101
|
Sim CB, Phipson B, Ziemann M, Rafehi H, Mills RJ, Watt KI, Abu-Bonsrah KD, Kalathur RK, Voges HK, Dinh DT, ter Huurne M, Vivien CJ, Kaspi A, Kaipananickal H, Hidalgo A, Delbridge LM, Robker RL, Gregorevic P, dos Remedios CG, Lal S, Piers AT, Konstantinov IE, Elliott DA, El-Osta A, Oshlack A, Hudson JE, Porrello ER. Sex-Specific Control of Human Heart Maturation by the Progesterone Receptor. Circulation 2021; 143:1614-1628. [PMID: 33682422 PMCID: PMC8055196 DOI: 10.1161/circulationaha.120.051921] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Despite in-depth knowledge of the molecular mechanisms controlling embryonic heart development, little is known about the signals governing postnatal maturation of the human heart. METHODS Single-nucleus RNA sequencing of 54 140 nuclei from 9 human donors was used to profile transcriptional changes in diverse cardiac cell types during maturation from fetal stages to adulthood. Bulk RNA sequencing and the Assay for Transposase-Accessible Chromatin using sequencing were used to further validate transcriptional changes and to profile alterations in the chromatin accessibility landscape in purified cardiomyocyte nuclei from 21 human donors. Functional validation studies of sex steroids implicated in cardiac maturation were performed in human pluripotent stem cell-derived cardiac organoids and mice. RESULTS Our data identify the progesterone receptor as a key mediator of sex-dependent transcriptional programs during cardiomyocyte maturation. Functional validation studies in human cardiac organoids and mice demonstrate that the progesterone receptor drives sex-specific metabolic programs and maturation of cardiac contractile properties. CONCLUSIONS These data provide a blueprint for understanding human heart maturation in both sexes and reveal an important role for the progesterone receptor in human heart development.
Collapse
Affiliation(s)
- Choon Boon Sim
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Belinda Phipson
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre (B.P., A.O.), University of Melbourne, Victoria, Australia
| | - Mark Ziemann
- Department of Diabetes, Central Clinical School, Alfred Centre, Monash University, Melbourne, Victoria, Australia (M.Z., H.R., A.K., H.K., A.E.-O.)
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, Australia (M.Z.)
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Haloom Rafehi
- Department of Diabetes, Central Clinical School, Alfred Centre, Monash University, Melbourne, Victoria, Australia (M.Z., H.R., A.K., H.K., A.E.-O.)
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Richard J. Mills
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia (R.J.M., J.E.H.)
| | - Kevin I. Watt
- Peter MacCallum Cancer Centre (B.P., A.O.), University of Melbourne, Victoria, Australia
- Centre for Muscle Research (K.I.W., P.G., E.R.P.), University of Melbourne, Victoria, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Kwaku D. Abu-Bonsrah
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences, and Department of Paediatrics (K.D.A.-B., H.K.V., A.H., I.E.K., D.A.E.), University of Melbourne, Victoria, Australia
| | - Ravi K.R. Kalathur
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Holly K. Voges
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences, and Department of Paediatrics (K.D.A.-B., H.K.V., A.H., I.E.K., D.A.E.), University of Melbourne, Victoria, Australia
| | - Doan T. Dinh
- Robinson Research Institute, The University of Adelaide, South Australia, Australia (D.T.D., R.L.R.)
| | - Menno ter Huurne
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Celine J. Vivien
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Antony Kaspi
- Department of Diabetes, Central Clinical School, Alfred Centre, Monash University, Melbourne, Victoria, Australia (M.Z., H.R., A.K., H.K., A.E.-O.)
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Harikrishnan Kaipananickal
- Department of Diabetes, Central Clinical School, Alfred Centre, Monash University, Melbourne, Victoria, Australia (M.Z., H.R., A.K., H.K., A.E.-O.)
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Alejandro Hidalgo
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences, and Department of Paediatrics (K.D.A.-B., H.K.V., A.H., I.E.K., D.A.E.), University of Melbourne, Victoria, Australia
| | - Leanne M.D. Delbridge
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology (K.I.W., L.M.D.D., P.G., E.R.P.), University of Melbourne, Victoria, Australia
| | - Rebecca L. Robker
- Robinson Research Institute, The University of Adelaide, South Australia, Australia (D.T.D., R.L.R.)
| | - Paul Gregorevic
- Department of Anatomy and Physiology (K.I.W., L.M.D.D., P.G., E.R.P.), University of Melbourne, Victoria, Australia
- Centre for Muscle Research (K.I.W., P.G., E.R.P.), University of Melbourne, Victoria, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
| | - Cristobal G. dos Remedios
- School of Medical Sciences, The University of Sydney, New South Wales, Australia (C.G.d.R., S.L.)
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia (C.G.d.R.)
| | - Sean Lal
- School of Medical Sciences, The University of Sydney, New South Wales, Australia (C.G.d.R., S.L.)
| | - Adam T. Piers
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Igor E. Konstantinov
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Department of Cardiac Surgery (I.E.K.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences, and Department of Paediatrics (K.D.A.-B., H.K.V., A.H., I.E.K., D.A.E.), University of Melbourne, Victoria, Australia
| | - David A. Elliott
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences, and Department of Paediatrics (K.D.A.-B., H.K.V., A.H., I.E.K., D.A.E.), University of Melbourne, Victoria, Australia
| | - Assam El-Osta
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Alicia Oshlack
- Department of Diabetes, Central Clinical School, Alfred Centre, Monash University, Melbourne, Victoria, Australia (M.Z., H.R., A.K., H.K., A.E.-O.)
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.Z., H.R., K.I.W., A.K., H.K., P.G., A.E.-O.)
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, Li Ka Shing Institute of Health Sciences, and The Chinese University of Hong Kong, China (A.E.-O.)
| | - James E. Hudson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia (R.J.M., J.E.H.)
- Centre for Cardiac and Vascular Biology, School of Biomedical Sciences, The University of Queensland, Brisbane, Australia (J.E.H., E.R.P.)
| | - Enzo R. Porrello
- Murdoch Children’s Research Institute (C.B.S., B.P., K.D.A.-B., R.K.R.K., H.K.V., M.t.H., C.J.V., A.H., A.T.P., I.E.K., D.A.E., A.O., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine (C.B.S., K.D.A.-B., R.K.R.K., M.t.H., C.J.V., L.M.D.D., A.T.P., I.E.K., D.A.E., E.R.P.), The Royal Children’s Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology (K.I.W., L.M.D.D., P.G., E.R.P.), University of Melbourne, Victoria, Australia
- Centre for Muscle Research (K.I.W., P.G., E.R.P.), University of Melbourne, Victoria, Australia
- Centre for Cardiac and Vascular Biology, School of Biomedical Sciences, The University of Queensland, Brisbane, Australia (J.E.H., E.R.P.)
| |
Collapse
|
102
|
Mills RJ, Humphrey SJ, Fortuna PRJ, Lor M, Foster SR, Quaife-Ryan GA, Johnston RL, Dumenil T, Bishop C, Rudraraju R, Rawle DJ, Le T, Zhao W, Lee L, Mackenzie-Kludas C, Mehdiabadi NR, Halliday C, Gilham D, Fu L, Nicholls SJ, Johansson J, Sweeney M, Wong NCW, Kulikowski E, Sokolowski KA, Tse BWC, Devilée L, Voges HK, Reynolds LT, Krumeich S, Mathieson E, Abu-Bonsrah D, Karavendzas K, Griffen B, Titmarsh D, Elliott DA, McMahon J, Suhrbier A, Subbarao K, Porrello ER, Smyth MJ, Engwerda CR, MacDonald KPA, Bald T, James DE, Hudson JE. BET inhibition blocks inflammation-induced cardiac dysfunction and SARS-CoV-2 infection. Cell 2021; 184:2167-2182.e22. [PMID: 33811809 PMCID: PMC7962543 DOI: 10.1016/j.cell.2021.03.026] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/10/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022]
Abstract
Cardiac injury and dysfunction occur in COVID-19 patients and increase the risk of mortality. Causes are ill defined but could be through direct cardiac infection and/or inflammation-induced dysfunction. To identify mechanisms and cardio-protective drugs, we use a state-of-the-art pipeline combining human cardiac organoids with phosphoproteomics and single nuclei RNA sequencing. We identify an inflammatory "cytokine-storm", a cocktail of interferon gamma, interleukin 1β, and poly(I:C), induced diastolic dysfunction. Bromodomain-containing protein 4 is activated along with a viral response that is consistent in both human cardiac organoids (hCOs) and hearts of SARS-CoV-2-infected K18-hACE2 mice. Bromodomain and extraterminal family inhibitors (BETi) recover dysfunction in hCOs and completely prevent cardiac dysfunction and death in a mouse cytokine-storm model. Additionally, BETi decreases transcription of genes in the viral response, decreases ACE2 expression, and reduces SARS-CoV-2 infection of cardiomyocytes. Together, BETi, including the Food and Drug Administration (FDA) breakthrough designated drug, apabetalone, are promising candidates to prevent COVID-19 mediated cardiac damage.
Collapse
Affiliation(s)
- Richard J Mills
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney 2006, NSW, Australia
| | | | - Mary Lor
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Simon R Foster
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | | | - Rebecca L Johnston
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Troy Dumenil
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Cameron Bishop
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Rajeev Rudraraju
- The WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne 3000, VIC, Australia; Department of Microbiology and Immunology, The University of Melbourne, Melbourne 3052, VIC, Australia; The Peter Doherty Institute for Infection and Immunity, Melbourne 3000, VIC, Australia
| | - Daniel J Rawle
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Thuy Le
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Wei Zhao
- The Peter Doherty Institute for Infection and Immunity, Melbourne 3000, VIC, Australia
| | - Leo Lee
- The Peter Doherty Institute for Infection and Immunity, Melbourne 3000, VIC, Australia
| | | | - Neda R Mehdiabadi
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne 3052, VIC, Australia
| | | | - Dean Gilham
- Resverlogix Corp., Calgary T3E 6L1, AB, Canada
| | - Li Fu
- Resverlogix Corp., Calgary T3E 6L1, AB, Canada
| | - Stephen J Nicholls
- Victorian Heart Hospital, Monash University, Clayton 3168, VIC, Australia
| | | | | | | | | | - Kamil A Sokolowski
- Preclinical Imaging Facility, Translational Research Institute, Brisbane, QLD, Australia
| | - Brian W C Tse
- Preclinical Imaging Facility, Translational Research Institute, Brisbane, QLD, Australia
| | - Lynn Devilée
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Holly K Voges
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Liam T Reynolds
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Sophie Krumeich
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Ellen Mathieson
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - Dad Abu-Bonsrah
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne 3052, VIC, Australia; Department of Paediatrics, The University of Melbourne, Melbourne 3052, VIC, Australia
| | - Kathy Karavendzas
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne 3052, VIC, Australia
| | - Brendan Griffen
- Dynomics Inc., San Mateo, CA 94401, USA; Dynomics Pty Ltd, Brisbane 4000, QLD, Australia
| | - Drew Titmarsh
- Dynomics Inc., San Mateo, CA 94401, USA; Dynomics Pty Ltd, Brisbane 4000, QLD, Australia
| | - David A Elliott
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne 3052, VIC, Australia
| | - James McMahon
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne 3004, VIC, Australia; Department of Infectious Diseases, Monash Medical Centre, Clayton 3168, VIC, Australia
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia; GVN Center of Excellence, Australian Infectious Diseases Research Centre, Brisbane, QLD, Australia
| | - Kanta Subbarao
- The WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne 3000, VIC, Australia; The Peter Doherty Institute for Infection and Immunity, Melbourne 3000, VIC, Australia
| | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne 3052, VIC, Australia; Department of Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne 3052, VIC, Australia
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | | | | | - Tobias Bald
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia; Institute of Experimental Oncology, University Hospital Bonn, Bonn 53127, Germany
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney 2006, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney 2006, NSW, Australia
| | - James E Hudson
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia.
| |
Collapse
|
103
|
Günthel M, van Duijvenboden K, Jeremiasse J, van den Hoff MJB, Christoffels VM. Early Postnatal Cardiac Stress Does Not Influence Ventricular Cardiomyocyte Cell-Cycle Withdrawal. J Cardiovasc Dev Dis 2021; 8:jcdd8040038. [PMID: 33917189 PMCID: PMC8068044 DOI: 10.3390/jcdd8040038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/31/2021] [Accepted: 04/05/2021] [Indexed: 12/27/2022] Open
Abstract
Congenital heart disease (CHD) is the most common birth defect. After birth, patients with CHD may suffer from cardiac stress resulting from abnormal loading conditions. However, it is not known how this cardiac burden influences postnatal development and adaptation of the ventricles. To study the transcriptional and cell-cycle response of neonatal cardiomyocytes to cardiac stress, we used a genetic mouse model that develops left ventricular volume overload within 2 weeks after birth. The increased volume load caused upregulation of the cardiac stress marker Nppa in the left ventricle and interventricular septum as early as 12 days after birth. Transcriptome analysis revealed that cardiac stress induced the expression of cell-cycle genes. This did not influence postnatal cell-cycle withdrawal of cardiomyocytes and other cell types in the ventricles as measured by Ki-67 immunostaining.
Collapse
|
104
|
Zheng L, Du J, Wang Z, Zhou Q, Zhu X, Xiong JW. Molecular regulation of myocardial proliferation and regeneration. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:13. [PMID: 33821373 PMCID: PMC8021683 DOI: 10.1186/s13619-021-00075-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/04/2021] [Indexed: 12/21/2022]
Abstract
Heart regeneration is a fascinating and complex biological process. Decades of intensive studies have revealed a sophisticated molecular network regulating cardiac regeneration in the zebrafish and neonatal mouse heart. Here, we review both the classical and recent literature on the molecular and cellular mechanisms underlying heart regeneration, with a particular focus on how injury triggers the cell-cycle re-entry of quiescent cardiomyocytes to replenish their massive loss after myocardial infarction or ventricular resection. We highlight several important signaling pathways for cardiomyocyte proliferation and propose a working model of how these injury-induced signals promote cardiomyocyte proliferation. Thus, this concise review provides up-to-date research progresses on heart regeneration for investigators in the field of regeneration biology.
Collapse
Affiliation(s)
- Lixia Zheng
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871, China
| | - Jianyong Du
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871, China
| | - Zihao Wang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871, China
| | - Qinchao Zhou
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871, China
| | - Xiaojun Zhu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871, China.
| | - Jing-Wei Xiong
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871, China
| |
Collapse
|
105
|
Campostrini G, Meraviglia V, Giacomelli E, van Helden RW, Yiangou L, Davis RP, Bellin M, Orlova VV, Mummery CL. Generation, functional analysis and applications of isogenic three-dimensional self-aggregating cardiac microtissues from human pluripotent stem cells. Nat Protoc 2021; 16:2213-2256. [PMID: 33772245 PMCID: PMC7611409 DOI: 10.1038/s41596-021-00497-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/11/2021] [Indexed: 02/01/2023]
Abstract
Tissue-like structures from human pluripotent stem cells containing multiple cell types are transforming our ability to model and understand human development and disease. Here we describe a protocol to generate cardiomyocytes (CMs), cardiac fibroblasts (CFs) and cardiac endothelial cells (ECs), the three principal cell types in the heart, from human induced pluripotent stem cells (hiPSCs) and combine them in three-dimensional (3D) cardiac microtissues (MTs). We include details of how to differentiate, isolate, cryopreserve and thaw the component cells and how to construct and analyze the MTs. The protocol supports hiPSC-CM maturation and allows replacement of one or more of the three heart cell types in the MTs with isogenic variants bearing disease mutations. Differentiation of each cell type takes ~30 d, while MT formation and maturation requires another 20 d. No specialist equipment is needed and the method is inexpensive, requiring just 5,000 cells per MT.
Collapse
Affiliation(s)
- Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Elisa Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ruben W.J. van Helden
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Richard P. Davis
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Department of Biology, University of Padua, 35121 Padua, Italy,Veneto Institute of Molecular Medicine, 35129 Padua, Italy,Correspondence to , or
| | - Valeria V. Orlova
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Correspondence to , or
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Department of Applied Stem Cell Technologies, University of Twente, The Netherlands,Correspondence to , or
| |
Collapse
|
106
|
Transcriptional Regulation of Postnatal Cardiomyocyte Maturation and Regeneration. Int J Mol Sci 2021; 22:ijms22063288. [PMID: 33807107 PMCID: PMC8004589 DOI: 10.3390/ijms22063288] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/17/2022] Open
Abstract
During the postnatal period, mammalian cardiomyocytes undergo numerous maturational changes associated with increased cardiac function and output, including hypertrophic growth, cell cycle exit, sarcomeric protein isoform switching, and mitochondrial maturation. These changes come at the expense of loss of regenerative capacity of the heart, contributing to heart failure after cardiac injury in adults. While most studies focus on the transcriptional regulation of embryonic or adult cardiomyocytes, the transcriptional changes that occur during the postnatal period are relatively unknown. In this review, we focus on the transcriptional regulators responsible for these aspects of cardiomyocyte maturation during the postnatal period in mammals. By specifically highlighting this transitional period, we draw attention to critical processes in cardiomyocyte maturation with potential therapeutic implications in cardiovascular disease.
Collapse
|
107
|
Application of genetic cell-lineage tracing technology to study cardiovascular diseases. J Mol Cell Cardiol 2021; 156:57-68. [PMID: 33745891 DOI: 10.1016/j.yjmcc.2021.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/03/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are leading causes that threaten people's life. To investigate cells that are involved in disease development and tissue repair, various technologies have been introduced. Among these technologies, lineage tracing is a powerful tool to track the fate of cells in vivo, providing deep insights into cellular behavior and plasticity. In cardiac diseases, newly formed cardiomyocytes and endothelial cells are found from proliferation of local cells, while fibroblasts and macrophages are originated from diverse cell sources. Similarly, in response to vascular injury, various sources of cells including media smooth muscle cells, endothelium, resident progenitors and bone marrow cells are involved in lesion formation and/or vessel regeneration. In summary, current review summarizes the development of lineage tracing techniques and their utilizations in investigating roles of different cell types in cardiovascular diseases.
Collapse
|
108
|
Taking Data Science to Heart: Next Scale of Gene Regulation. Curr Cardiol Rep 2021; 23:46. [PMID: 33721129 DOI: 10.1007/s11886-021-01467-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW Technical advances have facilitated high-throughput measurements of the genome in the context of cardiovascular biology. These techniques bring a deluge of gargantuan datasets, which in turn present two fundamentally new opportunities for innovation-data processing and knowledge integration-toward the goal of meaningful basic and translational discoveries. RECENT FINDINGS Big data, integrative analyses, and machine learning have brought cardiac investigations to the cutting edge of chromatin biology, not only to reveal basic principles of gene regulation in the heart, but also to aid in the design of targeted epigenetic therapies. SUMMARY Cardiac studies using big data are only beginning to integrate the millions of recorded data points and the tools of machine learning are aiding this process. Future experimental design should take into consideration insights from existing genomic datasets, thereby focusing on heretofore unexplored epigenomic contributions to disease pathology.
Collapse
|
109
|
Yuan X, Scott IC, Wilson MD. Heart Enhancers: Development and Disease Control at a Distance. Front Genet 2021; 12:642975. [PMID: 33777110 PMCID: PMC7987942 DOI: 10.3389/fgene.2021.642975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
Bound by lineage-determining transcription factors and signaling effectors, enhancers play essential roles in controlling spatiotemporal gene expression profiles during development, homeostasis and disease. Recent synergistic advances in functional genomic technologies, combined with the developmental biology toolbox, have resulted in unprecedented genome-wide annotation of heart enhancers and their target genes. Starting with early studies of vertebrate heart enhancers and ending with state-of-the-art genome-wide enhancer discovery and testing, we will review how studying heart enhancers in metazoan species has helped inform our understanding of cardiac development and disease.
Collapse
Affiliation(s)
- Xuefei Yuan
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ian C. Scott
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Michael D. Wilson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
110
|
Zhao H, Zhang Y, Xu X, Sun Q, Yang C, Wang H, Yang J, Yang Y, Yang X, Liu Y, Zhao Y. Sall4 and Myocd Empower Direct Cardiac Reprogramming From Adult Cardiac Fibroblasts After Injury. Front Cell Dev Biol 2021; 9:608367. [PMID: 33718351 PMCID: PMC7953844 DOI: 10.3389/fcell.2021.608367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 02/08/2021] [Indexed: 01/14/2023] Open
Abstract
Direct conversion of fibroblasts into induced cardiomyocytes (iCMs) holds promising potential to generate functional cardiomyocytes for drug development and clinical applications, especially for direct in situ heart regeneration by delivery of reprogramming genes into adult cardiac fibroblasts in injured hearts. For a decade, many cocktails of transcription factors have been developed to generate iCMs from fibroblasts of different tissues in vitro and some were applied in vivo. Here, we aimed to develop genetic cocktails that induce cardiac reprogramming directly in cultured cardiac fibroblasts isolated from adult mice with myocardial infarction (MICFs), which could be more relevant to heart diseases. We found that the widely used genetic cocktail, Gata4, Mef2c, and Tbx5 (GMT) were inefficient in reprogramming cardiomyocytes from MICFs. In a whole well of a 12-well plate, less than 10 mCherry+ cells (<0.1%) were observed after 2 weeks of GMT infection with Myh6-reporter transgenic MICFs. By screening 22 candidate transcription factors predicted through analyzing the gene regulatory network of cardiac development, we found that five factors, GMTMS (GMT plus Myocd and Sall4), induced more iCMs expressing the cardiac structural proteins cTnT and cTnI at a frequency of about 22.5 ± 2.7% of the transduced MICFs at day 21 post infection. What is more, GMTMS induced abundant beating cardiomyocytes at day 28 post infection. Specifically, Myocd contributed mainly to inducing the expression of cardiac proteins, while Sall4 accounted for the induction of functional properties, such as contractility. RNA-seq analysis of the iCMs at day 28 post infection revealed that they were reprogrammed to adopt a cardiomyocyte-like gene expression profile. Overall, we show here that Sall4 and Myocd play important roles in cardiac reprogramming from MICFs, providing a cocktail of genetic factors that have potential for further applications in in vivo cardiac reprogramming.
Collapse
Affiliation(s)
- Hong Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Yi Zhang
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Xiaochan Xu
- The Center for Models of Life, Niels Bohr Institute, Copenhagen, Denmark
| | - Qiushi Sun
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Chunyan Yang
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Hao Wang
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Junbo Yang
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Yang Yang
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Xiaochun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Yi Liu
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Yang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Plastech Pharmaceutical Technology Co., Ltd., Nanjing, China
| |
Collapse
|
111
|
Nicin L, Abplanalp WT, Schänzer A, Sprengel A, John D, Mellentin H, Tombor L, Keuper M, Ullrich E, Klingel K, Dettmeyer RB, Hoffmann J, Akintuerk H, Jux C, Schranz D, Zeiher AM, Rupp S, Dimmeler S. Single Nuclei Sequencing Reveals Novel Insights Into the Regulation of Cellular Signatures in Children With Dilated Cardiomyopathy. Circulation 2021; 143:1704-1719. [PMID: 33618539 DOI: 10.1161/circulationaha.120.051391] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a leading cause of death in children with heart failure. The outcome of pediatric heart failure treatment is inconsistent, and large cohort studies are lacking. Progress may be achieved through personalized therapy that takes age- and disease-related pathophysiology, pathology, and molecular fingerprints into account. We present single nuclei RNA sequencing from pediatric patients with DCM as the next step in identifying cellular signatures. METHODS We performed single nuclei RNA sequencing with heart tissues from 6 children with DCM with an age of 0.5, 0.75, 5, 6, 12, and 13 years. Unsupervised clustering of 18 211 nuclei led to the identification of 14 distinct clusters with 6 major cell types. RESULTS The number of nuclei in fibroblast clusters increased with age in patients with DCM, a finding that was confirmed by histological analysis and was consistent with an age-related increase in cardiac fibrosis quantified by cardiac magnetic resonance imaging. Fibroblasts of patients with DCM >6 years of age showed a profoundly altered gene expression pattern with enrichment of genes encoding fibrillary collagens, modulation of proteoglycans, switch in thrombospondin isoforms, and signatures of fibroblast activation. In addition, a population of cardiomyocytes with a high proregenerative profile was identified in infant patients with DCM but was absent in children >6 years of age. This cluster showed high expression of cell cycle activators such as cyclin D family members, increased glycolytic metabolism and antioxidative genes, and alterations in ß-adrenergic signaling genes. CONCLUSIONS Novel insights into the cellular transcriptomes of hearts from pediatric patients with DCM provide remarkable age-dependent changes in the expression patterns of fibroblast and cardiomyocyte genes with less fibrotic but enriched proregenerative signatures in infants.
Collapse
Affiliation(s)
- Luka Nicin
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany.,German Center for Cardiovascular Research, Frankfurt, Germany (L.N., W.T.A., S.D.).,Cardio-Pulmonary Institute, Frankfurt, Germany (L.N., W.T.A., S.D.)
| | - Wesley T Abplanalp
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany.,German Center for Cardiovascular Research, Frankfurt, Germany (L.N., W.T.A., S.D.).,Cardio-Pulmonary Institute, Frankfurt, Germany (L.N., W.T.A., S.D.)
| | - Anne Schänzer
- Institute of Neuropathology (A.S., M.K.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Anke Sprengel
- Pediatric Heart Center, Department of Pediatric Cardiac Surgery (A.S., H.A.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - David John
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany
| | - Hannah Mellentin
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany
| | - Lukas Tombor
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany
| | - Matthias Keuper
- Institute of Neuropathology (A.S., M.K.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Evelyn Ullrich
- Experimental Immunology, Division of Pediatric Stem Cell Transplantation and Immunology, Children and Adolescents Medicine, University Hospital Frankfurt (E.U.), Goethe University, Germany.,Frankfurt Cancer Institute (E.U.), Goethe University, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Germany (K.K.)
| | | | - Jedrzej Hoffmann
- Internal Medicine Clinic III, Department of Cardiology (J.H., A.M.Z.), Goethe University, Germany
| | - Hakan Akintuerk
- Pediatric Heart Center, Department of Pediatric Cardiac Surgery (A.S., H.A.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Christian Jux
- Department of Pediatric Cardiology and Congenital Heart Disease (C.J., D.S., S.R.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Dietmar Schranz
- Department of Pediatric Cardiology and Congenital Heart Disease (C.J., D.S., S.R.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Andreas M Zeiher
- Internal Medicine Clinic III, Department of Cardiology (J.H., A.M.Z.), Goethe University, Germany
| | - Stefan Rupp
- Department of Pediatric Cardiology and Congenital Heart Disease (C.J., D.S., S.R.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany.,German Center for Cardiovascular Research, Frankfurt, Germany (L.N., W.T.A., S.D.).,Cardio-Pulmonary Institute, Frankfurt, Germany (L.N., W.T.A., S.D.)
| |
Collapse
|
112
|
Farache Trajano L, Smart N. Immunomodulation for optimal cardiac regeneration: insights from comparative analyses. NPJ Regen Med 2021; 6:8. [PMID: 33589632 PMCID: PMC7884783 DOI: 10.1038/s41536-021-00118-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023] Open
Abstract
Despite decades of research, regeneration of the infarcted human heart remains an unmet ambition. A significant obstacle facing experimental regenerative therapies is the hostile immune response which arises following a myocardial infarction (MI). Upon cardiac damage, sterile inflammation commences via the release of pro-inflammatory meditators, leading to the migration of neutrophils, eosinophils and monocytes, as well as the activation of local vascular cells and fibroblasts. This response is amplified by components of the adaptive immune system. Moreover, the physical trauma of the infarction and immune-mediated tissue injury provides a supply of autoantigens, perpetuating a cycle of autoreactivity, which further contributes to adverse remodelling. A gradual shift towards an immune-resolving environment follows, culminating in the formation of a collagenous scar, which compromises cardiac function, ultimately driving the development of heart failure. Comparing the human heart with those of animal models that are capable of cardiac regeneration reveals key differences in the innate and adaptive immune responses to MI. By modulating key immune components to better resemble those of regenerative species, a cardiac environment may be established which would, either independently or via the synergistic application of emerging regenerative therapies, improve functional recovery post-MI.
Collapse
Affiliation(s)
- Luiza Farache Trajano
- British Heart Foundation Centre of Regenerative Medicine, Burdon Sanderson Cardiac Science centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Nicola Smart
- British Heart Foundation Centre of Regenerative Medicine, Burdon Sanderson Cardiac Science centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
113
|
Lother A, Bondareva O, Saadatmand AR, Pollmeier L, Härdtner C, Hilgendorf I, Weichenhan D, Eckstein V, Plass C, Bode C, Backs J, Hein L, Gilsbach R. Diabetes changes gene expression but not DNA methylation in cardiac cells. J Mol Cell Cardiol 2021; 151:74-87. [PMID: 33197445 DOI: 10.1016/j.yjmcc.2020.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 10/22/2020] [Accepted: 11/08/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Diabetes mellitus is a worldwide epidemic that causes high mortality due to cardiovascular complications, in particular heart failure. Diabetes is associated with profound pathophysiological changes in the heart. The aim of this study was to investigate the impact of diabetes on gene expression and DNA methylation in cardiac cells. METHODS AND RESULTS Transcriptome analysis of heart tissue from mice with streptozotocin-induced diabetes revealed only 39 genes regulated, whereas cell type-specific analysis of the diabetic heart was more sensitive and more specific than heart tissue analysis and revealed a total of 3205 differentially regulated genes in five cell types. Whole genome DNA methylation analysis with basepair resolution of distinct cardiac cell types identified highly specific DNA methylation signatures of genic and regulatory regions. Interestingly, despite marked changes in gene expression, DNA methylation remained stable in streptozotocin-induced diabetes. Integrated analysis of cell type-specific gene expression enabled us to assign the particular contribution of single cell types to the pathophysiology of the diabetic heart. Finally, analysis of gene regulation revealed ligand-receptor pairs as potential mediators of heterocellular interaction in the diabetic heart, with fibroblasts and monocytes showing the highest degree of interaction. CONCLUSION In summary, cell type-specific analysis reveals differentially regulated gene programs that are associated with distinct biological processes in diabetes. Interestingly, despite these changes in gene expression, cell type-specific DNA methylation signatures of genic and regulatory regions remain stable in diabetes. Analysis of heterocellular interactions in the diabetic heart suggest that the interplay between fibroblasts and monocytes is of pivotal importance.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany; Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany
| | - Olga Bondareva
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Ali R Saadatmand
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Luisa Pollmeier
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Carmen Härdtner
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany
| | - Ingo Hilgendorf
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany
| | - Dieter Weichenhan
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Volker Eckstein
- Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph Plass
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Bode
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany
| | - Johannes Backs
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany; Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany; German Centre of Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt am Main, Germany.
| |
Collapse
|
114
|
Valproic acid influences the expression of genes implicated with hyperglycaemia-induced complement and coagulation pathways. Sci Rep 2021; 11:2163. [PMID: 33495488 PMCID: PMC7835211 DOI: 10.1038/s41598-021-81794-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 01/11/2021] [Indexed: 01/04/2023] Open
Abstract
Because the liver plays a major role in metabolic homeostasis and secretion of clotting factors and inflammatory innate immune proteins, there is interest in understanding the mechanisms of hepatic cell activation under hyperglycaemia and whether this can be attenuated pharmacologically. We have previously shown that hyperglycaemia stimulates major changes in chromatin organization and metabolism in hepatocytes, and that the histone deacetylase inhibitor valproic acid (VPA) is able to reverse some of these metabolic changes. In this study, we have used RNA-sequencing (RNA-seq) to investigate how VPA influences gene expression in hepatocytes. Interesting, we observed that VPA attenuates hyperglycaemia-induced activation of complement and coagulation cascade genes. We also observe that many of the gene activation events coincide with changes to histone acetylation at the promoter of these genes indicating that epigenetic regulation is involved in VPA action.
Collapse
|
115
|
Hosseini FS, Amanlou A, Amanlou M. Tankyrase Inhibitor for Cardiac Tissue Regeneration: an In-silico Approach. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:315-328. [PMID: 35194449 PMCID: PMC8842603 DOI: 10.22037/ijpr.2021.115367.15339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Myocardial infarction causes heart tissue damages; therefore, using non-invasive methods to regenerate the heart tissue could be very helpful. Recent studies claimed that the inhibition of the Wnt signaling could promote cardiac remodeling and induce cardiac regeneration. Therefore, a tankyrase inhibitor to stabilize the AXIN and inhibit the Wnt/β-catenin signaling pathway will induce cardiac regeneration after injury. In this regard, virtual screening procedure, using molecular docking of 9127 FDA and world approved drugs, including herbal medicine, was done over the crystal structures of tankyrase 1 (TNKS1) and tankyrase 2 (TNKS2) catalytic poly (ADP-ribose) polymerase (PARP) domains with PDB ID: 2RF5 and 3KR7, respectively, to find potential small molecule inhibitors to regenerate injured heart tissue. Subsequently, molecular dynamics simulations were done to assess the stability of selected ligands phenothrin and ethyl rosinate in the binding pocket of TNKS1 and TNKS2 for 100 ns, respectively. Both compounds show suitable interaction in their binding pocket. The molecular dynamics simulation results confirm their stability. The binding free energy of complexes was carried out by the MM-PBSA method. ADME properties also indicate the potential of drug-likeness of both compounds. Taking together both drugs may be promising for inducing cardiac regeneration after injury. Nevertheless, clinical approval remains.
Collapse
Affiliation(s)
- Faezeh Sadat Hosseini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Arash Amanlou
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran. ,Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Corresponding author: E-mail:
| |
Collapse
|
116
|
Wang Z, Cui M, Shah AM, Tan W, Liu N, Bassel-Duby R, Olson EN. Cell-Type-Specific Gene Regulatory Networks Underlying Murine Neonatal Heart Regeneration at Single-Cell Resolution. Cell Rep 2020; 33:108472. [PMID: 33296652 PMCID: PMC7774872 DOI: 10.1016/j.celrep.2020.108472] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/09/2020] [Accepted: 11/11/2020] [Indexed: 12/22/2022] Open
Abstract
The adult mammalian heart has limited capacity for regeneration following injury, whereas the neonatal heart can readily regenerate within a short period after birth. Neonatal heart regeneration is orchestrated by multiple cell types intrinsic to the heart, as well as immune cells that infiltrate the heart after injury. To elucidate the transcriptional responses of the different cellular components of the mouse heart following injury, we perform single-cell RNA sequencing on neonatal hearts at various time points following myocardial infarction and couple the results with bulk tissue RNA-sequencing data collected at the same time points. Concomitant single-cell ATAC sequencing exposes underlying dynamics of open chromatin landscapes and regenerative gene regulatory networks of diverse cardiac cell types and reveals extracellular mediators of cardiomyocyte proliferation, angiogenesis, and fibroblast activation. Together, our data provide a transcriptional basis for neonatal heart regeneration at single-cell resolution and suggest strategies for enhancing cardiac function after injury.
Collapse
Affiliation(s)
- Zhaoning Wang
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Miao Cui
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Akansha M Shah
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Wei Tan
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ning Liu
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
117
|
Zhuang L, Lu L, Zhang R, Chen K, Yan X. Comprehensive Integration of Single-Cell Transcriptional Profiling Reveals the Heterogeneities of Non-cardiomyocytes in Healthy and Ischemic Hearts. Front Cardiovasc Med 2020; 7:615161. [PMID: 33365332 PMCID: PMC7750309 DOI: 10.3389/fcvm.2020.615161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/18/2020] [Indexed: 12/24/2022] Open
Abstract
Advances in single-cell RNA sequencing (scRNA-seq) technology have recently shed light on the molecular mechanisms of the spatial and temporal changes of thousands of cells simultaneously under homeostatic and ischemic conditions. The aim of this study is to investigate whether it is possible to integrate multiple similar scRNA-seq datasets for a more comprehensive understanding of diseases. In this study, we integrated three representative scRNA-seq datasets of 27,349 non-cardiomyocytes isolated at 3 and 7 days after myocardial infarction or sham surgery. In total, seven lineages, including macrophages, fibroblasts, endothelia, and lymphocytes, were identified in this analysis with distinct dynamic and functional properties in healthy and nonhealthy hearts. Myofibroblasts and endothelia were recognized as the central hubs of cellular communication via ligand-receptor interactions. Additionally, we showed that macrophages from different origins exhibited divergent transcriptional signatures, pathways, developmental trajectories, and transcriptional regulons. It was found that myofibroblasts predominantly expand at 7 days after myocardial infarction with pro-reparative characteristics. We identified signature genes of myofibroblasts, such as Postn, Cthrc1, and Ddah1, among which Ddah1 was exclusively expressed on activated fibroblasts and exhibited concordant upregulation in bulk RNA sequencing data and in vivo and in vitro experiments. Collectively, this compendium of scRNA-seq data provides a valuable entry point for understanding the transcriptional and dynamic changes of non-cardiomyocytes in healthy and nonhealthy hearts by integrating multiple datasets.
Collapse
Affiliation(s)
- Lingfang Zhuang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kang Chen
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxiang Yan
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
118
|
Proximity to injury, but neither number of nuclei nor ploidy define pathological adaptation and plasticity in cardiomyocytes. J Mol Cell Cardiol 2020; 152:95-104. [PMID: 33290769 DOI: 10.1016/j.yjmcc.2020.11.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 12/28/2022]
Abstract
The adult mammalian heart consists of mononuclear and binuclear cardiomyocytes (CMs) with various ploidies. However, it remains unclear whether a variation in ploidy or number of nuclei is associated with distinct functions and injury responses in CMs, including regeneration. Therefore, we investigated transcriptomes and cellular as well as nuclear features of mononucleated and binucleated CMs in adult mouse hearts with and without injury. To be able to identify the role of ploidy we analyzed control and failing human ventricular CMs because human CMs show a larger and disease-sensitive degree of polyploidization. Using transgenic Myh6-H2BmCh to identify mononucleated and binucleated mouse CMs, we found that cellular volume and RNA content were similar in both. On average nuclei of mononuclear CMs showed a 2-fold higher ploidy, as compared to binuclear CMs indicating that most mononuclear CMs are tetraploid. After myocardial infarction mononucleated and binucleated CMs in the border zone of the lesion responded with hypertrophy and corresponding changes in gene expression, as well as a low level of induction of cell cycle gene expression. Human CMs allowed us to study a wide range of polyploidy spanning from 2n to 16n. Notably, basal as well as pathological gene expression signatures and programs in failing CMs proved to be independent of ploidy. In summary, gene expression profiles were induced in proximity to injury, but independent of number of nuclei or ploidy levels in CMs.
Collapse
|
119
|
Quaife-Ryan GA, Mills RJ, Lavers G, Voges HK, Vivien CJ, Elliott DA, Ramialison M, Hudson JE, Porrello ER. β-Catenin drives distinct transcriptional networks in proliferative and nonproliferative cardiomyocytes. Development 2020; 147:dev.193417. [PMID: 33144401 DOI: 10.1242/dev.193417] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/18/2020] [Indexed: 12/22/2022]
Abstract
The inability of the adult mammalian heart to regenerate represents a fundamental barrier in heart failure management. By contrast, the neonatal heart retains a transient regenerative capacity, but the underlying mechanisms for the developmental loss of cardiac regenerative capacity in mammals are not fully understood. Wnt/β-catenin signalling has been proposed as a key cardioregenerative pathway driving cardiomyocyte proliferation. Here, we show that Wnt/β-catenin signalling potentiates neonatal mouse cardiomyocyte proliferation in vivo and immature human pluripotent stem cell-derived cardiomyocyte (hPSC-CM) proliferation in vitro By contrast, Wnt/β-catenin signalling in adult mice is cardioprotective but fails to induce cardiomyocyte proliferation. Transcriptional profiling and chromatin immunoprecipitation sequencing of neonatal mouse and hPSC-CMs revealed a core Wnt/β-catenin-dependent transcriptional network governing cardiomyocyte proliferation. By contrast, β-catenin failed to re-engage this neonatal proliferative gene network in the adult heart despite partial transcriptional re-activation of a neonatal glycolytic gene programme. These findings suggest that β-catenin might be repurposed from regenerative to protective functions in the adult heart in a developmental process dependent on the metabolic status of cardiomyocytes.
Collapse
Affiliation(s)
- Gregory A Quaife-Ryan
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4006, Australia.,School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Richard J Mills
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4006, Australia
| | - George Lavers
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4006, Australia
| | - Holly K Voges
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Celine J Vivien
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - David A Elliott
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia.,Department of Paediatrics, The Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - James E Hudson
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4006, Australia
| | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia .,Department of Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
120
|
Zhang DH, Zhang JL, Huang Z, Wu LM, Wang ZM, Li YP, Tian XY, Kong LY, Yao R, Zhang YZ. Deubiquitinase Ubiquitin-Specific Protease 10 Deficiency Regulates Sirt6 signaling and Exacerbates Cardiac Hypertrophy. J Am Heart Assoc 2020; 9:e017751. [PMID: 33170082 PMCID: PMC7763723 DOI: 10.1161/jaha.120.017751] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Cardiac hypertrophy (CH) is a physiological response that compensates for blood pressure overload. Under pathological conditions, hypertrophy can progress to heart failure as a consequence of the disorganized growth of cardiomyocytes and cardiac tissue. USP10 (ubiquitin‐specific protease 10) is a member of the ubiquitin‐specific protease family of cysteine proteases, which are involved in viral infection, oxidative stress, lipid drop formation, and heat shock. However, the role of USP10 in CH remains largely unclear. Here, we investigated the roles of USP10 in CH. Methods and Results Cardiac‐specific USP10 knockout (USP10‐CKO) mice and USP10‐transgenic (USP10‐TG) mice were used to examined the role of USP10 in CH following aortic banding. The specific functions of USP10 were further examined in isolated cardiomyocytes. USP10 expression was increased in murine hypertrophic hearts following aortic banding and in isolated cardiomyocytes in response to hypertrophic agonist. Mice deficient in USP10 in the heart exhibited exaggerated cardiac hypertrophy and fibrosis following pressure overload stress, which resulted in worsening of cardiac contractile function. In contrast, cardiac overexpression of USP10 protected against pressure overload‐induced maladaptive CH. Mechanistically, we demonstrated that USP10 activation and interaction with Sirt6 in response to angiotensin II led to a marked increase in the ubiquitination of Sirt6 and resulted in Akt signaling downregulation and attenuation of cardiomyocyte hypertrophy. Accordingly, inactivation of USP10 reduced Sirt6 abundance and stability and diminished Sirt6‐induced downstream signaling in cardiomyocytes. Conclusions USP10 functions as a Sirt6 deubiquitinase that induces cardiac myocyte hypertrophy and triggers maladaptive CH.
Collapse
Affiliation(s)
- Dian-Hong Zhang
- Cardiovascular Hospital the First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Jie-Lei Zhang
- Department of Endocrinology the First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Zhen Huang
- Cardiovascular Hospital the First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Lei-Ming Wu
- Cardiovascular Hospital the First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Zhong-Min Wang
- Department of Cardiology FuWai Central China Cardiovascular Hospital Zhengzhou China
| | - Ya-Peng Li
- Cardiovascular Hospital the First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Xin-Yu Tian
- Cardiovascular Hospital the First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Ling-Yao Kong
- Cardiovascular Hospital the First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Rui Yao
- Cardiovascular Hospital the First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Yan-Zhou Zhang
- Cardiovascular Hospital the First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| |
Collapse
|
121
|
Li H, Liu C, Bao M, Liu W, Nie Y, Lian H, Hu S. Optimized Langendorff perfusion system for cardiomyocyte isolation in adult mouse heart. J Cell Mol Med 2020; 24:14619-14625. [PMID: 33147379 PMCID: PMC7754046 DOI: 10.1111/jcmm.15773] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/28/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022] Open
Abstract
With the rapid development of single‐cell sequencing technology, the Langendorff perfusion system has emerged as a common approach to decompose cardiac tissue and obtain living cardiomyocytes to study cardiovascular disease with the mechanism of cardiomyocyte biology. However, the traditional Langendorff perfusion system is difficult to master, and further, the viability and purity of cardiomyocytes are frequently unable to meet sequencing requirements due to complicated devices and manipulate processes. Here, we provide an optimized Langendorff perfusion system with a simplified and standardized operating protocol which utilizes gravity as the perfusion pressure, includes a novel method for bubbles removing and standardizes the criteria for termination of digestion. We obtained stable cardiomyocyte with high viability and purity after multiple natural gravity sedimentation. The combination of the optimized Langendorff perfusion system and the multiple natural gravity sedimentation provides a stable system for isolating adult mouse heart, which will provide higher‐quality cardiomyocytes for further experiments.
Collapse
Affiliation(s)
- Haotong Li
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China
| | - Chungeng Liu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Minghui Bao
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Weijing Liu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China
| | - Hong Lian
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China
| |
Collapse
|
122
|
Waylen LN, Nim HT, Martelotto LG, Ramialison M. From whole-mount to single-cell spatial assessment of gene expression in 3D. Commun Biol 2020; 3:602. [PMID: 33097816 PMCID: PMC7584572 DOI: 10.1038/s42003-020-01341-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 09/10/2020] [Indexed: 12/31/2022] Open
Abstract
Unravelling spatio-temporal patterns of gene expression is crucial to understanding core biological principles from embryogenesis to disease. Here we review emerging technologies, providing automated, high-throughput, spatially resolved quantitative gene expression data. Novel techniques expand on current benchmark protocols, expediting their incorporation into ongoing research. These approaches digitally reconstruct patterns of embryonic expression in three dimensions, and have successfully identified novel domains of expression, cell types, and tissue features. Such technologies pave the way for unbiased and exhaustive recapitulation of gene expression levels in spatial and quantitative terms, promoting understanding of the molecular origin of developmental defects, and improving medical diagnostics.
Collapse
Affiliation(s)
- Lisa N Waylen
- Australian Regenerative Medicine Institute and Systems Biology Institute, Monash University, Clayton, VIC, Australia
| | - Hieu T Nim
- Australian Regenerative Medicine Institute and Systems Biology Institute, Monash University, Clayton, VIC, Australia
- Transcriptomics and Bioinformatics Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Luciano G Martelotto
- Single Cell Core Laboratory, Harvard Medical School, Department of System Biology, Boston, MA, USA
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute and Systems Biology Institute, Monash University, Clayton, VIC, Australia.
- Transcriptomics and Bioinformatics Group, Murdoch Children's Research Institute, Parkville, VIC, Australia.
| |
Collapse
|
123
|
Mills RJ, Hudson JE. There's No I in Team: Cellular Crosstalk Enhances In Vitro Cardiac Maturation. Cell Stem Cell 2020; 26:799-801. [PMID: 32502399 DOI: 10.1016/j.stem.2020.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Organoids and microtissues offer the unique opportunity to dissect cell-cell interactions in an organ-specific context without confounding effects of organ failure or organism viability. In a study in this issue of Cell Stem Cell, Giacomelli et al. (2020) add human cardiac fibroblasts into cardiac microtissues and reveal striking fibroblast-endothelial-cardiomyocyte crosstalk that promotes advanced cardiomyocyte maturation.
Collapse
Affiliation(s)
- Richard J Mills
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia
| | - James E Hudson
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia.
| |
Collapse
|
124
|
Sreejit G, Abdel Latif A, Murphy AJ, Nagareddy PR. Emerging roles of neutrophil-borne S100A8/A9 in cardiovascular inflammation. Pharmacol Res 2020; 161:105212. [PMID: 32991974 DOI: 10.1016/j.phrs.2020.105212] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Elevated neutrophil count is associated with higher risk of major adverse cardiac events including myocardial infarction and early development of heart failure. Neutrophils contribute to cardiac damage through a number of mechanisms, including attraction of other immune cells and release of inflammatory mediators. Recently, a number of independent studies have reported a causal role for neutrophil-derived alarmins (i.e. S100A8/A9) in inducing inflammation and cardiac injury following myocardial infarction (MI). Furthermore, a positive correlation between serum S100A8/A9 levels and major adverse cardiac events (MACE) in MI patients was also observed implying that targeting neutrophils or their inflammatory cargo could be beneficial in reducing heart failure. However, contradictory to this idea, neutrophils and neutrophil-derived S100A8/A9 also seem to play a vital role in the resolution of inflammation. Thus, a better understanding of how neutrophils balance these seemingly contrasting functions would allow us to develop effective therapies that preserve the inflammation-resolving function while restricting the damage caused by inflammation. In this review, we specifically discuss the mechanisms behind neutrophil-derived S100A8/A9 in promoting inflammation and resolution in the context of MI. We also provide a perspective on how neutrophils could be potentially targeted to ameliorate cardiac inflammation and the ensuing damage.
Collapse
Affiliation(s)
- Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ahmed Abdel Latif
- Division of Cardiovascular Medicine, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia
| | - Prabhakara R Nagareddy
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
125
|
Nagareddy PR, Sreejit G, Abo-Aly M, Jaggers RM, Chelvarajan L, Johnson J, Pernes G, Athmanathan B, Abdel-Latif A, Murphy AJ. NETosis Is Required for S100A8/A9-Induced Granulopoiesis After Myocardial Infarction. Arterioscler Thromb Vasc Biol 2020; 40:2805-2807. [PMID: 32878477 DOI: 10.1161/atvbaha.120.314807] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Prabhakara R Nagareddy
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus (P.R.N., G.S., R.M.J., J.J., B.A.)
| | - Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus (P.R.N., G.S., R.M.J., J.J., B.A.)
| | - Mohamed Abo-Aly
- Division of Cardiovascular Medicine, Department of Medicine, University of Kentucky, Lexington (M.A.-A., L.C., A.A.-L.)
| | - Robert M Jaggers
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus (P.R.N., G.S., R.M.J., J.J., B.A.)
| | - Lakshman Chelvarajan
- Division of Cardiovascular Medicine, Department of Medicine, University of Kentucky, Lexington (M.A.-A., L.C., A.A.-L.)
| | - Jillian Johnson
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus (P.R.N., G.S., R.M.J., J.J., B.A.)
| | - Gerard Pernes
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Australia (G.P., A.J.M.)
| | - Baskaran Athmanathan
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus (P.R.N., G.S., R.M.J., J.J., B.A.)
| | - Ahmed Abdel-Latif
- Division of Cardiovascular Medicine, Department of Medicine, University of Kentucky, Lexington (M.A.-A., L.C., A.A.-L.)
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Australia (G.P., A.J.M.)
| |
Collapse
|
126
|
Abstract
Regeneration is the process by which organisms replace lost or damaged tissue, and regenerative capacity can vary greatly among species, tissues and life stages. Tissue regeneration shares certain hallmarks of embryonic development, in that lineage-specific factors can be repurposed upon injury to initiate morphogenesis; however, many differences exist between regeneration and embryogenesis. Recent studies of regenerating tissues in laboratory model organisms - such as acoel worms, frogs, fish and mice - have revealed that chromatin structure, dedicated enhancers and transcriptional networks are regulated in a context-specific manner to control key gene expression programmes. A deeper mechanistic understanding of the gene regulatory networks of regeneration pathways might ultimately enable their targeted reactivation as a means to treat human injuries and degenerative diseases. In this Review, we consider the regeneration of body parts across a range of tissues and species to explore common themes and potentially exploitable elements.
Collapse
Affiliation(s)
- Joseph A Goldman
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, OH, USA.
| | - Kenneth D Poss
- Regeneration Next, Duke University, Durham, NC, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
127
|
Zhang S, Chen R, Chakrabarti S, Su Z. Resident macrophages as potential therapeutic targets for cardiac ageing and injury. Clin Transl Immunology 2020; 9:e1167. [PMID: 32874584 PMCID: PMC7450172 DOI: 10.1002/cti2.1167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/21/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac‐resident macrophages (CRMs) play critical roles in maintaining cardiac homoeostasis and removing senescent and dying cells. Recent preclinical data have re‐energised the area of cardioimmunology and provided improved understanding of the modulation of compositional and functional phenotypes of CRMs. These data can aid in achieving improved cardiac regeneration, repair and functional remodelling following cardiac injury. In this review, we discuss the composition and renewal of various subsets of CRMs. Specific attention has been given to delineate the roles of various CRM subsets with respect to (1) facilitation of cardiac development and maintenance of physiological function such as electrical conduction and rhythm; (2) promotion of cardiac regeneration, inflammation resolution and functional remodelling following a cardiac injury; and (3) therapeutic potential. We have also highlighted the relationship between CRM replenishment and cardiomyocyte senescence as well as cardiovascular diseases development. Finally, we have addressed future perspectives and directions in basic research and potentially clinical applications of CRMs.
Collapse
Affiliation(s)
- Shiqing Zhang
- International Genome Center Jiangsu University Zhenjiang China.,Department of Immunology Jiangsu University Zhenjiang China
| | - Rong Chen
- International Genome Center Jiangsu University Zhenjiang China.,Department of Immunology Jiangsu University Zhenjiang China
| | | | - Zhaoliang Su
- International Genome Center Jiangsu University Zhenjiang China.,Department of Immunology Jiangsu University Zhenjiang China.,Laboratory Center The Fourth Affiliated Hospital of Jiangsu University Zhenjiang China
| |
Collapse
|
128
|
Sanz-Morejón A, García-Redondo AB, Reuter H, Marques IJ, Bates T, Galardi-Castilla M, Große A, Manig S, Langa X, Ernst A, Piragyte I, Botos MA, González-Rosa JM, Ruiz-Ortega M, Briones AM, Salaices M, Englert C, Mercader N. Wilms Tumor 1b Expression Defines a Pro-regenerative Macrophage Subtype and Is Required for Organ Regeneration in the Zebrafish. Cell Rep 2020; 28:1296-1306.e6. [PMID: 31365871 PMCID: PMC6685527 DOI: 10.1016/j.celrep.2019.06.091] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/25/2019] [Accepted: 06/25/2019] [Indexed: 12/16/2022] Open
Abstract
Organ regeneration is preceded by the recruitment of innate immune cells, which play an active role during repair and regrowth. Here, we studied macrophage subtypes during organ regeneration in the zebrafish, an animal model with a high regenerative capacity. We identified a macrophage subpopulation expressing Wilms tumor 1b (wt1b), which accumulates within regenerating tissues. This wt1b+ macrophage population exhibited an overall pro-regenerative gene expression profile and different migratory behavior compared to the remainder of the macrophages. Functional studies showed that wt1b regulates macrophage migration and retention at the injury area. Furthermore, wt1b-null mutant zebrafish presented signs of impaired macrophage differentiation, delayed fin growth upon caudal fin amputation, and reduced cardiomyocyte proliferation following cardiac injury that correlated with altered macrophage recruitment to the regenerating areas. We describe a pro-regenerative macrophage subtype in the zebrafish and a role for wt1b in organ regeneration. Wt1b+ macrophages reveal a pro-regenerative gene expression prolife Wt1b controls migration behavior of macrophages during fin and heart regeneration Wt1b regulates differentiation of macrophages in the kidney marrow wt1b mutants reveal impaired fin and heart regeneration
Collapse
Affiliation(s)
- Andrés Sanz-Morejón
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Ana B García-Redondo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; Department of Pharmacology, Universidad Autónoma de Madrid, IIS-Hospital La Paz, Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Hanna Reuter
- Leibniz Institute on Aging-Fritz Lipmann Institute, 07745 Jena, Germany
| | - Inês J Marques
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | - Thomas Bates
- Leibniz Institute on Aging-Fritz Lipmann Institute, 07745 Jena, Germany
| | | | - Andreas Große
- Leibniz Institute on Aging-Fritz Lipmann Institute, 07745 Jena, Germany
| | - Steffi Manig
- Leibniz Institute on Aging-Fritz Lipmann Institute, 07745 Jena, Germany
| | - Xavier Langa
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | - Alexander Ernst
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | - Indre Piragyte
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | | | | | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma, 28040 Madrid, Spain
| | - Ana M Briones
- Department of Pharmacology, Universidad Autónoma de Madrid, IIS-Hospital La Paz, Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Mercedes Salaices
- Department of Pharmacology, Universidad Autónoma de Madrid, IIS-Hospital La Paz, Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Christoph Englert
- Leibniz Institute on Aging-Fritz Lipmann Institute, 07745 Jena, Germany; Institute of Biochemistry and Biophysics, Friedrich-Schiller-Universität, 07743 Jena, Germany
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.
| |
Collapse
|
129
|
Song J, Kim YK. Discovery and Functional Prediction of Long Non-Coding RNAs Common to Ischemic Stroke and Myocardial Infarction. J Lipid Atheroscler 2020; 9:449-459. [PMID: 33024736 PMCID: PMC7521976 DOI: 10.12997/jla.2020.9.3.449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/21/2020] [Accepted: 08/02/2020] [Indexed: 12/26/2022] Open
Abstract
Objective Ischemic stroke and myocardial infarction are 2 of the leading causes of mortality. Both conditions are caused by arterial occlusion, resulting in ischemic necrosis of the cells in the cortex and heart. Long non-coding RNAs (lncRNAs) are a group of non-coding RNAs longer than 200 nucleotides without protein-coding potential. Thousands of lncRNAs have been identified but their involvement in ischemic stroke and myocardial infarction has not been studied extensively. Therefore, this study aimed to identify the role of lncRNAs, particularly those that are commonly altered in these two ischemic injuries. Methods We combined diverse RNA sequencing data obtained from public databases and performed extensive bioinformatics analyses to determine reliable lncRNAs commonly identified from these datasets. Using sequence analysis, we also detected the lncRNAs that may act as microRNA (miRNA) regulators. Results We found several altered lncRNAs that were common in ischemic stroke and myocardial infarction models. Some of these lncRNAs, including zinc finger NFX1-type containing 1 antisense RNA 1 and small nucleolar RNA host gene 1, were previously reported to be involved in the pathogenesis of each of these models. Interestingly, several lncRNAs had binding sites for miRNAs that were previously reported to be involved in the hypoxic response, suggesting the possible role of these lncRNAs as regulators in ischemic responses. Conclusion The lncRNAs identified in this study will be useful in determining the regulatory networks in ischemic stroke and myocardial infarction and in identifying potential specific markers for each of these ischemic diseases.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Korea
| | - Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
130
|
Patrick R, Humphreys DT, Janbandhu V, Oshlack A, Ho JW, Harvey RP, Lo KK. Sierra: discovery of differential transcript usage from polyA-captured single-cell RNA-seq data. Genome Biol 2020; 21:167. [PMID: 32641141 PMCID: PMC7341584 DOI: 10.1186/s13059-020-02071-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
High-throughput single-cell RNA-seq (scRNA-seq) is a powerful tool for studying gene expression in single cells. Most current scRNA-seq bioinformatics tools focus on analysing overall expression levels, largely ignoring alternative mRNA isoform expression. We present a computational pipeline, Sierra, that readily detects differential transcript usage from data generated by commonly used polyA-captured scRNA-seq technology. We validate Sierra by comparing cardiac scRNA-seq cell types to bulk RNA-seq of matched populations, finding significant overlap in differential transcripts. Sierra detects differential transcript usage across human peripheral blood mononuclear cells and the Tabula Muris, and 3 'UTR shortening in cardiac fibroblasts. Sierra is available at https://github.com/VCCRI/Sierra .
Collapse
Affiliation(s)
- Ralph Patrick
- Victor Chang Cardiac Research Institute, 405 Liverpool St., Darlinghurst, 2010 Australia
- St. Vincent’s Clinical School, UNSW Sydney, Kensington, 2052 Australia
| | - David T. Humphreys
- Victor Chang Cardiac Research Institute, 405 Liverpool St., Darlinghurst, 2010 Australia
- St. Vincent’s Clinical School, UNSW Sydney, Kensington, 2052 Australia
| | - Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, 405 Liverpool St., Darlinghurst, 2010 Australia
- St. Vincent’s Clinical School, UNSW Sydney, Kensington, 2052 Australia
| | - Alicia Oshlack
- Murdoch Children’s Research Institute, Parkville, 3052 Victoria Australia
- Peter MacCallum Cancer Centre, Research Division, 305 Grattan Street, Melbourne, 3000 Victoria Australia
| | - Joshua W.K. Ho
- Victor Chang Cardiac Research Institute, 405 Liverpool St., Darlinghurst, 2010 Australia
- St. Vincent’s Clinical School, UNSW Sydney, Kensington, 2052 Australia
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, 405 Liverpool St., Darlinghurst, 2010 Australia
- St. Vincent’s Clinical School, UNSW Sydney, Kensington, 2052 Australia
- School of Biotechnology and Biomolecular Science, UNSW Sydney, Kensington, 2052 Australia
| | - Kitty K. Lo
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Camperdown, 2006 Australia
| |
Collapse
|
131
|
Abstract
PURPOSE OF REVIEW This review summarizes the important role that metabolism plays in driving maturation of human pluripotent stem cell-derived cardiomyocytes. RECENT FINDINGS Human pluripotent stem cell-derived cardiomyocytes provide a model system for human cardiac biology. However, these models have been unable to fully recapitulate the maturity observed in the adult heart. By simulating the glucose to fatty acid transition observed in neonatal mammals, human pluripotent stem cell-derived cardiomyocytes undergo structural and functional maturation also accompanied by transcriptional changes and cell cycle arrest. The role of metabolism in energy production, signaling, and epigenetic modifications illustrates that metabolism and cellular phenotype are intimately linked. Further understanding of key metabolic factors driving cardiac maturation will facilitate the generation of more mature human pluripotent stem cell-derived cardiomyocyte models. This will increase our understanding of cardiac biology and potentially lead to novel therapeutics to enhance heart function.
Collapse
|
132
|
Lock MC, Tellam RL, Darby JRT, Soo JY, Brooks DA, Seed M, Selvanayagam JB, Morrison JL. Identification of Novel miRNAs Involved in Cardiac Repair Following Infarction in Fetal and Adolescent Sheep Hearts. Front Physiol 2020; 11:614. [PMID: 32587529 PMCID: PMC7298149 DOI: 10.3389/fphys.2020.00614] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/15/2020] [Indexed: 01/14/2023] Open
Abstract
Aims Animal models have been used to show that there are critical molecular mechanisms that can be activated to induce myocardial repair at specific times in development. For example, specific miRNAs are critical for regulating the response to myocardial infarction (MI) and improving the response to injury. Manipulating these miRNAs in small animal models provides beneficial effects post-MI; however it is not known if these miRNAs are regulated similarly in large mammals. Studying a large animal where the timing of heart development in relation to birth is similar to humans may provide insights to better understand the capacity to repair a developing mammalian heart and its application to the adult heart. Methods We used a sheep model of MI that included permanent ligation of the left anterior descending (LAD) coronary artery. Surgery was performed on fetuses (at 105 days gestation when all cardiomyocytes are mononucleated and proliferative) and adolescent sheep (at 6 months of age when all cardiomyocytes contribute to heart growth by hypertrophy). A microarray was utilized to determine the expression of known miRNAs within the damaged and undamaged tissue regions in fetal and adolescent hearts after MI. Results 73 miRNAs were up-regulated and 58 miRNAs were down-regulated significantly within the fetal infarct compared to remote cardiac samples. From adolescent hearts 69 non-redundant miRNAs were up-regulated and 63 miRNAs were down-regulated significantly in the infarct area compared to remote samples. Opposite differential expression profiles of 10 miRNAs within tissue regions (Infarct area, Border zone and Remote area of the left ventricle) occurred between the fetuses and adolescent sheep. These included miR-558 and miR-1538, which when suppressed using LNA anti-miRNAs in cell culture, increased cardiomyoblast proliferation. Conclusion There were significant differences in miRNA responses in fetal and adolescent sheep hearts following a MI, suggesting that the modulation of novel miRNA expression may have therapeutic potential, by promoting proliferation or repair in a damaged heart.
Collapse
Affiliation(s)
- Mitchell C Lock
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia
| | - Ross L Tellam
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia
| | - Doug A Brooks
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia.,Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Mike Seed
- Division of Cardiology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Joseph B Selvanayagam
- Cardiac Imaging Research, Department of Heart Health, South Australian Health & Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
133
|
Cui M, Olson EN. Protocol for Single-Nucleus Transcriptomics of Diploid and Tetraploid Cardiomyocytes in Murine Hearts. STAR Protoc 2020; 1:100049. [PMID: 33111095 PMCID: PMC7580205 DOI: 10.1016/j.xpro.2020.100049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Murine cardiomyocytes undergo proliferation, multinucleation, and polyploidization during the first 3 weeks of postnatal life, resulting in a mixture of diploid and tetraploid cardiomyocytes in the heart. Understanding the molecular differences between diploid and tetraploid cardiomyocytes from these processes has been limited due to lack of unique markers and their heterogenous origins. Here, we apply single-nucleus RNA-sequencing to fluorescence-activated cell sorting-selected diploid and tetraploid cardiomyocytes to characterize their heterogeneity and molecular distinctions. For complete details on the use and execution of this protocol, please refer to Cui et al. (2020) PCM1 immunoselection isolates cardiomyocyte (CM) nuclei from murine hearts Protocol profiles transcriptome of 2n and 4n cardiomyocytes at single-nucleus resolution Integrating samples at various conditions identifies differential CM populations
Collapse
Affiliation(s)
- Miao Cui
- Department of Molecular Biology, the Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9148, USA
| | - Eric N Olson
- Department of Molecular Biology, the Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9148, USA
| |
Collapse
|
134
|
Ye L, Wang S, Xiao Y, Jiang C, Huang Y, Chen H, Zhang H, Zhang H, Liu J, Xu Z, Hong H. Pressure Overload Greatly Promotes Neonatal Right Ventricular Cardiomyocyte Proliferation: A New Model for the Study of Heart Regeneration. J Am Heart Assoc 2020; 9:e015574. [PMID: 32475201 PMCID: PMC7429015 DOI: 10.1161/jaha.119.015574] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Current mammalian models for heart regeneration research are limited to neonatal apex amputation and myocardial infarction, both of which are controversial. RNAseq has demonstrated a very limited set of differentially expressed genes between sham and operated hearts in myocardial infarction models. Here, we investigated in rats whether pressure overload in the right ventricle, a common phenomenon in children with congenital heart disease, could be used as a better animal model for heart regeneration studies when considering cardiomyocyte proliferation as the most important index. Methods and Results In the rat model, pressure overload was induced by pulmonary artery banding on postnatal day 1 and confirmed by echocardiography and hemodynamic measurements at postnatal day 7. RNA sequencing analyses of purified right ventricular cardiomyocytes at postnatal day 7 from pulmonary artery banding and sham-operated rats revealed that there were 5469 differentially expressed genes between these 2 groups. Gene ontology and Kyoto Encyclopedia of Genes and Genomes analysis showed that these genes mainly mediated mitosis and cell division. Cell proliferation assays indicated a continuous overproliferation of cardiomyocytes in the right ventricle after pulmonary artery banding, in particular for the first 3 postnatal days. We also validated the model using samples from overloaded right ventricles of human patients. There was an approximately 2-fold increase of Ki67/pHH3/aurora B-positive cardiomyocytes in human-overloaded right ventricles compared with nonoverloaded right ventricles. Other features of this animal model included cardiomyocyte hypotrophy with no fibrosis. Conclusions Pressure overload profoundly promotes cardiomyocyte proliferation in the neonatal stage in both rats and human beings. This activates a regeneration-specific gene program and may offer an alternative animal model for heart regeneration research.
Collapse
Affiliation(s)
- Lincai Ye
- Department of Thoracic and Cardiovascular Surgery Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China.,Institute of Pediatric Translational Medicine Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China.,Shanghai Institute for Pediatric Congenital Heart Disease Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China
| | - Shoubao Wang
- Department of Thoracic and Cardiovascular Surgery Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China.,Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital Shanghai Jiaotong University School of Medicine Shanghai China
| | - Yingying Xiao
- Department of Thoracic and Cardiovascular Surgery Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China
| | - Chuan Jiang
- Department of Thoracic and Cardiovascular Surgery Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China.,Institute of Pediatric Translational Medicine Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China.,Shanghai Institute for Pediatric Congenital Heart Disease Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China
| | - Yanhui Huang
- Department of Anesthesiology Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China
| | - Huiwen Chen
- Department of Thoracic and Cardiovascular Surgery Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China
| | - Haibo Zhang
- Department of Thoracic and Cardiovascular Surgery Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China
| | - Hao Zhang
- Department of Thoracic and Cardiovascular Surgery Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China.,Shanghai Institute for Pediatric Congenital Heart Disease Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China
| | - Jinfen Liu
- Department of Thoracic and Cardiovascular Surgery Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China
| | - Zhuoming Xu
- Department of Thoracic and Cardiovascular Surgery Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China
| | - Haifa Hong
- Institute of Pediatric Translational Medicine Shanghai Children's Medical Center Shanghai Jiaotong University School of Medicine Shanghai China
| |
Collapse
|
135
|
Gründl M, Walz S, Hauf L, Schwab M, Werner KM, Spahr S, Schulte C, Maric HM, Ade CP, Gaubatz S. Interaction of YAP with the Myb-MuvB (MMB) complex defines a transcriptional program to promote the proliferation of cardiomyocytes. PLoS Genet 2020; 16:e1008818. [PMID: 32469866 PMCID: PMC7286521 DOI: 10.1371/journal.pgen.1008818] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/10/2020] [Accepted: 05/01/2020] [Indexed: 01/14/2023] Open
Abstract
The Hippo signalling pathway and its central effector YAP regulate proliferation of cardiomyocytes and growth of the heart. Using genetic models in mice we show that the increased proliferation of embryonal and postnatal cardiomyocytes due to loss of the Hippo-signaling component SAV1 depends on the Myb-MuvB (MMB) complex. Similarly, proliferation of postnatal cardiomyocytes induced by constitutive active YAP requires MMB. Genome studies revealed that YAP and MMB regulate an overlapping set of cell cycle genes in cardiomyocytes. Protein-protein interaction studies in cell lines and with recombinant proteins showed that YAP binds directly to B-MYB, a subunit of MMB, in a manner dependent on the YAP WW domains and a PPXY motif in B-MYB. Disruption of the interaction by overexpression of the YAP binding domain of B-MYB strongly inhibits the proliferation of cardiomyocytes. Our results point to MMB as a critical downstream effector of YAP in the control of cardiomyocyte proliferation. YAP, the major downstream transducer of the Hippo pathway, is a potent inducer of proliferation. Here we show that the Myb-MuvB complex (MMB) mediates cardiomyocyte proliferation by YAP. We find that YAP and MMB regulate an overlapping set of pro-proliferative genes which involves binding of MMB to the promoters of these genes. We also identified a direct interaction between the B-MYB subunit of MMB and YAP. Based on the binding studies, we created a tool called MY-COMP that interferes with the association YAP to B-MYB and strongly inhibits proliferation of cardiomyocytes. Together, our data suggests that the YAP-MMB interaction is essential for division of cardiomyocytes, underscoring the functional relevance of the crosstalk between these two pathways for proper heart development.
Collapse
Affiliation(s)
- Marco Gründl
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Susanne Walz
- Comprehensive Cancer Center Mainfranken, Core Unit Bioinformatics, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| | - Laura Hauf
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Melissa Schwab
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Kerstin Marcela Werner
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Susanne Spahr
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Clemens Schulte
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Wuerzburg, Germany
| | - Hans Michael Maric
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Wuerzburg, Germany
| | - Carsten P. Ade
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Stefan Gaubatz
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| |
Collapse
|
136
|
Bywater MJ, Burkhart DL, Straube J, Sabò A, Pendino V, Hudson JE, Quaife-Ryan GA, Porrello ER, Rae J, Parton RG, Kress TR, Amati B, Littlewood TD, Evan GI, Wilson CH. Reactivation of Myc transcription in the mouse heart unlocks its proliferative capacity. Nat Commun 2020; 11:1827. [PMID: 32286286 PMCID: PMC7156407 DOI: 10.1038/s41467-020-15552-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 03/19/2020] [Indexed: 12/14/2022] Open
Abstract
It is unclear why some tissues are refractory to the mitogenic effects of the oncogene Myc. Here we show that Myc activation induces rapid transcriptional responses followed by proliferation in some, but not all, organs. Despite such disparities in proliferative response, Myc is bound to DNA at open elements in responsive (liver) and non-responsive (heart) tissues, but fails to induce a robust transcriptional and proliferative response in the heart. Using heart as an exemplar of a non-responsive tissue, we show that Myc-driven transcription is re-engaged in mature cardiomyocytes by elevating levels of the positive transcription elongation factor (P-TEFb), instating a large proliferative response. Hence, P-TEFb activity is a key limiting determinant of whether the heart is permissive for Myc transcriptional activation. These data provide a greater understanding of how Myc transcriptional activity is determined and indicate modification of P-TEFb levels could be utilised to drive regeneration of adult cardiomyocytes for the treatment of heart myopathies.
Collapse
Affiliation(s)
- Megan J Bywater
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Deborah L Burkhart
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Jasmin Straube
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Arianna Sabò
- Department of Experimental Oncology, European Institute of Oncology (IEO) - IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Vera Pendino
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Via Adamello 16, 20139, Milan, Italy
| | - James E Hudson
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | | | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, 3052, Australia
- Department of Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - James Rae
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, 4072, QLD, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, 4072, QLD, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, St Lucia, 4072, QLD, Australia
| | - Theresia R Kress
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Via Adamello 16, 20139, Milan, Italy
| | - Bruno Amati
- Department of Experimental Oncology, European Institute of Oncology (IEO) - IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Trevor D Littlewood
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Gerard I Evan
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK.
| | - Catherine H Wilson
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK.
- Department of Pharmacology, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1PD, UK.
| |
Collapse
|
137
|
Abstract
Maturation is the last phase of heart development that prepares the organ for strong, efficient, and persistent pumping throughout the mammal's lifespan. This process is characterized by structural, gene expression, metabolic, and functional specializations in cardiomyocytes as the heart transits from fetal to adult states. Cardiomyocyte maturation gained increased attention recently due to the maturation defects in pluripotent stem cell-derived cardiomyocyte, its antagonistic effect on myocardial regeneration, and its potential contribution to cardiac disease. Here, we review the major hallmarks of ventricular cardiomyocyte maturation and summarize key regulatory mechanisms that promote and coordinate these cellular events. With advances in the technical platforms used for cardiomyocyte maturation research, we expect significant progress in the future that will deepen our understanding of this process and lead to better maturation of pluripotent stem cell-derived cardiomyocyte and novel therapeutic strategies for heart disease.
Collapse
Affiliation(s)
- Yuxuan Guo
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - William Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
138
|
Cardiac regeneration as an environmental adaptation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118623. [DOI: 10.1016/j.bbamcr.2019.118623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/02/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022]
|
139
|
Tarkin JM, Calcagno C, Dweck MR, Evans NR, Chowdhury MM, Gopalan D, Newby DE, Fayad ZA, Bennett MR, Rudd JH. 68Ga-DOTATATE PET Identifies Residual Myocardial Inflammation and Bone Marrow Activation After Myocardial Infarction. J Am Coll Cardiol 2020; 73:2489-2491. [PMID: 31097170 PMCID: PMC6525109 DOI: 10.1016/j.jacc.2019.02.052] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | - James H.F. Rudd
- Division of Cardiovascular Medicine, University of Cambridge, Level 6, Box 110, ACCI, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom @jhfrudd
| |
Collapse
|
140
|
Flinn MA, Jeffery BE, O'Meara CC, Link BA. Yap is required for scar formation but not myocyte proliferation during heart regeneration in zebrafish. Cardiovasc Res 2020; 115:570-577. [PMID: 30295714 DOI: 10.1093/cvr/cvy243] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/26/2018] [Accepted: 10/04/2018] [Indexed: 12/17/2022] Open
Abstract
AIMS The Hippo signalling pathway regulates multiple cellular processes during organ development and maintenance by modulating activity of the transcriptional cofactor Yap. Core components of this pathway are required for neonatal mouse heart regeneration, however, investigations to date have typically focused on expression and activity in cardiomyocytes. Due to the regenerative capacity of zebrafish and the fact that global loss of Yap is not fully embryonic lethal in zebrafish, we leveraged a yap null mutant to investigate the impact of constitutive Yap deletion during zebrafish heart regeneration. METHODS AND RESULTS Following cryoinjury in adult hearts, myocyte proliferation was not decreased in yap mutants, contrary to expectations based on mouse data. Experiments in larval zebrafish (Danio rerio) revealed that deletion of either Yap or Taz had a modest effect on heart growth, reducing gross organ size, while their combined deletion was synergistic; thus, Yap and Taz share some overlapping roles in zebrafish heart development. Surprisingly, adult yap mutants exhibited decreased collagen composition at 7 days post-injury, suggesting a critical role for Yap in scar formation during heart regeneration. siRNA-mediated Yap knockdown in primary rat (Rattus norvegicus) cardiac cells revealed a fibroblast-specific role for Yap in controlling the expression of cytoskeletal and myofibroblast activation genes, as well as pro-inflammatory cyto/chemokines. Corroborating these RNAseq data, we observed increased macrophage infiltration in the scars of yap mutants at 7 days post-injury. CONCLUSION These results suggest that Yap deletion has minimal effect on myocyte proliferation in adults, but significantly influences scar formation and immune cell infiltration during zebrafish heart regeneration. Collectively, these data suggest an unexpected role for Yap in matrix formation and macrophage recruitment during heart regeneration.
Collapse
Affiliation(s)
- Michael A Flinn
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brooke E Jeffery
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Caitlin C O'Meara
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Genomics Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian A Link
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
141
|
Cui M, Wang Z, Chen K, Shah AM, Tan W, Duan L, Sanchez-Ortiz E, Li H, Xu L, Liu N, Bassel-Duby R, Olson EN. Dynamic Transcriptional Responses to Injury of Regenerative and Non-regenerative Cardiomyocytes Revealed by Single-Nucleus RNA Sequencing. Dev Cell 2020; 53:102-116.e8. [PMID: 32220304 DOI: 10.1016/j.devcel.2020.02.019] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/07/2020] [Accepted: 02/25/2020] [Indexed: 12/22/2022]
Abstract
The adult mammalian heart is incapable of regeneration following injury. In contrast, the neonatal mouse heart can efficiently regenerate during the first week of life. The molecular mechanisms that mediate the regenerative response and its blockade in later life are not understood. Here, by single-nucleus RNA sequencing, we map the dynamic transcriptional landscape of five distinct cardiomyocyte populations in healthy, injured, and regenerating mouse hearts. We identify immature cardiomyocytes that enter the cell cycle following injury and disappear as the heart loses the ability to regenerate. These proliferative neonatal cardiomyocytes display a unique transcriptional program dependent on nuclear transcription factor Y subunit alpha (NFYa) and nuclear factor erythroid 2-like 1 (NFE2L1) transcription factors, which exert proliferative and protective functions, respectively. Cardiac overexpression of these two factors conferred protection against ischemic injury in mature mouse hearts that were otherwise non-regenerative. These findings advance our understanding of the cellular basis of neonatal heart regeneration and reveal a transcriptional landscape for heart repair following injury.
Collapse
Affiliation(s)
- Miao Cui
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Zhaoning Wang
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Kenian Chen
- Quantitative Biomedical Research Center, Department of Population & Data Sciences and Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Akansha M Shah
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Wei Tan
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Lauren Duan
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Efrain Sanchez-Ortiz
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Hui Li
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Department of Population & Data Sciences and Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ning Liu
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
142
|
Bass-Stringer S, Ooi JYY, McMullen JR. Clusterin is regulated by IGF1–PI3K signaling in the heart: implications for biomarker and drug target discovery, and cardiotoxicity. Arch Toxicol 2020; 94:1763-1768. [DOI: 10.1007/s00204-020-02709-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
|
143
|
Locatelli P, Belaich MN, López AE, Olea FD, Uranga Vega M, Giménez CS, Simonin JA, Bauzá MDR, Castillo MG, Cuniberti LA, Crottogini A, Cerrudo CS, Ghiringhelli PD. Novel insights into cardiac regeneration based on differential fetal and adult ovine heart transcriptomic analysis. Am J Physiol Heart Circ Physiol 2020; 318:H994-H1007. [PMID: 32167779 DOI: 10.1152/ajpheart.00610.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The adult mammalian cardiomyocyte has a very limited capacity to reenter the cell cycle and advance into mitosis. Therefore, diseases characterized by lost contractile tissue usually evolve into myocardial remodeling and heart failure. Analyzing the cardiac transcriptome at different developmental stages in a large mammal closer to the human than laboratory rodents may serve to disclose positive and negative cardiomyocyte cell cycle regulators potentially targetable to induce cardiac regeneration in the clinical setting. Thus we aimed at characterizing the transcriptomic profiles of the early fetal, late fetal, and adult sheep heart by employing RNA-seq technique and bioinformatic analysis to detect protein-encoding genes that in some of the stages were turned off, turned on, or differentially expressed. Genes earlier proposed as positive cell cycle regulators such as cyclin A, cdk2, meis2, meis3, and PCNA showed higher expression in fetal hearts and lower in AH, as expected. In contrast, genes previously proposed as cell cycle inhibitors, such as meis1, p16, and sav1, tended to be higher in fetal than in adult hearts, suggesting that these genes are involved in cell processes other than cell cycle regulation. Additionally, we described Gene Ontology (GO) enrichment of different sets of genes. GO analysis revealed that differentially expressed gene sets were mainly associated with metabolic and cellular processes. The cell cycle-related genes fam64a, cdc20, and cdk1, and the metabolism-related genes pitx and adipoq showed strong differential expression between fetal and adult hearts, thus being potent candidates to be targeted in human cardiac regeneration strategies.NEW & NOTEWORTHY We characterized the transcriptomic profiles of the fetal and adult sheep hearts employing RNAseq technique and bioinformatic analyses to provide sets of transcripts whose variation in expression level may link them to a specific role in cell cycle regulation. It is important to remark that this study was performed in a large mammal closer to humans than laboratory rodents. In consequence, the results can be used for further translational studies in cardiac regeneration.
Collapse
Affiliation(s)
- Paola Locatelli
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Mariano N Belaich
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular; CONICET, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Ayelén E López
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Fernanda D Olea
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Martín Uranga Vega
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Carlos S Giménez
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Jorge Alejandro Simonin
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular; CONICET, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - María Del Rosario Bauzá
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Marta G Castillo
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Luis A Cuniberti
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Alberto Crottogini
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Carolina S Cerrudo
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular; CONICET, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Pablo D Ghiringhelli
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular; CONICET, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| |
Collapse
|
144
|
Windmueller R, Leach JP, Babu A, Zhou S, Morley MP, Wakabayashi A, Petrenko NB, Viatour P, Morrisey EE. Direct Comparison of Mononucleated and Binucleated Cardiomyocytes Reveals Molecular Mechanisms Underlying Distinct Proliferative Competencies. Cell Rep 2020; 30:3105-3116.e4. [PMID: 32130910 PMCID: PMC7194103 DOI: 10.1016/j.celrep.2020.02.034] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 11/25/2019] [Accepted: 02/07/2020] [Indexed: 11/26/2022] Open
Abstract
The mammalian heart is incapable of regenerating a sufficient number of cardiomyocytes to ameliorate the loss of contractile muscle after acute myocardial injury. Several reports have demonstrated that mononucleated cardiomyocytes are more responsive than are binucleated cardiomyocytes to pro-proliferative stimuli. We have developed a strategy to isolate and characterize highly enriched populations of mononucleated and binucleated cardiomyocytes at various times of development. Our results suggest that an E2f/Rb transcriptional network is central to the divergence of these two populations and that remnants of the differences acquired during the neonatal period remain in adult cardiomyocytes. Moreover, inducing binucleation by genetically blocking the ability of cardiomyocytes to complete cytokinesis leads to a reduction in E2f target gene expression, directly linking the E2f pathway with nucleation. These data identify key molecular differences between mononucleated and binucleated mammalian cardiomyocytes that can be used to leverage cardiomyocyte proliferation for promoting injury repair in the heart.
Collapse
Affiliation(s)
- Rebecca Windmueller
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John P Leach
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Apoorva Babu
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Su Zhou
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael P Morley
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aoi Wakabayashi
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nataliya B Petrenko
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Viatour
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
145
|
Abstract
PURPOSE OF REVIEW Comprehensive analyses of the genome, transcriptome, proteome and metabolome are instrumental in identifying biomarkers of disease, to gain insight into mechanisms underlying the development of cardiovascular disease, and show promise for better stratifying patients according to disease subtypes. This review highlights recent 'omics' studies, including integration of multiple 'omics' that have advanced mechanistic understanding and diagnosis in humans and animal models. RECENT FINDINGS Transcriptome-based discovery continues to be a primary method to obtain data for hypothesis generation and the understanding of disease pathogenesis has been enhanced by single cell-based methods capable of revealing heterogeneity in cellular responses. Advances in proteome coverage and quantitation of individual protein species, together with enhanced methods for detecting posttranslational modifications, have improved discovery of protein-based biomarkers. SUMMARY High-throughput assays capable of quantitating the vast majority of any particular type of biomolecule within a tissue sample, isolated cells or plasma are now available. In order to make best use of the large amount of data that can be generated on given molecule types, as well as their interrelationships in disease, continued development of pattern-recognition algorithms ('machine learning') will be required and the subclassification of disease that is made possible by such algorithms will be likely to inform clinical practice, and vice versa.
Collapse
|
146
|
Lock MC, Tellam RL, Darby JRT, Soo JY, Brooks DA, Macgowan CK, Selvanayagam JB, Porrello ER, Seed M, Keller-Wood M, Morrison JL. Differential gene responses 3 days following infarction in the fetal and adolescent sheep heart. Physiol Genomics 2020; 52:143-159. [PMID: 31961761 DOI: 10.1152/physiolgenomics.00092.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
There are critical molecular mechanisms that can be activated to induce myocardial repair, and in humans this is most efficient during fetal development. The timing of heart development in relation to birth and the size/electrophysiology of the heart are similar in humans and sheep, providing a model to investigate the repair capacity of the mammalian heart and how this can be applied to adult heart repair. Myocardial infarction was induced by ligation of the left anterior descending coronary artery in fetal (105 days gestation when cardiomyocytes are proliferative) and adolescent sheep (6 mo of age when all cardiomyocytes have switched to an adult phenotype). An ovine gene microarray was used to compare gene expression in sham and infarcted (remote, border and infarct areas) cardiac tissue from fetal and adolescent hearts. The gene response to myocardial infarction was less pronounced in fetal compared with adolescent sheep hearts and there were unique gene responses at each age. There were also region-specific changes in gene expression between each age, in the infarct tissue, tissue bordering the infarct, and tissue remote from the infarction. In total, there were 880 genes that responded to MI uniquely in the adolescent samples compared with 170 genes in the fetal response, as well as 742 overlap genes that showed concordant direction of change responses to infarction at both ages. In response to myocardial infarction, there were specific changes in genes within pathways of mitochondrial oxidation, muscle contraction, and hematopoietic cell lineages, suggesting that the control of energy utilization and immune function are critical for effective heart repair. The more restricted gene response in the fetus may be an important factor in its enhanced capacity for cardiac repair.
Collapse
Affiliation(s)
- Mitchell C Lock
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ross L Tellam
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Doug A Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | | | - Joseph B Selvanayagam
- Cardiac Imaging Research Group, Department of Heart Health, South Australian Health & Medical Research Institute, and Flinders University, Adelaide, South Australia, Australia
| | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Mike Seed
- Hospital for Sick Children, Division of Cardiology, Toronto, Ontario, Canada
| | | | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
147
|
Sreejit G, Abdel-Latif A, Athmanathan B, Annabathula R, Dhyani A, Noothi SK, Quaife-Ryan GA, Al-Sharea A, Pernes G, Dragoljevic D, Lal H, Schroder K, Hanaoka BY, Raman C, Grant MB, Hudson JE, Smyth SS, Porrello ER, Murphy AJ, Nagareddy PR. Neutrophil-Derived S100A8/A9 Amplify Granulopoiesis After Myocardial Infarction. Circulation 2020; 141:1080-1094. [PMID: 31941367 DOI: 10.1161/circulationaha.119.043833] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Myocardial infarction (MI) triggers myelopoiesis, resulting in heightened production of neutrophils. However, the mechanisms that sustain their production and recruitment to the injured heart are unclear. METHODS Using a mouse model of the permanent ligation of the left anterior descending artery and flow cytometry, we first characterized the temporal and spatial effects of MI on different myeloid cell types. We next performed global transcriptome analysis of different cardiac cell types within the infarct to identify the drivers of the acute inflammatory response and the underlying signaling pathways. Using a combination of genetic and pharmacological strategies, we identified the sequelae of events that led to MI-induced myelopoiesis. Cardiac function was assessed by echocardiography. The association of early indexes of neutrophilia with major adverse cardiovascular events was studied in a cohort of patients with acute MI. RESULTS Induction of MI results in rapid recruitment of neutrophils to the infarct, where they release specific alarmins, S100A8 and S100A9. These alarmins bind to the Toll-like receptor 4 and prime the nod-like receptor family pyrin domain-containing 3 inflammasome in naïve neutrophils and promote interleukin-1β secretion. The released interleukin-1β interacts with its receptor (interleukin 1 receptor type 1) on hematopoietic stem and progenitor cells in the bone marrow and stimulates granulopoiesis in a cell-autonomous manner. Genetic or pharmacological strategies aimed at disruption of S100A8/A9 and their downstream signaling cascade suppress MI-induced granulopoiesis and improve cardiac function. Furthermore, in patients with acute coronary syndrome, higher neutrophil count on admission and after revascularization correlates positively with major adverse cardiovascular disease outcomes. CONCLUSIONS Our study provides novel evidence for the primary role of neutrophil-derived alarmins (S100A8/A9) in dictating the nature of the ensuing inflammatory response after myocardial injury. Therapeutic strategies aimed at disruption of S100A8/A9 signaling or their downstream mediators (eg, nod-like receptor family pyrin domain-containing 3 inflammasome, interleukin-1β) in neutrophils suppress granulopoiesis and may improve cardiac function in patients with acute coronary syndrome.
Collapse
Affiliation(s)
- Gopalkrishna Sreejit
- Department of Surgery (G.S., B.A., P.R.N.), Ohio State University Wexner Medical Center, Columbus.,Departments of Pathology (G.S., B.A., A.D., S.K.N., P.R.N.), University of Alabama at Birmingham
| | - Ahmed Abdel-Latif
- Department of Medicine, University of Kentucky, Lexington (A.A.-L., R.A., S.S.S.)
| | - Baskaran Athmanathan
- Department of Surgery (G.S., B.A., P.R.N.), Ohio State University Wexner Medical Center, Columbus.,Departments of Pathology (G.S., B.A., A.D., S.K.N., P.R.N.), University of Alabama at Birmingham
| | - Rahul Annabathula
- Department of Medicine, University of Kentucky, Lexington (A.A.-L., R.A., S.S.S.)
| | - Ashish Dhyani
- Departments of Pathology (G.S., B.A., A.D., S.K.N., P.R.N.), University of Alabama at Birmingham
| | - Sunil K Noothi
- Departments of Pathology (G.S., B.A., A.D., S.K.N., P.R.N.), University of Alabama at Birmingham.,Ophthalmology and Visual Sciences (S.K.N., M.B.G.), University of Alabama at Birmingham
| | - Gregory A Quaife-Ryan
- School of Biomedical Sciences (G.A.Q.-R.), University of Queensland, St. Lucia, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, Australia (G.A.Q.-R., J.E.H.)
| | - Annas Al-Sharea
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia (A.A.-S., G.P., D.D., A.J.M.)
| | - Gerard Pernes
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia (A.A.-S., G.P., D.D., A.J.M.)
| | - Dragana Dragoljevic
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia (A.A.-S., G.P., D.D., A.J.M.)
| | - Hind Lal
- Medicine (H.L., B.Y.H., C.R.), University of Alabama at Birmingham
| | - Kate Schroder
- Institute for Molecular Bioscience (IMB) (K.S.), University of Queensland, St. Lucia, Australia.,IMB Centre for Inflammation and Disease Research (K.S.), University of Queensland, St. Lucia, Australia
| | - Beatriz Y Hanaoka
- Department of Medicine (B.Y.H.), Ohio State University Wexner Medical Center, Columbus.,Medicine (H.L., B.Y.H., C.R.), University of Alabama at Birmingham
| | - Chander Raman
- Medicine (H.L., B.Y.H., C.R.), University of Alabama at Birmingham
| | - Maria B Grant
- Ophthalmology and Visual Sciences (S.K.N., M.B.G.), University of Alabama at Birmingham
| | - James E Hudson
- QIMR Berghofer Medical Research Institute, Brisbane, Australia (G.A.Q.-R., J.E.H.)
| | - Susan S Smyth
- Department of Medicine, University of Kentucky, Lexington (A.A.-L., R.A., S.S.S.)
| | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, Australia (E.R.P.).,Department of Physiology, School of Biomedical Sciences, University of Melbourne, Australia (E.R.P.)
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia (A.A.-S., G.P., D.D., A.J.M.).,Department of Immunology, Monash University, Melbourne, Australia (A.J.M.)
| | - Prabhakara R Nagareddy
- Department of Surgery (G.S., B.A., P.R.N.), Ohio State University Wexner Medical Center, Columbus.,Departments of Pathology (G.S., B.A., A.D., S.K.N., P.R.N.), University of Alabama at Birmingham
| |
Collapse
|
148
|
Tan SM, Ziemann M, Thallas-Bonke V, Snelson M, Kumar V, Laskowski A, Nguyen TV, Huynh K, Clarke MV, Libianto R, Baker ST, Skene A, Power DA, MacIsaac RJ, Henstridge DC, Wetsel RA, El-Osta A, Meikle PJ, Wilson SG, Forbes JM, Cooper ME, Ekinci EI, Woodruff TM, Coughlan MT. Complement C5a Induces Renal Injury in Diabetic Kidney Disease by Disrupting Mitochondrial Metabolic Agility. Diabetes 2020; 69:83-98. [PMID: 31624141 DOI: 10.2337/db19-0043] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 10/08/2019] [Indexed: 11/13/2022]
Abstract
The sequelae of diabetes include microvascular complications such as diabetic kidney disease (DKD), which involves glucose-mediated renal injury associated with a disruption in mitochondrial metabolic agility, inflammation, and fibrosis. We explored the role of the innate immune complement component C5a, a potent mediator of inflammation, in the pathogenesis of DKD in clinical and experimental diabetes. Marked systemic elevation in C5a activity was demonstrated in patients with diabetes; conventional renoprotective agents did not therapeutically target this elevation. C5a and its receptor (C5aR1) were upregulated early in the disease process and prior to manifest kidney injury in several diverse rodent models of diabetes. Genetic deletion of C5aR1 in mice conferred protection against diabetes-induced renal injury. Transcriptomic profiling of kidney revealed diabetes-induced downregulation of pathways involved in mitochondrial fatty acid metabolism. Interrogation of the lipidomics signature revealed abnormal cardiolipin remodeling in diabetic kidneys, a cardinal sign of disrupted mitochondrial architecture and bioenergetics. In vivo delivery of an orally active inhibitor of C5aR1 (PMX53) reversed the phenotypic changes and normalized the renal mitochondrial fatty acid profile, cardiolipin remodeling, and citric acid cycle intermediates. In vitro exposure of human renal proximal tubular epithelial cells to C5a led to altered mitochondrial respiratory function and reactive oxygen species generation. These experiments provide evidence for a pivotal role of the C5a/C5aR1 axis in propagating renal injury in the development of DKD by disrupting mitochondrial agility, thereby establishing a new immunometabolic signaling pathway in DKD.
Collapse
Affiliation(s)
- Sih Min Tan
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Mark Ziemann
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
- School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia
| | - Vicki Thallas-Bonke
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Matthew Snelson
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Vinod Kumar
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Adrienne Laskowski
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | | | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Michele V Clarke
- Department of Endocrinology, Austin Health, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Renata Libianto
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Scott T Baker
- Department of Endocrinology, Austin Health, Melbourne, Victoria, Australia
| | - Alison Skene
- Department of Anatomical Pathology, Austin Health, Melbourne, Victoria, Australia
| | - David A Power
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Department of Nephrology and Institute for Breathing and Sleep, Austin Health, Melbourne, Victoria, Australia
| | - Richard J MacIsaac
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Department of Endocrinology and Diabetes, St Vincent's Hospital, Melbourne, Victoria, Australia
| | | | - Rick A Wetsel
- Research Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas-Houston, Houston, TX
| | - Assam El-Osta
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Scott G Wilson
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Renal Medicine, Alfred Health, Melbourne, Victoria, Australia
| | - Josephine M Forbes
- Glycation and Diabetes Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Mark E Cooper
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Elif I Ekinci
- Department of Endocrinology, Austin Health, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Melinda T Coughlan
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
149
|
Agnew EJ, Velayutham N, Matos Ortiz G, Alfieri CM, Hortells L, Moore V, Riggs KW, Baker RS, Gibson AM, Ponny SR, Alsaied T, Zafar F, Yutzey KE. Scar Formation with Decreased Cardiac Function Following Ischemia/Reperfusion Injury in 1 Month Old Swine. J Cardiovasc Dev Dis 2019; 7:E1. [PMID: 31861331 PMCID: PMC7151069 DOI: 10.3390/jcdd7010001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 02/07/2023] Open
Abstract
Studies in mice show a brief neonatal period of cardiac regeneration with minimal scar formation, but less is known about reparative mechanisms in large mammals. A transient cardiac injury approach (ischemia/reperfusion, IR) was used in weaned postnatal day (P)30 pigs to assess regenerative repair in young large mammals at a stage when cardiomyocyte (CM) mitotic activity is still detected. Female and male P30 pigs were subjected to cardiac ischemia (1 h) by occlusion of the left anterior descending artery followed by reperfusion, or to a sham operation. Following IR, myocardial damage occurred, with cardiac ejection fraction significantly decreased 2 h post-ischemia. No improvement or worsening of cardiac function to the 4 week study end-point was observed. Histology demonstrated CM cell cycling, detectable by phospho-histone H3 staining, at 2 months of age in multinucleated CMs in both sham-operated and IR pigs. Inflammation and regional scar formation in the epicardial region proximal to injury were observed 4 weeks post-IR. Thus, pigs subjected to cardiac IR at P30 show myocardial damage with a prolonged decrease in cardiac function, formation of a regional scar, and increased inflammation, but do not regenerate myocardium even in the presence of CM mitotic activity.
Collapse
Affiliation(s)
- Emma J Agnew
- Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229, USA; (E.J.A.); (N.V.); (G.M.O.); (C.M.A.); (L.H.)
| | - Nivedhitha Velayutham
- Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229, USA; (E.J.A.); (N.V.); (G.M.O.); (C.M.A.); (L.H.)
| | - Gabriela Matos Ortiz
- Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229, USA; (E.J.A.); (N.V.); (G.M.O.); (C.M.A.); (L.H.)
| | - Christina M Alfieri
- Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229, USA; (E.J.A.); (N.V.); (G.M.O.); (C.M.A.); (L.H.)
| | - Luis Hortells
- Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229, USA; (E.J.A.); (N.V.); (G.M.O.); (C.M.A.); (L.H.)
| | - Victoria Moore
- Cincinnati Children’s Hospital Heart Institute, Department of Pediatrics, University of Cincinnati College of Medicine Cincinnati, Cincinnati, OH 45229, USA; (V.M.); (T.A.)
| | - Kyle W Riggs
- Division of Pediatric Cardiothoracic Surgery, The Heart Institute, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA; (K.W.R.); (R.S.B.); (A.M.G.); (F.Z.)
| | - R. Scott Baker
- Division of Pediatric Cardiothoracic Surgery, The Heart Institute, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA; (K.W.R.); (R.S.B.); (A.M.G.); (F.Z.)
| | - Aaron M Gibson
- Division of Pediatric Cardiothoracic Surgery, The Heart Institute, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA; (K.W.R.); (R.S.B.); (A.M.G.); (F.Z.)
| | - Sithara Raju Ponny
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA;
| | - Tarek Alsaied
- Cincinnati Children’s Hospital Heart Institute, Department of Pediatrics, University of Cincinnati College of Medicine Cincinnati, Cincinnati, OH 45229, USA; (V.M.); (T.A.)
| | - Farhan Zafar
- Division of Pediatric Cardiothoracic Surgery, The Heart Institute, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA; (K.W.R.); (R.S.B.); (A.M.G.); (F.Z.)
| | - Katherine E Yutzey
- Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229, USA; (E.J.A.); (N.V.); (G.M.O.); (C.M.A.); (L.H.)
| |
Collapse
|
150
|
Wang M, Hu S, Nie Y, Song J. Proteomic profiling of key transcription factors in the process of neonatal mouse cardiac regeneration capacity loss. Cell Biol Int 2019; 43:1435-1442. [PMID: 31187911 DOI: 10.1002/cbin.11192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 06/06/2019] [Indexed: 01/24/2023]
Abstract
The heart lacks complete regeneration capacity. In mice, the cardiac apex can regenerate 1 day after birth, although 7 days after birth the repair occurs with a fibrous scar. However, the key transcription factors (TFs) related to this loss of regeneration capacity remain largely unknown. We aimed to find candidates for key TFs using proteomic profiling and comparison during loss of neonatal mouse cardiac regeneration capacity, with preliminary validation using western blotting (WB) and real-time quantitative polymerase chain reaction (RT-qPCR). A total of 69 common discrepant TFs with similar variation trends in two TF response element experiments were identified, 18 of which were matched to known key signaling pathways of cardiac regeneration after pathway enrichment of downstream genes. Validation using RT-qPCR-selected DACH1, RBL1 (P107), and TBX20, and further validation with WB-selected RBL1 (P107) and TBX20. We therefore identified two candidates for key TFs in the loss of mouse cardiac apex regeneration capacity. TBX20 has been biologically validated, and RBL1 (P107) needs to be validated in the future.
Collapse
Affiliation(s)
- Mangyuan Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, P.R. China
| | - ShengShou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, P.R. China
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, P.R. China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, P.R. China
| |
Collapse
|