1
|
Gersch S, Bengel P, Paul NB, Ravassa S, von Haehling S, Fischer A, Zeisberg EM, Puls M, Hasenfuß G, Schnelle M. Estimating myocardial fibrosis in aortic stenosis using the serum collagen type I C-terminal telopeptide to matrix metalloproteinase-1 ratio. MedComm (Beijing) 2025; 6:e70069. [PMID: 39830018 PMCID: PMC11739461 DOI: 10.1002/mco2.70069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/23/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Affiliation(s)
- Svante Gersch
- Department of Clinical ChemistryDepartment of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Philipp Bengel
- Department of Clinical ChemistryDepartment of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Niels B. Paul
- Department of Clinical ChemistryDepartment of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Susana Ravassa
- Program of Cardiovascular DiseaseCentro de Investigacion Medica Aplicada Universidad de Navarra (CIMA)PamplonaSpain
| | - Stephan von Haehling
- Department of Clinical ChemistryDepartment of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Andreas Fischer
- Department of Clinical ChemistryDepartment of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Elisabeth M. Zeisberg
- Department of Clinical ChemistryDepartment of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Miriam Puls
- Department of Clinical ChemistryDepartment of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Gerd Hasenfuß
- Department of Clinical ChemistryDepartment of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Moritz Schnelle
- Department of Clinical ChemistryDepartment of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| |
Collapse
|
2
|
Mouzarou A, Hadjigeorgiou N, Melanarkiti D, Plakomyti TE. The Role of NT-proBNP Levels in the Diagnosis of Hypertensive Heart Disease. Diagnostics (Basel) 2025; 15:113. [PMID: 39795641 PMCID: PMC11719755 DOI: 10.3390/diagnostics15010113] [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: 11/18/2024] [Revised: 12/29/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
Hypertension is a major risk factor of various cardiac complications, including hypertensive heart disease (HHD). This condition can lead to a number of structural and functional changes in the heart, such as left ventricular hypertrophy, diastolic dysfunction, and, eventually, systolic dysfunction. In the management of hypertensive heart disease, early diagnosis and appropriate treatment are crucial for preventing the progression to congestive heart failure. One potential diagnostic marker that has gained attention in recent years is the N-terminal pro-brain natriuretic peptide (NT-proBNP). The natriuretic peptides, including the brain natriuretic peptide (BNP) and its inactive N-terminal fragment, are secreted by the myocardium in response to increased wall stress and volume overload. In patients with hypertensive heart disease, increased NT-proBNP levels may reflect the structural and functional changes occurring in the myocardium as a result of chronic pressure overload. Several studies have investigated the diagnostic utility of NT-proBNP in hypertensive heart disease. NT-proBNP levels can be a useful adjunct in the diagnosis of hypertensive heart disease, particularly in the assessment of diastolic dysfunction and left ventricular hypertrophy. This review paper explores the role of NT-proBNP levels in the diagnosis of hypertensive heart disease.
Collapse
Affiliation(s)
- Angeliki Mouzarou
- Department of Cardiology, Paphos General Hospital, State Health Organization Services, Paphos 8026, Cyprus
| | | | | | | |
Collapse
|
3
|
Zhang Y, Lu F. Molecular mechanism of triptolide in myocardial fibrosis through the Wnt/β-catenin signaling pathway. SCAND CARDIOVASC J 2024; 58:2295785. [PMID: 38164796 DOI: 10.1080/14017431.2023.2295785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/24/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Objective. Myocardial fibrosis (MF) is a common manifestation of end-stage cardiovascular diseases. Triptolide (TP) provides protection against cardiovascular diseases. This study was to explore the functional mechanism of TP in MF rats via the Wnt/β-catenin pathway. Methods. The MF rat model was established via subcutaneous injection of isoproterenol (ISO) and treated with low/medium/high doses of TP (L-TP/M-TP/H-TP) or Wnt agonist BML-284. Cardiac function was examined by echocardiography. Pathological changes of myocardial tissues were observed by HE and Masson staining. Col-I/Col-III/Vimentin/α-SMA levels were detected by immunohistochemistry, RT-qPCR, and Western blot. Collagen volume fraction content was measured. Expression levels of the Wnt/β-catenin pathway-related proteins (β-catenin/c-myc/Cyclin D1) were detected by Western blot. Rat cardiac fibroblasts were utilized for in vitro validation experiments. Results. MF rats had enlarged left ventricle, decreased systolic and diastolic function and cardiac dysfunction, elevated collagen fiber distribution, collagen volume fraction and hydroxyproline content. Levels of Col-I/Col-III/Vimentin/α-SMA, and protein levels of β-catenin/c-myc/Cyclin D1 were increased in MF rats. The Wnt/β-catenin pathway was activated in the myocardial tissues of MF rats. TP treatment alleviated impairments of cardiac function and myocardial tissuepathological injury, decreased collagen fibers, collagen volume fraction, Col-I, Col-III, α-SMA and Vimentin levels, HYP content, inhibited Wnt/β-catenin pathway, with H-TP showing the most significant effects. Wnt agonist BML-284 antagonized the inhibitive effect of TP on MF. TP inhibited the Wnt/β-catenin pathway to repress the proliferation and differentiation of mouse cardiac fibroblasts in vitro. Conclusions. TP was found to ameliorate ISO-induced MF in rats by inhibiting the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yiwen Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Feng Lu
- Cardiovascular Internal Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
4
|
Rajah MR, Doubell AF, Herbst PG. Quantification of Replacement Fibrosis in Aortic Stenosis: A Narrative Review on the Utility of Cardiovascular Magnetic Resonance Imaging. Diagnostics (Basel) 2024; 14:2435. [PMID: 39518402 PMCID: PMC11544846 DOI: 10.3390/diagnostics14212435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Aortic stenosis (AS) is associated with the development of replacement myocardial fibrosis/scar. Given the dose-dependent relationship between scar and clinical outcomes after aortic valve replacement (AVR) surgery, scar quantity may serve as an important risk-stratification tool to aid decision-making on the optimal timing of AVR. Scar is non-invasively assessed and quantified by cardiovascular magnetic resonance (CMR) imaging. Several quantification techniques exist, and consensus on the optimal technique is lacking. These techniques range from a visual manual method to fully automated ones. This review describes the different scar quantification techniques used and highlights their strengths and shortfalls within the context of AS. The two most commonly used techniques in AS include the semi-automated signal threshold versus reference mean (STRM) and full-width half-maximum (FWHM) techniques. The accuracy and reproducibility of these techniques may be hindered in AS by the coexistence of diffuse interstitial fibrosis and the presence of relatively small, non-bright scars. The validation of these techniques against histology, which is the current gold standard for scar quantification in AS, is limited. Based on the best current evidence, the STRM method using a threshold of three standard deviations above the mean signal intensity of remote myocardium is recommended. The high reproducibility of the FWHM technique in non-AS cohorts has been shown and merits further evaluation within the context of AS. Future directions include the use of quantitative T1 mapping for the detection and quantification of scar, as well as the development of serum biomarkers that reflect the fibrotic status of the myocardium in AS.
Collapse
Affiliation(s)
- Megan R. Rajah
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town 7505, South Africa
| | | | | |
Collapse
|
5
|
Huang J, Shi Z, Huang Z, Lai S. Identification and Verification of Potential Markers Related to Myocardial Fibrosis by Bioinformatics Analysis. Biochem Genet 2024:10.1007/s10528-024-10937-9. [PMID: 39387979 DOI: 10.1007/s10528-024-10937-9] [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: 10/07/2023] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Mounting evidence indicates that myocardial fibrosis (MF) is frequently intertwined with immune and metabolic disorders. This comprehensive review aims to delve deeply into the crucial role of immune-related signature genes in the pathogenesis and progression of MF. This exploration holds significant importance as understanding the underlying mechanisms of MF is essential for developing effective diagnostic and therapeutic strategies. The dataset GSE9735 about myocardial fibrosis and non-fibrosis was downloaded from GEO database. Differentially expressed genes (DEGs) were identified by 'limma' package in R software. Then, the biological function of DEG was determined by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. XCell was used to estimate the composition pattern of matrix and immune cells. Protein-protein interaction (PPI) network was constructed based on STRING analysis software, and Hub genes were screened and functional modules were analyzed. The correlation between hub genes and immune cell subtypes was analyzed. Hub genes with |correlation coefficient|> 0.45 and p-value < 0.05 were used as characteristic biomarkers. Finally, the logistic regression model is used to verify the three markers in the training set and verification set (GSE97358 and GSE225336). A total of 635 DEGs were identified. Functional enrichment analysis shows that inflammation and immune response, extracellular matrix and structural remodeling play an important role in the pathological mechanism of MF. Immune cell infiltration analysis showed that immune cells (Plasma cells, Eosinophils, Chondrocytes and Th2 cells) significantly changed in MF pathological conditions. In PPI network analysis, IL1β, TTN, PTPRC, IGF1, ALDH1A1, CYP26A1, ALDH1A3, MYH11, CSF1R and CD80 were identified as hub genes, among which IL1β, CYP26A1 and GNG2 were regarded as immune-related characteristic markers. The AUC scores of the three biomarkers are all above 0.65, which proves that they have a good discrimination effect in MF. In this study, three immune-related genes were identified as diagnostic biomarkers of MF, which provided a new perspective for exploring the molecular mechanism of MF. This study takes a comprehensive approach to understanding the intricate relationship between myocardial fibrosis and immune metabolism. By identifying key immune-related biomarkers, this study not only reveals the molecular basis of myocardial fibrosis but also paves the way for the development of novel diagnostic tools and therapeutic strategies. These findings are critical for improving patient prognosis and may have broader implications for studying and treating other cardiovascular diseases associated with immune dysregulation.
Collapse
Affiliation(s)
- Jiazhuo Huang
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China
| | - Zhentao Shi
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China
| | - Zhifeng Huang
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China
| | - Shaobin Lai
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China.
| |
Collapse
|
6
|
Chaher N, Lacerda S, Digilio G, Padovan S, Gao L, Lavin B, Stefania R, Velasco C, Cruz G, Prieto C, Botnar RM, Phinikaridou A. Non-invasive in vivo imaging of changes in Collagen III turnover in myocardial fibrosis. NPJ IMAGING 2024; 2:33. [PMID: 39301014 PMCID: PMC11408249 DOI: 10.1038/s44303-024-00037-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/31/2024] [Indexed: 09/22/2024]
Abstract
Heart failure (HF) affects 64 million people globally with enormous societal and healthcare costs. Myocardial fibrosis, characterised by changes in collagen content drives HF. Despite evidence that collagen type III (COL3) content changes during myocardial fibrosis, in vivo imaging of COL3 has not been achieved. Here, we discovered the first imaging probe that binds to COL3 with high affinity and specificity, by screening candidate peptide-based probes. Characterisation of the probe showed favourable magnetic and biodistribution properties. The probe's potential for in vivo molecular cardiac magnetic resonance imaging was evaluated in a murine model of myocardial infarction. Using the new probe, we were able to map and quantify, previously undetectable, spatiotemporal changes in COL3 after myocardial infarction and monitor response to treatment. This innovative probe provides a promising tool to non-invasively study the unexplored roles of COL3 in cardiac fibrosis and other cardiovascular conditions marked by changes in COL3.
Collapse
Affiliation(s)
- Nadia Chaher
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor, Lambeth Wing, St Thomas' Hospital, London, SE17EH UK
| | - Sara Lacerda
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d'Orléans rue Charles Sadron, 45071 Orléans, France
| | - Giuseppe Digilio
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Alessandria, Italy
| | - Sergio Padovan
- Institute for Biostructures and Bioimages (CNR), Molecular Biotechnology Center, Torino, Italy
| | - Ling Gao
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor, Lambeth Wing, St Thomas' Hospital, London, SE17EH UK
| | - Begoña Lavin
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor, Lambeth Wing, St Thomas' Hospital, London, SE17EH UK
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Rachele Stefania
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Alessandria, Italy
| | - Carlos Velasco
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor, Lambeth Wing, St Thomas' Hospital, London, SE17EH UK
| | - Gastão Cruz
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor, Lambeth Wing, St Thomas' Hospital, London, SE17EH UK
- Department of Radiology, University of Michigan, Ann Arbor, MI USA
| | - Claudia Prieto
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor, Lambeth Wing, St Thomas' Hospital, London, SE17EH UK
- Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - René M Botnar
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor, Lambeth Wing, St Thomas' Hospital, London, SE17EH UK
- Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
- King's BHF Centre of Excellence, Cardiovascular Division, London, UK
- Instituto de Ingeniería Biológica y Médica, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alkystis Phinikaridou
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor, Lambeth Wing, St Thomas' Hospital, London, SE17EH UK
- King's BHF Centre of Excellence, Cardiovascular Division, London, UK
| |
Collapse
|
7
|
Kayvanpour E, Sedaghat-Hamedani F, Li DT, Miersch T, Weis T, Hoefer I, Frey N, Meder B. Prognostic Value of Circulating Fibrosis Biomarkers in Dilated Cardiomyopathy (DCM): Insights into Clinical Outcomes. Biomolecules 2024; 14:1137. [PMID: 39334904 PMCID: PMC11430616 DOI: 10.3390/biom14091137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 08/28/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) involves myocardial remodeling, characterized by significant fibrosis and extracellular matrix expansion. These changes impair heart function, increasing the risk of heart failure and sudden cardiac death. This study investigates the prognostic value of circulating fibrosis biomarkers as a less invasive method in DCM patients. METHODS Plasma samples from 185 patients with confirmed DCM were analyzed to measure 13 circulating biomarkers using Luminex bead-based multiplex assays and ELISA. The prognostic value of these biomarkers was evaluated concerning heart failure-associated events and all-cause mortality. RESULTS Elevated MMP-2 levels (>1519.3 ng/mL) were linked to older age, higher diabetes prevalence, lower HDL, increased NT-proBNP and hs-TnT levels, and severe systolic dysfunction. High TIMP-1 levels (>124.9 ng/mL) correlated with elevated NT-proBNP, more atrial fibrillation, reduced exercise capacity, and larger right ventricles. Increased GDF-15 levels (>1213.9 ng/mL) were associated with older age, systemic inflammation, renal impairment, and poor exercise performance. Elevated OPN levels (>81.7 ng/mL) were linked to higher serum creatinine and NT-proBNP levels. Over a median follow-up of 32.4 months, higher levels of these biomarkers predicted worse outcomes, including increased risks of heart failure-related events and mortality. CONCLUSIONS Circulating fibrosis biomarkers, particularly MMP-2, TIMP-1, GDF-15, and OPN, are valuable prognostic tools in DCM. They reflect the severity of myocardial remodeling and systemic disease burden, aiding in risk stratification and therapeutic intervention. Integrating these biomarkers into clinical practice could improve DCM management and patient prognosis.
Collapse
Affiliation(s)
- Elham Kayvanpour
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), 69120 Heidelberg, Germany
| | - Farbod Sedaghat-Hamedani
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), 69120 Heidelberg, Germany
| | - Daniel Tian Li
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), 69120 Heidelberg, Germany
| | - Tobias Miersch
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Tanja Weis
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), 69120 Heidelberg, Germany
| | - Imo Hoefer
- Experimental Cardiology Laboratory, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Norbert Frey
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), 69120 Heidelberg, Germany
| | - Benjamin Meder
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), 69120 Heidelberg, Germany
- Klaus Tschira Institute for Computational Cardiology, 69120 Heidelberg, Germany
| |
Collapse
|
8
|
Tanisha, Amudha C, Raake M, Samuel D, Aggarwal S, Bashir ZMD, Marole KK, Maryam I, Nazir Z. Diagnostic Modalities in Heart Failure: A Narrative Review. Cureus 2024; 16:e67432. [PMID: 39314559 PMCID: PMC11417415 DOI: 10.7759/cureus.67432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Heart failure (HF) can present acutely or progress over time. It can lead to morbidity and mortality affecting 6.5 million Americans over the age of 20. The HF type is described according to the ejection fraction classification, defined as the percentage of blood volume that exits the left ventricle after myocardial contraction, undergoing ejection into the circulation, also called stroke volume, and is proportional to the ejection fraction. Cardiac catheterization is an invasive procedure to evaluate coronary artery disease leading to HF. Several biomarkers are being studied that could lead to early detection of HF and better symptom management. Testing for various biomarkers in the patient's blood is instrumental in confirming the diagnosis and elucidating the etiology of HF. There are various biomarkers elevated in response to increased myocardial stress and volume overload, including B-type natriuretic peptide (BNP) and its N-terminal prohormone BNP. We explored online libraries such as PubMed, Google Scholar, and Cochrane to find relevant articles. Our narrative review aims to extensively shed light on diagnostic modalities and novel techniques for diagnosing HF.
Collapse
Affiliation(s)
- Tanisha
- Department of Internal Medicine No. 4, O.O. Bogomolets National Medical University, Kyiv, UKR
| | - Chaithanya Amudha
- Department of Medicine and Surgery, Saveetha Medical College and Hospital, Chennai, IND
| | - Mohammed Raake
- Department of Surgery, Annamalai University, Chennai, IND
| | - Dany Samuel
- Department of Radiology, Medical University of Varna, Varna, BGR
| | | | - Zainab M Din Bashir
- Department of Medicine and Surgery, Combined Military Hospital (CMH) Lahore Medical College and Institute of Dentistry, Lahore, PAK
| | - Karabo K Marole
- Department of Medicine and Surgery, St. George's University School of Medicine, St. George's, GRD
| | - Iqra Maryam
- Department of Radiology, Allama Iqbal Medical College, Lahore, PAK
| | - Zahra Nazir
- Department of Internal Medicine, Combined Military Hospital, Quetta, PAK
| |
Collapse
|
9
|
Bacmeister L, Cavus E, Bohnen S, Tahir E, Wolf H, Buellesbach A, Heidenreich A, Haacke VK, Weber S, Hilgendorf I, Zeller T, Ojeda F, Radunski UK, Lund GK, Adam G, Blankenberg S, Westermann D, Muellerleile K, Lindner D. Serum Concentrations of Matrix Metalloproteinase-1 and Procollagen Type I Carboxy Terminal Propeptide Discriminate Infarct-Like Myocarditis and Non-ST-Segment-Elevation Myocardial Infarction. J Am Heart Assoc 2024; 13:e034194. [PMID: 38989835 PMCID: PMC11292779 DOI: 10.1161/jaha.124.034194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/24/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Biomarkers simplifying the diagnostic workup by discriminating between non-ST-segment-elevation myocardial infarction (NSTEMI) and infarct-like myocarditis are an unmet clinical need. METHODS AND RESULTS A total of 105 subjects were categorized into groups as follows: ST-segment-elevation myocardial infarction (n=36), NSTEMI (n=22), infarct-like myocarditis (n=19), cardiomyopathy-like myocarditis (n=18), and healthy control (n=10). All subjects underwent cardiac magnetic resonance imaging, and serum concentrations of matrix metalloproteinase-1 (MMP-1) and procollagen type I carboxy terminal propeptide (PICP) were measured. Biomarker concentrations in subjects presenting with acute coronary syndrome and non-ST-segment-elevation, for example NSTEMI or infarct-like myocarditis, categorized as the non-ST-segment-elevation acute coronary syndrome-like cohort, were of particular interest for this study. Compared with healthy controls, subjects with myocarditis had higher serum concentrations of MMP-1 and PICP, while no difference was observed in individuals with myocardial infarction. In the non-ST-segment-elevation acute coronary syndrome-like cohort, MMP-1 concentrations discriminated infarct-like myocarditis and NSTEMI with an area under the receiver operating characteristic curve (AUC) of 0.95 (95% CI, 0.89-1.00), whereas high-sensitivity cardiac troponin T performed inferiorly (AUC, 0.74 [95% CI, 0.58-0.90]; P=0.012). Application of an optimal MMP-1 cutoff had 94.4% sensitivity (95% CI, 72.7%-99.9%) and 90.9% specificity (95% CI, 70.8%-98.9%) for the diagnosis of infarct-like myocarditis in this cohort. The AUC of PICP in this context was 0.82 (95% CI, 0.68-0.97). As assessed by likelihood ratio tests, incorporating MMP-1 or PICP with age and C-reactive protein into composite prediction models enhanced their diagnostic performance. CONCLUSIONS MMP-1 and PICP could potentially be useful biomarkers for differentiating between NSTEMI and infarct-like myocarditis in individuals with non-ST-segment-elevation acute coronary syndrome-like presentation, though further research is needed to validate their clinical applicability.
Collapse
Affiliation(s)
- Lucas Bacmeister
- Department of Cardiology and Angiology, Medical Centre, Faculty of MedicineUniversity Heart Centre Freiburg‐Bad Krozingen, University of FreiburgGermany
| | - Ersin Cavus
- Clinic of CardiologyUniversity Heart and Vascular Centre Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Sebastian Bohnen
- Clinic of CardiologyUniversity Heart and Vascular Centre Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Enver Tahir
- Department of Diagnostic and Interventional RadiologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Hanna Wolf
- Department of Cardiology and Angiology, Medical Centre, Faculty of MedicineUniversity Heart Centre Freiburg‐Bad Krozingen, University of FreiburgGermany
| | - Annette Buellesbach
- Department of Cardiology and Angiology, Medical Centre, Faculty of MedicineUniversity Heart Centre Freiburg‐Bad Krozingen, University of FreiburgGermany
| | - Adrian Heidenreich
- Department of Cardiology and Angiology, Medical Centre, Faculty of MedicineUniversity Heart Centre Freiburg‐Bad Krozingen, University of FreiburgGermany
| | - Virginia K. Haacke
- Department of Cardiology and Angiology, Medical Centre, Faculty of MedicineUniversity Heart Centre Freiburg‐Bad Krozingen, University of FreiburgGermany
| | - Susanne Weber
- Department of Cardiology and Angiology, Medical Centre, Faculty of MedicineUniversity Heart Centre Freiburg‐Bad Krozingen, University of FreiburgGermany
- Division Methods in Clinical Epidemiology (MICLEP)Medical Centre, Faculty of Medicine, Institute of Medical Biometry and Statistics, University of FreiburgFreiburgGermany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, Medical Centre, Faculty of MedicineUniversity Heart Centre Freiburg‐Bad Krozingen, University of FreiburgGermany
| | - Tanja Zeller
- Clinic of CardiologyUniversity Heart and Vascular Centre Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Francisco Ojeda
- Clinic of CardiologyUniversity Heart and Vascular Centre Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Ulf K. Radunski
- Clinic of CardiologyUniversity Heart and Vascular Centre Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Gunnar K. Lund
- Department of Diagnostic and Interventional RadiologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Gerhard Adam
- Department of Diagnostic and Interventional RadiologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Stefan Blankenberg
- Clinic of CardiologyUniversity Heart and Vascular Centre Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Dirk Westermann
- Department of Cardiology and Angiology, Medical Centre, Faculty of MedicineUniversity Heart Centre Freiburg‐Bad Krozingen, University of FreiburgGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Kai Muellerleile
- Clinic of CardiologyUniversity Heart and Vascular Centre Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Diana Lindner
- Department of Cardiology and Angiology, Medical Centre, Faculty of MedicineUniversity Heart Centre Freiburg‐Bad Krozingen, University of FreiburgGermany
- Clinic of CardiologyUniversity Heart and Vascular Centre Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| |
Collapse
|
10
|
Rubiś PP, Dziewięcka E, González A, Cleland JGF. High variability in assays of blood markers of collagen turnover in cardiovascular disease: Implications for research and clinical practice. Eur J Heart Fail 2024. [PMID: 38980205 DOI: 10.1002/ejhf.3375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/30/2024] [Accepted: 06/23/2024] [Indexed: 07/10/2024] Open
Abstract
AIMS Fibrosis is a common feature of many chronic diseases, including heart failure, which can have deleterious effects on cardiac structure and function that are associated with adverse outcomes. By-products of collagen synthesis and degradation, such as carboxy- and amino-terminal pro- or telo-peptides of collagen type I and III (PICP, PINP, PIIINP, and CITP) have been extensively investigated as markers of fibrosis. Although the majority of studies report on the reproducibility of their assay results, there is no a comparison of biomarker assays across studies. Therefore, we conducted a systematic review adhering to PRISMA guidelines. METHODS AND RESULTS The search terms employed in Medline were: 'collagen AND cardiac' or 'collagen AND heart'. This query yielded a total of 1049 articles. Thereafter, specific search criteria were applied: (i) original English-language papers; (ii) human studies; (iii) in-vivo investigations; and (iv) blood/serum/plasma samples. Overall, 89 studies were identified (42 on PIIINP, 32 on PICP, 29 on CITP, and 17 on PINP). The range of reported values for PIIINP was between 0.06 to 11 800 μg/l; for PICP 0.006 to 1265 μg/l; for CITP 0.3 to 5450 μg/l; for PINP 0.15 to 80 μg/l. Extreme variations in values for fibrosis biomarkers were observed across studies, especially when different assays were used, but also with the same assays. CONCLUSIONS Our findings show that it is challenging to ascertain normal ranges or compare studies for the measurement of fibrosis biomarkers. Given the potential implications for clinical practice and current lack of awareness of these issues, this subject warrants comprehensive acknowledgement and understanding.
Collapse
Affiliation(s)
- Pawel Piotr Rubiś
- Krakow Specialist Hospital named after St. John Paul II, Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, Krakow, Poland
| | - Ewa Dziewięcka
- Krakow Specialist Hospital named after St. John Paul II, Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, Krakow, Poland
| | - Arantxa González
- Program of Cardiovascular Disease, CIMA Universidad de Navarra, Department of Pathology, Anatomy and Physiology, Universidad de Navarra and IdiSNA, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
11
|
Fatehi Hassanabad A, Zarzycki AN, Fedak PWM. Cellular and molecular mechanisms driving cardiac tissue fibrosis: On the precipice of personalized and precision medicine. Cardiovasc Pathol 2024; 71:107635. [PMID: 38508436 DOI: 10.1016/j.carpath.2024.107635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
Cardiac fibrosis is a significant contributor to heart failure, a condition that continues to affect a growing number of patients worldwide. Various cardiovascular comorbidities can exacerbate cardiac fibrosis. While fibroblasts are believed to be the primary cell type underlying fibrosis, recent and emerging data suggest that other cell types can also potentiate or expedite fibrotic processes. Over the past few decades, clinicians have developed therapeutics that can blunt the development and progression of cardiac fibrosis. While these strategies have yielded positive results, overall clinical outcomes for patients suffering from heart failure continue to be dire. Herein, we overview the molecular and cellular mechanisms underlying cardiac tissue fibrosis. To do so, we establish the known mechanisms that drive fibrosis in the heart, outline the diagnostic tools available, and summarize the treatment options used in contemporary clinical practice. Finally, we underscore the critical role the immune microenvironment plays in the pathogenesis of cardiac fibrosis.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Anna N Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
12
|
Song MH, Yoo J, Kwon DA, Chepurko E, Cho S, Fargnoli A, Hajjar RJ, Park WJ, Zangi L, Jeong D. Modified mRNA-Mediated CCN5 Gene Transfer Ameliorates Cardiac Dysfunction and Fibrosis without Adverse Structural Remodeling. Int J Mol Sci 2024; 25:6262. [PMID: 38892449 PMCID: PMC11172546 DOI: 10.3390/ijms25116262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Modified mRNAs (modRNAs) are an emerging delivery method for gene therapy. The success of modRNA-based COVID-19 vaccines has demonstrated that modRNA is a safe and effective therapeutic tool. Moreover, modRNA has the potential to treat various human diseases, including cardiac dysfunction. Acute myocardial infarction (MI) is a major cardiac disorder that currently lacks curative treatment options, and MI is commonly accompanied by fibrosis and impaired cardiac function. Our group previously demonstrated that the matricellular protein CCN5 inhibits cardiac fibrosis (CF) and mitigates cardiac dysfunction. However, it remains unclear whether early intervention of CF under stress conditions is beneficial or more detrimental due to potential adverse effects such as left ventricular (LV) rupture. We hypothesized that CCN5 would alleviate the adverse effects of myocardial infarction (MI) through its anti-fibrotic properties under stress conditions. To induce the rapid expression of CCN5, ModRNA-CCN5 was synthesized and administrated directly into the myocardium in a mouse MI model. To evaluate CCN5 activity, we established two independent experimental schemes: (1) preventive intervention and (2) therapeutic intervention. Functional analyses, including echocardiography and magnetic resonance imaging (MRI), along with molecular assays, demonstrated that modRNA-mediated CCN5 gene transfer significantly attenuated cardiac fibrosis and improved cardiac function in both preventive and therapeutic models, without causing left ventricular rupture or any adverse cardiac remodeling. In conclusion, early intervention in CF by ModRNA-CCN5 gene transfer is an efficient and safe therapeutic modality for treating MI-induced heart failure.
Collapse
Affiliation(s)
- Min Ho Song
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea; (M.H.S.)
| | - Jimeen Yoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; (J.Y.); (E.C.); (A.F.)
| | - Do-A Kwon
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan-si 15588, Republic of Korea; (D.-A.K.); (S.C.)
| | - Elena Chepurko
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; (J.Y.); (E.C.); (A.F.)
| | - Sunghye Cho
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan-si 15588, Republic of Korea; (D.-A.K.); (S.C.)
| | - Anthony Fargnoli
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; (J.Y.); (E.C.); (A.F.)
| | - Roger J. Hajjar
- Mass General Brigham Gene and Cell Therapy Institute, Boston, MA 02139, USA;
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea; (M.H.S.)
| | - Lior Zangi
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; (J.Y.); (E.C.); (A.F.)
| | - Dongtak Jeong
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; (J.Y.); (E.C.); (A.F.)
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan-si 15588, Republic of Korea; (D.-A.K.); (S.C.)
| |
Collapse
|
13
|
Carland C, Zhao L, Salman O, Cohen JB, Zamani P, Xiao Q, Dongre A, Wang Z, Ebert C, Greenawalt D, van Empel V, Richards AM, Doughty RN, Rietzschel E, Javaheri A, Wang Y, Schafer PH, Hersey S, Carayannopoulos LN, Seiffert D, Chang C, Gordon DA, Ramirez‐Valle F, Mann DL, Cappola TP, Chirinos JA. Urinary Proteomics and Outcomes in Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2024; 13:e033410. [PMID: 38639358 PMCID: PMC11179922 DOI: 10.1161/jaha.123.033410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/01/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Although several studies have addressed plasma proteomics in heart failure with preserved ejection fraction, limited data are available on the prognostic value of urinary proteomics. The objective of our study was to identify urinary proteins/peptides associated with death and heart failure admission in patients with heart failure with preserved ejection fraction. METHODS AND RESULTS The study population included participants enrolled in TOPCAT (Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist Trial). The relationship between urine protein levels and the risk of death or heart failure admission was assessed using Cox regression, in both nonadjusted analyses and adjusting for urine creatinine levels, and the MAGGIC (Meta-Analysis Global Group in Chronic Heart Failure) score. A total of 426 (12.4%) TOPCAT participants had urinary protein data and were included. There were 40 urinary proteins/peptides significantly associated with death or heart failure admission in nonadjusted analyses, 21 of which were also significant adjusted analyses. Top proteins in the adjusted analysis included ANGPTL2 (angiopoietin-like protein 2) (hazard ratio [HR], 0.5731 [95% CI, 0.47-0.7]; P=3.13E-05), AMY2A (α amylase 2A) (HR, 0.5496 [95% CI, 0.44-0.69]; P=0.0001), and DNASE1 (deoxyribonuclease-1) (HR, 0.5704 [95% CI, 0.46-0.71]; P=0.0002). Higher urinary levels of proteins involved in fibrosis (collagen VI α-1, collagen XV α-1), metabolism (pancreatic α-amylase 2A/B, mannosidase α class 1A member 1), and inflammation (heat shock protein family D member 1, inducible T cell costimulatory ligand) were associated with a lower risk of death or heart failure admission. CONCLUSIONS Our study identifies several novel associations between urinary proteins/peptides and outcomes in heart failure with preserved ejection fraction. Many of these associations are independent of clinical risk scores and may aid in risk stratification in this patient population.
Collapse
Affiliation(s)
- Corinne Carland
- Hospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Lei Zhao
- Bristol‐Myers Squibb CompanyLawrencevilleNJUSA
| | - Oday Salman
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Jordana B. Cohen
- Hospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Payman Zamani
- Hospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Qing Xiao
- Bristol‐Myers Squibb CompanyLawrencevilleNJUSA
| | | | | | | | | | - Vanessa van Empel
- Department of CardiologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | - A. Mark Richards
- Cardiovascular Research Institute, National University of SingaporeSingapore
- Christchurch Heart Institute, University of OtagoChristchurchNew Zealand
| | - Robert N. Doughty
- Christchurch Heart Institute, University of OtagoChristchurchNew Zealand
| | - Ernst Rietzschel
- Department of Cardiovascular DiseasesGhent University Hospital and Ghent UniversityGhentBelgium
| | - Ali Javaheri
- Washington University School of MedicineSt. LouisMOUSA
| | - Yixin Wang
- Bristol‐Myers Squibb CompanyLawrencevilleNJUSA
| | | | | | | | | | | | | | | | | | - Thomas P. Cappola
- Hospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Julio A. Chirinos
- Hospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| |
Collapse
|
14
|
Kadoglou NPE, Mouzarou A, Hadjigeorgiou N, Korakianitis I, Myrianthefs MM. Challenges in Echocardiography for the Diagnosis and Prognosis of Non-Ischemic Hypertensive Heart Disease. J Clin Med 2024; 13:2708. [PMID: 38731238 PMCID: PMC11084735 DOI: 10.3390/jcm13092708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
It has been well established that arterial hypertension is considered as a predominant risk factor for the development of cardiovascular diseases. Despite the link between arterial hypertension and cardiovascular diseases, arterial hypertension may directly affect cardiac function, leading to heart failure, mostly with preserved ejection fraction (HFpEF). There are echocardiographic findings indicating hypertensive heart disease (HHD), defined as altered cardiac morphology (left ventricular concentric hypertrophy, left atrium dilatation) and function (systolic or diastolic dysfunction) in patients with persistent arterial hypertension irrespective of the cardiac pathologies to which it contributes, such as coronary artery disease and kidney function impairment. In addition to the classical echocardiographic parameters, novel indices, like speckle tracking of the left ventricle and left atrium, 3D volume evaluation, and myocardial work in echocardiography, may provide more accurate and reproducible diagnostic and prognostic data in patients with arterial hypertension. However, their use is still underappreciated. Early detection of and prompt therapy for HHD will greatly improve the prognosis. Hence, in the present review, we shed light on the role of echocardiography in the contemporary diagnostic and prognostic approaches to HHD.
Collapse
Affiliation(s)
- Nikolaos P. E. Kadoglou
- Medical School, University of Cyprus, 215/6 Old Road Lefkosias-Lemesou, Aglatzia, Nicosia CY 2029, Cyprus
| | - Angeliki Mouzarou
- Department of Cardiology, Pafos General Hospital, Paphos CY 8026, Cyprus
| | | | - Ioannis Korakianitis
- Medical School, University of Cyprus, 215/6 Old Road Lefkosias-Lemesou, Aglatzia, Nicosia CY 2029, Cyprus
| | | |
Collapse
|
15
|
Kobayashi M, Ferreira JP, Duarte K, Bresso E, Huttin O, Bozec E, Brunner La Rocca HP, Delles C, Clark AL, Edelmann F, González A, Heymans S, Pellicori P, Petutschnigg J, Verdonschot JAJ, Rossignol P, Cleland JGF, Zannad F, Girerd N. Proteomic profiles of left atrial volume and its influence on response to spironolactone: Findings from the HOMAGE trial and STANISLAS cohort. Eur J Heart Fail 2024; 26:1231-1241. [PMID: 38528728 DOI: 10.1002/ejhf.3202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 03/27/2024] Open
Abstract
AIMS High left ventricular filling pressure increases left atrial volume and causes myocardial fibrosis, which may decrease with spironolactone. We studied clinical and proteomic characteristics associated with left atrial volume indexed by body surface area (LAVi), and whether LAVi influences the response to spironolactone on biomarker expression and clinical variables. METHODS AND RESULTS In the HOMAGE trial, where people at risk of heart failure were randomized to spironolactone or control, we analysed 421 participants with available LAVi and 276 proteomic measurements (Olink) at baseline, month 1 and 9 (mean age 73 ± 6 years; women 26%; LAVi 32 ± 9 ml/m2). Circulating proteins associated with LAVi were also assessed in asymptomatic individuals from a population-based cohort (STANISLAS; n = 1640; mean age 49 ± 14 years; women 51%; LAVi 23 ± 7 ml/m2). In both studies, greater LAVi was significantly associated with greater left ventricular masses and volumes. In HOMAGE, after adjustment and correction for multiple testing, greater LAVi was associated with higher concentrations of matrix metallopeptidase-2 (MMP-2), insulin-like growth factor binding protein-2 (IGFBP-2) and N-terminal pro-B-type natriuretic peptide (NT-proBNP) (false discovery rates [FDR] <0.05). These associations were externally replicated in STANISLAS (all FDR <0.05). Among these biomarkers, spironolactone decreased concentrations of MMP-2 and NT-proBNP, regardless of baseline LAVi (pinteraction > 0.10). Spironolactone also significantly reduced LAVi, improved left ventricular ejection fraction, lowered E/e', blood pressure and serum procollagen type I C-terminal propeptide (PICP) concentration, a collagen synthesis marker, regardless of baseline LAVi (pinteraction > 0.10). CONCLUSION In individuals without heart failure, LAVi was associated with MMP-2, IGFBP-2 and NT-proBNP. Spironolactone reduced these biomarker concentrations as well as LAVi and PICP, irrespective of left atrial size.
Collapse
Affiliation(s)
- Masatake Kobayashi
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
- Department of Cardiology, Tokyo Medical University Hospital, Tokyo, Japan
| | - João Pedro Ferreira
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
- Cardiovascular Research and Development Center, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Kevin Duarte
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - Emmanuel Bresso
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - Olivier Huttin
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - Erwan Bozec
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | | | - Christian Delles
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Andrew L Clark
- Department of Cardiology, University of Hull, Castle Hill Hospital, Yorkshire, UK
| | - Frank Edelmann
- Department of Internal Medicine and Cardiology Campus Virchow Klinikum, Charité University Medicine Berlin and German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Arantxa González
- CIMA Universidad de Navarra, Department of Pathology, Anatomy and Physiology Universidad de Navarra and IdiSNA, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Stephane Heymans
- Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Pierpaolo Pellicori
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Johannes Petutschnigg
- Department of Internal Medicine and/Cardiology, Campus Virchow Klinikum, Charité University Medicine Berlin, and German Heart Center Berlin, and Berlin Institute of Health (BIH), and German Centre for Cardiovascular research (DZHK), Berlin, Germany
| | - Job A J Verdonschot
- Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Patrick Rossignol
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
- Medical Specialties and Nephrology Dialysis Departments, Monaco Princess Grace Hospital and Monaco Private Hemodialysis Centre, Monaco, Monaco
| | - John G F Cleland
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Faiez Zannad
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - Nicolas Girerd
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| |
Collapse
|
16
|
Amemiya K, Matsuyama TA, Ishibashi-Ueda H, Morita Y, Matsumoto M, Ohta-Ogo K, Ikeda Y, Tsukamoto Y, Fukushima N, Fukushima S, Fujita T, Hatakeyama K. Can right ventricular endomyocardial biopsy predict left ventricular fibrosis beforehand in dilated cardiomyopathy? ESC Heart Fail 2024; 11:1001-1008. [PMID: 38234242 DOI: 10.1002/ehf2.14642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/18/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024] Open
Abstract
AIMS Myocardial fibrosis of the left ventricle (LV) is a prognostic factor in dilated cardiomyopathy (DCM). This study aims to evaluate whether fibrosis of right ventricular (RV) endomyocardial biopsy (EMB) can predict the degree of LV fibrosis beforehand in DCM. METHODS AND RESULTS Fibrosis extent in 70 RV-EMB specimens of DCM diagnosis was compared with that in the whole cross-sectional LV of excised hearts in the same patients (52 explanted hearts for transplant and 18 autopsied hearts). The median interval between biopsy and excision was 4.1 (0.13-19.3) years. The fibrosis area ratio of the EMBs and excised hearts were evaluated via image analysis. The distribution of cardiovascular magnetic resonance-late gadolinium enhancement (LGE) in the intraventricular septum was classified into four quartile categories. The fibrosis area ratio in RV-EMB correlated significantly with that in the short-axis cut of the LV of excised hearts (r = 0.82, P < 0.0001) and with a diffuse pattern of LGE (r = 0.71, P = 0.003). In a multivariate model, after adjusting for the interval between biopsy performance and heart excision, the fibrosis area ratio in RV-EMB was associated with that in LV-excised heart (regression coefficient, 0.82; 95% confidence interval, 0.68-0.95; P < 0.0001). CONCLUSIONS The fibrosis observed in RV-EMB positively correlated with the extent of fibrosis in the LV of excised hearts in patients with DCM. The study findings may help predict LV fibrosis, considered a prognostic factor of DCM through relatively accessible biopsy techniques.
Collapse
Affiliation(s)
- Kisaki Amemiya
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
- Department of Legal Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Taka-Aki Matsuyama
- Department of Legal Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hatsue Ishibashi-Ueda
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
- Division of Pathology, Hokusetsu General Hospital, Osaka, Japan
| | - Yoshiaki Morita
- Department of Radiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Manabu Matsumoto
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Keiko Ohta-Ogo
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yoshihiko Ikeda
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yasumasa Tsukamoto
- Department of Transplant Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Norihide Fukushima
- Department of Transplant Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
- Senri Kinran University, Osaka, Japan
| | - Satsuki Fukushima
- Department of Cardiovascular Surgery, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Tomoyuki Fujita
- Department of Cardiovascular Surgery, National Cerebral and Cardiovascular Center, Osaka, Japan
- Department of Cardiovascular Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kinta Hatakeyama
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
| |
Collapse
|
17
|
Ciampi CM, Sultana A, Ossola P, Farina A, Fragasso G, Spoladore R. Current experimental and early investigational agents for cardiac fibrosis: where are we at? Expert Opin Investig Drugs 2024; 33:389-404. [PMID: 38426439 DOI: 10.1080/13543784.2024.2326024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/28/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION Myocardial fibrosis (MF) is induced by factors activating pro-fibrotic pathways such as acute and prolonged inflammation, myocardial ischemic events, hypertension, aging process, and genetically-linked cardiomyopathies. Dynamics and characteristics of myocardial fibrosis development are very different. The broad range of myocardial fibrosis presentations suggests the presence of multiple potential targets. AREA COVERED Heart failure treatment involves medications primarily aimed at counteracting neurohormonal activation. While these drugs have demonstrated efficacy against MF, not all specifically target inflammation or fibrosis progression with some exceptions such as RAAS inhibitors. Consequently, new therapies are being developed to address this issue. This article is aimed to describe anti-fibrotic drugs currently employed in clinical practice and emerging agents that target specific pathways, supported by evidence from both preclinical and clinical studies. EXPERT OPINION Despite various preclinical findings suggesting the potential utility of new drugs and molecules for treating cardiac fibrosis in animal models, there is a notable scarcity of clinical trials investigating these effects. However, the pathology of damage and repair in the heart muscle involves a complex network of interconnected inflammatory pathways and various types of immune cells. Our comprehension of the positive and negative roles played by specific immune cells and cytokines is an emerging area of research.
Collapse
Affiliation(s)
- Claudio M Ciampi
- Health Science Department, University of Milan Bicocca, Milano, Italy
| | - Andrea Sultana
- Health Science Department, University of Milan Bicocca, Milano, Italy
| | - Paolo Ossola
- Health Science Department, University of Milan Bicocca, Milano, Italy
| | - Andrea Farina
- Division of Cardiology, Alessandro Manzoni Hospital, ASST- Lecco, Italy
| | - Gabriele Fragasso
- Heart Failure Unit Head, Division of Cardiology, IRCCS Vita-Salute San Raffaele University Hospital, Milan, Italy
| | - Roberto Spoladore
- Division of Cardiology, Alessandro Manzoni Hospital, ASST- Lecco, Italy
| |
Collapse
|
18
|
Ferreira JP, Butler J, Anker SD, Januzzi JL, Panova-Noeva M, Reese-Petersen AL, Sattar N, Schueler E, Pocock SJ, Filippatos G, Packer M, Sumin M, Zannad F. Effects of empagliflozin on collagen biomarkers in patients with heart failure: Findings from the EMPEROR trials. Eur J Heart Fail 2024; 26:274-284. [PMID: 38037709 DOI: 10.1002/ejhf.3101] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/02/2023] Open
Abstract
AIMS Extracellular matrix remodelling is one of the key pathways involved in heart failure (HF) progression. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) may have a role in attenuating myocardial fibrosis. The impact of SGLT2i on blood markers of collagen turnover in humans is not fully elucidated. This study aimed to investigate the effect of empagliflozin on serum markers of collagen turnover in patients enrolled in the EMPEROR-Preserved and EMPEROR-Reduced trials. METHODS AND RESULTS Overall, 1084 patients (545 in empagliflozin and 539 in placebo) were included in the analysis. Procollagen type I carboxy-terminal propeptide (PICP), a fragment of N-terminal type III collagen (PRO-C3), procollagen type I amino-terminal peptide (PINP), a fragment of C-terminal type VIa3 collagen (PRO-C6), a fragment of type I collagen (C1M), and a fragment of type III collagen (C3M) were measured in serum at baseline, 12 and 52 weeks. A mixed model repeated measurements model was used to evaluate the effect of empagliflozin versus placebo on the analysed biomarkers. Higher baseline PICP, PRO-C6 and PINP levels were associated with older age, a more severe HF presentation, higher levels of natriuretic peptides and high-sensitivity troponin T, and the presence of comorbid conditions such as chronic kidney disease and atrial fibrillation. Higher PICP levels were associated with the occurrence of the study primary endpoint (a composite of HF hospitalization or cardiovascular death), and PRO-C6 and PINP were associated with the occurrence of sustained worsening of kidney function. On the other hand, PRO-C3, C1M, and C3M were not associated with worse HF severity or study outcomes. Compared to placebo, empagliflozin reduced PICP at week 12 by 5% and at week 52 by 8% (week 12: geometric mean ratio = 0.95, 95% confidence interval [CI] 0.91-0.99, p = 0.012; week 52: geometric mean ratio = 0.92, 95% CI 0.88-0.97, p = 0.003). Additionally, empagliflozin reduced PRO-C3 at week 52 by 7% (week 12: geometric mean ratio = 0.98, 95% CI 0.95-1.02, p = 0.42; week 52: geometric mean ratio = 0.93, 95% CI 0.89-0.98, p = 0.003), without impact on other collagen markers. CONCLUSION Our observations are consistent with experimental observations that empagliflozin down-regulates profibrotic signalling. The importance of such an effect for the clinical benefits of SGLT2i in HF remains to be elucidated.
Collapse
Affiliation(s)
- João Pedro Ferreira
- Department of Surgery and Physiology, Cardiovascular Research and Development Center (UnIC@RISE), Faculty of Medicine of the University of Porto, Porto, Portugal
- Centre d'Investigations Cliniques Plurithématique 14-33, Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, Nancy, France
- Cardiovascular Research and Development Center, Nancy, France
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- Department of Medicine, University of Mississippi School of Medicine, Jackson, MS, USA
| | - Stefan D Anker
- Department of Cardiology (CVK), Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Heart Diseases, Wrocław Medical University, Wrocław, Poland
| | - James L Januzzi
- Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | - Stuart J Pocock
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Gerasimos Filippatos
- National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Milton Packer
- Baylor University Medical Center, Dallas, TX, USA
- Imperial College, London, UK
| | - Mikhail Sumin
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Faiez Zannad
- Centre d'Investigations Cliniques Plurithématique 14-33, Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, Nancy, France
| |
Collapse
|
19
|
de Boer RA, Díez J. Advancing the fight against fibrosis in patients with heart failure: The contribution of sodium-glucose cotransporter 2 inhibition. Eur J Heart Fail 2024; 26:285-287. [PMID: 38196302 DOI: 10.1002/ejhf.3125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024] Open
Affiliation(s)
- Rudolf A de Boer
- Thorax Center, Department of Cardiology, Erasmus Medical Center, Erasmus MC, Cardiovascular Institute, Rotterdam, The Netherlands
| | - Javier Díez
- Center for Applied Medical Research (CIMA), and School of Medicine, University of Navarra, Pamplona, Spain
- Center for Network Biomedical Research of Cardiovascular Diseases (CIBERCV), Carlos III Institute of Health, Madrid, Spain
| |
Collapse
|
20
|
Yu YL, Huang QF, An DW, Raad J, Martens DS, Latosinska A, Stolarz-Skrzypek K, Van Cleemput J, Feng YQ, Mischak H, Allegaert K, Verhamme P, Janssens S, Nawrot TS, Staessen JA. OSTEO18, a novel urinary proteomic signature, associated with osteoporosis in heart transplant recipients. Heliyon 2024; 10:e24867. [PMID: 38312576 PMCID: PMC10835361 DOI: 10.1016/j.heliyon.2024.e24867] [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: 07/17/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 02/06/2024] Open
Abstract
Background Immunosuppressive treatment in heart transplant (HTx) recipient causes osteoporosis. The urinary proteomic profile (UPP) includes peptide fragments derived from the bone extracellular matrix. Study aims were to develop and validate a multidimensional UPP biomarker for osteoporosis in HTx patients from single sequenced urinary peptides identifying the parent proteins. Methods A single-center HTx cohort was analyzed. Urine samples were measured by capillary electrophoresis coupled with mass spectrometry. Cases with osteoporosis and matching controls were randomly selected from all available 389 patients. In derivation case-control dataset, 1576 sequenced peptides detectable in ≥30 % of patients. Applying statistical analysis on these, an 18-peptide multidimensional osteoporosis UPP biomarker (OSTEO18) was generated by support vector modeling. The 2 replication datasets included 118 and 94 patients. For further validation, the whole cohort was analyzed. Statistical methods included logistic regression and receiver operating characteristic curve (ROC) analysis. Results In derivation dataset, the AUC, sensitivity and specificity of OSTEO18 were 0.83 (95 % CI: 0.76-0.90), 74.3 % and 87.1 %, respectively. In replication datasets, results were confirmatory. In the whole cohort (154 osteoporotic patients [39.6 %]), the ORs for osteoporosis increased (p < 0.0001) across OSTEO18 quartiles from 0.39 (95 % CI: 0.25-0.61) to 3.14 (2.08-4.75). With full adjustment for known osteoporosis risk factors, OSTEO18 improved AUC from 0.708 to 0.786 (p = 0.0003) for OSTEO18 categorized (optimized threshold: 0.095) and to 0.784 (p = 0.0004) for OSTEO18 as continuously distributed classifier. Conclusion OSTEO18 is a clinically meaningful novel biomarker indicative of osteoporosis in HTx recipients and is being certified as in-vitro diagnostic.
Collapse
Affiliation(s)
- Yu-Ling Yu
- The Research Unit Environment and Health, KU Leuven Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
- Non-Profit Research Association Alliance for the Promotion of Preventive Medicine, Mechelen, Belgium
| | - Qi-Fang Huang
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - De-Wei An
- The Research Unit Environment and Health, KU Leuven Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
- Non-Profit Research Association Alliance for the Promotion of Preventive Medicine, Mechelen, Belgium
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Julia Raad
- Mosaiques Diagnostics GmbH, Hannover, Germany
| | - Dries S. Martens
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | | | - Katarzyna Stolarz-Skrzypek
- First Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University, Kraków, Poland
| | | | - Ying-Qing Feng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | | | - Karel Allegaert
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- KU Leuven Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Peter Verhamme
- Center for Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Stefan Janssens
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Tim S. Nawrot
- The Research Unit Environment and Health, KU Leuven Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Jan A. Staessen
- Non-Profit Research Association Alliance for the Promotion of Preventive Medicine, Mechelen, Belgium
- The Biomedical Sciences Group, Faculty of Medicine, University of Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Wagdy R, Assem H, Abd-Elmohsen AM, Fata A, Gendy WE, Gaber M. Altered ventricular longitudinal strain in children with sickle cell disease: Role of TGF-β and IL-18. Pediatr Blood Cancer 2024; 71:e30762. [PMID: 37933422 DOI: 10.1002/pbc.30762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/02/2023] [Accepted: 10/19/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Cardiovascular involvement in sickle cell disease (SCD) has a great impact on patients' morbidity and mortality. Recently, interleukin-18 (IL-18) and transforming growth factor beta (TGF-β) were suggested as potential biomarkers for sickle cell cardiomyopathy. Global longitudinal strain (GLS) is a reliable early parameter for estimation of deformed myocardium. This study evaluated the role of TGF-β and IL-18 as risk indicators of altered strain in patients with SCD. METHODS Forty children with SCD (age >5 years) and 40 healthy children as controls, matched in age and sex, were enrolled in the study. All participants were subjected to clinical examination, complete blood count, serum ferritin, TGF-β, IL-18, and assessment of cardiac function by echocardiography. RESULTS TGF-β, IL-18, and lactic acid dehydrogenase (LDH) were significantly higher among cases (mean age: 10.6 ± 3.5 years) when compared to controls (p < .001), at cutoff values 41.7 ng/mL, 128.9 pg/mL, and 340 unit, respectively. The LS of free wall of RV (FW-RV) was significantly lower among cases when compared to controls (-23.55% ± 5.55% vs. -28.73% ± 2.43%, p < .001). Free wall longitudinal strain of the right ventricle (FWLS-RV) was significantly correlated to IL-18 and LDH (p < .001), while GLS-RV was significantly correlated to TGF-β. The GLS-LV was correlated to frequency of vaso-occlusive crises (VOCs) per year (p < .001). Diastolic function, E/A of LV, and RV were negatively correlated to the hemoglobin and serum ferritin levels. CONCLUSIONS The TGF-β, IL-18, and LDH along with frequent VOCs are correlated to altered LS, especially the right ventricle, and could serve as risk indicators for subclinical cardiomyopathy in children with SCD.
Collapse
Affiliation(s)
- Reham Wagdy
- Department of Pediatrics, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Hala Assem
- Department of Pediatrics, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ali M Abd-Elmohsen
- Department of Pediatrics, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Aya Fata
- Department of Pediatrics, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Wessam El Gendy
- Department of Clinical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Marwa Gaber
- Department of Cardiology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
22
|
Baris Feldman H, Chai Gadot C, Zahler D, Mory A, Aviram G, Elhanan E, Shefer G, Goldiner I, Amir Y, Kurolap A, Ablin JN. Corin and Left Atrial Cardiomyopathy, Hypertension, Arrhythmia, and Fibrosis. N Engl J Med 2023; 389:1685-1692. [PMID: 37913506 DOI: 10.1056/nejmoa2301908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Two siblings presented with cardiomyopathy, hypertension, arrhythmia, and fibrosis of the left atrium. Each had a homozygous null variant in CORIN, the gene encoding atrial natriuretic peptide (ANP)-converting enzyme. A plasma sample obtained from one of the siblings had no detectable levels of corin or N-terminal pro-ANP but had elevated levels of B-type natriuretic peptide (BNP) and one of the two protein markers of fibrosis that we tested. These and other findings support the hypothesis that BNP cannot fully compensate for a lack of activation of the ANP pathway and that corin is critical to normal ANP activity, left atrial function, and cardiovascular homeostasis.
Collapse
Affiliation(s)
- Hagit Baris Feldman
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Chofit Chai Gadot
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - David Zahler
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Adi Mory
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Galit Aviram
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Emil Elhanan
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Gabi Shefer
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Ilana Goldiner
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Yam Amir
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Alina Kurolap
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Jacob N Ablin
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| |
Collapse
|
23
|
Buckley LF, Agha AM, Dorbala P, Claggett BL, Yu B, Hussain A, Nambi V, Chen LY, Matsushita K, Hoogeveen RC, Ballantyne CM, Shah AM. MMP-2 Associates With Incident Heart Failure and Atrial Fibrillation: The ARIC Study. Circ Heart Fail 2023; 16:e010849. [PMID: 37753653 PMCID: PMC10842537 DOI: 10.1161/circheartfailure.123.010849] [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] [Received: 05/08/2023] [Accepted: 08/16/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND MMP (matrix metalloproteinase)-2 participates in extracellular matrix regulation and may be involved in heart failure (HF), atrial fibrillation (AF), and coronary heart disease. METHODS Among the 4693 ARIC study (Atherosclerosis Risk in Communities) participants (mean age, 75±5 years; 42% women) without prevalent HF, multivariable Cox proportional hazard models were used to estimate associations of plasma MMP-2 levels with incident HF, HF with preserved ejection fraction (≥50%), HF with reduced ejection fraction (<50%), AF, and coronary heart disease. Mediation of the association between MMP-2 and HF was assessed by censoring participants who developed AF or coronary heart disease before HF. Multivariable linear regression models were used to assess associations of MMP-2 with measures of left ventricular and left atrial structure and function. RESULTS Compared with the 3 lower quartiles, the highest MMP-2 quartile associated with greater risk of incident HF overall (adjusted hazard ratio, 1.48 [95% CI, 1.21-1.81]), incident HF with preserved ejection fraction (1.44 [95% CI, 1.07-1.94]), incident heart failure with reduced ejection fraction (1.48 [95% CI, 1.08-2.02]), and incident AF (1.44 [95% CI, 1.18-1.77]) but not incident coronary heart disease (0.97 [95% CI, 0.71-1.34]). Censoring AF attenuated the MMP-2 association with HF with preserved ejection fraction. Higher plasma MMP-2 levels were associated with larger left ventricular end-diastolic volume index, greater left ventricular mass index, higher E/e' ratio, larger left atrial volume index, and worse left atrial reservoir and contractile strains (all P<0.001). CONCLUSIONS Higher plasma MMP-2 levels associate with diastolic dysfunction, left atrial dysfunction, and a higher risk of incident HF and AF. AF is a mediator of MMP-2-associated HF with preserved ejection fraction risk.
Collapse
Affiliation(s)
- Leo F Buckley
- Department of Pharmacy Services (L.F.B.), Brigham and Women's Hospital, Boston, MA
| | - Ali M Agha
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Pranav Dorbala
- Division of Cardiovascular Medicine (P.D., B.L.C.), Brigham and Women's Hospital, Boston, MA
| | - Brian L Claggett
- Division of Cardiovascular Medicine (P.D., B.L.C.), Brigham and Women's Hospital, Boston, MA
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health Science Center at Houston (B.Y.)
| | - Aliza Hussain
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Vijay Nambi
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
- Michael E. DeBakey Veterans Affairs Hospital, Houston, TX (V.N.)
| | - Lin Yee Chen
- Division of Cardiovascular Medicine, University of Minnesota, Minneapolis (L.Y.C.)
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.)
| | - Ron C Hoogeveen
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Christie M Ballantyne
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Amil M Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (A.M.S.)
| |
Collapse
|
24
|
Bengel FM, Diekmann J, Hess A, Jerosch-Herold M. Myocardial Fibrosis: Emerging Target for Cardiac Molecular Imaging and Opportunity for Image-Guided Therapy. J Nucl Med 2023; 64:49S-58S. [PMID: 37918842 DOI: 10.2967/jnumed.122.264867] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/25/2023] [Indexed: 11/04/2023] Open
Abstract
Myocardial fibrosis is a major contributor to the development and progression of heart failure. Significant progress in the understanding of its pathobiology has led to the introduction and preclinical testing of multiple highly specific antifibrotic therapies. Because the mechanisms of fibrosis are highly dynamic, and because the involved cell populations are heterogeneous and plastic, there is increasing emphasis that any therapy directed specifically against myocardial fibrosis will require personalization and guidance by equally specific diagnostic testing for successful clinical translation. Noninvasive imaging techniques have undergone significant progress and provide increasingly specific information about the quantity, quality, and activity of myocardial fibrosis. Cardiac MRI can precisely map the extracellular space of the myocardium, whereas nuclear imaging characterizes activated fibroblasts and immune cells as the cellular components contributing to fibrosis. Existing techniques may be used in complementarity to provide the imaging biomarkers needed for the success of novel targeted therapies. This review provides a road map on how progress in basic fibrosis research, antifibrotic drug development, and high-end noninvasive imaging may come together to facilitate the success of fibrosis-directed cardiovascular medicine.
Collapse
Affiliation(s)
- Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| | - Johanna Diekmann
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| | - Annika Hess
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| | | |
Collapse
|
25
|
Raafs AG, Adriaans BP, Henkens MTHM, Verdonschot JAJ, Abdul Hamid MA, Díez J, Knackstedt C, van Empel VPM, Brunner-La Rocca HP, González A, Wildberger JE, Heymans SRB, Hazebroek MR. Biomarkers of Collagen Metabolism Are Associated with Left Ventricular Function and Prognosis in Dilated Cardiomyopathy: A Multi-Modal Study. J Clin Med 2023; 12:5695. [PMID: 37685762 PMCID: PMC10488673 DOI: 10.3390/jcm12175695] [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: 07/12/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Collagen cross-linking is a fundamental process in dilated cardiomyopathy (DCM) and occurs when collagen deposition exceeds degradation, leading to impaired prognosis. This study investigated the associations of collagen-metabolism biomarkers with left ventricular function and prognosis in DCM. METHODS DCM patients who underwent endomyocardial biopsy, blood sampling, and cardiac MRI were included. The primary endpoint included death, heart failure hospitalization, or life-threatening arrhythmias, with a follow-up of 6 years (5-8). RESULTS In total, 209 DCM patients were included (aged 54 ± 13 years, 65% male). No associations were observed between collagen volume fraction, circulating carboxy-terminal propeptide of procollagen type-I (PICP), or collagen type I carboxy-terminal telopeptide [CITP] and matrix metalloproteinase [MMP]-1 ratio and cardiac function parameters. However, CITP:MMP-1 was significantly correlated with global longitudinal strain (GLS) in the total study sample (R = -0.40, p < 0.0001; lower CITP:MMP-1 ratio was associated with impaired GLS), with even stronger correlations in patients with LVEF > 40% (R = -0.70, p < 0.0001). Forty-seven (22%) patients reached the primary endpoint. Higher MMP-1 levels were associated with a worse outcome, even after adjustment for clinical and imaging predictors (1.026, 95% CI 1.002-1.051, p = 0.037), but CITP and CITP:MMP-1 were not. Combining MMP-1 and PICP improved the goodness-of-fit (LHR36.67, p = 0.004). CONCLUSION The degree of myocardial cross-linking (CITP:MMP-1) is associated with myocardial longitudinal contraction, and MMP-1 is an independent predictor of outcome in DCM patients.
Collapse
Affiliation(s)
- Anne G. Raafs
- Department of Cardiology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.P.A.); (M.T.H.M.H.); (J.A.J.V.); (C.K.); (V.P.M.v.E.); (H.-P.B.-L.R.); (S.R.B.H.); (M.R.H.)
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 HX Maastricht, The Netherlands;
| | - Bouke P. Adriaans
- Department of Cardiology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.P.A.); (M.T.H.M.H.); (J.A.J.V.); (C.K.); (V.P.M.v.E.); (H.-P.B.-L.R.); (S.R.B.H.); (M.R.H.)
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 HX Maastricht, The Netherlands;
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Michiel T. H. M. Henkens
- Department of Cardiology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.P.A.); (M.T.H.M.H.); (J.A.J.V.); (C.K.); (V.P.M.v.E.); (H.-P.B.-L.R.); (S.R.B.H.); (M.R.H.)
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 HX Maastricht, The Netherlands;
- Netherlands Heart Institute (NLHI), 3511 EP Utrecht, The Netherlands
- Department of Pathology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands;
| | - Job A. J. Verdonschot
- Department of Cardiology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.P.A.); (M.T.H.M.H.); (J.A.J.V.); (C.K.); (V.P.M.v.E.); (H.-P.B.-L.R.); (S.R.B.H.); (M.R.H.)
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 HX Maastricht, The Netherlands;
- Department of Clinical Genetics, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Myrurgia A. Abdul Hamid
- Department of Pathology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands;
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, 31008 Pamplona, Spain; (J.D.); (A.G.)
- CIBERCV, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Christian Knackstedt
- Department of Cardiology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.P.A.); (M.T.H.M.H.); (J.A.J.V.); (C.K.); (V.P.M.v.E.); (H.-P.B.-L.R.); (S.R.B.H.); (M.R.H.)
| | - Vanessa P. M. van Empel
- Department of Cardiology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.P.A.); (M.T.H.M.H.); (J.A.J.V.); (C.K.); (V.P.M.v.E.); (H.-P.B.-L.R.); (S.R.B.H.); (M.R.H.)
| | - Hans-Peter Brunner-La Rocca
- Department of Cardiology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.P.A.); (M.T.H.M.H.); (J.A.J.V.); (C.K.); (V.P.M.v.E.); (H.-P.B.-L.R.); (S.R.B.H.); (M.R.H.)
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, 31008 Pamplona, Spain; (J.D.); (A.G.)
- CIBERCV, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Joachim E. Wildberger
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 HX Maastricht, The Netherlands;
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Stephane R. B. Heymans
- Department of Cardiology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.P.A.); (M.T.H.M.H.); (J.A.J.V.); (C.K.); (V.P.M.v.E.); (H.-P.B.-L.R.); (S.R.B.H.); (M.R.H.)
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 HX Maastricht, The Netherlands;
- Department of Cardiovascular Research, University of Leuven, 3000 Leuven, Belgium
| | - Mark R. Hazebroek
- Department of Cardiology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.P.A.); (M.T.H.M.H.); (J.A.J.V.); (C.K.); (V.P.M.v.E.); (H.-P.B.-L.R.); (S.R.B.H.); (M.R.H.)
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 HX Maastricht, The Netherlands;
| |
Collapse
|
26
|
Neuber S, Ermer MR, Emmert MY, Nazari-Shafti TZ. Treatment of Cardiac Fibrosis with Extracellular Vesicles: What Is Missing for Clinical Translation? Int J Mol Sci 2023; 24:10480. [PMID: 37445658 PMCID: PMC10342089 DOI: 10.3390/ijms241310480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Heart failure is the leading cause of morbidity and mortality and currently affects more than 60 million people worldwide. A key feature in the pathogenesis of almost all forms of heart failure is cardiac fibrosis, which is characterized by excessive accumulation of extracellular matrix components in the heart. Although cardiac fibrosis is beneficial in the short term after acute myocardial injury to preserve the structural and functional integrity of the heart, persistent cardiac fibrosis contributes to pathological cardiac remodeling, leading to mechanical and electrical dysfunction of the heart. Despite its high prevalence, standard therapies specifically targeting cardiac fibrosis are not yet available. Cell-based approaches have been extensively studied as potential treatments for cardiac fibrosis, but several challenges have been identified during clinical translation. The observation that extracellular vesicles (EVs) derived from stem and progenitor cells exhibit some of the therapeutic effects of the parent cells has paved the way to overcome limitations associated with cell therapy. However, to make EV-based products a reality, standardized methods for EV production, isolation, characterization, and storage must be established, along with concrete evidence of their safety and efficacy in clinical trials. This article discusses EVs as novel therapeutics for cardiac fibrosis from a translational perspective.
Collapse
Affiliation(s)
- Sebastian Neuber
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353 Berlin, Germany; (M.R.E.); (M.Y.E.); (T.Z.N.-S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 13353 Berlin, Germany
| | - Miriam R. Ermer
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353 Berlin, Germany; (M.R.E.); (M.Y.E.); (T.Z.N.-S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Maximilian Y. Emmert
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353 Berlin, Germany; (M.R.E.); (M.Y.E.); (T.Z.N.-S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 13353 Berlin, Germany
- Institute for Regenerative Medicine, University of Zurich, 8044 Zurich, Switzerland
| | - Timo Z. Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353 Berlin, Germany; (M.R.E.); (M.Y.E.); (T.Z.N.-S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 13353 Berlin, Germany
| |
Collapse
|
27
|
Bonios MJ, Armenis I, Kogerakis N, Thodou A, Fragoulis S, Georgiadou P, Leontiadis E, Chamogeorgakis T, Drakos SG, Adamopoulos S. Prospective Phenotyping of Right Ventricle Function Following Intra-Aortic Balloon Pump Counterpulsation in Left Ventricular Assist Device Candidates: Outcomes and Predictors of Response. ASAIO J 2023; 69:e215-e222. [PMID: 37000672 DOI: 10.1097/mat.0000000000001927] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023] Open
Abstract
Intra-aortic balloon pump (IABP) may be applied to optimize advanced heart failure (AHF) patients and improve right ventricular (RV) function before left ventricular assist device (LVAD) implantation. We aimed to evaluate the outcome of this intervention and define RV response predictors. Decompensated AHF patients, not eligible for LVAD because of poor RV function, who required IABP for stabilization were enrolled. Echocardiography and invasive hemodynamics were serially applied to determine fulfillment of prespecified "LVAD eligibility RV function" criteria (right atrium pressure [RA] <12 mm Hg, pulmonary artery pulsatility index [PAPi] >2.00, RA/pulmonary capillary wedge pressure [PCWP] <0.67, RV strain <-14.0%). Right ventricular-free wall tissue was harvested to assess interstitial fibrosis. Eighteen patients (12 male), aged 38 ± 14 years were supported with IABP for 55 ± 51 (3-180) days. In 11 (61.1%), RV improved and fulfilled the prespecified criteria, while seven (38.9%) showed no substantial improvement. Histopathology revealed an inverse correlation between RV interstitial fibrosis and functional benefit following IABP: interstitial fibrosis correlated with post-IABP RA ( r = 0.63, p = 0.037), RA/PCWP ( r = 0.87, p = 0.001), PAPi ( r = -0.83, p = 0.003). Conclusively, IABP improves RV function in certain AHF patients facilitating successful LVAD implantation. Right ventricular interstitial fibrosis quantification may be applied to predict response and guide preoperative patient selection and optimization. http://links.lww.com/ASAIO/A995.
Collapse
Affiliation(s)
- Michael J Bonios
- From the Heart Failure and Transplant Units, Onassis Cardiac Surgery Center, Athens, Greece
- Division of Cardiovascular Medicine and Nora Eccles Treadwell Cardiovascular Research and Training Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Iakovos Armenis
- From the Heart Failure and Transplant Units, Onassis Cardiac Surgery Center, Athens, Greece
| | - Nektarios Kogerakis
- From the Heart Failure and Transplant Units, Onassis Cardiac Surgery Center, Athens, Greece
| | - Aspasia Thodou
- Division of Cardiovascular Medicine and Nora Eccles Treadwell Cardiovascular Research and Training Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Socrates Fragoulis
- From the Heart Failure and Transplant Units, Onassis Cardiac Surgery Center, Athens, Greece
| | - Panagiota Georgiadou
- From the Heart Failure and Transplant Units, Onassis Cardiac Surgery Center, Athens, Greece
| | - Evangelos Leontiadis
- From the Heart Failure and Transplant Units, Onassis Cardiac Surgery Center, Athens, Greece
| | | | - Stavros G Drakos
- Division of Cardiovascular Medicine and Nora Eccles Treadwell Cardiovascular Research and Training Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Stamatis Adamopoulos
- From the Heart Failure and Transplant Units, Onassis Cardiac Surgery Center, Athens, Greece
| |
Collapse
|
28
|
Samidurai A, Saravanan M, Ockaili R, Kraskauskas D, Lau SYV, Kodali V, Ramasamy S, Bhoopathi K, Nair M, Roh SK, Kukreja RC, Das A. Single-Dose Treatment with Rapamycin Preserves Post-Ischemic Cardiac Function through Attenuation of Fibrosis and Inflammation in Diabetic Rabbit. Int J Mol Sci 2023; 24:8998. [PMID: 37240345 PMCID: PMC10218967 DOI: 10.3390/ijms24108998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Robust activation of mTOR (mammalian target of rapamycin) signaling in diabetes exacerbates myocardial injury following lethal ischemia due to accelerated cardiomyocyte death with cardiac remodeling and inflammatory responses. We examined the effect of rapamycin (RAPA, mTOR inhibitor) on cardiac remodeling and inflammation following myocardial ischemia/reperfusion (I/R) injury in diabetic rabbits. Diabetic rabbits (DM) were subjected to 45 min of ischemia and 10 days of reperfusion by inflating/deflating a previously implanted hydraulic balloon occluder. RAPA (0.25 mg/kg, i.v.) or DMSO (vehicle) was infused 5 min before the onset of reperfusion. Post-I/R left ventricular (LV) function was assessed by echocardiography and fibrosis was evaluated by picrosirius red staining. Treatment with RAPA preserved LV ejection fraction and reduced fibrosis. Immunoblot and real-time PCR revealed that RAPA treatment inhibited several fibrosis markers (TGF-β, Galectin-3, MYH, p-SMAD). Furthermore, immunofluorescence staining revealed the attenuation of post-I/R NLRP3-inflammasome formation with RAPA treatment as shown by reduced aggregation of apoptosis speck-like protein with a caspase recruitment domain and active-form of caspase-1 in cardiomyocytes. In conclusion, our study suggests that acute reperfusion therapy with RAPA may be a viable strategy to preserve cardiac function with the alleviation of adverse post-infarct myocardial remodeling and inflammation in diabetic patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Rakesh C. Kukreja
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.)
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.)
| |
Collapse
|
29
|
Ferreira JP, Cleland JG, Girerd N, Rossignol P, Pellicori P, Cosmi F, Mariottoni B, González A, Diez J, Solomon SD, Claggett B, Pfeffer MA, Pitt B, Petutschnigg J, Pieske B, Edelmann F, Zannad F. Spironolactone effect on circulating procollagen type I carboxy-terminal propeptide: Pooled analysis of three randomized trials. Int J Cardiol 2023; 377:86-88. [PMID: 36738846 DOI: 10.1016/j.ijcard.2023.01.088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Spironolactone might improve the prognosis of patients with heart failure with preserved left ventricular ejection fraction (HFpEF), but the mechanisms by which it acts are uncertain. Serum concentrations of procollagen type I carboxy-terminal propeptide (PICP) reflect the synthesis of type I collagen and correlate well with histologically proven cardiac fibrosis. AIMS To investigate the effect of spironolactone on serum PICP concentration in patients with stage B and C HFpEF across three trials (HOMAGE, ALDO-DHF, and TOPCAT) for which measurements of serum PICP were available. METHODS Random-effects meta-analysis. RESULTS A total of 1038 patients with PICP measurements available both at baseline and 9-12 months were included in this analysis: 488 (47.0%) from HOMAGE, 386 (37.2%) from ALDO-DHF, and 164 (15.8%) from TOPCAT. The median (percentile25-75) serum PICP was 98 (76-128) ng/mL. Compared to placebo or usual care, administration of spironolactone for 9 to 12 months reduced serum PICP by -7.4 ng/mL, 95%CI -13.9 to -0.9, P-value =0.02. The effect was moderately heterogeneous (I2 = 64%) with the most pronounced effect seen in TOPCAT where PICP was reduced by -27.0 ng/mL, followed by HOMAGE where PICP was reduced by -8.1 ng/mL, and was least marked in ALDO-DHF where PICP changed by -2.9 ng/mL. The association between spironolactone and serum PICP was not mediated substantially by blood pressure. CONCLUSIONS Spironolactone reduced serum concentrations of PICP in patients with HFpEF with different severity and stages of disease. These findings are consistent with spironolactone having an anti-fibrotic effect.
Collapse
Affiliation(s)
- João Pedro Ferreira
- Cardiovascular R&D Centre - UnIC@RISE, Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto, Porto, Portugal & Internal Medicine Departament, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal; Université de Lorraine, Inserm, Centre d'Investigation Clinique Plurithématique 1433, U1116, CHRU de Nancy, F-CRIN INI-CRCT, Nancy, France.
| | - John G Cleland
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Nicolas Girerd
- Université de Lorraine, Inserm, Centre d'Investigation Clinique Plurithématique 1433, U1116, CHRU de Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Patrick Rossignol
- Université de Lorraine, Inserm, Centre d'Investigation Clinique Plurithématique 1433, U1116, CHRU de Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Pierpaolo Pellicori
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Franco Cosmi
- Department of Cardiology, Cortona Hospital, Arezzo, Italy
| | | | - Arantxa González
- Program of Cardiovascular Diseases, CIMA, Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Javier Diez
- Program of Cardiovascular Diseases, CIMA, Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Scott D Solomon
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Brian Claggett
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Marc A Pfeffer
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Bertram Pitt
- Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Johannes Petutschnigg
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité University Medicine Berlin, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité University Medicine Berlin, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Frank Edelmann
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité University Medicine Berlin, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Faiez Zannad
- Université de Lorraine, Inserm, Centre d'Investigation Clinique Plurithématique 1433, U1116, CHRU de Nancy, F-CRIN INI-CRCT, Nancy, France
| |
Collapse
|
30
|
Nemtsova V, Vischer AS, Burkard T. Hypertensive Heart Disease: A Narrative Review Series-Part 1: Pathophysiology and Microstructural Changes. J Clin Med 2023; 12:jcm12072606. [PMID: 37048689 PMCID: PMC10094934 DOI: 10.3390/jcm12072606] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
Sustained hypertension causes structural, functional, and neurohumoral abnormalities in the heart, a disease commonly termed hypertensive heart disease (HHD). Modern concepts of HHD, including processes of remodeling leading to the development of various LVH patterns, HF patterns accompanied by micro- and macrovasculopathies, and heart rhythm and conduction disturbances, are missing in the available definitions, despite copious studies being devoted to the roles of myocardial and vascular fibrosis, and neurohumoral and sympathetic regulation, in HHD development and progression. No comprehensive and generally accepted universal definition and classification of HHD is available to date, implementing diagnostic criteria that incorporate all the possible changes and adaptions to the heart. The aim of this review series is to summarize the relevant literature and data, leading to a proposal of a definition and classification of HHD. This first article reviews the processes of initial myocardial remodeling, and myocardial and vascular fibrosis, occurring in HHD. We discuss important pathophysiological and microstructural changes, the different patterns of fibrosis, and the biomarkers and imaging used to detect fibrosis in HHD. Furthermore, we review the possible methods of targeting myocardial fibrosis in HHD, and highlight areas for further research.
Collapse
Affiliation(s)
- Valeriya Nemtsova
- Medical Outpatient Department and Hypertension Clinic, ESH Hypertension Centre of Excellence, University Hospital Basel, 4031 Basel, Switzerland
- Internal Diseases and Family Medicine Department, Educational and Scientific Medical Institute, National Technical University "Kharkiv Polytechnic Institute", 61002 Kharkiv, Ukraine
| | - Annina S Vischer
- Medical Outpatient Department and Hypertension Clinic, ESH Hypertension Centre of Excellence, University Hospital Basel, 4031 Basel, Switzerland
| | - Thilo Burkard
- Medical Outpatient Department and Hypertension Clinic, ESH Hypertension Centre of Excellence, University Hospital Basel, 4031 Basel, Switzerland
- Department of Cardiology, University Hospital Basel, 4031 Basel, Switzerland
| |
Collapse
|
31
|
Effects of resveratrol supplementation on cardiac remodeling in hypertensive patients: a randomized controlled clinical trial. Hypertens Res 2023:10.1038/s41440-023-01231-z. [PMID: 36854725 DOI: 10.1038/s41440-023-01231-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/07/2023] [Accepted: 02/07/2023] [Indexed: 03/02/2023]
Abstract
Resveratrol (RES) has been demonstrated to be protective in the cardiovascular system in animal studies, but the evidence is limited in humans. The purpose of the study was to evaluate the effect of RES supplementation on cardiac remodeling in patients with hypertension. Eighty Subjects were randomly divided into RES group (plus RES 400 mg/d in addition to conventional therapy, n = 43) and control group (conventional therapy, n = 37). The main outcomes of the study were changes within cardiac-remodeling parameters. Secondary outcomes were changes in anthropometric parameters, arterial stiffness parameters and mechanism indices. There was no statistically significant difference between the RES group and control group in terms of baseline characteristics. After 6 months, the RES group had smaller left atrial, lower E/e', higher left ventricular global longitudinal strain and lower biomarkers indicating cardiac fibrosis (expressed by decreases in procollagen type I C-peptide and galectin-3) compared to the control group. However, there was no significant difference in left ventricular structure between the two groups. Although the RES group showed a significant decrease in brachial-ankle pulse wave velocity compared to the pre-intervention value, the difference between the RES and the control groups was not obvious. What's more, compared with the control group, the serum levels of sirtuin3, superoxide dismutase and klotho were significantly increased in the RES group. In conclusion, RES supplementation can alleviate left atrial remodeling, improve left ventricular diastolic function and may alleviate cardiac fibrosis in hypertensive patients, and could be used as an adjunct to conventional therapies of hypertensive heart disease.
Collapse
|
32
|
Masurkar N, Bouvet M, Logeart D, Jouve C, Dramé F, Claude O, Roux M, Delacroix C, Bergerot D, Mercadier JJ, Sirol M, Gellen B, Livrozet M, Fayol A, Robidel E, Trégouët DA, Marazzi G, Sassoon D, Valente M, Hulot JS. Novel Cardiokine GDF3 Predicts Adverse Fibrotic Remodeling After Myocardial Infarction. Circulation 2023; 147:498-511. [PMID: 36484260 DOI: 10.1161/circulationaha.121.056272] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Myocardial infarction (MI) induces a repair response that ultimately generates a stable fibrotic scar. Although the scar prevents cardiac rupture, an excessive profibrotic response impairs optimal recovery by promoting the development of noncontractile fibrotic areas. The mechanisms that lead to cardiac fibrosis are diverse and incompletely characterized. We explored whether the expansion of cardiac fibroblasts after MI can be regulated through a paracrine action of cardiac stromal cells. METHODS We performed a bioinformatic secretome analysis of cardiac stromal PW1+ cells isolated from normal and post-MI mouse hearts to identify novel secreted proteins. Functional assays were used to screen secreted proteins that promote fibroblast proliferation. The expressions of candidates were subsequently analyzed in mouse and human hearts and plasmas. The relationship between levels of circulating protein candidates and adverse post-MI cardiac remodeling was examined in a cohort of 80 patients with a first ST-segment-elevation MI and serial cardiac magnetic resonance imaging evaluations. RESULTS Cardiac stromal PW1+ cells undergo a change in paracrine behavior after MI, and the conditioned media from these cells induced a significant increase in the proliferation of fibroblasts. We identified a total of 12 candidates as secreted proteins overexpressed by cardiac PW1+ cells after MI. Among these factors, GDF3 (growth differentiation factor 3), a member of the TGF-β (transforming growth factor-β) family, was markedly upregulated in the ischemic hearts. Conditioned media specifically enriched with GDF3 induced fibroblast proliferation at a high level by stimulation of activin-receptor-like kinases. In line with the secretory nature of this protein, we next found that GDF3 can be detected in mice and human plasma samples, with a significant increase in the days after MI. In humans, higher GDF3 circulating levels (measured in the plasma at day 4 after MI) were significantly associated with an increased risk of adverse remodeling 6 months after MI (adjusted odds ratio, 1.76 [1.03-3.00]; P=0.037), including lower left ventricular ejection fraction and a higher proportion of akinetic segments. CONCLUSIONS Our findings define a mechanism for the profibrotic action of cardiac stromal cells through secreted cardiokines, such as GDF3, a candidate marker of adverse fibrotic remodeling after MI. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT01113268.
Collapse
Affiliation(s)
- Nihar Masurkar
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France
| | - Marion Bouvet
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France
| | - Damien Logeart
- Hôpital Lariboisière (D.L., M.S.), Université de Paris, Cité' France
| | - Charlène Jouve
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France
| | - Fatou Dramé
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France
| | - Olivier Claude
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France
| | - Maguelonne Roux
- Sorbonne Université, UPMC Univ Paris 06, INSERM, Institute of Cardio Metabolism and Nutrition, France (M.R.)
| | - Clément Delacroix
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France
| | - Damien Bergerot
- CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, Paris, France (D.B., M.L., A.F., J.-S.H.)
| | - Jean-Jacques Mercadier
- Signalisation and Cardiovascular Pathophysiology - Univ. Paris-Sud, INSERM, Université Paris-Saclay, Châtenay-Malabry, France (J.-J.M.)
| | - Marc Sirol
- Hôpital Lariboisière (D.L., M.S.), Université de Paris, Cité' France
| | - Barnabas Gellen
- ELSAN, Polyclinique de Poitiers, Service de Cardiologie, France (B.G.)
| | - Marine Livrozet
- CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, Paris, France (D.B., M.L., A.F., J.-S.H.)
| | - Antoine Fayol
- CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, Paris, France (D.B., M.L., A.F., J.-S.H.)
| | - Estelle Robidel
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France
| | - David-Alexandre Trégouët
- INSERM UMR_S 1219, Bordeaux Population Health Research Center, University of Bordeaux, France (D.-A.T.)
| | - Giovanna Marazzi
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France
| | - David Sassoon
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France
| | - Mariana Valente
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France
| | - Jean-Sébastien Hulot
- Paris Cardiovascular Research Center, INSERM (N.M., M.B., C.J., F.D., O.C., C.D., E.R., G.M., D.S., M.V., J.-S.H.), Université de Paris, Cité' France.,CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, Paris, France (D.B., M.L., A.F., J.-S.H.)
| |
Collapse
|
33
|
Aziz S, Yalan L, Raza MA, Lemin J, Akram HMB, Zhao W. GSK126 an inhibitor of epigenetic regulator EZH2 suppresses cardiac fibrosis by regulating the EZH2-PAX6-CXCL10 pathway. Biochem Cell Biol 2023; 101:87-100. [PMID: 36469862 DOI: 10.1139/bcb-2022-0224] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Myocardial fibrosis is a common pathological companion of various cardiovascular diseases. To date, the role of enhancer of zeste homolog 2 (EZH2) in cancer has been well demonstrated including in renal carcinoma and its inhibitors have entered the stage of phase I/II clinical trials. However, the precise mechanism of EZH2 in cardiac diseases is largely unclear. In the current study, we first found that EZH2 expression was increased in Ang-II-treated cardiac fibroblasts (CFs) and mouse heart homogenates following isoproterenol (ISO) administration for 21 days, respectively. Ang-II induces CFs activation and increased collagen-I, collagen-III, α-SMA, EZH2, and trimethylates lysine 27 on histone 3 (H3K27me3) expressions can be reversed by EZH2 inhibitor (GSK126) and EZH2 siRNA. The ISO-induced cardiac hypertrophy, and fibrosis in vivo which were also related to the upregulation of EZH2 and its downstream target, H3K27me3, could be recovered by GSK126. Furthermore, the upregulation of EZH2 induces the decrease of paired box 6 (PAX6) and C-X-C motif ligand 10 (CXCL10) "which" were also reversed by GSK126 treatment. In summary, the present evidence strongly suggests that GSK126 could be a therapeutic intervention, blunting the development and progression of myocardial fibrosis in an EZH2-PAX6-CXCL10-dependent manner.
Collapse
Affiliation(s)
- Shireen Aziz
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Li Yalan
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Muhammad Ahmer Raza
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiao Lemin
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Hafiz Muhamamd Bilal Akram
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Wen Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| |
Collapse
|
34
|
Gomes MFP, de Moura EDOC, Cardoso NM, da Silva GA, Dos Santos ACC, de Souza FS, Estadella D, Lambertucci RH, Lago JHG, Medeiros A. Supplementation with okra combined or not with exercise training is able to protect the heart of animals with metabolic syndrome. Sci Rep 2023; 13:1468. [PMID: 36702820 PMCID: PMC9879946 DOI: 10.1038/s41598-023-28072-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
The metabolic syndrome (MetS) is a clinical manifestation strongly associated with cardiovascular disease, the main cause of death worldwide. In view of this scenario, many therapeutic proposals have appeared in order to optimize the treatment of individuals with MetS, including the practice of exercise training (ET) and the consumption of okra (O). The aim of the present study was to evaluate the effect of O consumption and/or ET in animals with MetS. In all, 32 male Zucker rats (fa/fa) at 10 weeks old were randomly distributed into four groups of 8 animals each: MetS, MetS+O, MetS+ET and MetS+ET+O, and 8 lean Zucker rats (fa/ +) comprised the control group. Okra was administered by orogastric gavage 2x/day (morning and night, 100 mg/kg), 5 days/week, for 6 weeks. The ET was performed on a treadmill 1x/day (afternoon), 5 days/week, 60 min/day, in an intensity of 70% of maximal capacity, for the same days of O treatment. It was found that, O consumption alone was able to promote improved insulin sensitivity (MetS 93.93 ± 8.54 mg/dL vs. MetS+O 69.95 ± 18.7 mg/dL, p ≤ 0.05, d = 1.65, CI = 50.32 -89.58, triglyceride reduction (MetS 492.9 ± 97.8 mg/dL vs. MetS+O 334.9 ± 98.0 mg/dL, p ≤ 0.05, d = 1.61, CI = 193.2-398.7). In addition, it promoted a reduction in systolic blood pressure (MetS 149.0 ± 9.3 mmHg vs. MetS+O 132.0 ± 11.4 mmHg, p ≤ 0.05, d = 1.63, CI = 120-140), prevented an increase in cardiac collagen (MetS 12.60 ± 2.08% vs. MetS+O 7.52 ± 0.77%, p ≤ 0.05, d = 3.24, CI = 6.56-8.49). When associated with ET, the results were similar. Thus, we conclude that O consumption combined or not with aerobic ET can have a protective effect on the cardiac tissue of rats with MetS.
Collapse
Affiliation(s)
- Moisés Felipe Pereira Gomes
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil.
- Center for Applied Social Sciences and Health, Universidade Católica de Santos (Unisantos), Av. Conselheiro Nébias, 300, Vila Matias, Santos, SP, 11015-002, Brazil.
| | | | - Naiara Magalhães Cardoso
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| | - Graziele Aparecida da Silva
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| | - Ana Carolina Cardoso Dos Santos
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| | - Fernanda Samantha de Souza
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo (UNIFESP), Rua Prof. Artur Riedel, n° 275, Eldorado, Diadema, SP, 09972-270, Brazil
| | - Débora Estadella
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| | - Rafael Herling Lambertucci
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| | - João Henrique Ghilardi Lago
- Center of Natural and Human Sciences, Universidade Federal Do ABC, Av. Dos Estados, 500, Bangú, Santo André, SP, 09210-580, Brazil
| | - Alessandra Medeiros
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| |
Collapse
|
35
|
Clinical Utility of Strain Imaging in Assessment of Myocardial Fibrosis. J Clin Med 2023; 12:jcm12030743. [PMID: 36769393 PMCID: PMC9917743 DOI: 10.3390/jcm12030743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/26/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Myocardial fibrosis (MF) is a non-reversible process that occurs following acute or chronic myocardial damage. MF worsens myocardial deformation, remodels the heart and raises myocardial stiffness, and is a crucial pathological manifestation in patients with end-stage cardiovascular diseases and closely related to cardiac adverse events. Therefore, early quantitative analysis of MF plays an important role in risk stratification, clinical decision, and improvement in prognosis. With the advent and development of strain imaging modalities in recent years, MF may be detected early in cardiovascular diseases. This review summarizes the clinical usefulness of strain imaging techniques in the non-invasive assessment of MF.
Collapse
|
36
|
Bertaud A, Joshkon A, Heim X, Bachelier R, Bardin N, Leroyer AS, Blot-Chabaud M. Signaling Pathways and Potential Therapeutic Strategies in Cardiac Fibrosis. Int J Mol Sci 2023; 24:ijms24021756. [PMID: 36675283 PMCID: PMC9866199 DOI: 10.3390/ijms24021756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
Cardiac fibrosis constitutes irreversible necrosis of the heart muscle as a consequence of different acute (myocardial infarction) or chronic (diabetes, hypertension, …) diseases but also due to genetic alterations or aging. Currently, there is no curative treatment that is able to prevent or attenuate this phenomenon that leads to progressive cardiac dysfunction and life-threatening outcomes. This review summarizes the different targets identified and the new strategies proposed to fight cardiac fibrosis. Future directions, including the use of exosomes or nanoparticles, will also be discussed.
Collapse
|
37
|
Cong X, Tian B, Zhu X, Zhang X, Gu W, Zhao H, Hao S, Ning Z. Interleukin-11 Is Elevated in Patients with Atrial Fibrillation, Correlates with Serum Fibrosis Markers, and Represents a Therapeutic Target for Atrial Fibrosis. Cerebrovasc Dis 2023; 52:575-586. [PMID: 36599329 DOI: 10.1159/000527740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/11/2022] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Inflammatory cytokines are closely associated with developing cardiac fibrosis. This research aimed to explore the significant role of IL-11 in atrial fibrosis progression and potential therapeutic targets. METHODS 207 AF patients and 160 healthy subjects were included in the case-control study. Blood samples were analyzed for the level of IL-11 by enzyme-linked immunosorbent assay (ELISA). Angiotensin II (Ang II)-treated fibrosis mouse models were generated, and expression of IL-11 mRNA and protein was detected by RT-qPCR and Western blot. IL-11 antagonist was used to evaluating atrial fibrosis-related markers. RESULTS The persistent atrial fibrillation patients (n = 76) had significantly larger left atrial size, higher serum levels of hypertrophic protein BNP, proinflammatory cytokine high-sensitivity C-reactive protein (hs-CRP), and interleukin-6 (IL-6) compared to paroxysmal atrial fibrillation patients (n = 131), and healthy subjects (all p < 0.05). Pearson correlation analysis revealed significant positive correlation between serum IL-11 and cardiac fibrosis markers BNP (r = 0.394, p < 0.001), CTX-I (r = 0.418, p < 0.001), PICP (r = 0.306, p < 0.001), PIIINP (r = 0.335, p < 0.001), and TGF-β1 (r = 0.273, p < 0.001). In the fibrosis mouse model, Ang II infusion significantly upregulated IL-11 mRNA and protein expression in the left atrium of mice (p < 0.05), as well as staining intensity of Masson trichrome, the intensity of α-SMA, and it increased mRNA expression of collagen I and III in atrial tissue. IL-11 antagonist treatment significantly attenuated Masson trichrome, number of α-SMA-positive myofibroblasts in atrial tissue. Also, it significantly reduced the p-ERK1/2 in atrial tissue of mice infused with Ang II (p < 0.05). CONCLUSIONS IL-11 is upregulated in the serum of AF patients, and IL-11 inhibitor significantly inhibited Ang II-induced atrial fibrosis, a key pathological feature of AF. Therefore, IL-11 could be a potential therapeutic target for AF.
Collapse
Affiliation(s)
- Xinpeng Cong
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Bei Tian
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Xi Zhu
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Xiaogang Zhang
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Wei Gu
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Hanjun Zhao
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Shuwen Hao
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Zhongping Ning
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| |
Collapse
|
38
|
Abstract
Hypertensive heart disease (HHD) is currently the second leading cause of heart failure. The prevalence of HHD and its associated risk of heart failure have increased despite substantial improvement in arterial hypertension treatment and control in the recent decades. Therefore, the prevention of heart failure in patients with HHD represents an unmet medical need, due to its clinical, economic, and social impact. In this conceptual framework, we call to action because the time has come for diagnosis and treatment of patients with HHD not to be limited to assessment of morphological and functional left ventricular changes, blood pressure control, and left ventricular hypertrophy regression. We propose a further perspective incorporating also the detection and reversal of the histological changes that develop in the hypertensive heart and that lead to the structural remodeling of the myocardium. In particular, we focus on the diagnosis and treatment of myocardial interstitial fibrosis, likely the lesion most critically involved in the transition from subclinical HHD to clinically overt heart failure. In this context, it is worth considering whether the use of imaging and circulating biomarkers for the noninvasive diagnosis of myocardial interstitial fibrosis should be incorporated in the medical study of hypertensive patients, especially of those with HHD.
Collapse
Affiliation(s)
- Javier Díez
- Center for Applied Medical Research (CIMA), and School of Medicine, Universidad de Navarra, and Center for Network Biomedical Research of Cardiovascular Diseases (CIBERCV), Carlos III Institute of Health, Madrid, Spain (J.D.)
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, and Department of Medicine, University of Mississippi School of Medicine, Jackson (J.B.)
| |
Collapse
|
39
|
Morfino P, Aimo A, Castiglione V, Gálvez-Montón C, Emdin M, Bayes-Genis A. Treatment of cardiac fibrosis: from neuro-hormonal inhibitors to CAR-T cell therapy. Heart Fail Rev 2023; 28:555-569. [PMID: 36221014 PMCID: PMC9553301 DOI: 10.1007/s10741-022-10279-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/03/2022] [Indexed: 02/05/2023]
Abstract
Cardiac fibrosis is characterized by the deposition of extracellular matrix proteins in the spaces between cardiomyocytes following both acute and chronic tissue damage events, resulting in the remodeling and stiffening of heart tissue. Fibrosis plays an important role in the pathogenesis of many cardiovascular disorders, including heart failure and myocardial infarction. Several studies have identified fibroblasts, which are induced to differentiate into myofibroblasts in response to various types of damage, as the most important cell types involved in the fibrotic process. Some drugs, such as inhibitors of the renin-angiotensin-aldosterone system, have been shown to be effective in reducing cardiac fibrosis. There are currently no drugs with primarily anti-fibrotic action approved for clinical use, as well as the evidence of a clinical efficacy of these drugs is extremely limited, despite the numerous encouraging results from experimental studies. A new approach is represented by the use of CAR-T cells engineered in vivo using lipid nanoparticles containing mRNA coding for a receptor directed against the FAP protein, expressed by cardiac myofibroblasts. This strategy has proved to be safe and effective in reducing myocardial fibrosis and improving cardiac function in mouse models of cardiac fibrosis. Clinical studies are required to test this novel approach in humans.
Collapse
Affiliation(s)
- Paolo Morfino
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Alberto Aimo
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | - Vincenzo Castiglione
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Carolina Gálvez-Montón
- Institut del Cor, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Michele Emdin
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Antoni Bayes-Genis
- Institut del Cor, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
40
|
Feng WH, Chen PS, Chung HC, Lin YH, Li YH. Soluble tumor endothelial marker 1 in heart failure with reduced ejection fraction: A pilot study. Front Cardiovasc Med 2022; 9:1015471. [PMID: 36588577 PMCID: PMC9802008 DOI: 10.3389/fcvm.2022.1015471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Background Tumor endothelial marker 1 (TEM1/CD248) is a transmembrane protein that expresses in mesenchymal lineage derived cells during embryogenesis and becomes undetectable in normal adults after birth. Re-expression of TEM1 is found in organ fibrosis, wound healing and cardiac remodeling indicating its potential role in heart failure (HF). The purpose of this study is to explore the role of soluble TEM1 (sTEM1) in patients with HF with reduced ejection fraction. Methods We examined endomyocardial biopsy specimens from three HF patients and blood samples from 48 patients admitted for acute decompensated HF (age 72 years, men 61.7%). The expression of TEM1 in cardiac tissue and concentrations of sTEM1 in plasma were evaluated. Cultured rat cardiomyocytes (H9c2) and human cardiac fibroblasts (HCF) were stimulated with hypoxia or transforming growth factor beta (TGF-β) to observe the release of sTEM1 into culture media. The conditioned media of hypoxia-stimulated H9c2 cells was harvested and added into cultured cardiac fibroblast to evaluate its biological effect. Results Immunofluorescence study of biopsy specimens from three HF patients showed TEM1 expression in cardiomyocytes and cardiac fibroblasts. The plasma level of sTEM1 was significantly higher in patients (0.90 ± 0.23 vs. 0.33 ± 0.10 ng/mL, p = 0.032) with LVEF ≤ 35% compared with those with LVEF 36-49%. The sTEM1 levels had correlations with HF biomarkers of cardiac fibrosis, including growth differentiation factor-15 (GDF-15) and galectin-3. There was a significant increase in sTEM1 levels in the cultured media of H9c2 and HCF after being stressed with hypoxia or TGF-β. The conditioned media derived from hypoxia-stimulated H9c2 cells significantly increased cell proliferation of cardiac fibroblasts. This effect was partially reversed by anti-TEM1 antibody. Conclusion This pilot study demonstrated that cardiac TEM1 expression was upregulated in HF. The levels of sTEM1 were significantly higher in HF patients with LVEF ≤ 35% and correlated with other biomarkers of cardiac fibrosis. In vitro study proved that functional sTEM1 was released into cultured media after stressing cardiomyocytes and HCF.
Collapse
Affiliation(s)
- Wen-Han Feng
- College of Medicine, Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan,Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Sheng Chen
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Hsing-Chun Chung
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Hsiung Lin
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yi-Heng Li
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan,*Correspondence: Yi-Heng Li,
| |
Collapse
|
41
|
Proteomic Insights into Cardiac Fibrosis: From Pathophysiological Mechanisms to Therapeutic Opportunities. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248784. [PMID: 36557919 PMCID: PMC9781843 DOI: 10.3390/molecules27248784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic process in nearly all forms of heart disease which refers to excessive deposition of extracellular matrix proteins by cardiac fibroblasts. Activated fibroblasts are the central cellular effectors in cardiac fibrosis, and fibrotic remodelling can cause several cardiac dysfunctions either by reducing the ejection fraction due to a stiffened myocardial matrix, or by impairing electric conductance. Recently, there is a rising focus on the proteomic studies of cardiac fibrosis for pathogenesis elucidation and potential biomarker mining. This paper summarizes the current knowledge of molecular mechanisms underlying cardiac fibrosis, discusses the potential of imaging and circulating biomarkers available to recognize different phenotypes of this lesion, reviews the currently available and potential future therapies that allow individualized management in reversing progressive fibrosis, as well as the recent progress on proteomic studies of cardiac fibrosis. Proteomic approaches using clinical specimens and animal models can provide the ability to track pathological changes and new insights into the mechanisms underlining cardiac fibrosis. Furthermore, spatial and cell-type resolved quantitative proteomic analysis may also serve as a minimally invasive method for diagnosing cardiac fibrosis and allowing for the initiation of prophylactic treatment.
Collapse
|
42
|
Lv S, Zhang W, Yuan P, Lu C, Dong J, Zhang J. QiShenYiQi pill for myocardial collagen metabolism and apoptosis in rats of autoimmune cardiomyopathy. PHARMACEUTICAL BIOLOGY 2022; 60:722-728. [PMID: 35361037 PMCID: PMC8979511 DOI: 10.1080/13880209.2022.2056206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/14/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
CONTEXT QiShenYiQi pill (QSYQ) is a traditional Chinese medicine with a myocardial protective effect. OBJECTIVE To explore the effect of QSYQ on myocardial collagen metabolism in rats with autoimmune cardiomyopathy and explore the underlying mechanism from the aspect of apoptosis. MATERIALS AND METHODS We established an autoimmune cardiomyopathy model using Lewis rats. The rats were then randomly divided into six groups (n = 8): control, model, 3-methyladenine (15 mg/kg, intraperitoneal injection), QSYQ low-dose (135 mg/kg, gavage), QSYQ medium dose (270 mg/kg, gavage), and QSYQ high-dose (540 mg/kg, gavage) for four weeks. Van Gieson staining was applied for myocardial pathological characteristics, TUNEL fluorescence for myocardial cell apoptosis, enzyme-linked immunosorbent assay (ELISA) for serum PICP, PIIINP, and CTX-I levels, and western blot analysis for type I/III myocardial collagen, Bcl-2, Bax, and caspase-3 proteins. RESULTS Results showed that QSYQ (135, 270, or 540 mg/kg) significantly reduced the expression of myocardial type I/III collagen, and concentrations of serum PICP, PIIINP, and CTX-I in rats. Moreover, QSYQ could alleviate myocardial fibrosis more effectively at a higher dose. QSYQ could also inhibit myocardial apoptosis via downregulating Bcl-2 expression, and upregulating Bax and caspase-3 expression levels. DISCUSSION AND CONCLUSIONS The QSYQ can improve myocardial collagen metabolism by inhibiting apoptosis, which provides a potential therapeutic approach for autoimmune cardiomyopathy.
Collapse
Affiliation(s)
- Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
- Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin, China
| | - Wanqin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Peng Yuan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Chunmiao Lu
- Jiashan Hospital of Traditional Chinese Medicine, Zhejiang, China
| | | | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| |
Collapse
|
43
|
Sato R, Vatic M, da Fonseca GWP, von Haehling S. Sarcopenia and Frailty in Heart Failure: Is There a Biomarker Signature? Curr Heart Fail Rep 2022; 19:400-411. [PMID: 36261756 DOI: 10.1007/s11897-022-00575-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 10/24/2022]
Abstract
PURPOSE OF REVIEW Sarcopenia and frailty are common in patients with heart failure (HF) and are strongly associated with prognosis. This review aims to examine promising biomarkers that can guide physicians in identifying sarcopenia and frailty in HF. RECENT FINDINGS Traditional biomarkers including C-reactive protein, aminotransaminase, myostatin, and urinary creatinine as well as novel biomarkers including microRNAs, suppression of tumorigenicity 2 (ST2), galectin-3, and procollagen type III N-terminal peptide may help in predicting the development of sarcopenia and frailty in HF patients. Among those biomarkers, aminotransferase, urinary creatinine, and ST2 predicted the prognosis in HF patients with sarcopenia and frailty. This review outlines the current knowledge of biomarkers that are considered promising for diagnosing sarcopenia and frailty in HF. The listed biomarkers might support the diagnosis, prognosis, and therapeutic decisions for sarcopenia and frailty in HF patients.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075, Gottingen, Germany
| | - Mirela Vatic
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075, Gottingen, Germany
| | | | - Stephan von Haehling
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Gottingen, Germany.
| |
Collapse
|
44
|
Nezami Z, Holm H, Ohlsson M, Molvin J, Korduner J, Bachus E, Zaghi A, Dieden A, Platonov PG, Jujic A, Magnusson M. The impact of myocardial fibrosis biomarkers in a heart failure population with atrial fibrillation—The HARVEST-Malmö study. Front Cardiovasc Med 2022; 9:982871. [PMID: 36337899 PMCID: PMC9626526 DOI: 10.3389/fcvm.2022.982871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/30/2022] [Indexed: 11/26/2022] Open
Abstract
Background Several studies suggest that circulating biomarkers of myocardial fibrosis are associated with worse prognosis in subjects with atrial fibrillation (AF). Here, we aimed to explore associations between fibrosis biomarkers, prevalent AF, and left atrial volume (LAV) enlargement in subjects with heart failure (HF). Additionally, we evaluated the prognostic impact of fibrotic biomarkers in HF with co-existing AF. Materials and methods Patients hospitalized for HF (n = 316, mean age 75 years; 30% women) were screened for AF. Seven proteins previously associated with myocardial fibrosis [metalloproteinase inhibitor 4 (TIMP-4), suppression of tumorigenicity 2 (ST-2), galectin-3 (GAL-3), growth/differentiation factor-15 (GDF-15), and matrix metalloproteinase 2, 3, and 9 (MMP-3, MMP-3, and MMP-9, respectively)] were analyzed using a proximity extension assay. Proteins with significant Bonferroni-corrected associations with mortality and re-hospitalization risk were taken forward to multivariable Cox regression analyses. Further, Bonferroni-corrected multivariable logistic regression models were used to study associations between protein plasma levels, prevalent AF, and severely enlarged left atrial volume index (LAVI ≥ 48 ml/m2). Results Prevalent AF was observed in 194 patients at the hospitalization of whom 178 (92%) were re-hospitalized and 111 (57%) died during the follow-up period. In multivariable logistic regression models, increased plasma levels of TIMP-4, GDF-15, and ST-2 were associated with the prevalence of AF, whereas none of the seven proteins showed any significant association with severely enlarged LAVI. Increased plasma levels of five proteins yielded significant associations with all-cause mortality in patients with co-existing AF; TIMP-4 (HR 1.33; CI95% 1.07–1.66; p = 0.010), GDF-15 (HR 1.30; CI95% 1.05–1.62; p = 0.017), GAL-3 (HR 1.29; CI95% 1.03–1.61; p = 0.029), ST-2 (HR 1.48; CI95% 1.18–1.85; p < 0.001), and MMP-3 (HR 1.33; CI95% 1.09–1.63; p = 0.006). None of the proteins showed any significant association with re-hospitalization risk. Conclusion In this study, we were able to demonstrate that elevated levels of three plasma proteins previously linked to myocardial fibrosis are associated with prevalent AF in a HF population. Additionally, higher levels of five plasma proteins yielded an increased risk of mortality in the HF population with or without co-existing AF.
Collapse
Affiliation(s)
- Zainu Nezami
- Department of Internal Medicine, Sweden Lund University, Skane University Hospital, Lund, Sweden
| | - Hannes Holm
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Lund University, Skane University Hospital, Malmö, Sweden
- *Correspondence: Hannes Holm,
| | - Marcus Ohlsson
- Department of Internal Medicine, Sweden Lund University, Skane University Hospital, Lund, Sweden
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - John Molvin
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Lund University, Skane University Hospital, Malmö, Sweden
| | - Johan Korduner
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Erasmus Bachus
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Amir Zaghi
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Anna Dieden
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - Amra Jujic
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Lund University, Skane University Hospital, Malmö, Sweden
| | - Martin Magnusson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Lund University, Skane University Hospital, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Hypertension in Africa Research Team (HART), North West University Potchefstroom, Potchefstroom, South Africa
| |
Collapse
|
45
|
Cardiac fibrosis in oncologic therapies. CURRENT OPINION IN PHYSIOLOGY 2022; 29. [DOI: 10.1016/j.cophys.2022.100575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Zhu F, Li P, Sheng Y. Treatment of myocardial interstitial fibrosis in pathological myocardial hypertrophy. Front Pharmacol 2022; 13:1004181. [PMID: 36249793 PMCID: PMC9561344 DOI: 10.3389/fphar.2022.1004181] [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: 07/27/2022] [Accepted: 09/14/2022] [Indexed: 01/09/2023] Open
Abstract
Pathological myocardial hypertrophy can be caused by a variety of diseases, mainly accompanied by myocardial interstitial fibrosis (MIF), which is a diffuse and patchy process, appearing as a combination of interstitial micro-scars and perivascular collagen fiber deposition. Different stimuli may trigger MIF without cell death by activating a variety of fibrotic signaling pathways in mesenchymal cells. This manuscript summarizes the current knowledge about the mechanism and harmful outcomes of MIF in pathological myocardial hypertrophy, discusses the circulating and imaging biomarkers that can be used to identify this lesion, and reviews the currently available and potential future treatments that allow the individualized management of patients with pathological myocardial hypertrophy.
Collapse
Affiliation(s)
- Fuyu Zhu
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Peng Li
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Yanhui Sheng, ; Peng Li,
| | - Yanhui Sheng
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China,Department of Cardiology, Jiangsu Province Hospital, Nanjing, China,*Correspondence: Yanhui Sheng, ; Peng Li,
| |
Collapse
|
47
|
Zhu L, Wang Y, Zhao S, Lu M. Detection of myocardial fibrosis: Where we stand. Front Cardiovasc Med 2022; 9:926378. [PMID: 36247487 PMCID: PMC9557071 DOI: 10.3389/fcvm.2022.926378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Myocardial fibrosis, resulting from the disturbance of extracellular matrix homeostasis in response to different insults, is a common and important pathological remodeling process that is associated with adverse clinical outcomes, including arrhythmia, heart failure, or even sudden cardiac death. Over the past decades, multiple non-invasive detection methods have been developed. Laboratory biomarkers can aid in both detection and risk stratification by reflecting cellular and even molecular changes in fibrotic processes, yet more evidence that validates their detection accuracy is still warranted. Different non-invasive imaging techniques have been demonstrated to not only detect myocardial fibrosis but also provide information on prognosis and management. Cardiovascular magnetic resonance (CMR) is considered as the gold standard imaging technique to non-invasively identify and quantify myocardial fibrosis with its natural ability for tissue characterization. This review summarizes the current understanding of the non-invasive detection methods of myocardial fibrosis, with the focus on different techniques and clinical applications of CMR.
Collapse
Affiliation(s)
- Leyi Zhu
- State Key Laboratory of Cardiovascular Disease, Department of Magnetic Resonance Imaging, National Center for Cardiovascular Diseases, Fuwai Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Yining Wang
- State Key Laboratory of Cardiovascular Disease, Department of Magnetic Resonance Imaging, National Center for Cardiovascular Diseases, Fuwai Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shihua Zhao
- State Key Laboratory of Cardiovascular Disease, Department of Magnetic Resonance Imaging, National Center for Cardiovascular Diseases, Fuwai Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjie Lu
- State Key Laboratory of Cardiovascular Disease, Department of Magnetic Resonance Imaging, National Center for Cardiovascular Diseases, Fuwai Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Cardiovascular Imaging (Cultivation), Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Minjie Lu
| |
Collapse
|
48
|
Rationale and Protocol of the Multimodality Evaluation of Antibody-Mediated Injury in Heart Transplantation (LEONE-HT) Observational Cross-Sectional Study. Methods Protoc 2022; 5:mps5050075. [PMID: 36287047 PMCID: PMC9608829 DOI: 10.3390/mps5050075] [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: 07/31/2022] [Revised: 09/15/2022] [Accepted: 09/21/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction: Heart transplant (HT) survival has barely improved in the last decades, which is unsatisfactory for many HT recipients. The development of anti-human leukocyte antigen (anti-HLA) antibodies in HT patients is associated with a cardiac allograft dysfunction. The mechanisms leading to this damage are unclear. The Multimodality Evaluation Of Antibody-Mediated Injury In Heart Transplantation (LEONE-HT) study aimed to thoroughly describe the damage inflicted on the myocardium by anti-HLA antibodies. Methods and analysis: The LEONE-HT study is a cohort study with a cross-sectional approach in which HT patients with positive anti-HLA antibodies are compared with coetaneous HT patients with negative anti-HLA antibodies. All patients will undergo a state-of-the-art multimodal assessment, including imaging techniques, coronary anatomy and physiology evaluations and histological and immunological analyses. The individual and combined primary outcomes of structural graft injuries and longitudinal secondary outcomes are to be compared between the exposed and non-exposed groups with univariate and multivariable descriptive analyses. Ethics and dissemination: The LEONE-HT study is carried out in accordance with the principles set out in the Declaration of Helsinki and the International Conference on Harmonization guidelines for good clinical practice and following national laws and regulations. The study design, objectives and participant centers have been communicated to clinicaltrials.gov (NCT05184426). The LEONE-HT study counts on the support of patient associations to disseminate the objectives and results of the research. This study was funded by the Spanish Ministry of Science and Innovation and the Spanish Society of Cardiology.
Collapse
|
49
|
Assessment of Myocardial Diastolic Dysfunction as a Result of Myocardial Infarction and Extracellular Matrix Regulation Disorders in the Context of Mesenchymal Stem Cell Therapy. J Clin Med 2022; 11:jcm11185430. [PMID: 36143077 PMCID: PMC9502668 DOI: 10.3390/jcm11185430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
The decline in cardiac contractility due to damage or loss of cardiomyocytes is intensified by changes in the extracellular matrix leading to heart remodeling. An excessive matrix response in the ischemic cardiomyopathy may contribute to the elevated fibrotic compartment and diastolic dysfunction. Fibroproliferation is a defense response aimed at quickly closing the damaged area and maintaining tissue integrity. Balance in this process is of paramount importance, as the reduced post-infarction response causes scar thinning and more pronounced left ventricular remodeling, while excessive fibrosis leads to impairment of heart function. Under normal conditions, migration of progenitor cells to the lesion site occurs. These cells have the potential to differentiate into myocytes in vitro, but the changed micro-environment in the heart after infarction does not allow such differentiation. Stem cell transplantation affects the extracellular matrix remodeling and thus may facilitate the improvement of left ventricular function. Studies show that mesenchymal stem cell therapy after infarct reduces fibrosis. However, the authors did not specify whether they meant the reduction of scarring as a result of regeneration or changes in the matrix. Research is also necessary to rule out long-term negative effects of post-acute infarct stem cell therapy.
Collapse
|
50
|
New Insights into the Functions of MicroRNAs in Cardiac Fibrosis: From Mechanisms to Therapeutic Strategies. Genes (Basel) 2022; 13:genes13081390. [PMID: 36011301 PMCID: PMC9407613 DOI: 10.3390/genes13081390] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/16/2022] [Accepted: 08/03/2022] [Indexed: 02/06/2023] Open
Abstract
Cardiac fibrosis is a significant global health problem associated with almost all types of heart disease. Extensive cardiac fibrosis reduces tissue compliance and contributes to adverse outcomes, such as cardiomyocyte hypertrophy, cardiomyocyte apoptosis, and even heart failure. It is mainly associated with pathological myocardial remodeling, characterized by the excessive deposition of extracellular matrix (ECM) proteins in cardiac parenchymal tissues. In recent years, a growing body of evidence demonstrated that microRNAs (miRNAs) have a crucial role in the pathological development of cardiac fibrosis. More than sixty miRNAs have been associated with the progression of cardiac fibrosis. In this review, we summarized potential miRNAs and miRNAs-related regulatory mechanisms for cardiac fibrosis and discussed the potential clinical application of miRNAs in cardiac fibrosis.
Collapse
|