1
|
Davidson SM, Andreadou I, Antoniades C, Bartunek J, Basso C, Brundel BJJM, Byrne RA, Chiva-Blanch G, da Costa Martins P, Evans PC, Girão H, Giricz Z, Gollmann-Tepeköylü C, Guzik T, Gyöngyösi M, Hübner N, Joner M, Kleinbongard P, Krieg T, Liehn E, Madonna R, Maguy A, Paillard M, Pesce M, Petersen SE, Schiattarella GG, Sluijter JPG, Steffens S, Streckfuss-Bömeke K, Thielmann M, Tucker A, Van Linthout S, Wijns W, Wojta J, Wu JC, Perrino C. Opportunities and challenges for the use of human samples in translational cardiovascular research: a scientific statement of the ESC Working Group on Cellular Biology of the Heart, the ESC Working Group on Cardiovascular Surgery, the ESC Council on Basic Cardiovascular Science, the ESC Scientists of Tomorrow, the European Association of Percutaneous Cardiovascular Interventions of the ESC, and the Heart Failure Association of the ESC. Cardiovasc Res 2025; 121:702-729. [PMID: 40084813 PMCID: PMC12101359 DOI: 10.1093/cvr/cvaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 03/16/2025] Open
Abstract
Animal models offer invaluable insights into disease mechanisms but cannot entirely mimic the variability and heterogeneity of human populations, nor the increasing prevalence of multi-morbidity. Consequently, employing human samples-such as whole blood or fractions, valvular and vascular tissues, myocardium, pericardium, or human-derived cells-is essential for enhancing the translational relevance of cardiovascular research. For instance, myocardial tissue slices, which preserve crucial structural and functional characteristics of the human heart, can be used in vitro to examine drug responses. Human blood serves as a rich source of biomarkers, including extracellular vesicles, various types of RNA (miRNA, lncRNA, and circRNAs), circulating inflammatory cells, and endothelial colony-forming cells, facilitating detailed studies of cardiovascular diseases. Primary cardiomyocytes and vascular cells isolated from human tissues are invaluable for mechanistic investigations in vitro. In cases where these are unavailable, human induced pluripotent stem cells serve as effective substitutes, albeit with specific limitations. However, the use of human samples presents challenges such as ethical approvals, tissue procurement and storage, variability in patient genetics and treatment regimens, and the selection of appropriate control samples. Biobanks are central to the efficient use of these scarce and valuable resources. This scientific statement discusses opportunities to implement the use of human samples for cardiovascular research within specific clinical contexts, offers a practical framework for acquiring and utilizing different human materials, and presents examples of human sample applications for specific cardiovascular diseases, providing a valuable resource for clinicians, translational and basic scientists engaged in cardiovascular research.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Ioanna Andreadou
- School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Antoniades
- RDM Division of Cardiovascular Medicine, Acute Multidisciplinary Imaging and Interventional Centre, University of Oxford, Headley Way, Headington, Oxford OX3 9DU, UK
| | - Jozef Bartunek
- Cardiovascular Center Aalst, OLV Hospital, Aalst, Belgium
| | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, Cardiovascular Pathology, University of Padua, Padua, Italy
| | - Bianca J J M Brundel
- Physiology, Amsterdam UMC Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
| | - Robert A Byrne
- Cardiovascular Research Institute Dublin, Mater Private Network, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Gemma Chiva-Blanch
- Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Network Research Centre on Obesity and Nutrition Physiopathology, Instituto de Salud Carlos III, Madrid, Spain
| | - Paula da Costa Martins
- Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paul C Evans
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Henrique Girão
- Center for Innovative Biomedicine and Biotechnology, Clinical Academic Centre of Coimbra, Faculty of Medicine, University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Coimbra, Portugal
| | - Zoltan Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Can Gollmann-Tepeköylü
- Department for Cardiac Surgery, Cardiac Regeneration Research, Medical University of Innsbruck, Anichstraße 35 A, 6020 Innsbruck, Austria
| | - Tomasz Guzik
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Mariann Gyöngyösi
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Norbert Hübner
- Max Delbrück Center in the Helmholtz Association, Berlin, Germany
- Charite-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Michael Joner
- Department of Cardiology, German Heart Center Munich, Technical University of Munich, Lazarettstrasse 36, 80636 Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Petra Kleinbongard
- Faculty of Medicine University of Duisburg-Essen, Institute of Pathophysiology, Duisburg-Essen, Germany
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Elisa Liehn
- Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rosalinda Madonna
- Cardiology Division, Department of Pathology, University of Pisa, Pisa, Italy
| | - Ange Maguy
- Department of Physiology, University of Bern, Bern, Switzerland
| | - Melanie Paillard
- Laboratoire CarMeN—IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, Univ-Lyon, 69500 Bron, France
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Department of Aerospace and Mechanical Engineering, Politecnico di Torino, Italy
- Department of Cell Biology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Steffen E Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, UK
- Health Data Research UK, London, UK
- Alan Turing Institute, London, UK
| | - Gabriele G Schiattarella
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
- Deutsches Herzzentrum der Charité (DHZC), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Katrin Streckfuss-Bömeke
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen, Germany and German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Matthias Thielmann
- West-German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Art Tucker
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité, BIH Center for Regenerative Therapies, Universitätmedizin Berlin, Berlin, Germany
- Max Delbrück Center in the Helmholtz Association, Berlin, Germany
| | - William Wijns
- The Lambe Institute for Translational Research and Curam, University of Galway, Galway, Ireland
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Core Facilities, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
2
|
Kwan ED, Hardie BA, Garcia KM, Mu H, Wang TM, Valdez-Jasso D. Sex-dependent remodeling of right ventricular function in a rat model of pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol 2024; 327:H351-H363. [PMID: 38847755 PMCID: PMC11932540 DOI: 10.1152/ajpheart.00098.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/09/2024] [Accepted: 05/29/2024] [Indexed: 07/17/2024]
Abstract
Right ventricular (RV) function is an important prognostic indicator for pulmonary arterial hypertension (PAH), a vasculopathy that primarily and disproportionally affects women with distinct pre- and postmenopausal clinical outcomes. However, most animal studies have overlooked the impact of sex and ovarian hormones on RV remodeling in PAH. Here, we combined invasive measurements of RV hemodynamics and morphology with computational models of RV biomechanics in sugen-hypoxia (SuHx)-treated male, ovary-intact female, and ovariectomized female rats. Despite similar pressure overload levels, SuHx induced increases in end-diastolic elastance and passive myocardial stiffening, notably in male SuHx animals, corresponding to elevated diastolic intracellular calcium. Increases in end-systolic chamber elastance were largely explained by myocardial hypertrophy in male and ovary-intact female rats, whereas ovariectomized females exhibited contractility recruitment via calcium transient augmentation. Ovary-intact female rats primarily responded with hypertrophy, showing fewer myocardial mechanical alterations and less stiffening. These findings highlight sex-related RV remodeling differences in rats, affecting systolic and diastolic RV function in PAH.NEW & NOTEWORTHY Combining hemodynamic and morphological measurements from male, female, and ovariectomized female pulmonary arterial hypertension (PAH) rats revealed distinct adaptation mechanisms despite similar pressure overload. Males showed the most diastolic stiffening. Ovariectomized females had enhanced myocyte contractility and calcium transient upregulation. Ovary-intact females primarily responded with hypertrophy, experiencing milder passive myocardial stiffening and no changes in myocyte shortening. These findings suggest potential sex-specific pathways in right ventricular (RV) adaptation to PAH, with implications for targeted interventions.
Collapse
MESH Headings
- Animals
- Female
- Male
- Ventricular Function, Right
- Ventricular Remodeling
- Disease Models, Animal
- Rats, Sprague-Dawley
- Ovariectomy
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/etiology
- Sex Factors
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/pathology
- Rats
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/etiology
- Pulmonary Artery/physiopathology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Models, Cardiovascular
- Calcium Signaling
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/etiology
- Hemodynamics
Collapse
Affiliation(s)
- Ethan D Kwan
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| | - Becky A Hardie
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| | - Kristen M Garcia
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| | - Hao Mu
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| | - Tsui-Min Wang
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| | - Daniela Valdez-Jasso
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| |
Collapse
|
3
|
Braun JL, Fajardo VA. Spaceflight increases sarcoplasmic reticulum Ca 2+ leak and this cannot be counteracted with BuOE treatment. NPJ Microgravity 2024; 10:78. [PMID: 39030182 PMCID: PMC11271499 DOI: 10.1038/s41526-024-00419-y] [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: 03/01/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Spending time in a microgravity environment is known to cause significant skeletal muscle atrophy and weakness via muscle unloading, which can be partly attributed to Ca2+ dysregulation. The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) pump is responsible for bringing Ca2+ from the cytosol into its storage site, the sarcoplasmic reticulum (SR), at the expense of ATP. We have recently demonstrated that, in the soleus of space-flown mice, the Ca2+ uptake ability of the SERCA pump is severely impaired and this may be attributed to increases in reactive oxygen/nitrogen species (RONS), to which SERCA is highly susceptible. The purpose of this study was therefore to investigate whether treatment with the antioxidant, Manganese(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin, MnTnBuOE-2-PyP5+ (BuOE), could attenuate muscle atrophy and SERCA dysfunction. We received soleus muscles from the rodent research 18 mission which had male mice housed on the international space station for 35 days and treated with either saline or BuOE. Spaceflight significantly reduced the soleus:body mass ratio and significantly increased SERCA's ionophore ratio, a measure of SR Ca2+ leak, and 4-HNE content (marker of RONS), none of which could be rescued by BuOE treatment. In conclusion, we find that spaceflight induces significant soleus muscle atrophy and SR Ca2+ leak that cannot be counteracted with BuOE treatment. Future work should investigate alternative therapeutics that are specifically aimed at increasing SERCA activation or reducing Ca2+ leak.
Collapse
Affiliation(s)
- Jessica L Braun
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada.
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada.
| |
Collapse
|
4
|
Yao J, Hua X, Huo W, Xiao L, Wang Y, Tang Q, Valdivia CR, Valdivia HH, Dong W, Xiao L. The Effect of Acidic Residues on the Binding between Opicalcin1 and Ryanodine Receptor from the Structure-Functional Analysis. JOURNAL OF NATURAL PRODUCTS 2024; 87:104-112. [PMID: 38128916 PMCID: PMC10825818 DOI: 10.1021/acs.jnatprod.3c00821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/26/2023] [Accepted: 11/26/2023] [Indexed: 12/23/2023]
Abstract
Calcin is a group ligand with high affinity and specificity for the ryanodine receptors (RyRs). Little is known about the effect of its acidic residues on the spacial structure as well as the interaction with RyRs. We screened the opicalcin1 acidic mutants and investigated the effect of mutation on activity. The results indicated that all acidic mutants maintained the structural features, but their surface charge distribution underwent significant changes. Molecular docking and dynamics simulations were used to analyze the interaction between opicalcin1 mutants and RyRs, which demonstrated that all opicalcin1 mutants effectively bound to the channel domain of RyR1. This stable binding induced a pronounced asymmetry in the structure of the RyR tetramer, exhibiting a high degree of structural dissimilarity. [3H]Ryanodine binding to RyR1 was enhanced in D2A and D15A, which was similar to opicalcin1, but that effect was suppressed in E12A and E29A and reversed for the DE-4A, thereby inhibiting ryanodine binding. Opicalcin1 and DE-4A also exhibited the ability to form stable docking structures with RyR2. Acidic residues play a crucial role in the structure of calcin and its functional interaction with RyRs that is beneficial for the calcin optimization to develop more active peptide lead compounds for RyR-related diseases.
Collapse
Affiliation(s)
- Jinchi Yao
- School
of Life Sciences, Liaoning Normal University, Dalian116081, China
- Department
of Occupational and Environmental Health, Faculty of Naval Medicine, Naval Medical University (Second Military Medical
University), Shanghai 200433, China
| | - Xiaoyu Hua
- Department
of Occupational and Environmental Health, Faculty of Naval Medicine, Naval Medical University (Second Military Medical
University), Shanghai 200433, China
| | - Wenjing Huo
- The
305 Hospital of PLA, Beijing 100017, China
| | - Li Xiao
- Department
of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin 53188, United States
- Department
of Forensic Toxicological Analysis, West China School of Basic Medical
Sciences and Forensic Medicine, Sichuan
University, Chengdu 610017, China
| | - Yongfang Wang
- Department
of Occupational and Environmental Health, Faculty of Naval Medicine, Naval Medical University (Second Military Medical
University), Shanghai 200433, China
| | - Qinglong Tang
- Central
Medical District of Chinese, PLA General Hospital, Beijing 100120, China
| | - Carmen R. Valdivia
- Department
of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin 53188, United States
| | - Héctor H. Valdivia
- Department
of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin 53188, United States
| | - Weibing Dong
- School
of Life Sciences, Liaoning Normal University, Dalian116081, China
| | - Liang Xiao
- Department
of Occupational and Environmental Health, Faculty of Naval Medicine, Naval Medical University (Second Military Medical
University), Shanghai 200433, China
| |
Collapse
|
5
|
Brun C, Chalet L, Moulin F, Bochaton T, Ducreux S, Paillard M, Crola Da Silva C. A bibliometric analysis: Ca 2+ fluxes and inflammatory phenotyping by flow cytometry in peripheral blood mononuclear cells. Front Immunol 2023; 14:1272809. [PMID: 37901222 PMCID: PMC10611513 DOI: 10.3389/fimmu.2023.1272809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023] Open
Abstract
Background The immune system, composed of organs, tissues, cells, and proteins, is the key to protecting the body from external biological attacks and inflammation. The latter occurs in several pathologies, such as cancers, type 1 diabetes, and human immunodeficiency virus infection. Immunophenotyping by flow cytometry is the method of choice for diagnosing these pathologies. Under inflammatory conditions, the peripheral blood mononuclear cells (PBMCs) are partially activated and generate intracellular pathways involving Ca2+-dependent signaling cascades leading to transcription factor expression. Ca2+ signaling is typically studied by microscopy in cell lines but can present some limitations to explore human PBMCs, where flow cytometry can be a good alternative. Objective In this review, we dived into the research field of inflammation and Ca2+ signaling in PBMCs. We aimed to investigate the structure and evolution of this field in a physio-pathological context, and then we focused our review on flow cytometry analysis of Ca2+ fluxes in PBMCs. Methods From 1984 to 2022, 3865 articles on inflammation and Ca2+ signaling in PBMCs were published, according to The Clarivate Web of Science (WOS) database used in this review. A bibliometric study was designed for this collection and consisted of a co-citation and bibliographic coupling analysis. Results The co-citation analysis was performed on 133 articles: 4 clusters highlighted the global context of Ca2+ homeostasis, including chemical probe development, identification of the leading players in Ca2+ signaling, and the link with chemokine production in immune cell function. Next, the bibliographic coupling analysis combined 998 articles in 8 clusters. This analysis outlined the mechanisms of PBMC activation, from signal integration to cellular response. Further explorations of the bibliographic coupling network, focusing on flow cytometry, revealed 21 articles measuring cytosolic Ca2+ in PBMCs, with only 5 since 2016. This final query showed that Ca2+ signaling analysis in human PBMCs using flow cytometry is still underdeveloped and investigates mainly the cytosolic Ca2+ compartment. Conclusion Our review uncovers remaining knowledge gaps of intracellular players involved in Ca2+ signaling in PBMCs, such as reticulum and mitochondria, and presents flow cytometry as a solid option to supplement gold-standard microscopy studies.
Collapse
Affiliation(s)
- Camille Brun
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Lucie Chalet
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Bron, France
- Olea Medical, La Ciotat, France
| | - Florentin Moulin
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Thomas Bochaton
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Bron, France
- Hospices Civils de Lyon, Hôpital Louis Pradel, Services D’explorations Fonctionnelles Cardiovasculaires et CIC de Lyon, Lyon, France
| | - Sylvie Ducreux
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Melanie Paillard
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Claire Crola Da Silva
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Bron, France
| |
Collapse
|
6
|
Dridi H, Santulli G, Bahlouli L, Miotto MC, Weninger G, Marks AR. Mitochondrial Calcium Overload Plays a Causal Role in Oxidative Stress in the Failing Heart. Biomolecules 2023; 13:1409. [PMID: 37759809 PMCID: PMC10527470 DOI: 10.3390/biom13091409] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/13/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Heart failure is a serious global health challenge, affecting more than 6.2 million people in the United States and is projected to reach over 8 million by 2030. Independent of etiology, failing hearts share common features, including defective calcium (Ca2+) handling, mitochondrial Ca2+ overload, and oxidative stress. In cardiomyocytes, Ca2+ not only regulates excitation-contraction coupling, but also mitochondrial metabolism and oxidative stress signaling, thereby controlling the function and actual destiny of the cell. Understanding the mechanisms of mitochondrial Ca2+ uptake and the molecular pathways involved in the regulation of increased mitochondrial Ca2+ influx is an ongoing challenge in order to identify novel therapeutic targets to alleviate the burden of heart failure. In this review, we discuss the mechanisms underlying altered mitochondrial Ca2+ handling in heart failure and the potential therapeutic strategies.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Laith Bahlouli
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Marco C. Miotto
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Gunnar Weninger
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| |
Collapse
|
7
|
Jankauskas SS, Kansakar U, Sardu C, Varzideh F, Avvisato R, Wang X, Matarese A, Marfella R, Ziosi M, Gambardella J, Santulli G. COVID-19 Causes Ferroptosis and Oxidative Stress in Human Endothelial Cells. Antioxidants (Basel) 2023; 12:326. [PMID: 36829885 PMCID: PMC9952002 DOI: 10.3390/antiox12020326] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/03/2023] Open
Abstract
Oxidative stress and endothelial dysfunction have been shown to play crucial roles in the pathophysiology of COVID-19 (coronavirus disease 2019). On these grounds, we sought to investigate the impact of COVID-19 on lipid peroxidation and ferroptosis in human endothelial cells. We hypothesized that oxidative stress and lipid peroxidation induced by COVID-19 in endothelial cells could be linked to the disease outcome. Thus, we collected serum from COVID-19 patients on hospital admission, and we incubated these sera with human endothelial cells, comparing the effects on the generation of reactive oxygen species (ROS) and lipid peroxidation between patients who survived and patients who did not survive. We found that the serum from non-survivors significantly increased lipid peroxidation. Moreover, serum from non-survivors markedly regulated the expression levels of the main markers of ferroptosis, including GPX4, SLC7A11, FTH1, and SAT1, a response that was rescued by silencing TNFR1 on endothelial cells. Taken together, our data indicate that serum from patients who did not survive COVID-19 triggers lipid peroxidation in human endothelial cells.
Collapse
Affiliation(s)
- Stanislovas S. Jankauskas
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Urna Kansakar
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Celestino Sardu
- University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
| | - Fahimeh Varzideh
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Roberta Avvisato
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- “Federico II” University, 80131 Naples, Italy
| | - Xujun Wang
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | | | | | - Jessica Gambardella
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- “Federico II” University, 80131 Naples, Italy
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- “Federico II” University, 80131 Naples, Italy
- Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation (INI), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
8
|
Forzano I, Mone P, Mottola G, Kansakar U, Salemme L, De Luca A, Tesorio T, Varzideh F, Santulli G. Efficacy of the New Inotropic Agent Istaroxime in Acute Heart Failure. J Clin Med 2022; 11:7503. [PMID: 36556120 PMCID: PMC9786901 DOI: 10.3390/jcm11247503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Current therapeutic strategies for acute heart failure (AHF) are based on traditional inotropic agents that are often associated with untoward effects; therefore, finding new effective approaches with a safer profile is dramatically needed. Istaroxime is a novel compound, chemically unrelated to cardiac glycosides, that is currently being studied for the treatment of AHF. Its effects are essentially related to its inotropic and lusitropic positive properties exerted through a dual mechanism of action: activation of the sarcoplasmic reticulum Ca2+ ATPase isoform 2a (SERCA2a) and inhibition of the Na+/K+-ATPase (NKA) activity. The advantages of istaroxime over the available inotropic agents include its lower arrhythmogenic action combined with its capability of increasing systolic blood pressure without augmenting heart rate. However, it has a limited half-life (1 hour) and is associated with adverse effects including pain at the injection site and gastrointestinal issues. Herein, we describe the main mechanism of action of istaroxime and we present a systematic overview of both clinical and preclinical trials testing this drug, underlining the latest insights regarding its adoption in clinical practice for AHF.
Collapse
Affiliation(s)
- Imma Forzano
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
| | - Pasquale Mone
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Gaetano Mottola
- Casa di Cura “Montevergine”, Mercogliano, 83013 Avellino, Italy
| | - Urna Kansakar
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
| | - Luigi Salemme
- Casa di Cura “Montevergine”, Mercogliano, 83013 Avellino, Italy
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Vanvitelli”, 81100 Caserta, Italy
| | - Tullio Tesorio
- Casa di Cura “Montevergine”, Mercogliano, 83013 Avellino, Italy
| | - Fahimeh Varzideh
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
| | - Gaetano Santulli
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
9
|
Oknińska M, Mączewski M, Mackiewicz U. Ventricular arrhythmias in acute myocardial ischaemia-Focus on the ageing and sex. Ageing Res Rev 2022; 81:101722. [PMID: 36038114 DOI: 10.1016/j.arr.2022.101722] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 01/31/2023]
Abstract
Annually, approximately 17 million people die from cardiovascular diseases worldwide, half of them suddenly. The most common direct cause of sudden cardiac death is ventricular arrhythmia triggered by an acute coronary syndrome (ACS). The study summarizes the knowledge of the mechanisms of arrhythmia onset during ACS in humans and in animal models and factors that may influence the susceptibility to life-threatening arrhythmias during ACS with particular focus on the age and sex. The real impact of age and sex on the arrhythmic susceptibility within the setting of acute ischaemia is masked by the fact that ACSs result from coronary artery disease appearing with age much earlier among men than among women. However, results of researches show that in ageing process changes with potential pro-arrhythmic significance, such as increased fibrosis, cardiomyocyte hypertrophy, decrease number of gap junction channels, disturbances of the intracellular Ca2+ signalling or changes in electrophysiological parameters, occur independently of the development of cardiovascular diseases and are more severe in male individuals. A review of the literature also indicates a marked paucity of research in this area in female and elderly individuals. Greater awareness of sex differences in the aging process could help in the development of personalized prevention methods targeting potential pro-arrhythmic factors in patients of both sexes to reduce mortality during the acute phase of myocardial infarction. This is especially important in an era of aging populations in which women will predominate due to their longer lifespan.
Collapse
Affiliation(s)
- Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland.
| |
Collapse
|
10
|
Melville Z, Dridi H, Yuan Q, Reiken S, Wronska A, Liu Y, Clarke OB, Marks AR. A drug and ATP binding site in type 1 ryanodine receptor. Structure 2022; 30:1025-1034.e4. [PMID: 35580609 DOI: 10.1016/j.str.2022.04.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/02/2022] [Accepted: 04/21/2022] [Indexed: 12/12/2022]
Abstract
The ryanodine receptor (RyR)/calcium release channel on the sarcoplasmic reticulum (SR) is required for excitation-contraction coupling in skeletal and cardiac muscle. Inherited mutations and stress-induced post-translational modifications result in an SR Ca2+ leak that causes skeletal myopathies, heart failure, and exercise-induced sudden death. A class of therapeutics known as Rycals prevent the RyR-mediated leak, are effective in preventing disease progression and restoring function in animal models, and are in clinical trials for patients with muscle and heart disorders. Using cryogenic-electron microscopy, we present a model of RyR1 with a 2.45-Å resolution before local refinement, revealing a binding site in the RY1&2 domain (3.10 Å local resolution), where the Rycal ARM210 binds cooperatively with ATP and stabilizes the closed state of RyR1.
Collapse
Affiliation(s)
- Zephan Melville
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Haikel Dridi
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Anetta Wronska
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Yang Liu
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA; Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA; Clyde & Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA.
| |
Collapse
|
11
|
Dridi H, Forrester F, Umanskaya A, Xie W, Reiken S, Lacampagne A, Marks A. Role of oxidation of excitation-contraction coupling machinery in age-dependent loss of muscle function in C. elegans. eLife 2022; 11:75529. [PMID: 35506650 PMCID: PMC9113742 DOI: 10.7554/elife.75529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Age-dependent loss of body wall muscle function and impaired locomotion occur within 2 weeks in C. elegans; however, the underlying mechanism has not been fully elucidated. In humans, age-dependent loss of muscle function occurs at about 80 years of age and has been linked to dysfunction of ryanodine receptor (RyR)/intracellular calcium (Ca2+) release channels on the sarcoplasmic reticulum (SR). Mammalian skeletal muscle RyR1 channels undergo age-related remodeling due to oxidative overload, leading to loss of the stabilizing subunit calstabin1 (FKBP12) from the channel macromolecular complex. This destabilizes the closed state of the channel resulting in intracellular Ca2+ leak, reduced muscle function, and impaired exercise capacity. We now show that the C. elegans RyR homolog, UNC-68, exhibits a remarkable degree of evolutionary conservation with mammalian RyR channels and similar age-dependent dysfunction. Like RyR1 in mammals UNC-68 encodes a protein that comprises a macromolecular complex which includes the calstabin1 homolog FKB-2 and is immunoreactive with antibodies raised against the RyR1 complex. Further, as in aged mammals, UNC-68 is oxidized and depleted of FKB-2 in an age-dependent manner, resulting in 'leaky' channels, depleted SR Ca2+ stores, reduced body wall muscle Ca2+ transients, and age-dependent muscle weakness. FKB-2 (ok3007)-deficient worms exhibit reduced exercise capacity. Pharmacologically induced oxidization of UNC-68 and depletion of FKB-2 from the channel independently caused reduced body wall muscle Ca2+ transients. Preventing FKB-2 depletion from the UNC-68 macromolecular complex using the Rycal drug S107 improved muscle Ca2+ transients and function. Taken together, these data suggest that UNC-68 oxidation plays a role in age-dependent loss of muscle function. Remarkably, this age-dependent loss of muscle function induced by oxidative overload, which takes ~2 years in mice and ~80 years in humans, occurs in less than 2-3 weeks in C. elegans, suggesting that reduced antioxidant capacity may contribute to the differences in life span amongst species.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Frances Forrester
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Alisa Umanskaya
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Wenjun Xie
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Alain Lacampagne
- U1046, Montpellier University, INSERM, CNRS, Montpellier, France
| | - Andrew Marks
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| |
Collapse
|
12
|
Gambardella J, Jankauskas SS, D'Ascia SL, Sardu C, Matarese A, Minicucci F, Mone P, Santulli G. Glycation of ryanodine receptor in circulating lymphocytes predicts the response to cardiac resynchronization therapy. J Heart Lung Transplant 2022; 41:438-441. [PMID: 35042640 PMCID: PMC8977242 DOI: 10.1016/j.healun.2021.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/14/2021] [Accepted: 12/19/2021] [Indexed: 01/02/2023] Open
Abstract
Finding reliable parameters to identify patients with heart failure (HF) that will respond to cardiac resynchronization therapy (CRT) represents a major challenge. We and others have observed post-translational modifications of Ryanodine Receptor (RyR) in several tissues (including skeletal muscle and circulating lymphocytes) of patients with advanced HF. We designed a prospective study to test the hypothesis that RyR1 glycation in circulating lymphocytes could predict CRT responsiveness in patients with non-ischemic HF. We enrolled 94 patients who underwent CRT and 30 individuals without HF, examining RyR1 glycation in peripheral lymphocytes at enrollment and after 1 year. We found that baseline RyR1 glycation independently predicts CRT response at 1 year after adjusting for age, diabetes, QRS duration and morphology, echocardiographic dyssynchrony, and hypertension. Moreover, RyR1 glycation in circulating lymphocytes significantly correlated with pathologic intracellular calcium leak. Taken together, our data show for the first time that RyR1 glycation in circulating lymphocytes represents a novel biomarker to predict CRT responsiveness.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, University of Naples "Federico II" and International Translational Research and Medical Education (ITME) Consortium, Naples, Italy
| | - Stanislovas S Jankauskas
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York
| | | | | | | | - Fabio Minicucci
- Naples Local Health Unit (ASL) of the Italian Ministry of Health, Naples, Italy
| | - Pasquale Mone
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York; University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gaetano Santulli
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, University of Naples "Federico II" and International Translational Research and Medical Education (ITME) Consortium, Naples, Italy; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation (INI), Norman Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, New York.
| |
Collapse
|
13
|
Junho CVC, González-Lafuente L, Navarro-García JA, Rodríguez-Sánchez E, Carneiro-Ramos MS, Ruiz-Hurtado G. Unilateral Acute Renal Ischemia-Reperfusion Injury Induces Cardiac Dysfunction through Intracellular Calcium Mishandling. Int J Mol Sci 2022; 23:ijms23042266. [PMID: 35216382 PMCID: PMC8879526 DOI: 10.3390/ijms23042266] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 01/27/2023] Open
Abstract
Background: Acute renal failure (ARF) following renal ischemia-reperfusion (I/R) injury is considered a relevant risk factor for cardiac damage, but the underlying mechanisms, particularly those triggered at cardiomyocyte level, are unknown. Methods: We examined intracellular Ca2+ dynamics in adult ventricular cardiomyocytes isolated from C57BL/6 mice 7 or 15 days following unilateral renal I/R. Results: After 7 days of I/R, the cell contraction was significantly lower in cardiomyocytes compared to sham-treated mice. It was accompanied by a significant decrease in both systolic Ca2+ transients and sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) activity measured as Ca2+ transients decay. Moreover, the incidence of pro-arrhythmic events, measured as the number of Ca2+ sparks, waves or automatic Ca2+ transients, was greater in cardiomyocytes from mice 7 days after I/R than from sham-treated mice. Ca2+ mishandling related to systolic Ca2+ transients and contraction were recovered to sham values 15 days after I/R, but Ca2+ sparks frequency and arrhythmic events remained elevated. Conclusions: Renal I/R injury causes a cardiomyocyte Ca2+ cycle dysfunction at medium (contraction-relaxation dysfunction) and long term (Ca2+ leak), after 7 and 15 days of renal reperfusion, respectively.
Collapse
Affiliation(s)
- Carolina Victoria Cruz Junho
- Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-580, SP, Brazil;
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Community of Madrid, 28041 Madrid, Spain; (L.G.-L.); (J.A.N.-G.); (E.R.-S.)
| | - Laura González-Lafuente
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Community of Madrid, 28041 Madrid, Spain; (L.G.-L.); (J.A.N.-G.); (E.R.-S.)
| | - José Alberto Navarro-García
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Community of Madrid, 28041 Madrid, Spain; (L.G.-L.); (J.A.N.-G.); (E.R.-S.)
| | - Elena Rodríguez-Sánchez
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Community of Madrid, 28041 Madrid, Spain; (L.G.-L.); (J.A.N.-G.); (E.R.-S.)
| | - Marcela Sorelli Carneiro-Ramos
- Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-580, SP, Brazil;
- Correspondence: (M.S.C.-R.); (G.R.-H.); Tel.: +34-913908001 (G.R.-H.)
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Community of Madrid, 28041 Madrid, Spain; (L.G.-L.); (J.A.N.-G.); (E.R.-S.)
- CIBER-CV, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Correspondence: (M.S.C.-R.); (G.R.-H.); Tel.: +34-913908001 (G.R.-H.)
| |
Collapse
|
14
|
Dridi H, Santulli G, Gambardella J, Jankauskas SS, Yuan Q, Yang J, Reiken S, Wang X, Wronska A, Liu X, Lacampagne A, Marks AR. IP3 receptor orchestrates maladaptive vascular responses in heart failure. J Clin Invest 2022; 132:e152859. [PMID: 35166236 PMCID: PMC8843748 DOI: 10.1172/jci152859] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/21/2021] [Indexed: 12/02/2022] Open
Abstract
Patients with heart failure (HF) have augmented vascular tone, which increases cardiac workload, impairing ventricular output and promoting further myocardial dysfunction. The molecular mechanisms underlying the maladaptive vascular responses observed in HF are not fully understood. Vascular smooth muscle cells (VSMCs) control vasoconstriction via a Ca2+-dependent process, in which the type 1 inositol 1,4,5-trisphosphate receptor (IP3R1) on the sarcoplasmic reticulum (SR) plays a major role. To dissect the mechanistic contribution of intracellular Ca2+ release to the increased vascular tone observed in HF, we analyzed the remodeling of IP3R1 in aortic tissues from patients with HF and from controls. VSMC IP3R1 channels from patients with HF and HF mice were hyperphosphorylated by both serine and tyrosine kinases. VSMCs isolated from IP3R1VSMC-/- mice exhibited blunted Ca2+ responses to angiotensin II (ATII) and norepinephrine compared with control VSMCs. IP3R1VSMC-/- mice displayed significantly reduced responses to ATII, both in vivo and ex vivo. HF IP3R1VSMC-/- mice developed significantly less afterload compared with HF IP3R1fl/fl mice and exhibited significantly attenuated progression toward decompensated HF and reduced interstitial fibrosis. Ca2+-dependent phosphorylation of the MLC by MLCK activated VSMC contraction. MLC phosphorylation was markedly increased in VSMCs from patients with HF and HF mice but reduced in VSMCs from HF IP3R1VSMC-/- mice and HF WT mice treated with ML-7. Taken together, our data indicate that VSMC IP3R1 is a major effector of increased vascular tone, which contributes to increased cardiac afterload and decompensation in HF.
Collapse
MESH Headings
- Animals
- Calcium Signaling
- Heart Failure/genetics
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Humans
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Vasoconstriction
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Gaetano Santulli
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, New York, New York, USA
- Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Jessica Gambardella
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, New York, New York, USA
- International Translational Research and Medical Education (ITME) Consortium, Department of Advanced Biomedical Science, “Federico II” University, Naples, Italy
| | - Stanislovas S. Jankauskas
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, New York, New York, USA
- Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Jingyi Yang
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Xujun Wang
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, New York, New York, USA
- Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Anetta Wronska
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Xiaoping Liu
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, CNRS, INSERM, CHRU Montpellier, Montpellier, France
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
15
|
Giordanino EF. IMPROVING PATIENT SELECTION FOR CARDIAC RESYNCHRONIZATION THERAPY: THE ROLE OF PATHOPHYSIOLOGICAL BIOMARKERS. J Heart Lung Transplant 2022; 41:442-444. [DOI: 10.1016/j.healun.2022.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/08/2022] [Accepted: 01/12/2022] [Indexed: 11/30/2022] Open
|
16
|
Tang XH, Gambardella J, Jankauskas S, Wang X, Santulli G, Gudas LJ, Levi R. A Retinoic Acid Receptor β 2 Agonist Improves Cardiac Function in a Heart Failure Model. J Pharmacol Exp Ther 2021; 379:182-190. [PMID: 34389654 PMCID: PMC8626778 DOI: 10.1124/jpet.121.000806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 12/22/2022] Open
Abstract
We previously demonstrated that the selective retinoic acid receptor (RAR) β 2 agonist AC261066 reduces oxidative stress in an ex vivo murine model of ischemia/reperfusion. We hypothesized that by decreasing oxidative stress and consequent fibrogenesis, AC261066 could attenuate the development of contractile dysfunction in post-ischemic heart failure (HF). We tested this hypothesis in vivo using an established murine model of myocardial infarction (MI), obtained by permanent occlusion of the left anterior descending coronary artery. Treating mice with AC261066 in drinking water significantly attenuated the post-MI deterioration of echocardiographic indices of cardiac function, diminished remodeling, and reduced oxidative stress, as evidenced by a decrease in malondialdehyde level and p38 mitogen-activated protein kinase expression in cardiomyocytes. The effects of AC261066 were also associated with a decrease in interstitial fibrosis, as shown by a marked reduction in collagen deposition and α-smooth muscle actin expression. In cardiac murine fibroblasts subjected to hypoxia, AC261066 reversed hypoxia-induced decreases in superoxide dismutase 2 and angiopoietin-like 4 transcriptional levels as well as the increase in NADPH oxidase 2 mRNA, demonstrating that the post-MI cardioprotective effects of AC261066 are associated with an action at the fibroblast level. Thus, AC261066 alleviates post-MI cardiac dysfunction by modulating a set of genes involved in the oxidant/antioxidant balance. These AC261066 responsive genes diminish interstitial fibrogenesis and remodeling. Since MI is a recognized major cause of HF, our data identify RARβ 2 as a potential pharmacological target in the treatment of HF. SIGNIFICANCE STATEMENT: A previous report showed that the selective retinoic acid receptor (RAR) β 2 agonist AC261066 reduces oxidative stress in an ex vivo murine model of ischemia/reperfusion. This study shows that AC261066 attenuates the development of contractile dysfunction and maladaptive remodeling in post-ischemic heart failure (HF) by modulating a set of genes involved in oxidant/antioxidant balance. Since myocardial infarction is a recognized major cause of HF, these data identify RARβ 2 as a potential pharmacological target in the treatment of HF.
Collapse
Affiliation(s)
- Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Jessica Gambardella
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Stanislovas Jankauskas
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Xujun Wang
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Gaetano Santulli
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Roberto Levi
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| |
Collapse
|
17
|
Rathor R, Suryakumar G, Singh SN. Diet and redox state in maintaining skeletal muscle health and performance at high altitude. Free Radic Biol Med 2021; 174:305-320. [PMID: 34352371 DOI: 10.1016/j.freeradbiomed.2021.07.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/14/2021] [Accepted: 07/19/2021] [Indexed: 01/07/2023]
Abstract
High altitude exposure leads to compromised physical performance with considerable weight loss. The major stressor at high altitude is hypobaric hypoxia which leads to disturbance in redox homeostasis. Oxidative stress is a well-known trigger for many high altitude illnesses and regulates several key signaling pathways under stressful conditions. Altered redox homeostasis is considered the prime culprit of high altitude linked skeletal muscle atrophy. Hypobaric hypoxia disturbs redox homeostasis through increased RONS production and compromised antioxidant system. Increased RONS disturbs the cellular homeostasis via multiple ways such as inflammation generation, altered protein anabolic pathways, redox remodeling of RyR1 that contributed to dysregulated calcium homeostasis, enhanced protein degradation pathways via activation calcium-regulated protein, calpain, and apoptosis. Ultimately, all the cellular signaling pathways aggregately result in skeletal muscle atrophy. Dietary supplementation of phytochemicals could become a safe and effective intervention to ameliorate skeletal muscle atrophy and enhance the physical performance of the personnel who are staying at high altitude regions. The present evidence-based review explores few dietary supplementations which regulate several signaling mechanisms and ameliorate hypobaric hypoxia induced muscle atrophy and enhances physical performance. However, a clinical research trial is required to establish proof-of-concept.
Collapse
Affiliation(s)
- Richa Rathor
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, New Delhi, 110054, India.
| | - Geetha Suryakumar
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, New Delhi, 110054, India
| | - Som Nath Singh
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, New Delhi, 110054, India
| |
Collapse
|
18
|
Chacon Alberty L, Perin EC, Willerson JT, Gahremanpour A, Bolli R, Yang PC, Traverse JH, Lai D, Pepine CJ, Taylor DA. Peripheral Blood Biomarkers Associated With Improved Functional Outcome in Patients With Chronic Left Ventricular Dysfunction: A Biorepository Evaluation of the FOCUS-CCTRN Trial. Front Cardiovasc Med 2021; 8:698088. [PMID: 34540912 PMCID: PMC8446350 DOI: 10.3389/fcvm.2021.698088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Cell therapy trials for heart failure (HF) have shown modest improvement; however, the mechanisms underlying improvement in some patients but not others are not well understood. Although immune cells are important in the course of HF, our understanding of the immune processes in HF is limited. The objective of this study was to evaluate associations between temporal changes in peripheral blood (PB) cell subpopulations and improved outcome in patients with chronic ischemic cardiomyopathy after bone marrow-derived mononuclear cell therapy or placebo in the FOCUS-CCTRN trial. Peripheral blood was collected at days 0, 1, 30, 90, and 180 from consented participants. We used flow cytometry to compare PB populations in patients with the best (cohort 1) or worst functional outcome (cohort 2) in three primary endpoints: left ventricular (LV) ejection fraction, LV end-systolic volume, and maximal oxygen consumption (VO2 max). A linear mixed model was used to assess changes over time in 32 cell populations. The difference between each time point and baseline was calculated as linear contrast. Compared with cohort 2, patients who improved (cohort 1) had a higher frequency of CD45+CD19+ B cells at days 0, 1, 90, and 180. CD11B+ cells increased over baseline at day 1 in both cohorts and remained higher in cohort 2 until day 30. CD45+CD133+ progenitor cells decreased over baseline at day 30 in cohort 1. We identified specific cell subpopulations associated with improved cardiac function in patients with chronic LV dysfunction. These findings may improve patient selection and prediction of outcomes in cell therapy trials.
Collapse
Affiliation(s)
| | - Emerson C Perin
- Stem Cell Center, Texas Heart Institute, Houston, TX, United States
| | | | - Amir Gahremanpour
- Hospital Corporation of America-Houston Heart, Houston, TX, United States
| | - Roberto Bolli
- School of Medicine, University of Louisville, Louisville, KY, United States
| | - Phillip C Yang
- Stanford University School of Medicine, Stanford, CA, United States
| | - Jay H Traverse
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital and University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Dejian Lai
- UTHealth School of Public Health, Houston, TX, United States
| | - Carl J Pepine
- University of Florida College of Medicine, Gainesville, FL, United States
| | - Doris A Taylor
- Regenerative Medicine Department, Texas Heart Institute, Houston, TX, United States
| |
Collapse
|
19
|
Fomina AF. Neglected wardens: T lymphocyte ryanodine receptors. J Physiol 2021; 599:4415-4426. [PMID: 34411300 DOI: 10.1113/jp281722] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ryanodine receptors (RyRs) are intracellular Ca2+ release channels ubiquitously expressed in various cell types. RyRs were extensively studied in striated muscle cells due to their crucial role in muscle contraction. In contrast, the role of RyRs in Ca2+ signalling and functions in non-excitable cells, such as T lymphocytes, remains poorly understood. Expression of different isoforms of RyRs was shown in primary T cells and T cell lines. In T cells, RyRs co-localize with the plasmalemmal store-operated Ca2+ channels of the Orai family and endoplasmic reticulum Ca2+ sensing Stim family proteins and are activated by store-operated Ca2+ entry and pyridine nucleotide metabolites, the intracellular second messengers generated upon stimulation of T cell receptors. Experimental data indicate that together with d-myo-inositol 1,4,5-trisphosphate receptors, RyRs regulate intercellular Ca2+ dynamics by controlling Ca2+ concentration within the lumen of the endoplasmic reticulum and, consequently, store-operated Ca2+ entry. Gain-of-function mutations, genetic deletion or pharmacological inhibition of RyRs alters T cell Ca2+ signalling and effector functions. The picture emerging from the collective data shows that RyRs are the essential regulators of T cell Ca2+ signalling and can be potentially used as molecular targets for immunomodulation or T cell-based diagnostics of the disorders associated with RyRs dysregulation.
Collapse
Affiliation(s)
- Alla F Fomina
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA
| |
Collapse
|
20
|
Kim MJ, Kim MH, Park SH, Song YW. A case of dermatomyositis in a patient with central core disease: unusual association with autoimmunity and genetic muscle disease. Pediatr Rheumatol Online J 2021; 19:100. [PMID: 34193198 PMCID: PMC8243539 DOI: 10.1186/s12969-021-00598-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dermatomyositis is an inflammatory muscle disease caused by immune-mediated muscle injury, and central core disease (CCD) is a congenital myopathy associated with disturbed intracellular calcium homeostasis and excitation-contraction coupling. To date, CCD has not been reported to have autoantibodies or coexist with inflammatory myopathy. CASE PRESENTATION Here, we described the case of a 25-year-old woman who had progressive proximal muscle weakness, myalgia, pruritic macular rash, skin ulcers, and calcinosis. Dermatomyositis was initially suspected based on the clinical symptoms accompanied by elevated muscle enzyme levels, electromyography abnormalities, and a positive antinuclear antibody test. However, the patient's muscle biopsy revealed the characteristic findings of both dermatomyositis and CCD, suggesting that dermatomyositis occurred in this patient with previously asymptomatic CCD. The patient did not have any pathogenic gene mutations associated with congenital myopathy, including RYR1 and SEPN1 in targeted next-generation sequencing. She received high-dose glucocorticoid therapy and azathioprine with a significant improvement in muscle strength. CONCLUSIONS We present a case of rare coexistence of dermatomyositis and CCD. Clinicians should be aware that patients with CCD may have inflammatory myopathy that responds well to immunosuppressive therapy.
Collapse
Affiliation(s)
- Min Jung Kim
- grid.484628.4 0000 0001 0943 2764Division of Rheumatology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Hospital Boramae Medical Center, Seoul, South Korea
| | - Mi Hyeon Kim
- grid.412484.f0000 0001 0302 820XDivision of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Daehak-ro, Jongno-gu, Seoul, 03080 South Korea
| | - Sung-Hye Park
- grid.31501.360000 0004 0470 5905Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| | - Yeong Wook Song
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea. .,Medical Research Center, Institute of Human-Environment Interface Biology, Seoul, South Korea.
| |
Collapse
|
21
|
Tian CJ, Zhang JH, Liu J, Ma Z, Zhen Z. Ryanodine receptor and immune-related molecules in diabetic cardiomyopathy. ESC Heart Fail 2021; 8:2637-2646. [PMID: 34013670 PMCID: PMC8318495 DOI: 10.1002/ehf2.13431] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 04/04/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022] Open
Abstract
Hyperglycaemia is a major aetiological factor in the development of diabetic cardiomyopathy. Excessive hyperglycaemia increases the levels of reactive carbonyl species (RCS), reactive oxygen species (ROS) and reactive nitrogen species (RNS) in the heart and causes derangements in calcium homeostasis, inflammation and immune‐system disorders. Ryanodine receptor 2 (RyR2) plays a key role in excitation–contraction coupling during heart contractions, including rhythmic contraction and relaxation of the heart. Cardiac inflammation has been indicated in part though interleukin 1 (IL‐1) signals, supporting a role for B and T lymphocytes in diabetic cardiomyopathy. Some of the post‐translational modifications of the ryanodine receptor (RyR) by RCS, ROS and RNS stress are known to affect its gating and Ca2+ sensitivity, which contributes to RyR dysregulation in diabetic cardiomyopathy. RyRs and immune‐related molecules are important signalling species in many physiological and pathophysiological processes in various heart and cardiovascular diseases. However, little is known regarding the mechanistic relationship between RyRs and immune‐related molecules in diabetes, as well as the mechanisms mediating complex communication among cardiomyocytes, fibroblasts and immune cells. This review highlights new findings on the complex cellular communications in the pathogenesis and progression of diabetic cardiomyopathy. We discuss potential therapeutic applications targeting RyRs and immune‐related molecules in diabetic complications.
Collapse
Affiliation(s)
- Cheng-Ju Tian
- College of Rehabilitation and Sports Medicine, Jinzhou Medical University, Jinzhou, China
| | - Jing-Hua Zhang
- Department of Psychiatry, Tianjin Anding Hospital, Tianjin, China
| | - Jinfeng Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhuang Ma
- College of Rehabilitation and Sports Medicine, Jinzhou Medical University, Jinzhou, China
| | - Zhong Zhen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
22
|
Xu X, Xie X, Zhang H, Wang P, Li G, Chen J, Chen G, Cao X, Xiong L, Peng F, Peng C. Water-soluble alkaloids extracted from Aconiti Radix lateralis praeparata protect against chronic heart failure in rats via a calcium signaling pathway. Biomed Pharmacother 2021; 135:111184. [PMID: 33418305 DOI: 10.1016/j.biopha.2020.111184] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/15/2020] [Accepted: 12/26/2020] [Indexed: 11/16/2022] Open
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Many studies have shown the beneficial effects of aconite water-soluble alkaloid extract (AWA) in experimental models of heart disease, which have been ascribed to the presence of aconine, hypaconine, talatisamine, fuziline, neoline, and songorine. This study evaluated the effects of a chemically characterized AWA by chemical content, evaluated its effects in suprarenal abdominal aortic coarctation surgery (AAC)-induced chronic heart failure (CHF) in rats, and revealed the underlying mechanisms of action by proteomics. METHODS Rats were distributed into different groups: sham, model, and AWA-treated groups (10, 20, and 40 mg/kg/day). Sham rats received surgery without AAC, whereas model rats an AWA-treated groups underwent AAC surgery. after 8 weeks, the treatment group was fed AWA for 4 weeks, and body weight was assessed weekly. At the end of the treatment, heart function was tested by echocardiography. AAC-induced chronic heart failure, including myocardial fibrosis, cardiomyocyte hypertrophy, and apoptosis, was evaluated in heart tissue and plasma by RT-qPCR, ELISA, hematoxylin and eosin (H&E) staining, Masson's trichrome staining, TUNEL staining, and immunofluorescence staining of α-SMA, Col Ⅰ, and Col Ⅲ. Then, a proteomics approach was used to explore the underlying mechanisms of action of AWA in chronic heart failure. RESULTS AWA administration reduced body weight gain, myocardial fibrosis, cardiomyocyte hypertrophy, and apoptosis, and rats showed improvement in cardiac function compared to model group. The extract significantly ameliorated the AAC-induced altered expression of heart failure markers such as ANP, NT-proBNP, and β-MHC, as well as fibrosis, hypertrophy markers MMP-2 and MMP-9, and other heart failure-related factors including plasma levels of TNF-α and IL-6. Furthermore, the extract reduced the protein expression of α-SMA, Col Ⅰ, and Col Ⅲ in the left ventricular (LV), thus inhibiting the LV remodeling associated with CHF. In addition, proteomics characterization of differentially expressed proteins showed that AWA administration inhibited left ventricular remodeling in CHF rats via a calcium signaling pathway, and reversed the expression of RyR2 and SERCA2a. CONCLUSIONS AWA extract exerts beneficial effects in an AAC-induced CHF model in rats, which was associated with an improvement in LV function, hypertrophy, fibrosis, and apoptotic status. These effects may be related to the regulation of calcium signaling by the altered expression of RyR2 and SERCA2a.
Collapse
MESH Headings
- Aconitum/chemistry
- Animals
- Apoptosis/drug effects
- Calcium Signaling/drug effects
- Cardiovascular Agents/isolation & purification
- Cardiovascular Agents/pharmacology
- Chronic Disease
- Disease Models, Animal
- Fibrosis
- Heart Failure/drug therapy
- Heart Failure/metabolism
- Heart Failure/pathology
- Heart Failure/physiopathology
- Hypertrophy, Left Ventricular/drug therapy
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Plant Extracts/isolation & purification
- Plant Extracts/pharmacology
- Rats, Sprague-Dawley
- Ryanodine Receptor Calcium Release Channel/metabolism
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Solubility
- Solvents/chemistry
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
- Water/chemistry
- Rats
Collapse
Affiliation(s)
- Xin Xu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Xiaofang Xie
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Huiqiong Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Pei Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Gangmin Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Junren Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Guanru Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Xiaoyu Cao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Liang Xiong
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu 611137, China.
| | - Cheng Peng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China.
| |
Collapse
|
23
|
Agrawal A, Rathor R, Kumar R, Suryakumar G, Singh SN, Kumar B. Redox modification of ryanodine receptor contributes to impaired Ca 2+ homeostasis and exacerbates muscle atrophy under high altitude. Free Radic Biol Med 2020; 160:643-656. [PMID: 32916280 DOI: 10.1016/j.freeradbiomed.2020.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/17/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022]
Abstract
At extreme altitude, prolonged and severe hypoxia menaces human function and survival, and also associated with profound loss of muscle mass which results into a debilitating critical illness of skeletal muscle atrophy. Hypobaric hypoxia altered redox homeostasis and impaired calcium ion handling in skeletal muscles. Dysregulated Ca2+ homeostasis and activated calpain is the prime stressor in high altitude hypoxia while the reason for subsequent abnormal release of pathological Ca2+ into cytoplasm is largely unexplored. The present study identified the redox remodeling in the Ca2+ release channel, Ryanodine Receptor (RyR1) owing to its hypernitrosylation state in skeletal muscles in chronic hypobaric hypoxia exposed rats. RyR1-hypernitrosylation decreases the binding of FKBP12/calstabin-1 and other complexes from the channel, causing "leakiness" in RyR1 ion-channel. A strong RyR1 stabilizer, S107 enhanced binding affinity of FKBP12 with hypernitrosylated RyR1, reduced Sarco(endo)plasmic reticulum (SR) Ca2+ leak and improved muscle strength and function under chronic hypoxia. Administration of S107 inhibited the skeletal muscle damage, maintained ultrastructure of sarcomere and sarcolemmal integrity. Histological analysis proved the increase in cross-sectional area of myofibers. Further, the number of apoptotic cells was also reduced by S107 treatment. Conclusively, we proposed that the redox remodeling of RyR1 (hypernitrosylated-RyR1) might be responsible for dysregulated Ca2+ homeostasis which consequently impaired muscle strength and function in response to chronic hypoxic stress. Reduced SR Ca2+ leak and enhanced binding affinity of FKBP12 may provide a novel therapeutic avenue in ameliorating skeletal muscle atrophy at high altitude.
Collapse
Affiliation(s)
- Akanksha Agrawal
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, New Delhi, 110054, India
| | - Richa Rathor
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, New Delhi, 110054, India.
| | - Ravi Kumar
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, New Delhi, 110054, India
| | - Geetha Suryakumar
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, New Delhi, 110054, India
| | - Som Nath Singh
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, New Delhi, 110054, India
| | - Bhuvnesh Kumar
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, New Delhi, 110054, India
| |
Collapse
|
24
|
Dridi H, Kushnir A, Zalk R, Yuan Q, Melville Z, Marks AR. Intracellular calcium leak in heart failure and atrial fibrillation: a unifying mechanism and therapeutic target. Nat Rev Cardiol 2020; 17:732-747. [PMID: 32555383 PMCID: PMC8362847 DOI: 10.1038/s41569-020-0394-8] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/06/2020] [Indexed: 12/14/2022]
Abstract
Ca2+ is a fundamental second messenger in all cell types and is required for numerous essential cellular functions, including cardiac and skeletal muscle contraction. The intracellular concentration of free Ca2+ ([Ca2+]) is regulated primarily by ion channels, pumps (ATPases), exchangers and Ca2+-binding proteins. Defective regulation of [Ca2+] is found in a diverse spectrum of pathological states that affect all the major organs. In the heart, abnormalities in the regulation of cytosolic and mitochondrial [Ca2+] occur in heart failure (HF) and atrial fibrillation (AF), two common forms of heart disease and leading contributors to morbidity and mortality. In this Review, we focus on the mechanisms that regulate ryanodine receptor 2 (RYR2), the major sarcoplasmic reticulum (SR) Ca2+-release channel in the heart, how RYR2 becomes dysfunctional in HF and AF, and its potential as a therapeutic target. Inherited RYR2 mutations and/or stress-induced phosphorylation and oxidation of the protein destabilize the closed state of the channel, resulting in a pathological diastolic Ca2+ leak from the SR that both triggers arrhythmias and impairs contractility. On the basis of our increased understanding of SR Ca2+ leak as a shared Ca2+-dependent pathological mechanism in HF and AF, a new class of drugs developed in our laboratory, known as rycals, which stabilize RYR2 channels and prevent Ca2+ leak from the SR, are undergoing investigation in clinical trials.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Alexander Kushnir
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Ran Zalk
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Zephan Melville
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
25
|
García-Rivas G, Castillo EC, Gonzalez-Gil AM, Maravillas-Montero JL, Brunck M, Torres-Quintanilla A, Elizondo-Montemayor L, Torre-Amione G. The role of B cells in heart failure and implications for future immunomodulatory treatment strategies. ESC Heart Fail 2020; 7:1387-1399. [PMID: 32533765 PMCID: PMC7373901 DOI: 10.1002/ehf2.12744] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 12/17/2022] Open
Abstract
Despite numerous demonstrations that the immune system is activated in heart failure, negatively affecting patients' outcomes, no definitive treatment strategy exists directed to modulate the immune system. In this review, we present the evidence that B cells contribute to the development of hypertrophy, inflammation, and maladaptive tissue remodelling. B cells produce antibodies that interfere with cardiomyocyte function, which culminates as the result of recruitment and activation of a variety of innate and structural cell populations, including neutrophils, macrophages, fibroblasts, and T cells. As B cells appear as active players in heart failure, we propose here novel immunomodulatory therapeutic strategies that target B cells and their products.
Collapse
Affiliation(s)
- Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León, Mexico.,Tecnologico de Monterrey, Hospital Zambrano Hellion, TecSalud, Centro de Investigación Biomédica, San Pedro Garza García, Nuevo León, Mexico
| | - Elena Cristina Castillo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León, Mexico
| | - Adrian M Gonzalez-Gil
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León, Mexico
| | - José Luis Maravillas-Montero
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Marion Brunck
- Tecnologico de Monterrey, School of Engineering and Science, FEMSA Biotechnology Center, Monterrey, Nuevo León, Mexico
| | - Alejandro Torres-Quintanilla
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León, Mexico
| | - Leticia Elizondo-Montemayor
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León, Mexico.,Tecnologico de Monterrey, Hospital Zambrano Hellion, TecSalud, Centro de Investigación Biomédica, San Pedro Garza García, Nuevo León, Mexico
| | - Guillermo Torre-Amione
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León, Mexico.,Tecnologico de Monterrey, Hospital Zambrano Hellion, TecSalud, Centro de Investigación Biomédica, San Pedro Garza García, Nuevo León, Mexico.,Weill Cornell Medical College, Methodist DeBakey Heart & Vascular Center, The Methodist Hospital, Houston, TX, USA
| |
Collapse
|
26
|
Moellmann J, Klinkhammer BM, Droste P, Kappel B, Haj-Yehia E, Maxeiner S, Artati A, Adamski J, Boor P, Schütt K, Lopaschuk GD, Verma S, Marx N, Lehrke M. Empagliflozin improves left ventricular diastolic function of db/db mice. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165807. [PMID: 32353614 DOI: 10.1016/j.bbadis.2020.165807] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Investigation of the effect of SGLT2 inhibition by empagliflozin on left ventricular function in a model of diabetic cardiomyopathy. BACKGROUND SGLT2 inhibition is a new strategy to treat diabetes. In the EMPA-REG Outcome trial empagliflozin treatment reduced cardiovascular and overall mortality in patients with diabetes presumably due to beneficial cardiac effects, leading to reduced heart failure hospitalization. The relevant mechanisms remain currently elusive but might be mediated by a shift in cardiac substrate utilization leading to improved energetic supply to the heart. METHODS We used db/db mice on high-fat western diet with or without empagliflozin treatment as a model of severe diabetes. Left ventricular function was assessed by pressure catheter with or without dobutamine stress. RESULTS Treatment with empagliflozin significantly increased glycosuria, improved glucose metabolism, ameliorated left ventricular diastolic function and reduced mortality of mice. This was associated with reduced cardiac glucose concentrations and decreased calcium/calmodulin-dependent protein kinase (CaMKII) activation with subsequent less phosphorylation of the ryanodine receptor (RyR). No change of cardiac ketone bodies or branched-chain amino acid (BCAA) metabolites in serum was detected nor was cardiac expression of relevant catabolic enzymes for these substrates affected. CONCLUSIONS In a murine model of severe diabetes empagliflozin-dependent SGLT2 inhibition improved diastolic function and reduced mortality. Improvement of diastolic function was likely mediated by reduced spontaneous diastolic sarcoplasmic reticulum (SR) calcium release but independent of changes in cardiac ketone and BCAA metabolism.
Collapse
Affiliation(s)
- Julia Moellmann
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Barbara M Klinkhammer
- Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Patrick Droste
- Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Ben Kappel
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Elias Haj-Yehia
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Sebastian Maxeiner
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Anna Artati
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Centrum Munich, German Research Center for Environmental Health (GmbH), Munich-Neuherberg, Germany
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Centrum Munich, German Research Center for Environmental Health (GmbH), Munich-Neuherberg, Germany; Chair of Experimental Genetics, Technical University of Munich, Freising-Weihenstephan, Germany; German Center for Diabetes Research (DZD e.V.), Munich-Neuherberg, Germany; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Peter Boor
- Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Katharina Schütt
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Subodh Verma
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada
| | - Nikolaus Marx
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany.
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
27
|
Delrio-Lorenzo A, Rojo-Ruiz J, Alonso MT, García-Sancho J. Sarcoplasmic reticulum Ca 2+ decreases with age and correlates with the decline in muscle function in Drosophila. J Cell Sci 2020; 133:jcs240879. [PMID: 32005702 DOI: 10.1242/jcs.240879] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/24/2020] [Indexed: 08/31/2023] Open
Abstract
Sarcopenia, the loss of muscle mass and strength associated with age, has been linked to impairment of the cytosolic Ca2+ peak that triggers muscle contraction, but mechanistic details remain unknown. Here we explore the hypothesis that a reduction in sarcoplasmic reticulum (SR) Ca2+ concentration ([Ca2+]SR) is at the origin of this loss of Ca2+ homeostasis. We engineered Drosophila melanogaster to express the Ca2+ indicator GAP3 targeted to muscle SR, and we developed a new method to calibrate the signal into [Ca2+]SRin vivo [Ca2+]SR fell with age from ∼600 µM to 50 µM in close correlation with muscle function, which declined monotonically when [Ca2+]SR was <400 µM. [Ca2+]SR results from the pump-leak steady state at the SR membrane. However, changes in expression of the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump and of the ryanodine receptor leak were too modest to explain the large changes seen in [Ca2+]SR Instead, these changes are compatible with increased leakiness through the ryanodine receptor as the main determinant of the [Ca2+]SR decline in aging muscle. In contrast, there were no changes in endoplasmic reticulum [Ca2+] with age in brain neurons.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Alba Delrio-Lorenzo
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), c/Sanz y Forés 3, 47003 Valladolid, Spain
| | - Jonathan Rojo-Ruiz
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), c/Sanz y Forés 3, 47003 Valladolid, Spain
| | - María Teresa Alonso
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), c/Sanz y Forés 3, 47003 Valladolid, Spain
| | - Javier García-Sancho
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), c/Sanz y Forés 3, 47003 Valladolid, Spain
| |
Collapse
|
28
|
Sardu C, Santulli G, Guerra G, Trotta MC, Santamaria M, Sacra C, Testa N, Ducceschi V, Gatta G, Amico MD, Sasso FC, Paolisso G, Marfella R. Modulation of SERCA in Patients with Persistent Atrial Fibrillation Treated by Epicardial Thoracoscopic Ablation: The CAMAF Study. J Clin Med 2020; 9:544. [PMID: 32079238 PMCID: PMC7074346 DOI: 10.3390/jcm9020544] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES To evaluate atrial fibrillation (AF) recurrence and Sarcoplasmic Endoplasmic Reticulum Calcium ATPase (SERCA) levels in patients treated by epicardial thoracoscopic ablation for persistent AF. BACKGROUND Reduced levels of SERCA have been reported in the peripheral blood cells of patients with AF. We hypothesize that SERCA levels can predict the response to epicardial ablation. METHODS We designed a prospective, multicenter, observational study to recruit, from October 2014 to June 2016, patients with persistent AF receiving an epicardial thoracoscopic pulmonary vein isolation. RESULTS We enrolled 27 patients. Responders (n = 15) did not present AF recurrence after epicardial ablation at one-year follow-up; these patients displayed a marked remodeling of the left atrium, with a significant reduction of inflammatory cytokines, B type natriuretic peptide (BNP), and increased levels of SERCA compared to baseline and to nonresponders (p < 0.05). Furthermore, mean AF duration (Heart rate (HR) 1.235 (1.037-1.471), p < 0.05), Left atrium volume (LAV) (HR 1.755 (1.126-2.738), p < 0.05), BNP (HR 1.945 (1.895-1.999), p < 0.05), and SERCA (HR 1.763 (1.167-2.663), p < 0.05) were predictive of AF recurrence. CONCLUSIONS Our data indicate for the first time that baseline values of SERCA in patients with persistent AF might be predictive of failure to epicardial ablative approach. Intriguingly, epicardial ablation was associated with increased levels of SERCA in responders. Therefore, SERCA might be an innovative therapeutic target to improve the response to epicardial ablative treatments.
Collapse
Affiliation(s)
- Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (G.G.); (F.C.S.); (G.P.); (R.M.)
- Department of Medical Sciences, International University of Health and Medical Sciences “Saint Camillus”, 00131Rome, Italy
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, International Translational Research and Medical Education Academic Research Unit (ITME), “Federico II” University, 80138 Naples, Italy;
- Department of Medicine, Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Montefiore University Hospital, New York, NY 10461, USA
| | - Germano Guerra
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86010, Campobasso, Italy;
| | - Maria Consiglia Trotta
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (G.G.); (F.C.S.); (G.P.); (R.M.)
| | - Matteo Santamaria
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.S.); (M.D.A.)
| | - Cosimo Sacra
- Cardiovascular and Arrhythmias Department, Catholic University of Sacred Heart, 86010 Campobasso, Italy; (C.S.); (N.T.)
| | - Nicola Testa
- Cardiovascular and Arrhythmias Department, Catholic University of Sacred Heart, 86010 Campobasso, Italy; (C.S.); (N.T.)
| | - Valentino Ducceschi
- Cardiovascular and Arrhythmias Department, “Vecchio Pellegrini” Hospital; 80138 Naples, Italy;
| | - Gianluca Gatta
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (G.G.); (F.C.S.); (G.P.); (R.M.)
| | - Michele D’ Amico
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.S.); (M.D.A.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (G.G.); (F.C.S.); (G.P.); (R.M.)
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (G.G.); (F.C.S.); (G.P.); (R.M.)
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (G.G.); (F.C.S.); (G.P.); (R.M.)
| |
Collapse
|
29
|
Morelli MB, Shu J, Sardu C, Matarese A, Santulli G. Cardiosomal microRNAs Are Essential in Post-Infarction Myofibroblast Phenoconversion. Int J Mol Sci 2019; 21:201. [PMID: 31892162 PMCID: PMC6982041 DOI: 10.3390/ijms21010201] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 12/14/2022] Open
Abstract
The inclusion of microRNAs (miRNAs) in extracellular microvesicles/exosomes (named cardiosomes when deriving from cardiomyocytes) allows their active transportation and ensures cell-cell communication. We hypothesize that cardiosomal miRNAs play a pivotal role in the activation of myofibroblasts following ischemic injury. Using a murine model of myocardial infarction (MI), we tested our hypothesis by measuring in isolated fibroblasts and cardiosomes the expression levels of a set of miRNAs, which are upregulated in cardiomyocytes post-MI and involved in myofibroblast phenoconversion. We found that miR-195 was significantly upregulated in cardiosomes and in fibroblasts isolated after MI compared with SHAM conditions. Moreover, primary isolated cardiac fibroblasts were activated both when incubated with cardiosomes isolated from ischemic cardiomyocytes and when cultured in conditioned medium of post-MI cardiomyocytes, whereas no significant effect was observed following incubation with cardiosomes or medium from sham cardiomyocytes. Taken together, our findings indicate for the first time that a cardiomyocyte-specific miRNA, transferred to fibroblasts in form of exosomal cargo, is crucial in the activation of myofibroblasts.
Collapse
Affiliation(s)
- Marco B. Morelli
- Department of Medicine, Division of Cardiology and Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, Montefiore University Hospital, New York, NY 10461, USA; (M.B.M.); (J.S.)
| | - Jun Shu
- Department of Medicine, Division of Cardiology and Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, Montefiore University Hospital, New York, NY 10461, USA; (M.B.M.); (J.S.)
| | - Celestino Sardu
- Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, University of Campania “Luigi Vanvitelli”, 80100 Naples, Italy;
| | - Alessandro Matarese
- Department of Pneumology and Oncology, AORN “Ospedale dei Colli”, 80100 Naples, Italy;
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology and Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, Montefiore University Hospital, New York, NY 10461, USA; (M.B.M.); (J.S.)
- Department of Advanced Biomedical Science, “Federico II” University, and International Translational Research and Medical Education Consortium (ITME), 80131 Naples, Italy
| |
Collapse
|
30
|
Pathophysiology of Calcium Mediated Ventricular Arrhythmias and Novel Therapeutic Options with Focus on Gene Therapy. Int J Mol Sci 2019; 20:ijms20215304. [PMID: 31653119 PMCID: PMC6862059 DOI: 10.3390/ijms20215304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias constitute a major health problem with a huge impact on mortality rates and health care costs. Despite ongoing research efforts, the understanding of the molecular mechanisms and processes responsible for arrhythmogenesis remains incomplete. Given the crucial role of Ca2+-handling in action potential generation and cardiac contraction, Ca2+ channels and Ca2+ handling proteins represent promising targets for suppression of ventricular arrhythmias. Accordingly, we report the different roles of Ca2+-handling in the development of congenital as well as acquired ventricular arrhythmia syndromes. We highlight the therapeutic potential of gene therapy as a novel and innovative approach for future arrhythmia therapy. Furthermore, we discuss various promising cellular and mitochondrial targets for therapeutic gene transfer currently under investigation.
Collapse
|
31
|
Chen J, Xue R, Li L, Xiao LL, Shangguan J, Zhang W, Bai X, Liu G, Li L. Panax Notoginseng Saponins Protect Cardiac Myocytes Against Endoplasmic Reticulum Stress and Associated Apoptosis Through Mediation of Intracellular Calcium Homeostasis. Front Pharmacol 2019; 10:1013. [PMID: 31616293 PMCID: PMC6764115 DOI: 10.3389/fphar.2019.01013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/08/2019] [Indexed: 12/30/2022] Open
Abstract
Endoplasmic reticulum (ER) stress has been demonstrated to play important roles in the pathogenesis of various cardiovascular diseases. The ER stress pathway is therefore a promising therapeutic target in cardiovascular disease. Although Panax notoginseng saponins (PNS) are one of the patent medicines that are traditionally used to treat cardiovascular disorders, their effects on ER stress in cardiac myocytes remain unexploited so far. This study investigates the effects of PNS on ER stress and its associated cell apoptosis along with the related mechanism in cardiac myocytes. PNS compounds were identified via high-performance liquid chromatograph (HPLC) assay. PNS-pretreated H9c2 cells, HL-1 cells, and primary cultured neonatal rat cardiomyocytes were stimulated with thapsigargin (TG) to induce ER stress response and apoptosis. ER stress response was tested by immunofluorescence or immunoblot of the ER protein chaperones—calnexin, binding immunoglobulin protein (BiP) and the C/EBP homologous protein (CHOP). Cell viability was tested by methyl thiazolyl tetrazolium (MTT) assay. Cell apoptosis was detected by immunoblot of Cleaved caspase-3 and flow cytometry analysis of Annexin V/propidium iodide (PI) staining. Cytosolic, mitochondrial, and ER calcium dynamics were investigated by calcium imaging. Moreover, a ryanodine receptor type-2 (RyR2) overexpression stable cell line was generated to verify the mechanism of RyR2 involved in PNS in the inhibition of ER stress and cell apoptosis. We demonstrate here that PNS protected cardiac myocytes from ER stress response and associated cell death in a concentration-dependent manner. Importantly, PNS reduced the elevation of cytosolic calcium, mitochondria calcium, as well as ER calcium in response to either TG or histamine treatment. PNS protection in ER stress was regulated by RyR2 expression. In summary, PNS protection against TG-induced ER stress response and its associated cell apoptosis in cardiac myocytes is calcium dependent. Through the regulation of ER calcium release mediated by RyR2, a novel mechanism for PNS in the prevention of cardiovascular diseases is thereby identified.
Collapse
Affiliation(s)
- Jun Chen
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Li
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Li Xiao
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiahong Shangguan
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenjing Zhang
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueyang Bai
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gangqiong Liu
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ling Li
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Vaamonde-García C, López-Armada MJ. Role of mitochondrial dysfunction on rheumatic diseases. Biochem Pharmacol 2019; 165:181-195. [DOI: 10.1016/j.bcp.2019.03.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/07/2019] [Indexed: 02/09/2023]
|
33
|
Xie J, Du G, Zhang Y, Zhou F, Wu J, Jiao H, Li Y, Chen Y, Ouyang L, Bo D, Feng C, Yang W, Fan G. ECG conduction disturbances and ryanodine receptor expression levels in occupational lead exposure workers. Occup Environ Med 2019; 76:151-156. [DOI: 10.1136/oemed-2018-105463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/11/2018] [Accepted: 12/15/2018] [Indexed: 11/03/2022]
Abstract
ObjectivesA significant number of researches have evidenced that occupational lead (Pb) exposure increased risks of cardiovascular disease. However, evidences about the potential effects of Pb on the cardiac conduction system are sparse and inconclusive. Besides, ryanodine receptors (RyRs) induced dysfunction of cardiac excitation contraction coupling which is considered to be one of the mechanisms in cardiovascular diseases. Therefore, we examined the association between occupational Pb exposure and ECG conduction abnormalities, as well as RyRs in Pb-induced ECG abnormalities.MethodsWe investigated 529 Pb smelter workers, and measured blood lead (BPb), zinc protoporphyrin (ZPP), ECG outcomes and RyR expression levels. Based on BPb levels, the workers were divided into three groups: the BPb not elevated group, the BPb elevated group and the Pb poisoning group. Descriptive and multivariable analyses were performed.ResultsCompared with the BPb not elevated group, the Pb poisoning group had a higher incidence of high QRS voltage, and a lower level of RyR1 gene expression (p<0.05). Further unconditional multivariable logistic regression analyses showed that high QRS voltage was positively related to BPb (OR=1.045, 95% CI 1.014 to 1.078) and inversely associated with RyR1 expression (OR=0.042, 95% CI 0.002 to 0.980) after adjusting for potential confounders. In addition, multiple linear regression analyses showed that the QTc interval was positively associated with ZPP (β=0.299, 95% CI 0.130 to 0.468) after adjusting for potential confounders.ConclusionsOur study provided evidences that occupational exposure to Pb may be associated with worse ECG outcomes (high QRS voltage), which might be related to decreased levels of RyR1.
Collapse
|
34
|
Circulating level of fibroblast growth factor 21 is independently associated with the risks of unstable angina pectoris. Biosci Rep 2018; 38:BSR20181099. [PMID: 30185439 PMCID: PMC6153373 DOI: 10.1042/bsr20181099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/21/2018] [Accepted: 08/28/2018] [Indexed: 01/02/2023] Open
Abstract
There is increasing evidence that serum adipokine levels are associated with higher risks of cardiovascular diseases. As an important adipokine, fibroblast growth factor 21 (FGF21) has been demonstrated to be associated with atherosclerosis and coronary artery disease (CAD). However, circulating level of FGF21 in patients with angina pectoris has not yet been investigated. Circulating FGF21 level was examined in 197 patients with stable angina pectoris (SAP, n=66), unstable angina pectoris (UAP, n=76), and control subjects (n=55) along with clinical variables of cardiovascular risk factors. Serum FGF21 concentrations on admission were significantly increased more in patients with UAP than those with SAP (Ln-FGF21: 5.26 ± 0.87 compared with 4.85 ± 0.77, P<0.05) and control subjects (natural logarithm (Ln)-FGF21: 5.26 ± 0.87 compared with 4.54 ± 0.72, P<0.01). The correlation analysis revealed that serum FGF21 concentration was positively correlated with the levels of cardiac troponin I (cTnI) (r2 = 0.026, P=0.027) and creatine kinase-MB (CK-MB) (r2 = 0.023, P= 0.04). Furthermore, FGF21 level was identified as an independent factor associated with the risks of UAP (odds ratio (OR): 2.781; 95% CI: 1.476–5.239; P=0.002), after adjusting for gender, age, and body mass index (BMI). However, there were no correlations between serum FGF21 levels and the presence of SAP (OR: 1.248; 95% CI: 0.703–2.215; P=0.448). The present study indicates that FGF21 has a strong correlation and precise predictability for increased risks of UAP, that is independent of traditional risk factors of angina pectoris.
Collapse
|
35
|
Negative chronotropism, positive inotropism and lusitropism of 3,5-di-t-butyl-4-hydroxyanisole (DTBHA) on rat heart preparations occur through reduction of RyR2 Ca2+ leak. Biochem Pharmacol 2018; 155:434-443. [DOI: 10.1016/j.bcp.2018.07.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/19/2018] [Indexed: 12/31/2022]
|