1
|
Singh J, Adel FW, Scott CG, Chen HH. Natriuretic peptide levels and predicting risk of developing new diabetes mellitus and metabolic syndrome. Diabetes Obes Metab 2025; 27:777-784. [PMID: 39587371 DOI: 10.1111/dom.16073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/27/2024]
Abstract
AIMS Define the relationship between N-terminal atrial natriuretic peptide (NT-ANP) levels and incident metabolic syndrome and type 2 diabetes mellitus ('metabolic disease') in healthy adults and develop a risk prediction score. MATERIALS AND METHODS Retrospective cohort study of Olmsted County Heart Function Study participants, a random sampling of county residents aged 45 years and older (n = 2042). Clinical data were collected during enrolment between 1997 and 2000 and upon follow-up 4 years later. Outcomes were followed for 8 years. We studied 715 subjects without metabolic disease at enrolment who completed follow-up, assessing incident metabolic disease as the primary outcome. Youden's index was used to identify optimal cut-points and develop the risk score. RESULTS Upon multivariate analysis adjusting for age gender, HDL and triglycerides, higher baseline serum NT-ANP levels were associated with a lower risk of metabolic disease (OR: 0.65, CI 0.49-0.85, p = 0.002). Higher baseline serum insulin and aldosterone levels were associated with higher risk of incident metabolic disease (OR: 2.04, CI 1.57-2.65, p < 0.001; OR: 1.43, CI 1.14-1.81, p = 0.002, respectively). Baseline serum NT-ANP < 3337 pg/mL was 96.6% sensitive for future development of metabolic disease. A weighted score including all three biomarkers was 78.6% sensitive and 77.3% specific. CONCLUSIONS In healthy adults aged 45 years or older, higher baseline NT-ANP levels are associated with a lower four-year risk of developing metabolic disease. Serum NT-ANP levels are a sensitive biomarker of future risk of metabolic disease and have screening utility when combined with insulin and aldosterone levels into a composite score.
Collapse
Affiliation(s)
- Jasraj Singh
- Department of Internal Medicine, Mayo Clinic School of Graduate Medical Education, Mayo Clinic, Rochester, Minnesota, USA
| | - Fadi W Adel
- Department of Cardiovascular Disease, Mayo Clinic, Rochester, Minnesota, USA
| | - Christopher G Scott
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Horng H Chen
- Department of Cardiovascular Disease, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
2
|
Zhazykbayeva S, Budde H, Kaçmaz M, Zemedie Y, Osman H, Hassoun R, Jaquet K, Akin I, El-Battrawy I, Herwig M, Hamdani N. Exploring PKG signaling as a therapeutic avenue for pressure overload, ischemia, and HFpEF. Expert Opin Ther Targets 2024; 28:857-873. [PMID: 39329430 DOI: 10.1080/14728222.2024.2400093] [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: 01/10/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
INTRODUCTION Heart failure (HF) is a complex and heterogeneous syndrome resulting from any diastolic or systolic dysfunction of the cardiac muscle. In addition to comorbid conditions, pressure overload, and myocardial ischemia are associated with cardiac remodeling which manifests as extracellular matrix (ECM) perturbations, impaired cellular responses, and subsequent ventricular dysfunction. AREAS COVERED The current review discusses the main aspects of the cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) pathway (cGMP-PKG) pathway modulators and highlights the promising outcomes of its novel pharmacological boosters. EXPERT OPINION Among several signaling pathways involved in the pathogenesis of pressure overload, ischemia and HF with preserved ejection fraction (HFpEF) is cGMP-PKG pathway. This pathway plays a pivotal role in the regulation of cardiac contractility, and modulation of cGMP-PKG signaling, contributing to the development of the diseases. Ventricular cardiomyocytes of HF patients and animal models are known to exhibit reduced cGMP levels and disturbed cGMP signaling including hypophosphorylation of PKG downstream targets. However, restoration of cGMP-PKG signaling improves cardiomyocyte function and promotes cardioprotective effects.
Collapse
Affiliation(s)
- S Zhazykbayeva
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - H Budde
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - M Kaçmaz
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, Budapest, Hungary
| | - Y Zemedie
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - H Osman
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - R Hassoun
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - K Jaquet
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - I Akin
- Medical University Mannheim, Medical Faculty, Mannheim University, Heidelberg, Germany
| | - I El-Battrawy
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University, Bochum, Germany
| | - M Herwig
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - N Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, Budapest, Hungary
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University, Bochum, Germany
- Department of Physiology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
3
|
Munguia-Galaviz FJ, Miranda-Diaz AG, Gutierrez-Mercado YK, Ku-Centurion M, Gonzalez-Gonzalez RA, Portilla-de Buen E, Echavarria R. The Sigma-1 Receptor Exacerbates Cardiac Dysfunction Induced by Obstructive Nephropathy: A Role for Sexual Dimorphism. Biomedicines 2024; 12:1908. [PMID: 39200372 PMCID: PMC11351121 DOI: 10.3390/biomedicines12081908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/02/2024] Open
Abstract
The Sigma-1 Receptor (Sigmar1) is a stress-activated chaperone and a promising target for pharmacological modulation due to its ability to induce multiple cellular responses. Yet, it is unknown how Sigmar1 is involved in cardiorenal syndrome type 4 (CRS4) in which renal damage results in cardiac dysfunction. This study explored the role of Sigmar1 and its ligands in a CRS4 model induced by unilateral ureteral obstruction (UUO) in male and female C57BL/6 mice. We evaluated renal and cardiac dysfunction markers, Sigmar1 expression, and cardiac remodeling through time (7, 12, and 21 days) and after chronically administering the Sigmar1 agonists PRE-084 (1 mg/kg/day) and SA4503 (1 mg/kg/day), and the antagonist haloperidol (2 mg/kg/day), for 21 days after UUO using colorimetric analysis, RT-qPCR, histology, immunohistochemistry, enzyme-linked immunosorbent assay, RNA-seq, and bioinformatics. We found that obstructive nephropathy induces Sigmar1 expression in the kidneys and heart, and that Sigmar1 stimulation with its agonists PRE-084 and SA4503 aggravates cardiac dysfunction and remodeling in both sexes. Still, their effects are significantly more potent in males. Our findings reveal essential differences associated with sex in the development of CRS4 and should be considered when contemplating Sigmar1 as a pharmacological target.
Collapse
Affiliation(s)
- Francisco Javier Munguia-Galaviz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (F.J.M.-G.); (A.G.M.-D.)
- Division de Ciencias de la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzman 49000, Jalisco, Mexico
| | - Alejandra Guillermina Miranda-Diaz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (F.J.M.-G.); (A.G.M.-D.)
| | - Yanet Karina Gutierrez-Mercado
- Departamento de Clinicas, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlan 47620, Jalisco, Mexico;
| | - Marco Ku-Centurion
- Unidad de Biotecnologia Medica y Farmaceutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Jalisco, Mexico;
- Division de Investigacion Quirurgica, Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico; (R.A.G.-G.); (E.P.-d.B.)
| | - Ricardo Arturo Gonzalez-Gonzalez
- Division de Investigacion Quirurgica, Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico; (R.A.G.-G.); (E.P.-d.B.)
| | - Eliseo Portilla-de Buen
- Division de Investigacion Quirurgica, Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico; (R.A.G.-G.); (E.P.-d.B.)
| | - Raquel Echavarria
- Consejo Nacional de Humanidades, Ciencias y Tecnologias (CONAHCYT)—Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
4
|
Huang X, Hu L, Long Z, Wang X, Wu J, Cai J. Hypertensive Heart Disease: Mechanisms, Diagnosis and Treatment. Rev Cardiovasc Med 2024; 25:93. [PMID: 39076964 PMCID: PMC11263885 DOI: 10.31083/j.rcm2503093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 07/31/2024] Open
Abstract
Hypertensive heart disease (HHD) presents a substantial global health burden, spanning a spectrum from subtle cardiac functional alterations to overt heart failure. In this comprehensive review, we delved into the intricate pathophysiological mechanisms governing the onset and progression of HHD. We emphasized the significant role of neurohormonal activation, inflammation, and metabolic remodeling in HHD pathogenesis, offering insights into promising therapeutic avenues. Additionally, this review provided an overview of contemporary imaging diagnostic tools for precise HHD severity assessment. We discussed in detail the current potential treatments for HHD, including pharmacologic, lifestyle, and intervention devices. This review aimed to underscore the global importance of HHD and foster a deeper understanding of its pathophysiology, ultimately contributing to improved public health outcomes.
Collapse
Affiliation(s)
- Xuewei Huang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, 410013 Changsha, Hunan, China
| | - Lizhi Hu
- Xiangya School of Medicine, Central South University, 410013 Changsha, Hunan, China
| | - Zhuojun Long
- Xiangya School of Medicine, Central South University, 410013 Changsha, Hunan, China
| | - Xinyao Wang
- Xiangya School of Medicine, Central South University, 410013 Changsha, Hunan, China
| | - Junru Wu
- Department of Cardiology, The Third Xiangya Hospital, Central South University, 410013 Changsha, Hunan, China
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, 410013 Changsha, Hunan, China
| |
Collapse
|
5
|
Sousa MP, Bettencourt P, Brás-Silva C, Pereira C. Biosensors for natriuretic peptides in cardiovascular diseases. A review. Curr Probl Cardiol 2024; 49:102180. [PMID: 37907188 DOI: 10.1016/j.cpcardiol.2023.102180] [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: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 11/02/2023]
Abstract
Heart failure (HF) is a complex clinical syndrome associated with high rates of morbidity and mortality. Over the years, it has been crucial to find accurate biomarkers capable of doing a precise monitor of HF and provide an early diagnosis. Of these, it has been established an important role of natriuretic peptides in HF assessment. Moreover, the development of biosensors has been garnering interest as new diagnostic medical tools. In this review we first provide a general overview of HF, its pathogenesis, and diagnostic features. We then discuss the role of natriuretic peptides in heart failure by characterizing them and point out their potential as biomarkers. Finally, we adress the evolution of biosensors development and the available natriuretic peptides biosensors for disease monitoring.
Collapse
Affiliation(s)
- Mariana P Sousa
- Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Porto 4200-135, Portugal
| | - Paulo Bettencourt
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Carmen Brás-Silva
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Claudia Pereira
- FP-I3ID, Instituto de Investigação, Inovação e Desenvolvimento, FP-BHS, Biomedical and Health Sciences, Universidade Fernando Pessoa, Porto 4249-004, Portugal; HE-FP-Hospital Fernando Pessoa, CECLIN, Center of Clinical Studies, 4420-096 Gondomar, Portugal; FCS-Faculty of Health Sciences, Fernando Pessoa University, 4249-004 Porto, Portugal.
| |
Collapse
|
6
|
Juraver-Geslin H, Devotta A, Saint-Jeannet JP. Developmental roles of natriuretic peptides and their receptors. Cells Dev 2023; 176:203878. [PMID: 37742795 PMCID: PMC10841480 DOI: 10.1016/j.cdev.2023.203878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
Natriuretic peptides and their receptors are implicated in the physiological control of blood pressure, bone growth, and cardiovascular and renal homeostasis. They mediate their action through the modulation of intracellular levels of cGMP and cAMP, two second-messengers that have broad biological roles. In this review, we briefly describe the major players of this signaling pathway and their physiological roles in the adult, and discuss several reports describing their activity in the control of various aspects of embryonic development in several species. While the core components of this signaling pathway are well conserved, their functions have diverged in the embryo and the adult to control a diverse array of biological processes.
Collapse
Affiliation(s)
- Hugo Juraver-Geslin
- Department of Molecular Pathobiology, New York University, College of Dentistry, New York, NY 10010, USA
| | - Arun Devotta
- Department of Molecular Pathobiology, New York University, College of Dentistry, New York, NY 10010, USA
| | - Jean-Pierre Saint-Jeannet
- Department of Molecular Pathobiology, New York University, College of Dentistry, New York, NY 10010, USA.
| |
Collapse
|
7
|
Huang YM, Li TX, Li SY, Zhu XR, Li Y, Liu DY, Li WM, Yang LQ, Liu KS, Liu C. Glucocorticoids ameliorate cardiorenal syndrome through the NPR1/SGK1 pathway in natriuretic peptide receptor A‑heterozygous mice. Exp Ther Med 2023; 26:374. [PMID: 37415837 PMCID: PMC10320660 DOI: 10.3892/etm.2023.12073] [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: 02/11/2022] [Accepted: 04/28/2023] [Indexed: 07/08/2023] Open
Abstract
Natriuretic peptides, which are produced by the heart, bind to natriuretic peptide receptor A (NPR1 encoded by natriuretic peptide receptor 1 gene) and cause vasodilation and natriuresis. Thus, they serve an important role in regulating blood pressure. In the present study, microinjection of CRISPR associated protein 9/single guide RNA into fertilized C57BL/6N mouse eggs was performed to generate filial generation zero (F0) Npr1 knockout homozygous mice (Npr1-/-). F0 mice mated with wild-type (WT) mice to obtain F1 Npr1 knockout heterozygous mice with stable heredity (Npr1+/-). F1 self-hybridization was used to expand the population of heterozygous mice (Npr1+/-). The present study performed echocardiography to investigate the impact of NPR1 gene knockdown on cardiac function. Compared with those in the WT group (C57BL/6N male mice), the left ventricular ejection fraction, myocardial contractility and renal sodium and potassium excretion and creatinine-clearance rates were decreased, indicating that Npr1 knockdown induced cardiac and renal dysfunction. In addition, expression of serum glucocorticoid-regulated kinase 1 (SGK1) increased significantly compared with that in WT mice. However, glucocorticoids (dexamethasone) upregulated NPR1 and inhibited SGK1 and alleviated cardiac and renal dysfunction caused by Npr1 gene heterozygosity. SGK1 inhibitor GSK650394 ameliorate cardiorenal syndrome by suppressing SGK1. Briefly, glucocorticoids inhibited SGK1 by upregulating NPR1, thereby ameliorating cardiorenal impairment caused by Npr1 gene heterozygosity. The present findings provided novel insight into the understanding of cardiorenal syndrome and suggested that glucocorticoids targeting the NPR1/SGK1 pathway may be a potential therapeutic target to treat cardiorenal syndrome.
Collapse
Affiliation(s)
- Yao-Meng Huang
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Tong-Xin Li
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Shu-Yu Li
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
- Department of Cardiology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Xiao-Ran Zhu
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
- Department of Pharmacy, Hebei General Hospital, Shijiazhuang, Hebei 050057, P.R. China
| | - Ying Li
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Dang-Yang Liu
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Wei-Min Li
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Lin-Quan Yang
- Department of Pharmacy, Hebei General Hospital, Shijiazhuang, Hebei 050057, P.R. China
| | - Kun-Shen Liu
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Chao Liu
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| |
Collapse
|
8
|
Núñez-Marín G, Iraola D, Lorenzo M, de la Espriella R, Villar S, Santas E, Miñana G, Sanchis J, Carratalá A, Miró Ò, Bayés-Genís A, Núñez J. An update on utilising brain natriuretic peptide for risk stratification, monitoring and guiding therapy in heart failure. Expert Rev Mol Diagn 2023:1-13. [PMID: 37216616 DOI: 10.1080/14737159.2023.2216386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/04/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023]
Abstract
INTRODUCTION Heart failure (HF) is a dominant health problem with an overall poor prognosis. Natriuretic peptides (NPs) are upregulated in HF as a compensatory mechanism. They have extensively been used for diagnosis and risk stratification. AREAS COVERED This review addresses the history and physiology of NPs in order to understand their current role in clinical practice. It further provides a detailed and updated narrative review on the utility of those biomarkers for risk stratification, monitoring, and guiding therapy in HF. EXPERT OPINION NPs show excellent predictive ability in heart failure patients, both in acute and chronic settings. Understanding their pathophysiology and their modifications in specific situations is key for an adequate interpretation in specific clinical scenarios in which their prognostic value may be weaker or less well evaluated. To better promote risk stratification in HF, NPs should be integrated with other predictive tools to develop multiparametric risk models. Both inequalities of access to NPs and evidence caveats and limitations will need to be addressed by future research in the coming years.
Collapse
Affiliation(s)
- Gonzalo Núñez-Marín
- Cardiology Department, Hospital Clínico Universitario, INCLIVA, Universitat de Valencia. Valencia, Spain. Valencia, Spain
| | - Diego Iraola
- Cardiology Department, Hospital Clínico Universitario, INCLIVA, Universitat de Valencia. Valencia, Spain. Valencia, Spain
| | - Miguel Lorenzo
- Cardiology Department, Hospital Clínico Universitario, INCLIVA, Universitat de Valencia. Valencia, Spain. Valencia, Spain
| | - Rafael de la Espriella
- Cardiology Department, Hospital Clínico Universitario, INCLIVA, Universitat de Valencia. Valencia, Spain. Valencia, Spain
| | - Sandra Villar
- Cardiology Department, Hospital Clínico Universitario, INCLIVA, Universitat de Valencia. Valencia, Spain. Valencia, Spain
| | - Enrique Santas
- Cardiology Department, Hospital Clínico Universitario, INCLIVA, Universitat de Valencia. Valencia, Spain. Valencia, Spain
| | - Gema Miñana
- Cardiology Department, Hospital Clínico Universitario, INCLIVA, Universitat de Valencia. Valencia, Spain. Valencia, Spain
| | - Juan Sanchis
- Cardiology Department, Hospital Clínico Universitario, INCLIVA, Universitat de Valencia. Valencia, Spain. Valencia, Spain
| | - Arturo Carratalá
- Clinical Chemistry Department, Hospital Clínico Universitario, INCLIVA
| | - Òscar Miró
- Emergency Department, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Antoni Bayés-Genís
- Cardiology Department, Hospital Universitari Germas Trias i Pujol. Badalona, Spain
- CIBER Cardiovascular, Madrid, Spain
| | - Julio Núñez
- Cardiology Department, Hospital Clínico Universitario, INCLIVA, Universitat de Valencia. Valencia, Spain. Valencia, Spain
- Emergency Department, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
Martin TG, Juarros MA, Leinwand LA. Regression of cardiac hypertrophy in health and disease: mechanisms and therapeutic potential. Nat Rev Cardiol 2023; 20:347-363. [PMID: 36596855 PMCID: PMC10121965 DOI: 10.1038/s41569-022-00806-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 01/05/2023]
Abstract
Left ventricular hypertrophy is a leading risk factor for cardiovascular morbidity and mortality. Although reverse ventricular remodelling was long thought to be irreversible, evidence from the past three decades indicates that this process is possible with many existing heart disease therapies. The regression of pathological hypertrophy is associated with improved cardiac function, quality of life and long-term health outcomes. However, less than 50% of patients respond favourably to most therapies, and the reversibility of remodelling is influenced by many factors, including age, sex, BMI and disease aetiology. Cardiac hypertrophy also occurs in physiological settings, including pregnancy and exercise, although in these cases, hypertrophy is associated with normal or improved ventricular function and is completely reversible postpartum or with cessation of training. Studies over the past decade have identified the molecular features of hypertrophy regression in health and disease settings, which include modulation of protein synthesis, microRNAs, metabolism and protein degradation pathways. In this Review, we summarize the evidence for hypertrophy regression in patients with current first-line pharmacological and surgical interventions. We further discuss the molecular features of reverse remodelling identified in cell and animal models, highlighting remaining knowledge gaps and the essential questions for future investigation towards the goal of designing specific therapies to promote regression of pathological hypertrophy.
Collapse
Affiliation(s)
- Thomas G Martin
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Miranda A Juarros
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Leslie A Leinwand
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA.
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
10
|
Ghatage T, Singh S, Mandal K, Jadhav KB, Dhar A. Activation of Mas and pGCA receptor pathways protects renal epithelial cell damage against oxidative-stress-induced injury. Peptides 2023; 162:170959. [PMID: 36693526 DOI: 10.1016/j.peptides.2023.170959] [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: 10/26/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Over-activation of the renin-angiotensin-aldosterone system (RAAS) is a leading cause of cardio-renal complications. Oxidative stress is one of the major contributing factors in the over-activation of RAAS. Angiotensin-converting enzyme2/Angiotensin1-7/MasR and natriuretic peptide/particulate guanylyl cyclase receptor-A pathways play a key role in cardiorenal disease protection. Even though individual activation of these pathways possesses cardiorenal protective effects. However, the dual activation of these pathways under stress conditions and the underlying mechanism has not been explored. The study aimed to investigate whether activation of these pathways by dual-acting peptide (DAP) shows a protective effect in-vitro in oxidative stress-induced renal epithelial cells. Oxidative stress was induced in renal epithelial NRK-52E cells with H2O2. Co-treatment with Ang 1-7, BNP, and DAP was given for 30 min. AT1, MasR, and pGCA expression were measured by RT-PCR. The markers of oxidative stress and apoptosis were measured by confocal microscopy and FACS analysis. A significant increase in AT1, renin, α-SMA, and NFk-β expression and a significant decrease in MasR and pGCA expression was observed in H2O2-induced cells. DAP improved H2O2-induced pathological changes in NRK-52E cells. The effect of DAP was superior to that of Ang1-7 and BNP alone. Interestingly, MasR and pGCA inhibitors could block the effect of DAP in H2O2-induced cells. DAP shows superior anti-RAAS activity, and it is effective against H2O2-induced oxidative stress, apoptosis, fibrosis, and inflammation compared to Ang1-7 and BNP alone. The protective effect is mediated by the dual activation of MasR and pGCA.
Collapse
Affiliation(s)
- Trupti Ghatage
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad, Telangana 500078, India
| | - Sameer Singh
- Tata Institute of Fundamental Research Hyderabad, Hyderabad, Telangana 500046, India
| | - Kalyaneswar Mandal
- Tata Institute of Fundamental Research Hyderabad, Hyderabad, Telangana 500046, India
| | - Kirtikumar B Jadhav
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad, Telangana 500078, India.
| |
Collapse
|
11
|
Ushakov A, Ivanchenko V, Gagarina A. Heart Failure And Type 2 Diabetes Mellitus: Neurohumoral, Histological And Molecular Interconnections. Curr Cardiol Rev 2023; 19:e170622206132. [PMID: 35718961 PMCID: PMC10201898 DOI: 10.2174/1573403x18666220617121144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022] Open
Abstract
Heart failure (HF) is a global healthcare burden and a leading cause of morbidity and mortality worldwide. Type 2 diabetes mellitus (T2DM) appears to be one of the major risk factors that significantly worsen HF prognosis and increase the risk of fatal cardiovascular outcomes. Despite a great knowledge of pathophysiological mechanisms involved in HF development and progression, hospitalization rates in patients with HF and concomitant T2DM remain elevated. In this review, we discuss the complex interplay between systemic neurohumoral regulation and local cardiac mechanisms participating in myocardial remodeling and HF development in T2DM with special attention to cardiomyocyte energy metabolism, mitochondrial function and calcium metabolism, cardiomyocyte hypertrophy and death, extracellular matrix remodeling.
Collapse
Affiliation(s)
- A. Ushakov
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - V. Ivanchenko
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - A. Gagarina
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| |
Collapse
|
12
|
Lu G, Hu R, Tao T, Hu M, Dong Z, Wang C. Regulatory role of atrial natriuretic peptide in brown adipose tissue: A narrative review. Obes Rev 2023; 24:e13522. [PMID: 36336901 DOI: 10.1111/obr.13522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/15/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022]
Abstract
Atrial natriuretic peptide (ANP) has been considered to exert an essential role as a cardiac secretory hormone in the regulation of hemodynamic homeostasis. As the research progresses, the role of ANP in the crosstalk between heart and lipid metabolism has become an interesting topic that is attracting the interest of researchers. The regulation of ANP in lipid metabolism shows favorable effects, particularly the activation of brown adipose tissue (BAT). The complex regulatory network of ANP on BAT has not been fully outlined. This narrative review critically evaluated the existing literature on the regulatory effects of ANP on BAT. In general, we have summarized the expression of ANP and its receptors in various human tissues, analyzed the progress of research on the relationship between the ANP and BAT, and described several potential pathways of ANP to BAT. Exogenous ANP, natriuretic peptide receptor C (NPRC) deficiency, cold exposure, bariatric surgery, and cardiac or renal insufficiency could all contribute to BAT expression by increasing circulating ANP levels.
Collapse
Affiliation(s)
- Guanhua Lu
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Ruixiang Hu
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Tian Tao
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Min Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Zhiyong Dong
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
13
|
García-Carrizo F, Galmés S, Picó C, Palou A, Rodríguez AM. Supplementation with the Prebiotic High-Esterified Pectin Improves Blood Pressure and Cardiovascular Risk Biomarker Profile, Counteracting Metabolic Malprogramming. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:13200-13211. [PMID: 36214580 PMCID: PMC9585587 DOI: 10.1021/acs.jafc.2c03143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 06/01/2023]
Abstract
Supplementation with the prebiotic pectin is associated with beneficial health effects. We aimed to characterize the cardioprotective actions of chronic high-esterified pectin (HEP) supplementation (10%) in a model of metabolic malprogramming in rats, prone to obesity and associated disorders: the progeny of mild calorie-restricted dams during the first half of pregnancy. Results show that pectin supplementation reverses metabolic malprogramming associated with gestational undernutrition. In this sense, HEP supplementation improved blood pressure, reduced heart lipid content, and regulated cardiac gene expression of atrial natriuretic peptide and lipid metabolism-related genes. Moreover, it caused an elevation in circulating levels of fibroblast growth factor 21 and a higher expression of its co-receptor β-klotho in the heart. Most effects are correlated with the gut levels of beneficial bacteria promoted by HEP. Therefore, chronic HEP supplementation shows cardioprotective actions, and hence, it is worth considering as a strategy to prevent programmed cardiometabolic alterations.
Collapse
Affiliation(s)
- Francisco García-Carrizo
- Laboratory
of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics,
Biomarkers and Risk Evaluation−NuBE), University of the Balearic Islands, 07122 Palma, Spain
- Department
of Adipocyte Development and Nutrition (ADE), German Institute of Human Nutrition (DIfE), 14558 Potsdam-Rehbrücke, Germany
| | - Sebastià Galmés
- Laboratory
of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics,
Biomarkers and Risk Evaluation−NuBE), University of the Balearic Islands, 07122 Palma, Spain
- Health
Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- Centro
de Investigación Biomédica en Red de Fisiopatología
de la Obesidad y Nutrición, Instituto
de Salud Carlos III, 28029 Madrid, Spain
| | - Catalina Picó
- Laboratory
of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics,
Biomarkers and Risk Evaluation−NuBE), University of the Balearic Islands, 07122 Palma, Spain
- Health
Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- Centro
de Investigación Biomédica en Red de Fisiopatología
de la Obesidad y Nutrición, Instituto
de Salud Carlos III, 28029 Madrid, Spain
| | - Andreu Palou
- Laboratory
of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics,
Biomarkers and Risk Evaluation−NuBE), University of the Balearic Islands, 07122 Palma, Spain
- Health
Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- Centro
de Investigación Biomédica en Red de Fisiopatología
de la Obesidad y Nutrición, Instituto
de Salud Carlos III, 28029 Madrid, Spain
| | - Ana María Rodríguez
- Laboratory
of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics,
Biomarkers and Risk Evaluation−NuBE), University of the Balearic Islands, 07122 Palma, Spain
- Health
Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- Centro
de Investigación Biomédica en Red de Fisiopatología
de la Obesidad y Nutrición, Instituto
de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
14
|
Omatsu-Kanbe M, Fukunaga R, Mi X, Matsuura H. Atypically Shaped Cardiomyocytes (ACMs): The Identification, Characterization and New Insights into a Subpopulation of Cardiomyocytes. Biomolecules 2022; 12:biom12070896. [PMID: 35883452 PMCID: PMC9313223 DOI: 10.3390/biom12070896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 02/01/2023] Open
Abstract
In the adult mammalian heart, no data have yet shown the existence of cardiomyocyte-differentiable stem cells that can be used to practically repair the injured myocardium. Atypically shaped cardiomyocytes (ACMs) are found in cultures of the cardiomyocyte-removed fraction obtained from cardiac ventricles from neonatal to aged mice. ACMs are thought to be a subpopulation of cardiomyocytes or immature cardiomyocytes, most closely resembling cardiomyocytes due to their spontaneous beating, well-organized sarcomere and the expression of cardiac-specific proteins, including some fetal cardiac gene proteins. In this review, we focus on the characteristics of ACMs compared with ventricular myocytes and discuss whether these cells can be substitutes for damaged cardiomyocytes. ACMs reside in the interstitial spaces among ventricular myocytes and survive under severely hypoxic conditions fatal to ventricular myocytes. ACMs have not been observed to divide or proliferate, similar to cardiomyocytes, but they maintain their ability to fuse with each other. Thus, it is worthwhile to understand the role of ACMs and especially how these cells perform cell fusion or function independently in vivo. It may aid in the development of new approaches to cell therapy to protect the injured heart or the clarification of the pathogenesis underlying arrhythmia in the injured heart.
Collapse
|
15
|
Corin: A Key Mediator in Sodium Homeostasis, Vascular Remodeling, and Heart Failure. BIOLOGY 2022; 11:biology11050717. [PMID: 35625445 PMCID: PMC9138375 DOI: 10.3390/biology11050717] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/01/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022]
Abstract
Simple Summary Atrial natriuretic peptide (ANP) is an important hormone that regulates many physiological and pathological processes, including electrolyte and body fluid balance, blood volume and pressure, cardiac channel activity and function, inflammatory response, lipid metabolism, and vascular remodeling. Corin is a transmembrane serine protease that activates ANP. Variants in the CORIN gene are associated with cardiovascular disease, including hypertension, cardiac hypertrophy, atrial fibrillation, heart failure, and preeclampsia. The current data indicate a key role of corin-mediated ANP production and signaling in the maintenance of cardiovascular homeostasis. In this review, we discuss the latest findings regarding the molecular and cellular mechanisms underlying the role of corin in sodium homeostasis, uterine spiral artery remodeling, and heart failure. Abstract Atrial natriuretic peptide (ANP) is a crucial element of the cardiac endocrine function that promotes natriuresis, diuresis, and vasodilation, thereby protecting normal blood pressure and cardiac function. Corin is a type II transmembrane serine protease that is highly expressed in the heart, where it converts the ANP precursor to mature ANP. Corin deficiency prevents ANP activation and causes hypertension and heart disease. In addition to the heart, corin is expressed in other tissues, including those of the kidney, skin, and uterus, where corin-mediated ANP production and signaling act locally to promote sodium excretion and vascular remodeling. These results indicate that corin and ANP function in many tissues via endocrine and autocrine mechanisms. In heart failure patients, impaired natriuretic peptide processing is a common pathological mechanism that contributes to sodium and body fluid retention. In this review, we discuss most recent findings regarding the role of corin in non-cardiac tissues, including the kidney and skin, in regulating sodium homeostasis and body fluid excretion. Moreover, we describe the molecular mechanisms underlying corin and ANP function in supporting orderly cellular events in uterine spiral artery remodeling. Finally, we assess the potential of corin-based approaches to enhance natriuretic peptide production and activity as a treatment of heart failure.
Collapse
|
16
|
Inside the pathophysiological mechanisms of cardiometabolic diseases: the other pandemic to fight. Pflugers Arch 2021; 474:1-4. [PMID: 34961913 DOI: 10.1007/s00424-021-02658-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 10/19/2022]
|
17
|
Mishra S, Sadagopan N, Dunkerly-Eyring B, Rodriguez S, Sarver DC, Ceddia RP, Murphy SA, Knutsdottir H, Jani VP, Ashok D, Oeing CU, O'Rourke B, Gangoiti JA, Sears DD, Wong GW, Collins S, Kass DA. Inhibition of phosphodiesterase type 9 reduces obesity and cardiometabolic syndrome in mice. J Clin Invest 2021; 131:148798. [PMID: 34618683 DOI: 10.1172/jci148798] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 09/16/2021] [Indexed: 12/16/2022] Open
Abstract
Central obesity with cardiometabolic syndrome (CMS) is a major global contributor to human disease, and effective therapies are needed. Here, we show that cyclic GMP-selective phosphodiesterase 9A inhibition (PDE9-I) in both male and ovariectomized female mice suppresses preestablished severe diet-induced obesity/CMS with or without superimposed mild cardiac pressure load. PDE9-I reduces total body, inguinal, hepatic, and myocardial fat; stimulates mitochondrial activity in brown and white fat; and improves CMS, without significantly altering activity or food intake. PDE9 localized at mitochondria, and its inhibition in vitro stimulated lipolysis in a PPARα-dependent manner and increased mitochondrial respiration in both adipocytes and myocytes. PPARα upregulation was required to achieve the lipolytic, antiobesity, and metabolic effects of PDE9-I. All these PDE9-I-induced changes were not observed in obese/CMS nonovariectomized females, indicating a strong sexual dimorphism. We found that PPARα chromatin binding was reoriented away from fat metabolism-regulating genes when stimulated in the presence of coactivated estrogen receptor-α, and this may underlie the dimorphism. These findings have translational relevance given that PDE9-I is already being studied in humans for indications including heart failure, and efficacy against obesity/CMS would enhance its therapeutic utility.
Collapse
Affiliation(s)
| | | | | | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ryan P Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Hildur Knutsdottir
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vivek P Jani
- Division of Cardiology, Department of Medicine, and.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | - Jon A Gangoiti
- UCSD Biochemical Genetics and Metabolomics Laboratory and
| | - Dorothy D Sears
- Department of Medicine, UCSD, La Jolla, California, USA.,College of Health Solutions, Arizona State University, Phoenix, Arizona, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, and.,Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Diabetes Mellitus and Heart Failure. J Clin Med 2021; 10:jcm10163682. [PMID: 34441977 PMCID: PMC8396967 DOI: 10.3390/jcm10163682] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus (DM) is a major risk factor for new-onset heart failure (HF) and vice versa. The pathogenesis of new-onset HF in DM is complex and has been largely attributed to the toxic cardiovascular effects of hyperglycemia and relevant metabolic abnormalities (diabetic cardiomyopathy) as well as the frequently coexisting morbidities such as hypertension (HTN), coronary artery disease (CAD), and diabetic nephropathy. In patients with type 1 DM (T1DM), HF develops in the setting of a dysregulated immune response, whereas in most patients with type 2 DM (T2DM), against a background of overweight/obesity. HF prevention in DM is feasible with rigorous treatment of cardiovascular risk factors and selective antidiabetic agents. Conversely, development of new-onset T2DM in HF (cardiogenic DM) is common and has been attributed to an increase in the resistance to insulin, especially in the skeletal muscle, liver, and adipose tissue as well as in diminished insulin secretory response to hyperglycemia by pancreatic β-cells. Cardiogenic DM further deteriorates cardiac dysfunction and adversely affects outcome in HF. Novel lifesaving medications employed in HF management such as sacubitril/valsartan and sodium glucose cotransporter 2 inhibitors (SGLT-2i) have a favorable metabolic profile and lower the incidence of cardiogenic diabetes. Whether mitigation of cardiogenic DM should be a treatment target in HF deserves further investigation.
Collapse
|
19
|
Function and regulation of corin in physiology and disease. Biochem Soc Trans 2021; 48:1905-1916. [PMID: 33125488 DOI: 10.1042/bst20190760] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/19/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Atrial natriuretic peptide (ANP) is of major importance in the maintenance of electrolyte balance and normal blood pressure. Reduced plasma ANP levels are associated with the increased risk of cardiovascular disease. Corin is a type II transmembrane serine protease that converts the ANP precursor to mature ANP. Corin deficiency prevents ANP generation and alters electrolyte and body fluid homeostasis. Corin is synthesized as a zymogen that is proteolytically activated on the cell surface. Factors that disrupt corin folding, intracellular trafficking, cell surface expression, and zymogen activation are expected to impair corin function. To date, CORIN variants that reduce corin activity have been identified in hypertensive patients. In addition to the heart, corin expression has been detected in non-cardiac tissues, where corin and ANP participate in diverse physiological processes. In this review, we summarize the current knowledge in corin biosynthesis and post-translational modifications. We also discuss tissue-specific corin expression and function in physiology and disease.
Collapse
|
20
|
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 2021; 474:5-19. [PMID: 34173888 DOI: 10.1007/s00424-021-02596-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [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
|
21
|
Bełtowski J. Salt Intake, Aldosterone Secretion, and Obesity: Role in the Pathogenesis of Resistant Hypertension. Am J Hypertens 2021; 34:588-590. [PMID: 33438728 DOI: 10.1093/ajh/hpab015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jerzy Bełtowski
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
22
|
Natriuretic Peptides Regulate Prostate Cells Inflammatory Behavior: Potential Novel Anticancer Agents for Prostate Cancer. Biomolecules 2021; 11:biom11060794. [PMID: 34070682 PMCID: PMC8228623 DOI: 10.3390/biom11060794] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 02/05/2023] Open
Abstract
Inflammation, by inducing a tumor-promoting microenvironment, is a hallmark for prostate cancer (PCa) progression. NOD-like receptor protein 3 (NLRP3)-inflammasome activation, interleukin-1β (IL-1β) secretion, and cancer cell-released extracellular vesicles (EVs) contribute to the establishment of tumor microenvironment. We have shown that PC3-derived EVs (PC3-EVs) activate inflammasome cascade in non-cancerous PNT2 cells. It is known that the endogenous biomolecules and Natriuretic Peptides (NPs), such as ANP and BNP, inhibit inflammasome activation in immune cells. Here we investigated whether ANP and BNP modify PCa inflammatory phenotype in vitro. By using PNT2, LNCaP, and PC3 cell lines, which model different PCa progression stages, we analyzed inflammasome activation and the related pathways by Western blot and IL-1β secretion by ELISA. We found that tumor progression is characterized by constitutive inflammasome activation, increased IL-1β secretion, and reduced endogenous NPs expression. The administration of exogenous ANP and BNP, via p38-MAPK or ERK1/2-MAPK, by inducing NLRP3 phosphorylation, counteract inflammasome activation and IL-1β maturation in PC3 and PC3-EVs-treated PNT2 cells, respectively. Our results demonstrate that NPs, by interfering with cell-specific signaling pathways, exert pleiotropic anti-inflammatory effects converging toward inflammasome phosphorylation and suggest that NPs can be included in a drug repurposing process for PCa.
Collapse
|
23
|
Dubé MP, Lemaçon A, Barhdadi A, Lemieux Perreault LP, Oussaïd E, Asselin G, Provost S, Sun M, Sandoval J, Legault MA, Mongrain I, Dubois A, Valois D, Dedelis E, Lousky J, Choi J, Goulet E, Savard C, Chicoine LM, Cossette M, Chabot-Blanchet M, Guertin MC, de Denus S, Bouabdallaoui N, Marchand R, Bassevitch Z, Nozza A, Gaudet D, L'Allier PL, Hussin J, Boivin G, Busseuil D, Tardif JC. Genetics of symptom remission in outpatients with COVID-19. Sci Rep 2021; 11:10847. [PMID: 34035401 PMCID: PMC8149390 DOI: 10.1038/s41598-021-90365-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/11/2021] [Indexed: 01/09/2023] Open
Abstract
We conducted a genome-wide association study of time to remission of COVID-19 symptoms in 1723 outpatients with at least one risk factor for disease severity from the COLCORONA clinical trial. We found a significant association at 5p13.3 (rs1173773; P = 4.94 × 10-8) near the natriuretic peptide receptor 3 gene (NPR3). By day 15 of the study, 44%, 54% and 59% of participants with 0, 1, or 2 copies of the effect allele respectively, had symptom remission. In 851 participants not treated with colchicine (placebo), there was a significant association at 9q33.1 (rs62575331; P = 2.95 × 10-8) in interaction with colchicine (P = 1.19 × 10-5) without impact on risk of hospitalisations, highlighting a possibly shared mechanistic pathway. By day 15 of the study, 46%, 62% and 64% of those with 0, 1, or 2 copies of the effect allele respectively, had symptom remission. The findings need to be replicated and could contribute to the biological understanding of COVID-19 symptom remission.
Collapse
Affiliation(s)
- Marie-Pierre Dubé
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada. .,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada. .,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada.
| | - Audrey Lemaçon
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Amina Barhdadi
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Louis-Philippe Lemieux Perreault
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Essaïd Oussaïd
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Géraldine Asselin
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Sylvie Provost
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Maxine Sun
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Johanna Sandoval
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Marc-André Legault
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada.,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Ian Mongrain
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Anick Dubois
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Diane Valois
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Emma Dedelis
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Jennifer Lousky
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Julie Choi
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | - Elisabeth Goulet
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada
| | - Christiane Savard
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada
| | - Lea-Mei Chicoine
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada
| | - Mariève Cossette
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Montreal Health Innovations Coordinating Centre, Montreal, Canada
| | - Malorie Chabot-Blanchet
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Montreal Health Innovations Coordinating Centre, Montreal, Canada
| | - Marie-Claude Guertin
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Montreal Health Innovations Coordinating Centre, Montreal, Canada
| | - Simon de Denus
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada.,Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | | | - Richard Marchand
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada
| | - Zohar Bassevitch
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Montreal Health Innovations Coordinating Centre, Montreal, Canada
| | - Anna Nozza
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Montreal Health Innovations Coordinating Centre, Montreal, Canada
| | - Daniel Gaudet
- Ecogene-21 and Department of Medicine, Université de Montréal, Chicoutimi, Canada
| | | | - Julie Hussin
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada.,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Guy Boivin
- Centre Hospitalier de l'Université Laval, Quebec City, Canada
| | - David Busseuil
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8, Canada. .,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada.
| |
Collapse
|
24
|
Stacey RB, Hundley WG. Integrating Measures of Myocardial Fibrosis in the Transition from Hypertensive Heart Disease to Heart Failure. Curr Hypertens Rep 2021; 23:22. [PMID: 33881630 DOI: 10.1007/s11906-021-01135-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2021] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW This review aims to summarize recent developments in identifying and quantifying both the presence and amount of myocardial fibrosis by imaging and biomarkers. Further, this review seeks to describe in general ways how this information may be used to identify hypertension and the transition to heart failure with preserved ejection fraction. RECENT FINDINGS Recent studies using cardiac magnetic resonance imaging highlight the progressive nature of fibrosis from normal individuals to those with hypertension to those with clinical heart failure. However, separating hypertensive patients from those with heart failure remains challenging. Recent studies involving echocardiography show the subclinical myocardial strain changes between hypertensive heart disease and heart failure. Lastly, recent studies highlight the potential use of biomarkers to identify those with hypertension at the greatest risk of developing heart failure. In light of the heterogeneous nature between hypertension and heart failure with preserved ejection fraction, an integrated approach with cardiac imaging and biomarker analysis may enable clinicians and investigators to more accurately characterize, prevent, and treat heart failure in those with hypertension.
Collapse
Affiliation(s)
- R Brandon Stacey
- Division of Cardiovascular Medicine, Wake Forest University School of Medicine, Watlington Hall, Medical Center Boulevard, Winston-Salem, NC, 27157-1045, USA.
| | - W Gregory Hundley
- Division of Cardiovascular Medicine, Wake Forest University School of Medicine, Watlington Hall, Medical Center Boulevard, Winston-Salem, NC, 27157-1045, USA.,Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| |
Collapse
|
25
|
Niu Y, Zhang S, Gu X, Zhou T, Li F, Liu M, Wu Q, Dong N. Recombinant Soluble Corin Improves Cardiac Function in Mouse Models of Heart Failure. J Am Heart Assoc 2021; 10:e019961. [PMID: 33759549 PMCID: PMC8174325 DOI: 10.1161/jaha.120.019961] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Corin is a transmembrane protease that activates ANP and BNP (atrial and B‐type natriuretic peptides). Impaired corin expression and function are associated with heart failure. In this study, we characterized a soluble form of corin (sCorin) and examined its effects on cardiac morphology and function in mouse heart failure models. Methods and Results sCorin, consisting of the full‐length extracellular fragment of human corin with an engineered activation site, was expressed in Chinese hamster ovary cells, purified from the conditioned medium with affinity chromatography, and characterized in pro‐ANP processing assays in vitro and pharmacokinetic studies in mice. Effects of sCorin on mouse models of heart failure induced by left coronary artery ligation and transverse aortic constriction were assessed by ELISA analysis of plasma markers, histologic examination, and echocardiography. We showed that purified and activated sCorin converted pro‐ANP to ANP that stimulated cGMP production in cultured cells. In mice, intravenously and intraperitoneally administered sCorin had plasma half‐lives of 3.5±0.1 and 8.3±0.3 hour, respectively. In the mouse heart failure models, intraperitoneal injection of sCorin increased plasma ANP, BNP, and cGMP levels; lowered plasma levels of NT‐proANP (N‐terminal‐pro‐ANP), angiotensin II, and aldosterone; reduced cardiac hypertrophy and fibrosis; and improved cardiac function. Conclusions We show that sCorin treatment enhanced natriuretic peptide processing and activity, suppressed the renin‐angiotensin‐aldosterone system, and improved cardiac morphology and function in mice with failing hearts.
Collapse
Affiliation(s)
- Yayan Niu
- Cyrus Tang Hematology Center Collaborative Innovation Center of Hematology State Key Laboratory of Radiation Medicine and Prevention The First Affiliated HospitalMedical CollegeSoochow University Suzhou China.,MOH Key Laboratory of Thrombosis and Hemostasis Jiangsu Institute of HematologySoochow University Suzhou China
| | - Shengnan Zhang
- Cyrus Tang Hematology Center Collaborative Innovation Center of Hematology State Key Laboratory of Radiation Medicine and Prevention The First Affiliated HospitalMedical CollegeSoochow University Suzhou China.,MOH Key Laboratory of Thrombosis and Hemostasis Jiangsu Institute of HematologySoochow University Suzhou China
| | - Xiabing Gu
- Cyrus Tang Hematology Center Collaborative Innovation Center of Hematology State Key Laboratory of Radiation Medicine and Prevention The First Affiliated HospitalMedical CollegeSoochow University Suzhou China.,MOH Key Laboratory of Thrombosis and Hemostasis Jiangsu Institute of HematologySoochow University Suzhou China
| | - Tiantian Zhou
- Cyrus Tang Hematology Center Collaborative Innovation Center of Hematology State Key Laboratory of Radiation Medicine and Prevention The First Affiliated HospitalMedical CollegeSoochow University Suzhou China
| | - Feng Li
- Cyrus Tang Hematology Center Collaborative Innovation Center of Hematology State Key Laboratory of Radiation Medicine and Prevention The First Affiliated HospitalMedical CollegeSoochow University Suzhou China.,MOH Key Laboratory of Thrombosis and Hemostasis Jiangsu Institute of HematologySoochow University Suzhou China
| | - Meng Liu
- Cyrus Tang Hematology Center Collaborative Innovation Center of Hematology State Key Laboratory of Radiation Medicine and Prevention The First Affiliated HospitalMedical CollegeSoochow University Suzhou China
| | - Qingyu Wu
- Cyrus Tang Hematology Center Collaborative Innovation Center of Hematology State Key Laboratory of Radiation Medicine and Prevention The First Affiliated HospitalMedical CollegeSoochow University Suzhou China.,Cardiovascular & Metabolic Sciences Lerner Research InstituteCleveland Clinic Cleveland OH
| | - Ningzheng Dong
- Cyrus Tang Hematology Center Collaborative Innovation Center of Hematology State Key Laboratory of Radiation Medicine and Prevention The First Affiliated HospitalMedical CollegeSoochow University Suzhou China.,MOH Key Laboratory of Thrombosis and Hemostasis Jiangsu Institute of HematologySoochow University Suzhou China
| |
Collapse
|
26
|
Mezzasoma L, Talesa VN, Romani R, Bellezza I. ANP and BNP Exert Anti-Inflammatory Action via NPR-1/cGMP Axis by Interfering with Canonical, Non-Canonical, and Alternative Routes of Inflammasome Activation in Human THP1 Cells. Int J Mol Sci 2020; 22:ijms22010024. [PMID: 33375031 PMCID: PMC7792787 DOI: 10.3390/ijms22010024] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/11/2022] Open
Abstract
Dysregulated inflammasome activation and interleukin (IL)-1β production are associated with several inflammatory disorders. Three different routes can lead to inflammasome activation: a canonical two-step, a non-canonical Caspase-4/5- and Gasdermin D-dependent, and an alternative Caspase-8-mediated pathway. Natriuretic Peptides (NPs), Atrial Natriuretic Peptide (ANP) and B-type Natriuretic Peptide (BNP), binding to Natriuretic Peptide Receptor-1 (NPR-1), signal by increasing cGMP (cyclic guanosine monophosphate) levels that, in turn, stimulate cGMP-dependent protein kinase-I (PKG-I). We previously demonstrated that, by counteracting inflammasome activation, NPs inhibit IL-1β secretion. Here we aimed to decipher the molecular mechanism underlying NPs effects on THP-1 cells stimulated with lipopolysaccharide (LPS) + ATP. Involvement of cGMP and PKG-I were assessed pre-treating THP-1 cells with the membrane-permeable analogue, 8-Br-cGMP, and the specific inhibitor KT-5823, respectively. We found that NPs, by activating NPR-1/cGMP/PKG-I axis, lead to phosphorylation of NLRP3 at Ser295 and to inflammasome platform disassembly. Moreover, by increasing intracellular cGMP levels and activating phosphodiesterases, NPs interfere with both Gasdermin D and Caspase-8 cleavage, indicating that they disturb non-canonical and alternative routes of inflammasome activation. These results showed that ANP and BNP anti-inflammatory and immunomodulatory actions may involve the inhibition of all the known routes of inflammasome activation. Thus, NPs might be proposed for the treatment of the plethora of diseases caused by a dysregulated inflammasome activation.
Collapse
|
27
|
Senesi P, Luzi L, Terruzzi I. Adipokines, Myokines, and Cardiokines: The Role of Nutritional Interventions. Int J Mol Sci 2020; 21:ijms21218372. [PMID: 33171610 PMCID: PMC7664629 DOI: 10.3390/ijms21218372] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
It is now established that adipose tissue, skeletal muscle, and heart are endocrine organs and secrete in normal and in pathological conditions several molecules, called, respectively, adipokines, myokines, and cardiokines. These secretory proteins constitute a closed network that plays a crucial role in obesity and above all in cardiac diseases associated with obesity. In particular, the interaction between adipokines, myokines, and cardiokines is mainly involved in inflammatory and oxidative damage characterized obesity condition. Identifying new therapeutic agents or treatment having a positive action on the expression of these molecules could have a key positive effect on the management of obesity and its cardiac complications. Results from recent studies indicate that several nutritional interventions, including nutraceutical supplements, could represent new therapeutic agents on the adipo-myo-cardiokines network. This review focuses the biological action on the main adipokines, myokines and cardiokines involved in obesity and cardiovascular diseases and describe the principal nutraceutical approaches able to regulate leptin, adiponectin, apelin, irisin, natriuretic peptides, and follistatin-like 1 expression.
Collapse
Affiliation(s)
- Pamela Senesi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20131 Milan, Italy; (P.S.); (L.L.)
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, 20138 Milan, Italy
| | - Livio Luzi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20131 Milan, Italy; (P.S.); (L.L.)
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, 20138 Milan, Italy
| | - Ileana Terruzzi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20131 Milan, Italy; (P.S.); (L.L.)
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, 20138 Milan, Italy
- Correspondence:
| |
Collapse
|