1
|
Lopes MES, Marcantonio CC, Salmon CR, Mofatto LS, Nociti Junior FH, Eick S, Deschner J, Cirelli JA, Nogueira AVB. Effects of periodontal disease on the proteomic profile of the periodontal ligament. J Proteomics 2025; 314:105384. [PMID: 39800186 DOI: 10.1016/j.jprot.2025.105384] [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: 09/19/2024] [Revised: 11/20/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Periodontal disease affects over 1 billion people globally. This study investigated how periodontitis affects the protein profile of the periodontal ligament (PDL) in rats. Eight Holtzman rats were divided into control and experimental periodontitis groups. The PDL was isolated using laser capture microdissection and protein extracts were analyzed by mass spectrometry. Data analysis utilized specialized software, and Gene Ontology enrichment analysis identified significant protein functions. The data are available via ProteomeXchange with identifier PXD055817. Proteins such as SerpinB1, C5, and Lgals3 were validated through immunohistochemistry, and their gene expression was examined in an in vitro human PDL cell line. This study identified 1326 proteins, with 156 unique to the control group, 294 unique to the periodontitis group, and 876 common to both groups. Enrichment analysis revealed that proteins associated with the regulation of enzyme activity and RNA binding were significantly represented in the periodontitis group. There were increased levels of SerpinB1, C5, and Lgals3 in the periodontitis group based on proteomic and immunohistochemical analyses. Furthermore, these targets showed increased gene expression in stimulated human PDL cells. This study provides insights into the periodontitis-related alterations in the protein composition of the PDL and PDL cells, identifying both novel and previously known disease-associated proteins. SIGNIFICANCE: The periodontal ligament plays a crucial role in oral functions by providing structural support to the tooth. Due to the presence of undifferentiated mesenchymal cells, research into its regenerative capacity is ongoing. Pathological conditions can affect these functions and protein composition. Currently, there is a lack of comprehensive research specifically focusing on evaluating the periodontal ligament in both healthy and diseased states. This pioneering study screened for protein alterations and the mechanisms related to periodontitis. The possibility of using proteomic analysis to evaluate the protein alterations that occur in periodontitis, a disease with a high global incidence, could provide therapeutic targets and new biomarkers for future clinical studies.
Collapse
Affiliation(s)
- Maria Eduarda Scordamaia Lopes
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
| | - Camila Chierici Marcantonio
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
| | - Cristiane Ribeiro Salmon
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, São Paulo, Brazil; Dental School, Centro Universitário N. Sra do Patrocínio - CEUNSP, Itu, São Paulo, Brazil
| | - Luciana Souto Mofatto
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | - Francisco Humberto Nociti Junior
- ADA Forsyth Institute, Cambridge, MA, USA; Dental School, São Leopoldo Mandic, Department of Research, Campinas, São Paulo, Brazil
| | - Sigrun Eick
- Laboratory of Oral Microbiology, Department of Periodontology, University of Bern, Bern, Switzerland
| | - James Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Joni Augusto Cirelli
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil.
| | - Andressa Vilas Boas Nogueira
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil; Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
2
|
Miyara S, Adler M, Umansky KB, Häußler D, Bassat E, Divinsky Y, Elkahal J, Kain D, Lendengolts D, Ramirez Flores RO, Bueno-Levy H, Golani O, Shalit T, Gershovits M, Weizman E, Genzelinakh A, Kimchi DM, Shakked A, Zhang L, Wang J, Baehr A, Petrover Z, Sarig R, Dorn T, Moretti A, Saez-Rodriguez J, Kupatt C, Tanaka EM, Medzhitov R, Krüger A, Mayo A, Alon U, Tzahor E. Cold and hot fibrosis define clinically distinct cardiac pathologies. Cell Syst 2025; 16:101198. [PMID: 39970910 PMCID: PMC11922821 DOI: 10.1016/j.cels.2025.101198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 09/28/2024] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
Fibrosis remains a major unmet medical need. Simplifying principles are needed to better understand fibrosis and to yield new therapeutic approaches. Fibrosis is driven by myofibroblasts that interact with macrophages. A mathematical cell-circuit model predicts two types of fibrosis: hot fibrosis driven by macrophages and myofibroblasts and cold fibrosis driven by myofibroblasts alone. Testing these concepts in cardiac fibrosis resulting from myocardial infarction (MI) and heart failure (HF), we revealed that acute MI leads to cold fibrosis whereas chronic injury (HF) leads to hot fibrosis. MI-driven cold fibrosis is conserved in pigs and humans. We computationally identified a vulnerability of cold fibrosis: the myofibroblast autocrine growth factor loop. Inhibiting this loop by targeting TIMP1 with neutralizing antibodies reduced myofibroblast proliferation and fibrosis post-MI in mice. Our study demonstrates the utility of the concepts of hot and cold fibrosis and the feasibility of a circuit-to-target approach to pinpoint a treatment strategy that reduces fibrosis. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Shoval Miyara
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Miri Adler
- Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Kfir B Umansky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel Häußler
- TUM School of Medicine and Health, Institute of Experimental Oncology and Therapy Research, Technical University of Munich, Munich, Germany
| | - Elad Bassat
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria; Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Yalin Divinsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob Elkahal
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - David Kain
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daria Lendengolts
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ricardo O Ramirez Flores
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
| | - Hanna Bueno-Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofra Golani
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Shalit
- The Mantoux Bioinformatics institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Gershovits
- The Mantoux Bioinformatics institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Eviatar Weizman
- The Mantoux Bioinformatics institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Genzelinakh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Danielle M Kimchi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Lingling Zhang
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jingkui Wang
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria; Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Andrea Baehr
- Klinik und Poliklinik für Innere Medizin I, University Clinic rechts der Isar, Technical University of Munich, Munich, Germany; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Zachary Petrover
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Rachel Sarig
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tatjana Dorn
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Alessandra Moretti
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany; European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridgeshire, UK
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, University Clinic rechts der Isar, Technical University of Munich, Munich, Germany; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Elly M Tanaka
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria; Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Ruslan Medzhitov
- Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, CT, USA; Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, Yale, New Haven, CT, USA
| | - Achim Krüger
- TUM School of Medicine and Health, Institute of Experimental Oncology and Therapy Research, Technical University of Munich, Munich, Germany
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
3
|
Misirlioglu NF, Orucoglu GG, Bıcakhan B, Kucuk SH, Himmetoglu S, Sayili SB, Ozen GD, Uzun H. Evaluation of Thrombomodulin, Heart-Type Fatty-Acid-Binding Protein, Pentraxin-3 and Galectin-3 Levels in Patients with Myocardial Infarction, with and Without ST Segment Elevation. J Clin Med 2025; 14:1015. [PMID: 39941683 PMCID: PMC11818096 DOI: 10.3390/jcm14031015] [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: 12/08/2024] [Revised: 01/19/2025] [Accepted: 02/02/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Medical history, ECG findings and cardiac markers are used in the diagnosis of myocardial infarction (MI). Biomarkers used especially for the diagnosis of MI include high-sensitivity troponins (hsTns), creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), myoglobin, cardiac myosin-binding protein C and new cardiac biomarkers. This study evaluated the levels of serum thrombomodulin (TM), heart-type fatty-acid-binding protein (H-FABP), pentraxin-3 (PTX-3) and galectin-3 (Gal-3) to determine their utility in distinguishing between ST-elevation myocardial infarction (STEMI) and non-ST-elevation myocardial infarction (NSTEMI). Methods: This study included a total of 180 patients (90 patients with acute STEMI and 90 patients with NSTEMI) who presented to the Gaziosmanpaşa Training and Research Hospital, Cardiovascular Surgery and Emergency Department, with ischemic chest pain lasting longer than 30 min. Ninety healthy volunteers were included as the control group. Results: Serum levels of N-terminal pro-brain natriuretic peptide (NT-proBNP), TM, H-FABP, PTX-3 and Gal-3 were significantly different across the STEMI, NSTEMI and control groups (p < 0.001). Strong positive correlations were observed between NT-proBNP and TM, H-FABP, PTX-3 and Gal-3 in the STEMI group. ROC analysis demonstrated excellent diagnostic accuracy for these biomarkers in distinguishing STEMI from NSTEMI and control groups. Conclusions: Vascular inflammation plays an important role in the pathophysiology of STEMI and NSTEMI. A comprehensive cardiac biomarker panel enhances diagnostic accuracy and risk stratification, particularly when distinguishing between STEMI and NSTEMI. The biomarkers hs-TnI, CK-MB, NT-proBNP, TM, H-FABP, PTX-3 and Gal-3 offer complementary information when used together as a panel. Further research and validation are essential to establish standardized protocols for their widespread use.
Collapse
Affiliation(s)
- Naile Fevziye Misirlioglu
- Department of Biochemistry, Gaziosmanpaşa Training and Research Hospital, University of Health Sciences, 34098 Istanbul, Turkey
| | - Gulbahar Guler Orucoglu
- Department of Emergency, Gaziosmanpaşa Training and Research Hospital, University of Health Sciences, 34098 Istanbul, Turkey;
| | - Burcu Bıcakhan
- Department of Cardiovascular Surgery, Gaziosmanpaşa Training and Research Hospital, University of Health Sciences, 34098 Istanbul, Turkey;
| | - Suat Hayri Kucuk
- Department of Biochemistry, Istanbul Physical Medicine and Rehabilitation Training and Research Hospital, University of Health Sciences, 34186 Istanbul, Turkey;
| | - Solen Himmetoglu
- Department of Medical Biochemistry, Faculty of Medicine, Biruni University, 34015 Istanbul, Turkey;
- Biruni University Research Center (B@MER), Biruni University, 34015 Istanbul, Turkey
| | - Sena Baykara Sayili
- Department of Emergency Medicine, Istanbul Training and Research Hospital, 34098 Istanbul, Turkey;
| | - Gulenay Defne Ozen
- Department of Psychology, McGill University, Montreal, QC H3A 1G1, Canada;
| | - Hafize Uzun
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Atlas University, 34403 Istanbul, Turkey;
| |
Collapse
|
4
|
Jiménez-González S, Delgado-Valero B, Romero-Miranda A, Islas F, Luaces M, Ramchandani B, Cuesta-Corral M, Montoro-Garrido A, Nieto ML, Martínez-Martínez E, Cachofeiro V. The mechanisms underlying the cardiac effects of modified citrus pectin in obese rats with myocardial ischemia: Role of galectin-3. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024:S0214-9168(24)00114-1. [PMID: 39638645 DOI: 10.1016/j.arteri.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Modified citrus pectin (MCP) is used as a nutritional supplement that inhibits galectin-3 activity, a central player in the cardiac damage associated with different pathological situations. In fact, we have previously observed that MCP improved cardiac function in obese infarcted rats that was associated with a reduction in cardiac fibrosis. Therefore, the aim of the present study was to further explore whether this effect could involve the modulation of gene expression of ECM components and their mediators as well as whether it could affect another two mechanisms involved in cardiac damage: mitochondrial dynamics and autophagic flux. METHODS Male Wistar rats were fed an atherogenic diet with a high content of saturated fat (35%). MI was induced by the ligation of left anterior descendant (LAD) coronary artery 6 weeks after and MCP (100mg/kg/day) or vehicle were administered for 4 weeks more. A group of rats fed a standard diet (5.3% fat) and subjected to a sham operation was used as controls. RESULTS Obese infarcted animals presented an increase in cross-linked collagen that was not affected by the administration of galectin-3 inhibitor. However, MCP reduced the increase in gene expression observed in obese infarcted rats of ECM components and mediators (collagen I, fibronectin, transforming growth factor-β and connective tissue growth factor), of components of endoplasmic reticulum stress (binding immunoglobulin protein, CCAAT-enhancer-binding homologous protein and activating transcription factor 4), of oxidative stress mediator (NADPH oxidase-4) and normalized those of the interleukin 33/ST2 system. MCP is also able to increase the levels of the mitochondrial protein Dynamin-1-like and those of both proteins involved in autophagic flux (p62 and LC3) that were reduced by the myocardial ischemia in the context of obesity. CONCLUSIONS The data show that the beneficial effect of the nutritional supplement MCP on the cardiac consequences associated with myocardial ischemia in the context of obesity could rely on its capacity to inhibit galectin-3 and to consequently modulate different downstream mechanisms, including inflammation, ER stress, oxidative stress, autophagy and mitochondrial function, which can facilitate fibrosis and cardiac remodeling in this pathological context.
Collapse
Affiliation(s)
- Sara Jiménez-González
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Beatriz Delgado-Valero
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Ana Romero-Miranda
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Fabian Islas
- Unidad de Imagen Cardíaca, Hospital General Universitario de Talavera de la Reina, Toledo, Spain
| | - María Luaces
- Servicio de Cardiología, Instituto Cardiovascular, Hospital Clínico San Carlos, Madrid, Spain
| | - Bunty Ramchandani
- Servicio de Cirugía Cardiaca Infantil, Hospital La Paz, Madrid, Spain
| | - María Cuesta-Corral
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Alejandro Montoro-Garrido
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - María Luisa Nieto
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Majadahonda, Spain; Institute of Molecular Biology and Genetics (IBGM-CSIC/Uva), (IBGM-CSIC/Uva), Valladolid, Spain
| | - Ernesto Martínez-Martínez
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain; Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Majadahonda, Spain.
| | - Victoria Cachofeiro
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain; Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Majadahonda, Spain.
| |
Collapse
|
5
|
Seropian IM, El-Diasty M, El-Sherbini AH, González GE, Rabinovich GA. Central role of Galectin-3 at the cross-roads of cardiac inflammation and fibrosis: Implications for heart failure and transplantation. Cytokine Growth Factor Rev 2024; 80:47-58. [PMID: 39482190 DOI: 10.1016/j.cytogfr.2024.10.002] [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: 09/02/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 11/03/2024]
Abstract
Cardiac inflammation and fibrosis are central pathogenic mechanisms leading to heart failure. Transplantation is still the treatment of choice for many patients undergoing end-stage heart failure who remain symptomatic despite optimal medical therapy. In spite of considerable progress, the molecular mechanisms linking inflammation, fibrosis and heart failure remain poorly understood. Galectin-3 (GAL3), a chimera-type member of the galectin family, has emerged as a critical mediator implicated in cardiac inflammatory, vascular and fibrotic processes through modulation of different cellular compartments including monocytes and macrophages, fibroblasts, endothelial cells and vascular smooth muscle cells via glycan-dependent or independent mechanisms. GAL3-driven circuits may hierarchically amplify cytokine production and function, immune cell activation and fibrosis cascades, influencing a wide range of cardiovascular disorders. Thus, GAL3 emerges as a potential therapeutic target to counteract aberrant inflammation and fibrosis during heart failure and a potential biomarker of heart failure and clinical outcome of heart transplantation.
Collapse
Affiliation(s)
- Ignacio M Seropian
- Servicio de Hemodinamia y Cardiología Intervencionista, Hospital Italiano de Buenos Aires, Ciudad de Buenos Aires C1199, Argentina; Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Medicina, Pontificia Universidad Católica Argentina, Ciudad de Buenos Aires & Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Ciudad de Buenos Aires, Argentina
| | - Mohammad El-Diasty
- Harrington Heart and Vascular Institute, University Hospital Cleveland Medical Center, Cleveland, OH 44106, USA; Faculty of Health Sciences, Queen's University, Kingston, ON K7L 2V7, Canada
| | | | - Germán E González
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Medicina, Pontificia Universidad Católica Argentina, Ciudad de Buenos Aires & Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Ciudad de Buenos Aires, Argentina.
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad de Buenos Aires C1428, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, C1428, Argentina.
| |
Collapse
|
6
|
Xie Z, Xie H, Xie C, Yang S, Feng Y, Su Z, Tang T, Zhang B, Yang J, Wang Y, Huang L, Zhu H, Cao J, Jiang R, Li T, Lu W. A combined analysis of bulk RNA-seq and scRNA-seq was performed to investigate the molecular mechanisms associated with the occurrence of myocardial infarction. BMC Genomics 2024; 25:921. [PMID: 39363266 PMCID: PMC11448016 DOI: 10.1186/s12864-024-10813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Myocardial infarction (MI) induces complex transcriptional changes across diverse cardiac cell types. Single-cell RNA sequencing (scRNA-seq) provides an unparalleled ability to discern cellular diversity during infarction, yet the veracity of these discoveries necessitates confirmation. This investigation sought to elucidate MI mechanisms by integrating scRNA-seq and bulk RNA-seq data. METHODS Publicly available scRNA-seq (GSE136088) and bulk RNA-seq (GSE153485) data from mice MI models were analyzed. Cell types were annotated, and differential expression analysis conducted. Bulk RNA-seq underwent quality control, principal component analysis, and differential expression analysis. RESULTS In scRNA-seq data, the comparison between MI and sham groups unveiled a reduction in endothelial cell populations, but macrophages and monocytes increased. Within fibroblast subgroups, three distinct categories were discerned, with two exhibiting upregulation in MI. Notably, endothelial cells exhibited an elevated expression of genes associated with apoptosis and ferroptosis. In bulk RNA-seq analysis, distinct patterns emerged when comparing MI and sham groups. Specifically, six genes linked to endothelial ferroptosis exhibited heightened expression in MI group, thereby corroborating the scRNA-seq findings. Moreover, the examination of isolated cardiac macrophages from mice MI model revealed increased expression of Spp1, Col1a2, Col3a1, Ctsd, and Lgals3 compared to sham group, thus substantiating the dysregulation of macrophage apoptosis-related proteins following MI. CONCLUSION MI altered the transcriptomic landscapes of cardiac cells with increased expression of apoptotic genes. Moreover, the upregulation of macrophage apoptosis marker was confirmed within MI models. The presence of endothelial cell depletion and ferroptosis in MI has been demonstrated.
Collapse
Affiliation(s)
- Zheng Xie
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Huicong Xie
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Chen Xie
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Saichao Yang
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Yun Feng
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Zhaohai Su
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Tao Tang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Bilong Zhang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Jiangyong Yang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Yueting Wang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Ling Huang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Hengqing Zhu
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Jun Cao
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Rengui Jiang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Weiling Lu
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China.
| |
Collapse
|
7
|
Jiménez-González S, Delgado-Valero B, Islas F, Romero-Miranda A, Luaces M, Ramchandani B, Cuesta-Corral M, Montoro-Garrido A, Martínez-Martínez E, Cachofeiro V. The detrimental role of galectin-3 and endoplasmic reticulum stress in the cardiac consequences of myocardial ischemia in the context of obesity. FASEB J 2024; 38:e23818. [PMID: 38989572 DOI: 10.1096/fj.202400747r] [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: 04/03/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
The association between cardiac fibrosis and galectin-3 was evaluated in patients with acute myocardial infarction (MI). The role of galectin-3 and its association with endoplasmic reticulum (ER) stress activation in the progression of cardiovascular fibrosis was also evaluated in obese-infarcted rats. The inhibitor of galectin-3 activity, modified citrus pectin (MCP; 100 mg/kg/day), and the inhibitor of the ER stress activation, 4-phenylbutyric acid (4-PBA; 500 mg/kg/day), were administered for 4 weeks after MI in obese rats. Overweight-obese patients who suffered a first MI showed higher circulating galectin-3 levels, higher extracellular volume, and LV infarcted size, as well as lower E/e'ratio and LVEF compared with normal-weight patients. A correlation was observed between galectin-3 levels and extracellular volume. Obese-infarcted animals presented cardiac hypertrophy and reduction in LVEF, and E/A ratio as compared with control animals. They also showed an increase in galectin-3 gene expression, as well as cardiac fibrosis and reduced autophagic flux. These alterations were associated with ER stress activation characterized by enhanced cardiac levels of binding immunoglobulin protein, which were correlated with those of galectin-3. Both MCP and 4-PBA not only reduced cardiac fibrosis, oxidative stress, galectin-3 levels, and ER stress activation, but also prevented cardiac functional alterations and ameliorated autophagic flux. These results show the relevant role of galectin-3 in the development of diffuse fibrosis associated with MI in the context of obesity in both the animal model and patients. Galectin-3 in tandem with ER stress activation could modulate different downstream mechanisms, including inflammation, oxidative stress, and autophagy.
Collapse
Affiliation(s)
- Sara Jiménez-González
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Beatriz Delgado-Valero
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Fabian Islas
- Unidad de Imagen Cardíaca, Hospital General Universitario de Talavera de la Reina, Toledo, Spain
| | - Ana Romero-Miranda
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - María Luaces
- Servicio de Cardiología, Instituto Cardiovascular, Hospital Clínico San Carlos, Madrid, Spain
| | - Bunty Ramchandani
- Servicio de Cirugía Cardiaca Infantil, Hospital La Paz, Madrid, Spain
| | - María Cuesta-Corral
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Alejandro Montoro-Garrido
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Ernesto Martínez-Martínez
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Majadahonda, Spain
| | - Victoria Cachofeiro
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Majadahonda, Spain
| |
Collapse
|
8
|
Gao R, Lu Y, Zhang W, Zhang Z. The Application of Berberine in Fibrosis and the Related Diseases. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:753-773. [PMID: 38716621 DOI: 10.1142/s0192415x24500307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
The formation of fibrotic tissue, characterized by the excessive accumulation of extracellular matrix (ECM) components such as collagen and fibronectin, is a normal and crucial stage of tissue repair in all organs. The over-synthesis, deposition, and remodeling of ECM components lead to organ dysfunction, posing a significant medical burden. Berberine, an isoquinoline alkaloid, is commonly used in the treatment of gastrointestinal diseases. With the deepening of scientific research, it has been gradually discovered that berberine also plays an important role in fibrotic diseases. In this review, we systematically introduce the effective role of berberine in fibrosis-related diseases. Specifically, this paper aims to provide a comprehensive review of the therapeutic role of berberine in treating fibrosis in organs such as the heart, liver, lungs, and kidneys. By summarizing its various pathways and mechanisms of action, including the inhibition of the transforming growth factor-[Formula: see text]/Smad signaling pathway, PI3K/Akt signaling pathway, MAPK signaling pathway, RhoA/ROCK signaling, and mTOR/p70S6K signaling pathway, as well as its activation of the Nrf2-ARE signaling pathway, AMPK signaling pathway, phosphorylated Smad 2/3 and Smad 7, and other signaling pathways, this review offers additional evidence to support the treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Rongmao Gao
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, P. R. China
| | - Yuanyu Lu
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, P. R. China
| | - Wei Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610057, P. R. China
| | - Zhao Zhang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, P. R. China
| |
Collapse
|
9
|
Seropian IM, Cassaglia P, Miksztowicz V, González GE. Unraveling the role of galectin-3 in cardiac pathology and physiology. Front Physiol 2023; 14:1304735. [PMID: 38170009 PMCID: PMC10759241 DOI: 10.3389/fphys.2023.1304735] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Galectin-3 (Gal-3) is a carbohydrate-binding protein with multiple functions. Gal-3 regulates cell growth, proliferation, and apoptosis by orchestrating cell-cell and cell-matrix interactions. It is implicated in the development and progression of cardiovascular disease, and its expression is increased in patients with heart failure. In atherosclerosis, Gal-3 promotes monocyte recruitment to the arterial wall boosting inflammation and atheroma. In acute myocardial infarction (AMI), the expression of Gal-3 increases in infarcted and remote zones from the beginning of AMI, and plays a critical role in macrophage infiltration, differentiation to M1 phenotype, inflammation and interstitial fibrosis through collagen synthesis. Genetic deficiency of Gal-3 delays wound healing, impairs cardiac remodeling and function after AMI. On the contrary, Gal-3 deficiency shows opposite results with improved remodeling and function in other cardiomyopathies and in hypertension. Pharmacologic inhibition with non-selective inhibitors is also protective in cardiac disease. Finally, we recently showed that Gal-3 participates in normal aging. However, genetic absence of Gal-3 in aged mice exacerbates pathological hypertrophy and increases fibrosis, as opposed to reduced fibrosis shown in cardiac disease. Despite some gaps in understanding its precise mechanisms of action, Gal-3 represents a potential therapeutic target for the treatment of cardiovascular diseases and the management of cardiac aging. In this review, we summarize the current knowledge regarding the role of Gal-3 in the pathophysiology of heart failure, atherosclerosis, hypertension, myocarditis, and ischemic heart disease. Furthermore, we describe the physiological role of Gal-3 in cardiac aging.
Collapse
Affiliation(s)
- Ignacio M. Seropian
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
- Servicio de Hemodinamia, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Cassaglia
- Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Buenos Aires, Argentina
| | - Verónica Miksztowicz
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
| | - Germán E. González
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
- Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Buenos Aires, Argentina
| |
Collapse
|
10
|
Katsioupa M, Kourampi I, Oikonomou E, Tsigkou V, Theofilis P, Charalambous G, Marinos G, Gialamas I, Zisimos K, Anastasiou A, Katsianos E, Kalogeras K, Katsarou O, Vavuranakis M, Siasos G, Tousoulis D. Novel Biomarkers and Their Role in the Diagnosis and Prognosis of Acute Coronary Syndrome. Life (Basel) 2023; 13:1992. [PMID: 37895374 PMCID: PMC10608753 DOI: 10.3390/life13101992] [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: 09/04/2023] [Revised: 09/23/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
The burden of cardiovascular diseases and the critical role of acute coronary syndrome (ACS) in their progression underscore the need for effective diagnostic and prognostic tools. Biomarkers have emerged as crucial instruments for ACS diagnosis, risk stratification, and prognosis assessment. Among these, high-sensitivity troponin (hs-cTn) has revolutionized ACS diagnosis due to its superior sensitivity and negative predictive value. However, challenges regarding specificity, standardization, and interpretation persist. Beyond troponins, various biomarkers reflecting myocardial injury, neurohormonal activation, inflammation, thrombosis, and other pathways are being explored to refine ACS management. This review article comprehensively explores the landscape of clinically used biomarkers intricately involved in the pathophysiology, diagnosis, and prognosis of ACS (i.e., troponins, creatine kinase MB (CK-MB), B-type natriuretic peptides (BNP), copeptin, C-reactive protein (CRP), interleukin-6 (IL-6), d-dimers, fibrinogen), especially focusing on the prognostic role of natriuretic peptides and of inflammatory indices. Research data on novel biomarkers (i.e., endocan, galectin, soluble suppression of tumorigenicity (sST2), microRNAs (miRNAs), soluble oxidized low-density lipoprotein receptor-1 (sLOX-1), F2 isoprostanes, and growth differentiation factor 15 (GDF-15)) are further analyzed, aiming to shed light on the multiplicity of pathophysiologic mechanisms implicated in the evolution of ACS. By elucidating the complex interplay of these biomarkers in ACS pathophysiology, diagnosis, and outcomes, this review aims to enhance our understanding of the evolving trajectory and advancements in ACS management. However, further research is necessary to establish the clinical utility and integration of these biomarkers into routine practice to improve patient outcomes.
Collapse
Affiliation(s)
- Maria Katsioupa
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Islam Kourampi
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Vasiliki Tsigkou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Panagiotis Theofilis
- 1st Department of Cardiology, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (D.T.)
| | - Georgios Charalambous
- Department of Emergency Medicine, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - George Marinos
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Ioannis Gialamas
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Konstantinos Zisimos
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Artemis Anastasiou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Efstratios Katsianos
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Ourania Katsarou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.K.); (I.K.); (E.O.); (V.T.); (I.G.); (K.Z.); (A.A.); (E.K.); (K.K.); (O.K.); (M.V.)
| | - Dimitris Tousoulis
- 1st Department of Cardiology, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (D.T.)
| |
Collapse
|
11
|
Screever EM, Gorter TM, Willems TP, Aboumsallem JP, Suthahar N, Mahmoud B, van Veldhuisen DJ, de Boer RA, Meijers WC. Diffuse Myocardial Fibrosis on Cardiac Magnetic Resonance Imaging Is Related to Galectin-3 and Predicts Outcome in Heart Failure. Biomolecules 2023; 13:410. [PMID: 36979345 PMCID: PMC10046101 DOI: 10.3390/biom13030410] [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: 12/16/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
AIMS Ongoing adverse remodeling is a hallmark of heart failure (HF), which might be reflected by either focal or diffuse myocardial fibrosis. Therefore, in (pre)clinical settings, we used immunohistochemistry or cardiac magnetic resonance imaging (CMR) to investigate the association of (focal or diffuse) fibrosis with cardiac biomarkers and adverse events in HF. METHODS AND RESULTS In C57Bl/6J mice, we determined the presence and extent of myocardial fibrosis 6 weeks post-myocardial infarction (MI). Furthermore, we studied 159 outpatient HF patients who underwent CMR, and determined focal and diffuse fibrosis by late gadolinium enhancement (LGE) and post-contrast T1 time of the non-LGE myocardium, respectively. HF patients were categorized based on the presence of LGE, and by the median post-contrast T1 time. Kaplan-Meier and Cox regression analyses were used to determine the association of fibrosis with HF hospitalization and all-cause mortality. LGE was detected in 61 (38%) patients. Cardiac biomarker levels were comparable between LGE-positive and LGE-negative patients. LGE-positive patients with a short T1 time had elevated levels of both NT-proBNP and galectin-3 (1611 vs. 453 ng/L, p = 0.026 and 20 vs. 15 μg/L, p = 0.004, respectively). This was not observed in LGE-negative patients. Furthermore, a short T1 time in LGE-positive patients was associated with a higher risk of adverse events (log-rank p = 0.01). CONCLUSION This study implies that cardiac biomarkers reflect active remodeling of the non-infarcted myocardium of patients with focal myocardial scarring. Diffuse fibrosis, in contrast to focal scarring, might have a higher prognostic value regarding adverse outcomes in HF patients.
Collapse
Affiliation(s)
- Elles M. Screever
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Thomas M. Gorter
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Tineke P. Willems
- Department of Radiology, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Navin Suthahar
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Belend Mahmoud
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Dirk J. van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Wouter C. Meijers
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
12
|
Chalise U, Becirovic-Agic M, Rodriguez-Paar JR, Konfrst SR, de Morais SDB, Johnson CS, Flynn ER, Hall ME, Anderson DR, Cook LM, DeLeon-Pennell KY, Lindsey ML. Harnessing the Plasma Proteome to Mirror Current and Predict Future Cardiac Remodeling After Myocardial Infarction. J Cardiovasc Transl Res 2023; 16:3-16. [PMID: 36197585 PMCID: PMC9944212 DOI: 10.1007/s12265-022-10326-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 09/15/2022] [Indexed: 12/01/2022]
Abstract
To identify plasma proteins that mirror current and predict future remodeling after myocardial infarction (MI), we retrospectively interrogated plasma proteomes of day (D)0 control (n = 16) and D3 MI (n = 15) from C57BL/6 J mice (20 ± 1 months). A total of 165 unique proteins were correlated with cardiac physiology variables. We prospectively tested the hypothesis that candidates identified retrospectively would predict cardiac physiology at an extended timepoint (D7 MI) in a second cohort of mice (n = 4 ± 1 months). We also examined human plasma from healthy controls (n = 18) and patients 48 h after presentation for MI (n = 41). Retrospectively, we identified 5 strong reflectors of remodeling (all r ≥ 0.60 and p < 0.05). Prospectively, ApoA1, IgA, IL-17E, and TIMP-1 mirrored current and predicted future remodeling. In humans, cytokine-cytokine receptor signaling was the top enriched KEGG pathway for all candidates. In summary, we identified plasma proteins that serve as useful prognostic indicators of adverse remodeling and progression to heart failure.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68198, USA
| | - Mediha Becirovic-Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68198, USA
| | - Jocelyn R Rodriguez-Paar
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68198, USA
| | - Shelby R Konfrst
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68198, USA
| | - Sharon D B de Morais
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68198, USA
| | - Catherine S Johnson
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, 68198, USA
| | - Elizabeth R Flynn
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Michael E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Daniel R Anderson
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Leah M Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kristine Y DeLeon-Pennell
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC, 29425, USA
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, 29401, USA
| | - Merry L Lindsey
- School of Graduate Studies and Research, Meharry Medical College, 1005 Dr DB Todd Jr Blvd, Nashville, TN, 37208, USA.
- Nashville VA Medical Center, Nashville, TN, 37212, USA.
| |
Collapse
|
13
|
Djordjevic A, Zivkovic M, Boskovic M, Dekleva M, Stankovic G, Stankovic A, Djuric T. Variants Tagging LGALS-3 Haplotype Block in Association with First Myocardial Infarction and Plasma Galectin-3 Six Months after the Acute Event. Genes (Basel) 2022; 14:genes14010109. [PMID: 36672849 PMCID: PMC9859409 DOI: 10.3390/genes14010109] [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: 10/28/2022] [Revised: 12/09/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Galectin-3 is encoded by LGALS-3, located in a unique haplotype block in Caucasians. According to the Tagger server, rs4040064, rs11628437, and rs7159490 cover 82% (r2 > 0.8) of the genetic variance of this HapBlock. Our aims were to examine the association of their haplotypes with first myocardial infarction (MI), changes in left ventricular echocardiographic parameters over time, and impact on plasma galectin-3 and LGALS-3 mRNA in peripheral blood mononuclear cells, both 6 months post-MI. The study group consisted of 546 MI patients and 323 controls. Gene expression was assessed in 92 patients and plasma galectin-3 in 189 patients. Rs4040064, rs11628437, rs7159490, and LGALS-3 mRNA expression were detected using TaqMan® technology. Plasma galectin-3 concentrations were determined by the ELISA method. We found that the TGC haplotype could have a protective effect against MI (adjusted OR 0.19 [0.05-0.72], p = 0.015) and that the GAC haplotype had significantly higher galectin-3 concentrations (48.3 [37.3-59.4] ng/mL vs. 18.9 [14.5-23.4] ng/mL, p < 0.0001), both in males and compared to the referent haplotype GGC. Higher plasma Gal-3 was also associated with higher NYHA class and systolic dysfunction. Our results suggest that variants tagging LGALS-3 HapBlock could reflect plasma Gal-3 levels 6 months post-MI and may have a potential protective effect against MI in men. Further replication, validation, and functional studies are needed.
Collapse
Affiliation(s)
- Ana Djordjevic
- Department of Radiobiology and Molecular Genetics, “Vinca” Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
- Correspondence: ; Tel.: +381-113-408-566 or +381-116-447-485
| | - Maja Zivkovic
- Department of Radiobiology and Molecular Genetics, “Vinca” Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Maja Boskovic
- Department of Radiobiology and Molecular Genetics, “Vinca” Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Milica Dekleva
- Department of Cardiology, University Clinical Centre “Zvezdara”, 11120 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Goran Stankovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Department of Cardiology, Clinical Centre of Serbia, 11000 Belgrade, Serbia
| | - Aleksandra Stankovic
- Department of Radiobiology and Molecular Genetics, “Vinca” Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Tamara Djuric
- Department of Radiobiology and Molecular Genetics, “Vinca” Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| |
Collapse
|
14
|
Yang X, Li J, Hu X, Zhang Y, Kuang Y, Liu Y, Liu C, Gao H, Ma L, Tang J, Ma Q. Identification of PFKFB2 as a key gene for the transition from acute to old myocardial infarction in peripheral blood. Front Cardiovasc Med 2022; 9:993579. [PMID: 36561770 PMCID: PMC9763698 DOI: 10.3389/fcvm.2022.993579] [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: 07/26/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Objective This study aims to analyze the gene expression profile of peripheral blood in different stages of myocardial infarction (MI) by transcriptome sequencing, and to study the gene expression characteristics of peripheral blood after MI. Methods Differentially expressed genes (DEGs) and weighted gene co-expression network analysis (WGCNA) were used to identify genes and modules associated with old myocardial infarction (OMI). Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation were applied to analyze the potential functions of genes. Hub genes were identified by Random Forest Classifier. CIBERSORT was used to provide an estimate of the abundance of 22 immune cells in peripheral blood. Quantitative polymerase chain reaction (qPCR) was used to detect gene expression levels in clinical samples. The cellular components (CC) of peripheral blood were counted by an automatic hematology analyzer. Results Through differential gene analysis and co-expression network analysis, 11 candidate genes were obtained. A random forest classifier identified 10 hub genes. Immune cell distribution of peripheral blood was found that T cell CD4 memory resting, NK cells resting, Dendritic cells activated, Mast cells resting, Monocytes and Neutrophils were correlated with OMI. Spearman correlation analysis found that PFKFB2 is related to the above immune cells. Low expression of PFKFB2 in peripheral blood of OMI was detected in clinical samples, and the relationship between PFKFB2 and peripheral blood immune cell counts was analyzed, which showed monocytes were associated with PFKFB2 in our study. Conclusion PFKFB2 was low expressed in OMI, and related to the distribution of immune cells. PFKFB2 may play a key role in reflecting the transition from AMI to OMI, and predicting the distribution of immune cells, which provided a new perspective for improving myocardial fibrosis and adverse remodeling.
Collapse
Affiliation(s)
- Xiangyu Yang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Li
- Department of Reproductive Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyao Hu
- Department of Reproductive Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yinzhuang Zhang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Kuang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yubo Liu
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chenxi Liu
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Haodong Gao
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Li Ma
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jia Tang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qilin Ma
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Qilin Ma,
| |
Collapse
|
15
|
Hojda SE, Chis IC, Clichici S. Biomarkers in Pulmonary Arterial Hypertension. Diagnostics (Basel) 2022; 12:diagnostics12123033. [PMID: 36553040 PMCID: PMC9776459 DOI: 10.3390/diagnostics12123033] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe medical condition characterized by elevated pulmonary vascular resistance (PVR), right ventricular (RV) failure, and death in the absence of appropriate treatment. The progression and prognosis are strictly related to the etiology, biochemical parameters, and treatment response. The gold-standard test remains right-sided heart catheterization, but dynamic monitoring of systolic pressure in the pulmonary artery is performed using echocardiography. However, simple and easily accessible non-invasive assays are also required in order to monitor this pathology. In addition, research in this area is in continuous development. In recent years, more and more biomarkers have been studied and included in clinical guidelines. These biomarkers can be categorized based on their associations with inflammation, endothelial cell dysfunction, cardiac fibrosis, oxidative stress, and metabolic disorders. Moreover, biomarkers can be easily detected in blood and urine and correlated with disease severity, playing an important role in diagnosis, prognosis, and disease progression.
Collapse
|
16
|
Chen Y, Fu W, Zheng Y, Yang J, Liu Y, Qi Z, Wu M, Fan Z, Yin K, Chen Y, Gao W, Ding Z, Dong J, Li Q, Zhang S, Hu L. Galectin 3 enhances platelet aggregation and thrombosis via Dectin-1 activation: a translational study. Eur Heart J 2022; 43:3556-3574. [PMID: 35165707 PMCID: PMC9989600 DOI: 10.1093/eurheartj/ehac034] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/25/2021] [Accepted: 01/18/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS Galectin-3, a β-galactoside-binding lectin, is abnormally increased in cardiovascular disease. Plasma Galectin-3 receives a Class II recommendation for heart failure management and has been extensively studied for multiple cellular functions. The direct effects of Galectin-3 on platelet activation remain unclear. This study explores the direct effects of Galectin-3 on platelet activation and thrombosis. METHODS AND RESULTS A strong positive correlation between plasma Galectin-3 concentration and platelet aggregation or whole blood thrombus formation was observed in patients with coronary artery disease (CAD). Multiple platelet function studies demonstrated that Galectin-3 directly potentiated platelet activation and in vivo thrombosis. Mechanistic studies using the Dectin-1 inhibitor, laminarin, and Dectin-1-/- mice revealed that Galectin-3 bound to and activated Dectin-1, a receptor not previously reported in platelets, to phosphorylate spleen tyrosine kinase and thus increased Ca2+ influx, protein kinase C activation, and reactive oxygen species production to regulate platelet hyperreactivity. TD139, a Galectin-3 inhibitor in a Phase II clinical trial, concentration dependently suppressed Galectin-3-potentiated platelet activation and inhibited occlusive thrombosis without exacerbating haemorrhage in ApoE-/- mice, which spontaneously developed increased plasma Galectin-3 levels. TD139 also suppressed microvascular thrombosis to protect the heart from myocardial ischaemia-reperfusion injury in ApoE-/- mice. CONCLUSION Galectin-3 is a novel positive regulator of platelet hyperreactivity and thrombus formation in CAD. As TD139 has potent antithrombotic effects without bleeding risk, Galectin-3 inhibitors may have therapeutic advantages as potential antiplatelet drugs for patients with high plasma Galectin-3 levels.
Collapse
Affiliation(s)
- Yufei Chen
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wanrong Fu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunbo Zheng
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yangyang Liu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiyong Qi
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Meiling Wu
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, USA
| | - Kanhua Yin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yunfeng Chen
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Wen Gao
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhongren Ding
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianzeng Dong
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Li
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Liang Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Zhuang J, Zhang X, Liu Q, Zhu M, Huang X. Targeted delivery of nanomedicines for promoting vascular regeneration in ischemic diseases. Am J Cancer Res 2022; 12:6223-6241. [PMID: 36168632 PMCID: PMC9475455 DOI: 10.7150/thno.73421] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022] Open
Abstract
Ischemic diseases, the leading cause of disability and death, are caused by the restriction or blockage of blood flow in specific tissues, including ischemic cardiac, ischemic cerebrovascular and ischemic peripheral vascular diseases. The regeneration of functional vasculature network in ischemic tissues is essential for treatment of ischemic diseases. Direct delivery of pro-angiogenesis factors, such as VEGF, has demonstrated the effectiveness in ischemic disease therapy but suffering from several obstacles, such as low delivery efficacy in disease sites and uncontrolled modulation. In this review, we summarize the molecular mechanisms of inducing vascular regeneration, providing the guidance for designing the desired nanomedicines. We also introduce the delivery of various nanomedicines to ischemic tissues by passive or active targeting manner. To achieve the efficient delivery of nanomedicines in various ischemic diseases, we highlight targeted delivery of nanomedicines and controllable modulation of disease microenvironment using nanomedicines.
Collapse
Affiliation(s)
- Jie Zhuang
- School of Medicine, Nankai University, Tianjin 300071, China.,Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiangyun Zhang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qiqi Liu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mingsheng Zhu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
18
|
Wang Q, Huai W, Ye X, Pan Y, Yang X, Chen M, Ma QB, Gao Y, Zhang Y. Circulating plasma galectin-3 predicts new-onset atrial fibrillation in patients after acute myocardial infarction during hospitalization. BMC Cardiovasc Disord 2022; 22:392. [PMID: 36057558 PMCID: PMC9440583 DOI: 10.1186/s12872-022-02827-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Background New-onset atrial fibrillation (NOAF) is a common complication in patients with acute myocardial infarction (AMI) during hospitalization. Galectin-3 (Gal-3) is a novel inflammation marker that is significantly associated with AF. The association between post-AMI NOAF and Gal-3 during hospitalization is yet unclear. Objective The present study aimed to investigate the predictive value of plasma Gal-3 for post-AMI NOAF. Methods A total of 217 consecutive patients admitted with AMI were included in this retrospective study. Peripheral venous blood samples were obtained within 24 h after admission and plasma Gal-3 concentrations were measured. Results Post-AMI NOAF occurred in 18 patients in this study. Patients with NOAF were older (p < 0.001) than those without. A higher level of the peak brain natriuretic peptide (BNP) (p < 0.001) and Gal-3 (p < 0.001) and a lower low-density lipoprotein cholesterol level (LDL-C) (p = 0.030), and an estimated glomerular filtration rate (e-GFR) (p = 0.030) were recorded in patients with post-AMI NOAF. Echocardiographic information revealed that patients with NOAF had a significantly decreased left ventricular eject fraction (LVEF) (p < 0.001) and an increased left atrial diameter (LAD) (p = 0.004) than those without NOAF. The receiver operating characteristic (ROC) curve analysis revealed a significantly higher value of plasma Gal-3 in the diagnosis of NOAF for patients with AMI during hospitalization (area under the curve (p < 0.001), with a sensitivity of 72.22% and a specificity of 72.22%, respectively. Multivariate logistic regression model analysis indicated that age (p = 0.045), plasma Gal-3 (p = 0.018), and LAD (p = 0.014) were independent predictors of post-MI NOAF. Conclusions Plasma Gal-3 concentration is an independent predictor of post-MI NOAF.
Collapse
Affiliation(s)
- Qianhui Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Department of Cardiology, Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China
| | - Wei Huai
- Emergency Department, Third Clinical Medical College, Peking University, Beijing, 100191, China
| | - Xiaoguang Ye
- Heart Center and Beijing Key Laboratory of Hypertension, Department of Cardiology, Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China
| | - Yuxia Pan
- Heart Center and Beijing Key Laboratory of Hypertension, Department of Cardiology, Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China
| | - Xinchun Yang
- Heart Center and Beijing Key Laboratory of Hypertension, Department of Cardiology, Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China
| | - Mulei Chen
- Heart Center and Beijing Key Laboratory of Hypertension, Department of Cardiology, Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China
| | - Qing-Bian Ma
- Emergency Department, Third Clinical Medical College, Peking University, Beijing, 100191, China
| | - Yuanfeng Gao
- Heart Center and Beijing Key Laboratory of Hypertension, Department of Cardiology, Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China.
| | - Yuan Zhang
- Heart Center and Beijing Key Laboratory of Hypertension, Department of Cardiology, Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
19
|
Leancă SA, Crișu D, Petriș AO, Afrăsânie I, Genes A, Costache AD, Tesloianu DN, Costache II. Left Ventricular Remodeling after Myocardial Infarction: From Physiopathology to Treatment. Life (Basel) 2022; 12:1111. [PMID: 35892913 PMCID: PMC9332014 DOI: 10.3390/life12081111] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/11/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of death and morbidity worldwide, with an incidence relatively high in developed countries and rapidly growing in developing countries. The most common cause of MI is the rupture of an atherosclerotic plaque with subsequent thrombotic occlusion in the coronary circulation. This causes cardiomyocyte death and myocardial necrosis, with subsequent inflammation and fibrosis. Current therapies aim to restore coronary flow by thrombus dissolution with pharmaceutical treatment and/or intravascular stent implantation and to counteract neurohormonal activation. Despite these therapies, the injury caused by myocardial ischemia leads to left ventricular remodeling; this process involves changes in cardiac geometry, dimension and function and eventually progression to heart failure (HF). This review describes the pathophysiological mechanism that leads to cardiac remodeling and the therapeutic strategies with a role in slowing the progression of remodeling and improving cardiac structure and function.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Daniela Crișu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antoniu Octavian Petriș
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| | - Irina Afrăsânie
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antonia Genes
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Dan Nicolae Tesloianu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Irina Iuliana Costache
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| |
Collapse
|
20
|
Aimo A, Vergaro G, González A, Barison A, Lupón J, Delgado V, Richards AM, de Boer RA, Thum T, Arfsten H, Hülsmann M, Falcao-Pires I, Díez J, Foo RSY, Chan MYY, Anene-Nzelu CG, Abdelhamid M, Adamopoulos S, Anker SD, Belenkov Y, Ben Gal T, Cohen-Solal A, Böhm M, Chioncel O, Jankowska EA, Gustafsson F, Hill L, Jaarsma T, Januzzi JL, Jhund P, Lopatin Y, Lund LH, Metra M, Milicic D, Moura B, Mueller C, Mullens W, Núñez J, Piepoli MF, Rakisheva A, Ristić AD, Rossignol P, Savarese G, Tocchetti CG, van Linthout S, Volterrani M, Seferovic P, Rosano G, Coats AJS, Emdin M, Bayes-Genis A. Cardiac remodelling - Part 2: Clinical, imaging and laboratory findings. A review from the Study Group on Biomarkers of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2022; 24:944-958. [PMID: 35488811 DOI: 10.1002/ejhf.2522] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 11/06/2022] Open
Abstract
In patients with heart failure, the beneficial effects of drug and device therapies counteract to some extent ongoing cardiac damage. According to the net balance between these two factors, cardiac geometry and function may improve (reverse remodelling, RR) and even completely normalize (remission), or vice versa progressively deteriorate (adverse remodelling, AR). RR or remission predict a better prognosis, while AR has been associated with worsening clinical status and outcomes. The remodelling process ultimately involves all cardiac chambers, but has been traditionally evaluated in terms of left ventricular volumes and ejection fraction. This is the second part of a review paper by the Study Group on Biomarkers of the Heart Failure Association of the European Society of Cardiology dedicated to ventricular remodelling. This document examines the proposed criteria to diagnose RR and AR, their prevalence and prognostic value, and the variables predicting remodelling in patients managed according to current guidelines. Much attention will be devoted to RR in patients with heart failure with reduced ejection fraction because most studies on cardiac remodelling focused on this setting.
Collapse
Affiliation(s)
- Alberto Aimo
- Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Giuseppe Vergaro
- Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Arantxa González
- CIMA Universidad de Navarra, and IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Andrea Barison
- Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Josep Lupón
- Institut del Cor, Hospital Universitari Germans Trias i Pujol Badalona, Barcelona, Spain
| | - Victoria Delgado
- Institut del Cor, Hospital Universitari Germans Trias i Pujol Badalona, Barcelona, Spain
| | | | - Rudolf A de Boer
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Thomas Thum
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Henrike Arfsten
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Martin Hülsmann
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | | | - Javier Díez
- Center for Applied Medical Research, Pamplona, Spain
| | - Roger S Y Foo
- Department of Medicine, Yong Loo-Lin School of Medicine, National University Hospital, Singapore, Singapore
| | - Mark Yan Yee Chan
- Department of Medicine, Yong Loo-Lin School of Medicine, National University Hospital, Singapore, Singapore
| | - Chukwuemeka G Anene-Nzelu
- Department of Medicine, Yong Loo-Lin School of Medicine, National University Hospital, Singapore, Singapore
| | | | - Stamatis Adamopoulos
- 2nd Department of Cardiovascular Medicine, Onassis Cardiac Surgery Center, Athens, Greece
| | - Stefan D Anker
- Department of Cardiology (CVK), and Berlin Institute of Health Center for Regenerative Therapy (BCRT), German Center for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | | | - Tuvia Ben Gal
- Cardiology Department, Rabin Medical Center, Beilinson, Israel
| | | | - Michael Böhm
- University of the Saarland, Homburg/Saar, Germany
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases 'Prof. C.C. Iliescu' Bucharest, University of Medicine Carol Davila, Bucharest, Romania
| | - Ewa A Jankowska
- Institute of Heart Disases, Wroclaw Medical University, Wroclaw, Poland
| | - Finn Gustafsson
- Heart Centre, Department of Cardiology, Rigshospitalet - Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - James L Januzzi
- Massachusetts General Hospital and Baim Institute for Clinical Research, Boston, MA, USA
| | | | - Yuri Lopatin
- Volgograd State Medical University, Volgograd, Russia
| | - Lars H Lund
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Marco Metra
- Cardiology, ASST Spedali Civili; Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Davor Milicic
- University of Zagreb School of Medicine, Zagreb, Croatia
| | - Brenda Moura
- Faculty of Medicine, University of Porto, Porto, Portugal
- Cardiology Department, Porto Armed Forces Hospital, Porto, Portugal
| | | | | | - Julio Núñez
- Hospital Clínico Universitario de Valencia, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - Massimo F Piepoli
- Cardiology Division, Castelsangiovanni Hospital, Castelsangiovanni, Italy
| | - Amina Rakisheva
- Scientific Research Institute of Cardiology and Internal Medicine, Almaty, Kazakhstan
| | - Arsen D Ristić
- Department of Cardiology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Patrick Rossignol
- Université de Lorraine, Centre d'Investigations Cliniques-Plurithématique 1433 and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Gianluigi Savarese
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Carlo G Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Sophie van Linthout
- Berlin Institute of Health (BIH) at Charité - Universitätmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | | | - Petar Seferovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Giuseppe Rosano
- St. George's Hospitals, NHS Trust, University of London, London, UK
| | | | - Michele Emdin
- Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Antoni Bayes-Genis
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Institut del Cor, Hospital Universitari Germans Trias i Pujol Badalona, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Cheng Z, Cai K, Xu C, Zhan Q, Xu X, Xu D, Zeng Q. Prognostic Value of Serum Galectin-3 in Chronic Heart Failure: A Meta-Analysis. Front Cardiovasc Med 2022; 9:783707. [PMID: 35252382 PMCID: PMC8894589 DOI: 10.3389/fcvm.2022.783707] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To evaluate the association between serum galectin-3 and all-cause death (ACD) and cardiovascular death (CVD) in patients with chronic heart failure (CHF). Methods The PubMed and Embase databases and Clinical Trials Registry (www.clinicaltrials.gov) were searched for studies with data on serum galectin-3 and ACD and CVD in CHF patients. The hazard ratios (HRs) of ACD and CVD were calculated and presented with 95% CIs. HRs were pooled using fixed effects or random effects models when appropriate. Sensitivity analysis, meta-regression and subgroup analysis were applied to find the origin of heterogeneity. Visual inspection of Begg's funnel plot and Egger's test were performed to assess the possibility publication bias. Results Pooled data included the results from 6,440 patients from 12 studies in the meta-analysis. Higher serum galectin-3 was associated with a higher risk of ACD (HR, 1.38; 95% CI, 1.14–1.67) and CVD (HR, 1.13; 95% CI, 1.02–1.25) in CHF patients. In the subgroup analyses, higher serum galectin-3 was associated with an increased risk of ACD in all subgroups. The pooled HR of the shorter follow-up group (1.78; 95% CI, 1.50–2.11) was significantly higher than the pooled HR of the longer follow-up group (1.15; 95% CI, 1.05–1.25). Sensitivity analysis of eliminating one study in each turn indicated that Koukoui et al.'s study had the largest influence on the risk of all-cause death. All-cause death publication bias was not detected (Pr>|z| = 0.35 for Begg's test and P>|t| = 0.15 for Egger's test). Conclusions Serum galectin-3 has prognostic value of both all-cause death and cardiovascular death in CHF. Serum galectin-3 could be useful for risk classification in patients with CHF. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=193399.
Collapse
Affiliation(s)
- Zhendong Cheng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Kefeng Cai
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chaoxian Xu
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qiong Zhan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Xingbo Xu
- Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August-University, Göttingen, Germany
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
- Dingli Xu
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
- *Correspondence: Qingchun Zeng
| |
Collapse
|
22
|
Banaszkiewicz M, Gąsecka A, Darocha S, Florczyk M, Pietrasik A, Kędzierski P, Piłka M, Torbicki A, Kurzyna M. Circulating Blood-Based Biomarkers in Pulmonary Hypertension. J Clin Med 2022; 11:jcm11020383. [PMID: 35054082 PMCID: PMC8779219 DOI: 10.3390/jcm11020383] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/01/2022] [Accepted: 01/09/2022] [Indexed: 12/23/2022] Open
Abstract
Pulmonary hypertension (PH) is a serious hemodynamic condition, characterized by increased pulmonary vascular resistance (PVR), leading to right heart failure (HF) and death when not properly treated. The prognosis of PH depends on etiology, hemodynamic and biochemical parameters, as well as on response to specific treatment. Biomarkers appear to be useful noninvasive tools, providing information about the disease severity, treatment response, and prognosis. However, given the complexity of PH, it is impossible for a single biomarker to be adequate for the broad assessment of patients with different types of PH. The search for novel emerging biomarkers is still ongoing, resulting in a few potential biomarkers mirroring numerous pathophysiological courses. In this review, markers related to HF, myocardial remodeling, inflammation, hypoxia and tissue damage, and endothelial and pulmonary smooth muscle cell dysfunction are discussed in terms of diagnosis and prognosis. Extracellular vesicles and other markers with complex backgrounds are also reviewed. In conclusion, although many promising biomarkers have been identified and studied in recent years, there are still insufficient data on the application of multimarker strategies for monitoring and risk stratification in PH patients.
Collapse
Affiliation(s)
- Marta Banaszkiewicz
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, 05-400 Warsaw, Poland; (S.D.); (M.F.); (P.K.); (M.P.); (A.T.); (M.K.)
- Correspondence:
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.G.); (A.P.)
| | - Szymon Darocha
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, 05-400 Warsaw, Poland; (S.D.); (M.F.); (P.K.); (M.P.); (A.T.); (M.K.)
| | - Michał Florczyk
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, 05-400 Warsaw, Poland; (S.D.); (M.F.); (P.K.); (M.P.); (A.T.); (M.K.)
| | - Arkadiusz Pietrasik
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.G.); (A.P.)
| | - Piotr Kędzierski
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, 05-400 Warsaw, Poland; (S.D.); (M.F.); (P.K.); (M.P.); (A.T.); (M.K.)
| | - Michał Piłka
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, 05-400 Warsaw, Poland; (S.D.); (M.F.); (P.K.); (M.P.); (A.T.); (M.K.)
| | - Adam Torbicki
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, 05-400 Warsaw, Poland; (S.D.); (M.F.); (P.K.); (M.P.); (A.T.); (M.K.)
| | - Marcin Kurzyna
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, 05-400 Warsaw, Poland; (S.D.); (M.F.); (P.K.); (M.P.); (A.T.); (M.K.)
| |
Collapse
|
23
|
Inflammatory Burden and Immunomodulative Therapeutics of Cardiovascular Diseases. Int J Mol Sci 2022; 23:ijms23020804. [PMID: 35054989 PMCID: PMC8775955 DOI: 10.3390/ijms23020804] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 02/04/2023] Open
Abstract
Phenotyping cardiovascular illness and recognising heterogeneities within are pivotal in the contemporary era. Besides traditional risk factors, accumulated evidence suggested that a high inflammatory burden has emerged as a key characteristic modulating both the pathogenesis and progression of cardiovascular diseases, inclusive of atherosclerosis and myocardial infarction. To mechanistically elucidate the correlation, signalling pathways downstream to Toll-like receptors, nucleotide oligomerisation domain-like receptors, interleukins, tumour necrosis factor, and corresponding cytokines were raised as central mechanisms exerting the effect of inflammation. Other remarkable adjuvant factors include oxidative stress and secondary ferroptosis. These molecular discoveries have propelled pharmaceutical advancements. Statin was suggested to confer cardiovascular benefits not only by lowering cholesterol levels but also by attenuating inflammation. Colchicine was repurposed as an immunomodulator co-administered with coronary intervention. Novel interleukin-1β and −6 antagonists exhibited promising cardiac benefits in the recent trials as well. Moreover, manipulation of gut microbiota and associated metabolites was addressed to antagonise inflammation-related cardiovascular pathophysiology. The gut-cardio-renal axis was therein established to explain the mutual interrelationship. As for future perspectives, artificial intelligence in conjunction with machine learning could better elucidate the sequencing of the microbiome and data mining. Comprehensively understanding the interplay between the gut microbiome and its cardiovascular impact will help identify future therapeutic targets, affording holistic care for patients with cardiovascular diseases.
Collapse
|
24
|
Novel multi-marker proteomics in phenotypically matched patients with ST-segment myocardial infarction: association with clinical outcomes. J Thromb Thrombolysis 2021; 53:841-850. [PMID: 34708315 DOI: 10.1007/s11239-021-02582-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/03/2021] [Indexed: 12/22/2022]
Abstract
Early prediction of significant morbidity or mortality in patients with acute ST-segment elevation myocardial infarction (STEMI) represents an unmet clinical need. In phenotypically matched population of 139 STEMI patients (72 cases, 67 controls) treated with primary percutaneous coronary intervention, we explored associations between a 24-h relative change from baseline in the concentration of 91 novel biomarkers and the composite outcome of death, heart failure, or shock within 90 days. Additionally, we used random forest models to predict the 90-day outcomes. After adjustment for false discovery rate, the 90-day composite was significantly associated with concentration changes in 14 biomarkers involved in various pathophysiologic processes including: myocardial fibrosis/remodeling (collagen alpha-1, cathepsin Z, metalloproteinase inhibitor 4, protein tyrosine phosphatase subunits), inflammation, angiogenesis and signaling (interleukin 1 and 2 subunits, growth differentiation factor 15, galectin 4, trefoil factor 3), bone/mineral metabolism (osteoprotegerin, matrix extracellular phosphoglycoprotein and tartrate-resistant acid phosphatase), thrombosis (tissue factor pathway inhibitor) and cholesterol metabolism (LDL-receptor). Random forest models suggested an independent association when inflammatory markers are included in models predicting the outcomes within 90 days. Substantial heterogeneity is apparent in the early proteomic responses among patients with acutely reperfused STEMI patients who develop death, heart failure or shock within 90 days. These findings suggest the need to consider synergistic multi-biomarker strategies for risk stratification and to inform future development of novel post-myocardial infarction therapies.
Collapse
|
25
|
Meijers WC, Bayes-Genis A, Mebazaa A, Bauersachs J, Cleland JGF, Coats AJS, Januzzi JL, Maisel AS, McDonald K, Mueller T, Richards AM, Seferovic P, Mueller C, de Boer RA. Circulating heart failure biomarkers beyond natriuretic peptides: review from the Biomarker Study Group of the Heart Failure Association (HFA), European Society of Cardiology (ESC). Eur J Heart Fail 2021; 23:1610-1632. [PMID: 34498368 PMCID: PMC9292239 DOI: 10.1002/ejhf.2346] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 08/13/2021] [Accepted: 09/07/2021] [Indexed: 12/18/2022] Open
Abstract
New biomarkers are being evaluated for their ability to advance the management of patients with heart failure. Despite a large pool of interesting candidate biomarkers, besides natriuretic peptides virtually none have succeeded in being applied into the clinical setting. In this review, we examine the most promising emerging candidates for clinical assessment and management of patients with heart failure. We discuss high-sensitivity cardiac troponins (Tn), procalcitonin, novel kidney markers, soluble suppression of tumorigenicity 2 (sST2), galectin-3, growth differentiation factor-15 (GDF-15), cluster of differentiation 146 (CD146), neprilysin, adrenomedullin (ADM), and also discuss proteomics and genetic-based risk scores. We focused on guidance and assistance with daily clinical care decision-making. For each biomarker, analytical considerations are discussed, as well as performance regarding diagnosis and prognosis. Furthermore, we discuss potential implementation in clinical algorithms and in ongoing clinical trials.
Collapse
Affiliation(s)
- Wouter C Meijers
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, CIBERCV, Barcelona, Spain
| | - Alexandre Mebazaa
- Inserm U942-MASCOT; Université de Paris; Department of Anesthesia and Critical Care, Hôpitaux Saint Louis & Lariboisière; FHU PROMICE, Paris, France.,Université de Paris, Paris, France.,Department of Anesthesia and Critical Care, Hôpitaux Saint Louis & Lariboisière, Paris, France.,FHU PROMICE, Paris, France
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - John G F Cleland
- Robertson Centre for Biostatistics and Clinical Trials, University of Glasgow; National Heart & Lung Institute, Imperial College London, London, UK
| | - Andrew J S Coats
- Monash University, Melbourne, Australia.,University of Warwick, Coventry, UK
| | | | | | | | - Thomas Mueller
- Department of Clinical Pathology, Hospital of Bolzano, Bolzano, Italy
| | - A Mark Richards
- Christchurch Heart Institute, Christchurch, New Zealand.,Cardiovascular Research Institute, National University of Singapore, Singapore
| | - Petar Seferovic
- Faculty of Medicine, Belgrade University, Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgarde, Serbia
| | | | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
26
|
Guo Q, Zhao Y, Li J, Huang C, Wang H, Zhao X, Wang M, Zhu W. Galectin-3 Derived from HucMSC Exosomes Promoted Myocardial Fibroblast-to-Myofibroblast Differentiation Associated with β-catenin Upregulation. Int J Stem Cells 2021; 14:320-330. [PMID: 33906979 PMCID: PMC8429944 DOI: 10.15283/ijsc20186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/01/2021] [Accepted: 02/01/2021] [Indexed: 01/01/2023] Open
Abstract
Background and Objectives Galectin-3 promotes fibroblast-to-myofibroblast differentiation and facilitates injury repair. Previous studies have shown that exosomes derived from human umbilical cord mesenchymal stem cells (hucMSC-ex) promote the differentiation of myocardial fibroblasts into myofibroblasts under inflammatory environment. Whether hucMSC-ex derived Galectin-3 (hucMSC-ex-Galectin-3) plays an important role in fibroblast-to-myofibroblast differentiation is the focus of this study. Methods and Results Galectin-3 was knocked-down by siRNA in hucMSCs, and then exosomes were extracted. Fibroblasts were treated with LPS, LPS+hucMSC-ex, LPS+negative control-siRNA-ex (NC-ex), or LPS+Galectin-3-siRNA-ex (si-ex) in vitro. The coronary artery of the left anterior descending (LAD) branch was permanently ligated, followed by intramyocardial injection with phosphate buffered saline(PBS), hucMSC-ex, hucMSC-NC-ex, or hucMSC-si-ex in vivo. Western blot, RT-PCR, and immunohistochemistry were used to detect the expression of markers related to fibroblast-to-myofibroblast differentiation and inflammatory factors. Migration and contraction functions of fibroblasts were evaluated using Transwell migration and collagen contraction assays, respectively. β-catenin expression was detected by western blot and immunofluorescence. The results showed that hucMSC-ex increased the protein expression of myofibroblast markers, anti-inflammatory factors, and β-catenin. HucMSC-ex also reduced the migration and promoted the contractility of fibroblasts. However, hucMSC-si-ex did not show these activities. Conclusions HucMSC-ex-Galectin-3 promoted the differentiation of cardiac fibroblasts into myofibroblasts in an inflammatory environment, which was associated with increased β-catenin levels.
Collapse
Affiliation(s)
- Qinyu Guo
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuanyuan Zhao
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jiejie Li
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Chao Huang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hua Wang
- Department of Obstetrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiangdong Zhao
- Department of Clinical Laboratory, Zhenjiang Provincial Blood Center, Zhenjiang, China
| | - Mei Wang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Wei Zhu
- School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
27
|
Kalstad AA, Myhre PL, Laake K, Opstad TB, Tveit A, Solheim S, Arnesen H, Seljeflot I. Biomarkers of ageing and cardiac remodeling are associated with atrial fibrillation. SCAND CARDIOVASC J 2021; 55:213-219. [PMID: 33650449 DOI: 10.1080/14017431.2021.1889653] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Objectives. Ageing is one of the strongest risk factors for atrial fibrillation (AF), and additional risk factors are also closely related to ageing. Remodeling is part of the pathophysiology of AF, and a possible common denominator of ageing and other AF risk factors. The aim of this study was to investigate any association between the presence of AF and the ageing biomarkers, leukocyte telomere length (LTL) and sirtuin-1 (SIRT-1), and the cardiac remodeling biomarkers Galectin-3 and sST2 in elderly myocardial infarction (MI) patients. Design. Patients were included after admission for MI. Diagnosis of AF was retrieved from medical records and classified as either history of AF before MI or new onset from admission to study inclusion. SIRT-1, sST2 and Galectin-3 were analyzed by ELISAs and LTL by qPCR. Results. In total, 299 patients were included, median age 75 years, 70.2% male. A history of AF was recorded in 38 patients and 30 patients experienced new onset AF. Higher levels of SIRT-1 were associated with lower risk of having a history of AF (OR = 0.46 (95% CI 0.26, 0.81), p = 0.007), whereas higher sST2 levels were associated with higher risk of AF (OR = 4.13 (95% CI 1.69, 10.13), p = 0.002). Results remained significant after adjustment for other AF risk factors. No significant associations with AF were found for Galectin-3 or LTL. None of the biomarkers associated with new onset AF. Conclusion. In elderly patients with MI, higher ST2 and lower SIRT-2 levels were associated with higher prevalence of AF, possibly reflecting both ageing and the remodeling phenomena in AF. Clinical trials registration: ClinicalTrials.gov (NCT01841944).
Collapse
Affiliation(s)
- Are A Kalstad
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Peder L Myhre
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Medicine, Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Kristian Laake
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Trine B Opstad
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Arnljot Tveit
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Medical Research, Vestre Viken Hospital Trust, Baerum Hospital, Gjettum, Norway
| | - Svein Solheim
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Harald Arnesen
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingebjørg Seljeflot
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
28
|
Karsiyaka Hendek M, Olgun E, Kisa U. The effect of initial periodontal treatment on gingival crevicular fluid galectin-3 levels in participants with periodontal disease. Aust Dent J 2021; 66:169-174. [PMID: 33378559 DOI: 10.1111/adj.12815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND The aim of study was to evaluate galectin-3 levels in gingival crevicular fluid (GCF) from periodontally healthy (H) patients and those with periodontitis (P), gingivitis (G) and the effect of initial periodontal treatment on GCF galectin-3 level. METHODS A total of 75 participants, 25 patients with periodontitis, 25 with gingivitis and 25 periodontally healthy subjects were included into the study. Patients with periodontal disease received initial periodontal treatment. GCF galectin-3 level was assessed at baseline and at the 6th-8th weeks after completion of periodontal treatment. GCF galectin-3 level was evaluated by enzyme-linked immunosorbent assay. RESULTS GCF galectin-3 level was the lowest in the H group (102.31[63.07] μg/30 s), followed by the G group (241.45[145.89] μg/30 s) and the highest in the P group (338.27[219.37] μg/30 s). These differences were statistically significant between H and the other groups (P < 0.001). After initial periodontal treatment, GCF galectin-3 level significantly decreased in the G and P groups compared to baseline values (P < 0.01). CONCLUSION The results of this study suggest that GCF galectin-3 level is a potential biomarker for the evaluation of gingival inflammation and initial periodontal treatment is effective in decreasing GCF galectin-3 level.
Collapse
Affiliation(s)
- M Karsiyaka Hendek
- Department of Periodontology, Faculty of Dentistry, Kirikkale University, Kirikkale, Turkey
| | - E Olgun
- Department of Periodontology, Faculty of Dentistry, Kirikkale University, Kirikkale, Turkey
| | - U Kisa
- Department of Biochemistry, Faculty of Medicine, Kirikkale University, Kirikkale, Turkey
| |
Collapse
|
29
|
Li F, Yang Y, Xue C, Tan M, Xu L, Gao J, Xu L, Zong J, Qian W. Zinc Finger Protein ZBTB20 protects against cardiac remodelling post-myocardial infarction via ROS-TNFα/ASK1/JNK pathway regulation. J Cell Mol Med 2020; 24:13383-13396. [PMID: 33063955 PMCID: PMC7701508 DOI: 10.1111/jcmm.15961] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 02/03/2023] Open
Abstract
This study aims to determine the efficacy of Zinc finger protein ZBTB20 in treatment of post‐infarction cardiac remodelling. For this purpose, left anterior descending (LAD) ligation was operated on mice to induce myocardial infarction (MI) with sham control group as contrast and adeno‐associated virus (AAV9) system was used to deliver ZBTB20 to mouse heart by myocardial injection with vehicle‐injected control group as contrast two weeks before MI surgery. Then four weeks after MI, vehicle‐treated mice with left ventricular (LV) remodelling underwent deterioration of cardiac function, with symptoms of hypertrophy, interstitial fibrosis, inflammation and apoptosis. The vehicle‐injected mice also showed increase of infarct size and decrease of survival rate. Meanwhile, the ZBTB20‐overexpressed mice displayed improvement after MI. Moreover, the anti‐apoptosis effect of ZBTB20 was further confirmed in H9c2 cells subjected to hypoxia in vitro. Further study suggested that ZBTB20 exerts cardioprotection by inhibiting tumour necrosis factor α/apoptosis signal‐regulating kinase 1 (ASK1)/c‐Jun N‐terminal kinase 1/2 (JNK1/2) signalling, which was confirmed by shRNA‐JNK adenoviruses transfection or a JNK activator in vitro as well as ASK1 overexpression in vivo. In summary, our data suggest that ZBTB20 could alleviate cardiac remodelling post‐MI. Thus, administration of ZBTB20 can be considered as a promising treatment strategy for heart failure post‐MI. Significance Statement: ZBTB20 could alleviate cardiac remodelling post‐MI via inhibition of ASK1/JNK1/2 signalling.
Collapse
Affiliation(s)
- Fangfang Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Yiming Yang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Chuanyou Xue
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Mengtong Tan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Lu Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Jianbo Gao
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Luhong Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Jing Zong
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Wenhao Qian
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
30
|
Zhang J, Dong L. Status and prospects: personalized treatment and biomarker for airway remodeling in asthma. J Thorac Dis 2020; 12:6090-6101. [PMID: 33209441 PMCID: PMC7656354 DOI: 10.21037/jtd-20-1024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Airway remodeling, as a major characteristic of bronchial asthma, is critical to the progression of this disease, whereas it is of less importance in clinical management. Complying with the current stepwise treatment standard for asthma, the choice of intervention on the clinical status is primarily determined by the patient’s treatment response to airway inflammation. However, a considerable number of asthmatic patients, especially severe asthmatic subjects, remain uncontrolled though they have undergone fortified anti-inflammation treatment. In the past few years, a growing number of biologics specific to asthma phenotypes have emerged, bringing new hope for patients with refractory asthma and severe asthma. While at the same time, the effect of airway remodeling on asthma treatment has become progressively prominent. In the era of personalized treatment, it has become one of the development directions for asthma treatment to find reliable airway remodeling biomarkers to assist in asthma phenotypes classification, and to further combine multiple phenotypes to accurately treat patients. In the present study, the research status of airway remodeling in asthma is reviewed to show the basis for classifying and treating such disease. Besides, several selected airway remodeling biomarkers and possibility to use them in individual treatment are discussed as well. This study considers that continuously optimized mechanisms and emerging biomarkers for airway remodeling in the future may further support individual therapy for asthma patients.
Collapse
Affiliation(s)
- Jintao Zhang
- Department of Respiratory and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liang Dong
- Department of Respiratory and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
31
|
Abstract
Galectins are an ancient family of lectins characterized by evolutionarily conserved amino acid sequences and β-galactoside recognition and binding sites. Galectin-3 (Gal-3) is one of 15 known galectins. This protein has important functions in numerous biological activities, including cardiac fibrosis and heart failure. In recent years, many studies have shown that Gal-3 is closely associated with acute myocardial infarction (AMI) and may be a promising biomarker for the assessment of severity as well as prognosis prediction in AMI patients, but controversy still exists. In this review, we summarize the latest literature on the relationship between Gal-3 and unstable plaques, the secretion kinetics of Gal-3 during the acute phase of AMI, and the value of Gal-3 in the prediction of post-AMI remodeling. Finally, the possible value of Gal-3 as a biological target for AMI therapy is examined.
Collapse
Affiliation(s)
- Mingxing Li
- Department of Cardiology, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Yong Yuan
- Department of Cardiology, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Kai Guo
- Department of Cardiology, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Yi Lao
- Department of Cardiology, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Xuansheng Huang
- Department of Cardiology, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Li Feng
- Department of Cardiology, Zhongshan People's Hospital, Zhongshan, Guangdong, China.
| |
Collapse
|
32
|
Wang C, Zhang C, Wu D, Guo L, Zhao F, Lv J, Fu L. Cholecystokinin octapeptide reduces myocardial fibrosis and improves cardiac remodeling in post myocardial infarction rats. Int J Biochem Cell Biol 2020; 125:105793. [PMID: 32554056 DOI: 10.1016/j.biocel.2020.105793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 06/03/2020] [Accepted: 06/13/2020] [Indexed: 12/09/2022]
Abstract
BACKGROUND/AIMS Myocardial infarction (MI) increases myocardial fibrosis (MF) and subsequent cardiac remodeling. Cholecystokinin octapeptide (CCK-8) is expressed in cardiomyocytes and plays an important role in cardiovascular regulation. In this study, we intend to use a rat model of myocardial infarction to evaluate the effects of CCK-8 on myocardial fibrosis and cardiac remodeling. METHODS Male Sprague-Dawley rats were separated into 3 groups: sham operation, MI + NaCl, and MI + CCK-8. All rats were subjected to left coronary artery ligation to induce MI or sham operation and then treated with CCK-8 or saline for 28 days. After 4 weeks, echocardiography was performed to assess cardiac function and myocardial fibrosis was evaluated using H&E and Masson's Trichrome-stained sections. The levels of BNP, CCK-8 in the plasma of all rats were detected by ELISA; RNA sequencing (RNA-seq) analysis was also adapted to detect differentially expressed genes in myocardial tissues of each group. Myocardial expression of fibrosis markers was analyzed by western blotting, immunohistochemistry and qRT-PCR. RESULTS CCK-8 was demonstrated to improve left ventricular function and results of H&E staining, Masson's trichrome staining, immunohistochemistry and western blotting showed that CCK-8 attenuated MF. Gene expression profiles of the left ventricles were analysed by RNA-seq and validated by qRT-PCR. Cardiac fibrosis genes were downregulated by CCK-8 in the left ventricle. SIGNIFICANCE CCK-8 can alleviate fibrosis in the noninfarcted regions and delay the left ventricular remodeling and the progress of heart failure in a MI rat model.
Collapse
Affiliation(s)
- Can Wang
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Cuili Zhang
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongdong Wu
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lu Guo
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fali Zhao
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinxin Lv
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lu Fu
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
33
|
Cassaglia P, Penas F, Betazza C, Fontana Estevez F, Miksztowicz V, Martínez Naya N, Llamosas MC, Noli Truant S, Wilensky L, Volberg V, Cevey ÁC, Touceda V, Cicale E, Berg G, Fernández M, Goren N, Morales C, González GE. Genetic Deletion of Galectin-3 Alters the Temporal Evolution of Macrophage Infiltration and Healing Affecting the Cardiac Remodeling and Function after Myocardial Infarction in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1789-1800. [PMID: 32473918 DOI: 10.1016/j.ajpath.2020.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/22/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022]
Abstract
We studied the role of galectin-3 (Gal-3) in the expression of alternative activation markers (M2) on macrophage, cytokines, and fibrosis through the temporal evolution of healing, ventricular remodeling, and function after myocardial infarction (MI). C57BL/6J and Gal-3 knockout mice (Lgals3-/-) were subjected to permanent coronary ligation or sham. We studied i) mortality, ii) macrophage infiltration and expression of markers of alternative activation, iii) cytokine, iv) matrix metalloproteinase-2 activity, v) fibrosis, and vi) cardiac function and remodeling. At 1 week post-MI, lack of Gal-3 markedly attenuated F4/80+ macrophage infiltration and significantly increased the expression of Mrc1 and Chil1, markers of M2 macrophages at the MI zone. Levels of IL-10, IL-6, and matrix metalloproteinase-2 were significantly increased, whereas tumor necrosis factor-α, transforming growth factor-β, and fibrosis were remarkably attenuated at the infarct zone. In Gal-3 knockout mice, scar thinning ratio, expansion, and cardiac remodeling and function were severely affected from the onset of MI. At 4 weeks post-MI, the natural evolution of fibrosis in Gal-3 knockout mice was also affected. Our results suggest that Gal-3 is essential for wound healing because it regulates the dynamics of macrophage infiltration, proinflammatory and anti-inflammatory cytokine expression, and fibrosis along the temporal evolution of MI in mice. The deficit of Gal-3 affected the dynamics of wound healing, thus aggravating the evolution of remodeling and function.
Collapse
Affiliation(s)
- Pablo Cassaglia
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Federico Penas
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Celeste Betazza
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina; Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina
| | - Florencia Fontana Estevez
- Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina
| | - Verónica Miksztowicz
- Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina; Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica-INFIBIOC, Laboratorio de Lípidos y Aterosclerosis, Buenos Aires, Argentina
| | - Nadia Martínez Naya
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - María Clara Llamosas
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Sofía Noli Truant
- Facultad de Farmacia y Bioquímica-CONICET, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, Argentina
| | - Luciana Wilensky
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Verónica Volberg
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Ágata C Cevey
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Vanessa Touceda
- Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica-INFIBIOC, Laboratorio de Lípidos y Aterosclerosis, Buenos Aires, Argentina
| | - Eliana Cicale
- Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriela Berg
- Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica-INFIBIOC, Laboratorio de Lípidos y Aterosclerosis, Buenos Aires, Argentina
| | - Marisa Fernández
- Facultad de Farmacia y Bioquímica-CONICET, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, Argentina
| | - Nora Goren
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Celina Morales
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Germán E González
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina; Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina.
| |
Collapse
|
34
|
Lorenzo-Almorós A, Pello A, Aceña Á, Martínez-Milla J, González-Lorenzo Ó, Tarín N, Cristóbal C, Blanco-Colio LM, Martín-Ventura JL, Huelmos A, Gutiérrez-Landaluce C, López-Castillo M, Kallmeyer A, Cánovas E, Alonso J, López Bescós L, Egido J, Lorenzo Ó, Tuñón J. Galectin-3 Is Associated with Cardiovascular Events in Post-Acute Coronary Syndrome Patients with Type-2 Diabetes. J Clin Med 2020; 9:jcm9041105. [PMID: 32294902 PMCID: PMC7230213 DOI: 10.3390/jcm9041105] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction: Type-2 diabetes mellitus (T2DM) is associated with early and severe atherosclerosis. However, few biomarkers can predict cardiovascular events in this population. Methods: We followed 964 patients with coronary artery disease (CAD), assessing plasma levels of galectin-3, monocyte chemoattractant protein-1 (MCP-1), and N-terminal fragment of brain natriuretic peptide (NT-proBNP) at baseline. The secondary outcomes were acute ischemia and heart failure or death. The primary outcome was the combination of the secondary outcomes. Results. Two hundred thirty-two patients had T2DM. Patients with T2DM showed higher MCP-1 (144 (113–195) vs. 133 (105–173) pg/mL, p = 0.006) and galectin-3 (8.3 (6.5–10.5) vs. 7.8 (5.9–9.8) ng/mL, p = 0.049) levels as compared to patients without diabetes. Median follow-up was 5.39 years (2.81–6.92). Galectin-3 levels were associated with increased risk of the primary outcome in T2DM patients (Hazard ratio (HR) 1.57 (1.07–2.30); p = 0.022), along with a history of cerebrovascular events. Treatment with clopidogrel was associated with lower risk. In contrast, NT-proBNP and MCP-1, but not galectin-3, were related to increased risk of the event in nondiabetic patients (HR 1.21 (1.04–1.42); p = 0.017 and HR 1.23 (1.05–1.44); p = 0.012, respectively), along with male sex and age. Galectin-3 was also the only biomarker associated with the development of acute ischemic events and heart failure or death in T2DM patients, while, in nondiabetics, MCP-1 and NT-proBNP, respectively, were related to these events. Conclusion: In CAD patients, galectin-3 plasma levels are associated with cardiovascular events in patients with T2DM, and MCP-1 and NT-proBNP in those without T2DM.
Collapse
Affiliation(s)
- Ana Lorenzo-Almorós
- Department of Internal Medicine, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain;
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
| | - Ana Pello
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
| | - Álvaro Aceña
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
| | - Juan Martínez-Milla
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - Óscar González-Lorenzo
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - Nieves Tarín
- Department of Cardiology, Hospital Universitario de Móstoles, 28935 Madrid, Spain;
| | - Carmen Cristóbal
- Department of Cardiology, Hospital de Fuenlabrada, 28942 Madrid, Spain; (C.C.); (C.G.-L.)
- Department of Medicine, Rey Juan Carlos University, Alcorcón, 28943 Madrid, Spain; (J.A.); (L.L.B.)
| | - Luis M Blanco-Colio
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28040 Madrid, Spain
| | - José Luis Martín-Ventura
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28040 Madrid, Spain
| | - Ana Huelmos
- Department of Cardiology, Hospital Universitario Fundación Alcorcón, 28922 Madrid, Spain;
| | | | - Marta López-Castillo
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
| | - Andrea Kallmeyer
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - Ester Cánovas
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - Joaquín Alonso
- Department of Medicine, Rey Juan Carlos University, Alcorcón, 28943 Madrid, Spain; (J.A.); (L.L.B.)
- Department of Cardiology, Hospital de Getafe, 28905 Madrid, Spain
| | - Lorenzo López Bescós
- Department of Medicine, Rey Juan Carlos University, Alcorcón, 28943 Madrid, Spain; (J.A.); (L.L.B.)
| | - Jesús Egido
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de diabetes y enfermedades metabólicas asociadas (CIBERDEM), 28040 Madrid, Spain
| | - Óscar Lorenzo
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de diabetes y enfermedades metabólicas asociadas (CIBERDEM), 28040 Madrid, Spain
| | - Jose Tuñón
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28040 Madrid, Spain
- Correspondence:
| |
Collapse
|
35
|
New clues arising from hunt of saccharides binding to galectin 3 via 3D QSAR and docking studies. INFORMATICS IN MEDICINE UNLOCKED 2020. [DOI: 10.1016/j.imu.2020.100411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
36
|
Cardiac Chagas Disease: MMPs, TIMPs, Galectins, and TGF- β as Tissue Remodelling Players. DISEASE MARKERS 2019; 2019:3632906. [PMID: 31885735 PMCID: PMC6899287 DOI: 10.1155/2019/3632906] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/01/2019] [Indexed: 01/13/2023]
Abstract
A century after the discovery of Chagas disease, studies are still needed to establish the complex pathophysiology of this disease. However, it is known that several proteins and molecules are related to the establishment of this disease, its evolution, and the appearance of its different clinical forms. Metalloproteinases and their tissue inhibitors, galectins, and TGF-β are involved in the process of infection and consequently the development of myocarditis, tissue remodeling, and fibrosis upon infection with Trypanosoma cruzi. Thus, considering that the heart is one of the main target organs in Chagas disease, knowledge regarding the mechanisms of action of these molecules is essential to understand how they interact and trigger local and systemic reactions and, consequently, determine whether they contribute to the development of Chagas' heart disease. In this sense, it is believed that the inflammatory microenvironment caused by the infection alters the expression of these proteins favoring progression of the host-parasite cycle and thereby stimulating cardiac tissue remodeling mechanisms and fibrosis. The aim of this review was to gather information on metalloproteinases and their tissue inhibitors, galectins, and TGF-β and discuss how these molecules and their different interrelationships contribute to the development of Chagas' heart disease.
Collapse
|
37
|
Shirakawa K, Endo J, Kataoka M, Katsumata Y, Yoshida N, Yamamoto T, Isobe S, Moriyama H, Goto S, Kitakata H, Hiraide T, Fukuda K, Sano M. IL (Interleukin)-10-STAT3-Galectin-3 Axis Is Essential for Osteopontin-Producing Reparative Macrophage Polarization After Myocardial Infarction. Circulation 2019; 138:2021-2035. [PMID: 29967195 DOI: 10.1161/circulationaha.118.035047] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Both osteopontin (OPN) and galectin-3 have been implicated in phagocytic clearance of dead cells and reparative fibrosis during wound healing. CD206+ macrophages are involved in tissue repair through phagocytosis and fibrosis after myocardial infarction (MI). However, the relationship among OPN, galectin-3, and macrophage polarization in the context of MI remains unclear. METHODS The time course of Spp1 (encoding OPN) expression in the heart after MI showed a strong activation of Spp1 on day 3 after MI. To identify where in the body and in which cells the transcriptional activity of Spp1 increased after MI, we analyzed EGFP (enhanced green fluorescent protein)- Spp1 knockin reporter mice on day 3 after MI. RESULTS The transcriptional activity of Spp1 increased only in CD206+ macrophages in the infarct myocardium, and most of CD206+ macrophages have strong transcriptional activation of Spp1 after MI. The temporal expression pattern of Lgal3 (encoding galectin-3) in cardiac macrophages after MI was similar to that of Spp1, and OPN is almost exclusively produced by galectin-3hiCD206+ macrophages. Although both interleukin (IL)-4 and IL-10 were reported to promote CD206+ macrophage-mediated cardiac repair after MI, IL-10- but not IL-4-stimulated CD11b+Ly6G- cells could differentiate into OPN-producing galectin-3hiCD206+ macrophages and showed enhanced phagocytic ability. Inhibition of STAT3 tyrosine phosphorylation suppressed IL-10-induced expression of intracellular galectin-3 and transcriptional activation of Spp1. Knockdown of galectin-3 suppressed their ability to differentiate into OPN-producing cells, but not STAT3 activation. The tyrosine phosphorylation of STAT3 and the appearance rate of galectin-3hiCD206+ cells on cardiac CD11b+Ly6G- cells in Spp1 knockout mice were the same as those in wild-type mice. Spp1 knockout mice showed vulnerability to developing post-MI left ventricular chamber dilatation and the terminal deoxynucleo-tidyltransferase 2'-Deoxyuridine-5'-triphosphate nick-end labeling (TUNEL)-positive cells in the infarcted myocardium after MI remained higher in number in Spp1 knockout mice than in wild-type mice. CONCLUSIONS OPN is almost exclusively produced by galectin-3hiCD206+ macrophages, which specifically appear in the infarct myocardium after MI. The IL-10-STAT3-galectin-3 axis is essential for OPN-producing reparative macrophage polarization after myocardial infarction, and these macrophages contribute to tissue repair by promoting fibrosis and clearance of apoptotic cells. These results suggest that galectin-3 may contribute to reparative fibrosis in the infarct myocardium by controlling OPN levels.
Collapse
Affiliation(s)
- Kohsuke Shirakawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Jin Endo
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Masaharu Kataoka
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Yoshinori Katsumata
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Naohiro Yoshida
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.).,Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Japan (N.Y.)
| | - Tsunehisa Yamamoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Sarasa Isobe
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Hidenori Moriyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Shinichi Goto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Hiroki Kitakata
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Takahiro Hiraide
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.)
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (K.S., J.E., M.K., Y.K., N.Y., T.Y., S.I., H.M., S.G., H.K., T.H., K.F., M.S.).,Japan Science and Technology Agency, Tokyo, Japan (M.S.)
| |
Collapse
|
38
|
Association between Galectin-3 levels within central and peripheral venous blood, and adverse left ventricular remodelling after first acute myocardial infarction. Sci Rep 2019; 9:13145. [PMID: 31511537 PMCID: PMC6739356 DOI: 10.1038/s41598-019-49511-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/08/2019] [Indexed: 12/24/2022] Open
Abstract
Our study investigates association between Galectin-3 levels and adverse left ventricular remodelling (LVR) at six months. Fifty-seven patients following first acute myocardial infarction (AMI) were enrolled in this study and blood samples collected on day 1 from the femoral vein and artery, the right atrium near the coronary sinus and the aortic root, and on day 30, from the cubital vein. Patients with LVESV ≥20% at six months, were included in the LVR group. On day 1, Galectin-3 plasma levels in the femoral vein (10.34 ng/ml ± 3.81 vs 8.22 ng/ml ± 2.34, p = 0.01), and near coronary sinus (10.7 ng/ml ± 3.97 vs 8.41 ng/ml ± 2.56, p = 0.007) were higher in the LVR group. Positive correlations between Galectin-3 levels from aortic root and coronary sinus, aortic root and femoral vein, and coronary sinus and femoral vein, were observed in both groups. On day 30, Galectin-3 concentration in the cubital vein was an independent risk factor of LVR six months post-AMI, demonstrating 1.5-fold increased risk. Day-30 Galectin-3 also showed positive correlations with echocardiography parameters indicative of diastolic and systolic dysfunction. Determining Galectin-3 plasma concentration on day 30 following AMI could have beneficial prognostic value in predicting LVR.
Collapse
|
39
|
Yalta K, Yilmaz MB, Yalta T, Palabiyik O, Taylan G, Zorkun C. Late Versus Early Myocardial Remodeling After Acute Myocardial Infarction: A Comparative Review on Mechanistic Insights and Clinical Implications. J Cardiovasc Pharmacol Ther 2019; 25:15-26. [DOI: 10.1177/1074248419869618] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the setting of acute myocardial infarction (AMI), adverse myocardial remodeling (AMR) has been universally regarded as an early-onset phenomenon generally arising within the first few weeks (usually within days in the infarct zone) following myocardial injury. On the other hand, onset of cardiac morphological changes in this setting may potentially extend far beyond this time frame (usually beyond several months after the index AMI), suggesting a prolonged latent period in certain cases. In clinical practice, this delayed form of post-AMI remodeling, namely late AMR, has emerged as an interesting and underrecognized phenomenon with poorly understood mechanisms. Notably, systemic inflammation and associated growth factors seem to play a pivotal role in this setting. Accordingly, the present article primarily aims to discuss potential mechanisms and clinical implications of late AMR (in a comparative manner with its classical early counterpart) among AMI survivors along with a particular emphasis on potential benefits of certain anti-inflammatory strategies in this setting.
Collapse
Affiliation(s)
- Kenan Yalta
- Cardiology Department, Trakya University, Edirne, Turkey
| | | | - Tulin Yalta
- Pathology Department, Trakya University, Edirne, Turkey
| | | | - Gokay Taylan
- Cardiology Department, Trakya University, Edirne, Turkey
| | - Cafer Zorkun
- Cardiology Department, Trakya University, Edirne, Turkey
| |
Collapse
|
40
|
Myocardial Injury After Ischemia/Reperfusion Is Attenuated By Pharmacological Galectin-3 Inhibition. Sci Rep 2019; 9:9607. [PMID: 31270370 PMCID: PMC6610618 DOI: 10.1038/s41598-019-46119-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/17/2019] [Indexed: 12/19/2022] Open
Abstract
Although optimal therapy for myocardial infarction includes reperfusion to restore blood flow to the ischemic region, ischemia/reperfusion (IR) also initiates an inflammatory response likely contributing to adverse left ventricular (LV) extracellular matrix (ECM) remodeling. Galectin-3 (Gal-3), a β-galactoside-binding-lectin, promotes cardiac remodeling and dysfunction. Our aim is to investigate whether Gal-3 pharmacological inhibition using modified citrus pectin (MCP) improves cardiac remodeling and functional changes associated with IR. Wistar rats were treated with MCP from 1 day before until 8 days after IR (coronary artery ligation) injury. Invasive hemodynamics revealed that both LV contractility and LV compliance were impaired in IR rats. LV compliance was improved by MCP treatment 8 days after IR. Cardiac magnetic resonance imaging showed decreased LV perfusion in IR rats, which was improved with MCP. There was no difference in LV hypertrophy in MCP-treated compared to untreated IR rats. However, MCP treatment decreased the ischemic area as well as Gal-3 expression. Gal-3 blockade paralleled lower myocardial inflammation and reduced fibrosis. These novel data showing the benefits of MCP in compliance and ECM remodeling in IR reinforces previously published data showing the therapeutic potential of Gal-3 inhibition.
Collapse
|
41
|
Du X, Zhao W, Nguyen M, Lu Q, Kiriazis H. β-Adrenoceptor activation affects galectin-3 as a biomarker and therapeutic target in heart disease. Br J Pharmacol 2019; 176:2449-2464. [PMID: 30756388 PMCID: PMC6592856 DOI: 10.1111/bph.14620] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/11/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Myocardial fibrosis is a key histopathological component that drives the progression of heart disease leading to heart failure and constitutes a therapeutic target. Recent preclinical and clinical studies have implicated galectin-3 (Gal-3) as a pro-fibrotic molecule and a biomarker of heart disease and fibrosis. However, our knowledge is poor on the mechanism(s) that determine the blood level or regulate cardiac expression of Gal-3. Recent studies have demonstrated that enhanced β-adrenoceptor activity is a determinant of both circulating concentration and cardiac expression of Gal-3. Pharmacological or transgenic activation of β-adrenoceptors leads to increased blood levels of Gal-3 and up-regulated cardiac Gal-3 expression, effect that can be reversed with the use of β-adrenoceptor antagonists. Conversely, Gal-3 gene deletion confers protection against isoprenaline-induced cardiotoxicity and fibrogenesis. At the transcription level, β-adrenoceptor stimulation activates cardiac mammalian sterile-20-like kinase 1, a pivotal kinase of the Hippo signalling pathway, which is associated with Gal-3 up-regulation. Recent studies have suggested a role for the β-adrenoceptor-Hippo signalling pathway in the regulation of cardiac Gal-3 expression thereby contributing to the onset and progression of heart disease. This implies a therapeutic potential of the suppression of Gal-3 expression. In this review, we discuss the effects of β-adrenoceptor activity on Gal-3 as a biomarker and causative mediator in the setting of heart disease and point out pivotal knowledge gaps. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Xiao‐Jun Du
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Physiology and Pathophysiology, School of Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Wei‐Bo Zhao
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - My‐Nhan Nguyen
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - Qun Lu
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Cardiovascular Medicine, First HospitalXi'an Jiaotong University Health Science CenterXi'anChina
| | - Helen Kiriazis
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| |
Collapse
|
42
|
Zhang T, Cao S, Yang H, Li J. Prognostic impact of galectin-3 in chronic kidney disease patients: a systematic review and meta-analysis. Int Urol Nephrol 2019; 51:1005-1011. [PMID: 30963453 DOI: 10.1007/s11255-019-02123-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 03/04/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Galectin-3 as a β-galactoside-binding lectin, which has served important functions in numerous biological activities including cell growth, apoptosis, pre-mRNA splicing, differentiation, transformation, angiogenesis, inflammation, fibrosis, and host defense, may be used in prediction of clinical outcomes in CKD patients. However, the given results remain debatable and inconclusive. Hence, we performed a comprehensive meta-analysis to clarify the predictive value of galectin-3 in patients with CKD, especially ESRD patients going on dialysis. METHODS PubMed and Embase electronic databases were searched to identify eligible studies reporting the association between galectin-3 and adverse outcomes in CKD patients. We searched the literatures published October 2018 or earlier. We used both fix-effects and random-effects models to calculate the overall effect estimate. An I2 > 50% indicates at least moderate statistical heterogeneity. A sensitivity analysis and subgroup analysis were performed to find the origin of heterogeneity. RESULTS We ultimately enrolled five studies with a total of 5226 patients in this meta-analysis. The result showed that high galectin-3 levels were associated with increased risk of all-cause mortality and cardiovascular (CV) events in CKD patients. For every 1% increased in galectin-3, the risk of all-cause mortality increased by 37.9% (HR 1.379, 95% CI 1.090-1.744). Much more, the risk of CV events in CKD patients was also significantly increased (HR 1.054, 95% CI 1.007-1.102) with no statistical heterogeneity among the studies (I2 = 0.0%, p = 0.623). However, there was no statistical difference between the risk of all-cause mortality and galectin-3 in HD patients (HR 1.171, 95% CI 0.963-1.425). CONCLUSIONS Our meta-analysis suggests that high levels of galectin-3 may increase the risk of all-cause mortality and CV events in CKD patients, however, probably not a sensitive biomarker for outcomes in HD patients. Further studies were warranted to validate our findings.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300112, Tianjin, People's Republic of China
| | - Shili Cao
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300112, Tianjin, People's Republic of China
| | - Hongtao Yang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300112, Tianjin, People's Republic of China.
| | - Jing Li
- Department of Nephrology, First Central Hospital of Tianjin, 300192, Tianjin, People's Republic of China.
| |
Collapse
|
43
|
Li S, Li S, Hao X, Zhang Y, Deng W. Perindopril and a Galectin-3 Inhibitor Improve Ischemic Heart Failure in Rabbits by Reducing Gal-3 Expression and Myocardial Fibrosis. Front Physiol 2019; 10:267. [PMID: 30967790 PMCID: PMC6438875 DOI: 10.3389/fphys.2019.00267] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/28/2019] [Indexed: 11/13/2022] Open
Abstract
Objective: Ventricular remodeling is considered the basis of heart failure and is involved in myocardial fibrosis. This study aimed to assess perindopril and a galectin-3 inhibitor (modified citrus pectin, MCP) for their effects on ventricular remodeling and myocardial fibrosis in rabbits with ischemic heart failure. Methods: Rabbits were divided into sham, heart failure (model), MCP, and perindopril groups, respectively. A rabbit model of ischemic heart failure was established by ligating the anterior descending coronary artery. Then, the rabbits were orally administered MCP, perindopril, or saline (all at 2 ml/kg/d) for 4 weeks. Sham animals only underwent open heart surgery without further treatment. After 4 weeks, cardiac function was examined by ultrasound, and myocardial Gal-3, collagen type I, and collagen type III expression was assessed, at the gene and protein levels, by real-time PCR and Western-Blot, respectively; serum Gal-3 was detected by ELISA, and fibrosis in the infarct zone was evaluated by H&E and Masson staining. Results: In model animals, myocardial Gal-3, collagen type I, and collagen type III gene and protein expression levels were increased compared with control values, as well as serum Gal-3 amounts. Treatment with perindopril and MCP significantly alleviated the above effects, with no significant differences between the treatment groups. Pathological analyses showed that compared with model animals, treatment with MCP or perindopril resulted in relatively neatly arranged myocardial cells in the infarct zone, with significantly decreased fibrosis. Conclusion: Perindopril and the galectin-3 inhibitor MCP comparably improve ischemic heart failure in rabbits, by downregulating Gal-3 and reducing myocardial fibrosis.
Collapse
Affiliation(s)
- Sha Li
- Department of Examination Center, Hebei General Hospital, Shijiazhuang, China
| | - Shuren Li
- Department of Cardiovascular Division 1, Hebei General Hospital, Shijiazhuang, China
| | - Xiao Hao
- Department of Cardiovascular Division 1, Hebei General Hospital, Shijiazhuang, China
| | - Yuehua Zhang
- Department of Cardiovascular Division 1, Hebei General Hospital, Shijiazhuang, China
| | - Wenhao Deng
- Department of Cardiovascular Division 1, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
44
|
Seropian IM, González GE, Maller SM, Berrocal DH, Abbate A, Rabinovich GA. Galectin-1 as an Emerging Mediator of Cardiovascular Inflammation: Mechanisms and Therapeutic Opportunities. Mediators Inflamm 2018; 2018:8696543. [PMID: 30524200 PMCID: PMC6247465 DOI: 10.1155/2018/8696543] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/30/2018] [Indexed: 01/07/2023] Open
Abstract
Galectin-1 (Gal-1), an evolutionarily conserved β-galactoside-binding lectin, controls immune cell homeostasis and tempers acute and chronic inflammation by blunting proinflammatory cytokine synthesis, engaging T-cell apoptotic programs, promoting expansion of T regulatory (Treg) cells, and deactivating antigen-presenting cells. In addition, this lectin promotes angiogenesis by co-opting the vascular endothelial growth factor receptor (VEGFR) 2 signaling pathway. Since a coordinated network of immunomodulatory and proangiogenic mediators controls cardiac homeostasis, this lectin has been proposed to play a key hierarchical role in cardiac pathophysiology via glycan-dependent regulation of inflammatory responses. Here, we discuss the emerging roles of Gal-1 in cardiovascular diseases including acute myocardial infarction, heart failure, Chagas cardiomyopathy, pulmonary hypertension, and ischemic stroke, highlighting underlying anti-inflammatory mechanisms and therapeutic opportunities. Whereas Gal-1 administration emerges as a potential novel treatment option in acute myocardial infarction and ischemic stroke, Gal-1 blockade may contribute to attenuate pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Ignacio M. Seropian
- Servicio de Hemodinamia y Cardiología Intervencionista, Instituto de Medicina Cardiovascular, Hospital Italiano de Buenos Aires, Buenos Aires C1199, Argentina
| | - Germán E. González
- Instituto de Biología y Medicina Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiopatología Cardiovascular, Departamento de Patología, Universidad de Buenos Aires, Buenos Aires C1428, Argentina
| | - Sebastián M. Maller
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina
| | - Daniel H. Berrocal
- Servicio de Hemodinamia y Cardiología Intervencionista, Instituto de Medicina Cardiovascular, Hospital Italiano de Buenos Aires, Buenos Aires C1199, Argentina
| | - Antonio Abbate
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Gabriel A. Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428, Argentina
| |
Collapse
|
45
|
Left ventricular remodeling after the first myocardial infarction in association with LGALS-3 neighbouring variants rs2274273 and rs17128183 and its relative mRNA expression: a prospective study. Mol Biol Rep 2018; 45:2227-2236. [PMID: 30229476 DOI: 10.1007/s11033-018-4384-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/12/2018] [Indexed: 02/07/2023]
Abstract
Post-infarct left ventricular remodeling (LVR) process increases the risk of heart failure (HF). Circulating galectin-3 has been associated with fibrosis, inflammation and cardiac dysfunction during the remodeling process after myocardial infarction (MI). The aims of this prospective case study were to investigate the association of potentially functional variants in the vicinity of LGALS-3 locus, rs2274273 and rs17128183 with maladaptive LVR and whether these variants could affect LGALS-3 mRNA expression in peripheral blood mononuclear cells of patients 6 months after the first MI. This study encompassed 167 patients with acute MI that were followed up for 6 months. Evidence of LVR was obtained by repeated 2D Doppler echocardiography. Rs2274273, rs17128183 and LGALS-3 mRNA expression were detected by TaqMan® technology. Rs2274273 and rs17128183 rare allele bearing genotypes, according to the dominant model (CT+TT vs. CC and AG+GG vs. AA, respectively), were significantly and independently associated with maladaptive LVR (adjusted OR = 3.02, P = 0.016; adjusted OR = 3.14, P = 0.019, respectively) and higher LGALS-3 mRNA expression (fold induction 1.203, P = 0.03 and 1.214, P = 0.03, respectively). Our exploratory results suggest that rs2274273 and rs17128183 variants affect LGALS-3 mRNA and bear the risk for maladaptive LVR post-MI remodeling. Further replication and validation in a larger group of patients is inevitable.
Collapse
|
46
|
Asensio-Lopez MDC, Lax A, Fernandez Del Palacio MJ, Sassi Y, Hajjar RJ, Pascual-Figal DA. Pharmacological inhibition of the mitochondrial NADPH oxidase 4/PKCα/Gal-3 pathway reduces left ventricular fibrosis following myocardial infarction. Transl Res 2018; 199:4-23. [PMID: 29753686 DOI: 10.1016/j.trsl.2018.04.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 12/30/2022]
Abstract
Although the initial reparative fibrosis after myocardial infarction (MI) is crucial for preventing rupture of the ventricular wall, an exaggerated fibrotic response and reactive fibrosis outside the injured area are detrimental. Although metformin prevents adverse cardiac remodeling, as well as provides glycemic control, the underlying mechanisms remain poorly documented. This study describes the effect of mitochondrial NADPH oxidase 4 (mitoNox) and protein kinase C-alpha (PKCα) on the cardiac fibrosis and galectin 3 (Gal-3) expression. Randomly rats underwent MI, received metformin or saline solution. A model of biomechanical strain and co-culturewas used to enable cross talk between cardiomyocytes and fibroblasts. Long-term metformin treatment after MIwas associated with (1) a reduction in myocardial fibrosis and Gal-3 levels; (2) an increase in adenosine monophosphate-activated protein kinase (AMPK) α1/α2 levels; and (3) an inhibition of both mRNA expression and enzymatic activities of mitoNox and PKCα. These findings were replicated in the cellular model, where the silencing of AMPK expression blocked the ability of metformin to protect cardiomyocytes from strain. The use of specific inhibitors or small interference RNA provided evidence that PKCα is downstream of mitoNox, and that the activation of this pathway results in Gal-3 upregulation.The Gal-3 secreted by cardiomyocytes has a paracrine effect on cardiac fibroblasts, inducing their activation. In conclusion, a metformin-induced increase in AMPK improves myocardial remodeling post-MI, which is related to the inhibition of the mitoNox/PKCα/Gal-3 pathway. Manipulation of this pathway might offer new therapeutic options against adverse cardiac remodeling, in terms of preventing the activation of the present fibroblast population.
Collapse
Affiliation(s)
| | - Antonio Lax
- Biomedical Research Institute Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain.
| | | | - Yassine Sassi
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Domingo A Pascual-Figal
- Cardiology Department, Clinic and Universitary Hospital Virgen de la Arrixaca, Murcia, Spain; CIBER in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| |
Collapse
|
47
|
Liao Y, Chen K, Dong X, Li W, Li G, Huang G, Song W, Chen L, Fang Y. Berberine inhibits cardiac remodeling of heart failure after myocardial infarction by reducing myocardial cell apoptosis in rats. Exp Ther Med 2018; 16:2499-2505. [PMID: 30186485 PMCID: PMC6122433 DOI: 10.3892/etm.2018.6438] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 06/18/2018] [Indexed: 11/06/2022] Open
Abstract
The effects of berberine on cardiac function of heart failure after myocardial infarction and its possible mechanism were investigated. The anterior descending branches of 50 female Wistar rats were ligatured to establish the model of heart failure after myocardial infarction. At 4 weeks after successful modeling, the rats were randomly divided into two groups receiving 4-week gavage with saline (Sal group) and berberine (Ber group), while the sham-operation group (Sham group) was set up. After 4 weeks, the hemodynamics and serum BNP in rats were measured. The hearts of rats were taken to detect the degree of myocardial fibrosis. The myocardial cell apoptosis was detected. The expressions and changes in myocardial apoptosis-related proteins, including Bcl-2, Bax and caspase-3, were detected. The expression and changes in GRP78, CHOP and caspase-12 in myocardial tissue were detected. The results showed that Berberine improved the cardiac function of rats after myocardial infarction. After myocardial infarction, myocardial fibrosis and apoptosis were observed around the infarction area, berberine improved the myocardial fibrosis and reduced cell apoptosis. Furthermore, berberine alleviated endoplasmic reticulum stress (ERS) after myocardial infarction. In conclusion, Berberine can inhibit the myocardium cell apoptosis of heart failure after myocardial infarction, and its mechanism may be realized by affecting the ERS in myocardial tissue of heart failure after myocardial infarction and CHOP and caspase-12 apoptotic signaling pathway, upregulating Bcl-2/Bax expression and downregulating caspase-3 expression, thus inhibiting the cardiac remodeling and protecting the cardiac function.
Collapse
Affiliation(s)
- Ying Liao
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, Fujian 364000, P.R. China
| | - Kaihong Chen
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, Fujian 364000, P.R. China
| | - Xingmo Dong
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, Fujian 364000, P.R. China
| | - Weiguo Li
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, Fujian 364000, P.R. China
| | - Ganyang Li
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, Fujian 364000, P.R. China
| | - Guoyong Huang
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, Fujian 364000, P.R. China
| | - Wei Song
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, Fujian 364000, P.R. China
| | - Liling Chen
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, Fujian 364000, P.R. China
| | - Yong Fang
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, Fujian 364000, P.R. China
| |
Collapse
|
48
|
Zivlas C, Triposkiadis F, Psarras S, Giamouzis G, Skoularigis I, Chryssanthopoulos S, Kapelouzou A, Ramcharitar S, Barnes E, Papasteriadis E, Cokkinos D. Cystatin C and galectin-3 as therapeutic targets in heart failure. Ther Adv Cardiovasc Dis 2018; 12:233-235. [PMID: 29848191 DOI: 10.1177/1753944718778470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Christos Zivlas
- Wiltshire Cardiac Centre, Great Western Hospitals NHS Foundation Trusts, Marlborough Road, Swindon, SN3 6BB, UK
| | | | - Stelios Psarras
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Gregory Giamouzis
- Department of Cardiology, Larissa University Hospital, Larissa, Greece
| | | | | | | | - Steve Ramcharitar
- Wiltshire Cardiac Centre, Great Western Hospitals NHS Foundation Trusts, Swindon, UK
| | - Edward Barnes
- Wiltshire Cardiac Centre, Great Western Hospitals NHS Foundation Trusts, Swindon, UK
| | | | - Dennis Cokkinos
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
49
|
Nguyen MN, Su Y, Kiriazis H, Yang Y, Gao XM, McMullen JR, Dart AM, Du XJ. Upregulated galectin-3 is not a critical disease mediator of cardiomyopathy induced by β2-adrenoceptor overexpression. Am J Physiol Heart Circ Physiol 2018; 314:H1169-H1178. [DOI: 10.1152/ajpheart.00337.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preclinical studies have demonstrated that anti-galectin-3 (Gal-3) interventions are effective in attenuating cardiac remodeling, fibrosis, and dysfunction. We determined, in a transgenic (TG) mouse model of fibrotic cardiomyopathy, whether Gal-3 expression was elevated and whether Gal-3 played a critical role in disease development. We studied mice with fibrotic cardiomyopathy attributable to cardiac overexpression of human β2-adrenoceptors (β2-TG). Cardiac expression levels of Gal-3 and fibrotic or inflammatory genes were determined. The effect of Gal-3 inhibition in β2-TG mice was studied by treatment with Gal-3 inhibitors ( N-acetyllactosamine and modified citrus pectin) or by deletion of Gal-3 through crossing β2-TG and Gal-3 knockout mice. Changes in cardiomyopathy phenotypes were assessed by echocardiography and biochemical assays. In β2-TG mice at 3, 6, and 9 mo of age, upregulation of Gal-3 expression was observed at mRNA (~6- to 15-fold) and protein (~4- to 8-fold) levels. Treatment of β2-TG mice with N-acetyllactosamine (3 wk) or modified citrus pectin (3 mo) did not reverse cardiac fibrosis, inflammation, and cardiomyopathy. Similarly, Gal-3 gene deletion in β2-TG mice aged 3 and 9 mo did not rescue the cardiomyopathy phenotype. In conclusion, the β2-TG model of cardiomyopathy showed a robust upregulation of Gal-3 that correlated with disease severity, but Gal-3 inhibitors or Gal-3 gene deletion had no effect in halting myocardial fibrosis, remodeling, and dysfunction. Gal-3 may not be critical for cardiac fibrogenesis and remodeling in this cardiomyopathy model. NEW & NOTEWORTHY We showed a robust upregulation of cardiac galectin-3 (Gal-3) expression in a mouse model of cardiomyopathy attributable to cardiomyocyte-restricted transgenic activation of β2-adrenoceptors. However, pharmacological and genetic inhibition of Gal-3 did not confer benefit in this model, implying that Gal-3 may not be a critical disease mediator of cardiac remodeling in this model.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Yidan Su
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yan Yang
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Alfred Heart Centre, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Julie R. McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Anthony M. Dart
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Heart Centre, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
50
|
Di Tano G, Caretta G, De Maria R, Bettari L, Parolini M, Testa S, Pirelli S. Galectin-3 and outcomes after anterior-wall myocardial infarction treated by primary percutaneous coronary intervention. Biomark Med 2017; 12:21-26. [PMID: 29243525 DOI: 10.2217/bmm-2017-0178] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Galectin-3 (Gal-3), a biomarker of inflammation, tissue repair and fibrogenesis, is associated to left ventricular remodeling after ST-elevated myocardial infarction (STEMI), but its relation with long-term outcomes is unclear. METHODS In 103 consecutive patients with a first anterior STEMI treated by primary angioplasty, we assayed Gal-3 and NT-proBNP. RESULTS Age was 65 (56-76) years, 28% were women. During 18 ± 13 months, 20 patients (19.4%) died or were admitted for heart failure. After adjustment for age, gender, renal and ventricular function, troponin, NT-proBNP and Gal-3 independently predicted the combined end point (hazard ratio: 1.11; 95% CI: 1.05-1.17; per 1 ng/ml increase). Event-free survival was 42.3 versus 93.5% for Gal-3≥ versus <16.8 ng/ml (p < 0.001). CONCLUSION Among anterior STEMI patients, early postangioplasty Gal-3 levels may be useful for risk stratification.
Collapse
Affiliation(s)
- Giuseppe Di Tano
- Division of Cardiology, ASST - Hospital of Cremona, Cremona, Italy
| | - Giorgio Caretta
- Division of Cardiology, Sant'Andrea Hospital, ASL 5 Liguria - La Spezia, Italy
| | - Renata De Maria
- CNR Clinical Physiology Institute Cardiothoracic & Vascular Department ASST-Great Metropolitan Hospital Niguarda, Milan, Italy
| | - Luca Bettari
- Division of Cardiology, ASST - Hospital of Cremona, Cremona, Italy
| | - Marina Parolini
- CNR Clinical Physiology Institute Cardiothoracic & Vascular Department ASST-Great Metropolitan Hospital Niguarda, Milan, Italy
| | - Sophie Testa
- Division of Laboratory Medicine, ASST - Hospital of Cremona, Cremona, Italy
| | | |
Collapse
|