1
|
Vinícius de Paula da Silva M, Vieira Alves I, Rodrigues Pereira Alves A, Soares Lemos V, Assis Lopes do Carmo G, Morato de Castilho F, Léo Gelape C. Crosstalk between cytokines, inflammation and pulmonary arterial hypertension in heart transplant patients. Cytokine 2024; 182:156709. [PMID: 39079217 DOI: 10.1016/j.cyto.2024.156709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 08/25/2024]
Abstract
BACKGROUND Heart transplant (HT) is a therapeutic option for patients with advanced heart failure (HF) refractory to optimized treatment. Patients with advanced HF often develop pulmonary arterial hypertension (PAH). PAH is defined as a condition in which the mean pulmonary artery pressure is greater than 20 mmHg. Inflammation is an important aspect of PAH development. In this context, the objective of this work was to evaluate the relationship between the inflammatory process and the development of HAP in patients undergoing HT. METHODS The levels of interleukins IL-6, IL-1β and TNF-α were obtained by ELISA and associated with CD68+ and CD66b neutrophil counts using the immunofluorescence technique in fragments of the pulmonary arteries of donors and patients with or without chagasic cardiomyopathy subjected to HT. RESULTS The results showed a positive, statistically significant correlation (p < 0.05) between right atrium pressure levels and IL-6. Furthermore, negative, moderate, and statistically significant correlations (p < 0.05) were observed between the variables cardiac index and TNF-α, and between the levels of transpulmonary pressure grandient and TNF-α. The study also revealed the presence of a statistically significant difference (p < 0.05) between patients who died within 30 days and the highest number of CD68 cells per square micrometer in the vessel of the donor and recipient patient. CONCLUSION Suggesting the presence of a pro-inflammatory profile in HT patients, independent of measured pulmonary artery pressure levels.
Collapse
Affiliation(s)
- Marcus Vinícius de Paula da Silva
- Department of Cardiovascular Surgery, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Ildernandes Vieira Alves
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais
| | | | - Virginia Soares Lemos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais
| | - Gabriel Assis Lopes do Carmo
- Department of Cardiovascular Surgery, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fábio Morato de Castilho
- Department of Cardiovascular Surgery, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cláudio Léo Gelape
- Department of Cardiovascular Surgery, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
2
|
Crisci G, Bobbio E, Gentile P, Bromage DI, Bollano E, Ferone E, Israr MZ, Heaney LM, Polte CL, Cannatà A, Salzano A. Biomarkers in Acute Myocarditis and Chronic Inflammatory Cardiomyopathy: An Updated Review of the Literature. J Clin Med 2023; 12:7214. [PMID: 38068265 PMCID: PMC10706911 DOI: 10.3390/jcm12237214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 02/15/2024] Open
Abstract
Myocarditis is a disease caused by cardiac inflammation that can progress to dilated cardiomyopathy, heart failure, and eventually death. Several etiologies, including autoimmune, drug-induced, and infectious, lead to inflammation, which causes damage to the myocardium, followed by remodeling and fibrosis. Although there has been an increasing understanding of pathophysiology, early and accurate diagnosis, and effective treatment remain challenging due to the high heterogeneity. As a result, many patients have poor prognosis, with those surviving at risk of long-term sequelae. Current diagnostic methods, including imaging and endomyocardial biopsy, are, at times, expensive, invasive, and not always performed early enough to affect disease progression. Therefore, the identification of accurate, cost-effective, and prognostically informative biomarkers is critical for screening and treatment. The review then focuses on the biomarkers currently associated with these conditions, which have been extensively studied via blood tests and imaging techniques. The information within this review was retrieved through extensive literature research conducted on major publicly accessible databases and has been collated and revised by an international panel of experts. The biomarkers discussed in the article have shown great promise in clinical research studies and provide clinicians with essential tools for early diagnosis and improved outcomes.
Collapse
Affiliation(s)
- Giulia Crisci
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy;
- Italian Clinical Outcome Research and Reporting Program (I-CORRP), 80131 Naples, Italy
| | - Emanuele Bobbio
- Department of Cardiology, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (E.B.); (E.B.)
- Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, 41390 Gothenburg, Sweden;
| | - Piero Gentile
- De Gasperis Cardio Center, Niguarda Hospital, 20162 Milan, Italy;
| | - Daniel I. Bromage
- Department of Cardiology, King’s College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK; (D.I.B.); (E.F.)
- Department of Cardiovascular Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 8AF, UK
| | - Entela Bollano
- Department of Cardiology, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (E.B.); (E.B.)
- Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, 41390 Gothenburg, Sweden;
| | - Emma Ferone
- Department of Cardiology, King’s College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK; (D.I.B.); (E.F.)
- Department of Cardiovascular Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 8AF, UK
| | - Muhammad Zubair Israr
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Groby Road, Leicester LE3 9QP, UK;
| | - Liam M. Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
| | - Christian L. Polte
- Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, 41390 Gothenburg, Sweden;
- Department of Clinical Physiology, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Antonio Cannatà
- Department of Cardiology, King’s College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK; (D.I.B.); (E.F.)
- Department of Cardiovascular Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 8AF, UK
| | - Andrea Salzano
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Groby Road, Leicester LE3 9QP, UK;
- Cardiology Unit, AORN A Cardarelli, 80131 Naples, Italy
| |
Collapse
|
3
|
Solberg OG, Aaberge L, Bosse G, Ueland T, Gullestad L, Aukrust P, Stavem K. Microvascular function and inflammatory activation in Takotsubo cardiomyopathy. ESC Heart Fail 2023; 10:3216-3222. [PMID: 37537779 PMCID: PMC10567652 DOI: 10.1002/ehf2.14461] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/14/2023] [Accepted: 06/21/2023] [Indexed: 08/05/2023] Open
Abstract
AIMS The aim of this study was to determine microvascular function in the acute phase of Takotsubo syndrome (TTS) and to identify inflammatory mediators that could reflect TTS-induced pathology. METHODS AND RESULTS The study included 20 females [median age 65 years; interquarile range (IQR) = 58-70 years] with TTS according to the Mayo diagnostic criteria. During heart catheterization, we determined the index of microvascular resistance (IMR) and drew blood samples almost simultaneously from the aorta and coronary sinus. Cardiac magnetic resonance imaging (MRI) was done in the acute phase. We present descriptive coronary physiology and cardiac MRI data and compare inflammatory biomarkers between samples from the aorta, coronary sinus, and venous samples after 3 months using the Wilcoxon signed-rank test. For comparison, we also analysed the actual biomarkers in venous blood from 15 healthy female controls. A supplementary analysis explored Spearman's rank correlation between the inflammatory biomarkers, IMR, MRI data, and cardiac biomarkers. The median IMR was 16.5 mmHg·s (IQR = 10.5-28.2 mmHg·s), which was only slightly higher than that in the reference populations. Seven (35%) of the study subjects had IMR > 25 mmHg·s, suggesting a microvascular dysfunction. IMR was not affected by time from symptom onset. According to MRI, the apical region of the left ventricle was affected in 65% of the subjects. The median ejection fraction was 41% (IQR = 31-48%). Biomarker analyses revealed elevation of markers for extracellular matrix remodelling and fibrosis, inflammation, immune activation, and upstream inflammation as compared with healthy controls. Only the levels of interleukin (IL)-1 receptor antagonist and soluble T-cell immunoglobulin mucin domain-3 (sTIM-3) were higher in the coronary sinus than in the aorta. No variable was significantly correlated with IMR. The IL-6 level in the aorta was inversely correlated with the left ventricular ejection fraction. Growth differentiation factor-15, osteoprotegerin, and von Willebrand factor levels in both aorta and coronary sinus were positively correlated with N-terminal-pro-brain-natriuretic peptide, while the correlations of IL-6 and sTIM-3 with N-terminal-pro-brain-natriuretic peptide were restricted to the aorta and coronary sinus, respectively. While most of the markers were within normal limits after 3 months, matrix metalloproteinase-9 increased during follow-up to reach levels higher than those in the healthy controls. CONCLUSION The median IMR was only slightly elevated in this study, but about one-third of the patients had values indicating microvascular dysfunction. The present study supports the involvement of several inflammatory pathways in TTS, including monocyte/macrophage activation, with sTIM-3 as a potential novel marker.
Collapse
Affiliation(s)
| | - Lars Aaberge
- Department of CardiologyOslo University HospitalOsloNorway
| | - Gerhard Bosse
- Department of RadiologyOslo University HospitalOsloNorway
| | - Thor Ueland
- Faculty of Medicine, Institute of Clinical MedicineUniversity of OsloOsloNorway
- K.G. Jebsen TRECUniversity of TromsøTromsøNorway
- Section of Clinical Immunology and Infectious DiseasesOslo University HospitalOsloNorway
| | - Lars Gullestad
- Department of CardiologyOslo University HospitalOsloNorway
- Faculty of Medicine, Institute of Clinical MedicineUniversity of OsloOsloNorway
- K.G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, Faculty of MedicineOslo University HospitalOsloNorway
| | - Pål Aukrust
- Faculty of Medicine, Institute of Clinical MedicineUniversity of OsloOsloNorway
- Section of Clinical Immunology and Infectious DiseasesOslo University HospitalOsloNorway
- Research Institute of Internal MedicineOslo University HospitalOsloNorway
| | - Knut Stavem
- Faculty of Medicine, Institute of Clinical MedicineUniversity of OsloOsloNorway
- Department of Pulmonary MedicineAkershus University HospitalLørenskogNorway
- Department of Health Services ResearchAkershus University HospitalLørenskogNorway
| |
Collapse
|
4
|
Crisci G, Israr MZ, Cittadini A, Bossone E, Suzuki T, Salzano A. Heart failure and trimethylamine N-oxide: time to transform a 'gut feeling' in a fact? ESC Heart Fail 2023; 10:1-7. [PMID: 36237140 PMCID: PMC9871667 DOI: 10.1002/ehf2.14205] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 01/27/2023] Open
Affiliation(s)
- Giulia Crisci
- Department of Translational Medical Sciences, Federico II University, Via S Pansini 5, Naples, 80131, Italy.,Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, 80131, Italy
| | - Muhammad Zubair Israr
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Groby road, Leicester, LE3 9QP, United Kingdom
| | - Antonio Cittadini
- Department of Translational Medical Sciences, Federico II University, Via S Pansini 5, Naples, 80131, Italy.,Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, 80131, Italy
| | - Eduardo Bossone
- Department of Public Health, Federico II University, Naples, 80138, Italy
| | - Toru Suzuki
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Groby road, Leicester, LE3 9QP, United Kingdom
| | - Andrea Salzano
- IRCCS Synlab SDN, Diagnostic and Nuclear Research Institute, Via E Gianturco 113, Naples, 80143, Italy
| |
Collapse
|
5
|
Ruozi G, Bortolotti F, Mura A, Tomczyk M, Falcione A, Martinelli V, Vodret S, Braga L, Dal Ferro M, Cannatà A, Zentilin L, Sinagra G, Zacchigna S, Giacca M. Cardioprotective factors against myocardial infarction selected in vivo from an AAV secretome library. Sci Transl Med 2022; 14:eabo0699. [PMID: 36044596 DOI: 10.1126/scitranslmed.abo0699] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Therapies for patients with myocardial infarction and heart failure are urgently needed, in light of the breadth of these conditions and lack of curative treatments. To systematically identify previously unidentified cardioactive biologicals in an unbiased manner in vivo, we developed cardiac FunSel, a method for the systematic, functional selection of effective factors using a library of 1198 barcoded adeno-associated virus (AAV) vectors encoding for the mouse secretome. By pooled vector injection into the heart, this library was screened to functionally select for factors that confer cardioprotection against myocardial infarction. After two rounds of iterative selection in mice, cardiac FunSel identified three proteins [chordin-like 1 (Chrdl1), family with sequence similarity 3 member C (Fam3c), and Fam3b] that preserve cardiomyocyte viability, sustain cardiac function, and prevent pathological remodeling. In particular, Chrdl1 exerted its protective activity by binding and inhibiting extracellular bone morphogenetic protein 4 (BMP4), which resulted in protection against cardiomyocyte death and induction of autophagy in cardiomyocytes after myocardial infarction. Chrdl1 also inhibited fibrosis and maladaptive cardiac remodeling by binding transforming growth factor-β (TGF-β) and preventing cardiac fibroblast differentiation into myofibroblasts. Production of secreted and circulating Chrdl1, Fam3c, and Fam3b from the liver also protected the heart from myocardial infarction, thus supporting the use of the three proteins as recombinant factors. Together, these findings disclose a powerful method for the in vivo, unbiased selection of tissue-protective factors and describe potential cardiac therapeutics.
Collapse
Affiliation(s)
- Giulia Ruozi
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Francesca Bortolotti
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy.,Cardiovascular Department, ASUGI, 34149 Trieste, Italy
| | - Antonio Mura
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Mateusz Tomczyk
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy.,British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, UK
| | - Antonella Falcione
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Valentina Martinelli
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Simone Vodret
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Luca Braga
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy.,British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, UK
| | | | - Antonio Cannatà
- Cardiovascular Department, ASUGI, 34149 Trieste, Italy.,British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, UK
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Gianfranco Sinagra
- Cardiovascular Department, ASUGI, 34149 Trieste, Italy.,Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Serena Zacchigna
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy.,Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy.,British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, UK.,Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| |
Collapse
|
6
|
Salzano A, D'Assante R, Iacoviello M, Triggiani V, Rengo G, Cacciatore F, Maiello C, Limongelli G, Masarone D, Sciacqua A, Filardi PP, Mancini A, Volterrani M, Vriz O, Castello R, Passantino A, Campo M, Modesti PA, De Giorgi A, Arcopinto M, Gargiulo P, Perticone M, Colao A, Milano S, Garavaglia A, Napoli R, Suzuki T, Bossone E, Marra AM, Cittadini A. Progressive right ventricular dysfunction and exercise impairment in patients with heart failure and diabetes mellitus: insights from the T.O.S.CA. Registry. Cardiovasc Diabetol 2022; 21:108. [PMID: 35710369 PMCID: PMC9204878 DOI: 10.1186/s12933-022-01543-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/16/2022] [Indexed: 01/01/2023] Open
Abstract
Background Findings from the T.O.S.CA. Registry recently reported that patients with concomitant chronic heart failure (CHF) and impairment of insulin axis (either insulin resistance—IR or diabetes mellitus—T2D) display increased morbidity and mortality. However, little information is available on the relative impact of IR and T2D on cardiac structure and function, cardiopulmonary performance, and their longitudinal changes in CHF. Methods Patients enrolled in the T.O.S.CA. Registry performed echocardiography and cardiopulmonary exercise test at baseline and at a patient-average follow-up of 36 months. Patients were divided into three groups based on the degree of insulin impairment: euglycemic without IR (EU), euglycemic with IR (IR), and T2D. Results Compared with EU and IR, T2D was associated with increased filling pressures (E/e′ratio: 15.9 ± 8.9, 12.0 ± 6.5, and 14.5 ± 8.1 respectively, p < 0.01) and worse right ventricular(RV)-arterial uncoupling (RVAUC) (TAPSE/PASP ratio 0.52 ± 0.2, 0.6 ± 0.3, and 0.6 ± 0.3 in T2D, EU and IR, respectively, p < 0.05). Likewise, impairment in peak oxygen consumption (peak VO2) in TD2 vs EU and IR patients was recorded (respectively, 15.8 ± 3.8 ml/Kg/min, 18.4 ± 4.3 ml/Kg/min and 16.5 ± 4.3 ml/Kg/min, p < 0.003). Longitudinal data demonstrated higher deterioration of RVAUC, RV dimension, and peak VO2 in the T2D group (+ 13% increase in RV dimension, − 21% decline in TAPSE/PAPS ratio and − 20% decrease in peak VO2). Conclusion The higher risk of death and CV hospitalizations exhibited by HF-T2D patients in the T.O.S.CA. Registry is associated with progressive RV ventricular dysfunction and exercise impairment when compared to euglycemic CHF patients, supporting the pivotal importance of hyperglycaemia and right chambers in HF prognosis. Trial registration ClinicalTrials.gov identifier: NCT023358017
Collapse
Affiliation(s)
- Andrea Salzano
- IRCCS SYNLAB SDN, Diagnostic and Nuclear Research Institute, Naples, Italy
| | - Roberta D'Assante
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, Italy
| | - Massimo Iacoviello
- Cardiology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122, Foggia, Italy
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari 'A Moro', Bari, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB) - IRCCS - Scientific Institute of Telese Terme, Telese Terme, BN, Italy
| | - Francesco Cacciatore
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Ciro Maiello
- Heart Transplantation Unit, Monaldi Hospital, Azienda Ospedaliera Dei Colli, Naples, Italy
| | - Giuseppe Limongelli
- Division of Cardiology, Monaldi Hospital, Azienda Ospedaliera Dei Colli, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Daniele Masarone
- Division of Cardiology, Monaldi Hospital, Azienda Ospedaliera Dei Colli, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Pasquale Perrone Filardi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.,Mediterranea Cardiocentro, Naples, Italy
| | - Antonio Mancini
- Operative Unit of Endocrinology, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Olga Vriz
- Heart Center Department, King Faisal Hospital & Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Roberto Castello
- Division of General Medicine, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | | | - Michela Campo
- Unit of Endocrinology and Metabolic Diseases, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Pietro A Modesti
- Dipartimento Di Medicina Sperimentale E Clinica, Università Degli Studi Di Firenze, Florence, Italy
| | - Alfredo De Giorgi
- Department of Medical Sciences, School of Medicine, Pharmacy and Prevention, University of Ferrara, Ferrara, Italy
| | - Michele Arcopinto
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, Italy
| | - Paola Gargiulo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Maria Perticone
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Annamaria Colao
- Clinical Medicine and Surgery Department, Federico II University, Naples, Italy
| | - Salvatore Milano
- Department of Laboratory Medicine, AOUP P. Giaccone, Palermo, Italy
| | | | - Raffaele Napoli
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Toru Suzuki
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, UK
| | - Eduardo Bossone
- Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, Italy.,Cardiology Division, A Cardarelli Hospital, Naples, Italy
| | - Alberto M Marra
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, Italy.,Center for Pulmonary Hypertension, Thoraxclinic at Heidelberg University Hospital, Heidelberg, Germany
| | - Antonio Cittadini
- Department of Translational Medical Sciences, Federico II University, Naples, Italy. .,Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, Italy. .,Department of Translational Medical Sciences, Division of Internal Medicine and Metabolism and Rehabilitation, Federico II University of Naples, Via S. Pansini 5, Bld.18, 1stfloor, 80131, Naples, Italy.
| | | |
Collapse
|
7
|
|
8
|
Salzano A, Cassambai S, Yazaki Y, Israr MZ, Bernieh D, Wong M, Suzuki T. The Gut Axis Involvement in Heart Failure: Focus on Trimethylamine N-oxide. Cardiol Clin 2022; 40:161-169. [PMID: 35465890 DOI: 10.1016/j.ccl.2021.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A novel pathophysiological model of interest is the association between heart failure (HF) and the gastrointestinal system, the 'gut hypothesis'. The choline and carnitine metabolic by-product, Trimethylamine N-oxide (TMAO) is one of the more prominent molecules associated with the link between HF and the gut. Indeed, TMAO levels are increased in HF populations and higher TMAO levels are associated with poor prognosis, whereas low TMAO levels either at baseline/follow up confer better prognosis. Considering that TMAO levels seem not to be affected by guideline-HF treatment, this model could represent a novel and independent therapeutic target for HF.
Collapse
Affiliation(s)
- Andrea Salzano
- IRCCS SDN, Diagnostic and Nuclear Research Institute, Via E Gianturco, 80143, Naples, Italy
| | - Shabana Cassambai
- Department of Cardiovascular Sciences, NIHR Leicester Cardiovascular Biomedical Research Centre, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Yoshiyuki Yazaki
- Department of Cardiovascular Sciences, NIHR Leicester Cardiovascular Biomedical Research Centre, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Muhammad Zubair Israr
- Department of Cardiovascular Sciences, NIHR Leicester Cardiovascular Biomedical Research Centre, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Dennis Bernieh
- Department of Cardiovascular Sciences, NIHR Leicester Cardiovascular Biomedical Research Centre, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Max Wong
- Department of Cardiovascular Sciences, NIHR Leicester Cardiovascular Biomedical Research Centre, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Toru Suzuki
- Department of Cardiovascular Sciences, NIHR Leicester Cardiovascular Biomedical Research Centre, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK.
| |
Collapse
|
9
|
Prausmüller S, Spinka G, Stasek S, Arfsten H, Bartko PE, Goliasch G, Hülsmann M, Pavo N. Neutrophil Activation/Maturation Markers in Chronic Heart Failure with Reduced Ejection Fraction. Diagnostics (Basel) 2022; 12:444. [PMID: 35204534 PMCID: PMC8871325 DOI: 10.3390/diagnostics12020444] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Neutrophils are critically involved in the immune response. Inflammatory stimuli alter the expression status of their surface molecule toolset, while inflammation-stimulated granulopoiesis might also influence their maturation status. Data on neutrophil status in heart failure with reduced ejection fraction (HFrEF) are scarce. The present study aims to evaluate the role of neutrophil CD11b, CD66b and CD64 expression in HFrEF. METHODS A total of 135 HFrEF patients and 43 controls were recruited. Mean fluorescence intensity of the activation/maturation markers CD11b, CD66b and CD64 was measured on neutrophils by flow cytometry. CD10 (neprilysin) expression was simultaneously determined. RESULTS Neutrophil CD64 expression was higher in HFrEF compared with controls, while CD11b/CD66b levels were similar. Neutrophil CD11b and CD66b showed a significant direct correlation to neutrophil CD10 expression (rs = 0.573, p < 0.001 and rs = 0.184, p = 0.033). Neutrophil CD11b and CD66b correlated inversely with heart failure severity reflected by NT-proBNP and NYHA class (NT-proBNP: rs = -0.243, p = 0.005 and rs = -0.250, p = 0.004; NYHA class: p = 0.032 and p = 0.055), whereas no association for CD64 could be found. Outcome analysis did not reveal a significant association between the expression of CD11b, CD66b and CD64 and all-cause mortality (p = ns). CONCLUSIONS The results underline the potential role of neutrophils in HFrEF disease pathophysiology and risk stratification and should stimulate further research, characterizing subpopulations of neutrophils and searching for key molecules involved in the downward spiral of inflammation and heart failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Martin Hülsmann
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (G.S.); (S.S.); (H.A.); (P.E.B.); (G.G.); (N.P.)
| | | |
Collapse
|
10
|
Wang Q, Zheng B, Chen P, Lei Y. Leptin and PCSK9 concentrations are associated with vascular endothelial cytokines in patients with stable coronary heart disease. Open Med (Wars) 2022; 17:185-190. [PMID: 35087951 PMCID: PMC8768505 DOI: 10.1515/med-2021-0400] [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: 10/08/2020] [Revised: 06/27/2021] [Accepted: 10/28/2021] [Indexed: 12/03/2022] Open
Abstract
Leptin and proprotein convertase subtilisin kexin 9 (PCSK9) play an important role in regulating blood lipid concentration. Recently, they have been found to show the ability to independently regulate the immune response. Vascular immune response has an important pathological function in the development of coronary heart disease (CHD) and thrombosis. The aim of this study was to explore the relationship between leptin, PCSK9, and vascular endothelial cell related inflammatory factors. First, detailed clinical information were collected and analyzed for 27 patients with stable CHD and corresponding 27 healthy controls. Second, using liquid-phase protein chip technology, leptin, PCSK9, and vascular-related inflammatory factors, such as E-selectin, vascular cell adhesion protein 1 (VCAM-1), intercellular cell adhesion molecule-1 (ICAM-1), interferon-gamma (IFN-γ), and interleukin-17 (IL-17), were detected on the same platform. Finally, the correlation between leptin, PCSK9, and the inflammatory factors was analyzed. Through collecting clinical information of patients, it was suggested that there was a significant positive correlation between leptin and blood lipid level in CHD. Compared with healthy people, the levels of leptin, PCSK9, E-selectin, and ICAM-1 were significantly high in patients with CHD. There was a high positive correlation between leptin and E-selectin, ICAM-1, IFN-γ, and IL-17. Also, a high positive correlation between PCSK9 and E-selectin, IFN-γ, and IL-17 concentrations was observed. In general, leptin and PCSK9 may not only be able to regulate lipid metabolism, but may also be able to regulate inflammation in CHD.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Cardiovascular Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Chaoyang District , Beijing 100102 , China
| | - Bo Zheng
- Department of Cardiovascular Medicine, Affiliated Hospital of Binzhou Medical University , BinZhou City , China
| | - Peng Chen
- Department of Molecular Biology, Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Dongcheng District , Beijing 100700 , China
| | - Yan Lei
- Department of Molecular Biology, Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Dongcheng District , Beijing 100700 , China
| |
Collapse
|
11
|
Zha L, Dong J, Chen Q, Liao Y, Zhang H, Xie T, Tang T, Xia N, Zhang M, Jiao J, Zhou Y, Wu J, Yang X, Xu C, Wang QK, Tu X, Cheng X, Nie S. Genetic association analysis between IL9 and coronary artery disease in a Chinese Han population. Cytokine 2021; 150:155761. [PMID: 34814015 DOI: 10.1016/j.cyto.2021.155761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/04/2021] [Accepted: 11/02/2021] [Indexed: 11/28/2022]
Abstract
Interleukin-9 (IL-9) plays important role in coronary artery disease (CAD). However, the exact relationship between them is not explored yet. Here, four tag SNPs covering IL9 (rs31563, rs2069868, rs2069870 and rs31564) were selected to conduct case-control association analyses in a total of 3704 individuals from Chinese Han population (1863 CAD vs 1841 control). Results showed that: first, rs2069868 was associated with CAD combined with hypertension (Padj = 0.027); second, IL9 haplotype (CGAT) was associated with CAD (Padj = 0.035), and the combination genotype of "rs31563_CC/rs31564_TT" would remarkably decrease the risk of CAD (Padj = 0.001); third, significant associations were found between rs2069870 and decreased LDL-c levels and decreased total cholesterol levels, and between rs31563 and increased HDL-c levels (Padj < 0.05). Therefore, we conclude that IL9 might play a causal role in CAD by interacted with CAD traditional risk factors, which might confer a new way to improve the prevention and treatment of CAD.
Collapse
Affiliation(s)
- Lingfeng Zha
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiangtao Dong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qianwen Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Maternal and Child Health Hospital, Wuhan 430070, China
| | - Yuhua Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongsong Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Tian Xie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tingting Tang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ni Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Min Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiao Jiao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yingchao Zhou
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jianfei Wu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangping Yang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qing K Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xin Tu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shaofang Nie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
12
|
De Luca M, Crisci G, Giardino F, Valente V, Amaranto I, Iacono O, D'Assante R, Giallauria F, Marra AM. Anabolic hormones and heart failure with preserved ejection fraction: looking for Ariadne's thread. Monaldi Arch Chest Dis 2021; 92. [PMID: 34351104 DOI: 10.4081/monaldi.2021.1743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/29/2021] [Indexed: 11/23/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex clinical syndrome that accounts for more than half of all heart failure patients. Identification, early diagnosis and management of patients are still complex, and no targeted treatment is available, since all tested drugs were not able to lower hard clinical outcomes. A multi-hormonal deficiency syndrome has been described in HFpEF patients suggesting that different hormones may represent new biomarkers of the disease, but their clinical utility is still debated. The natriuretic peptides are the cornerstone biomarker in heart failure, predicting cardiovascular death and heart failure hospitalization. Testosterone and DHEA-S deficiencies have been reported in HFpEF and associated with right ventricular impairment and diastolic dysfunction. IGFBP-1/IGF-1 axis correlates with echocardiographic parameters of HFpEF patients and with several prognostic biomarkers including NT-proBNP and C reactive protein. Low triiodothyronine syndrome is frequently found in HFpEF and thyroid hormones should represent a potential biomarker of risk stratification and prognosis.
Collapse
Affiliation(s)
| | - Giulia Crisci
- Department of Translational Medical Sciences, "Federico II" University, Naples.
| | - Federica Giardino
- Department of Translational Medical Sciences, "Federico II" University, Naples.
| | - Valeria Valente
- Department of Translational Medical Sciences, "Federico II" University, Naples.
| | - Ilaria Amaranto
- Department of Translational Medical Sciences, "Federico II" University, Naples.
| | - Olimpia Iacono
- Department of Translational Medical Sciences, "Federico II" University, Naples.
| | - Roberta D'Assante
- Department of Translational Medical Sciences, "Federico II" University, Naples.
| | | | - Alberto M Marra
- Department of Translational Medical Sciences, "Federico II" University, Naples.
| |
Collapse
|
13
|
Abstract
Exercise intolerance represents a typical feature of heart failure with preserved ejection fraction (HFpEF), and is associated with a poor quality of life, frequent hospitalizations, and increased all-cause mortality. The cardiopulmonary exercise test is the best method to quantify exercise intolerance, and allows detection of the main mechanism responsible for the exercise limitation, influencing treatment and prognosis. Exercise training programs improve exercise tolerance in HFpEF. However, studies are needed to identify appropriate type and duration. This article discusses the pathophysiology of exercise limitation in HFpEF, describes methods of determining exercise tolerance class, and evaluates prognostic implications and potential therapeutic strategies.
Collapse
|
14
|
Cittadini A, Salzano A, Iacoviello M, Triggiani V, Rengo G, Cacciatore F, Maiello C, Limongelli G, Masarone D, Perticone F, Cimellaro A, Perrone Filardi P, Paolillo S, Mancini A, Volterrani M, Vriz O, Castello R, Passantino A, Campo M, Modesti PA, De Giorgi A, Monte IP, Puzzo A, Ballotta A, D'Assante R, Arcopinto M, Gargiulo P, Sciacqua A, Bruzzese D, Colao A, Napoli R, Suzuki T, Eagle KA, Ventura HO, Marra AM, Bossone E. Multiple hormonal and metabolic deficiency syndrome predicts outcome in heart failure: the T.O.S.CA. Registry. Eur J Prev Cardiol 2021; 28:1691-1700. [PMID: 33693736 DOI: 10.1093/eurjpc/zwab020] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/04/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
AIMS Recent evidence supports the occurrence of multiple hormonal and metabolic deficiency syndrome (MHDS) in chronic heart failure (CHF). However, no large observational study has unequivocally demonstrated its impact on CHF progression and outcome. The T.O.S.CA. (Trattamento Ormonale nello Scompenso CArdiaco; Hormone Treatment in Heart Failure) Registry has been specifically designed to test the hypothesis that MHDS affects morbidity and mortality in CHF patients. METHODS AND RESULTS The T.O.S.CA. Registry is a prospective, multicentre, observational study involving 19 Italian centres. Thyroid hormones, insulin-like growth factor-1, total testosterone, dehydropianoandrosterone sulfate, insulin resistance, and the presence of diabetes were evaluated. A MHDS was defined as the presence of ≥2 hormone deficiencies (HDs). Primary endpoint was a composite of all-cause mortality and cardiovascular hospitalizations. Four hundred and eighty heart failure patients with ejection fraction ≤45% were enrolled. MHDS or diabetes was diagnosed in 372 patients (77.5%). A total of 271 events (97 deaths and 174 cardiovascular hospitalizations) were recorded, 41% in NO-MHDS and 62% in MHDS (P < 0.001). Median follow-up was of 36 months. MHDS was independently associated with the occurrence of the primary endpoint [hazard ratio 95% (confidence interval), 1.93 (1.37-2.73), P < 0.001] and identified a group of patients with a higher mortality [2.2 (1.28-3.83), P = 0.01], with a graded relation between HDs and cumulative events (P < 0.01). CONCLUSION MHDS is common in CHF and independently associated with increased all-cause mortality and cardiovascular hospitalization, representing a promising therapeutic target. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT023358017.
Collapse
Affiliation(s)
- Antonio Cittadini
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini, 5, 80131 Naples, Italy.,Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, Italy
| | - Andrea Salzano
- IRCCS SDN, Diagnostic and Nuclear Research Institute, Naples, Italy
| | - Massimo Iacoviello
- Cardiology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari 'A Moro', Bari, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini, 5, 80131 Naples, Italy.,Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB), IRCCS, Scientific Institute of Telese Terme, Telese BN, Italy
| | - Francesco Cacciatore
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Ciro Maiello
- Heart Transplantation Unit, Monaldi Hospital, Azienda Ospedaliera dei Colli, Naples, Italy
| | - Giuseppe Limongelli
- Division of Cardiology, Monaldi Hospital, Azienda Ospedaliera dei Colli, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Daniele Masarone
- Division of Cardiology, Monaldi Hospital, Azienda Ospedaliera dei Colli, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Antonio Cimellaro
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Pasquale Perrone Filardi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.,Mediterranea Cardiocentro, Naples, Italy
| | - Stefania Paolillo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.,Mediterranea Cardiocentro, Naples, Italy
| | - Antonio Mancini
- Operative Unit of Endocrinology, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Olga Vriz
- Heart Center Department, King Faisal Hospital & Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Roberto Castello
- Division of General Medicine, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | | | - Michela Campo
- Department of Medical and Surgical Sciences, Unit of Endocrinology and Metabolic Diseases, University of Foggia, Foggia, Italy
| | - Pietro A Modesti
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Florence, Italy
| | - Alfredo De Giorgi
- Department of Medical Sciences, School of Medicine, Pharmacy and Prevention, University of Ferrara, Ferrara, Italy
| | - Ines P Monte
- Department of General Surgery and Medical-Surgery Specialties, University of Catania, Catania, Italy
| | | | - Andrea Ballotta
- IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Roberta D'Assante
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini, 5, 80131 Naples, Italy.,Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, Italy
| | - Michele Arcopinto
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini, 5, 80131 Naples, Italy.,Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, Italy
| | - Paola Gargiulo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Dario Bruzzese
- Department of Public Health, Federico II University, Naples, Italy
| | - Annamaria Colao
- Clinical Medicine and Surgery Department, Federico II University, Naples, Italy
| | - Raffaele Napoli
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Toru Suzuki
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Kim A Eagle
- Michigan Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Hector O Ventura
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School, The University of Queensland School of Medicine, New Orleans, LA, USA
| | - Alberto M Marra
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini, 5, 80131 Naples, Italy.,Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, Italy.,Center for Pulmonary Hypertension, Thoraxclinic at Heidelberg University Hospital, Heidelberg, Germany
| | - Eduardo Bossone
- Italian Clinical Outcome Research and Reporting Program (I-CORRP), Naples, Italy.,Cardiology Division, A Cardarelli Hospital, Naples, Italy
| |
Collapse
|
15
|
Salzano A, D'Assante R, Israr MZ, Eltayeb M, D'Agostino A, Bernieh D, De Luca M, Rega S, Ranieri B, Mauro C, Bossone E, Squire IB, Suzuki T, Marra AM. Biomarkers in Heart Failure: Clinical Insights. Heart Fail Clin 2021; 17:223-243. [PMID: 33673947 DOI: 10.1016/j.hfc.2021.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Andrea Salzano
- IRCCS SDN Nuclear and Diagnostic Research Institute, Naples, Italy.
| | - Roberta D'Assante
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | | | - Mohamed Eltayeb
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Anna D'Agostino
- IRCCS SDN Nuclear and Diagnostic Research Institute, Naples, Italy
| | - Dennis Bernieh
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Mariarosaria De Luca
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Salvatore Rega
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Brigida Ranieri
- IRCCS SDN Nuclear and Diagnostic Research Institute, Naples, Italy
| | - Ciro Mauro
- AORN A Cardarelli, Cardiac Rehabilitation Unit, Naples, Italy
| | - Eduardo Bossone
- AORN A Cardarelli, Cardiac Rehabilitation Unit, Naples, Italy
| | - Iain B Squire
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Toru Suzuki
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Alberto M Marra
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
16
|
Ye D, Wang Z, Xu Y, Ye J, Wang M, Liu J, Zhang J, Zhao M, Chen J, Wan J. Interleukin-9 aggravates doxorubicin-induced cardiotoxicity by promoting inflammation and apoptosis in mice. Life Sci 2020; 255:117844. [PMID: 32464124 DOI: 10.1016/j.lfs.2020.117844] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 01/03/2023]
Abstract
AIMS Interleukin (IL) 9 is a pleiotropic cytokine, and recent studies have demonstrated that IL-9 is associated with several cardiovascular diseases, via regulation of the inflammatory response. Doxorubicin (DOX) is known to induce severe cardiac injury and dysfunction by enhancing inflammation. This study aimed to investigate the role of IL-9 in DOX-induced cardiotoxicity. MATERIALS AND METHODS DOX was used to induce cardiac dysfunction and the expression of IL-9 in the murine cardiac tissues was measured. The mice were intraperitoneally injected with recombinant mouse IL-9 (rmIL-9) or anti-IL-9 neutralizing antibody (IL-9nAb) for investigating the effect of IL-9 on DOX-induced cardiac injury and dysfunction. The messenger ribonucleic acid (mRNA) expression levels of the pro-inflammatory cytokines were determined in each group by quantitative real-time polymerase chain reaction (RT-qPCR). The effect of rmIL-9 or IL-9nAb on DOX-induced apoptosis was determined both in vivo and vitro. KEY FINDINGS IL-9 levels significantly increased in the heart following DOX injection. Cardiac injury and dysfunction were induced by DOX, and treatment with IL-9nAb significantly alleviated DOX-induced injury, whereas rmIL-9 administration aggravated the cardiac damage. IL-9nAb decreased the expression of pro-inflammatory cytokines in the DOX-treated mice, while rmIL-9 administration increased the levels of pro-inflammatory cytokines. IL-9nAb reduced DOX-induced myocardial apoptosis, whereas rmIL-9 administration produced the opposite results. Additionally, IL-9nAb mitigated the DOX-induced apoptosis in H9C2 cells, while administration of rmIL-9 produced the opposite effect. SIGNIFICANCE Our results demonstrated that IL-9 aggravated DOX-induced cardiac injury and dysfunction by promoting the inflammatory response and cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jiangbin Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
17
|
Abstract
Heart failure (HF) is the leading cause of morbidity and mortality in developed countries, and it is the primary cause of mortality in the elderly worldwide. The processes of inflammatory response activation, production and release of pro-inflammatory cytokines, activation of the complement system, synthesis of autoantibodies, and overexpression of Class II major histocompatibility complex molecules contribute to the HF development and progression. High levels of circulating cytokines correlate with the severity of HF, measured with the use of New York Heart Association's classification, and prognosis of the disease. In HF, there is an imbalance between pro-inflammatory and anti-inflammatory cytokines. Concentrations of several interleukins are increased in HF, including IL-1β, IL-6, IL-8, IL-9, IL-10, IL-13, IL-17, and IL-18, whereas the levels of IL-5, IL-7, or IL-33 are down-regulated. Concentrations of inflammatory mediators are associated with cardiac function and can be HF markers and predictors of adverse outcomes or mortality. This review presents the role of interleukins, which contribute to the HF initiation and progression, the importance of their pathways in transition from myocardial injury to HF, and the role of interleukins as markers of disease severity and outcome predictors.
Collapse
|
18
|
Yang Y, Xu C, Tang S, Xia Z. Interleukin-9 Aggravates Isoproterenol-Induced Heart Failure by Activating Signal Transducer and Activator of Transcription 3 Signalling. Can J Cardiol 2020; 36:1770-1781. [PMID: 32621886 DOI: 10.1016/j.cjca.2020.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/18/2019] [Accepted: 01/08/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Previous studies have demonstrated that inflammation is closely related to the occurrence and development of heart failure (HF). As an inflammation-related cytokine, interleukin (IL)-9 has been reported to be involved in the development of cardiovascular diseases. However, the role of IL-9 in HF in response to isoproterenol (ISO) stimulation has barely been explored. Thus, this study aimed to investigate whether IL-9 participates in HF and the possible associated mechanisms. METHODS Chronic ISO infusion was used to establish an HF model, and the IL-9 levels in mice and isolated cardiomyocytes were measured. In addition, ISO-treated mice received an injection of recombinant mouse IL-9 (rIL-9) or an antimouse IL-9 neutralizing monoclonal antibody (mAb) to investigate the effects of IL-9 on cardiac function, hypertrophy, and fibrosis. RESULTS IL-9 levels were significantly increased in mice and isolated cardiomyocytes after ISO treatment. Treatment with rIL-9 resulted in aggravated cardiac dysfunction and amplified cardiac hypertrophy and fibrosis, whereas treatment with the anti-IL-9 neutralizing mAb ameliorated cardiac dysfunction and reduced cardiac hypertrophy and fibrosis in ISO-treated mice. In addition, ISO infusion-induced cardiac inflammation and cardiomyocyte apoptosis was aggravated by rIL-9 but prevented by the anti-IL-9 mAb. IL-9 did not activate signal transducer and activator of transcription (STAT)1 or STAT5 but induced STAT3 phosphorylation in ISO-induced HF. Moreover, S31-201, a specific STAT3 inhibitor, nearly abolished rIL-9-induced increases in cardiac dysfunction, hypertrophy, and fibrosis in response to ISO stimulation. CONCLUSIONS IL-9 aggravated cardiac dysfunction and amplified cardiac hypertrophy and fibrosis in the ISO-induced HF model by activating STAT3 signalling. These data indicate that blocking IL-9 may be an attractive pharmacotherapeutic strategy for the treatment of cardiac hypertrophy and fibrosis induced by chronic β-adrenergic receptor activation to limit the progression of HF.
Collapse
Affiliation(s)
- Yunzhao Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cheng Xu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shaoqun Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
19
|
|
20
|
Salzano A, Cassambai S, Yazaki Y, Israr MZ, Bernieh D, Wong M, Suzuki T. The Gut Axis Involvement in Heart Failure. Heart Fail Clin 2020; 16:23-31. [DOI: 10.1016/j.hfc.2019.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
21
|
Huang J, Xiang Y, Zhang H, Wu N, Chen X, Wu L, Xu B, Li C, Zhang Z, Tong S, Zhong L, Li Y. Plasma Level of Interferon-γ Predicts the Prognosis in Patients With New-Onset Atrial Fibrillation. Heart Lung Circ 2019; 29:e168-e176. [PMID: 31813744 DOI: 10.1016/j.hlc.2019.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/25/2019] [Accepted: 11/02/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Patients with atrial fibrillation are at increased risk of stroke and mortality. It is not clear if inflammatory biomarkers are associated with stroke and mortality in patients with atrial fibrillation. We aimed to evaluate the predictive value of three inflammatory biomarkers (interleukin [IL]-9, IL-10, and interferon [IFN]-γ) for stroke and mortality in atrial fibrillation. METHOD A total of 232 patients with new-onset atrial fibrillation were enrolled and 217 patients were completely followed-up. Peripheral plasma concentrations of cytokines (IL-9, IL-10, and IFN-γ) were measured using Luminex xMAP assays. The association between dichotomous groups of cytokines and outcomes were evaluated by a Cox proportional hazards model. The incremental value of inflammatory biomarkers, in addition to the CHA2DS2-VASc score, was also assessed. RESULTS Patients were followed-up for a median duration of 27 (interquartile range [IQR], 23-30) months. The elevated plasma level of IFN-γ was an independent risk factor for stroke (hazard ratio [HR], 4.02 [IQR, 1.06-15.34]; p=0.042) and all-cause mortality (HR, 3.93 [IQR, 1.43-10.78]; p=0.008) in patients with atrial fibrillation. Adding high IFN-γ to the CHA2DS2-VASc score showed improvement in discrimination and reclassification prediction for stroke and mortality. However, IL-9 and IL-10 had no statistically significant association with stroke and all-cause mortality in patients with atrial fibrillation. CONCLUSIONS In this "real-world" cohort of patients with atrial fibrillation, we have shown for the first time that plasma levels of IFN-γ could provide incremental prognostic value supplementary to that obtained from the CHA2DS2-VASc scores for predicting of stroke and all-cause mortality.
Collapse
Affiliation(s)
- Jiaqi Huang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China; Evidence-based Medicine and Clinical Epidemiology Center, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Ying Xiang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China; Evidence-based Medicine and Clinical Epidemiology Center, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Huan Zhang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China; Evidence-based Medicine and Clinical Epidemiology Center, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Na Wu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China; Evidence-based Medicine and Clinical Epidemiology Center, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Xinghua Chen
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Long Wu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China; Evidence-based Medicine and Clinical Epidemiology Center, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Bin Xu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China; Evidence-based Medicine and Clinical Epidemiology Center, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Chengying Li
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China; Evidence-based Medicine and Clinical Epidemiology Center, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Zhihui Zhang
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Shifei Tong
- Cardiovascular Disease Center, Third Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Li Zhong
- Cardiovascular Disease Center, Third Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yafei Li
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China; Evidence-based Medicine and Clinical Epidemiology Center, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China.
| |
Collapse
|
22
|
Salzano A, D'Assante R, Lander M, Arcopinto M, Bossone E, Suzuki T, Cittadini A. Hormonal Replacement Therapy in Heart Failure: Focus on Growth Hormone and Testosterone. Heart Fail Clin 2019; 15:377-391. [PMID: 31079696 DOI: 10.1016/j.hfc.2019.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A growing body of evidence led to the hypothesis that heart failure (HF) could be considered a multiple hormone deficiency syndrome. Deficiencies in the main anabolic axes cannot be considered as mere epiphenomena, are very common in HF, and are clearly associated with poor cardiovascular performance and outcomes. Growth hormone deficiency and testosterone deficiency play a pivotal role and the replacement treatment is an innovative therapy that should be considered. This article appraises the current evidence regarding growth hormone and testosterone deficiencies in HF and reviews novel findings about the treatment of these conditions in HF.
Collapse
Affiliation(s)
- Andrea Salzano
- Department of Cardiovascular Sciences, NIHR Leicester Biomedical Research Centre, University of Leicester, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK; Department of Translational Medical Sciences, Federico II University, Via Pansini 5, Naples 80138, Italy
| | | | - Mark Lander
- Department of Acute Medicine, University College London Hospitals NHS Foundation Trust, 235 Euston Road, London NW1 2BU, UK
| | - Michele Arcopinto
- Department of Translational Medical Sciences, Federico II University, Via Pansini 5, Naples 80138, Italy; Emergency Department, A Cardarelli Hospital, Via Cardarelli 9, Naples 80131, Italy
| | - Eduardo Bossone
- Cardiology Division, A Cardarelli Hospital, Via Cardarelli 9, Naples 80131, Italy
| | - Toru Suzuki
- Department of Cardiovascular Sciences, NIHR Leicester Biomedical Research Centre, University of Leicester, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Antonio Cittadini
- Department of Translational Medical Sciences, Federico II University, Via Pansini 5, Naples 80138, Italy; Interdisciplinary Research Centre in Biomedical Materials (CRIB), Piazzale Tecchio 80, Naples 80125, Italy.
| |
Collapse
|
23
|
Salzano A, Marra AM, D’Assante R, Arcopinto M, Bossone E, Suzuki T, Cittadini A. Biomarkers and Imaging. Heart Fail Clin 2019; 15:321-331. [DOI: 10.1016/j.hfc.2018.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
24
|
Biomarkers in Heart Failure and Associated Diseases. DISEASE MARKERS 2019; 2019:8768624. [PMID: 30881524 PMCID: PMC6383412 DOI: 10.1155/2019/8768624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 12/20/2022]
|
25
|
Salzano A, Marra AM, D’Assante R, Arcopinto M, Suzuki T, Bossone E, Cittadini A. Growth Hormone Therapy in Heart Failure. Heart Fail Clin 2018; 14:501-515. [DOI: 10.1016/j.hfc.2018.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
26
|
Bossone E, Arcopinto M, Iacoviello M, Triggiani V, Cacciatore F, Maiello C, Limongelli G, Masarone D, Perticone F, Sciacqua A, Perrone-Filardi P, Mancini A, Volterrani M, Vriz O, Castello R, Passantino A, Campo M, Modesti PA, De Giorgi A, Monte I, Puzzo A, Ballotta A, Caliendo L, D'Assante R, Marra AM, Salzano A, Suzuki T, Cittadini A. Multiple hormonal and metabolic deficiency syndrome in chronic heart failure: rationale, design, and demographic characteristics of the T.O.S.CA. Registry. Intern Emerg Med 2018; 13:661-671. [PMID: 29619769 DOI: 10.1007/s11739-018-1844-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/24/2018] [Indexed: 12/20/2022]
Abstract
Recent evidence supports the concept that progression of chronic heart failure (CHF) depends upon an imbalance of catabolic forces over the anabolic drive. In this regard, multiple hormonal deficiency syndrome (MHDS) significantly has impacts upon CHF progression, and is associated with a worse clinical status and increased mortality. The T.O.S.CA. (Trattamento Ormonale nello Scompenso CArdiaco; Hormone Therapy in Heart Failure) Registry (clinicaltrial.gov = NCT02335801) tests the hypothesis that anabolic deficiencies reduce survival in a large population of mild-to-moderate CHF patients. The T.O.S.CA. Registry is a prospective multicenter observational study coordinated by "Federico II" University of Naples, and involves 19 centers situated throughout Italy. Thyroid hormones, insulin-like growth factor-1, total testosterone, dehydroepiandrosterone , and insulin are measured at baseline and every year for a patient-average follow-up of 3 years. Subjects with CHF are divided into two groups: patients with one or no anabolic deficiency, and patients with two or more anabolic deficiencies at baseline. The primary endpoint is the composite of all-cause mortality and cardiovascular hospitalization. Secondary endpoints include the composite of all-cause mortality and hospitalization, the composite of cardiovascular mortality and cardiovascular hospitalization, and change of VO2 peak. Patient enrollment started in April 2013, and was completed in July 2017. Demographics and main clinical characteristics of enrolled patients are provided in this article. Detailed cross-sectional results will be available in late 2018. The T.O.S.CA. Registry represents the most robust prospective observational trial on MHDS in the field of CHF. The study findings will advance our knowledge with regard to the intimate mechanisms of CHF progression and hopefully pave the way for future randomized clinical trials of single or multiple hormonal replacement therapies in CHF.
Collapse
Affiliation(s)
- E Bossone
- Heart Department, Cardiology Division, "Cava de' Tirreni and Amalfi Coast" Hospital, University of Salerno, Salerno, Italy
| | - M Arcopinto
- Department of Translational Medical Sciences, Federico II University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - M Iacoviello
- Cardiology Unit, Cardiothoracic Department, University of Bari "Aldo Moro", Bari, Italy
| | - V Triggiani
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "A. Moro", Bari, Italy
| | - F Cacciatore
- Heart Transplantation Unit, Monaldi Hospital, Azienda Ospedaliera dei Colli, Naples, Italy
| | - C Maiello
- Heart Transplantation Unit, Monaldi Hospital, Azienda Ospedaliera dei Colli, Naples, Italy
| | - G Limongelli
- Division of Cardiology SUN, Monaldi Hospital, Azienda Ospedaliera dei Colli, Second University of Naples, Naples, Italy
| | - D Masarone
- Division of Cardiology SUN, Monaldi Hospital, Azienda Ospedaliera dei Colli, Second University of Naples, Naples, Italy
| | - F Perticone
- Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - A Sciacqua
- Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - P Perrone-Filardi
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - A Mancini
- Operative Unit of Endocrinology, Catholic University of the Sacred Heart, Rome, Italy
| | - M Volterrani
- Department of Medical Sciences, IRCCS San Raffaele Pisana, Rome, Italy
| | - O Vriz
- Heart Center Department, King Faisal Hospital & Research Center, Riyadh, Kingdom of Saudi Arabia
| | - R Castello
- Division of General Medicine, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - A Passantino
- Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Cassano Murge, Bari, Italy
| | - M Campo
- Unit of Endocrinology and Metabolic Diseases, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - P A Modesti
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Florence, Italy
| | - A De Giorgi
- Department of Medical Sciences, School of Medicine, Pharmacy and Prevention, University of Ferrara, Ferrara, Italy
| | - I Monte
- Department of General Surgery and Medical-Surgery Specialties, University of Catania, Catania, Italy
| | - A Puzzo
- IRCSS. Oasi Maria SS, Troina, Italy
| | - A Ballotta
- IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - L Caliendo
- Ospedale Santa Maria della Pietà, Nola, Naples, Italy
| | | | | | - A Salzano
- Department of Translational Medical Sciences, Federico II University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Glenfield Hospital, Leicester, UK
| | - T Suzuki
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Glenfield Hospital, Leicester, UK
| | - A Cittadini
- Department of Translational Medical Sciences, Federico II University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy.
| |
Collapse
|
27
|
Marra AM, Bobbio E, D'Assante R, Salzano A, Arcopinto M, Bossone E, Cittadini A. Growth Hormone as Biomarker in Heart Failure. Heart Fail Clin 2018; 14:65-74. [PMID: 29153202 DOI: 10.1016/j.hfc.2017.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The impairment of growth hormone (GH)/insulin growth factor-1(IGF-1) plays a crucial role in chronic heart failure (CHF). Several studies have shown that patients affected by this condition display a more aggressive disease, with impaired functional capacity and poor outcomes. Interestingly, GH replacement therapy represents a possible future therapeutic option in CHF. In this review, the authors focus on the assessment of the main abnormalities in GH/IGF-1 axis in CHF, the underlying molecular background, and their impact on disease progression and outcomes.
Collapse
Affiliation(s)
| | - Emanuele Bobbio
- Department of Translational Medical Sciences, Federico II University, Via Pansini, 5, 80131 Naples, Italy
| | | | - Andrea Salzano
- Department of Translational Medical Sciences, Federico II University, Via Pansini, 5, 80131 Naples, Italy; Department of Cardiovascular Sciences and NIHR Biomedical Research Centre, University of Leicester, Glenfield Hospital, Groby Road LE3 9QP, Leicester, UK
| | - Michele Arcopinto
- Department of Translational Medical Sciences, Federico II University, Via Pansini, 5, 80131 Naples, Italy
| | - Eduardo Bossone
- Heart Department, University Hospital Salerno, Via Enrico de Marinis, 84013 Cava de' Tirreni SA, Italy
| | - Antonio Cittadini
- Department of Translational Medical Sciences, Federico II University, Via Pansini, 5, 80131 Naples, Italy; Interdisciplinary Research Centre in Biomedical Materials (CRIB), Via Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|