventricular ejection fraction (LVEF) < 50% [(Area under the curve (AUC)-values of 0.79 (0.69; 0.87) vs. 0.59 (0.48; 0.69), P = 0.0453]. Conclusions NT-proCNP and STS-Score were the independent prognostic factors of mortality among TAVR patients. Furthermore, NT-proCNP was superior to NT-proBNP for TAVR risk evaluation in patients with LVEF < 50%. Trial registration NCT02803294, 16/06/2016. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02615-8.
Collapse
Affiliation(s)
- Po Hu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Han Chen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Li-Han Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Ju-Bo Jiang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Jia-Min Li
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China
| | - Meng-Yao Tang
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yu-Chao Guo
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Qi-Feng Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Zhao-Xia Pu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Department of Echocardiography, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Xin-Ping Lin
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Department of Echocardiography, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Stella Ng
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China
| | - Xian-Bao Liu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China. .,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China.
| | - Jian-An Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China. .,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
23
|
|
24
|
Rukavina Mikusic NL, Kouyoumdzian NM, Puyó AM, Fernández BE, Choi MR. Role of natriuretic peptides in the cardiovascular-adipose communication: a tale of two organs. Pflugers Arch 2022; 474:5-19. [PMID: 34173888 DOI: 10.1007/s00424-021-02596-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 05/31/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022]
Abstract
Natriuretic peptides have long been known for their cardiovascular function. However, a growing body of evidence emphasizes the role of natriuretic peptides in the energy metabolism of several substrates in humans and animals, thus interrelating the heart, as an endocrine organ, with various insulin-sensitive tissues and organs such as adipose tissue, muscle skeletal, and liver. Adipose tissue dysfunction is associated with altered regulation of the natriuretic peptide system, also indicated as a natriuretic disability. Evidence points to a contribution of this natriuretic disability to the development of obesity, type 2 diabetes mellitus, and cardiometabolic complications; although the causal relationship is not fully understood at present. However, targeting the natriuretic peptide pathway may improve metabolic health in obesity and type 2 diabetes mellitus. This review will focus on the current literature on the metabolic functions of natriuretic peptides with emphasis on lipid metabolism and insulin sensitivity. Natriuretic peptide system alterations could be proposed as one of the linking mechanisms between adipose tissue dysfunction and cardiovascular disease.
Collapse
Affiliation(s)
- Natalia Lucía Rukavina Mikusic
- Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Nicolás Martín Kouyoumdzian
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana María Puyó
- Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Marcelo Roberto Choi
- Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto Universitario de Ciencias de la Salud, Fundación H.A. Barceló, Buenos Aires, Argentina
| |
Collapse
|
25
|
Vitiello A, Ferrara F. Pharmacological agents modifying the renin angiotensin and natriuretic peptide systems in COVID-19 patients. Wien Klin Wochenschr 2021; 133:983-988. [PMID: 33877436 PMCID: PMC8055751 DOI: 10.1007/s00508-021-01855-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/18/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Antonio Vitiello
- Pharmaceutical Department, Usl Umbria 1, A. Migliorati street, 06132 Perugia, Italy
| | - Francesco Ferrara
- Pharmaceutical Department, Usl Umbria 1, A. Migliorati street, 06132 Perugia, Italy
| |
Collapse
|
26
|
Prognostic Value of Urinary and Plasma C-Type Natriuretic Peptide in Acute Decompensated Heart Failure. JACC-HEART FAILURE 2021; 9:613-623. [PMID: 34246604 DOI: 10.1016/j.jchf.2021.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVES This study sought to characterize urinary and plasma C-type natriuretic peptide (CNP) in acute decompensated heart failure (ADHF) to define their relationship with clinical variables and to determine whether urinary and plasma CNP together add prognostic value. BACKGROUND CNP is a protective hormone that is synthesized in the kidney and endothelium and possesses antiremodeling properties. Urinary and plasma CNP levels are elevated in pathophysiological conditions; however, their regulation and prognostic value in heart failure (HF) is unclear. METHODS Urinary and plasma CNP were measured in 109 healthy subjects and 208 patients with ADHF; the 95th percentile of CNP values from healthy subjects established the normal contemporary cutoffs. Patients with ADHF were stratified based on urinary and plasma CNP levels for clinical characterization and the assessment of risk for adverse outcomes. RESULTS There was no significant correlation between urinary and plasma CNP in both cohorts. Urinary and plasma CNP were significantly elevated in patients with ADHF, and both increased with disease severity and were positively correlated with plasma N-terminal pro-B-type natriuretic peptide (NT-proBNP). Of the patients with ADHF, 23% had elevations in both urinary and plasma CNP, whereas 24% had normal CNP levels. During a median follow-up of 3 years, patients with elevated urinary and plasma CNP had a significantly higher risk of rehospitalization and/or death (HR: 1.79; P = 0.03) and rehospitalization (HR: 2.16; P = 0.01) after adjusting for age, sex, left ventricular ejection fraction, renal function, and plasma NT-proBNP. The C-statistic and integrated discrimination analyses further supported that the addition of urinary and plasma CNP to established risk models improved the prediction of adverse outcomes in patients with ADHF. CONCLUSIONS Urinary and plasma CNP are differentially regulated in ADHF, and elevations in both provided independent prognostic value for predicting adverse outcomes.
Collapse
|
27
|
Cardiac morphological and functional changes induced by C-type natriuretic peptide are different in normotensive and spontaneously hypertensive rats. J Hypertens 2021; 38:2305-2317. [PMID: 32649642 DOI: 10.1097/hjh.0000000000002570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Inflammation and fibrosis are key mechanisms in cardiovascular remodeling. C-type natriuretic peptide (CNP) is an endothelium-derived factor with a cardiovascular protective role, although its in-vivo effect on cardiac remodeling linked to hypertension has not been investigated. The aim of this study was to determine the effects of chronic administration of CNP on inflammatory and fibrotic cardiac mechanisms in normotensive Wistar rats and spontaneously hypertensive rats (SHR). METHODS Twelve-week-old male SHR and normotensive rats were infused with CNP (0.75 μg/h/100 g) or isotonic saline (NaCl 0.9%) for 14 days (subcutaneous micro-osmotic pumps). Echocardiograms and electrocardiograms were performed, and SBP was measured. After treatment, transforming growth factor-beta 1, Smad proteins, tumor necrosis factor-alpha, interleukin-1 and interleukin-6, nitric oxide (NO) system and 2-thiobarbituric acid-reactive substances were evaluated in left ventricle. Histological studies were also performed. RESULTS SHR showed lower cardiac output with signs of fibrosis and hypertrophy in left ventricle, higher NO-system activity and more oxidative damage, as well as higher pro-inflammatory and pro-fibrotic markers than normotensive rats. Chronic CNP treatment-attenuated hypertension and ventricular hypertrophy in SHR, with no changes in normotensive rats. In left ventricle, CNP induced an anti-inflammatory and antifibrotic response, decreasing both pro-fibrotic and pro-inflammatory cytokines in SHR. In addition, CNP reduced oxidative damage as well as collagen content, and upregulated the NO system in both groups. CONCLUSION Chronic CNP treatment appears to attenuate hypertension and associated end-organ damage in the heart by reducing inflammation and fibrosis.
Collapse
|
28
|
Lukowski R, Cruz Santos M, Kuret A, Ruth P. cGMP and mitochondrial K + channels-Compartmentalized but closely connected in cardioprotection. Br J Pharmacol 2021; 179:2344-2360. [PMID: 33991427 DOI: 10.1111/bph.15536] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 01/01/2023] Open
Abstract
The 3',5'-cGMP pathway triggers cytoprotective responses and improves cardiomyocyte survival during myocardial ischaemia and reperfusion (I/R) injury. These beneficial effects were attributed to NO-sensitive GC induced cGMP production leading to activation of cGMP-dependent protein kinase I (cGKI). cGKI in turn phosphorylates many substrates, which eventually facilitate opening of mitochondrial ATP-sensitive potassium channels (mitoKATP ) and Ca2+ -activated potassium channels of the BK type (mitoBK). Accordingly, agents activating mitoKATP or mitoBK provide protection against I/R-induced damages. Here, we provide an up-to-date summary of the infarct-limiting actions exhibited by the GC/cGMP axis and discuss how mitoKATP and mitoBK, which are present at the inner mitochondrial membrane, confer mito- and cytoprotective effects on cardiomyocytes exposed to I/R injury. In view of this, we believe that the functional connection between the cGMP cascade and mitoK+ channels should be exploited further as adjunct to reperfusion therapy in myocardial infarction.
Collapse
Affiliation(s)
- Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Melanie Cruz Santos
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Anna Kuret
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
29
|
Murphy SP, Prescott MF, Maisel AS, Butler J, Piña IL, Felker GM, Ward JH, Williamson KM, Camacho A, Kandanelly RR, Solomon SD, Januzzi JL. Association Between Angiotensin Receptor-Neprilysin Inhibition, Cardiovascular Biomarkers, and Cardiac Remodeling in Heart Failure With Reduced Ejection Fraction. Circ Heart Fail 2021; 14:e008410. [PMID: 33998243 DOI: 10.1161/circheartfailure.120.008410] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Sacubitril/valsartan (S/V) treatment is associated with reverse cardiac remodeling and reductions in biomarkers reflecting ventricular wall stress and myocardial injury, such as NT-proBNP (N-terminal pro-B-type natriuretic peptide), hs-cTnT (high-sensitivity cardiac troponin T), and soluble suppressor of tumorigenicity 2 (sST2). How longitudinal changes in these biomarkers analyzed collectively are associated with cardiac remodeling in patients with heart failure with reduced ejection fraction treated with S/V is uncertain. METHODS In a prospective study of S/V in patients with heart failure with reduced ejection fraction, this prespecified exploratory analysis included patients with serially collected biomarkers and echocardiographic measures of cardiac remodeling through 12 months of treatment. A multivariate latent growth curve model assessed associations between simultaneous changes in biomarkers and left ventricular ejection fraction and left atrial volume index. RESULTS Seven hundred fifteen out of 794 total study participants were included (mean age 65 years, 73% male). Mean baseline left ventricular ejection fraction and left atrial volume index were 29% and 40 mL/m2, respectively. Adjusted geometric mean baseline concentrations for biomarkers included NT-proBNP of 649 pg/mL, hs-cTnT of 15.9 ng/L, and sST2 of 24.7 ng/mL. Following initiation of S/V, circulating concentrations of NT-proBNP, hs-cTnT, and sST2 significantly decreased within 30 days and remained significantly different than baseline at all subsequent timepoints. From baseline to month 12, decreases in adjusted biomarker concentrations averaged -27.9% (95% CI, -35.1% to -20.7%; P<0.001) for NT-proBNP; -6.7% (95% CI, -8.8% to -4.7%; P<0.001) for hs-cTnT; and -1.6% (95% CI, -2.9% to -0.4%; P<0.001) for sST2. NT-proBNP concentrations were predictive of later changes in hs-cTnT. The magnitude of reductions in NT-proBNP and hs-cTnT concentrations associated with improvements in left ventricular ejection fraction and left atrial volume index. There was no association between changes in sST2 and changes in other measures. CONCLUSIONS Following initiation of S/V, NT-proBNP, hs-cTnT, and sST2 concentrations decreased significantly. Longitudinal changes in NT-proBNP and hs-cTnT together associated with left atrial and left ventricular reverse remodeling. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT02887183.
Collapse
Affiliation(s)
- Sean P Murphy
- Massachusetts General Hospital, Boston (S.P.M., A.C., R.R.K., J.L.J.)
| | | | - Alan S Maisel
- University of California, San Diego School of Medicine (A.S.M.)
| | - Javed Butler
- University of Mississippi Medical Center, Jackson (J.B.)
| | | | - G Michael Felker
- Duke University Medical Center and Duke Clinical Research Institute, Durham, NC (G.M.F.)
| | - Jonathan H Ward
- Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., J.H.W., K.M.W.)
| | | | - Alexander Camacho
- Massachusetts General Hospital, Boston (S.P.M., A.C., R.R.K., J.L.J.)
| | | | - Scott D Solomon
- Brigham and Women's Hospital, Boston, MA (S.D.S.).,Harvard Medical School, Boston, MA (S.D.S., J.L.J.)
| | - James L Januzzi
- Massachusetts General Hospital, Boston (S.P.M., A.C., R.R.K., J.L.J.).,Harvard Medical School, Boston, MA (S.D.S., J.L.J.).,Baim Institute for Clinical Research, Boston, MA (J.L.J.)
| |
Collapse
|
30
|
Kuwahara K. The natriuretic peptide system in heart failure: Diagnostic and therapeutic implications. Pharmacol Ther 2021; 227:107863. [PMID: 33894277 DOI: 10.1016/j.pharmthera.2021.107863] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
Natriuretic peptides, which are activated in heart failure, play an important cardioprotective role. The most notable of the cardioprotective natriuretic peptides are atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), which are abundantly expressed and secreted in the atrium and ventricles, respectively, and C-type natriuretic peptide (CNP), which is expressed mainly in the vasculature, central nervous system, and bone. ANP and BNP exhibit antagonistic effects against angiotensin II via diuretic/natriuretic actions, vasodilatory actions, and inhibition of aldosterone secretion, whereas CNP is involved in the regulation of vascular tone and blood pressure, among other roles. ANP and BNP are of particular interest with respect to heart failure, as their levels, most notably BNP and N-terminal proBNP-a cleavage product produced when proBNP is processed to mature BNP-are increased in patients with heart failure. Furthermore, the identification of natriuretic peptides as sensitive markers of cardiac load has driven significant research into their physiological roles in cardiovascular homeostasis and disease, as well as their potential use as both biomarkers and therapeutics. In this review, I discuss the physiological functions of the natriuretic peptide family, with a particular focus on the basic research that has led to our current understanding of its roles in maintaining cardiovascular homeostasis, and the pathophysiological implications for the onset and progression of heart failure. The clinical significance and potential of natriuretic peptides as diagnostic and/or therapeutic agents are also discussed.
Collapse
Affiliation(s)
- Koichiro Kuwahara
- Department of Cardiovascular Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| |
Collapse
|
31
|
An JS, Tsuji K, Onuma H, Araya N, Isono M, Hoshino T, Inomata K, Hino J, Miyazato M, Hosoda H, Kangawa K, Nakagawa Y, Katagiri H, Miyatake K, Sekiya I, Muneta T, Koga H. Inhibition of fibrotic changes in infrapatellar fat pad alleviates persistent pain and articular cartilage degeneration in monoiodoacetic acid-induced rat arthritis model. Osteoarthritis Cartilage 2021; 29:380-388. [PMID: 33388431 DOI: 10.1016/j.joca.2020.12.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 12/05/2020] [Accepted: 12/15/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE We have reported that fibrotic changes in infrapatellar fat pad (IFP) after acute joint inflammation are closely associated with persistent pain in rats. In this study, to examine the effects of anti-fibrotic treatment on persistent pain, we used C-type natriuretic peptides (CNP) at the recovery phase after acute joint inflammation. DESIGN Thirty-two male Wistar rats were used in this study. Monoiodoacetic acid (MIA) was injected intra-articularly to induce IFP fibrosis and persistent pain. CNP was injected after acute inflammatory phase in the same knee joint. Time-course pain-avoidance behavior tests and histological analyses were performed to examine the effects of CNP. RESULTS Histological evaluations indicated that intra-articular injection of CNP inhibited fibrotic changes in IFP after acute inflammation. Incapacitance tests indicated that MIA injection into rat knee joint quickly decreased the percent weight on ipsilateral limb. In the vehicle group, the decrease was maintained up to day 28, suggesting that pain persistence occurred after acute inflammation (Day 0/Day 28, Est Dif -8.15, CI -10.78∼-5.53, Linear mixed-effect model). In contrast, the pain was alleviated in the CNP group after day 14 (Day0/Day 14, -0.51, -2.62-1.59). In addition, we observed significant improvement in the degree of articular cartilage degeneration at day 14 in the CNP group (OARSI score: vehicle 16.14 ± 4.37 vs CNP 6.87 ± 3.44, P < 0.01; Wilcoxon rank sum test). CONCLUSION Fibrotic changes in IFP may play important roles in both persistent pain and articular cartilage degeneration.
Collapse
Affiliation(s)
- J-S An
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - K Tsuji
- Department of Cartilage Regeneration, Tokyo Medical and Dental University, Tokyo, Japan.
| | - H Onuma
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - N Araya
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - M Isono
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - T Hoshino
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - K Inomata
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - J Hino
- Department of Biochemistry, Japan; National Cerebral and Cardiovascular Center Research Institute, Japan.
| | - M Miyazato
- Department of Biochemistry, Japan; National Cerebral and Cardiovascular Center Research Institute, Japan.
| | - H Hosoda
- Department of Regenerative Medicine and Tissue Engineering, Japan; National Cerebral and Cardiovascular Center Research Institute, Japan.
| | - K Kangawa
- National Cerebral and Cardiovascular Center Research Institute, Japan.
| | - Y Nakagawa
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Department of Cartilage Regeneration, Tokyo Medical and Dental University, Tokyo, Japan.
| | - H Katagiri
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - K Miyatake
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - I Sekiya
- Center for Stem Cells and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - T Muneta
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - H Koga
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
32
|
Vitiello A, La Porta R, Ferrara F. Scientific hypothesis and rational pharmacological for the use of sacubitril/valsartan in cardiac damage caused by COVID-19. Med Hypotheses 2021; 147:110486. [PMID: 33460992 PMCID: PMC7788318 DOI: 10.1016/j.mehy.2021.110486] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/30/2020] [Accepted: 01/02/2021] [Indexed: 12/17/2022]
Abstract
On March 11, 2020 the World Health Organization (WHO) declared the state of global pandemic caused by the new SARS-CoV-2 (COVID-19). To date, no antivirals directed against SARS-CoV-2 or effective vaccines to combat the viral infection are available. Severe acute respiratory syndrome caused by SARS-CoV-2 is treated empirically with antivirals, anti-inflammatory, anticoagulants. The approval of an effective vaccine still takes time. In this state, it may be useful to find new therapeutic solutions from drugs already on the market. Recent hypotheses suggest that the use of AT-1 receptor antagonists (ARB) in combination with neprilisin inhibitors (NEPi) could indirectly provide clinical benefits to patients with SARS-CoV-2 and cardiac involvement. In this article we investigate and describe a possible innovative pharmacological approach for the treatment of the most severe stages of COVID-19 infection.
Collapse
Affiliation(s)
- Antonio Vitiello
- Pharmaceutical Department, Usl Umbria 1, XIV Settembre Street, 06132 Perugia, Italy.
| | - Raffaele La Porta
- Clinical Pathology, Asur Marche, Viale Guido Da Montefeltro, Urbino, Italy.
| | - Francesco Ferrara
- Pharmaceutical Department, Usl Umbria 1, A. Migliorati Street, 06132 Perugia, Italy.
| |
Collapse
|
33
|
Vitiello A, Ferrara F. Pharmacological agents to therapeutic treatment of cardiac injury caused by Covid-19. Life Sci 2020; 262:118510. [PMID: 32991879 PMCID: PMC7521881 DOI: 10.1016/j.lfs.2020.118510] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 01/08/2023]
Abstract
SARS-CoV-2 is responsible for the 2019 coronavirus disease (COVID-19), a global pandemic that began in March 2020 and is currently in progress. To date, COVID-19 has caused about 935,000 deaths in more than 200 countries. The respiratory system is most affected by injuries caused by COVID-19, but other organs may be involved, including the cardiovascular system. SARS-CoV-2 penetrates host cells through the angiotensin 2 conversion enzyme (ACE-2). ACE-2 is expressed not only in the lungs, but also in other organs, including the cardiovascular system. Several studies have found that a good percentage of patients with severe COVID-19 have cardiac lesions, including myocardial fibrosis, edema and pericarditis. Pathological remodeling of the extracellular matrix caused by viral infection leads to myocardial fibrotic lesions. These fibrotic scars can cause cardiac dysfunction, reducing the ejection fraction caused by the presence of stiffened myocardial matrix, or cardiac arrhythmias that cause an alteration in the electrical conduction system of the heart. These cardiac dysfunctions can cause death. It is therefore essential to identify cardiac involvement early in order to act with appropriate therapeutic treatments. In this review, we describe what is known about cardiac injury from COVID-19, highlighting effective pharmacological therapeutic solutions to combat cardiac injury, particularly cardiac fibrosis, caused by COVID-19.
Collapse
Affiliation(s)
- A Vitiello
- Pharmaceutical Department, Usl Umbria 1, A. Migliorati street, 06132 Perugia, Italy.
| | - F Ferrara
- Pharmaceutical Department, Usl Umbria 1, A. Migliorati street, 06132 Perugia, Italy.
| |
Collapse
|
34
|
Michel K, Herwig M, Werner F, Špiranec Spes K, Abeßer M, Schuh K, Dabral S, Mügge A, Baba HA, Skryabin BV, Hamdani N, Kuhn M. C-type natriuretic peptide moderates titin-based cardiomyocyte stiffness. JCI Insight 2020; 5:139910. [PMID: 33055420 PMCID: PMC7710274 DOI: 10.1172/jci.insight.139910] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Heart failure is often accompanied by titin-dependent myocardial stiffness. Phosphorylation of titin by cGMP-dependent protein kinase I (PKGI) increases cardiomyocyte distensibility. The upstream pathways stimulating PKGI-mediated titin phosphorylation are unclear. We studied whether C-type natriuretic peptide (CNP), via its guanylyl cyclase-B (GC-B) receptor and cGMP/PKGI signaling, modulates titin-based ventricular compliance. To dissect GC-B–mediated effects of endogenous CNP in cardiomyocytes, we generated mice with cardiomyocyte-restricted GC-B deletion (CM GC-B–KO mice). The impact on heart morphology and function, myocyte passive tension, and titin isoform expression and phosphorylation was studied at baseline and after increased afterload induced by transverse aortic constriction (TAC). Pressure overload increased left ventricular endothelial CNP expression, with an early peak after 3 days. Concomitantly, titin phosphorylation at Ser4080, the site phosphorylated by PKGI, was augmented. Notably, in CM GC-B–KO mice this titin response was abolished. TAC-induced hypertrophy and fibrosis were not different between genotypes. However, the KO mice presented mild systolic and diastolic dysfunction together with myocyte stiffness, which were not observed in control littermates. In vitro, recombinant PKGI rescued reduced titin-Ser4080 phosphorylation and reverted passive stiffness of GC-B–deficient cardiomyocytes. CNP-induced activation of GC-B/cGMP/PKGI signaling in cardiomyocytes provides a protecting regulatory circuit preventing titin-based myocyte stiffening during early phases of pressure overload. C-type natriuretic peptide via GC-B/cGMP/PKGI signalling in cardiomyocytes attenuates titin-based cardiomyocyte stiffening during early phases of pressure-overload.
Collapse
Affiliation(s)
- Konstanze Michel
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Melissa Herwig
- Institute of Physiology and.,Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Franziska Werner
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | | | - Marco Abeßer
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Kai Schuh
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Swati Dabral
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Andreas Mügge
- Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Hideo A Baba
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Boris V Skryabin
- Medical Faculty, Core Facility TRAnsgenic animal and genetic engineering Models (TRAM), University of Münster, Münster, Germany
| | - Nazha Hamdani
- Institute of Physiology and.,Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
35
|
Affiliation(s)
- S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Yang Chen
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
36
|
Murphy SP, Prescott MF, Camacho A, Iyer SR, Maisel AS, Felker GM, Butler J, Piña IL, Ibrahim NE, Abbas C, Burnett JC, Solomon SD, Januzzi JL. Atrial Natriuretic Peptide and Treatment With Sacubitril/Valsartan in Heart Failure With Reduced Ejection Fraction. JACC-HEART FAILURE 2020; 9:127-136. [PMID: 33189632 DOI: 10.1016/j.jchf.2020.09.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVES This study sought to assess associations between longitudinal change in atrial natriuretic peptide (ANP) and reverse cardiac remodeling following initiation of sacubitril/valsartan in patients with heart failure with reduced ejection fraction (HFrEF). BACKGROUND Neprilysin inhibition results in an increase of several vasoactive peptides that may mediate the beneficial effects of sacubitril/valsartan, including ANP. METHODS In a prospective study of initiation and titration of sacubitril/valsartan in patients with HFrEF, blood was collected at scheduled time points into tubes containing protease inhibitors. This pre-specified exploratory analysis included patients in whom ANP was measured at baseline and serially through 12 months of treatment. RESULTS Among 144 participants (mean age: 64.5 years; left ventricular ejection fraction: 30.8%), following initiation of sacubitril/valsartan, there was an early and significant increase in ANP, with the majority of rise from 99 pg/ml at baseline to 156 pg/ml at day 14 (p < 0.001). There was a further trend toward a second increase from day 30 to day 45 (p = 0.07). At maximal rise, ANP had doubled. In longitudinal analyses, early rise in ANP was followed by a subsequent increase in urinary cycle guanosine monophosphate. Larger early increase in ANP was associated with larger later improvements in left ventricular ejection fraction and left atrial volume index (p < 0.001 for both). CONCLUSIONS Concentrations of ANP doubled after initiation of sacubitril/valsartan in patients with HFrEF. Larger early increases in ANP were associated with a greater magnitude of subsequent reverse cardiac remodeling. (Effects of Sacubitril/Valsartan Therapy on Biomarkers, Myocardial Remodeling and Outcomes [PROVE-HF]; NCT02887183).
Collapse
Affiliation(s)
- Sean P Murphy
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | | - Alan S Maisel
- University of California, San Diego School of Medicine, San Diego, California, USA
| | - G Michael Felker
- Duke University Medical Center and Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Javed Butler
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | - Nasrien E Ibrahim
- Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Cheryl Abbas
- Novartis Pharmaceuticals, East Hanover, New Jersey, USA
| | | | - Scott D Solomon
- Harvard Medical School, Boston, Massachusetts, USA; Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - James L Januzzi
- Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; Baim Institute for Clinical Research, Boston, Massachusetts, USA.
| |
Collapse
|
37
|
Zhao J, Pei L. Cardiac Endocrinology: Heart-Derived Hormones in Physiology and Disease. ACTA ACUST UNITED AC 2020; 5:949-960. [PMID: 33015416 PMCID: PMC7524786 DOI: 10.1016/j.jacbts.2020.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022]
Abstract
The heart plays a central role in the circulatory system and provides essential oxygen, nutrients, and growth factors to the whole organism. The heart can synthesize and secrete endocrine signals to communicate with distant target organs. Studies of long-known and recently discovered heart-derived hormones highlight a shared theme and reveal a unified mechanism of heart-derived hormones in coordinating cardiac function and target organ biology. This paper reviews the biochemistry, signaling, function, regulation, and clinical significance of representative heart-derived hormones, with a focus on the cardiovascular system. This review also discusses important and exciting questions that will advance the field of cardiac endocrinology.
Collapse
Key Words
- ANP, atrial natriuretic peptide
- ActR, activin receptor
- BNP, brain natriuretic peptide
- CNP, C-type natriuretic peptide
- FGF, fibroblast growth factor
- FSTL, follistatin-like
- GDF, growth differentiation factor
- GDF15
- GFRAL, GDNF family receptor α-like
- NPR, natriuretic peptide receptors
- PCSK, proprotein convertase subtilisin/kexin type
- ST2, suppression of tumorigenesis-2
- TGF, transforming growth factor
- cardiac endocrinology
- heart
- heart-derived hormones
Collapse
Affiliation(s)
- Juanjuan Zhao
- Center for Mitochondrial and Epigenomic Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Liming Pei
- Center for Mitochondrial and Epigenomic Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
38
|
Zeigler AC, Nelson AR, Chandrabhatla AS, Brazhkina O, Holmes JW, Saucerman JJ. Computational model predicts paracrine and intracellular drivers of fibroblast phenotype after myocardial infarction. Matrix Biol 2020; 91-92:136-151. [PMID: 32209358 PMCID: PMC7434705 DOI: 10.1016/j.matbio.2020.03.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/14/2020] [Accepted: 03/16/2020] [Indexed: 01/09/2023]
Abstract
The fibroblast is a key mediator of wound healing in the heart and other organs, yet how it integrates multiple time-dependent paracrine signals to control extracellular matrix synthesis has been difficult to study in vivo. Here, we extended a computational model to simulate the dynamics of fibroblast signaling and fibrosis after myocardial infarction (MI) in response to time-dependent data for nine paracrine stimuli. This computational model was validated against dynamic collagen expression and collagen area fraction data from post-infarction rat hearts. The model predicted that while many features of the fibroblast phenotype at inflammatory or maturation phases of healing could be recapitulated by single static paracrine stimuli (interleukin-1 and angiotensin-II, respectively), mimicking the reparative phase required paired stimuli (e.g. TGFβ and endothelin-1). Virtual overexpression screens simulated with either static cytokine pairs or post-MI paracrine dynamic predicted phase-specific regulators of collagen expression. Several regulators increased (Smad3) or decreased (Smad7, protein kinase G) collagen expression specifically in the reparative phase. NADPH oxidase (NOX) overexpression sustained collagen expression from reparative to maturation phases, driven by TGFβ and endothelin positive feedback loops. Interleukin-1 overexpression had mixed effects, both enhancing collagen via the TGFβ positive feedback loop and suppressing collagen via NFκB and BAMBI (BMP and activin membrane-bound inhibitor) incoherent feed-forward loops. These model-based predictions reveal network mechanisms by which the dynamics of paracrine stimuli and interacting signaling pathways drive the progression of fibroblast phenotypes and fibrosis after myocardial infarction.
Collapse
Affiliation(s)
- Angela C Zeigler
- Department of Biomedical Engineering, University of Virginia, PO Box 800759, Charlottesville, VA 22908-0759, USA
| | - Anders R Nelson
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Anirudha S Chandrabhatla
- Department of Biomedical Engineering, University of Virginia, PO Box 800759, Charlottesville, VA 22908-0759, USA
| | - Olga Brazhkina
- Department of Biomedical Engineering, University of Virginia, PO Box 800759, Charlottesville, VA 22908-0759, USA; Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA, USA
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, PO Box 800759, Charlottesville, VA 22908-0759, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA; Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, PO Box 800759, Charlottesville, VA 22908-0759, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
39
|
Krylatov AV, Tsibulnikov SY, Mukhomedzyanov AV, Boshchenko AA, Goldberg VE, Jaggi AS, Erben RG, Maslov LN. The Role of Natriuretic Peptides in the Regulation of Cardiac Tolerance to Ischemia/Reperfusion and Postinfarction Heart Remodeling. J Cardiovasc Pharmacol Ther 2020; 26:131-148. [PMID: 32840121 DOI: 10.1177/1074248420952243] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In the past 10 years, mortality from acute myocardial infarction has not decreased despite the widespread introduction of percutaneous coronary intervention. The reason for this situation is the absence in clinical practice of drugs capable of preventing reperfusion injury of the heart with high efficiency. In this regard, noteworthy natriuretic peptides (NPs) which have the infarct-limiting effect, prevent reperfusion cardiac injury, prevent adverse post-infarction remodeling of the heart. Atrial natriuretic peptide does not have the infarct-reducing effect in rats with alloxan-induced diabetes mellitus. NPs have the anti-apoptotic and anti-inflammatory effects. There is indirect evidence that NPs inhibit pyroptosis and autophagy. Published data indicate that NPs inhibit reactive oxygen species production in cardiomyocytes, aorta, heart, kidney and the endothelial cells. NPs can suppress aldosterone, angiotensin II, endothelin-1 synthesize and secretion. NPs inhibit the effects aldosterone, angiotensin II on the post-receptor level through intracellular signaling events. NPs activate guanylyl cyclase, protein kinase G and protein kinase A, and reduce phosphodiesterase 3 activity. NO-synthase and soluble guanylyl cyclase are involved in the cardioprotective effect of NPs. The cardioprotective effect of natriuretic peptides is mediated via activation of kinases (AMPK, PKC, PI3 K, ERK1/2, p70s6 k, Akt) and inhibition of glycogen synthase kinase 3β. The cardioprotective effect of NPs is mediated via sarcolemmal KATP channel and mitochondrial KATP channel opening. The cardioprotective effect of brain natriuretic peptide is mediated via MPT pore closing. The anti-fibrotic effect of NPs may be mediated through inhibition TGF-β1 expression. Natriuretic peptides can inhibit NF-κB activity and activate GATA. Hemeoxygenase-1 and peroxisome proliferator-activated receptor γ may be involved in the infarct-reducing effect of NPs. NPs exhibit the infarct-limiting effect in patients with acute myocardial infarction. NPs prevent post-infarction remodeling of the heart. To finally resolve the question of the feasibility of using NPs in AMI, a multicenter, randomized, blind, placebo-controlled study is needed to assess the effect of NPs on the mortality of patients after AMI.
Collapse
Affiliation(s)
- Andrey V Krylatov
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Sergey Y Tsibulnikov
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | | | - Alla A Boshchenko
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Victor E Goldberg
- Cancer Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Amteshwar S Jaggi
- 429174Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Reinhold G Erben
- Department of Biomedical Research, Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Leonid N Maslov
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| |
Collapse
|
40
|
Vinnakota S, Chen HH. The Importance of Natriuretic Peptides in Cardiometabolic Diseases. J Endocr Soc 2020; 4:bvaa052. [PMID: 32537542 PMCID: PMC7278279 DOI: 10.1210/jendso/bvaa052] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/07/2020] [Indexed: 12/11/2022] Open
Abstract
The natriuretic peptide (NP) system is composed of 3 distinct peptides (atrial natriuretic peptide or ANP, B-type natriuretic peptide or BNP, and C-type natriuretic peptide or CNP) and 3 receptors (natriuretic peptide receptor-A or NPR-A or particulate guanynyl cyclase-A natriuretic peptide receptor-B or NPR-B or particulate guanynyl cyclase-B, and natriuretic peptide receptor-C or NPR-C or clearance receptor). ANP and BNP function as defense mechanisms against ventricular stress and the deleterious effects of volume and pressure overload on the heart. Although the role of NPs in cardiovascular homeostasis has been extensively studied and well established, much remains uncertain about the signaling pathways in pathological states like heart failure, a state of impaired natriuretic peptide function. Elevated levels of ANP and BNP in heart failure correlate with disease severity and have a prognostic value. Synthetic ANP and BNP have been studied for their therapeutic role in hypertension and heart failure, and promising trials are under way. In recent years, the expression of ANP and BNP in human adipocytes has come to light. Through their role in promotion of adipocyte browning, lipolysis, lipid oxidation, and modulation of adipokine secretion, they have emerged as key regulators of energy consumption and metabolism. NPR-A signaling in skeletal muscles and adipocytes is emerging as pivotal to the maintenance of long-term insulin sensitivity, which is disrupted in obesity and reduced glucose-tolerance states. Genetic variants in the genes encoding for ANP and BNP have been associated with a favorable cardiometabolic profile. In this review, we discuss several pathways that have been proposed to explain the role of NPs as endocrine networkers. There is much to be explored about the therapeutic role of NPs in improving metabolic milieu.
Collapse
Affiliation(s)
- Shravya Vinnakota
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Horng H Chen
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
41
|
He Y, Liu Y, Zhou M, Xie K, Tang Y, Huang H, Huang C. C-type natriuretic peptide suppresses ventricular arrhythmias in rats with acute myocardial ischemia. Peptides 2020; 126:170238. [PMID: 31870937 DOI: 10.1016/j.peptides.2019.170238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 11/23/2022]
Abstract
This study aimed to investigate the effects of C-type natriuretic peptide (CNP) on ventricular arrhythmias in rats with acute myocardial ischemia (AMI). Forty male Sprague-Dawley rats were randomly divided into sham group (n = 10), AMI group (n = 15) and AMI + CNP group (n = 15). AMI model was induced by ligating the left anterior descending branch of the coronary artery, and CNP was pumped through the femoral vein starting 30 min before ischemia and continuing until 1 h after AMI. The occurrence of ventricular arrhythmias after ischemia and heart rate variability (HRV) were recorded and analyzed. The plasma norepinephrine level was detected at 15 min after AMI. Ventricular electrophysiological parameters including ventricular effective refractory period (ERP), ERP dispersion, ventricular action potential duration (APD) alternans and ventricular fibrillation threshold (VFT) were measured one hour after AMI. Then, the expressions of cyclic guanosine monophosphate in myocardial tissue and left stellate ganglion were examined. Compared to sham group, AMI significantly shortened the ERP, augmented ERP dispersion, elevated APD alternans cycle length, reduced VFT, and increased the incidence of ventricular arrhythmias. Moreover, AMI increased the sympathetic component of HRV, raised plasma norepinephrine levels, and decreased the cyclic guanosine monophosphate levels in myocardium and left stellate ganglion. All those changes were attenuated by CNP treatment. These findings suggest that CNP protected against ventricular arrhythmias in rats with AMI, potentially by inhibiting ischemia-induced cardiac sympathetic hyperactivity and cardiac electrophysiology instability.
Collapse
Affiliation(s)
- Yan He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yu Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Mingmin Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ke Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
42
|
Moyes AJ, Chu SM, Aubdool AA, Dukinfield MS, Margulies KB, Bedi KC, Hodivala-Dilke K, Baliga RS, Hobbs AJ. C-type natriuretic peptide co-ordinates cardiac structure and function. Eur Heart J 2020; 41:1006-1020. [PMID: 30903134 PMCID: PMC7068173 DOI: 10.1093/eurheartj/ehz093] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/21/2018] [Accepted: 02/26/2019] [Indexed: 12/11/2022] Open
Abstract
AIMS C-type natriuretic peptide (CNP) is an essential endothelium-derived signalling species that governs vascular homoeostasis; CNP is also expressed in the heart but an intrinsic role for the peptide in cardiac function is not established. Herein, we employ unique transgenic strains with cell-specific deletion of CNP to define a central (patho)physiological capacity of CNP in maintaining heart morphology and contractility. METHODS AND RESULTS Cardiac structure and function were explored in wild type (WT), cardiomyocyte (cmCNP-/-), endothelium (ecCNP-/-), and fibroblast (fbCNP-/-)-specific CNP knockout mice, and global natriuretic peptide receptor (NPR)-B-/-, and NPR-C-/- animals at baseline and in experimental models of myocardial infarction and heart failure (HF). Endothelium-specific deletion of CNP resulted in impaired coronary responsiveness to endothelium-dependent- and flow-mediated-dilatation; changes mirrored in NPR-C-/- mice. Ex vivo, global ischaemia resulted in larger infarcts and diminished functional recovery in cmCNP-/- and NPR-C-/-, but not ecCNP-/-, vs. WT. The cardiac phenotype of cmCNP-/-, fbCNP-/-, and NPR-C-/- (but not ecCNP-/- or NPR-B-/-) mice was more severe in pressure overload- and sympathetic hyperactivation-induced HF compared with WT; these adverse effects were rescued by pharmacological CNP administration in WT, but not NPR-C-/-, mice. At a molecular level, CNP/NPR-C signalling is impaired in human HF but attenuates activation of well-validated pro-hypertrophic and pro-fibrotic pathways. CONCLUSION C-type natriuretic peptide of cardiomyocyte, endothelial and fibroblast origins co-ordinates and preserves cardiac structure, function, and coronary vasoreactivity via activation of NPR-C. Targeting NPR-C may prove an innovative approach to treating HF and ischaemic cardiovascular disorders.
Collapse
Affiliation(s)
- Amie J Moyes
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Sandy M Chu
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Aisah A Aubdool
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Matthew S Dukinfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Kenneth B Margulies
- Heart Failure and Transplant Program, Perelman School of Medicine, University of Pennsylvania, Translational Research Center, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Kenneth C Bedi
- Heart Failure and Transplant Program, Perelman School of Medicine, University of Pennsylvania, Translational Research Center, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Reshma S Baliga
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
43
|
Chen W, Werner F, Illerhaus A, Knopp T, Völker K, Potapenko T, Hofmann U, Frantz S, Baba HA, Rösch M, Zernecke A, Karbach S, Wenzel P, Kuhn M. Stabilization of Perivascular Mast Cells by Endothelial CNP (C-Type Natriuretic Peptide). Arterioscler Thromb Vasc Biol 2020; 40:682-696. [PMID: 31893950 DOI: 10.1161/atvbaha.119.313702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Activated perivascular mast cells (MCs) participate in different cardiovascular diseases. Many factors provoking MC degranulation have been described, while physiological counterregulators are barely known. Endothelial CNP (C-type natriuretic peptide) participates in the maintenance of vascular barrier integrity, but the target cells and mechanisms are unclear. Here, we studied whether MCs are regulated by CNP. Approach and Results: In cultured human and murine MCs, CNP activated its specific GC (guanylyl cyclase)-B receptor and cyclic GMP signaling. This enhanced cyclic GMP-dependent phosphorylation of the cytoskeleton-associated VASP (vasodilator-stimulated phosphoprotein) and inhibited ATP-evoked degranulation. To elucidate the relevance in vivo, mice with a floxed GC-B (Npr2) gene were interbred with a Mcpt5-CreTG line to generate mice lacking GC-B in connective tissue MCs (MC GC-B knockout). In anesthetized mice, acute ischemia-reperfusion of the cremaster muscle microcirculation provoked extensive MC degranulation and macromolecule extravasation. Superfusion of CNP markedly prevented MC activation and endothelial barrier disruption in control but not in MC GC-B knockout mice. Notably, already under resting conditions, such knockout mice had increased numbers of degranulated MCs in different tissues, together with elevated plasma chymase levels. After transient coronary occlusion, their myocardial areas at risk and with infarction were enlarged. Moreover, MC GC-B knockout mice showed augmented perivascular neutrophil infiltration and deep vein thrombosis in a model of inferior vena cava ligation. CONCLUSIONS CNP, via GC-B/cyclic GMP signaling, stabilizes resident perivascular MCs at baseline and prevents their excessive activation under pathological conditions. Thereby CNP contributes to the maintenance of vascular integrity in physiology and disease.
Collapse
Affiliation(s)
- Wen Chen
- From the Institute of Physiology, University of Würzburg, Germany (W.C., F.W., K.V., T.P., M.K.).,Comprehensive Heart Failure Center (W.C., U.H., S.F., M.K.), University Hospital Würzburg, Germany
| | - Franziska Werner
- From the Institute of Physiology, University of Würzburg, Germany (W.C., F.W., K.V., T.P., M.K.)
| | - Anja Illerhaus
- Institute of Experimental Biomedicine (M.R., A.Z.), University Hospital Würzburg, Germany
| | - Tanja Knopp
- Department of Dermatology, University of Cologne, Germany (A.I.)
| | - Katharina Völker
- From the Institute of Physiology, University of Würzburg, Germany (W.C., F.W., K.V., T.P., M.K.)
| | - Tamara Potapenko
- From the Institute of Physiology, University of Würzburg, Germany (W.C., F.W., K.V., T.P., M.K.)
| | - Ulrich Hofmann
- Comprehensive Heart Failure Center (W.C., U.H., S.F., M.K.), University Hospital Würzburg, Germany
| | - Stefan Frantz
- Comprehensive Heart Failure Center (W.C., U.H., S.F., M.K.), University Hospital Würzburg, Germany
| | - Hideo A Baba
- Center of Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Germany (T.K., S.K., P.W.)
| | - Melanie Rösch
- Institute of Experimental Biomedicine (M.R., A.Z.), University Hospital Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine (M.R., A.Z.), University Hospital Würzburg, Germany
| | - Susanne Karbach
- Department of Dermatology, University of Cologne, Germany (A.I.).,Institute of Pathology, University Hospital Essen, University Duisburg-Essen (H.A.B.)
| | - Philip Wenzel
- Department of Dermatology, University of Cologne, Germany (A.I.).,Institute of Pathology, University Hospital Essen, University Duisburg-Essen (H.A.B.)
| | - Michaela Kuhn
- From the Institute of Physiology, University of Würzburg, Germany (W.C., F.W., K.V., T.P., M.K.).,Comprehensive Heart Failure Center (W.C., U.H., S.F., M.K.), University Hospital Würzburg, Germany
| |
Collapse
|
44
|
Ding DZ, Jia YN, Zhang B, Guan CM, Zhou S, Li X, Cui X. C‑type natriuretic peptide prevents angiotensin II‑induced atrial connexin 40 and 43 dysregulation by activating AMP‑activated kinase signaling. Mol Med Rep 2019; 20:5091-5099. [PMID: 31638216 PMCID: PMC6854524 DOI: 10.3892/mmr.2019.10744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 08/14/2019] [Indexed: 12/13/2022] Open
Abstract
C‑type natriuretic peptide (CNP), from the family of natriuretic peptides (NPs), has been shown to induce antihypertrophic and antifibrotic effects in cardiomyocytes. However, the roles of CNP in the atrial dysregulation of connexin (Cx)40 and Cx43 remain to be elucidated. The present study aimed to investigate the effects of CNP on angiotensin (Ang) II‑induced Cx40 and Cx43 dysregulation in isolated perfused beating rat left atria. A rat isolated perfused beating atrial model was used and the protein levels were determined via western blotting. Ang II significantly upregulated NF‑κB, activator protein‑1, transforming growth factor‑β1 (TGF‑β1), collagen I and matrix metalloproteinase 2, leading to atrial fibrosis, and downregulated expression of Cx40 and Cx43. The changes in Cx40 and Cx43 induced by Ang II were abolished by CNP through upregulation of phosphorylated AMP‑activated kinase a1 (AMPK) and downregulation of TGF‑β1. The effects of CNP on AMPK and TGF‑β1 levels were inhibited by KT5823 and pertussis toxin, inhibitors of protein kinase G (PKG) and NP receptor type C (NPR‑C), respectively. Thus, CNP can prevent Ang II‑induced dysregulation of Cx40 and Cx43 through activation of AMPK via the CNP‑PKG and CNP‑NPR‑C pathways in isolated beating rat atria. The present findings suggested that CNP may be therapeutically useful for clinical conditions involving cardiac dysregulation of Cx expression‑related diseases.
Collapse
Affiliation(s)
- Da-Zhi Ding
- Department of Cardiology, Institute of Clinical Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Ya-Nan Jia
- Department of Cardiology, Institute of Clinical Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Bo Zhang
- Department of Physiology, College of Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Cheng-Ming Guan
- Department of Cardiology, Institute of Clinical Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Shuai Zhou
- Department of Physiology, College of Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Xiang Li
- Department of Physiology, College of Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Xun Cui
- Department of Physiology, College of Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
- Key Laboratory of Organism Functional Factors of The Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, P.R. China
- Cellular Function Research Center, Yanbian University, Yanji, Jilin 133002, P.R. China
| |
Collapse
|
45
|
Tuscany Sangiovese grape juice imparts cardioprotection by regulating gene expression of cardioprotective C-type natriuretic peptide. Eur J Nutr 2019; 59:2953-2968. [PMID: 31707544 DOI: 10.1007/s00394-019-02134-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 10/29/2019] [Indexed: 01/02/2023]
Abstract
PURPOSE A regular intake of red grape juice has cardioprotective properties, but its role on the modulation of natriuretic peptides (NPs), in particular of C-type NP (CNP), has not yet been proven. The aims were to evaluate: (1) in vivo the effects of long-term intake of Tuscany Sangiovese grape juice (SGJ) on the NPs system in a mouse model of myocardial infarction (MI); (2) in vitro the response to SGJ small RNAs of murine MCEC-1 under physiological and ischemic condition; (3) the activation of CNP/NPR-B/NPR-C in healthy human subjects after 7 days' SGJ regular intake. METHODS (1) C57BL/6J male and female mice (n = 33) were randomly subdivided into: SHAM (n = 7), MI (n = 15) and MI fed for 4 weeks with a normal chow supplemented with Tuscany SGJ (25% vol/vol, 200 µl/per day) (MI + SGJ, n = 11). Echocardiography and histological analyses were performed. Myocardial NPs transcriptional profile was investigated by Real-Time PCR. (2) MCEC-1 were treated for 24 h with a pool of SGJ small RNAs and cell viability under 24 h exposure to H2O2 was evaluated by MTT assay. (3) Human blood samples were collected from seven subjects before and after the 7 days' intake of Tuscany SGJ. NPs and miRNA transcriptional profile were investigated by Real-Time PCR in MCEC-1 and human blood. RESULTS Our experimental data, obtained in a multimodal pipeline, suggest that the long-term intake of SGJ promotes an adaptive response of the myocardium to the ischemic microenvironment through the modulation of the cardiac CNP/NPR-B/NPR-C system. CONCLUSIONS Our results open new avenue in the development of functional foods aimed at enhancing cardioprotection of infarcted hearts through action on the myocardial epigenome.
Collapse
|
46
|
Oatmen KE, Zile MR, Burnett JC, Spinale FG. Bioactive Signaling in Next-Generation Pharmacotherapies for Heart Failure: A Review. JAMA Cardiol 2019; 3:1232-1243. [PMID: 30484834 DOI: 10.1001/jamacardio.2018.3789] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Importance The standard pharmacotherapy for heart failure (HF), particularly HF with reduced ejection fraction (HFrEF), is primarily through the use of receptor antagonists, notably inhibition of the renin-angiotensin system by either angiotensin-converting enzyme inhibition or angiotensin II receptor blockade (ARB). However, the completed Prospective Comparison of ARNI With an ACE-Inhibitor to Determine Impact on Global Mortality and Morbidity in Heart Failure (PARADIGM-HF) trial identified that the use of a single molecule (sacubitril/valsartan), which is an ARB and the neutral endopeptidase inhibitor (NEPi) neprilysin, yielded improved clinical outcomes in HFrEF compared with angiotensin-converting enzyme inhibition alone. Observations This review examined specific bioactive signaling pathways that would be potentiated by NEPi and how these would affect key cardiovascular processes relevant to HFrEF. It also addressed potential additive/synergistic effects of ARB. A number of biological signaling pathways that may be potentiated by sacubitril/valsartan were identified, including some novel candidate molecules, which will act in a synergistic manner to favorably alter the natural history of HFrEF. Conclusions and Relevance This review identified that activation rather than inhibition of specific receptor pathways provided favorable cardiovascular effects that cannot be achieved by renin-angiotensin system inhibition alone. Thus, an entirely new avenue of translational and clinical research lies ahead in which HF pharmacotherapies will move beyond receptor antagonist strategies.
Collapse
Affiliation(s)
- Kelsie E Oatmen
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia
| | - Michael R Zile
- Medical University of South Carolina, Charleston.,Ralph H. Johnson Department of VA Medical Center, Charleston, South Carolina
| | - John C Burnett
- Cardiorenal Research Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia.,William Jennings Bryan Dorn VA Medical Center, Columbia, South Carolina
| |
Collapse
|
47
|
Forte M, Madonna M, Schiavon S, Valenti V, Versaci F, Zoccai GB, Frati G, Sciarretta S. Cardiovascular Pleiotropic Effects of Natriuretic Peptides. Int J Mol Sci 2019; 20:3874. [PMID: 31398927 PMCID: PMC6719167 DOI: 10.3390/ijms20163874] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/03/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023] Open
Abstract
Atrial natriuretic peptide (ANP) is a cardiac hormone belonging to the family of natriuretic peptides (NPs). ANP exerts diuretic, natriuretic, and vasodilatory effects that contribute to maintain water-salt balance and regulate blood pressure. Besides these systemic properties, ANP displays important pleiotropic effects in the heart and in the vascular system that are independent of blood pressure regulation. These functions occur through autocrine and paracrine mechanisms. Previous works examining the cardiac phenotype of loss-of-function mouse models of ANP signaling showed that both mice with gene deletion of ANP or its receptor natriuretic peptide receptor A (NPR-A) developed cardiac hypertrophy and dysfunction in response to pressure overload and chronic ischemic remodeling. Conversely, ANP administration has been shown to improve cardiac function in response to remodeling and reduces ischemia-reperfusion (I/R) injury. ANP also acts as a pro-angiogenetic, anti-inflammatory, and anti-atherosclerotic factor in the vascular system. Pleiotropic effects regarding brain natriuretic peptide (BNP) and C-type natriuretic peptide (CNP) were also reported. In this review, we discuss the current evidence underlying the pleiotropic effects of NPs, underlying their importance in cardiovascular homeostasis.
Collapse
Affiliation(s)
| | | | - Sonia Schiavon
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Valentina Valenti
- Department of Cardiology, Santa Maria Goretti Hospital, 04100 Latina, Italy
| | - Francesco Versaci
- Department of Cardiology, Santa Maria Goretti Hospital, 04100 Latina, Italy
| | - Giuseppe Biondi Zoccai
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
- Mediterranea Cardiocentro, 80122 Napoli, Italy
| | - Giacomo Frati
- IRCCS NEUROMED, 86077 Pozzilli, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Sebastiano Sciarretta
- IRCCS NEUROMED, 86077 Pozzilli, Italy.
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy.
| |
Collapse
|
48
|
Hansen LH, Madsen TD, Goth CK, Clausen H, Chen Y, Dzhoyashvili N, Iyer SR, Sangaralingham SJ, Burnett JC, Rehfeld JF, Vakhrushev SY, Schjoldager KT, Goetze JP. Discovery of O-glycans on atrial natriuretic peptide (ANP) that affect both its proteolytic degradation and potency at its cognate receptor. J Biol Chem 2019; 294:12567-12578. [PMID: 31186350 DOI: 10.1074/jbc.ra119.008102] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/03/2019] [Indexed: 12/11/2022] Open
Abstract
Atrial natriuretic peptide (ANP) is a peptide hormone that in response to atrial stretch is secreted from atrial myocytes into the circulation, where it stimulates vasodilatation and natriuresis. ANP is an important biomarker of heart failure where low plasma concentrations exclude cardiac dysfunction. ANP is a member of the natriuretic peptide (NP) family, which also includes the B-type natriuretic peptide (BNP) and the C-type natriuretic peptide. The proforms of these hormones undergo processing to mature peptides, and for proBNP, this process has previously been demonstrated to be regulated by O-glycosylation. It has been suggested that proANP also may undergo post-translational modifications. Here, we conducted a targeted O-glycoproteomics approach to characterize O-glycans on NPs and demonstrate that all NP members can carry O-glycans. We identified four O-glycosites in proANP in the porcine heart, and surprisingly, two of these were located on the mature bioactive ANP itself. We found that one of these glycans is located within a conserved sequence motif of the receptor-binding region, suggesting that O-glycans may serve a function beyond intracellular processing and maturation. We also identified an O-glycoform of proANP naturally occurring in human circulation. We demonstrated that site-specific O-glycosylation shields bioactive ANP from proteolytic degradation and modifies potency at its cognate receptor in vitro Furthermore, we showed that ANP O-glycosylation attenuates acute renal and cardiovascular ANP actions in vivo The discovery of novel glycosylated ANP proteoforms reported here significantly improves our understanding of cardiac endocrinology and provides important insight into the etiology of heart failure.
Collapse
Affiliation(s)
- Lasse H Hansen
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, 2100 Copenhagen, Denmark,Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, School of Dentistry, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Thomas Daugbjerg Madsen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, School of Dentistry, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christoffer K Goth
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, School of Dentistry, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, School of Dentistry, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Yang Chen
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Nina Dzhoyashvili
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Seethalakshmi R Iyer
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, School of Dentistry, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Katrine T Schjoldager
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, School of Dentistry, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jens P Goetze
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, 2100 Copenhagen, Denmark .,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 3 Blegdamsvej, 2200 Copenhagen, Denmark
| |
Collapse
|
49
|
Moyes AJ, Hobbs AJ. C-type Natriuretic Peptide: A Multifaceted Paracrine Regulator in the Heart and Vasculature. Int J Mol Sci 2019; 20:E2281. [PMID: 31072047 PMCID: PMC6539462 DOI: 10.3390/ijms20092281] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/11/2022] Open
Abstract
C-type natriuretic peptide (CNP) is an autocrine and paracrine mediator released by endothelial cells, cardiomyocytes and fibroblasts that regulates vital physiological functions in the cardiovascular system. These roles are conveyed via two cognate receptors, natriuretic peptide receptor B (NPR-B) and natriuretic peptide receptor C (NPR-C), which activate different signalling pathways that mediate complementary yet distinct cellular responses. Traditionally, CNP has been deemed the endothelial component of the natriuretic peptide system, while its sibling peptides, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), are considered the endocrine guardians of cardiac function and blood volume. However, accumulating evidence indicates that CNP not only modulates vascular tone and blood pressure, but also governs a wide range of cardiovascular effects including the control of inflammation, angiogenesis, smooth muscle and endothelial cell proliferation, atherosclerosis, cardiomyocyte contractility, hypertrophy, fibrosis, and cardiac electrophysiology. This review will focus on the novel physiological functions ascribed to CNP, the receptors/signalling mechanisms involved in mediating its cardioprotective effects, and the development of therapeutics targeting CNP signalling pathways in different disease pathologies.
Collapse
Affiliation(s)
- Amie J Moyes
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
50
|
Chen Y, Zheng Y, Iyer SR, Harders GE, Pan S, Chen HH, Ichiki T, Burnett JC, Sangaralingham SJ. C53: A novel particulate guanylyl cyclase B receptor activator that has sustained activity in vivo with anti-fibrotic actions in human cardiac and renal fibroblasts. J Mol Cell Cardiol 2019; 130:140-150. [PMID: 30954448 DOI: 10.1016/j.yjmcc.2019.03.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/08/2019] [Accepted: 03/31/2019] [Indexed: 01/01/2023]
Abstract
The native particulate guanylyl cyclase B receptor (pGC-B) activator, C-type natriuretic peptide (CNP), induces anti-remodeling actions in the heart and kidney through the generation of the second messenger 3', 5' cyclic guanosine monophosphate (cGMP). Indeed fibrotic remodeling, particularly in cardiorenal disease states, contributes to disease progression and thus, has been a key target for drug discovery and development. Although the pGC-B/cGMP system has been perceived as a promising anti-fibrotic pathway, its therapeutic potential is limited due to the rapid degradation and catabolism of CNP by neprilysin (NEP) and natriuretic peptide clearance receptor (NPRC). The goal of this study was to bioengineer and test in vitro and in vivo a novel pGC-B activator, C53. Here we established that C53 selectively generates cGMP via the pGC-B receptor and is highly resistant to NEP and has less interaction with NPRC in vitro. Furthermore in vivo, C53 had enhanced cGMP-generating actions that paralleled elevated plasma CNP-like levels, thus indicating a longer circulating half-life compared to CNP. Importantly in human cardiac fibroblasts (HCFs) and renal fibroblasts (HRFs), C53 exerted robust cGMP-generating actions, inhibited TGFβ-1 stimulated HCFs and HRFs proliferation chronically and suppressed the differentiation of HCFs and HRFs to myofibroblasts. The current findings advance innovation in drug discovery and highlight C53 as a novel pGC-B activator with sustained in vivo activity and anti-fibrotic actions in vitro. Future studies are warranted to explore the efficacy and therapeutic opportunity of C53 targeting fibrosis in cardiorenal disease states and beyond.
Collapse
Affiliation(s)
- Yang Chen
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States.
| | - Ye Zheng
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - Seethalakshmi R Iyer
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - Gerald E Harders
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - Shuchong Pan
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - Horng H Chen
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - Tomoko Ichiki
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States; Department of Physiology and Biomedical Engineering, United States
| | - S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States; Department of Physiology and Biomedical Engineering, United States.
| |
Collapse
|