151
|
Hill MC, Kadow ZA, Long H, Morikawa Y, Martin TJ, Birks EJ, Campbell KS, Nerbonne J, Lavine K, Wadhwa L, Wang J, Turaga D, Adachi I, Martin JF. Integrated multi-omic characterization of congenital heart disease. Nature 2022; 608:181-191. [PMID: 35732239 PMCID: PMC10405779 DOI: 10.1038/s41586-022-04989-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/16/2022] [Indexed: 11/09/2022]
Abstract
The heart, the first organ to develop in the embryo, undergoes complex morphogenesis that when defective results in congenital heart disease (CHD). With current therapies, more than 90% of patients with CHD survive into adulthood, but many suffer premature death from heart failure and non-cardiac causes1. Here, to gain insight into this disease progression, we performed single-nucleus RNA sequencing on 157,273 nuclei from control hearts and hearts from patients with CHD, including those with hypoplastic left heart syndrome (HLHS) and tetralogy of Fallot, two common forms of cyanotic CHD lesions, as well as dilated and hypertrophic cardiomyopathies. We observed CHD-specific cell states in cardiomyocytes, which showed evidence of insulin resistance and increased expression of genes associated with FOXO signalling and CRIM1. Cardiac fibroblasts in HLHS were enriched in a low-Hippo and high-YAP cell state characteristic of activated cardiac fibroblasts. Imaging mass cytometry uncovered a spatially resolved perivascular microenvironment consistent with an immunodeficient state in CHD. Peripheral immune cell profiling suggested deficient monocytic immunity in CHD, in agreement with the predilection in CHD to infection and cancer2. Our comprehensive phenotyping of CHD provides a roadmap towards future personalized treatments for CHD.
Collapse
MESH Headings
- Bone Morphogenetic Protein Receptors/metabolism
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/immunology
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cardiomyopathy, Hypertrophic/genetics
- Cardiomyopathy, Hypertrophic/immunology
- Cardiomyopathy, Hypertrophic/metabolism
- Cardiomyopathy, Hypertrophic/pathology
- Disease Progression
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Forkhead Transcription Factors/metabolism
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/immunology
- Heart Defects, Congenital/metabolism
- Heart Defects, Congenital/pathology
- Humans
- Hypoplastic Left Heart Syndrome/genetics
- Hypoplastic Left Heart Syndrome/immunology
- Hypoplastic Left Heart Syndrome/metabolism
- Hypoplastic Left Heart Syndrome/pathology
- Image Cytometry
- Insulin Resistance
- Monocytes/immunology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phenotype
- RNA-Seq
- Signal Transduction/genetics
- Single-Cell Analysis
- Tetralogy of Fallot/genetics
- Tetralogy of Fallot/immunology
- Tetralogy of Fallot/metabolism
- Tetralogy of Fallot/pathology
- YAP-Signaling Proteins/metabolism
Collapse
Affiliation(s)
- Matthew C Hill
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Zachary A Kadow
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | - Hali Long
- Interdepartmental Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
| | | | - Thomas J Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Emma J Birks
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Kenneth S Campbell
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Jeanne Nerbonne
- Center for Cardiovascular Research, Departmental of Medicine, Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Kory Lavine
- Center for Cardiovascular Research, Departmental of Medicine, Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Lalita Wadhwa
- Section of Cardiothoracic Surgery, Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Diwakar Turaga
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Iki Adachi
- Section of Cardiothoracic Surgery, Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - James F Martin
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.
- Interdepartmental Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA.
- Texas Heart Institute, Houston, TX, USA.
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA.
- Center for Organ Repair and Renewal, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
152
|
Walls GM, O'Kane R, Ghita M, Kuburas R, McGarry CK, Cole AJ, Jain S, Butterworth KT. Murine models of radiation cardiotoxicity: A systematic review and recommendations for future studies. Radiother Oncol 2022; 173:19-31. [PMID: 35533784 DOI: 10.1016/j.radonc.2022.04.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/13/2022] [Accepted: 04/29/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE The effects of radiation on the heart are dependent on dose, fractionation, overall treatment time, and pre-existing cardiovascular pathology. Murine models have played a central role in improving our understanding of the radiation response of the heart yet a wide range of exposure parameters have been used. We evaluated the study design of published murine cardiac irradiation experiments to assess gaps in the literature and to suggest guidance for the harmonisation of future study reporting. METHODS AND MATERIALS A systematic review of mouse/rat studies published 1981-2021 that examined the effect of radiation on the heart was performed. The protocol was published on PROSPERO (CRD42021238921) and the findings were reported in accordance with the PRISMA guidance. Risk of bias was assessed using the SYRCLE checklist. RESULTS 159 relevant full-text original articles were reviewed. The heart only was the target volume in 67% of the studies and simulation details were unavailable for 44% studies. Dosimetry methods were reported in 31% studies. The pulmonary effects of whole and partial heart irradiation were reported in 13% studies. Seventy-eight unique dose-fractionation schedules were evaluated. Large heterogeneity was observed in the endpoints measured, and the reporting standards were highly variable. CONCLUSIONS Current murine models of radiation cardiotoxicity cover a wide range of irradiation configurations and latency periods. There is a lack of evidence describing clinically relevant dose-fractionations, circulating biomarkers and radioprotectants. Recommendations for the consistent reporting of methods and results of in vivo cardiac irradiation studies are made to increase their suitability for informing the design of clinical studies.
Collapse
Affiliation(s)
- Gerard M Walls
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland; Cancer Centre Belfast City Hospital, Belfast Health & Social Care Trust, Lisburn Road, Belfast, Northern Ireland.
| | - Reagan O'Kane
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland
| | - Mihaela Ghita
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland
| | - Refik Kuburas
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland
| | - Conor K McGarry
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland; Cancer Centre Belfast City Hospital, Belfast Health & Social Care Trust, Lisburn Road, Belfast, Northern Ireland
| | - Aidan J Cole
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland; Cancer Centre Belfast City Hospital, Belfast Health & Social Care Trust, Lisburn Road, Belfast, Northern Ireland
| | - Suneil Jain
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland; Cancer Centre Belfast City Hospital, Belfast Health & Social Care Trust, Lisburn Road, Belfast, Northern Ireland
| | - Karl T Butterworth
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland
| |
Collapse
|
153
|
Exogenous ANP Treatment Ameliorates Myocardial Insulin Resistance and Protects against Ischemia-Reperfusion Injury in Diet-Induced Obesity. Int J Mol Sci 2022; 23:ijms23158373. [PMID: 35955507 PMCID: PMC9369294 DOI: 10.3390/ijms23158373] [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: 07/04/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
Increasing evidence suggests natriuretic peptides (NPs) coordinate interorgan metabolic crosstalk. We recently reported exogenous ANP treatment ameliorated systemic insulin resistance by inducing adipose tissue browning and attenuating hepatic steatosis in diet-induced obesity (DIO). We herein investigated whether ANP treatment also ameliorates myocardial insulin resistance, leading to cardioprotection during ischemia-reperfusion injury (IRI) in DIO. Mice fed a high-fat diet (HFD) or normal-fat diet for 13 weeks were treated with or without ANP infusion subcutaneously for another 3 weeks. Left ventricular BNP expression was substantially reduced in HFD hearts. Intraperitoneal-insulin-administration-induced Akt phosphorylation was impaired in HFD hearts, which was restored by ANP treatment, suggesting that ANP treatment ameliorated myocardial insulin resistance. After ischemia-reperfusion using the Langendorff model, HFD impaired cardiac functional recovery with a corresponding increased infarct size. However, ANP treatment improved functional recovery and reduced injury while restoring impaired IRI-induced Akt phosphorylation in HFD hearts. Myocardial ultrastructural analyses showed increased peri-mitochondrial lipid droplets with concomitantly decreased ATGL and HSL phosphorylation levels in ANP-treated HFD, suggesting that ANP protects mitochondria from lipid overload by trapping lipids. Accordingly, ANP treatment attenuated mitochondria cristae disruption after IRI in HFD hearts. In summary, exogenous ANP treatment ameliorates myocardial insulin resistance and protects against IRI associated with mitochondrial ultrastructure modifications in DIO. Replenishing biologically active NPs substantially affects HFD hearts in which endogenous NP production is impaired.
Collapse
|
154
|
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: 21] [Impact Index Per Article: 10.5] [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
|
155
|
Zhang X, Li W, Zhou T, Liu M, Wu Q, Dong N. Corin Deficiency Alters Adipose Tissue Phenotype and Impairs Thermogenesis in Mice. BIOLOGY 2022; 11:biology11081101. [PMID: 35892957 PMCID: PMC9329919 DOI: 10.3390/biology11081101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022]
Abstract
Atrial natriuretic peptide (ANP) is a key regulator in body fluid balance and cardiovascular biology. In addition to its role in enhancing natriuresis and vasodilation, ANP increases lipolysis and thermogenesis in adipose tissue. Corin is a protease responsible for ANP activation. It remains unknown if corin has a role in regulating adipose tissue function. Here, we examined adipose tissue morphology and function in corin knockout (KO) mice. We observed increased weights and cell sizes in white adipose tissue (WAT), decreased levels of uncoupling protein 1 (Ucp1), a brown adipocyte marker in WAT and brown adipose tissue (BAT), and suppressed thermogenic gene expression in BAT from corin KO mice. At regular room temperature, corin KO and wild-type mice had similar metabolic rates. Upon cold exposure at 4 °C, corin KO mice exhibited impaired thermogenic responses and developed hypothermia. In BAT from corin KO mice, the signaling pathway of p38 mitogen-activated protein kinase, peroxisome proliferator-activated receptor c coactivator 1a, and Ucp1 was impaired. In cell culture, ANP treatment increased Ucp1 expression in BAT-derived adipocytes from corin KO mice. These data indicate that corin mediated-ANP activation is an important hormonal mechanism in regulating adipose tissue function and body temperature upon cold exposure in mice.
Collapse
Affiliation(s)
- Xianrui Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- MOH Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wenguo Li
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- MOH Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
| | - Meng Liu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- Correspondence: (Q.W.); (N.D.)
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- MOH Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Correspondence: (Q.W.); (N.D.)
| |
Collapse
|
156
|
Palà E, Bustamante A, Pagola J, Juega J, Francisco-Pascual J, Penalba A, Rodriguez M, De Lera Alfonso M, Arenillas JF, Cabezas JA, Pérez-Sánchez S, Moniche F, de Torres R, González-Alujas T, Clúa-Espuny JL, Ballesta-Ors J, Ribas D, Acosta J, Pedrote A, Gonzalez-Loyola F, Gentile Lorente D, Ángel Muñoz M, Molina CA, Montaner J. Blood-Based Biomarkers to Search for Atrial Fibrillation in High-Risk Asymptomatic Individuals and Cryptogenic Stroke Patients. Front Cardiovasc Med 2022; 9:908053. [PMID: 35859587 PMCID: PMC9289129 DOI: 10.3389/fcvm.2022.908053] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/15/2022] [Indexed: 11/24/2022] Open
Abstract
Background Atrial fibrillation (AF) increases the risk of ischemic stroke in asymptomatic individuals and may be the underlying cause of many cryptogenic strokes. We aimed to test the usefulness of candidate blood-biomarkers related to AF pathophysiology in two prospective cohorts representative of those populations. Methods Two hundred seventy-four subjects aged 65–75 years with hypertension and diabetes from the AFRICAT cohort, and 218 cryptogenic stroke patients aged >55 years from the CRYPTO-AF cohort were analyzed. AF was assessed by 4 weeks of monitoring with a wearable Holter device (NuuboTM™). Blood was collected immediately before monitoring started. 10 candidate biomarkers were measured by automated immunoassays (Roche, Penzberg) in the plasma of all patients. Univariate and logistic regression analyses were performed in each cohort separately. Results Atrial fibrillation detection rate was 12.4% (AFRICAT cohort) and 22.9% (CRYPTO-AF cohort). 4 biomarkers were significantly increased in asymptomatic individuals with AF [Troponin-T, Angiopoietin-2 (Ang-2), Endocan, and total N-terminal pro-B type natriuretic peptide (NT-proBNP)] and 7 biomarkers showed significantly higher concentrations in cryptogenic stroke patients with AF detection [growth differentiation factor 15, interleukin 6, Troponin-T, Ang-2, Bone morphogenic protein 10, Dickkopf-related protein 3 (DKK-3), and total NT-proBNP]. The models including Ang-2 and total NT-proBNP [AUC 0.764 (0.665–0.863)], and Ang-2 and DKK-3 [AUC = 0.733 (0.654–0.813)], together with age and sex, showed the best performance to detect AF in high-risk asymptomatic individuals, and in cryptogenic stroke patients, respectively. Conclusion Blood-biomarkers, in particular, total NT-proBNP, DKK-3, and Ang-2, were associated with AF reflecting two mechanistically different pathways involved in AF pathophysiology (AF stretch and vascular changes). The combination of these biomarkers could be useful in AF screening strategies in the primary care setting and also for searching AF after cryptogenic stroke.
Collapse
Affiliation(s)
- Elena Palà
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Stroke Unit, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Jorge Pagola
- Stroke Unit, Medicine Department, Vall d'Hebrón Hospital and Autonomous University of Barcelona, Barcelona, Spain
| | - Jesus Juega
- Stroke Unit, Medicine Department, Vall d'Hebrón Hospital and Autonomous University of Barcelona, Barcelona, Spain
| | - Jaume Francisco-Pascual
- Arrhythmia Unit-Cardiology Department, Vall d'Hebrón Hospital, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - Anna Penalba
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maite Rodriguez
- Stroke Unit, Medicine Department, Vall d'Hebrón Hospital and Autonomous University of Barcelona, Barcelona, Spain
| | | | - Juan F Arenillas
- Stroke Unit, University Hospital of Valladolid, Valladolid, Spain
| | | | | | | | - Reyes de Torres
- Stroke Unit, University Hospital Virgen Macarena, Seville, Spain
| | - Teresa González-Alujas
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain.,Echocardiography Lab Cardiology Department, Vall d'Hebrón Hospital, Barcelona, Spain
| | - Josep Lluís Clúa-Espuny
- Equip d'Atenció Primària Tortosa Est, SAP Terres de l'Ebre, Institut Català de la Salut, Tortosa, Spain.,Institut d'Investigació en Atenció Primària IDIAP Jordi Gol, Ebrictus Group, Barcelona, Spain
| | - Juan Ballesta-Ors
- Institut d'Investigació en Atenció Primària IDIAP Jordi Gol, Ebrictus Group, Barcelona, Spain
| | - Domingo Ribas
- EAP Sant Pere i Sant Pau, DAP Camp de Tarragona, Institut Català de la Salut, Tarragona, Spain
| | - Juan Acosta
- Department of Cardiology, Hospital Universitario Virgen del Rocio, Seville, Spain
| | - Alonso Pedrote
- Department of Cardiology, Hospital Universitario Virgen del Rocio, Seville, Spain
| | - Felipe Gonzalez-Loyola
- Gerència Atenció Primària de Barcelona, Institut Català de la Salut, Barcelona, Spain.,Institut d'Investigació en Atenció Primària IDIAP Jordi Gol, Unitat Suport Recerca Barcelona, Barcelona, Spain
| | - Delicia Gentile Lorente
- Institut d'Investigació en Atenció Primària IDIAP Jordi Gol, Ebrictus Group, Barcelona, Spain.,Cardiology Department, Hospital Verge de la Cinta, Institut Català de la Salut, Tortosa, Spain
| | - Miguel Ángel Muñoz
- Gerència Atenció Primària de Barcelona, Institut Català de la Salut, Barcelona, Spain.,Institut d'Investigació en Atenció Primària IDIAP Jordi Gol, Unitat Suport Recerca Barcelona, Barcelona, Spain
| | - Carlos A Molina
- Stroke Unit, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
157
|
Tang HM, Talbot CC, Fung MC, Tang HL. Transcriptomic study of anastasis for reversal of ethanol-induced apoptosis in mouse primary liver cells. Sci Data 2022; 9:418. [PMID: 35851273 PMCID: PMC9293995 DOI: 10.1038/s41597-022-01470-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 05/27/2022] [Indexed: 11/30/2022] Open
Abstract
Anastasis is a cell recovery mechanism that rescues dying cells from the brink of death. Reversal of apoptosis is the first example of anastasis. Here, we describe a comprehensive dataset containing time-course mRNA expression profiles for reversal of ethanol-induced apoptosis in mouse primary liver cells in νitro. This transcriptome dataset includes the conditions of the untreated cells, cells undergoing apoptosis triggered by incubating with cell death inducer of 4.5% ethanol for 5 hours, and apoptosis reversal of ethanol-induced cells at the early (3rd hour), middle (6th hour), and late (24th, 48th hour) stages after being washed with and incubated in fresh cell culture medium. By comparing this dataset with the transcriptomic profiles of other anastasis models generated with different combinations of cell types and cell death inducers, investigators can identify the key regulators governing reversal of apoptosis and other reversible cell death processes. Therefore, reusing or reanalysing this dataset will facilitate the future studies on the physiological, pathological, and therapeutic implications of anastasis.
Collapse
Affiliation(s)
- Ho Man Tang
- School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - C Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ming Chiu Fung
- School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| | - Ho Lam Tang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
158
|
Nishikimi T, Nakagawa Y. B-Type Natriuretic Peptide (BNP) Revisited—Is BNP Still a Biomarker for Heart Failure in the Angiotensin Receptor/Neprilysin Inhibitor Era? BIOLOGY 2022; 11:biology11071034. [PMID: 36101415 PMCID: PMC9312360 DOI: 10.3390/biology11071034] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/28/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Active BNP-32, less active proBNP-108, and inactive N-terminal proBNP-76 all circulate in the blood. The circulating protease neprilysin has lower substrate specificity for BNP than ANP, while proBNP and N-terminal proBNP are not degraded by neprilysin. Currently available BNP immunoassays react with both mature BNP and proBNP; therefore, measured plasma BNP is mature BNP + proBNP. Because ARNI administration increases mature BNP, measured plasma BNP initially increases with ARNI administration by the amount of the increase in mature BNP. Later, ARNI administration reduces myocardial wall stress, and the resultant reduction in BNP production more than offsets the increase of mature BNP due to inhibition of degradation by neprilysin, resulting in lower plasma BNP levels. In the ARNI era, BNP remains a useful biomarker for heart failure, though mild increases early during ARNI administration should be taken into consideration. Abstract Myocardial wall stress, cytokines, hormones, and ischemia all stimulate B-type (or brain) natriuretic peptide (BNP) gene expression. Within the myocardium, ProBNP-108, a BNP precursor, undergoes glycosylation, after which a portion is cleaved by furin into mature BNP-32 and N-terminal proBNP-76, depending on the glycosylation status. As a result, active BNP, less active proBNP, and inactive N-terminal proBNP all circulate in the blood. There are three major pathways for BNP clearance: (1) cellular internalization via natriuretic peptide receptor (NPR)-A and NPR-C; (2) degradation by proteases in the blood, including neprilysin, dipeptidyl-peptidase-IV, insulin degrading enzyme, etc.; and (3) excretion in the urine. Because neprilysin has lower substrate specificity for BNP than atrial natriuretic peptide (ANP), the increase in plasma BNP after angiotensin receptor neprilysin inhibitor (ARNI) administration is much smaller than the increase in plasma ANP. Currently available BNP immunoassays react with both mature BNP and proBNP. Therefore, BNP measured with an immunoassay is mature BNP + proBNP. ARNI administration increases mature BNP but not proBNP, as the latter is not degraded by neprilysin. Consequently, measured plasma BNP initially increases with ARNI administration by the amount of the increase in mature BNP. Later, ARNI reduces myocardial wall stress, and the resultant reduction in BNP production more than offsets the increase in mature BNP mediated by inhibiting degradation by neprilysin, which lowers plasma BNP levels. These results suggest that even in the ARNI era, BNP can be used for diagnosis and assessment of the pathophysiology and prognosis of heart failure, though the mild increases early during ARNI administration should be taken into consideration.
Collapse
Affiliation(s)
- Toshio Nishikimi
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan;
- Department of Medicine, Wakakusa Tatsuma Rehabilitation Hospital, 1580 Ooaza Tatsuma, Daito City 574-0012, Japan
- Correspondence: ; Tel.: +81-75-751-4287
| | - Yasuaki Nakagawa
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan;
| |
Collapse
|
159
|
Nakagawa H, Saito Y. Roles of Natriuretic Peptides and the Significance of Neprilysin in Cardiovascular Diseases. BIOLOGY 2022; 11:1017. [PMID: 36101398 PMCID: PMC9312343 DOI: 10.3390/biology11071017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022]
Abstract
Atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) activate the guanylyl cyclase A receptor (GC-A), which synthesizes the second messenger cGMP in a wide variety of tissues and cells. C-type natriuretic peptide (CNP) activates the cGMP-producing guanylyl cyclase B receptor (GC-B) in chondrocytes, endothelial cells, and possibly smooth muscle cells, cardiomyocytes, and cardiac fibroblasts. The development of genetically modified mice has helped elucidate the physiological roles of natriuretic peptides via GC-A or GC-B. These include the hormonal effects of ANP/BNP in the vasculature, autocrine effects of ANP/BNP in cardiomyocytes, and paracrine effects of CNP in the vasculature and cardiomyocytes. Neprilysin (NEP) is a transmembrane neutral endopeptidase that degrades the three natriuretic peptides. Recently, mice overexpressing NEP, specifically in cardiomyocytes, revealed that local cardiac NEP plays a vital role in regulating natriuretic peptides in the heart tissue. Since NEP inhibition is a clinically accepted approach for heart failure treatment, the physiological roles of natriuretic peptides have regained attention. This article focuses on the physiological roles of natriuretic peptides elucidated in mice with GC-A or GC-B deletion, the significance of NEP in natriuretic peptide metabolism, and the long-term effects of angiotensin receptor-neprilysin inhibitor (ARNI) on cardiovascular diseases.
Collapse
Affiliation(s)
- Hitoshi Nakagawa
- Cardiovascular Medicine, Nara Medical University, Kashihara 634-8522, Nara, Japan;
| | - Yoshihiko Saito
- Nara Prefecture Seiwa Medical Center, Mimuro 636-0802, Nara, Japan
| |
Collapse
|
160
|
Huang J, Zhang W, Pan C, Zhu S, Mead RH, Li R, He B. Mobile Cardiac Acoustic Monitoring System to Evaluate Left Ventricular Systolic Function in Pacemaker Patients. J Clin Med 2022; 11:jcm11133862. [PMID: 35807146 PMCID: PMC9267668 DOI: 10.3390/jcm11133862] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/17/2022] [Accepted: 06/30/2022] [Indexed: 11/16/2022] Open
Abstract
The mobile cardiac acoustic monitoring system is a promising tool to enable detection and assist the diagnosis of left ventricular systolic dysfunction (LVSD). The objective of the study was to evaluate the diagnostic value of electromechanical activation time (EMAT), an important cardiac acoustic biomarker, in quantifying LVSD among left bundle branch pacing (LBBP) and right ventricular apical pacing (RVAP) patients using a mobile acoustic cardiography monitoring system. In this prospective single-center observational study, pacemaker-dependent patients were consecutively enrolled. EMAT, the time from the start of the pacing QRS wave to first heart sound (S1) peak; left ventricular systolic time (LVST), the time from S1 peak to S2 peak; and ECG were recorded simultaneously by the mobile cardiac acoustic monitoring system. LVEF was measured by echocardiography. A logistic regression model was applied to evaluate the association between EMAT and reduced EF (LVEF < 50%). A total of 105 pacemaker-dependent patients participated. The RVAP group (n = 58) displayed a significantly higher EMAT than the LBBP group (n = 47) (150.95 ± 19.46 vs. 108.23 ± 12.26 ms, p < 0.001). Pearson correlation analysis revealed a statistically significant negative correlation between EMAT and LVEF (p < 0.001). Survival analysis showed the sensitivity and specificity of detecting LVEF to be < 50% when EMAT ≥ 151 ms were 96.00% and 96.97% in the RVAP group. In LBBP patients, the sensitivity and specificity of using EMAT ≥ 110 ms as the cutoff value for the detection of LVEF < 50% were 75.00% and 100.00%. There was no significant difference in LVST with or without LVSD in the RVAP group (p = 0.823) and LBBP group (p = 0.086). Compared to LVST, EMAT was more helpful to identify LVSD in pacemaker-dependent patients. The cutoff point of EMAT for diagnosing LVEF < 50% differed regarding the pacing type. Therefore, the mobile cardiac acoustic monitoring system can be used to identify the progress of LVSD in pacemaker patients.
Collapse
Affiliation(s)
- Jingjuan Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China; (J.H.); (W.Z.); (C.P.); (S.Z.); (B.H.)
| | - Weiwei Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China; (J.H.); (W.Z.); (C.P.); (S.Z.); (B.H.)
| | - Changqing Pan
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China; (J.H.); (W.Z.); (C.P.); (S.Z.); (B.H.)
| | - Shiwei Zhu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China; (J.H.); (W.Z.); (C.P.); (S.Z.); (B.H.)
| | | | - Ruogu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China; (J.H.); (W.Z.); (C.P.); (S.Z.); (B.H.)
- Correspondence:
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China; (J.H.); (W.Z.); (C.P.); (S.Z.); (B.H.)
| |
Collapse
|
161
|
Cholecystokinin Octapeptide Promotes ANP Secretion through Activation of NOX4-PGC-1 α-PPAR α/PPAR γ Signaling in Isolated Beating Rat Atria. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5905374. [PMID: 35770043 PMCID: PMC9236793 DOI: 10.1155/2022/5905374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/15/2022] [Accepted: 05/19/2022] [Indexed: 11/18/2022]
Abstract
Atrial natriuretic peptide (ANP), a canonical cardiac hormone, is mainly secreted from atrial myocytes and is involved in the regulation of body fluid, blood pressure homeostasis, and antioxidants. Cholecystokinin (CCK) is also found in cardiomyocytes as a novel cardiac hormone and induces multiple cardiovascular regulations. However, the direct role of CCK on the atrial mechanical dynamics and ANP secretion is unclear. The current study was to investigate the effect of CCK octapeptide (CCK-8) on the regulation of atrial dynamics and ANP secretion. Experiments were performed in isolated perfused beating rat atria. ANP was measured using radioimmunoassay. The levels of hydrogen peroxide (H2O2) and arachidonic acid (AA) were determined using ELISA Kits. The levels of relative proteins and mRNA were detected by Western blot and RT-qPCR. The results showed that sulfated CCK-8 (CCK-8s) rather than desulfated CCK-8 increased the levels of phosphorylated cytosolic phospholipase A2 and AA release through activation of CCK receptors. This led to the upregulation of NADPH oxidase 4 (NOX4) expression levels and H2O2 production and played a negative inotropic effect on atrial mechanical dynamics via activation of ATP-sensitive potassium channels and large-conductance calcium-activated potassium channels. In addition, CCK-8s-induced NOX4 subsequently upregulated peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α) expression levels through activation of p38 mitogen-activated protein kinase as well as the serine/threonine kinase signaling, ultimately promoting the secretion of ANP via activation of PPARα and PPARγ. In the presence of the ANP receptor inhibitor, the CCK-8-induced increase of AA release, H2O2 production, and the upregulation of NOX4 and CAT expressions was augmented but the SOD expression induced by CCK-8s was repealed. These findings indicate that CCK-8s promotes the secretion of ANP through activation of NOX4-PGC-1α-PPARα/PPARγ signaling, in which ANP is involved in resistance for NOX4 expression and ROS production and regulation of SOD expression.
Collapse
|
162
|
Xi X, Li H, Chen S, Lv T, Ma T, Jiang R, Zhang P, Wong WH, Zhang X. Unfolding the genotype-to-phenotype black box of cardiovascular diseases through cross-scale modeling. iScience 2022; 25:104790. [PMID: 35992073 PMCID: PMC9386115 DOI: 10.1016/j.isci.2022.104790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/26/2022] [Accepted: 07/14/2022] [Indexed: 12/01/2022] Open
Abstract
Complex traits such as cardiovascular diseases (CVD) are the results of complicated processes jointly affected by genetic and environmental factors. Genome-wide association studies (GWAS) identified genetic variants associated with diseases but usually did not reveal the underlying mechanisms. There could be many intermediate steps at epigenetic, transcriptomic, and cellular scales inside the black box of genotype-phenotype associations. In this article, we present a machine-learning-based cross-scale framework GRPath to decipher putative causal paths (pcPaths) from genetic variants to disease phenotypes by integrating multiple omics data. Applying GRPath on CVD, we identified 646 and 549 pcPaths linking putative causal regions, variants, and gene expressions in specific cell types for two types of heart failure, respectively. The findings suggest new understandings of coronary heart disease. Our work promoted the modeling of tissue- and cell type-specific cross-scale regulation to uncover mechanisms behind disease-associated variants, and provided new findings on the molecular mechanisms of CVD. We defined one type of cross-scale genotype-to-phenotype regulation path We designed a framework GRPath to uncover putative regulation paths for diseases GRPath helped uncover molecular mechanisms for two major types of heart failure
Collapse
Affiliation(s)
- Xi Xi
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing 100084, China
| | - Haochen Li
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shengquan Chen
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing 100084, China
| | - Tingting Lv
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Tianxing Ma
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing 100084, China
| | - Rui Jiang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing 100084, China
| | - Ping Zhang
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Wing Hung Wong
- Departments of Statistics and Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Xuegong Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing 100084, China
- School of Medicine, Tsinghua University, Beijing 100084, China
- Corresponding author
| |
Collapse
|
163
|
Goetze JP, Bartels ED, Shalmi TW, Andraud-Dang L, Rehfeld JF. Biochemistry of the Endocrine Heart. BIOLOGY 2022; 11:biology11070971. [PMID: 36101352 PMCID: PMC9311610 DOI: 10.3390/biology11070971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/25/2022]
Abstract
Simple Summary Besides being a muscle and an electrochemically active organ, the heart is a true endocrine organ. As endocrine cells, cardiac myocytes possess all the needed chemical necessities for translation, post-translational modifications, and complex peptide proteolysis. In addition, intracellular granules in the cells contain not only peptides destined for secretion but also important granin molecules involved in maintaining a regulated secretory pathway. In this review, we highlight the biochemical phenotype of the endocrine heart, recapitulating that the cardiac myocytes are truly and fully capable endocrine cells. Abstract Production and release of natriuretic peptides and other vasoactive peptides are tightly regulated in mammalian physiology and involved in cardiovascular homeostasis. As endocrine cells, the cardiac myocytes seem to possess almost all known chemical necessities for translation, post-translational modifications, and complex peptide proteolysis. In several ways, intracellular granules in the cells contain not only peptides destined for secretion but also important granin molecules involved in maintaining a regulated secretory pathway. In this review, we will highlight the biochemical phenotype of the endocrine heart recapitulating that the cardiac myocytes are capable endocrine cells. Understanding the basal biochemistry of the endocrine heart in producing and secreting peptides to circulation could lead to new discoveries concerning known peptide products as well as hitherto unidentified cardiac peptide products. In perspective, studies on natriuretic peptides in the heart have shown that the post-translational phase of gene expression is not only relevant for human physiology but may prove implicated also in the development and, perhaps one day, cure of human cardiovascular disease.
Collapse
Affiliation(s)
- Jens P Goetze
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark
| | - Emil D Bartels
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark
| | - Theodor W Shalmi
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark
| | - Lilian Andraud-Dang
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark
| |
Collapse
|
164
|
Parcha V, Irvin MR, Lange LA, Armstrong ND, Pampana A, Meyer M, Judd SE, Arora G, Arora P. Corin Missense Variants, Blood Pressure, and Hypertension in 11 322 Black Individuals: Insights From REGARDS and the Jackson Heart Study. J Am Heart Assoc 2022; 11:e025582. [PMID: 35699180 PMCID: PMC9238660 DOI: 10.1161/jaha.121.025582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/27/2022] [Indexed: 11/18/2022]
Abstract
Background Corin enzyme contributes to the processing of inactive natriuretic peptides to bioactive hormones. In Black individuals, Corin gene variants (rs111253292 [Q568P] and rs75770792 [T555I]) have been previously reported to have a modest association with blood pressure (BP) and hypertension. Methods and Results We evaluated the association of Corin genotype with BP traits, prevalent hypertension, and incident hypertension among self-identified 11 322 Black Americans in the REGARDS (Reasons for Geographic and Racial Differences in Stroke) study and the JHS (Jackson Heart Study) using multivariable-adjusted regression modeling. Multivariable-adjusted genotype-stratified differences in NT-proBNP (N-terminal pro-B-type natriuretic peptide) and BNP (B-type natriuretic peptide) levels were assessed. Genotype-stratified NPPA and NPPB expression differences in healthy organ donor left atrial and left ventricular heart tissue (N=15) were also examined. The rs111253292 genotype was not associated with systolic BP (β±SE, 0.42±0.58; -1.24±0.82), diastolic BP (0.51±0.33; -0.41±0.46), mean arterial pressure (0.48±0.38; -0.68±0.51), and prevalent hypertension (odds ratio [OR], 0.93 [95% CI, 0.80-1.09]; OR, 0.79 [95% CI, 0.61-1.01]) in both REGARDS and JHS, respectively. The rs75770792 genotype was not associated with systolic BP (0.48±0.58; -1.26±0.81), diastolic BP (0.52±0.33; -0.33±0.45), mean arterial pressure (0.50±0.38; -0.63±0.50), and prevalent hypertension (OR, 1.02 [95% CI, 0.84-1.23]; OR, 0.87 [95% CI, 0.67-1.13]) in both cohorts, respectively. The Corin genotype was also not associated with incident hypertension (OR, 1.35 [95% CI, 0.94-1.93]; OR, 0.95 [95% CI, 0.64-1.39]) in the study cohorts. The NT-proBNP levels in REGARDS and BNP levels in JHS were similar between the Corin genotype groups. In heart tissue, the NPPA and NPPB expression was similar between the genotype groups. Conclusions Corin gene variants observed more commonly in Black individuals are not associated with differences in NP expression, circulating NP levels, and BP or hypertension as previously reported in candidate gene studies. Understanding the genetic determinants of complex cardiovascular traits in underrepresented populations requires further evaluation.
Collapse
Affiliation(s)
- Vibhu Parcha
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
| | | | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized MedicineDepartment of MedicineUniversity of Colorado School of MedicineAuroraCO
- Department of EpidemiologyUniversity of Colorado School of Public HealthAuroraCO
| | | | - Akhil Pampana
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
| | - Mariah Meyer
- Division of Biomedical Informatics and Personalized MedicineDepartment of MedicineUniversity of Colorado School of MedicineAuroraCO
| | - Suzanne E. Judd
- Department of BiostatisticsUniversity of Alabama at BirminghamBirminghamAL
| | - Garima Arora
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
| | - Pankaj Arora
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
- Section of CardiologyBirmingham Veterans Affairs Medical CenterBirminghamAL
| |
Collapse
|
165
|
Castro-Portillo E, López-Izquierdo R, Sanz-García A, Ortega GJ, Delgado-Benito JF, Castro Villamor MA, Sánchez-Soberón I, Del Pozo Vegas C, Martín-Rodríguez F. Role of prehospital point-of-care N-terminal pro-brain natriuretic peptide in acute life-threatening cardiovascular disease. Int J Cardiol 2022; 364:126-132. [PMID: 35716940 DOI: 10.1016/j.ijcard.2022.06.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/18/2022] [Accepted: 06/10/2022] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The evidence about the use of natriuretic peptides (NP) to predict mortality in the pre-hospital setting is limited. The main objective of this study is to assess the ability of point-of-care testing (POCT) N-terminal portion of B-type natriuretic peptide (NT-proBNP) to predict 2-day in-hospital mortality of acute cardiovascular diseases (ACVD). METHODS We conducted a multicentric, prospective, observational study in adults with ACVD transferred by ambulance to emergency departments (ED). The primary outcome was 2-day in-hospital mortality. The discrimination capacity of the NT-proBNP was performed through a prediction model trained using a derivation cohort and evaluated by the area under the curve (AUC) of the receiver operating characteristic on a validation cohort. RESULTS A total of 1006 patients were recruited. The median age was 75 (IQR 63-84) years and 421 (41.85%) were females. The 2-day in-hospital mortality was 5.8% (58 cases). The predictive validity of NT-proBNP, for 2-day mortality reached the following AUC: 0.823 (95%CI: 0.758-0.889, p < 0.001), and the optimal specificity and sensitivity were 73.1 and 82.7. Predictive power of NT-proBNP obtained an AUC 0.549 (95%CI: 0.432-0.865, p 0.215) for acute heart failure, AUC 0.893 (95%CI: 0.617-0.97, p < 0.001) for ischemic heart disease, AUC 0.714 (95%CI: 0.55-0.87, p = 0.0069) for arrhythmia and AUC 0.927 (95%CI: 0.877-0.978, p < 0,001) for syncope. CONCLUSION POCT NT-proBNP has proven to be a strong predictor of early mortality in ACVD, showing an excellent predictive capacity in cases of syncope. However, this biomarker does not appear to be useful for predicting outcome in patients with acute heart failure.
Collapse
Affiliation(s)
| | - Raúl López-Izquierdo
- Emergency Department, Hospital Universitario Rio Hortega, Valladolid, Spain; Faculty of Medicine, Universidad de Valladolid, Valladolid, Spain; Prehospital Early Warning Scoring-System Investigation Group, Valladolid, Spain
| | - Ancor Sanz-García
- Prehospital Early Warning Scoring-System Investigation Group, Valladolid, Spain; Data Analysis Unit, Health Research Institute, Hospital de la Princesa, Madrid (IIS-IP), Spain.
| | - Guillermo J Ortega
- Data Analysis Unit, Health Research Institute, Hospital de la Princesa, Madrid (IIS-IP), Spain; CONICET, Argentina; Science and Technology department, National University of Quilmes, Argentina
| | - Juan F Delgado-Benito
- Advanced Life Support, Emergency Medical Services (SACYL), Valladolid, Spain; Prehospital Early Warning Scoring-System Investigation Group, Valladolid, Spain
| | - Miguel A Castro Villamor
- Faculty of Medicine, Universidad de Valladolid, Valladolid, Spain; Prehospital Early Warning Scoring-System Investigation Group, Valladolid, Spain
| | | | - Carlos Del Pozo Vegas
- Faculty of Medicine, Universidad de Valladolid, Valladolid, Spain; Prehospital Early Warning Scoring-System Investigation Group, Valladolid, Spain; Emergency Department, Hospital Clínico Universitario, Valladolid, Spain
| | - Francisco Martín-Rodríguez
- Faculty of Medicine, Universidad de Valladolid, Valladolid, Spain; Advanced Life Support, Emergency Medical Services (SACYL), Valladolid, Spain; Prehospital Early Warning Scoring-System Investigation Group, Valladolid, Spain
| |
Collapse
|
166
|
Harbaum L, Rhodes CJ, Wharton J, Lawrie A, Karnes JH, Desai AA, Nichols WC, Humbert M, Montani D, Girerd B, Sitbon O, Boehm M, Novoyatleva T, Schermuly RT, Ghofrani HA, Toshner M, Kiely DG, Howard LS, Swietlik EM, Gräf S, Pietzner M, Morrell NW, Wilkins MR. Mining the Plasma Proteome for Insights into the Molecular Pathology of Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2022; 205:1449-1460. [PMID: 35394406 PMCID: PMC9875902 DOI: 10.1164/rccm.202109-2106oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 04/07/2022] [Indexed: 01/29/2023] Open
Abstract
Rationale: Pulmonary arterial hypertension (PAH) is characterized by structural remodeling of pulmonary arteries and arterioles. Underlying biological processes are likely reflected in a perturbation of circulating proteins. Objectives: To quantify and analyze the plasma proteome of patients with PAH using inherited genetic variation to inform on underlying molecular drivers. Methods: An aptamer-based assay was used to measure plasma proteins in 357 patients with idiopathic or heritable PAH, 103 healthy volunteers, and 23 relatives of patients with PAH. In discovery and replication subgroups, the plasma proteomes of PAH and healthy individuals were compared, and the relationship to transplantation-free survival in PAH was determined. To examine causal relationships to PAH, protein quantitative trait loci (pQTL) that influenced protein levels in the patient population were used as instruments for Mendelian randomization (MR) analysis. Measurements and Main Results: From 4,152 annotated plasma proteins, levels of 208 differed between patients with PAH and healthy subjects, and 49 predicted long-term survival. MR based on cis-pQTL located in proximity to the encoding gene for proteins that were prognostic and distinguished PAH from health estimated an adverse effect for higher levels of netrin-4 (odds ratio [OR], 1.55; 95% confidence interval [CI], 1.16-2.08) and a protective effect for higher levels of thrombospondin-2 (OR, 0.83; 95% CI, 0.74-0.94) on PAH. Both proteins tracked the development of PAH in previously healthy relatives and changes in thrombospondin-2 associated with pulmonary arterial pressure at disease onset. Conclusions: Integrated analysis of the plasma proteome and genome implicates two secreted matrix-binding proteins, netrin-4 and thrombospondin-2, in the pathobiology of PAH.
Collapse
Affiliation(s)
- Lars Harbaum
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Christopher J. Rhodes
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - John Wharton
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, United Kingdom
| | - Jason H. Karnes
- Department of Pharmacy Practice and Science, University of Arizona College of Pharmacy, Tucson, Arizona
| | - Ankit A. Desai
- Department of Medical and Molecular Genetics, and Krannert Institute of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - William C. Nichols
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marc Humbert
- Université Paris–Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - David Montani
- Université Paris–Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Barbara Girerd
- Université Paris–Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Olivier Sitbon
- Université Paris–Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Mario Boehm
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Tatyana Novoyatleva
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Ralph T. Schermuly
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | | | - Mark Toshner
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - David G. Kiely
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, United Kingdom
| | - Luke S. Howard
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Emilia M. Swietlik
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Stefan Gräf
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- National Institute for Health Research BioResource for Translational Research, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Maik Pietzner
- Computational Medicine, Berlin Institute of Health (BIH) at Charité–Universitätsmedizin Berlin, Germany; and
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Martin R. Wilkins
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
167
|
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: 2.5] [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
|
168
|
González A, Richards AM, de Boer RA, Thum T, Arfsten H, Hülsmann M, Falcao-Pires I, Díez J, Foo RSY, Chan MY, Aimo A, Anene-Nzelu CG, Abdelhamid M, Adamopoulos S, Anker SD, Belenkov Y, Ben Gal T, Cohen-Solal A, Böhm M, Chioncel O, Delgado V, Emdin M, Jankowska EA, Gustafsson F, Hill L, Jaarsma T, Januzzi JL, Jhund PS, Lopatin Y, Lund LH, Metra M, Milicic D, Moura B, Mueller C, Mullens W, Núñez J, Piepoli MF, Rakisheva A, Ristić AD, Rossignol P, Savarese G, Tocchetti CG, Van Linthout S, Volterrani M, Seferovic P, Rosano G, Coats AJS, Bayés-Genís A. Cardiac remodelling - Part 1: From cells and tissues to circulating biomarkers. A review from the Study Group on Biomarkers of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2022; 24:927-943. [PMID: 35334137 DOI: 10.1002/ejhf.2493] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
Cardiac remodelling refers to changes in left ventricular structure and function over time, with a progressive deterioration that may lead to heart failure (HF) development (adverse remodelling) or vice versa a recovery (reverse remodelling) in response to HF treatment. Adverse remodelling predicts a worse outcome, whilst reverse remodelling predicts a better prognosis. The geometry, systolic and diastolic function and electric activity of the left ventricle are affected, as well as the left atrium and on the long term even right heart chambers. At a cellular and molecular level, remodelling involves all components of cardiac tissue: cardiomyocytes, fibroblasts, endothelial cells and leucocytes. The molecular, cellular and histological signatures of remodelling may differ according to the cause and severity of cardiac damage, and clearly to the global trend toward worsening or recovery. These processes cannot be routinely evaluated through endomyocardial biopsies, but may be reflected by circulating levels of several biomarkers. Different classes of biomarkers (e.g. proteins, non-coding RNAs, metabolites and/or epigenetic modifications) and several biomarkers of each class might inform on some aspects on HF development, progression and long-term outcomes, but most have failed to enter clinical practice. This may be due to the biological complexity of remodelling, so that no single biomarker could provide great insight on remodelling when assessed alone. Another possible reason is a still incomplete understanding of the role of biomarkers in the pathophysiology of cardiac remodelling. Such role will be investigated in the first part of this review paper on biomarkers of cardiac remodelling.
Collapse
Affiliation(s)
- Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, and IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - A Mark Richards
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
- Christchurch Heart Institute, University of Otago, Dunedin, New Zealand
| | - Rudolf A de Boer
- University Medical Center Groningen, University of Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) and Rebirth Center for Translational Regenerative Therapies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Henrike Arfsten
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Martin Hülsmann
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Inês Falcao-Pires
- Department od Surgery and Physiology, Cardiovascular Research and Development Center, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, and IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Departments of Cardiology and Cardiac Surgery, and Nephrology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Roger S Y Foo
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
| | - Mark Y Chan
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
| | - Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Chukwuemeka G Anene-Nzelu
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
- Montreal Heart Institute, Montreal, Canada
| | | | - Stamatis Adamopoulos
- 2nd Department of Cardiovascular Medicine, Onassis Cardiac Surgery Center, Athens, Greece
| | - Stefan D Anker
- Department of Cardiology (CVK), and Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | | | - Tuvia Ben Gal
- Cardiology Department, Rabin Medical Center, Beilinson, Israel
| | | | - Michael Böhm
- Universitätsklinikum des Saarlandes, Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Saarland University, Homburg/Saar, Germany
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases 'Prof. C.C. Iliescu' Bucharest, University of Medicine Carol Davila, Bucharest, Romania
| | - Victoria Delgado
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Ewa A Jankowska
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Finn Gustafsson
- Rigshospitalet-Copenhagen University Hospital, Heart Centre, Department of Cardiology, Copenhagen, Denmark
| | | | | | - James L Januzzi
- Massachusetts General Hospital and Baim Institute for Clinical Research, Boston, MA, USA
| | - Pardeep S Jhund
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland
| | - Yuri Lopatin
- Volgograd State Medical University, Volgograd, Russia
| | - Lars H Lund
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Marco Metra
- Cardiology, ASST Spedali Civili; Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Davor Milicic
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Brenda Moura
- Faculty of Medicine, University of Porto, Porto, Portugal
- Cardiology Department, Porto Armed Forces Hospital, Portugal
| | | | | | - Julio Núñez
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Hospital Clínico Universitario de Valencia, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - Massimo F Piepoli
- Cardiology Division, Castelsangiovanni Hospital, Castelsangiovanni, Italy
| | - Amina Rakisheva
- Scientific Research Institute of Cardiology and Internal Medicine, Almaty, Kazakhstan
| | - Arsen D Ristić
- Department of Cardiology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Patrick Rossignol
- Université de Lorraine, Centre d'Investigations Cliniques- Plurithématique 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Gianluigi Savarese
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Carlo G Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Sophie Van Linthout
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | | | - Petar Seferovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Giuseppe Rosano
- St. George's Hospitals, NHS Trust, University of London, London, UK
| | | | - Antoni Bayés-Genís
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
169
|
Aimo A, Vergaro G, González A, Barison A, Lupón J, Delgado V, Richards AM, de Boer RA, Thum T, Arfsten H, Hülsmann M, Falcao-Pires I, Díez J, Foo RSY, Chan MYY, Anene-Nzelu CG, Abdelhamid M, Adamopoulos S, Anker SD, Belenkov Y, Ben Gal T, Cohen-Solal A, Böhm M, Chioncel O, Jankowska EA, Gustafsson F, Hill L, Jaarsma T, Januzzi JL, Jhund P, Lopatin Y, Lund LH, Metra M, Milicic D, Moura B, Mueller C, Mullens W, Núñez J, Piepoli MF, Rakisheva A, Ristić AD, Rossignol P, Savarese G, Tocchetti CG, van Linthout S, Volterrani M, Seferovic P, Rosano G, Coats AJS, Emdin M, Bayes-Genis A. Cardiac remodelling - Part 2: Clinical, imaging and laboratory findings. A review from the Study Group on Biomarkers of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2022; 24:944-958. [PMID: 35488811 DOI: 10.1002/ejhf.2522] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 11/06/2022] Open
Abstract
In patients with heart failure, the beneficial effects of drug and device therapies counteract to some extent ongoing cardiac damage. According to the net balance between these two factors, cardiac geometry and function may improve (reverse remodelling, RR) and even completely normalize (remission), or vice versa progressively deteriorate (adverse remodelling, AR). RR or remission predict a better prognosis, while AR has been associated with worsening clinical status and outcomes. The remodelling process ultimately involves all cardiac chambers, but has been traditionally evaluated in terms of left ventricular volumes and ejection fraction. This is the second part of a review paper by the Study Group on Biomarkers of the Heart Failure Association of the European Society of Cardiology dedicated to ventricular remodelling. This document examines the proposed criteria to diagnose RR and AR, their prevalence and prognostic value, and the variables predicting remodelling in patients managed according to current guidelines. Much attention will be devoted to RR in patients with heart failure with reduced ejection fraction because most studies on cardiac remodelling focused on this setting.
Collapse
Affiliation(s)
- Alberto Aimo
- Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Giuseppe Vergaro
- Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Arantxa González
- CIMA Universidad de Navarra, and IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Andrea Barison
- Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Josep Lupón
- Institut del Cor, Hospital Universitari Germans Trias i Pujol Badalona, Barcelona, Spain
| | - Victoria Delgado
- Institut del Cor, Hospital Universitari Germans Trias i Pujol Badalona, Barcelona, Spain
| | | | - Rudolf A de Boer
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Thomas Thum
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Henrike Arfsten
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Martin Hülsmann
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | | | - Javier Díez
- Center for Applied Medical Research, Pamplona, Spain
| | - Roger S Y Foo
- Department of Medicine, Yong Loo-Lin School of Medicine, National University Hospital, Singapore, Singapore
| | - Mark Yan Yee Chan
- Department of Medicine, Yong Loo-Lin School of Medicine, National University Hospital, Singapore, Singapore
| | - Chukwuemeka G Anene-Nzelu
- Department of Medicine, Yong Loo-Lin School of Medicine, National University Hospital, Singapore, Singapore
| | | | - Stamatis Adamopoulos
- 2nd Department of Cardiovascular Medicine, Onassis Cardiac Surgery Center, Athens, Greece
| | - Stefan D Anker
- Department of Cardiology (CVK), and Berlin Institute of Health Center for Regenerative Therapy (BCRT), German Center for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | | | - Tuvia Ben Gal
- Cardiology Department, Rabin Medical Center, Beilinson, Israel
| | | | - Michael Böhm
- University of the Saarland, Homburg/Saar, Germany
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases 'Prof. C.C. Iliescu' Bucharest, University of Medicine Carol Davila, Bucharest, Romania
| | - Ewa A Jankowska
- Institute of Heart Disases, Wroclaw Medical University, Wroclaw, Poland
| | - Finn Gustafsson
- Heart Centre, Department of Cardiology, Rigshospitalet - Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - James L Januzzi
- Massachusetts General Hospital and Baim Institute for Clinical Research, Boston, MA, USA
| | | | - Yuri Lopatin
- Volgograd State Medical University, Volgograd, Russia
| | - Lars H Lund
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Marco Metra
- Cardiology, ASST Spedali Civili; Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Davor Milicic
- University of Zagreb School of Medicine, Zagreb, Croatia
| | - Brenda Moura
- Faculty of Medicine, University of Porto, Porto, Portugal
- Cardiology Department, Porto Armed Forces Hospital, Porto, Portugal
| | | | | | - Julio Núñez
- Hospital Clínico Universitario de Valencia, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - Massimo F Piepoli
- Cardiology Division, Castelsangiovanni Hospital, Castelsangiovanni, Italy
| | - Amina Rakisheva
- Scientific Research Institute of Cardiology and Internal Medicine, Almaty, Kazakhstan
| | - Arsen D Ristić
- Department of Cardiology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Patrick Rossignol
- Université de Lorraine, Centre d'Investigations Cliniques-Plurithématique 1433 and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Gianluigi Savarese
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Carlo G Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Sophie van Linthout
- Berlin Institute of Health (BIH) at Charité - Universitätmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | | | - Petar Seferovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Giuseppe Rosano
- St. George's Hospitals, NHS Trust, University of London, London, UK
| | | | - Michele Emdin
- Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Antoni Bayes-Genis
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Institut del Cor, Hospital Universitari Germans Trias i Pujol Badalona, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
170
|
Chen Q, Chen Y, Qin F, Du H, Gan C, Zhou B, Wang N, Xiao M, Ou Z, Zhao W, Cui B, Liu Z, Yin Y. Effect of Sacubitril-Valsartan on Restoration and Maintenance of Sinus Rhythm in Patients With Persistent Atrial Fibrillation. Front Cardiovasc Med 2022; 9:870203. [PMID: 35707128 PMCID: PMC9189349 DOI: 10.3389/fcvm.2022.870203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/02/2022] [Indexed: 11/22/2022] Open
Abstract
Background Existing studies have shown that sacubitril-valsartan ameliorated atrial remodeling in atrial fibrillation (AF) and favored maintenance of sinus rhythm in patients with AF and heart failure. However, the effect of sacubitril-valsartan in patients with persistent AF is yet unknown. We aimed to evaluate the effect of sacubitril-valsartan on restoration and maintenance of sinus rhythm in patients with persistent AF who underwent electrical cardioversion (ECV). Method Consecutive patients with persistent AF who underwent ECV between 1 January 2016 and 30 September 2020 were investigated in this retrospective cohort study. All eligible patients were categorized into sacubitril-valsartan users and sacubitril-valsartan non-users based on whether they received treatment with sacubitril-valsartan or not. The endpoint was ineffictive ECV, defined as the composite of failure to terminate AF or any recurrence of AF during 30 days follow-up. Results A total of 76 patients were enrolled in this study, including 28 sacubitril-valsartan users and 48 non-users. Within a follow-up of 30 days after ECV, the endpoint had occurred in 7 (25%) of 28 sacubitril-valsartan users and 25 (52%) of 48 non-users. Significantly lower rate of ineffictive ECV in sacubitril-valsartan users compared with non-users was shown in Kaplan-Meier survival curves (P = 0.02; Log-rank test). Multivariate Cox regression analysis indicated that sacubitril-valsartan use (hazard ratio [HR], 0.35; 95% confidence interval [CI], 0.14–0.91), amiodarone use (HR, 0.32; 95% CI, 0.13–0.78), left atrial diameter ≤ 39 mm (HR, 0.21; 95% CI, 0.06–0.71) were independently associated with a decreased rate of ineffective electrical cardioversion. Conclusion Use of sacubitril-valsartan is associated with a significantly decreased risk of ineffective ECV compared with non-users in patients with persistent AF.
Collapse
|
171
|
Senekovič Kojc T, Marčun Varda N. Novel Biomarkers of Heart Failure in Pediatrics. CHILDREN 2022; 9:children9050740. [PMID: 35626917 PMCID: PMC9139970 DOI: 10.3390/children9050740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023]
Abstract
Novel biomarkers of heart failure are the subject of numerous studies. Biomarkers of heart failure can be determined in the blood and in the urine. Seven groups of biomarkers of heart failure based on pathophysiological mechanisms are presented in this review, namely biomarkers of myocardial stretch, myocyte injury, myocardial remodeling, biomarkers of inflammation, renal dysfunction, neurohumoral activation, and oxidative stress. Studies of biomarkers in the pediatric population are scarce, therefore, further investigation is needed for reliable prognostic and therapeutic implications. The future of biomarker use is in multimarker panels that include a combination of biomarkers with different pathophysiological mechanisms in order to improve their diagnostic and prognostic predictive value.
Collapse
Affiliation(s)
- Teja Senekovič Kojc
- Department of Perinatology, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia
- Correspondence:
| | - Nataša Marčun Varda
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia;
- Medical Faculty, University of Maribor, Taborska 8, 2000 Maribor, Slovenia
| |
Collapse
|
172
|
Ravi R, Fernandes Silva L, Vangipurapu J, Maria M, Raivo J, Helisalmi S, Laakso M. Metabolite Signature in the Carriers of Pathogenic Genetic Variants for Cardiomyopathy: A Population-Based METSIM Study. Metabolites 2022; 12:437. [PMID: 35629941 PMCID: PMC9143630 DOI: 10.3390/metabo12050437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/04/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Hypertrophic (HCM) and dilated (DCM) cardiomyopathies are among the leading causes of sudden cardiac death. We identified 38 pathogenic or likely pathogenic variant carriers for HCM in three sarcomere genes (MYH7, MYBPC3, TPMI) among 9.928 participants of the METSIM Study having whole exome sequencing data available. Eight of them had a clinical diagnosis of HCM. We also identified 20 pathogenic or likely pathogenic variant carriers for DCM in the TTN gene, and six of them had a clinical diagnosis of DCM. The aim of our study was to investigate the metabolite signature in the carriers of the pathogenic or likely pathogenic genetic variants for HCM and DCM, compared to age- and body-mass-index-matched controls. Our novel findings were that the carriers of pathogenic or likely pathogenic variants for HCM had significantly increased concentrations of bradykinin (des-arg 9), vanillactate, and dimethylglycine and decreased concentrations of polysaturated fatty acids (PUFAs) and lysophosphatidylcholines compared with the controls without HCM. Additionally, our novel findings were that the carriers of pathogenic or likely pathogenic variants for DCM had significantly decreased concentrations of 1,5-anhydrogluticol, histidine betaine, N-acetyltryptophan, and methylsuccinate and increased concentrations of trans-4-hydroxyproline compared to the controls without DCM. Our population-based study shows that the metabolite signature of the genetic variants for HCM and DCM includes several novel metabolic pathways not previously described.
Collapse
Affiliation(s)
- Rowmika Ravi
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (R.R.); (L.F.S.); (J.V.); (J.R.); (S.H.)
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (R.R.); (L.F.S.); (J.V.); (J.R.); (S.H.)
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (R.R.); (L.F.S.); (J.V.); (J.R.); (S.H.)
| | - Maleeha Maria
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Joose Raivo
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (R.R.); (L.F.S.); (J.V.); (J.R.); (S.H.)
| | - Seppo Helisalmi
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (R.R.); (L.F.S.); (J.V.); (J.R.); (S.H.)
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (R.R.); (L.F.S.); (J.V.); (J.R.); (S.H.)
- Department of Medicine, Kuopio University Hospital, 70210 Kuopio, Finland
| |
Collapse
|
173
|
Johansson M, Tangruksa B, Heydarkhan-Hagvall S, Jeppsson A, Sartipy P, Synnergren J. Data Mining Identifies CCN2 and THBS1 as Biomarker Candidates for Cardiac Hypertrophy. Life (Basel) 2022; 12:life12050726. [PMID: 35629393 PMCID: PMC9147176 DOI: 10.3390/life12050726] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 12/02/2022] Open
Abstract
Cardiac hypertrophy is a condition that may contribute to the development of heart failure. In this study, we compare the gene-expression patterns of our in vitro stem-cell-based cardiac hypertrophy model with the gene expression of biopsies collected from hypertrophic human hearts. Twenty-five differentially expressed genes (DEGs) from both groups were identified and the expression of selected corresponding secreted proteins were validated using ELISA and Western blot. Several biomarkers, including CCN2, THBS1, NPPA, and NPPB, were identified, which showed significant overexpressions in the hypertrophic samples in both the cardiac biopsies and in the endothelin-1-treated cells, both at gene and protein levels. The protein-interaction network analysis revealed CCN2 as a central node among the 25 overlapping DEGs, suggesting that this gene might play an important role in the development of cardiac hypertrophy. GO-enrichment analysis of the 25 DEGs revealed many biological processes associated with cardiac function and the development of cardiac hypertrophy. In conclusion, we identified important similarities between ET-1-stimulated human-stem-cell-derived cardiomyocytes and human hypertrophic cardiac tissue. Novel putative cardiac hypertrophy biomarkers were identified and validated on the protein level, lending support for further investigations to assess their potential for future clinical applications.
Collapse
Affiliation(s)
- Markus Johansson
- Systems Biology Research Center, School of Bioscience, University of Skövde, SE-541 28 Skövde, Sweden; (S.H.-H.); (P.S.); (J.S.)
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, SE-413 45 Gothenburg, Sweden;
- Correspondence: (M.J.); (B.T.)
| | - Benyapa Tangruksa
- Systems Biology Research Center, School of Bioscience, University of Skövde, SE-541 28 Skövde, Sweden; (S.H.-H.); (P.S.); (J.S.)
- Correspondence: (M.J.); (B.T.)
| | - Sepideh Heydarkhan-Hagvall
- Systems Biology Research Center, School of Bioscience, University of Skövde, SE-541 28 Skövde, Sweden; (S.H.-H.); (P.S.); (J.S.)
- Bioscience, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, SE-413 83 Gothenburg, Sweden
| | - Anders Jeppsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, SE-413 45 Gothenburg, Sweden;
- Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden
| | - Peter Sartipy
- Systems Biology Research Center, School of Bioscience, University of Skövde, SE-541 28 Skövde, Sweden; (S.H.-H.); (P.S.); (J.S.)
| | - Jane Synnergren
- Systems Biology Research Center, School of Bioscience, University of Skövde, SE-541 28 Skövde, Sweden; (S.H.-H.); (P.S.); (J.S.)
| |
Collapse
|
174
|
Shen X, Dong N, Xu Y, Han L, Yang R, Liao J, Zhang X, Xie T, Wang Y, Chen C, Liu M, Jiang Y, Yu L, Fang Q. Analyzing Corin–BNP–NEP Protein Pathway Revealing Differential Mechanisms in AF-Related Ischemic Stroke and No AF-Related Ischemic Stroke. Front Aging Neurosci 2022; 14:863489. [PMID: 35615592 PMCID: PMC9125077 DOI: 10.3389/fnagi.2022.863489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background The incidence of atrial fibrillation (AF)-related stroke increases with aging. Natriuretic peptides (NPs) family, including Corin-B type natriuretic peptide (BNP)-neprilysin (NEP) protein levels increased with age and are risk markers of cardiovascular and cerebrovascular diseases, such as AF and cardioembolic stroke. Aging is also linked to epigenetics, specifically DNA methylation. However, only a few studies have investigated the effect of DNA methylation on the NP system. Thus, the present study aimed to investigate whether the Corin-BNP-NEP protein pathway is involved in the pathogenesis of AF-stroke and CpG methylation in the promoter region of the Corin protein gene has an effect on AF-related ischemic stroke. Methods A total of 82 patients hospitalized with acute ischemic strokes were enrolled in this study. The differences in clinical information were compared between the AF-stroke (n = 37) and no AF-stroke groups (n = 45). Plasma-soluble Corin and NEP were detected using an ELISA kit. CpG methylation in the promoter region of the gene was assessed by a next-generation sequencing-based bisulfite sequencing polymerase chain reaction (BSP). Results (1) Patients in AF-stroke were older, had higher initial NIHSS score, 90-day mRs, higher D2-dimer, INR, and APTT, and low TG, TC, and HbA1c (all p < 0.05). (2) Serum levels of Corin and BNP in the AF-stroke group were significantly higher than that in the no AF-stroke group (p < 0.05). No significant difference was detected in the serum levels of NEP between the two groups. (3) The levels of CpG methylation in the promoter region of the Corin protein gene in the AF-stroke group was significantly lower than that in the no AF-stroke group (p < 0.05). The CpG sites with maximal methylation differences between the two groups were CORIN:678, CORIN:682, CORIN:694, and CORIN:700. Conclusion The current findings raise the possibility that the Corin–BNP–NEP protein pathway may be involved in the pathogenesis of AF-related ischemic stroke. Deficient CpG methylation in the promoter region of the Corin protein gene is associated with AF-related ischemic stroke.
Collapse
Affiliation(s)
- Xiaozhu Shen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Geriatrics, Lianyungang Second People’s Hospital, Lianyungang, China
| | - Nan Dong
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurology, Suzhou Industrial Park Xinghai Hospital, Suzhou, China
| | - Yiwen Xu
- Department of General Medicine, Lianyungang Hospital, Affiliated to Jiangsu University (Lianyungang Second People’s Hospital), Lianyungang, China
- *Correspondence: Yiwen Xu,
| | - Lin Han
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Rui Yang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Juan Liao
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xianxian Zhang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tao Xie
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yugang Wang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chen Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mengqian Liu
- Department of General Medicine, Lianyungang Hospital, Affiliated to Jiangsu University (Lianyungang Second People’s Hospital), Lianyungang, China
| | - Yi Jiang
- Bengbu Medical College, Bengbu, China
| | - Liqiang Yu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Qi Fang,
| |
Collapse
|
175
|
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: 2.0] [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
|
176
|
Frantz S, Hundertmark MJ, Schulz-Menger J, Bengel FM, Bauersachs J. Left ventricular remodelling post-myocardial infarction: pathophysiology, imaging, and novel therapies. Eur Heart J 2022; 43:2549-2561. [PMID: 35511857 PMCID: PMC9336586 DOI: 10.1093/eurheartj/ehac223] [Citation(s) in RCA: 181] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/08/2022] [Accepted: 04/13/2022] [Indexed: 12/11/2022] Open
Abstract
Most patients survive acute myocardial infarction (MI). Yet this encouraging development has certain drawbacks: heart failure (HF) prevalence is increasing and patients affected tend to have more comorbidities worsening economic strain on healthcare systems and impeding effective medical management. The heart’s pathological changes in structure and/or function, termed myocardial remodelling, significantly impact on patient outcomes. Risk factors like diabetes, chronic obstructive pulmonary disease, female sex, and others distinctly shape disease progression on the ‘road to HF’. Despite the availability of HF drugs that interact with general pathways involved in myocardial remodelling, targeted drugs remain absent, and patient risk stratification is poor. Hence, in this review, we highlight the pathophysiological basis, current diagnostic methods and available treatments for cardiac remodelling following MI. We further aim to provide a roadmap for developing improved risk stratification and novel medical and interventional therapies.
Collapse
Affiliation(s)
- Stefan Frantz
- Department of Internal Medicine I, Universitätsklinikum Würzburg, University Hospital Würzburg, University of Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Moritz Jens Hundertmark
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Jeanette Schulz-Menger
- Department of Cardiology and Nephrology, Experimental and Clinical Research Center, a Joint Cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine and HELIOS Hospital Berlin Buch, Berlin, Germany
| | | | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
177
|
Rhodes CJ, Wharton J, Swietlik EM, Harbaum L, Girerd B, Coghlan JG, Lordan J, Church C, Pepke-Zaba J, Toshner M, Wort SJ, Kiely DG, Condliffe R, Lawrie A, Gräf S, Montani D, Boucly A, Sitbon O, Humbert M, Howard LS, Morrell NW, Wilkins MR. Using the Plasma Proteome for Risk Stratifying Patients with Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2022; 205:1102-1111. [PMID: 35081018 PMCID: PMC9851485 DOI: 10.1164/rccm.202105-1118oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Rationale: NT-proBNP (N-terminal pro-brain natriuretic peptide), a biomarker of cardiac origin, is used to risk stratify patients with pulmonary arterial hypertension (PAH). Its limitations include poor sensitivity to early vascular pathology. Other biomarkers of vascular or systemic origin may also be useful in the management of PAH. Objectives: Identify prognostic proteins in PAH that complement NT-proBNP and clinical risk scores. Methods: An aptamer-based assay (SomaScan version 4) targeting 4,152 proteins was used to measure plasma proteins in patients with idiopathic, heritable, or drug-induced PAH from the UK National Cohort of PAH (n = 357) and the French EFORT (Evaluation of Prognostic Factors and Therapeutic Targets in PAH) study (n = 79). Prognostic proteins were identified in discovery-replication analyses of UK samples. Proteins independent of 6-minute-walk distance and NT-proBNP entered least absolute shrinkage and selection operator modeling, and the best combination in a single score was evaluated against clinical targets in EFORT. Measurements and Main Results: Thirty-one proteins robustly informed prognosis independent of NT-proBNP and 6-minute-walk distance in the UK cohort. A weighted combination score of six proteins was validated at baseline (5-yr mortality; area under the curve [AUC], 0.73; 95% confidence interval [CI], 0.63-0.85) and follow-up in EFORT (AUC, 0.84; 95% CI, 0.75-0.94; P = 9.96 × 10-6). The protein score risk stratified patients independent of established clinical targets and risk equations. The addition of the six-protein model score to NT-proBNP improved prediction of 5-year outcomes from AUC 0.762 (0.702-0.821) to 0.818 (0.767-0.869) by receiver operating characteristic analysis (P = 0.00426 for difference in AUC) in the UK replication and French samples combined. Conclusions: The plasma proteome informs prognosis beyond established factors in PAH and may provide a more sensitive measure of therapeutic response.
Collapse
Affiliation(s)
- Christopher J Rhodes
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Emilia M Swietlik
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Lars Harbaum
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Barbara Girerd
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - J Gerry Coghlan
- Department of Cardiology, Royal Free Campus, University College London, London, United Kingdom
| | - James Lordan
- University of Newcastle upon Tyne, Newcastle upon Tyne, United Kingdom
| | - Colin Church
- University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Joanna Pepke-Zaba
- Pulmonary Vascular Disease Unit, Royal Papworth Hospital, Cambridge, United Kingdom
| | - Mark Toshner
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J Wort
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - David G Kiely
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom.,Sheffield Pulmonary Vascular Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom; and
| | - Robin Condliffe
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom.,Sheffield Pulmonary Vascular Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom; and
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,BioResource for Translational Research, National Institute for Health Research Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - David Montani
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Athénaïs Boucly
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Olivier Sitbon
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Marc Humbert
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Luke S Howard
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | |
Collapse
|
178
|
Salido-Medina AB, Gil A, Expósito V, Martínez F, Redondo JM, Hurlé MA, Nistal JF, García R. BMP7-based peptide agonists of BMPR1A protect the left ventricle against pathological remodeling induced by pressure overload. Biomed Pharmacother 2022; 149:112910. [PMID: 35616049 DOI: 10.1016/j.biopha.2022.112910] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/26/2022] Open
Abstract
Aortic stenosis (AS) exposes the left ventricle (LV) to pressure overload leading to detrimental LV remodeling and heart failure. In animal models of cardiac injury or hemodynamic stress, bone morphogenetic protein-7 (BMP7) protects LV against remodeling by counteracting TGF-β effects. BMP receptor 1A (BMPR1A) might mediate BMP7 antifibrotic effects. Herein we evaluated BMP7-based peptides, THR123 and THR184, agonists of BMPR1A, as cardioprotective drugs in a pressure overload model. We studied patients with AS, mice subjected to four-week transverse aortic constriction (TAC) and TAC release (de-TAC). The LV of AS patients and TAC mice featured Bmpr1a downregulation. Also, pSMAD1/5/(8)9 was reduced in TAC mice. Pre-emptive treatment of mice with THR123 and THR184, during the four-week TAC period, normalized pSMAD1/5/(8)9 levels in the LV, attenuated overexpression of remodeling-related genes (Col 1α1, β-MHC, BNP), palliated structural damage (hypertrophy and fibrosis) and alleviated LV dysfunction (systolic and diastolic). THR184 administration, starting fifteen days after TAC, halted the ongoing remodeling and partially reversed LV dysfunction. The reverse remodeling after pressure overload release was facilitated by THR184. Both peptides diminished the TGF-β1-induced hypertrophic gene program in cardiomyocytes, collagen transcriptional activation in fibroblasts, and differentiation of cardiac fibroblasts to myofibroblasts. Molecular docking suggests that both peptides bind with similar binding energies to the BMP7 binding domain at the BMPR1A. The present study results provide a preclinical proof-of-concept of potential therapeutic benefits of BMP7-based small peptides, which function as agonists of BMPR1A, against the pathological LV remodeling in the context of aortic stenosis.
Collapse
Affiliation(s)
| | - Aritz Gil
- Instituto de Investigación Sanitaria Valdecillla (IDIVAL), Santander, Spain; Servicio de Cardiología, Hospital Universitario Marqués de Valdecilla (HUMV), Santander, Spain
| | - Víctor Expósito
- Instituto de Investigación Sanitaria Valdecillla (IDIVAL), Santander, Spain; Servicio de Cardiología, Hospital Universitario Marqués de Valdecilla (HUMV), Santander, Spain
| | - Fernando Martínez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Centro de Investigación Biomédica en RED en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan M Redondo
- Centro de Investigación Biomédica en RED en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Gene regulation in cardiovascular remodeling and inflammation group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María A Hurlé
- Instituto de Investigación Sanitaria Valdecillla (IDIVAL), Santander, Spain; Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
| | - J Francisco Nistal
- Instituto de Investigación Sanitaria Valdecillla (IDIVAL), Santander, Spain; Centro de Investigación Biomédica en RED en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Departamento de Ciencias Médicas y Quirúrgicas, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Servicio de Cirugía Cardiovascular, Hospital Universitario Marqués de Valdecilla (HUMV), Santander, Spain.
| | - Raquel García
- Instituto de Investigación Sanitaria Valdecillla (IDIVAL), Santander, Spain; Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain.
| |
Collapse
|
179
|
Srivastava H, Pozzoli M, Lau E. Defining the Roles of Cardiokines in Human Aging and Age-Associated Diseases. FRONTIERS IN AGING 2022; 3:884321. [PMID: 35821831 PMCID: PMC9261440 DOI: 10.3389/fragi.2022.884321] [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: 02/26/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022]
Abstract
In recent years an expanding collection of heart-secreted signaling proteins have been discovered that play cellular communication roles in diverse pathophysiological processes. This minireview briefly discusses current evidence for the roles of cardiokines in systemic regulation of aging and age-associated diseases. An analysis of human transcriptome and secretome data suggests the possibility that many other cardiokines remain to be discovered that may function in long-range physiological regulations. We discuss the ongoing challenges and emerging technologies for elucidating the identity and function of cardiokines in endocrine regulations.
Collapse
Affiliation(s)
- Himangi Srivastava
- Department of Medicine/Cardiology, School of Medicine, University of Colorado, Aurora, CO, United States
- Consortium for Fibrosis Research and Translation, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Marina Pozzoli
- Department of Medicine/Cardiology, School of Medicine, University of Colorado, Aurora, CO, United States
- Consortium for Fibrosis Research and Translation, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Edward Lau
- Department of Medicine/Cardiology, School of Medicine, University of Colorado, Aurora, CO, United States
- Consortium for Fibrosis Research and Translation, School of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
180
|
Numata G, Takimoto E. Cyclic GMP and PKG Signaling in Heart Failure. Front Pharmacol 2022; 13:792798. [PMID: 35479330 PMCID: PMC9036358 DOI: 10.3389/fphar.2022.792798] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclic guanosine monophosphate (cGMP), produced by guanylate cyclase (GC), activates protein kinase G (PKG) and regulates cardiac remodeling. cGMP/PKG signal is activated by two intrinsic pathways: nitric oxide (NO)-soluble GC and natriuretic peptide (NP)-particulate GC (pGC) pathways. Activation of these pathways has emerged as a potent therapeutic strategy to treat patients with heart failure, given cGMP-PKG signaling is impaired in heart failure with reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF). Large scale clinical trials in patients with HFrEF have shown positive results with agents that activate cGMP-PKG pathways. In patients with HFpEF, however, benefits were observed only in a subgroup of patients. Further investigation for cGMP-PKG pathway is needed to develop better targeting strategies for HFpEF. This review outlines cGMP-PKG pathway and its modulation in heart failure.
Collapse
Affiliation(s)
- Genri Numata
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
- Department of Advanced Translational Research and Medicine in Management of Pulmonary Hypertension, The University of Tokyo Hospital, Tokyo, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, United States
| |
Collapse
|
181
|
Remodeling and Fibrosis of the Cardiac Muscle in the Course of Obesity-Pathogenesis and Involvement of the Extracellular Matrix. Int J Mol Sci 2022; 23:ijms23084195. [PMID: 35457013 PMCID: PMC9032681 DOI: 10.3390/ijms23084195] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/09/2022] [Indexed: 12/16/2022] Open
Abstract
Obesity is a growing epidemiological problem, as two-thirds of the adult population are carrying excess weight. It is a risk factor for the development of cardiovascular diseases (hypertension, ischemic heart disease, myocardial infarct, and atrial fibrillation). It has also been shown that chronic obesity in people may be a cause for the development of heart failure with preserved ejection fraction (HFpEF), whose components include cellular hypertrophy, left ventricular diastolic dysfunction, and increased extracellular collagen deposition. Several animal models with induced obesity, via the administration of a high-fat diet, also developed increased heart fibrosis as a result of extracellular collagen accumulation. Excessive collagen deposition in the extracellular matrix (ECM) in the course of obesity may increase the stiffness of the myocardium and thereby deteriorate the heart diastolic function and facilitate the occurrence of HFpEF. In this review, we include a rationale for that process, including a discussion about possible putative factors (such as increased renin–angiotensin–aldosterone activity, sympathetic overdrive, hemodynamic alterations, hypoadiponectinemia, hyperleptinemia, and concomitant heart diseases). To address the topic clearly, we include a description of the fundamentals of ECM turnover, as well as a summary of studies assessing collagen deposition in obese individuals.
Collapse
|
182
|
Meems L, Artola Arita V, Velt M, Dudink E, Crijns H, Van Gelder I, Rienstra M. Increased plasma levels of NT-proBNP, Troponin T and GDF-15 are driven by persistent AF and associated comorbidities: Data from the AF-RISK study. IJC HEART & VASCULATURE 2022; 39:100987. [PMID: 35281756 PMCID: PMC8914333 DOI: 10.1016/j.ijcha.2022.100987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/26/2022] [Indexed: 11/18/2022]
Affiliation(s)
- L.M.G. Meems
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - V. Artola Arita
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M. Velt
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - E.A.M.P. Dudink
- Maastricht University Medical Center+ and Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - H.J.G.M. Crijns
- Maastricht University Medical Center+ and Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - I.C. Van Gelder
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M. Rienstra
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Corresponding author at: Department of Cardiology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, the Netherlands.
| |
Collapse
|
183
|
Vasileiou PVS, Siasos G, Gorgoulis VG. Molecular biomarkers in cardio-oncology: Where we stand and where we are heading. Bioessays 2022; 44:e2100234. [PMID: 35352831 DOI: 10.1002/bies.202100234] [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: 10/04/2021] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 11/08/2022]
Abstract
Until recently, cardiotoxicity in the setting of a malignant disease was attributed solely to the detrimental effects of chemo- and/or radio-therapy to the heart. On this account, the focus was on the evaluation of well-established cardiac biomarkers for the early detection of myocardial damage. Currently, this view has been revised. Cardiotoxicity is not restricted to a single organ but instead affects the endothelium as a whole. Indeed, it has come into light that not only cancer therapy but also malignant cells per se can impair the cardiovascular system, through a paracrine and endocrine mode of action. Even more intriguingly, a clear interplay between molecular pathways involved in cancer and cardiovascular disease has become prevalent, suggesting a common nominator that governs the pathophysiology of these two entities. Taken together, our strategy in the quest of novel biomarkers in the emerging field of cardio-oncology should be critically reshaped.
Collapse
Affiliation(s)
- Panagiotis V S Vasileiou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerasimos Siasos
- Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK.,Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK
| |
Collapse
|
184
|
Fan S, Hu Y. Integrative analyses of biomarkers and pathways for heart failure. BMC Med Genomics 2022; 15:72. [PMID: 35346191 PMCID: PMC8962515 DOI: 10.1186/s12920-022-01221-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
Abstract
Background
Heart failure (HF) is the most common potential cause of death, causing a huge health and economic burden all over the world. So far, some impressive progress has been made in the study of pathogenesis. However, the underlying molecular mechanisms leading to this disease remain to be fully elucidated.
Methods
The microarray data sets of GSE76701, GSE21610 and GSE8331 were retrieved from the gene expression comprehensive database (GEO). After merging all microarray data and adjusting batch effects, differentially expressed genes (DEG) were determined. Functional enrichment analysis was performed based on Gene Ontology (GO) resources, Kyoto Encyclopedia of Genes and Genomes (KEGG) resources, gene set enrichment analysis (GSEA), response pathway database and Disease Ontology (DO). Protein protein interaction (PPI) network was constructed using string database. Combined with the above important bioinformatics information, the potential key genes were selected. The comparative toxicological genomics database (CTD) is used to explore the interaction between potential key genes and HF.
Results
We identified 38 patients with heart failure and 16 normal controls. There were 315 DEGs among HF samples, including 278 up-regulated genes and 37 down-regulated genes. Pathway enrichment analysis showed that most DEGs were significantly enriched in BMP signal pathway, transmembrane receptor protein serine/threonine kinase signal pathway, extracellular matrix, basement membrane, glycosaminoglycan binding, sulfur compound binding and so on. Similarly, GSEA enrichment analysis showed that DEGs were mainly enriched in extracellular matrix and extracellular matrix related proteins. BBS9, CHRD, BMP4, MYH6, NPPA and CCL5 are central genes in PPI networks and modules.
Conclusions
The enrichment pathway of DEGs and GO may reveal the molecular mechanism of HF. Among them, target genes EIF1AY, RPS4Y1, USP9Y, KDM5D, DDX3Y, NPPA, HBB, TSIX, LOC28556 and XIST are expected to become new targets for heart failure. Our findings provide potential biomarkers or therapeutic targets for the further study of heart failure and contribute to the development of advanced prediction, diagnosis and treatment strategies.
Collapse
|
185
|
Wang Y, Wang Y, Han X, Sun J, Li C, Adhikari BK, Zhang J, Miao X, Chen Z. Cardio-Oncology: A Myriad of Relationships Between Cardiovascular Disease and Cancer. Front Cardiovasc Med 2022; 9:727487. [PMID: 35369296 PMCID: PMC8968416 DOI: 10.3389/fcvm.2022.727487] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 02/21/2022] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease (CVD) and cancer are the leading causes of death worldwide. With an increasing number of the elderly population, and early cancer screening and treatment, the number of cancers cases are rising, while the mortality rate is decreasing. However, the number of cancer survivors is increasing yearly. With the prolonged life span of cancer patients, the adverse effects of anti-tumor therapy, especially CVD, have gained enormous attention. The incidence of cardiovascular events such as cardiac injury or cardiovascular toxicity is higher than malignant tumors' recurrence rate. Numerous clinical studies have also shifted their focus from the study of a single disease to the interdisciplinary study of oncology and cardiology. Previous studies have confirmed that anti-tumor therapy can cause CVD. Additionally, the treatment of CVD is also related to the tumors incidence. It is well established that the increased incidence of CVD in cancer patients is probably due to an unmodified unhealthy lifestyle among cancer survivors or cardiotoxicity caused by anti-cancer therapy. Nevertheless, some patients with CVD have a relatively increased cancer risk because CVD and malignant tumors are highly overlapping risk factors, including gender, age, hypertension, diabetes, hyperlipidemia, inflammation, and obesity. With advancements in the diagnosis and treatment, many patients simultaneously suffer from CVD and cancer, and most of them have a poor prognosis. Therefore, clinicians should understand the relationship between CVD and tumors, effectively identify the primary and secondary prevention for these diseases, and follow proper treatment methods.
Collapse
Affiliation(s)
- Yinghui Wang
- Department of Cardiovascular Centre, The First Hospital of Jilin University, Changchun, China
| | - Yonggang Wang
- Department of Cardiovascular Centre, The First Hospital of Jilin University, Changchun, China
| | - Xiaorong Han
- Department of Cardiovascular Centre, The First Hospital of Jilin University, Changchun, China
| | - Jian Sun
- Department of Cardiovascular Centre, The First Hospital of Jilin University, Changchun, China
| | - Cheng Li
- Department of Cardiovascular Centre, The First Hospital of Jilin University, Changchun, China
| | | | - Jin Zhang
- Department of Cardiovascular Centre, The First Hospital of Jilin University, Changchun, China
| | - Xiao Miao
- The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Xiao Miao
| | - Zhaoyang Chen
- Department of Cardiology, Union Hospital, Fujian Medical University, Fuzhou, China
- Zhaoyang Chen
| |
Collapse
|
186
|
Li M, Xu Y, Wu J, Wu C, Li A, Ji X. Circulating N-Terminal Probrain Natriuretic Peptide Levels in Relation to Ischemic Stroke and Its Subtypes: A Mendelian Randomization Study. Front Genet 2022; 13:795479. [PMID: 35273636 PMCID: PMC8902306 DOI: 10.3389/fgene.2022.795479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/10/2022] [Indexed: 12/01/2022] Open
Abstract
Mendelian randomization was used to evaluate the potential causal association between N-terminal probrain natriuretic peptide (NT-proBNP) and ischemic stroke based on summary statistics data from large-scale genome-wide association studies. Three single-nucleotide polymorphisms (SNPs) rs198389, rs13107325, and rs11105306 associated with NT-proBNP levels found in large general populations and in patients with acute heart disease were used as instrumental variables. The results of genetic association analysis of each single SNP show that there is no significant association between NT-proBNP levels and ischemic stroke or its subtypes, whereas rs198389 alone has a suggestive association with large-artery atherosclerosis stroke. The MR analysis of three SNPs shows that NT-proBNP levels may reduce the risk of small-vessel occlusion stroke suggestively. This genetic analysis provides insights into the pathophysiology and treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ming Li
- China-America Institute of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yi Xu
- China-America Institute of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jiaqi Wu
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Chuanjie Wu
- Department of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ang Li
- Department of Biomedical Engineering, Columbia University, New York City, NY, United States
| | - Xunming Ji
- China-America Institute of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
187
|
Li W, Lee SH, Kim SH. Carbon monoxide releasing molecule-2 suppresses stretch-activated atrial natriuretic peptide secretion by activating large-conductance calcium-activated potassium channels. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2022; 26:125-133. [PMID: 35203062 PMCID: PMC8890946 DOI: 10.4196/kjpp.2022.26.2.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 11/15/2022]
Abstract
Carbon monoxide (CO) is a known gaseous bioactive substance found across a wide array of body systems. The administration of low concentrations of CO has been found to exert an anti-inflammatory, anti-apoptotic, anti-hypertensive, and vaso-dilatory effect. To date, however, it has remained unknown whether CO influences atrial natriuretic peptide (ANP) secretion. This study explores the effect of CO on ANP secretion and its associated signaling pathway using isolated beating rat atria. Atrial perfusate was collected for 10 min for use as a control, after which high atrial stretch was induced by increasing the height of the outflow catheter. Carbon monoxide releasing molecule-2 (CORM-2; 10, 50, 100 µM) and hemin (HO-1 inducer; 0.1, 1, 50 µM), but not CORM-3 (10, 50, 100 µM), decreased high stretch-induced ANP secretion. However, zinc porphyrin (HO-1 inhibitor) did not affect ANP secretion. The order of potency for the suppression of ANP secretion was found to be hemin > CORM-2 >> CORM-3. The suppression of ANP secretion by CORM-2 was attenuated by pretreatment with 5-hydroxydecanoic acid, paxilline, and 1H-[1,2,4] oxadiazolo [4,3-a] quinoxalin-1-one, but not by diltiazem, wortmannin, LY-294002, or NG-nitro-L-arginine methyl ester. Hypoxic conditions attenuated the suppressive effect of CORM-2 on ANP secretion. In sum, these results suggest that CORM-2 suppresses ANP secretion via mitochondrial KATP channels and large conductance Ca2+-activated K+ channels.
Collapse
Affiliation(s)
- Weijian Li
- Departments of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Korea
| | - Sun Hwa Lee
- Departments of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54907, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University Jeonju 54907, Korea
| | - Suhn Hee Kim
- Departments of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University Jeonju 54907, Korea
| |
Collapse
|
188
|
Cao YX, Li S, Liu HH, Zhang M, Guo YL, Wu NQ, Zhu CG, Dong Q, Sun J, Dou KF, Li JJ. Prognostic Value of N-Terminal Pro-B-Type Natriuretic Peptide and High-Sensitivity C-Reactive Protein in Patients With Previous Myocardial Infarction. Front Cardiovasc Med 2022; 9:797297. [PMID: 35282356 PMCID: PMC8907519 DOI: 10.3389/fcvm.2022.797297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background Patients with previous myocardial infarction (MI) have a poor prognosis and stratification for recurrent major adverse cardiovascular events (MACE) among these patients is of considerable interest. N-terminal pro-B-type natriuretic peptide (NT-proBNP) and high-sensitivity C-reactive protein (hs-CRP) are considered to be potential cardiovascular risk factors, but less is known about their prognostic importance in post-MI patients. This study aimed to evaluate the prognostic value of NT-proBNP and hs-CRP alone or together in patients who reported a prior MI. Methods In this prospective study, we consecutively enrolled 3,306 post-MI patients to assess the recurrent MACE. The predictive values of NT-proBNP and hs-CRP alone and together were assessed by multivariable Cox regression using hazard ratios (HR) and 95% confidence intervals (CI). Results During the 4-year follow-up period, 335 patients developed recurrent MACE. Multivariate Cox regression analysis showed a significant correlation between NT-proBNP levels and MACE (HR: 2.99, 95%CI: 2.06–4.36, p < 0.001), hard endpoints (HR: 5.44, 95%CI: 2.99–9.90, p < 0.001), cardiac mortality (HR: 5.92, 95%CI: 2.34–14.96, p < 0.001) and all-cause mortality (HR: 5.03, 95%CI: 2.51–10.09, p < 0.001). However, hs-CRP was not an independent predictor after adjusting for NT-proBNP. When patients were divided into six groups by using tertiles values of NT-proBNP and median values of hsCRP, patients with high NT-proBNP/hs-CRP values were 3.27 times more likely to experience MACE than patients with low NT-proBNP/hs-CRP values. The addition of NT-proBNP and hs-CRP to a prognostic model revealed a significant improvement in C-statistic, net reclassification, and integrated discrimination. Conclusions Increased NT-proBNP levels were associated with long-term worse outcomes and the combination of NT-proBNP and hs-CRP has an incremental value in the further risk stratification of post-MI patients.
Collapse
Affiliation(s)
- Ye-Xuan Cao
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Beijing Chaoyang Hospital Affiliated to Capital University of Medical Science, Beijing, China
| | - Sha Li
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui-Hui Liu
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Zhang
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan-Lin Guo
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na-Qiong Wu
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng-Gang Zhu
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Dong
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Sun
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ke-Fei Dou
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Ke-Fei Dou
| | - Jian-Jun Li
- Cardiovascular Metabolism Center, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Jian-Jun Li ; orcid.org/0000-0003-2536-4364
| |
Collapse
|
189
|
Multi-Omics Characterization of a Human Stem Cell-Based Model of Cardiac Hypertrophy. Life (Basel) 2022; 12:life12020293. [PMID: 35207580 PMCID: PMC8875317 DOI: 10.3390/life12020293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiac hypertrophy is an important and independent risk factor for the development of cardiac myopathy that may lead to heart failure. The mechanisms underlying the development of cardiac hypertrophy are yet not well understood. To increase the knowledge about mechanisms and regulatory pathways involved in the progression of cardiac hypertrophy, we have developed a human induced pluripotent stem cell (hiPSC)-based in vitro model of cardiac hypertrophy and performed extensive characterization using a multi-omics approach. In a series of experiments, hiPSC-derived cardiomyocytes were stimulated with Endothelin-1 for 8, 24, 48, and 72 h, and their transcriptome and secreted proteome were analyzed. The transcriptomic data show many enriched canonical pathways related to cardiac hypertrophy already at the earliest time point, e.g., cardiac hypertrophy signaling. An integrated transcriptome–secretome analysis enabled the identification of multimodal biomarkers that may prove highly relevant for monitoring early cardiac hypertrophy progression. Taken together, the results from this study demonstrate that our in vitro model displays a hypertrophic response on both transcriptomic- and secreted-proteomic levels. The results also shed novel insights into the underlying mechanisms of cardiac hypertrophy, and novel putative early cardiac hypertrophy biomarkers have been identified that warrant further investigation to assess their potential clinical relevance.
Collapse
|
190
|
Cannone V, Burnett JC. Natriuretic Peptides and Blood Pressure Homeostasis: Implications for MANP, a Novel Guanylyl Cyclase a Receptor Activator for Hypertension. Front Physiol 2022; 12:815796. [PMID: 35222065 PMCID: PMC8878907 DOI: 10.3389/fphys.2021.815796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/24/2021] [Indexed: 12/11/2022] Open
Abstract
The heart serves as an endocrine organ producing the hormones atrial natriuretic peptide (ANP) and b-type natriuretic peptide (BNP) which via the guanylyl cyclase A (GC-A) receptor and the second messenger cGMP participate in blood pressure homeostasis under physiologic conditions. Genetic models of the ANP gene or the GCA receptor together with genomic medicine have solidified the concept that both cardiac hormones are fundamental for blood pressure homeostasis and when deficient or disrupted they may contribute to human hypertension. Advances in peptide engineering have led to novel peptide therapeutics including the ANP-analog MANP for human hypertension. Most importantly a first in human study of MANP in essential hypertension has demonstrated its unique properties of aldosterone suppression and blood pressure reduction. Physiology and pharmacology ultimately lead us to innovative peptide-based therapeutics to reduce the burden of cardiovascular disease.
Collapse
Affiliation(s)
- Valentina Cannone
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- *Correspondence: Valentina Cannone,
| | - John C. Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
191
|
Bicciato G, Arnold M, Gebhardt A, Katan M. Precision medicine in secondary prevention of ischemic stroke: how may blood-based biomarkers help in clinical routine? An expert opinion. Curr Opin Neurol 2022; 35:45-54. [PMID: 34839341 DOI: 10.1097/wco.0000000000001011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW One in eight patients unfortunately suffers a new stroke within 5 years of their first stroke, even today. Research in precision medicine could lead to a more individualized treatment allocation, possibly achieving lower recurrence rates of ischemic stroke. In this narrative review, we aim to discuss potential clinical implementation of several promising candidate blood biomarkers. RECENT FINDINGS We discuss specifically some promising blood-based biomarkers, which may improve the identification of underlying causes as well as risk stratification of patients according to their specific cerebrovascular risk factor pattern. SUMMARY Multimodal profiling of ischemic stroke patients by means of blood biomarkers, in addition to established clinical and neuroradiological data, may allow in the future a refinement of decision algorithms for treatment allocation in secondary ischemic stroke prevention.
Collapse
Affiliation(s)
- Giulio Bicciato
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | | | | | | |
Collapse
|
192
|
Lisco G, Giagulli VA, Iovino M, Zupo R, Guastamacchia E, De Pergola G, Iacoviello M, Triggiani V. Endocrine system dysfunction and chronic heart failure: a clinical perspective. Endocrine 2022; 75:360-376. [PMID: 34713389 PMCID: PMC8553109 DOI: 10.1007/s12020-021-02912-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/13/2021] [Indexed: 11/01/2022]
Abstract
Chronic heart failure (CHF) leads to an excess of urgent ambulatory visits, recurrent hospital admissions, morbidity, and mortality regardless of medical and non-medical management of the disease. This excess of risk may be attributable, at least in part, to comorbid conditions influencing the development and progression of CHF. In this perspective, the authors examined and described the most common endocrine disorders observed in patients with CHF, particularly in individuals with reduced ejection fraction, aiming to qualify the risks, quantify the epidemiological burden and discuss about the potential role of endocrine treatment. Thyroid dysfunction is commonly observed in patients with CHF, and sometimes it could be the consequence of certain medications (e.g., amiodarone). Male and female hypogonadism may also coexist in this clinical context, contributing to deteriorating the prognosis of these patients. Furthermore, growth hormone deficiency may affect the development of adult myocardium and predispose to CHF. Limited recommendation suggests to screen endocrine disorders in CHF patients, but it could be interesting to evaluate possible endocrine dysfunction in this setting, especially when a high suspicion coexists. Data referring to long-term safety and effectiveness of endocrine treatments in patients with CHF are limited, and their impact on several "hard" endpoints (such as hospital admission, all-cause, and cardiovascular mortality) are still poorly understood.
Collapse
Affiliation(s)
- Giuseppe Lisco
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Vito Angelo Giagulli
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Michele Iovino
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Roberta Zupo
- National Institute of Gastroenterology, Saverio de Bellis, Research Hospital, Castellana Grotte, Bari, Italy
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Giovanni De Pergola
- National Institute of Gastroenterology, Saverio de Bellis, Research Hospital, Castellana Grotte, Bari, Italy
- Clinical Nutrition Unit, Medical Oncology, Department of Internal Medicine and Clinical Oncology, University of Bari, School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Massimo Iacoviello
- Department of Medical and Surgical Sciences, Cardiology Department, University of Foggia, Foggia, Italy
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
193
|
Yu B, Yao S, Liu L, Li H, Zhu J, Li M, Han S, Yu Z. The role of polypeptide PDTLN1 in suppression of PI3K/AKT signaling causes cardiogenetic disorders in vitro and in vivo. Life Sci 2022; 289:120244. [PMID: 34922940 DOI: 10.1016/j.lfs.2021.120244] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/04/2021] [Accepted: 12/13/2021] [Indexed: 11/28/2022]
Abstract
AIMS A new polypeptide, PDTLN1, derived from the human Talin-1 protein, which is highly expressed in both myocardial tissue and maternal peripheral blood of aborted fetuses with congenital heart disease (CHD). However, its role in cardiac developmental disorders has not been disclosed till now. In the present study, we aim to assess the functions of PDTLN1 in heart development of zebrafish and cellular viability, proliferation, and apoptosis of P19 cells. MAIN METHODS Cellular viability was assessed by Cell Counting Kit-8, the EdU Kit was used to evaluate cellular proliferation, and apoptosic rate of P19 was examined using FITC Annexin-V staining followed by flow cytometry. The zebrafish embryos were divided into three groups: PEP group and NC group were microinjected with polypeptides, WT group without any intervention. The protein expression of PI3K/AKT were evaluated by western blotting. KEY FINDINGS PDTLN1 could suppress the proliferation, and facilitate apoptosis. PDTLN1 caused abnormal heart development of zebrafish embryos and the PDTLN1 (50 μM)-injected group showed an aberrant expression pattern of vmhc, amhc and cmlc2. Compared to the CTL group and SC79 group of P19 cells, the PDTLN1 group had a lower phosphorylated PI3K/AKT proteins level, decreased cellular viability and lower proliferation activity. SIGNIFICANCE PDTLN1 caused cardiac developmental defects in zebrafish, inhibited cellular viability, proliferation, and promoted apoptosis of P19 cells via suppressing the PI3K/AKT signaling pathway. Our findings provide a fresh perspective on the functional mechanism of human-derived peptides and may promote novel diagnostic biomarkers detection and therapeutic targets in CHD.
Collapse
Affiliation(s)
- Boshi Yu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, China; Nanjing Maternity and Child Health Care Hospital, Tian Fei Xiang, Nanjing, Jiangsu, China
| | - Shuwen Yao
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, China; Nanjing Maternity and Child Health Care Hospital, Tian Fei Xiang, Nanjing, Jiangsu, China
| | - Linjie Liu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, China; Nanjing Maternity and Child Health Care Hospital, Tian Fei Xiang, Nanjing, Jiangsu, China
| | - Huimin Li
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, China; Nanjing Maternity and Child Health Care Hospital, Tian Fei Xiang, Nanjing, Jiangsu, China
| | - Jingai Zhu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, China; Nanjing Maternity and Child Health Care Hospital, Tian Fei Xiang, Nanjing, Jiangsu, China
| | - Mengmeng Li
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, China; Nanjing Maternity and Child Health Care Hospital, Tian Fei Xiang, Nanjing, Jiangsu, China
| | - Shuping Han
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, China; Nanjing Maternity and Child Health Care Hospital, Tian Fei Xiang, Nanjing, Jiangsu, China.
| | - Zhangbin Yu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, China; Nanjing Maternity and Child Health Care Hospital, Tian Fei Xiang, Nanjing, Jiangsu, China.
| |
Collapse
|
194
|
Wu Y, Zhou Y, Huang J, Ma K, Yuan T, Jiang Y, Ye M, Li J. The Role of Sorting Nexin 17 in Cardiac Development. Front Cardiovasc Med 2022; 8:748891. [PMID: 34988124 PMCID: PMC8720881 DOI: 10.3389/fcvm.2021.748891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/31/2021] [Indexed: 11/13/2022] Open
Abstract
Sorting nexin 17 (SNX17), a member of sorting nexin (SNX) family, acts as a modulator for endocytic recycling of membrane proteins. Results from our previous study demonstrated the embryonic lethality of homozygous defect of SNX17. In this study, we investigated the role of SNX17 in rat fetal development. Specifically, we analyzed patterns of SNX17 messenger RNA (mRNA) expression in multiple rat tissues and found high expression in the cardiac outflow tract (OFT). This expression was gradually elevated during the cardiac OFT morphogenesis. Homozygous deletion of the SNX17 gene in rats resulted in mid-gestational embryonic lethality, which was accompanied by congenital heart defects, including the double-outlet right ventricle and atrioventricular and ventricular septal defects, whereas heterozygotes exhibited normal fetal development. Moreover, we found normal migration distance and the number of cardiac neural crest cells during the OFT morphogenesis. Although cellular proliferation in the cardiac OFT endocardial cushion was not affected, cellular apoptosis was significantly suppressed. Transcriptomic profiles and quantitative real-time PCR data in the cardiac OFT showed that SNX17 deletion resulted in abnormal expression of genes associated with cardiac development. Overall, these findings suggest that SNX17 plays a crucial role in cardiac development.
Collapse
Affiliation(s)
- Yufei Wu
- School of Medicine, Tongji University, Shanghai, China
| | - Yaqun Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Huang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ke Ma
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tianyou Yuan
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Jiang
- Department of Echocardiography, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, China
| | - Maoqing Ye
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jun Li
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
195
|
Takei Y. Evolution of the membrane/particulate guanylyl cyclase: From physicochemical sensors to hormone receptors. Gen Comp Endocrinol 2022; 315:113797. [PMID: 33957096 DOI: 10.1016/j.ygcen.2021.113797] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 12/26/2022]
Abstract
Guanylyl cyclase (GC) is an enzyme that produces 3',5'-cyclic guanosine monophosphate (cGMP), one of the two canonical cyclic nucleotides used as a second messenger for intracellular signal transduction. The GCs are classified into two groups, particulate/membrane GCs (pGC) and soluble/cytosolic GCs (sGC). In relation to the endocrine system, pGCs include hormone receptors for natriuretic peptides (GC-A and GC-B) and guanylin peptides (GC-C), while sGC is a receptor for nitric oxide and carbon monoxide. Comparing the functions of pGCs in eukaryotes, it is apparent that pGCs perceive various environmental factors such as light, temperature, and various external chemical signals in addition to endocrine hormones, and transmit the information into the cell using the intracellular signaling cascade initiated by cGMP, e.g., cGMP-dependent protein kinases, cGMP-sensitive cyclic nucleotide-gated ion channels and cGMP-regulated phosphodiesterases. Among vertebrate pGCs, GC-E and GC-F are localized on retinal epithelia and are involved in modifying signal transduction from the photoreceptor, rhodopsin. GC-D and GC-G are localized in olfactory epithelia and serve as sensors at the extracellular domain for external chemical signals such as odorants and pheromones. GC-G also responds to guanylin peptides in the urine, which alters sensitivity to other chemicals. In addition, guanylin peptides that are secreted into the intestinal lumen, a pseudo-external environment, act on the GC-C on the apical membrane for regulation of epithelial transport. In this context, GC-C and GC-G appear to be in transition from exocrine pheromone receptor to endocrine hormone receptor. The pGCs also exist in various deuterostome and protostome invertebrates, and act as receptors for environmental, exocrine and endocrine factors including hormones. Tracing the evolutionary history of pGCs, it appears that pGCs first appeared as a sensor for physicochemical signals in the environment, and then evolved to function as hormone receptors. In this review, the author proposes an evolutionary history of pGCs that highlights the emerging role of the GC/cGMP system for signal transduction in hormone action.
Collapse
Affiliation(s)
- Yoshio Takei
- Laboratory of Physiology, Department of Marine Bioscience, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba 277-8564, Japan.
| |
Collapse
|
196
|
|
197
|
Diaz-Arocutipa C, Saucedo-Chinchay J, Imazio M, Argulian E. Natriuretic peptides to differentiate constrictive pericarditis and restrictive cardiomyopathy: A systematic review and meta-analysis. Clin Cardiol 2021; 45:251-257. [PMID: 34967020 PMCID: PMC8922532 DOI: 10.1002/clc.23772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/06/2021] [Accepted: 12/21/2021] [Indexed: 11/11/2022] Open
Abstract
Previous studies have shown that natriuretic peptide levels are increased in patients with restrictive cardiomyopathy (RCM) but not in constrictive pericarditis (CP). We performed a systematic review and meta-analysis to evaluate the diagnostic utility of B-type natriuretic peptide (BNP) and N-terminal pro-brain natriuretic peptide (NT-proBNP) to differentiate CP and RCM. We searched electronic databases from inception to January 07, 2021. Studies involving adult patients that assessed the utility of natriuretic peptides to differentiate CP and RCM were included. All meta-analyses were performed using a random-effects model. Seven studies (four case-control and three cohorts) involving 204 patients were included. The mean age ranged between 25.7 and 64.1 years and 77% of patients were men. BNP levels were significantly lower (standardized median difference [SMD], -1.48; 95% confidence interval [CI], -2.33 to -0.63) in patients with CP compared to RCM. The pooled area under the curve (AUC) of the BNP level was 0.81 (95% CI, 0.70-0.92). NT-proBNP (SMD, -0.86; 95% CI, -1.38 to -0.33) and log NT-proBNP (SMD, -1.89; 95% CI, -2.59 to -1.20) levels were significantly lower in patients with CP compared to RCM. Our review shows that BNP and NT-proBNP levels were significantly lower in patients with CP compared to RCM. The pooled AUC of BNP level showed a good diagnostic accuracy to differentiate both conditions.
Collapse
Affiliation(s)
- Carlos Diaz-Arocutipa
- Vicerrectorado de Investigación, Universidad San Ignacio de Loyola, Lima, Peru.,Programa de Atención Domiciliaria, EsSalud, Lima, Peru.,Asociación para el Desarrollo de la Investigación Estudiantil en Ciencias de la Salud (ADIECS), Lima, Peru
| | | | - Massimo Imazio
- Cardiothoracic Department, University Hospital "Santa Maria della Misericordia", Udine, Italy
| | - Edgar Argulian
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
198
|
Imamura T, Gonoi W, Hori M, Ueno Y, Narang N, Onoda H, Tanaka S, Nakamura M, Kataoka N, Ushijima R, Sobajima M, Fukuda N, Ueno H, Kinugawa K. Validation of Noninvasive Remote Dielectric Sensing System to Quantify Lung Fluid Levels. J Clin Med 2021; 11:jcm11010164. [PMID: 35011905 PMCID: PMC8745965 DOI: 10.3390/jcm11010164] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/28/2021] [Accepted: 12/28/2021] [Indexed: 01/10/2023] Open
Abstract
Background: The accuracy of the remote dielectric sensing (ReDSTM) system, which is a noninvasive electromagnetic-based technology to quantify lung fluid levels, particularly among those with small body size, remains uncertain. Methods: Hospitalized patients with and without heart failure underwent assessment of lung fluid levels with ReDS and successive chest computed tomography imaging. We performed a correlation analysis of the ReDS measurement, representing lung fluid levels, and computed tomography-derived high attenuation area percentage, which also provides a spatial quantification of lung fluid level. Results: A total of 46 patients (median 76 years old, 28 men), including 28 patients with heart failure, were included. The median ReDS value was 28% (interquartile: 23%, 33%), and the median percentage of high attenuation area was 21.6% (14.4%, 28.5%). ReDS values and percentage of high attenuation area were moderately correlated (r = 0.65, p < 0.001), irrespective of the existence of heart failure. ReDS value independently predicted the percentage of high attenuation area seen on computed tomography (p < 0.001). Conclusions: The ReDS system may be a promising, noninvasive tool to quantify fluid lung levels, as validated by comparison with chest computed tomography imaging. Further studies are warranted to validate the utility and applicability of this technology to a variety of clinical scenarios.
Collapse
Affiliation(s)
- Teruhiko Imamura
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
- Correspondence: ; Tel.: +81-76-434-2281; Fax: +81-76-434-5026
| | - Wataru Gonoi
- Department of Radiology, Graduate School of Medicine, University of Tokyo, Tokyo 1138654, Japan;
| | - Masakazu Hori
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| | - Yohei Ueno
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| | - Nikhil Narang
- Advocate Christ Medical Center, Oak Lawn, IL 60453, USA;
| | - Hiroshi Onoda
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| | - Shuhei Tanaka
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| | - Makiko Nakamura
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| | - Naoya Kataoka
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| | - Ryuichi Ushijima
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| | - Mitsuo Sobajima
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| | - Nobuyuki Fukuda
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| | - Hiroshi Ueno
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| | - Koichiro Kinugawa
- Second Department of Internal Medicine, University of Toyama, Toyama 9300194, Japan; (M.H.); (Y.U.); (H.O.); (S.T.); (M.N.); (N.K.); (R.U.); (M.S.); (N.F.); (H.U.); (K.K.)
| |
Collapse
|
199
|
Discovery of small molecule guanylyl cyclase A receptor positive allosteric modulators. Proc Natl Acad Sci U S A 2021; 118:2109386118. [PMID: 34930837 DOI: 10.1073/pnas.2109386118] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2021] [Indexed: 12/13/2022] Open
Abstract
The particulate guanylyl cyclase A receptor (GC-A), via activation by its endogenous ligands atrial natriuretic peptide (ANP) and b-type natriuretic peptide (BNP), possesses beneficial biological properties such as blood pressure regulation, natriuresis, suppression of adverse remodeling, inhibition of the renin-angiotensin-aldosterone system, and favorable metabolic actions through the generation of its second messenger cyclic guanosine monophosphate (cGMP). Thus, the GC-A represents an important molecular therapeutic target for cardiovascular disease and its associated risk factors. However, a small molecule that is orally bioavailable and directly targets the GC-A to potentiate cGMP has yet to be discovered. Here, we performed a cell-based high-throughput screening campaign of the NIH Molecular Libraries Small Molecule Repository, and we successfully identified small molecule GC-A positive allosteric modulator (PAM) scaffolds. Further medicinal chemistry structure-activity relationship efforts of the lead scaffold resulted in the development of a GC-A PAM, MCUF-651, which enhanced ANP-mediated cGMP generation in human cardiac, renal, and fat cells and inhibited cardiomyocyte hypertrophy in vitro. Further, binding analysis confirmed MCUF-651 binds to GC-A and selectively enhances the binding of ANP to GC-A. Moreover, MCUF-651 is orally bioavailable in mice and enhances the ability of endogenous ANP and BNP, found in the plasma of normal subjects and patients with hypertension or heart failure, to generate GC-A-mediated cGMP ex vivo. In this work, we report the discovery and development of an oral, small molecule GC-A PAM that holds great potential as a therapeutic for cardiovascular, renal, and metabolic diseases.
Collapse
|
200
|
Liu Y, Li B, Ma Y, Huang Y, Ouyang F, Liu Q. Mendelian Randomization Integrating GWAS, eQTL, and mQTL Data Identified Genes Pleiotropically Associated With Atrial Fibrillation. Front Cardiovasc Med 2021; 8:745757. [PMID: 34977172 PMCID: PMC8719596 DOI: 10.3389/fcvm.2021.745757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Atrial fibrillation (AF) is the most common arrhythmia. Genome-wide association studies (GWAS) have identified more than 100 loci associated with AF, but the underlying biological interpretation remains largely unknown. The goal of this study is to identify gene expression and DNA methylation (DNAm) that are pleiotropically or potentially causally associated with AF, and to integrate results from transcriptome and methylome.Methods: We used the summary data-based Mendelian randomization (SMR) to integrate GWAS with expression quantitative trait loci (eQTL) studies and methylation quantitative trait loci (mQTL) studies. The HEIDI (heterogeneity in dependent instruments) test was introduced to test against the null hypothesis that there is a single causal variant underlying the association.Results: We prioritized 22 genes by eQTL analysis and 50 genes by mQTL analysis that passed the SMR & HEIDI test. Among them, 6 genes were overlapped. By incorporating consistent SMR associations between DNAm and AF, between gene expression and AF, and between DNAm and gene expression, we identified several mediation models at which a genetic variant exerted an effect on AF by altering the DNAm level, which regulated the expression level of a functional gene. One example was the genetic variant-cg18693985-CPEB4-AF axis.Conclusion: In conclusion, our integrative analysis identified multiple genes and DNAm sites that had potentially causal effects on AF. We also pinpointed plausible mechanisms in which the effect of a genetic variant on AF was mediated by genetic regulation of transcription through DNAm. Further experimental validation is necessary to translate the identified genes and possible mechanisms into clinical practice.
Collapse
Affiliation(s)
- Yaozhong Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Biao Li
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yingxu Ma
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yunying Huang
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Feifan Ouyang
- Department of Cardiology, Asklepios Klinik St. Georg, Hamburg, Germany
| | - Qiming Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Qiming Liu
| |
Collapse
|