1
|
Karadas H, Tosun H, Ceylan H. Identification of dilated cardiomyopathy-linked key genes by bioinformatics methods and evaluating the impact of tannic acid and monosodium glutamate in rats. Biotechnol Appl Biochem 2025; 72:377-387. [PMID: 39318238 PMCID: PMC11975261 DOI: 10.1002/bab.2670] [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: 07/17/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024]
Abstract
Dilated cardiomyopathy (DCM) is the most common type of myocardial dysfunction, affecting mostly young adults, but its therapeutic diagnosis and biomarkers for prognosis are lacking. This study aimed to investigate the possible effect of the common food additive monosodium glutamate (MSG) and tannic acid (TA), a phenolic compound, on the key molecular actors responsible for DCM. DCM-related publicly available microarray datasets (GSE120895, GSE17800, and GSE19303) were downloaded from the comprehensive Gene Expression Omnibus (GEO) database, and analyzed to identify differentially expressed genes (DEGs). By integrating DEGs and gene-disease validity curation results, overlapping genes were screened and identified as hub genes. Protein-protein interaction (PPI) network and ontology analysis were performed to make sense of the identified biological data. Finally, mRNA expression changes of identified hub genes in the heart tissues of rats treated with MSG and TA were measured by the qPCR method. Six upregulated (IGF1, TTN, ACTB, LMNA, EDN1, and NPPB) DEGs were identified between the DCM and healthy control samples as the hub genes. qPCR results revealed that the mRNA levels of these genes involved in DCM development increased significantly in rat heart tissues exposed to MSG. In contrast, this increase was remarkably alleviated by TA treatment. Our results provide new insights into critical molecular mechanisms that should be focused on in future DCM studies. Moreover, MSG may play a critical role in DCM formation, and TA may be used as a promising therapeutic agent in DCM.
Collapse
Affiliation(s)
- Habibe Karadas
- Department of Molecular Biology and Genetics, Faculty of ScienceAtatürk UniversityErzurumTurkey
| | - Hilal Tosun
- Department of Molecular Biology and Genetics, Faculty of ScienceAtatürk UniversityErzurumTurkey
| | - Hamid Ceylan
- Department of Molecular Biology and Genetics, Faculty of ScienceAtatürk UniversityErzurumTurkey
| |
Collapse
|
2
|
Luo Y, Zhang F, Zhu L, Ye J, Pan HY, Lu X, Fan X. Efficacy and compatibility mechanism of bear bile powder in Shexiang Tongxin dropping pills for acute myocardial infarction treatment. Chin Med 2025; 20:14. [PMID: 39863867 PMCID: PMC11763157 DOI: 10.1186/s13020-025-01060-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Bear bile powder (BBP), a unique animal-derived medicine with anti-inflammatory and antioxidant effects, is used in Shexiang Tongxin dropping pills (STDP), which is applied to treat cardiovascular diseases, including acute myocardial infarction (AMI). The efficacy and compatibility mechanisms of action of BBP in STDP against cardiovascular diseases remain unclear. This study aimed to investigate the compatibility effects of BBP in STDP in rats with AMI. METHODS We investigated the compatibility effects of BBP in STDP in rats with AMI. Non-targeted metabonomics, 16S rRNA analysis, RNA sequencing, and network pharmacology were performed to explore the underlying mechanisms. RESULTS The combination of BBP and CF (STDP without BBP) significantly reduced AMI-induced infarction size, pathological alterations of cardiac tissues, and serum lactate dehydrogenase and creatine kinase levels in rats, compared with CF or BBP treatment alone. Gut microbiota and metabonomics results revealed that the combination treatment could upregulate the relative abundance of Lactobacillus and downregulate that of Helicobacter, Bilophila, and Butyricimonas, thereby rebalancing the gut microbiota dysbiosis induced by AMI. Consequently, the intestinal metabolite levels of oleoylcholine, glutamylalanine, isokobusone, and hemorphin-4 were altered. However, treatment with CF or BBP alone has a weaker effect on these bacteria. Additionally, the combination treatment induced a 62.34% gene reversion rate compared with 55.56% for BBP and 30.20% for CF treatment alone. Modulation of endothelin 1 and growth factor receptor-bound protein 2 was identified as a key synergistic mechanism underlying the anti-AMI effects of BBP in STDP. CONCLUSION This research provides a scientific explanation of the compatibility of BBP in STDP. Our findings suggested that combination treatment with CF and BBP synergistically attenuates AMI by altering gene expression, gut microbiota, and intestinal metabolite profiles.
Collapse
Affiliation(s)
- Yu Luo
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Fangmin Zhang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lidan Zhu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianfeng Ye
- Zhejiang Conba Pharmaceutical Co., Ltd, Hangzhou, 310051, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Pharmaceutical Technology, Hangzhou, 310051, China
| | - Hong-Ye Pan
- Zhejiang Conba Pharmaceutical Co., Ltd, Hangzhou, 310051, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Pharmaceutical Technology, Hangzhou, 310051, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delt, Zhejiang University, Jiaxing, 314100, China.
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China.
| |
Collapse
|
3
|
Balaraman AK, Altamimi ASA, Babu MA, Goyal K, PadmaPriya G, Bansal P, Rajotiya S, Kumar MR, Rajput P, Imran M, Gupta G, Thangavelu L. The interplay of senescence and MMPs in myocardial infarction: implications for cardiac aging and therapeutics. Biogerontology 2025; 26:46. [PMID: 39832057 DOI: 10.1007/s10522-025-10190-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Aging is associated with a marked increase in cardiovascular diseases, such as myocardial infarction (MI). Cellular senescence is also a crucial factor in the development of age-related MI. Matrix metalloproteinases (MMPs) interaction with cellular senescence is a critical determinant of MI development and outcomes, most notably in the aged heart. After experiencing a heart attack, senescent cells exhibit a Senescence-Associated Secretory Phenotype (SASP) and are involved in tissue regeneration and chronic inflammation. MMPs are necessary for extracellular matrix proteolysis and have a biphasic effect, promoting early heart healing and detrimental change if overexpressed shortly. This review analyses the complex connection between senescence and MMPs in MI and how it influences elderly cardiac performance. Critical findings suggest that increasing cellular senescence in aged hearts elevates MMP activity and aggravates extended ventricular remodeling and dysfunction. Additionally, we explore potential therapeutics that address MMPs and senescence to enhance old MI patient myocardial performance and regeneration.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari Cyber 11, Cyberjaya, Selangor, 63000, Malaysia
| | | | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Uttar Pradesh, Mathura, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - G PadmaPriya
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Pooja Bansal
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Sumit Rajotiya
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Pranchal Rajput
- Division of Research and Innovation, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Punjab, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
4
|
Adao DMT, Ching C, Fish JE, Simmons CA, Billia F. Endothelial cell-cardiomyocyte cross-talk: understanding bidirectional paracrine signaling in cardiovascular homeostasis and disease. Clin Sci (Lond) 2024; 138:1395-1419. [PMID: 39492693 DOI: 10.1042/cs20241084] [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: 06/09/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
To maintain homeostasis in the heart, endothelial cells and cardiomyocytes engage in dynamic cross-talk through paracrine signals that regulate both cardiac development and function. Here, we review the paracrine signals that endothelial cells release to regulate cardiomyocyte growth, hypertrophy and contractility, and the factors that cardiomyocytes release to influence angiogenesis and vascular tone. Dysregulated communication between these cell types can drive pathophysiology of disease, as seen in ischemia-reperfusion injury, diabetes, maladaptive hypertrophy, and chemotherapy-induced cardiotoxicity. Investingating the role of cross-talk is critical in developing an understanding of tissue homeostasis, regeneration, and disease pathogenesis, with the potential to identify novel targets for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Doris M T Adao
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario, Canada, M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave., Toronto, Ontario, Canada, M5G 1M1
- Toronto General Hospital Research Institute, University Health Network, 100 College St., Toronto, Ontario Canada, M5G 1L7
| | - Crizza Ching
- Toronto General Hospital Research Institute, University Health Network, 100 College St., Toronto, Ontario Canada, M5G 1L7
- Institute of Medical Science, University of Toronto, 1 King's College Cir., Toronto, Ontario, Canada, M5G 1A8
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, 100 College St., Toronto, Ontario Canada, M5G 1L7
- Institute of Medical Science, University of Toronto, 1 King's College Cir., Toronto, Ontario, Canada, M5G 1A8
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Cir., Toronto, Ontario, Canada, M5G 1A8
- Peter Munk Cardiac Centre, University Health Network, 585 University Ave., Toronto, Ontario, Canada, M5G 2N2
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario, Canada, M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave., Toronto, Ontario, Canada, M5G 1M1
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Rd., Toronto, Ontario, Canada, M5S 3G8
| | - Filio Billia
- Toronto General Hospital Research Institute, University Health Network, 100 College St., Toronto, Ontario Canada, M5G 1L7
- Institute of Medical Science, University of Toronto, 1 King's College Cir., Toronto, Ontario, Canada, M5G 1A8
- Peter Munk Cardiac Centre, University Health Network, 585 University Ave., Toronto, Ontario, Canada, M5G 2N2
| |
Collapse
|
5
|
Hojda SE, Chis IC, Clichici S. Magnesium Sulfate, Rosuvastatin, Sildenafil and Their Combination in Chronic Hypoxia-Induced Pulmonary Hypertension in Male Rats. Life (Basel) 2024; 14:1193. [PMID: 39337975 PMCID: PMC11433049 DOI: 10.3390/life14091193] [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: 08/16/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Previous experimental findings have led to considerable interest in the beneficial effects on pulmonary hypertension (PH) produced by sildenafil and in the pleiotropic effects of rosuvastatin and their positive role in the process of pulmonary angiogenesis. However, magnesium sulfate, the most abundant intracellular cation, is essential in vascular endothelial functionality due to its anti-inflammatory and vasodilatory effects. Therefore, the present study aims to assess these treatment regimens and how they could potentially provide some additional benefits in PH therapy. Fourteen days after chronic-hypoxia PH was induced, rosuvastatin, sildenafil and magnesium sulfate were administered for an additional fourteen days to male Wistar rats. The Fulton Index, right ventricle (RV) anterior wall thickness, RV internal diameter and pulmonary arterial (PA) acceleration time/ejection time were evaluated, and another four biochemical parameters were calculated: brain natriuretic peptide, vascular endothelial growth factor, nitric oxide metabolites and endothelin 1. The present study demonstrates that sildenafil and rosuvastatin have modest effects in reducing RV hypertrophy and RV systolic pressure. The drug combination of sildenafil + rosuvastatin + magnesium sulfate recorded statistically very highly significant results on all parameters; through their positive synergistic effects on vascular endothelial function, oxidative stress and pathological RV remodeling, they attenuated PH in the chronic hypoxia pulmonary hypertension (CHPH) rat model.
Collapse
Affiliation(s)
- Silvana-Elena Hojda
- Department of Physiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Number 1-3, Clinicilor Street, RO-400023 Cluj-Napoca, Romania
| | - Irina Camelia Chis
- Department of Physiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Number 1-3, Clinicilor Street, RO-400023 Cluj-Napoca, Romania
| | - Simona Clichici
- Department of Physiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Number 1-3, Clinicilor Street, RO-400023 Cluj-Napoca, Romania
| |
Collapse
|
6
|
Redwanz C, Pires RH, Biedenweg D, Groß S, Otto O, Könemann S. Endothelin-1 influences mechanical properties and contractility of hiPSC derived cardiomyocytes resulting in diastolic dysfunction. J Mol Cell Cardiol 2024; 194:105-117. [PMID: 39019395 DOI: 10.1016/j.yjmcc.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
A better understanding of the underlying pathomechanisms of diastolic dysfunction is crucial for the development of targeted therapeutic options with the aim to increase the patients' quality of life. In order to shed light on the processes involved, suitable models are required. Here, effects of endothelin-1 (ET-1) treatment on cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) were investigated. While it is well established, that ET-1 treatment induces hypertrophy in cardiomyocytes, resulting changes in cell mechanics and contractile behavior with focus on relaxation have not been examined before. Cardiomyocytes were treated with 10 nM of ET-1 for 24 h and 48 h, respectively. Hypertrophy was confirmed by real-time deformability cytometry (RT-DC) which was also used to assess the mechanical properties of cardiomyocytes. For investigation of the contractile behavior, 24 h phase contrast video microscopy was applied. To get a deeper insight into changes on the molecular biological level, gene expression analysis was performed using the NanoString nCounter® cardiovascular disease panel. Besides an increased cell size, ET-1 treated cardiomyocytes are stiffer and show an impaired relaxation. Gene expression patterns in ET-1 treated hiPSC derived cardiomyocytes showed that pathways associated with cardiovascular diseases, cardiac hypertrophy and extracellular matrix were upregulated while those associated with fatty acid metabolism were downregulated. We conclude that alterations in cardiomyocytes after ET-1 treatment go far beyond hypertrophy and represent a useful model for diastolic dysfunction.
Collapse
Affiliation(s)
- Caterina Redwanz
- Department for Internal Medicine B, Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany.
| | - Ricardo H Pires
- Institute of Physics, University of Greifswald, Felix-Hausdorff-Straße 6, 17489 Greifswald, Germany; Centre for Innovation Competence - Humoral Immune Response in Cardiovascular Diseases, University Medicine Greifswald, Friedrich-Ludwig-Jahn-Straße 15a, 17489 Greifswald, Germany
| | - Doreen Biedenweg
- Institute of Physics, University of Greifswald, Felix-Hausdorff-Straße 6, 17489 Greifswald, Germany; Centre for Innovation Competence - Humoral Immune Response in Cardiovascular Diseases, University Medicine Greifswald, Friedrich-Ludwig-Jahn-Straße 15a, 17489 Greifswald, Germany.
| | - Stefan Groß
- Department for Internal Medicine B, Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany.
| | - Oliver Otto
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany; Institute of Physics, University of Greifswald, Felix-Hausdorff-Straße 6, 17489 Greifswald, Germany; Centre for Innovation Competence - Humoral Immune Response in Cardiovascular Diseases, University Medicine Greifswald, Friedrich-Ludwig-Jahn-Straße 15a, 17489 Greifswald, Germany.
| | - Stephanie Könemann
- Department for Internal Medicine B, Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany.
| |
Collapse
|
7
|
Bonet F, Campuzano O, Córdoba-Caballero J, Alcalde M, Sarquella-Brugada G, Braza-Boïls A, Brugada R, Hernández-Torres F, Quezada-Feijoo M, Ramos M, Mangas A, Ranea JAG, Toro R. Role of miRNA-mRNA Interactome in Pathophysiology of Arrhythmogenic Cardiomyopathy. Biomedicines 2024; 12:1807. [PMID: 39200271 PMCID: PMC11351583 DOI: 10.3390/biomedicines12081807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Arrhythmogenic cardiomyopathy is an inherited entity characterized by irregular cell-cell adhesion, cardiomyocyte death and fibro-fatty replacement of ventricular myocytes, leading to malignant ventricular arrythmias, contractile dysfunction and sudden cardiac death. Pathogenic variants in genes that encode desmosome are the predominant cause of arrhythmogenic cardiomyopathy. Moreover, signalling pathways such as Wnt/ß-catenin and transforming growth factor-β have been involved in the disease progression. However, still little is known about the molecular pathophysiological mechanisms that underlie arrhythmogenic cardiomyopathy pathogenesis. We used mRNA and small RNA sequencing to analyse the transcriptome of health and arrhythmogenic cardiomyopathy of autopsied human hearts. Our results showed 697 differentially expressed genes and eight differentially expressed miRNAs. Functional enrichment revealed mitochondrial respiratory-related pathways, impaired response to oxidative stress, apoptotic signalling pathways and inflammatory response-related and extracellular matrix response pathways. Furthermore, analysis of the miRNA-mRNA interactome identified eleven negatively correlated miRNA-target pairs for arrhythmogenic cardiomyopathy. Our finding revealed novel arrhythmogenic cardiomyopathy-related miRNAs with important regulatory function in disease pathogenesis, highlighting their value as potential key targets for therapeutic approaches.
Collapse
Affiliation(s)
- Fernando Bonet
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
| | - Oscar Campuzano
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain; (G.S.-B.); (R.B.)
- Institut d’Investigació Biomèdica de Girona (IDIBGI-CERCA), 17190 Salt, Spain;
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - José Córdoba-Caballero
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
- Department of Molecular Biology and Biochemistry, University of Málaga, 29071 Málaga, Spain;
| | - Mireia Alcalde
- Institut d’Investigació Biomèdica de Girona (IDIBGI-CERCA), 17190 Salt, Spain;
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - Georgia Sarquella-Brugada
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain; (G.S.-B.); (R.B.)
- Pediatric Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Cardiology Department, Sant Joan de Déu Hospital, 08950 Barcelona, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - Aitana Braza-Boïls
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
- Cardiopatías Familiares, Muerte Súbita y Mecanismos de Enfermedad (CAFAMUSME) Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Ramon Brugada
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain; (G.S.-B.); (R.B.)
- Institut d’Investigació Biomèdica de Girona (IDIBGI-CERCA), 17190 Salt, Spain;
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
- Cardiology Service, Hospital Josep Trueta de Girona, 17007 Girona, Spain
| | - Francisco Hernández-Torres
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain;
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Maribel Quezada-Feijoo
- Cardiology Department, Hospital Central de la Cruz Roja, 28003 Madrid, Spain; (M.Q.-F.)
- Medicine School, Alfonso X el Sabio University, 28007 Madrid, Spain
| | - Monica Ramos
- Cardiology Department, Hospital Central de la Cruz Roja, 28003 Madrid, Spain; (M.Q.-F.)
- Medicine School, Alfonso X el Sabio University, 28007 Madrid, Spain
| | - Alipio Mangas
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
- Medicine Department, School of Medicine, University of Cadiz, 11003 Cádiz, Spain
- Lipid and Atherosclerotic Unit, Puerta del Mar University Hospital, 11009 Cadiz, Spain
| | - Juan A. G. Ranea
- Department of Molecular Biology and Biochemistry, University of Málaga, 29071 Málaga, Spain;
- Institute of Biomedical Research in Málaga and platform of nanomedicine (IBIMA Plataforma BIONAND), 29071 Málaga, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Spanish National Bioinformatics Institute (INB/ELIXIR-ES), Instituto de Salud Carlos III (ISCIII), 28020 Madrid, Spain
| | - Rocío Toro
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
- Medicine Department, School of Medicine, University of Cadiz, 11003 Cádiz, Spain
| |
Collapse
|
8
|
Wang Z, Wu Z, Hu Z, Zheng H, Chen Z. ET1 acts as a potential plasma biomarker and therapeutic target in deep venous thrombosis rat model. J Thromb Thrombolysis 2024; 57:1067-1075. [PMID: 38824487 PMCID: PMC11315785 DOI: 10.1007/s11239-024-02981-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/07/2024] [Indexed: 06/03/2024]
Abstract
Deep venous thrombosis (DVT) is the third leading cause of death in cardiovascular disease, following heart attacks and strokes. Early diagnosis and intervention are crucial for effective DVT therapy. We aim to investigate whether endothelin-1 (ET-1) could serve as an early diagnostic marker or a potential therapeutic target in a DVT rat model. CCK8 assay, invasion assay, and flow cytometry were used to detect the proliferation, migration and apoptosis of HUVECs, respectively. Elisa assay was used to detect ET-1 and coagulation factor VII in cell supernatant and rat?s plasma. Western blot was used to detect antioxidant signaling protein. Inferior vena cava stenosis was used to construct the DVT rat model. Lentivirus mediated overexpression of ET-1 in HUVECs impaired the cell proliferation and migration, increased cell apoptosis, inhibited the antioxidant signaling pathway proteins expression (e.g., NQO1, GCLC, Nrf-2), and upregulated coagulation factor VII. Furthermore, overexpression of ET-1 further impaired antioxidant signaling pathway protein in response to H2O2 treatment. However, lentivirus mediated ET-1 knockdown and BQ123 (an ET-1 inhibitor), showed the opposite results with ET-1 overexpression. We then established a DVT rat model by inferior vena cava stenosis. The stenosis induced early expression of ET-1 and coagulation factor VII in plasma at day 1 and restore their level at day 10. BQ123 could downregulate the coagulation factor VII to ameliorate the stenosis effects. Our findings suggest that ET-1 might serve as an early diagnostic marker for DVT rat model and a potential therapeutic target for treating DVT.
Collapse
Affiliation(s)
- Zhanqi Wang
- Department of Vascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital of Capital Medical University, Beijing, 100029, China
| | - Zhangmin Wu
- Department of Vascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital of Capital Medical University, Beijing, 100029, China
| | - Zhongzhou Hu
- Department of Vascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital of Capital Medical University, Beijing, 100029, China
| | - Huanqin Zheng
- Department of Vascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital of Capital Medical University, Beijing, 100029, China
| | - Zhong Chen
- Department of Vascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital of Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
9
|
Qu Y, Zhang D, Hu Y, Wang J, Tan H, Qin F, Liu Y. Long-term prognostic value of big endothelin-1 and its combination with late gadolinium enhancement in patients with idiopathic restrictive cardiomyopathy. Clin Chim Acta 2024; 561:119755. [PMID: 38821338 DOI: 10.1016/j.cca.2024.119755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/28/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND AND AIMS Idiopathic restrictive cardiomyopathy (RCM) has a low incidence. This study aimed to determine the prognostic value of big endothelin-1 (ET-1) in idiopathic RCM. MATERIALS AND METHODS We prospectively enrolled patients with idiopathic RCM from 2009 to 2017 and followed them up. The primary outcome was a composite of all-cause mortality and cardiac transplantation, and the secondary outcome was a composite of cardiac death and cardiac transplantation. RESULTS Ninety-one patients were divided into the high big ET-1 (>0.85 pmol/L, n = 56) and low big ET-1 (≤0.85 pmol/L, n = 35) groups, and 87 of them completed the follow-up. Big ET-1 concentrations (hazard ratio: 1.756, 95 % confidence interval [CI]: 1.117-2.760) and late gadolinium enhancement (LGE) (hazard ratio: 3.851, 95 % CI: 1.238-11.981) were independent risk factors for the primary outcome. Big ET-1 concentrations (C-statistic estimation: 0.764, 95 % CI: 0.657-0.871) and the combination of LGE and big ET-1 concentrations (C-statistic estimation: 0.870, 95 % CI: 0.769-0.970) could accurately predict the 5-year transplant-free survival rate, and 0.85 pmol/L was a suitable cutoff for big ET-1. CONCLUSION Big ET-1 and its combination with LGE may be useful to predict an adverse prognosis in patients with idiopathic RCM.
Collapse
Affiliation(s)
- Yi Qu
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Beijing, China
| | - Di Zhang
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Beijing, China
| | - Yuxiao Hu
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Beijing, China
| | - Jiayi Wang
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Beijing, China
| | - Huiqiong Tan
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Beijing, China
| | - Fuzhong Qin
- The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, Shanxi Province, China
| | - Yaxin Liu
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Beijing, China.
| |
Collapse
|
10
|
Rao RS, Sharma P, padhy M, Sharma R, Gupta R, Bhatacharjee J, Singh M. Circulatory Maternal Endothelin 1 and Matrix Metalloproteinase-9 Gene Expression in PREECLAMPSIA: A Study in Western Uttar Pradesh, India. J Obstet Gynaecol India 2023; 73:97-102. [PMID: 37916024 PMCID: PMC10616056 DOI: 10.1007/s13224-022-01720-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction Preeclampsia (PE) is a multiorgan disease of pregnant women. The main pathophysiology of PE is a trophoblastic invasion into maternal circulation leading to alterations in circulatory levels of matrix metalloproteinases (MMPs), inflammatory markers, and endothelin 1(ET1) levels. Therefore, the present study has explored the role of MMP-9 and ET1 and their association in PE. The advantage of the study is to provide insight into the pathology of PE. These markers may help in the early diagnosis and prognosis of PE. Objective To investigate MMP-9 gene expression, ET1 level in PE cases and their correlation with blood pressure (BP), gestational age, weight, and height. Methods The study design was a case-control observational study, which included 70 subjects in each case (PE) and controls (normal pregnant women (NPW)). Whole blood (250 ul) was utilized for RNA extraction (Trizol method) and synthesized cDNA as per manufacturer protocol. MMP-9 gene expression was analyzed by real-time PCR. Serum was utilized for ET1 estimation by sandwich ELISA. Results The ET1 levels and MMP-9 gene expression were significantly increased in preeclamptic women as compared to controls. There was no significant correlation between MMP-9 gene expression and serum ET1 levels. However, a significant moderate association between systolic BP and diastolic BP with ET1 levels and MMP9 gene expression was seen in both PE and NPW. Conclusion A significantly increased circulatory concentration of ET1 and MMP-9 gene expression in PE might be used as an early diagnostic as well as a prognostic marker of PE.
Collapse
Affiliation(s)
- Ravoori Saideswar Rao
- Department of Biochemistry, Santosh Medical College and Hospital, Ghaziabad, UP India
| | - Preeti Sharma
- Department of Biochemistry, Santosh Medical College and Hospital, Ghaziabad, UP India
| | - Mamta padhy
- Department of Biochemistry, Government Institute of Medical Sciences, Greater Noida, UP India
| | - Ritu Sharma
- Department of Obstetrics and Gynecology, Government Institute of Medical Sciences, Greater Noida, UP India
| | - Rakesh Gupta
- Department of Pediatrics, Government Institute of Medical Sciences, Greater Noida, UP India
| | - Jayashree Bhatacharjee
- Department of Biochemistry, Government Institute of Medical Sciences, Greater Noida, UP India
| | - Manisha Singh
- Department of Biochemistry, Government Institute of Medical Sciences, Greater Noida, UP India
| |
Collapse
|
11
|
Dmour BA, Costache AD, Dmour A, Huzum B, Duca ȘT, Chetran A, Miftode RȘ, Afrăsânie I, Tuchiluș C, Cianga CM, Botnariu G, Șerban LI, Ciocoiu M, Bădescu CM, Costache II. Could Endothelin-1 Be a Promising Neurohormonal Biomarker in Acute Heart Failure? Diagnostics (Basel) 2023; 13:2277. [PMID: 37443671 DOI: 10.3390/diagnostics13132277] [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: 05/24/2023] [Revised: 06/20/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Acute heart failure (AHF) is a life-threatening condition with high morbidity and mortality. Even though this pathology has been extensively researched, there are still challenges in establishing an accurate and early diagnosis, determining the long- and short-term prognosis and choosing a targeted therapeutic strategy. The use of reliable biomarkers to support clinical judgment has been shown to improve the management of AHF patients. Despite a large pool of interesting candidate biomarkers, endothelin-1 (ET-1) appears to be involved in multiple aspects of AHF pathogenesis that include neurohormonal activation, cardiac remodeling, endothelial dysfunction, inflammation, atherosclerosis and alteration of the renal function. Since its discovery, numerous studies have shown that the level of ET-1 is associated with the severity of symptoms and cardiac dysfunction in this pathology. The purpose of this paper is to review the existing information on ET-1 and answer the question of whether this neurohormone could be a promising biomarker in AHF.
Collapse
Affiliation(s)
- Bianca-Ana Dmour
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alexandru Dan Costache
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Awad Dmour
- Department of Orthopedics and Traumatology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Orthopaedics and Traumatology, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Bogdan Huzum
- Department of Orthopaedics and Traumatology, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ștefania Teodora Duca
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Cardiology Clinic, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Adriana Chetran
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Cardiology Clinic, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Radu Ștefan Miftode
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Cardiology Clinic, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Irina Afrăsânie
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Cardiology Clinic, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Cristina Tuchiluș
- Department of Microbiology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Microbiology Laboratory, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Corina Maria Cianga
- Immunology Laboratory, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Immunology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Gina Botnariu
- Unit of Diabetes, Nutrition and Metabolic Diseases, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Lăcrămioara Ionela Șerban
- Department of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Manuela Ciocoiu
- Department of Morpho-Functional Sciences II, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Codruța Minerva Bădescu
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Irina Iuliana Costache
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Cardiology Clinic, "St. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| |
Collapse
|
12
|
Feng J, Liang L, Chen Y, Tian P, Zhao X, Huang B, Wu Y, Wang J, Guan J, Huang L, Li X, Zhang Y, Zhang J. Big Endothelin-1 as a Predictor of Reverse Remodeling and Prognosis in Dilated Cardiomyopathy. J Clin Med 2023; 12:jcm12041363. [PMID: 36835899 PMCID: PMC9967115 DOI: 10.3390/jcm12041363] [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: 01/06/2023] [Revised: 01/24/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
This study aimed to investigate the predictive value of Big endothelin-1(ET-1) for left ventricular reverse remodeling (LVRR) and prognosis in patients with dilated cardiomyopathy (DCM). Patients with DCM and a left ventricular ejection fraction (LVEF) ≤ 50% from 2008 to 2017 were included. LVRR was defined as the LVEF increased by at least 10% or follow-up LVEF increased to at least 50% with a minimum improvement of 5%; meanwhile, the index of left ventricular end-diastolic diameter (LVEDDi) decreased by at least 10% or LVEDDi decreased to ≤33 mm/m2. The composite outcome for prognostic analysis consisted of death and heart transplantations. Of the 375 patients included (median age 47 years, 21.1% female), 135 patients (36%) had LVRR after a median of 14 months of treatment. An independent association was found between Big ET-1 at baseline and LVRR in the multivariate model (OR 0.70, 95% CI 0.55-0.89, p = 0.003, per log increase). Big ET-1, body mass index, systolic blood pressure, diagnosis of type 2 diabetes mellitus (T2DM) and treatment with ACEI/ARB were significant predictors for LVRR after stepwise selection. Adding Big ET-1 to the model improved the discrimination (∆AUC = 0.037, p = 0.042 and reclassification (IDI, 3.29%; p = 0.002; NRI, 35%; p = 0.002) for identifying patients with LVRR. During a median follow-up of 39 (27-68) months, Big ET-1 was also independently associated with the composite outcome of death and heart transplantations (HR 1.45, 95% CI 1.13-1.85, p = 0.003, per log increase). In conclusion, Big ET-1 was an independent predictor for LVRR and had prognostic implications, which might help to improve the risk stratification of patients with DCM.
Collapse
Affiliation(s)
- Jiayu Feng
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Lin Liang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yuyi Chen
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Pengchao Tian
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xuemei Zhao
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Boping Huang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yihang Wu
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jing Wang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jingyuan Guan
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Liyan Huang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xinqing Li
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yuhui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Correspondence: (Y.Z.); (J.Z.)
| | - Jian Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Key Laboratory of Clinical Research for Cardiovascular Medications, National Health Committee, Beijing 100037, China
- Correspondence: (Y.Z.); (J.Z.)
| |
Collapse
|
13
|
De Bortoli M, Meraviglia V, Mackova K, Frommelt LS, König E, Rainer J, Volani C, Benzoni P, Schlittler M, Cattelan G, Motta BM, Volpato C, Rauhe W, Barbuti A, Zacchigna S, Pramstaller PP, Rossini A. Modeling incomplete penetrance in arrhythmogenic cardiomyopathy by human induced pluripotent stem cell derived cardiomyocytes. Comput Struct Biotechnol J 2023; 21:1759-1773. [PMID: 36915380 PMCID: PMC10006475 DOI: 10.1016/j.csbj.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023] Open
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are commonly used to model arrhythmogenic cardiomyopathy (ACM), a heritable cardiac disease characterized by severe ventricular arrhythmias, fibrofatty myocardial replacement and progressive ventricular dysfunction. Although ACM is inherited as an autosomal dominant disease, incomplete penetrance and variable expressivity are extremely common, resulting in different clinical manifestations. Here, we propose hiPSC-CMs as a powerful in vitro model to study incomplete penetrance in ACM. Six hiPSC lines were generated from blood samples of three ACM patients carrying a heterozygous deletion of exon 4 in the PKP2 gene, two asymptomatic (ASY) carriers of the same mutation and one healthy control (CTR), all belonging to the same family. Whole exome sequencing was performed in all family members and hiPSC-CMs were examined by ddPCR, western blot, Wes™ immunoassay system, patch clamp, immunofluorescence and RNASeq. Our results show molecular and functional differences between ACM and ASY hiPSC-CMs, including a higher amount of mutated PKP2 mRNA, a lower expression of the connexin-43 protein, a lower overall density of sodium current, a higher intracellular lipid accumulation and sarcomere disorganization in ACM compared to ASY hiPSC-CMs. Differentially expressed genes were also found, supporting a predisposition for a fatty phenotype in ACM hiPSC-CMs. These data indicate that hiPSC-CMs are a suitable model to study incomplete penetrance in ACM.
Collapse
Key Words
- ABC, active ß-catenin
- ACM, arrhythmogenic cardiomyopathy
- ASY, asymptomatic
- Arrhythmogenic cardiomyopathy
- BBB, bundle-branch block
- CMs, cardiomyocytes
- CTR, control
- Cx43, connexin-43
- DEGs, differentially expressed genes
- GATK, Genome Analysis Toolkit
- Human induced pluripotent stem cell derived cardiomyocytes
- ICD, implantable cardioverter-defibrillator
- ID, intercalated disk
- Incomplete penetrance
- LBB, left bundle-branch block
- MRI, magnetic resonance imagingmut, mutated
- NSVT, non-sustained ventricular tachycardia
- RV, right ventricle
- hiPSC, human induced pluripotent stem cell
- wt, wild type
Collapse
Affiliation(s)
- Marzia De Bortoli
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Viviana Meraviglia
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy.,Department of Anatomy and Embryology, Leiden University Medical Center, 2316 Leiden, the Netherlands
| | - Katarina Mackova
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Laura S Frommelt
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Eva König
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Johannes Rainer
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Chiara Volani
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy.,Universita` degli Studi di Milano, The Cell Physiology MiLab, Department of Biosciences, Milano, Italy
| | - Patrizia Benzoni
- Universita` degli Studi di Milano, The Cell Physiology MiLab, Department of Biosciences, Milano, Italy
| | - Maja Schlittler
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Giada Cattelan
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Benedetta M Motta
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Claudia Volpato
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Werner Rauhe
- San Maurizio Hospital, Department of Cardiology, Bolzano, Italy
| | - Andrea Barbuti
- Universita` degli Studi di Milano, The Cell Physiology MiLab, Department of Biosciences, Milano, Italy
| | - Serena Zacchigna
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cardiovascular Biology Laboratory, Trieste, Italy
| | - Peter P Pramstaller
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Alessandra Rossini
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| |
Collapse
|
14
|
Moxonidine ameliorates cardiac injury in rats with metabolic syndrome by regulating autophagy. Life Sci 2022; 312:121210. [PMID: 36410408 DOI: 10.1016/j.lfs.2022.121210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022]
Abstract
AIMS Reduced cardiac autophagy, ischemic injury, sympathetic overactivity, and apoptosis all contribute to metabolic syndrome (MetS)-associated cardiovascular risks. NR4A2, an orphan nuclear receptor NR4A family member, induces autophagy while suppressing apoptosis in myocardial infarction. Moxonidine, a sympathoinhibitor imidazoline1 receptor (I1R) agonist, has beneficial metabolic and hemodynamic effects; however, whether autophagy and/or NR4A2 signaling are involved in moxonidine's cardiovascular effects via I1R activation, is unknown, and is the aim of this study. MATERIALS AND METHODS To induce MetS, rats were fed 3 % salt in their diet and 10 % fructose in their drinking water for 12 weeks. MetS-rats were given either moxonidine (6 mg/kg/day, gavage), efaroxan (I1R antagonist, 0.6 mg/kg/day, i.p), both treatments, or vehicles for the last two weeks. Blood pressure, lipid profile, and glycemic control were evaluated. Histopathological examination, circulating cardiac troponin I (c-TnI), proinflammatory interleukin-6 (IL-6), apoptosis (active caspase-3 and Fas-immunostaining), interstitial fibrosis [transforming growth factor-β1 (TGF-β1), Mallory's trichrome staining], and extracellular matrix remodeling [matrix metalloproteinase-9 (MMP-9)], were used to assess cardiac pathology. Cardiac NR4A2 and its downstream factor, p53, as well as autophagic flux markers, SQSTM1/p62, LC3, and Beclin-1 were also determined. KEY FINDINGS Moxonidine significantly ameliorated MetS-induced metabolic and hemodynamic derangements and the associated cardiac pathology. Moxonidine restored NR4A2 and p53 myocardial levels and enhanced autophagic flux via modulating SQSTM1/p62, LC3, and Beclin-1. Efaroxan reversed the majority of the moxonidine-induced improvements. SIGNIFICANCE The current study suggests that autophagy modulation via I1R activation is involved in moxonidine-mediated cardiac beneficial effects in MetS.
Collapse
|
15
|
Prediction of Regulatory SNPs in Putative Minor Genes of the Neuro-Cardiovascular Variant in Fabry Reveals Insights into Autophagy/Apoptosis and Fibrosis. BIOLOGY 2022; 11:biology11091287. [PMID: 36138766 PMCID: PMC9495465 DOI: 10.3390/biology11091287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
Even though a mutation in monogenic diseases leads to a “classic” manifestation, many disorders exhibit great clinical variability that could be due to modifying genes also called minor genes. Fabry disease (FD) is an X-linked inborn error resulting from the deficient or absent activity of alpha-galactosidase A (α-GAL) enzyme, that leads to deposits of globotriaosylceramide. With our proprietary software SNPclinic v.1.0, we analyzed 110 single nucleotide polymorphisms (SNPs) in the proximal promoter of 14 genes that could modify the FD phenotype FD. We found seven regulatory-SNP (rSNPs) in three genes (IL10, TGFB1 and EDN1) in five cell lines relevant to FD (Cardiac myocytes and fibroblasts, Astrocytes-cerebellar, endothelial cells and T helper cells 1-TH1). Each SNP was confirmed as a true rSNP in public eQTL databases, and additional software suggested the prediction of variants. The two proposed rSNPs in IL10, could explain components for the regulation of active B cells that influence the fibrosis process. The three predicted rSNPs in TGFB1, could act in apoptosis-autophagy regulation. The two putative rSNPs in EDN1, putatively regulate chronic inflammation. The seven rSNPs described here could act to modulate Fabry’s clinical phenotype so we propose that IL10, TGFB1 and EDN1 be considered minor genes in FD.
Collapse
|
16
|
Reichart D, Lindberg EL, Maatz H, Miranda AMA, Viveiros A, Shvetsov N, Gärtner A, Nadelmann ER, Lee M, Kanemaru K, Ruiz-Orera J, Strohmenger V, DeLaughter DM, Patone G, Zhang H, Woehler A, Lippert C, Kim Y, Adami E, Gorham JM, Barnett SN, Brown K, Buchan RJ, Chowdhury RA, Constantinou C, Cranley J, Felkin LE, Fox H, Ghauri A, Gummert J, Kanda M, Li R, Mach L, McDonough B, Samari S, Shahriaran F, Yapp C, Stanasiuk C, Theotokis PI, Theis FJ, van den Bogaerdt A, Wakimoto H, Ware JS, Worth CL, Barton PJR, Lee YA, Teichmann SA, Milting H, Noseda M, Oudit GY, Heinig M, Seidman JG, Hubner N, Seidman CE. Pathogenic variants damage cell composition and single cell transcription in cardiomyopathies. Science 2022; 377:eabo1984. [PMID: 35926050 PMCID: PMC9528698 DOI: 10.1126/science.abo1984] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pathogenic variants in genes that cause dilated cardiomyopathy (DCM) and arrhythmogenic cardiomyopathy (ACM) convey high risks for the development of heart failure through unknown mechanisms. Using single-nucleus RNA sequencing, we characterized the transcriptome of 880,000 nuclei from 18 control and 61 failing, nonischemic human hearts with pathogenic variants in DCM and ACM genes or idiopathic disease. We performed genotype-stratified analyses of the ventricular cell lineages and transcriptional states. The resultant DCM and ACM ventricular cell atlas demonstrated distinct right and left ventricular responses, highlighting genotype-associated pathways, intercellular interactions, and differential gene expression at single-cell resolution. Together, these data illuminate both shared and distinct cellular and molecular architectures of human heart failure and suggest candidate therapeutic targets.
Collapse
Affiliation(s)
- Daniel Reichart
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.,Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115, USA.,Department of Medicine I, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Eric L Lindberg
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Henrike Maatz
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Antonio M A Miranda
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK.,British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London WC2R 2LS, UK
| | - Anissa Viveiros
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada.,Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Nikolay Shvetsov
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Anna Gärtner
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Emily R Nadelmann
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Lee
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Kazumasa Kanemaru
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Jorge Ruiz-Orera
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Viktoria Strohmenger
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.,Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilian University of Munich, 81377 Munich, Germany
| | - Daniel M DeLaughter
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.,Howard Hughes Medical Institute, Bethesda, MD 20815, USA
| | - Giannino Patone
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Hao Zhang
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada.,Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Andrew Woehler
- Systems Biology Imaging Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 10115 Berlin, Germany
| | - Christoph Lippert
- Digital Health-Machine Learning group, Hasso Plattner Institute for Digital Engineering, University of Potsdam, 14482 Potsdam, Germany.,Hasso Plattner Institute for Digital Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yuri Kim
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.,Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Eleonora Adami
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Joshua M Gorham
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Sam N Barnett
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Kemar Brown
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.,Cardiac Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Rachel J Buchan
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK.,Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London SW3 6NR, UK
| | - Rasheda A Chowdhury
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | | | - James Cranley
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Leanne E Felkin
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK.,Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London SW3 6NR, UK
| | - Henrik Fox
- Heart and Diabetes Center NRW, Clinic for Thoracic and Cardiovascular Surgery, University Hospital of the Ruhr-University, 32545 Bad Oeynhausen, Germany
| | - Ahla Ghauri
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Jan Gummert
- Heart and Diabetes Center NRW, Clinic for Thoracic and Cardiovascular Surgery, University Hospital of the Ruhr-University, 32545 Bad Oeynhausen, Germany
| | - Masatoshi Kanda
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany.,Department of Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Ruoyan Li
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Lukas Mach
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK.,Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London SW3 6NR, UK
| | - Barbara McDonough
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115, USA.,Howard Hughes Medical Institute, Bethesda, MD 20815, USA
| | - Sara Samari
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Farnoush Shahriaran
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
| | - Clarence Yapp
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Caroline Stanasiuk
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Pantazis I Theotokis
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK.,MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, UK
| | - Fabian J Theis
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
| | | | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK.,Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London SW3 6NR, UK.,MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, UK
| | - Catherine L Worth
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Paul J R Barton
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK.,Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London SW3 6NR, UK.,MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, UK
| | - Young-Ae Lee
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany.,Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Sarah A Teichmann
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK.,Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge CB3 0HE, UK
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK.,British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London WC2R 2LS, UK
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada.,Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Matthias Heinig
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany.,Department of Informatics, Technische Universitaet Muenchen (TUM), 85748 Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Munich Heart Association, Partner Site Munich, 10785 Berlin, Germany
| | | | - Norbert Hubner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany.,Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.,Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115, USA.,Howard Hughes Medical Institute, Bethesda, MD 20815, USA
| |
Collapse
|
17
|
Tonry C, Russel-Hallinan A, McCune C, Collier P, Harbinson M, Dixon L, Watson CJ. Circulating biomarkers for management of cancer therapeutics related cardiac dysfunction. Cardiovasc Res 2022; 119:710-728. [PMID: 35640873 PMCID: PMC10153425 DOI: 10.1093/cvr/cvac087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 05/09/2022] [Accepted: 05/15/2022] [Indexed: 11/14/2022] Open
Abstract
Cancer therapeutics related cardiac dysfunction (CTRCD) has emerged as a major cause of morbidity and mortality in cancer survivors. Effective clinical management of CTRCD is impeded by a lack of sensitive diagnostic and prognostic strategies. Circulating molecular markers could potentially address this need as they are often indicative of cardiac stress before cardiac damage can be detected clinically. A growing understanding of the underlying physiological mechanisms for CTRCD has inspired research efforts to identify novel pathophysiologically-relevant biomarkers that may also guide development of cardio-protective therapeutic approaches. The purpose of this review is to evaluate current circulating biomarkers of cardiac stress and their potential role in diagnosis and management of CTRCD. We also discuss some emerging avenues for CTRCD-focused biomarker investigations.
Collapse
Affiliation(s)
- Claire Tonry
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Adam Russel-Hallinan
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Claire McCune
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | | | | | | | - Chris J Watson
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
18
|
Endothelin and the Cardiovascular System: The Long Journey and Where We Are Going. BIOLOGY 2022; 11:biology11050759. [PMID: 35625487 PMCID: PMC9138590 DOI: 10.3390/biology11050759] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022]
Abstract
Simple Summary In this review, we describe the basic functions of endothelin and related molecules, including their receptors and enzymes. Furthermore, we discuss the important role of endothelin in several cardiovascular diseases, the relevant clinical evidence for targeting the endothelin pathway, and the scope of endothelin-targeting treatments in the future. We highlight the present uses of endothelin receptor antagonists and the advancements in the development of future treatment options, thereby providing an overview of endothelin research over the years and its future scope. Abstract Endothelin was first discovered more than 30 years ago as a potent vasoconstrictor. In subsequent years, three isoforms, two canonical receptors, and two converting enzymes were identified, and their basic functions were elucidated by numerous preclinical and clinical studies. Over the years, the endothelin system has been found to be critical in the pathogenesis of several cardiovascular diseases, including hypertension, pulmonary arterial hypertension, heart failure, and coronary artery disease. In this review, we summarize the current knowledge on endothelin and its role in cardiovascular diseases. Furthermore, we discuss how endothelin-targeting therapies, such as endothelin receptor antagonists, have been employed to treat cardiovascular diseases with varying degrees of success. Lastly, we provide a glimpse of what could be in store for endothelin-targeting treatment options for cardiovascular diseases in the future.
Collapse
|
19
|
Guo Q, Zhu Q, Zhang T, Qu Q, Cheang I, Liao S, Chen M, Zhu X, Shi M, Li X. Integrated bioinformatic analysis reveals immune molecular markers and potential drugs for diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2022; 13:933635. [PMID: 36046789 PMCID: PMC9421304 DOI: 10.3389/fendo.2022.933635] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a pathophysiological condition induced by diabetes mellitus that often causes heart failure (HF). However, their mechanistic relationships remain unclear. This study aimed to identify immune gene signatures and molecular mechanisms of DCM. Microarray data from the Gene Expression Omnibus (GEO) database from patients with DCM were subjected to weighted gene co-expression network analysis (WGCNA) identify co-expression modules. Core expression modules were intersected with the immune gene database. We analyzed and mapped protein-protein interaction (PPI) networks using the STRING database and MCODE and filtering out 17 hub genes using cytoHubba software. Finally, potential transcriptional regulatory factors and therapeutic drugs were identified and molecular docking between gene targets and small molecules was performed. We identified five potential immune biomarkers: proteosome subunit beta type-8 (PSMB8), nuclear factor kappa B1 (NFKB1), albumin (ALB), endothelin 1 (EDN1), and estrogen receptor 1 (ESR1). Their expression levels in animal models were consistent with the changes observed in the datasets. EDN1 showed significant differences in expression in both the dataset and the validation model by real-time quantitative PCR (qPCR) and Western blotting(WB). Subsequently, we confirmed that the potential transcription factors upstream of EDN1 were PRDM5 and KLF4, as its expression was positively correlated with the expression of the two transcription factors. To repurpose known therapeutic drugs, a connectivity map (CMap) database was retrieved, and nine candidate compounds were identified. Finally, molecular docking simulations of the proteins encoded by the five genes with small-molecule drugs were performed. Our data suggest that EDN1 may play a key role in the development of DCM and is a potential DCM biomarker.
Collapse
|
20
|
Matyas C, Haskó G, Liaudet L, Trojnar E, Pacher P. Interplay of cardiovascular mediators, oxidative stress and inflammation in liver disease and its complications. Nat Rev Cardiol 2021; 18:117-135. [PMID: 32999450 DOI: 10.1038/s41569-020-0433-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/11/2020] [Indexed: 12/11/2022]
Abstract
The liver is a crucial metabolic organ that has a key role in maintaining immune and endocrine homeostasis. Accumulating evidence suggests that chronic liver disease might promote the development of various cardiac disorders (such as arrhythmias and cardiomyopathy) and circulatory complications (including systemic, splanchnic and pulmonary complications), which can eventually culminate in clinical conditions ranging from portal and pulmonary hypertension to pulmonary, cardiac and renal failure, ascites and encephalopathy. Liver diseases can affect cardiovascular function during the early stages of disease progression. The development of cardiovascular diseases in patients with chronic liver failure is associated with increased morbidity and mortality, and cardiovascular complications can in turn affect liver function and liver disease progression. Furthermore, numerous infectious, inflammatory, metabolic and genetic diseases, as well as alcohol abuse can also influence both hepatic and cardiovascular outcomes. In this Review, we highlight how chronic liver diseases and associated cardiovascular effects can influence different organ pathologies. Furthermore, we explore the potential roles of inflammation, oxidative stress, vasoactive mediator imbalance, dysregulated endocannabinoid and autonomic nervous systems and endothelial dysfunction in mediating the complex interplay between the liver and the systemic vasculature that results in the development of the extrahepatic complications of chronic liver disease. The roles of ageing, sex, the gut microbiome and organ transplantation in this complex interplay are also discussed.
Collapse
Affiliation(s)
- Csaba Matyas
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Lucas Liaudet
- Department of Intensive Care Medicine and Burn Center, University Hospital Medical Center, Faculty of Biology and Medicine, Lausanne, Switzerland
| | - Eszter Trojnar
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD, USA.
| |
Collapse
|
21
|
Kastner N, Zlabinger K, Spannbauer A, Traxler D, Mester-Tonczar J, Hašimbegović E, Gyöngyösi M. New Insights and Current Approaches in Cardiac Hypertrophy Cell Culture, Tissue Engineering Models, and Novel Pathways Involving Non-Coding RNA. Front Pharmacol 2020; 11:1314. [PMID: 32973530 PMCID: PMC7472597 DOI: 10.3389/fphar.2020.01314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/07/2020] [Indexed: 12/23/2022] Open
Abstract
Cardiac hypertrophy is an ongoing clinical challenge, as risk factors such as obesity, smoking and increasing age become more widespread, which lead to an increasing prevalence of developing hypertrophy. Pathological hypertrophy is a maladaptive response to stress conditions, such as pressure overload, and involve a number of changes in cellular mechanisms, gene expression and pathway regulations. Although several important pathways involved in the remodeling and hypertrophy process have been identified, further research is needed to achieve a better understanding and explore new and better treatment options. More recently discovered pathways showed the involvement of several non-coding RNAs, including micro RNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), which either promote or inhibit the remodeling process and pose a possible target for novel therapy approaches. In vitro modeling serves as a vital tool for this further pathway analysis and treatment testing and has vastly improved over the recent years, providing a less costly and labor-intensive alternative to in vivo animal models.
Collapse
Affiliation(s)
- Nina Kastner
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Katrin Zlabinger
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | - Denise Traxler
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | - Ena Hašimbegović
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Youk H, Kim M, Lee CJ, Oh J, Park S, Kang SM, Kim JH, Ann SJ, Lee SH. Nlrp3, Csf3, and Edn1 in Macrophage Response to Saturated Fatty Acids and Modified Low-Density Lipoprotein. Korean Circ J 2020; 51:68-80. [PMID: 32975056 PMCID: PMC7779813 DOI: 10.4070/kcj.2020.0117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/09/2020] [Accepted: 07/28/2020] [Indexed: 12/17/2022] Open
Abstract
Background and Objectives The relationship between metabolic stress, inflammation, and cardiovascular disease is being studied steadily. The aim of this study was to evaluate the effect of palmitate (PA) and minimally modified low-density lipoprotein (mmLDL) on macrophages and to identify the associated pathways. Methods J774 macrophages were incubated with PA or mmLDL and lipopolysaccharide (LPS). Secretion of inflammatory chemokines and the expression of corresponding genes were determined. The phosphorylation of extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase was also assessed. RNA sequencing of macrophages was performed to identify the genes regulated by PA or mmLDL. Some of the genes regulated by the 2 agents were validated by knocking down the cells using small interfering RNA. Results PA or mmLDL promoted the secretion of interleukin (IL)-6 and IL-1β in LPS-stimulated macrophages, and this was accompanied by higher phosphorylation of ERK. RNA sequencing revealed dozens of genes that were regulated in this process, such as Csf3 and Edn1, which were affected by PA and mmLDL, respectively. These agents also increased Nlrp3 expression. The effect of Csf3 or Edn1 silencing on inflammation was modest, whereas toll-like receptor (TLR) 4 inhibition reduced a large proportion of macrophage activation. Conclusions We demonstrated that the proinflammatory milieu with high levels of PA or mmLDL promoted macrophage activation and the expression of associated genes such as Nlrp3, Csf3, and Edn1. Although the TLR4 pathway appeared to be most relevant, additional role of other genes in this process provided insights regarding the potential targets for intervention.
Collapse
Affiliation(s)
- Harin Youk
- Graduate Program of Science for Aging, Graduate School of Yonsei University, Seoul, Korea
| | - Miso Kim
- Graduate Program of Science for Aging, Graduate School of Yonsei University, Seoul, Korea
| | - Chan Joo Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jaewon Oh
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sungha Park
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seok Min Kang
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong Ho Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Jin Ann
- Graduate Program of Science for Aging, Graduate School of Yonsei University, Seoul, Korea.
| | - Sang Hak Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
23
|
Leary PJ, Jenny NS, Bluemke DA, Kawut SM, Kronmal RA, Lima JA, Maron BA, Ralph DD, Rayner SG, Ryan JJ, Steinberg ZL, Hinckley Stukovsky KD, Tedford RJ. Endothelin-1, cardiac morphology, and heart failure: the MESA angiogenesis study. J Heart Lung Transplant 2020; 39:45-52. [PMID: 31515065 PMCID: PMC6942224 DOI: 10.1016/j.healun.2019.07.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/10/2019] [Accepted: 07/31/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Circulating levels of endothelin-1 (ET1) are elevated in heart failure and predict poor prognosis. However, it is not clear whether ET1 elevation is an adaptive response, maladaptive response, or an epiphenomenon of heart failure. In this study, we evaluated the relationships between ET1, cardiac morphology, and incident heart failure or cardiovascular death in participants with no evidence of clinical cardiovascular disease at the time ET1 was measured. METHODS AND RESULTS ET1 was measured in 1,361 participants in the Multi-Ethnic Study of Atherosclerosis Angiogenesis Sub-Study. As suggested by linear regression, participants with lower circulating ET1 levels tended to be older, non-white, more likely to have smoked heavily, and less likely to report intentional exercise. Participants with higher ET1 levels had smaller left ventricular end-diastolic volumes (8.9 ml smaller per log increase in ET1, 95% confidence interval 17.1-0.7, p = 0.03) with an increased left ventricular ejection fraction (2.8% per log increase in ET1, 95% confidence interval 0.5%-5.2%, p = 0.02). As suggested by Cox Proportional Hazards estimates, participants with higher ET1 levels had a lower risk for the composite outcome of heart failure or cardiovascular death in models that were unadjusted or had limited adjustment (p = 0.03 and p = 0.05, respectively). Lower risk for heart failure with higher ET1 levels could not be clearly shown in a model including health behaviors. CONCLUSIONS These results suggest, but do not confirm, that elevated levels of circulating ET1 are associated with a more favorable cardiac phenotype. The relationship between ET1 and outcomes was not fully independent of one or more covariates.
Collapse
Affiliation(s)
- Peter J Leary
- University of Washington, Department of Medicine, Seattle, Washington.
| | - Nancy S Jenny
- University of Vermont, Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont
| | | | - Steven M Kawut
- Departments of Medicine and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Richard A Kronmal
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Joao A Lima
- Departments of Medicine and Radiology, Johns Hopkins Hospital, Baltimore, Maryland
| | - Bradley A Maron
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts; Department of Cardiology, Brigham and Women's Hospital, Boston, Massachusetts; Division of Cardiology, Harvard Medical School, Boston, Massachusetts
| | - David D Ralph
- University of Washington, Department of Medicine, Seattle, Washington
| | - Samuel G Rayner
- University of Washington, Department of Medicine, Seattle, Washington
| | - John J Ryan
- Department of Medicine, University of Utah, Salt Lake City, Utah
| | | | | | - Ryan J Tedford
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
24
|
Andersson C, Lin H, Liu C, Levy D, Mitchell GF, Larson MG, Vasan RS. Integrated Multiomics Approach to Identify Genetic Underpinnings of Heart Failure and Its Echocardiographic Precursors. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 12:e002489. [DOI: 10.1161/circgen.118.002489] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background:
Heart failure (HF) may arise from alterations in metabolic, structural, and signaling pathways, but its genetic architecture is incompletely understood. To elucidate potential genetic contributors to cardiac remodeling and HF, we integrated genome-wide single-nucleotide polymorphisms, gene expression, and DNA methylation using a transomics analytical approach.
Methods:
We used robust rank aggregation (where the position of a certain gene in a rank order list [based on statistical significance level] is tested against a randomly shuffled rank order list) to derive an integrative transomic score for each annotated gene associated with a HF trait.
Results:
We evaluated ≤8372 FHS (Framingham Heart Study) participants (54% women; mean age, 55±17 years). Of these, 62 (0.7%) and 35 (0.4%) had prevalent HF with reduced ejection fraction and HF with preserved left ventricular ejection fraction, respectively. During a mean follow-up of 8.5 years (minimum–maximum, 0.005–18.6 years), 223 (2.7%) and 234 (2.8%) individuals developed incident HF with reduced ejection fraction and HF with reduced ejection fraction, respectively. Top genes included
MMP20
and
MTSS1
(promotes actin assembly at intercellular junctions) for left ventricular systolic function;
ITGA9
(receptor for
VCAM1
[vascular cell protein 1]) and
C5
for left ventricular remodeling;
NUP210
(expressed during myogenic differentiation) and
ANK1
(cytoskeletal protein) for diastolic function;
TSPAN16
and
RAB11FIP3
(involved in regulation of actin cytoskeleton) for prevalent HF with reduced ejection fraction;
ANKRD13D
and
TRIM69
for incident HF with reduced ejection fraction;
HPCAL1
and
PTTG1IP
for prevalent HF with reduced ejection fraction; and
ZNF146
(close to the
COX7A1
enzyme) and
ZFP3
(close to
SLC52A1
—the riboflavin transporter) for incident HF with reduced ejection fraction. We tested the HF-related top single-nucleotide polymorphisms in the UK biobank, where
rs77059055
in
TPM1
(minor allele frequency, 0.023; odds ratio, 0.83;
P
=0.002) remained statistically significant upon Bonferroni correction.
Conclusions:
Our integrative transomics approach offers insights into potential molecular and genetic contributors to HF and its precursors. Although several of our candidate genes have been implicated in HF in animal models, independent replication is warranted.
Collapse
Affiliation(s)
- Charlotte Andersson
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Department of Cardiology, Herlev and Gentofte Hospital, Herlev, Denmark (C.A.)
| | - Honghuang Lin
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Section of Computational Biomedicine, Department of Medicine (H.L.), Boston University School of Medicine, MA
| | - Chunyu Liu
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Department of Biostatistics (C.L., M.G.L.), Boston University School of Public Health, MA
| | - Daniel Levy
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (D.L.)
| | | | - Martin G. Larson
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Department of Biostatistics (C.L., M.G.L.), Boston University School of Public Health, MA
| | - Ramachandran S. Vasan
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Sections of Preventive Medicine and Epidemiology and Cardiology, Department of Medicine (R.S.V.), Boston University School of Medicine, MA
- Department of Epidemiology (R.S.V.), Boston University School of Public Health, MA
| |
Collapse
|
25
|
Fujisawa N, Tohyama C, Yoshioka W. Cardiotoxicity induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure through lactation in mice. J Toxicol Sci 2019; 44:505-513. [PMID: 31270306 DOI: 10.2131/jts.44.505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Dioxins are a group of structurally related chemicals that persist in the environment. Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), the most toxic congener, is a suspected risk factor for cardiac diseases in humans. TCDD induces signs of cardiotoxicity in various animals. Mouse models of TCDD exposure suggest cardiotoxicity phenotypes develop differently depending on the timing and time-course of exposure. In order to clarify and characterize the TCDD-induced cardiotoxicity in the developing period, we utilized mouse pups exposed to TCDD. One day after delivery, groups of nursing C57BL/6J dams were orally administered TCDD at a dose of 0 (Control), 20 (TCDD-20), or 80 μg/kg (TCDD-80) body weight (BW). On postnatal days (PNDs) 7 and 21, pups' hearts were examined by histological and gene expression analyses. The TCDD-80 group was found to have a left ventricular remodeling on PND 7, and to develop heart hypertrophy on PND 21. It was accompanied by fibrosis and increased expression of associated genes, such as those for atrial natriuretic peptide (ANP), β-myosin heavy chain (β-MHC), and endothelin-1 (ET-1). These results revealed that TCDD directly induces cardiotoxicity in the postnatal period represented by progressive hypertrophy in which ANP, β-MHC, and ET-1 have potentials to mediate the cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Nozomi Fujisawa
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo.,Faculty of Medicine, University of Tsukuba
| | - Wataru Yoshioka
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo.,Department of Public Health & Environmental Medicine, The Jikei University School of Medicine
| |
Collapse
|
26
|
Sun Y, Fan W, Xue R, Dong B, Liang Z, Chen C, Li J, Wang Y, Zhao J, Huang H, Jiang J, Wu Z, Dai G, Fang R, Yan Y, Yang T, Huang ZP, Dong Y, Liu C. Transcribed Ultraconserved Regions, Uc.323, Ameliorates Cardiac Hypertrophy by Regulating the Transcription of CPT1b (Carnitine Palmitoyl transferase 1b). Hypertension 2019; 75:79-90. [PMID: 31735087 DOI: 10.1161/hypertensionaha.119.13173] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transcribed ultraconserved regions (T-UCRs) are a novel class of long noncoding RNAs transcribed from UCRs, which exhibit 100% DNA sequence conservation among humans, mice, and rats. However, whether T-UCRs regulate cardiac hypertrophy remains unclear. We aimed to explore the effects of T-UCRs on cardiac hypertrophy. First, we performed long noncoding RNA microarray analysis on hearts of mice subjected to sham surgery or aortic banding and found that the T-UCR uc.323 was decreased significantly in mice with aortic banding-induced cardiac hypertrophy. In vitro loss- and gain-of-function experiments demonstrated that uc.323 protected cardiomyocytes against hypertrophy induced by phenylephrine. Additionally, we discovered that mammalian target of rapamycin 1 contributed to phenylephrine-induced uc.323 downregulation and uc.323-mediated cardiomyocyte hypertrophy. We further mapped the possible target genes of uc.323 through global microarray mRNA expression analysis after uc.323 knockdown and found that uc.323 regulated the expression of cardiac hypertrophy-related genes such as CPT1b (Carnitine Palmitoyl transferase 1b). Then, chromatin immunoprecipitation proved that EZH2 (enhancer of zeste homolog 2) bound to the promoter of CPT1b via H3K27me3 (trimethylation of lysine 27 of histone H3) to induce CPT1b downregulation. And overexpression of CPT1b could block uc.323-mediated cardiomyocyte hypertrophy. Finally, we found that uc.323 deficiency induced cardiac hypertrophy. Our results reveal that uc.323 is a conserved T-UCR that inhibits cardiac hypertrophy, potentially by regulating the transcription of CPT1b via interaction with EZH2.
Collapse
Affiliation(s)
- Yu Sun
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,Department of Cardiology, the Second People's Hospital of Guangdong Province, Guangzhou, Guangdong, China (Y.S.).,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Wendong Fan
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Ruicong Xue
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Bin Dong
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Zhuomin Liang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Chen Chen
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Jiayong Li
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Yan Wang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Jingjing Zhao
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Huiling Huang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Jingzhou Jiang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Zexuan Wu
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Gang Dai
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Rong Fang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Youchen Yan
- Department of Cardiology, Center for Translational Medicine (Y.Y., T.Y.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Tiqun Yang
- Department of Cardiology, Center for Translational Medicine (Y.Y., T.Y.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhan-Peng Huang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Yugang Dong
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Chen Liu
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| |
Collapse
|
27
|
Affiliation(s)
- Takumi J Matsubara
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Katsuhito Fujiu
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo.,Department of Advanced Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| |
Collapse
|
28
|
de Lucia C, Gambino G, Petraglia L, Elia A, Komici K, Femminella GD, D'Amico ML, Formisano R, Borghetti G, Liccardo D, Nolano M, Houser SR, Leosco D, Ferrara N, Koch WJ, Rengo G. Long-Term Caloric Restriction Improves Cardiac Function, Remodeling, Adrenergic Responsiveness, and Sympathetic Innervation in a Model of Postischemic Heart Failure. Circ Heart Fail 2019. [PMID: 29535114 DOI: 10.1161/circheartfailure.117.004153] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Caloric restriction (CR) has been described to have cardioprotective effects and improve functional outcomes in animal models and humans. Chronic ischemic heart failure (HF) is associated with reduced cardiac sympathetic innervation, dysfunctional β-adrenergic receptor signaling, and decreased cardiac inotropic reserve. We tested the effects of a long-term CR diet, started late after myocardial infarction on cardiac function, sympathetic innervation, and β-adrenergic receptor responsiveness in a rat model of postischemic HF. METHODS AND RESULTS Adult male rats were randomly assigned to myocardial infarction or sham operation and 4 weeks later were further randomized to a 1-year CR or normal diet. One year of CR resulted in a significant reduction in body weight, heart weight, and heart weight/tibia length ratio when compared with normal diet in HF groups. At the end of the study period, echocardiography and histology revealed that HF animals under the CR diet had ameliorated left ventricular remodeling compared with HF rats fed with normal diet. Invasive hemodynamic showed a significant improvement of cardiac inotropic reserve in CR HF rats compared with HF-normal diet animals. Importantly, CR dietary regimen was associated with a significant increase of cardiac sympathetic innervation and with normalized cardiac β-adrenergic receptor levels in HF rats when compared with HF rats on the standard diet. CONCLUSIONS We demonstrate, for the first time, that chronic CR, when started after HF established, can ameliorate cardiac dysfunction and improve inotropic reserve. At the molecular level, we find that chronic CR diet significantly improves sympathetic cardiac innervation and β-adrenergic receptor levels in failing myocardium.
Collapse
Affiliation(s)
- Claudio de Lucia
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Giuseppina Gambino
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Laura Petraglia
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Andrea Elia
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Klara Komici
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Grazia Daniela Femminella
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Maria Loreta D'Amico
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Roberto Formisano
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Giulia Borghetti
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Daniela Liccardo
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Maria Nolano
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Steven R Houser
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Dario Leosco
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Nicola Ferrara
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.)
| | - Walter J Koch
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.).
| | - Giuseppe Rengo
- From the Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (C.d.L., G.G., L.P., A.E., K.K., G.D.F., M.L.D., R.F., D. Liccardo, D. Leosco, N.F., G.R.); Center for Translational Medicine (C.d.L., D. Liccardo, W.J.K.), Department of Pharmacology (C.d.L., D. Liccardo, W.J.K.) and Cardiovascular Research Center (G.B., S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.G., A.E., M.L.D., M.N., N.F., G.R.); and Neurology Imaging Unit, Imperial College London, United Kingdom (G.D.F.).
| |
Collapse
|
29
|
He M, Wang J, Yin Z, Zhao Y, Hou H, Fan J, Li H, Wen Z, Tang J, Wang Y, Wang DW, Chen C. MiR-320a induces diabetic nephropathy via inhibiting MafB. Aging (Albany NY) 2019; 11:3055-3079. [PMID: 31102503 PMCID: PMC6555468 DOI: 10.18632/aging.101962] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/06/2019] [Indexed: 12/17/2022]
Abstract
Multiple studies indicate that microRNAs (miRNAs) are involved in diabetes. However, the roles of miRNA in the target organ damages in diabetes remain unclear. This study investigated the functions of miR-320a in diabetic nephropathy (DN). In this study, db/db mice were used to observe the changes in podocytes and their function in vivo, as well as in cultured mouse podocyte cells (MPC5) exposed to high glucose in vitro. To further explore the role of miR-320a in DN, recombinant adeno-associated viral particle was administered intravenously to manipulate the expression of miR-320a in db/db mice. Overexpression of miR-320a markedly promoted podocyte loss and dysfunction in DN, including mesangial expansion and increased levels of proteinuria, serum creatinine and urea nitrogen. Furthermore, MafB was identified as a direct target of miR-320a through AGO2 co-immunoprecipitation, luciferase reporter assay, and Western blotting. Moreover, re-expression of MafB rescued miR-320a-induced podocyte loss and dysfunction by upregulating the expressions of Nephrin and glutathione peroxidase 3 (Gpx3). Our data indicated that miR-320a aggravated renal disfunction in DN by targeting MafB and downregulating Nephrin and Gpx3 in podocytes, which suggested that miR-320a could be a potential therapeutic target of diabetic nephropathy.
Collapse
Affiliation(s)
- Mengying He
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jin Wang
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhongwei Yin
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanru Zhao
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huiying Hou
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiahui Fan
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huaping Li
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zheng Wen
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiarong Tang
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Wang
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chen Chen
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
30
|
Beneficial effects of nicotinamide on the mouse model of preeclampsia. OA JOURNAL OF PREGNANCY AND CHILD CARE 2018; 1. [PMID: 34268502 PMCID: PMC8278325 DOI: 10.33118/oaj.preg.2019.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Preeclampsia (PE) is a pregnancy related disorder that is characterized by hypertension and proteinuria in the mother. It is associated with impaired coagulation and liver function, and a variety of other detrimental effects. In severe cases without treatment, PE can progress to eclampsia and result in seizures, a life-threatening condition. Although the etiology of PE is largely unknown, sFlt-1 (soluble vascular endothelial growth factor receptor 1) released by the impaired placenta resulting from insufficient perfusion plays a critical role in PE, and phenotypes of PE can be induced by experimentally increasing sFlt-1. We and other investigators have proposed that endothelin-1 (ET-1) system is the mediator of the pathological effects of excess sFlt-1, and antagonists of ET-1 receptor block the effects of sFlt-1. Unfortunately, this class of drugs is teratogenic and unsuitable for treating pregnant women. Nicotinamide is a naturally occurring derivative of vitamin B3 in the body and inhibits ADP-ribosyl cyclase, which is activated by the ET-1 receptor. Therefore, if utilized, it would be expected to play a beneficial role in PE. In mouse models of PE, a high dose of nicotinamide shows great success in lowering blood pressure, correcting renal function and structure, prolonging pregnancy as well as increasing fetal weight/number. Nicotinamide, being generally regarded as safe, could be a promising substance to further investigate for use in clinical trials.
Collapse
|
31
|
Cannata' A, Merlo M, Artico J, Gentile P, Camparini L, Cristallini J, Porcari A, Loffredo F, Sinagra G. Cardiovascular aging: the unveiled enigma from bench to bedside. J Cardiovasc Med (Hagerstown) 2018; 19:517-526. [PMID: 30024423 DOI: 10.2459/jcm.0000000000000694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
: The rapid increase in the median age of the world's population requires particular attention towards older and more fragile people. Cardiovascular risk factors, time and comorbidities play a vicious role in the development of heart failure, both with reduced and preserved ejection fraction, in the elderly. Understanding the mechanisms underlying the pathophysiological processes observed with aging is pivotal to target those patients and their therapeutic needs properly. This review aims to investigate and to dissect the main pathways leading to the aging cardiomyopathy, helping to understand the relationship from bench to bedside of the clinical phenotype.
Collapse
Affiliation(s)
- Antonio Cannata'
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste.,International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Marco Merlo
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Jessica Artico
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Piero Gentile
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Luca Camparini
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Jacopo Cristallini
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Aldostefano Porcari
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Francesco Loffredo
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste.,International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Gianfranco Sinagra
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| |
Collapse
|
32
|
Hippo Signaling Plays an Essential Role in Cell State Transitions during Cardiac Fibroblast Development. Dev Cell 2018; 45:153-169.e6. [PMID: 29689192 DOI: 10.1016/j.devcel.2018.03.019] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 02/02/2018] [Accepted: 03/26/2018] [Indexed: 12/14/2022]
Abstract
During development, progenitors progress through transition states. The cardiac epicardium contains progenitors of essential non-cardiomyocytes. The Hippo pathway, a kinase cascade that inhibits the Yap transcriptional co-factor, controls organ size in developing hearts. Here, we investigated Hippo kinases Lats1 and Lats2 in epicardial diversification. Epicardial-specific deletion of Lats1/2 was embryonic lethal, and mutant embryos had defective coronary vasculature remodeling. Single-cell RNA sequencing revealed that Lats1/2 mutant cells failed to activate fibroblast differentiation but remained in an intermediate cell state with both epicardial and fibroblast characteristics. Lats1/2 mutant cells displayed an arrested developmental trajectory with persistence of epicardial markers and expanded expression of Yap targets Dhrs3, an inhibitor of retinoic acid synthesis, and Dpp4, a protease that modulates extracellular matrix (ECM) composition. Genetic and pharmacologic manipulation revealed that Yap inhibits fibroblast differentiation, prolonging a subepicardial-like cell state, and promotes expression of matricellular factors, such as Dpp4, that define ECM characteristics.
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW To review the most recent data on the development of endothelin receptor antagonists (ERAs) for the treatment of hypertension and the management of diabetic nephropathy RECENT FINDINGS: Recent reviews and meta-analyses of experimental and clinical data obtained with ERAs confirmed that endothelin receptor blockade is associated with significant decreases in blood pressure in essential hypertension but also in resistant hypertension. In addition, in patients with diabetic nephropathy, ERAs induce significant 30-40% decreases in albuminuria when administered on top of blockers of the renin-angiotensin system. Yet, the benefits of ERAs have often been limited by their tolerability profile, essentially fluid retention and the development of edema and liver toxicity. Hence, several programs have been interrupted. Today, only one ERA, aprocitentan, is still under development for the treatment of resistant hypertension. Regarding the place of ERAs in the management of diabetic nephropathy, the results of the SONAR trial with atrasentan are eagerly awaited but the recent interruption of this trial because of insufficient events is worrisome, as one might not obtain all the expected information for this major trial. Blockade of endothelin receptor have a high potential in the treatment of hypertension and the prevention of the progression of renal diseases such as diabetic nephropathy. Today, the number of clinical programs investigating the potential benefits of ERAs is limited and more data must be obtained to define the real place of ERAs in these indications.
Collapse
Affiliation(s)
- Michel Burnier
- Department of Medicine, Service of Nephrology and Hypertension, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon 17, 1011, Lausanne, Switzerland.
| |
Collapse
|
34
|
Segers VFM, Brutsaert DL, De Keulenaer GW. Cardiac Remodeling: Endothelial Cells Have More to Say Than Just NO. Front Physiol 2018; 9:382. [PMID: 29695980 PMCID: PMC5904256 DOI: 10.3389/fphys.2018.00382] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/28/2018] [Indexed: 12/12/2022] Open
Abstract
The heart is a highly structured organ consisting of different cell types, including myocytes, endothelial cells, fibroblasts, stem cells, and inflammatory cells. This pluricellularity provides the opportunity of intercellular communication within the organ, with subsequent optimization of its function. Intercellular cross-talk is indispensable during cardiac development, but also plays a substantial modulatory role in the normal and failing heart of adults. More specifically, factors secreted by cardiac microvascular endothelial cells modulate cardiac performance and either positively or negatively affect cardiac remodeling. The role of endothelium-derived small molecules and peptides—for instance NO or endothelin-1—has been extensively studied and is relatively well defined. However, endothelial cells also secrete numerous larger proteins. Information on the role of these proteins in the heart is scattered throughout the literature. In this review, we will link specific proteins that modulate cardiac contractility or cardiac remodeling to their expression by cardiac microvascular endothelial cells. The following proteins will be discussed: IL-6, periostin, tenascin-C, thrombospondin, follistatin-like 1, frizzled-related protein 3, IGF-1, CTGF, dickkopf-3, BMP-2 and−4, apelin, IL-1β, placental growth factor, LIF, WISP-1, midkine, and adrenomedullin. In the future, it is likely that some of these proteins can serve as markers of cardiac remodeling and that the concept of endothelial function and dysfunction might have to be redefined as we learn more about other factors secreted by ECs besides NO.
Collapse
Affiliation(s)
- Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| | - Dirk L Brutsaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, Middelheim Hospital, Antwerp, Belgium
| |
Collapse
|
35
|
Li F, Kakoki M, Smid M, Boggess K, Wilder J, Hiller S, Bounajim C, Parnell SE, Sulik KK, Smithies O, Maeda-Smithies N. Causative Effects of Genetically Determined High Maternal/Fetal Endothelin-1 on Preeclampsia-Like Conditions in Mice. Hypertension 2018; 71:894-903. [PMID: 29610266 DOI: 10.1161/hypertensionaha.117.10849] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 01/22/2018] [Accepted: 03/06/2018] [Indexed: 12/20/2022]
Abstract
Endothelin-1 (ET-1) is implicated in the pathophysiology of preeclampsia. An association between an EDN1 gene polymorphism with high ET-1 and preeclampsia was reported in humans, but their cause and effect relationships have not been defined. We examined the pregnancy effects in mice with a modified Edn1 allele that increases mRNA stability and thus ET-1 production. Heterozygous Edn1H/+ females showed no obvious abnormalities before pregnancy, but when mated with wild-type (WT) males developed a full spectrum of preeclampsia-like phenotypes, including increased systolic blood pressure, proteinuria, glomerular endotheliosis, and intrauterine fetal growth restriction. At 7.5 days post-coitus, the embryos from Edn1H/+ dams, regardless of their Edn1 genotype, lagged 12 hours in development compared with embryos from WT dams, had disoriented ectoplacental cones, and retained high E-cadherin expression. In contrast, WT females mated with Edn1H/+ males, which also carried half of the fetuses with Edn1H/+ genotype, showed a mild systolic blood pressure increase only. These WT dams had 2× higher plasma soluble fms-like tyrosine kinase-1 than WT dams mated with WT males. In human first trimester trophoblast cells, pharmacological doses of ET-1 increased the cellular sFlt1 transcripts and protein secretion via both type A and B ET-1 receptors. Our data demonstrate that high maternal ET-1 production causes preeclampsia-like phenotypes during pregnancy, affecting both initial stage of trophoblast differentiation/invasion and maternal peripheral vasculature during late gestation. High fetal ET-1 production, however, could cause increased soluble fms-like tyrosine kinase-1 in the maternal circulation and contribute to blood pressure elevation.
Collapse
Affiliation(s)
- Feng Li
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.).
| | - Masao Kakoki
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.)
| | - Marcela Smid
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.)
| | - Kim Boggess
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.)
| | - Jennifer Wilder
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.)
| | - Sylvia Hiller
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.)
| | - Carol Bounajim
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.)
| | - Scott E Parnell
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.)
| | - Kathleen K Sulik
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.)
| | - Oliver Smithies
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.)
| | - Nobuyo Maeda-Smithies
- From the Department of Pathology and Laboratory Medicine (F.L., M.K., J.W., S.H., O.S., N.M.-S.), Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology (K.B.), School of Medicine (C.B.), and Department of Cell Biology and Physiology (S.E.P., K.K.S.), University of North Carolina; and Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City (M.S.)
| |
Collapse
|
36
|
Xu M, Lu YP, Hasan A, Hocher B. Plasma ET-1 Concentrations are Elevated in Patients with Hypertension – Meta-Analysis of Clinical Studies. Kidney Blood Press Res 2017; 42:304-313. [DOI: 10.1159/000477572] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/08/2017] [Indexed: 11/19/2022] Open
|
37
|
Wang X, Chen W, Zhang J, Khan A, Li L, Huang F, Qiu Z, Wang L, Chen X. Critical Role of ADAMTS2 (A Disintegrin and Metalloproteinase With Thrombospondin Motifs 2) in Cardiac Hypertrophy Induced by Pressure Overload. Hypertension 2017; 69:1060-1069. [PMID: 28373586 DOI: 10.1161/hypertensionaha.116.08581] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 10/29/2016] [Accepted: 03/04/2017] [Indexed: 12/31/2022]
Abstract
ADAMTS2 (A Disintegrin and Metalloproteinase With Thrombospondin Motifs 2) is recognized as a metalloproteinase that promotes the cleavage of amino propeptides of types I, II, III, and V procollagens. However, the role of ADAMTS2 in the heart has not yet been defined. Herein, we observed the upregulated expression of ADAMTS2 in failing human hearts and hypertrophic murine hearts. Mice lacking ADAMTS2 display exacerbated cardiac hypertrophy on pressure overload-induced hypertrophic response, whereas mice with cardiac-specific overexpression of ADAMTS2 display alleviation of this detrimental phenotype. Consistent with these results, in vitro loss or gain of function experiments in neonatal rat cardiomyocytes confirmed that ADAMTS2 negatively regulates cardiomyocyte hypertrophy in response to Ang II. Mechanistically, blockage of the PI3K (phosphoinositide 3-kinase)/AKT (protein kinase B)-dependent signaling pathway with specific inhibitors both in vivo and in vitro could rescue the aggravated hypertrophic response to the loss of ADAMTS2. Collectively, we propose that ADAMTS2 regulates the hypertrophic response through inhibiting the activation of the PI3K/AKT-dependent signaling pathway. Because ADAMTS2 is an extracellular protein, it could be effectively manipulated using pharmacological means to modulate cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiaodi Wang
- From the Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, People's Republic of China
| | - Wen Chen
- From the Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, People's Republic of China
| | - Jie Zhang
- From the Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, People's Republic of China
| | - Aiman Khan
- From the Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, People's Republic of China
| | - Liangpeng Li
- From the Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, People's Republic of China
| | - Fuhua Huang
- From the Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, People's Republic of China
| | - Zhibing Qiu
- From the Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, People's Republic of China
| | - Liming Wang
- From the Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, People's Republic of China
| | - Xin Chen
- From the Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, People's Republic of China.
| |
Collapse
|
38
|
Jia W, Zhang Y, Sui M, Zheng J, Guo Q, Sun Q, Guo Q, Ji Z, Wang Z, Liu Q. Effect of acupuncture on the genetic expression of myocardial endothelin-1 and atrial natriuretic peptide in rats with stress-induced prehypertension. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2017. [DOI: 10.1016/j.jtcms.2017.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
39
|
Cheng Z, Zhu Q, Dee R, Opheim Z, Mack CP, Cyr DM, Taylor JM. Focal Adhesion Kinase-mediated Phosphorylation of Beclin1 Protein Suppresses Cardiomyocyte Autophagy and Initiates Hypertrophic Growth. J Biol Chem 2016; 292:2065-2079. [PMID: 27994061 DOI: 10.1074/jbc.m116.758268] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/15/2016] [Indexed: 01/11/2023] Open
Abstract
Autophagy is an evolutionarily conserved intracellular degradation/recycling system that is essential for cellular homeostasis but is dysregulated in a number of diseases, including myocardial hypertrophy. Although it is clear that limiting or accelerating autophagic flux can result in pathological cardiac remodeling, the physiological signaling pathways that fine-tune cardiac autophagy are poorly understood. Herein, we demonstrated that stimulation of cardiomyocytes with phenylephrine (PE), a well known hypertrophic agonist, suppresses autophagy and that activation of focal adhesion kinase (FAK) is necessary for PE-stimulated autophagy suppression and subsequent initiation of hypertrophic growth. Mechanistically, we showed that FAK phosphorylates Beclin1, a core autophagy protein, on Tyr-233 and that this post-translational modification limits Beclin1 association with Atg14L and reduces Beclin1-dependent autophagosome formation. Remarkably, although ectopic expression of wild-type Beclin1 promoted cardiomyocyte atrophy, expression of a Y233E phosphomimetic variant of Beclin1 failed to affect cardiomyocyte size. Moreover, genetic depletion of Beclin1 attenuated PE-mediated/FAK-dependent initiation of myocyte hypertrophy in vivo Collectively, these findings identify FAK as a novel negative regulator of Beclin1-mediated autophagy and indicate that this pathway can facilitate the promotion of compensatory hypertrophic growth. This novel mechanism to limit Beclin1 activity has important implications for treating a variety of pathologies associated with altered autophagic flux.
Collapse
Affiliation(s)
| | | | | | | | | | - Douglas M Cyr
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Joan M Taylor
- From the Department of Pathology, .,McAllister Heart Institute, and
| |
Collapse
|
40
|
Weng CH, Chung FP, Chen YC, Lin SF, Huang PH, Kuo TBJ, Hsu WH, Su WC, Sung YL, Lin YJ, Chang SL, Lo LW, Yeh HI, Chen YJ, Hong YR, Chen SA, Hu YF. Pleiotropic Effects of Myocardial MMP-9 Inhibition to Prevent Ventricular Arrhythmia. Sci Rep 2016; 6:38894. [PMID: 27966586 PMCID: PMC5155273 DOI: 10.1038/srep38894] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/14/2016] [Indexed: 01/01/2023] Open
Abstract
Observational studies have established a strong association between matrix metalloproteinase-9 (MMP-9) and ventricular arrhythmia. However, whether MMP-9 has a causal link to ventricular arrhythmia, as well as the underlying mechanism, remains unclear. Here, we investigated the mechanistic involvement of myocardial MMP-9 in the pathophysiology of ventricular arrhythmia. Increased levels of myocardial MMP-9 are linked to ventricular arrhythmia attacks after angiotensin II (Ang II) treatment. MMP-9-deficient mice were protected from ventricular arrhythmia. Increased expressions of protein kinase A (PKA) and ryanodine receptor phosphorylation at serine 2808 (pS2808) were correlated with inducible ventricular arrhythmia. MMP-9 deficiency consistently prevented PKA and pS2808 increases after Ang II treatment and reduced ventricular arrhythmia. Calcium dynamics were examined via confocal imaging in isolated murine cardiomyocytes. MMP-9 inhibition prevents calcium leakage from the sarcoplasmic reticulum and reduces arrhythmia-like irregular calcium transients via protein kinase A and ryanodine receptor phosphorylation. Human induced pluripotent stem cell-derived cardiomyocytes similarly show that MMP-9 inhibition prevents abnormal calcium leakage. Myocardial MMP-9 inhibition prevents ventricular arrhythmia through pleiotropic effects, including the modulation of calcium homeostasis and reduced calcium leakage.
Collapse
Affiliation(s)
- Ching-Hui Weng
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Fa-Po Chung
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Shien-Fong Lin
- Institute of Biomedical Engineering, National Chiao-Tung University, Hsinchu, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Terry B. J. Kuo
- Institute of Brain Science, National Yang Ming University, Taipei, Taiwan
| | - Wei-Hsuan Hsu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen-Cheng Su
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Ling Sung
- Institute of Biomedical Engineering, National Chiao-Tung University, Hsinchu, Taiwan
| | - Yenn-Jiang Lin
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Lin Chang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Li-Wei Lo
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hung-I Yeh
- Division of Cardiology, Department of Internal Medicine, Mackay Memorial Hospital, Mackay Medical College, Taipei, Taiwan
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei Taiwan
| | - Yi-Ren Hong
- Faculty of Medicine, Department of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Ann Chen
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Feng Hu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
41
|
Wang Y, Zhang Y, Zhu CG, Guo YL, Huang QJ, Wu NQ, Gao Y, Qing P, Liu G, Dong Q, Li JJ. Big endothelin-1 level is a useful marker for predicting the presence of isolated coronary artery ectasia. Biomarkers 2016; 22:331-336. [PMID: 27885846 DOI: 10.1080/1354750x.2016.1265001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
CONTEXT Endothelin-1(ET-1) has been implicated in coronary artery disease (CAD) and may be associated with coronary artery ectasia (CAE). OBJECTIVE To clarify the relationship between big ET-1 and isolated CAE. METHODS We measured big ET-1 with ELISA in 216 patients (CAE, n = 72; CAD, n = 72; normal, n = 72) and evaluated the link with isolated CAE. RESULTS The level of plasma big ET-1 was significantly higher in patients with isolated CAE (p < 0.001). Big ET-1 was strongly and independently associated with CAE by multivariate analysis (OR 95%CI: 1.026 (1.018-1.034), p = 0.000). CONCLUSIONS Big ET-1 may be a useful predictor for the presence of isolated CAE.
Collapse
Affiliation(s)
- Yao Wang
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Yan Zhang
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Cheng-Gang Zhu
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Yuan-Lin Guo
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Qiao-Juan Huang
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Na-Qiong Wu
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Ying Gao
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Ping Qing
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Geng Liu
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Qian Dong
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Jian-Jun Li
- a Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| |
Collapse
|
42
|
Antoniak S, Cardenas JC, Buczek LJ, Church FC, Mackman N, Pawlinski R. Protease-Activated Receptor 1 Contributes to Angiotensin II-Induced Cardiovascular Remodeling and Inflammation. Cardiology 2016; 136:258-268. [PMID: 27880950 DOI: 10.1159/000452269] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 09/23/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Angiotensin II (Ang II) plays an important role in cardiovascular disease. It also leads to the activation of coagulation. The coagulation protease thrombin induces cellular responses by activating protease-activated receptor 1 (PAR-1). We investigated whether PAR-1 contributes to Ang II-induced cardiovascular remodeling and inflammation. METHODS AND RESULTS PAR-1+/+ (wild-type; WT) and PAR-1-/- mice were infused with Ang II (600 ng/kg/min) for up to 4 weeks. In WT mice, this dose of Ang II did not cause a significant increase in blood pressure but it did cause pathological changes in both the aorta and the heart. Ang II infusion resulted in vascular remodeling of the aorta, demonstrated by a significant increase in medial wall thickening and perivascular fibrosis. Importantly, both parameters were significantly attenuated by PAR-1 deficiency. Furthermore, perivascular fibrosis around coronary vessels was reduced in Ang II-treated PAR-1-/- mice compared to WT mice. In addition, PAR-1 deficiency significantly attenuated Ang II induction of inflammatory cytokines and profibrotic genes in the aortas compared to WT mice. Finally, PAR-1 deficiency had no effect on Ang II-induced heart hypertrophy. However, the heart function measured by fractional shortening was less impaired in PAR-1-/- mice than in WT mice. CONCLUSION Our data indicate that PAR-1 plays a significant role in cardiovascular remodeling mediated by a blood pressure-independent action of Ang II.
Collapse
Affiliation(s)
- Silvio Antoniak
- UNC McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | | | | | | |
Collapse
|
43
|
Fattori V, Serafim KGG, Zarpelon AC, Borghi SM, Pinho-Ribeiro FA, Alves-Filho JC, Cunha TM, Cunha FQ, Casagrande R, Verri WA. Differential regulation of oxidative stress and cytokine production by endothelin ET A and ET B receptors in superoxide anion-induced inflammation and pain in mice. J Drug Target 2016; 25:264-274. [PMID: 27701898 DOI: 10.1080/1061186x.2016.1245308] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The present study investigated whether endothelin-1 acts via ETA or ETB receptors to mediate superoxide anion-induced pain and inflammation. Mice were treated with clazosentan (ETA receptor antagonist) or BQ-788 (ETB receptor antagonist) prior to stimulation with the superoxide anion donor, KO2. Intraplantar treatment with 30 nmol of clazosentan or BQ-788 reduced mechanical hyperalgesia (47% and 42%), thermal hyperalgesia (68% and 76%), oedema (50% and 30%); myeloperoxidase activity (64% and 32%), and overt-pain like behaviours, such as paw flinching (42% and 42%) and paw licking (38% and 62%), respectively. Similarly, intraperitoneal treatment with 30 nmol of clazosentan or BQ-788 reduced leukocyte recruitment to the peritoneal cavity (58% and 32%) and abdominal writhing (81% and 77%), respectively. Additionally, intraplantar treatment with clazosentan or BQ-788 decreased spinal (45% and 41%) and peripheral (47% and 47%) superoxide anion production as well as spinal (47% and 47%) and peripheral (33% and 54%) lipid peroxidation, respectively. Intraplantar treatment with clazosentan, but not BQ-788, reduced spinal (71%) and peripheral (51%) interleukin-1 beta as well as spinal (59%) and peripheral (50%) tumor necrosis factor-alpha production. Therefore, the present study unveils the differential mechanisms by which ET-1, acting on ETA or ETB receptors, regulates superoxide anion-induced inflammation and pain.
Collapse
Affiliation(s)
- Victor Fattori
- a Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina , Londrina , Brazil
| | - Karla G G Serafim
- a Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina , Londrina , Brazil
| | - Ana C Zarpelon
- a Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina , Londrina , Brazil
| | - Sergio M Borghi
- a Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina , Londrina , Brazil
| | - Felipe A Pinho-Ribeiro
- a Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina , Londrina , Brazil
| | - José C Alves-Filho
- b Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto , Universidade de São Paulo , Ribeirão Preto , Brazil
| | - Thiago M Cunha
- b Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto , Universidade de São Paulo , Ribeirão Preto , Brazil
| | - Fernando Q Cunha
- b Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto , Universidade de São Paulo , Ribeirão Preto , Brazil
| | - Rúbia Casagrande
- c Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde , Universidade Estadual de Londrina , Londrina , Brazil
| | - Waldiceu A Verri
- a Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina , Londrina , Brazil
| |
Collapse
|
44
|
Cannatà A, Camparini L, Sinagra G, Giacca M, Loffredo FS. Pathways for salvage and protection of the heart under stress: novel routes for cardiac rejuvenation. Cardiovasc Res 2016; 111:142-53. [DOI: 10.1093/cvr/cvw106] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/10/2016] [Indexed: 01/07/2023] Open
|
45
|
Davenport AP, Hyndman KA, Dhaun N, Southan C, Kohan DE, Pollock JS, Pollock DM, Webb DJ, Maguire JJ. Endothelin. Pharmacol Rev 2016; 68:357-418. [PMID: 26956245 PMCID: PMC4815360 DOI: 10.1124/pr.115.011833] [Citation(s) in RCA: 523] [Impact Index Per Article: 58.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The endothelins comprise three structurally similar 21-amino acid peptides. Endothelin-1 and -2 activate two G-protein coupled receptors, ETA and ETB, with equal affinity, whereas endothelin-3 has a lower affinity for the ETA subtype. Genes encoding the peptides are present only among vertebrates. The ligand-receptor signaling pathway is a vertebrate innovation and may reflect the evolution of endothelin-1 as the most potent vasoconstrictor in the human cardiovascular system with remarkably long lasting action. Highly selective peptide ETA and ETB antagonists and ETB agonists together with radiolabeled analogs have accurately delineated endothelin pharmacology in humans and animal models, although surprisingly no ETA agonist has been discovered. ET antagonists (bosentan, ambrisentan) have revolutionized the treatment of pulmonary arterial hypertension, with the next generation of antagonists exhibiting improved efficacy (macitentan). Clinical trials continue to explore new applications, particularly in renal failure and for reducing proteinuria in diabetic nephropathy. Translational studies suggest a potential benefit of ETB agonists in chemotherapy and neuroprotection. However, demonstrating clinical efficacy of combined inhibitors of the endothelin converting enzyme and neutral endopeptidase has proved elusive. Over 28 genetic modifications have been made to the ET system in mice through global or cell-specific knockouts, knock ins, or alterations in gene expression of endothelin ligands or their target receptors. These studies have identified key roles for the endothelin isoforms and new therapeutic targets in development, fluid-electrolyte homeostasis, and cardiovascular and neuronal function. For the future, novel pharmacological strategies are emerging via small molecule epigenetic modulators, biologicals such as ETB monoclonal antibodies and the potential of signaling pathway biased agonists and antagonists.
Collapse
Affiliation(s)
- Anthony P Davenport
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Kelly A Hyndman
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Neeraj Dhaun
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Christopher Southan
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Donald E Kohan
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Jennifer S Pollock
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - David M Pollock
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - David J Webb
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Janet J Maguire
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| |
Collapse
|
46
|
Liu X, Bai C, Ding X, Wei Z, Guo H, Li G. Microarray Analysis of the Gene Expression Profile and Lipid Metabolism in Fat-1 Transgenic Cattle. PLoS One 2015; 10:e0138874. [PMID: 26426396 PMCID: PMC4591129 DOI: 10.1371/journal.pone.0138874] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 09/04/2015] [Indexed: 11/30/2022] Open
Abstract
Long-chain n-3 polyunsaturated fatty acids (n-3 PUFAs) are beneficial for human health. However, humans and mammals are unable to synthesize n-3 PUFAs because they lack the n-3 desaturase gene fat-1 and must therefore obtain this type of fatty acid through their diet. Through the production of fat-1 transgenic animals, it is possible to obtain animal products that are rich in n-3 PUFAs, such as meat and milk. The aim of this study was to analyze the gene expression profile and the mechanism of lipid metabolism in fat-1 transgenic cattle and to accumulate important basic data that are required to obtain more efficient fat-1 transgenic cattle. Transcriptome profiling of fat-1 transgenic and wild-type cattle identified differentially expressed genes that are involved in 90 biological pathways, eight pathways of which were related to lipid metabolism processes 36 genes of which were related to lipid metabolism. This analysis also identified 11 significantly enriched genes that were involved in the peroxisome proliferator-activated receptor signaling pathway. These findings were verified by quantitative polymerase chain reaction. The information obtained in this study indicated that the introduction of an exogenous fat-1 gene into cattle affects the gene expression profile and the process of lipid metabolism in these animals. These results may provide important insights into how an exogenous fat-1 gene synthesizes n-3 PUFAs in transgenic cattle and other mammals.
Collapse
Affiliation(s)
- Xinfeng Liu
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
- College of Animal Science and Animal Medicine, Tianjin Agriculture University, Tianjin, China
| | - Chunling Bai
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Xiangbin Ding
- College of Animal Science and Animal Medicine, Tianjin Agriculture University, Tianjin, China
| | - Zhuying Wei
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Hong Guo
- College of Animal Science and Animal Medicine, Tianjin Agriculture University, Tianjin, China
- * E-mail: (HG); (GPL)
| | - Guangpeng Li
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
- * E-mail: (HG); (GPL)
| |
Collapse
|
47
|
Yu AP, Tam BT, Yau WY, Chan KS, Yu SS, Chung TL, Siu PM. Association of endothelin-1 and matrix metallopeptidase-9 with metabolic syndrome in middle-aged and older adults. Diabetol Metab Syndr 2015; 7:111. [PMID: 26692905 PMCID: PMC4676096 DOI: 10.1186/s13098-015-0108-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/25/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) contains a cluster of cardiovascular risk factors. People with MetS are more susceptible to cardiovascular disease, diabetes mellitus, and cancer. Endothelin-1 (ET-1) and matrix metallopeptidase-9 (MMP-9) have been implicated in the development of cardiovascular diseases, diabetes mellitus and cancers. This cross-sectional study aimed to examine the association of ET-1 and MMP-9 with MetS in middle-aged and older Hong Kong Chinese adults. METHODS 149 adults aged 50 to 92 (n = 75 for non-MetS group and n = 74 for MetS group) were examined. All subjects were screened for MetS according to the diagnostic guideline of the United States National Cholesterol Education Program (NCEP) Expert Panel Adult Treatment Panel (ATP) III criteria. Serum levels of ET-1 and MMP-9 were measured. Independent t test was used to detect differences between non-MetS and MetS groups and between subjects with or without certain metabolic abnormality. The association of the serum concentration of MMP-9 and ET-1 with MetS parameters were examined by Pearson's correlation analysis. RESULTS Serum level of ET-1 is higher in MetS-positive subjects and in subjects with high blood pressure, elevated fasting blood glucose, and central obesity. The serum concentration of MMP-9 is higher in subjects positively diagnosed with MetS and subjects with high blood pressure, elevated fasting blood glucose, low blood high-density lipoprotein-cholesterol (HDL-C), high blood triglycerides, and central obesity. Correlation analyses revealed that serum concentration of ET-1 is positively correlated to systolic blood pressure, waist circumference, fasting blood glucose, and age whereas it is negatively correlated to HDL-C. MMP-9 is positively correlated to systolic blood pressure, waist circumference, fasting blood glucose, and age whereas it is negatively correlated to HDL-C. CONCLUSION Serum ET-1 is higher in subjects with hypertension, hyperglycemia, central obesity or MetS. Serum MMP-9 is higher in subjects diagnosed with MetS or having either one of the MetS parameters. Both circulating levels of ET-1 and MMP-9 are correlated to systolic blood pressure, waist circumference, fasting blood glucose, HDL-C, and age. Further research is needed to fully dissect the role of ET-1 and MMP-9 in the development of cancers, diabetes and cardiovascular disease in relation to MetS.
Collapse
Affiliation(s)
- A. P. Yu
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - B. T. Tam
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - W. Y. Yau
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - K. S. Chan
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - S. S. Yu
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - T. L. Chung
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - P. M. Siu
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| |
Collapse
|