51
|
Sladitschek-Martens HL, Guarnieri A, Brumana G, Zanconato F, Battilana G, Xiccato RL, Panciera T, Forcato M, Bicciato S, Guzzardo V, Fassan M, Ulliana L, Gandin A, Tripodo C, Foiani M, Brusatin G, Cordenonsi M, Piccolo S. YAP/TAZ activity in stromal cells prevents ageing by controlling cGAS-STING. Nature 2022; 607:790-798. [PMID: 35768505 PMCID: PMC7613988 DOI: 10.1038/s41586-022-04924-6] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 06/01/2022] [Indexed: 02/06/2023]
Abstract
Ageing is intimately connected to the induction of cell senescence1,2, but why this is so remains poorly understood. A key challenge is the identification of pathways that normally suppress senescence, are lost during ageing and are functionally relevant to oppose ageing3. Here we connected the structural and functional decline of ageing tissues to attenuated function of the master effectors of cellular mechanosignalling YAP and TAZ. YAP/TAZ activity declines during physiological ageing in stromal cells, and mimicking such decline through genetic inactivation of YAP/TAZ in these cells leads to accelerated ageing. Conversely, sustaining YAP function rejuvenates old cells and opposes the emergence of ageing-related traits associated with either physiological ageing or accelerated ageing triggered by a mechano-defective extracellular matrix. Ageing traits induced by inactivation of YAP/TAZ are preceded by induction of tissue senescence. This occurs because YAP/TAZ mechanotransduction suppresses cGAS-STING signalling, to the extent that inhibition of STING prevents tissue senescence and premature ageing-related tissue degeneration after YAP/TAZ inactivation. Mechanistically, YAP/TAZ-mediated control of cGAS-STING signalling relies on the unexpected role of YAP/TAZ in preserving nuclear envelope integrity, at least in part through direct transcriptional regulation of lamin B1 and ACTR2, the latter of which is involved in building the peri-nuclear actin cap. The findings demonstrate that declining YAP/TAZ mechanotransduction drives ageing by unleashing cGAS-STING signalling, a pillar of innate immunity. Thus, sustaining YAP/TAZ mechanosignalling or inhibiting STING may represent promising approaches for limiting senescence-associated inflammation and improving healthy ageing.
Collapse
Affiliation(s)
| | | | - Giulia Brumana
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Giusy Battilana
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Tito Panciera
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Mattia Forcato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Matteo Fassan
- Department of Medicine, University of Padua, Padua, Italy
| | - Lorenzo Ulliana
- Department of Industrial Engineering, University of Padua, Padua, Italy
| | - Alessandro Gandin
- Department of Industrial Engineering, University of Padua, Padua, Italy
| | - Claudio Tripodo
- Department of Health Sciences Unit, Human Pathology Section, University of Palermo, Palermo, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marco Foiani
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- University of Milan, Milan, Italy
| | - Giovanna Brusatin
- Department of Industrial Engineering, University of Padua, Padua, Italy
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy.
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
52
|
Bruton FA, Kaveh A, Ross-Stewart KM, Matrone G, Oremek MEM, Solomonidis EG, Tucker CS, Mullins JJ, Lucas CD, Brittan M, Taylor JM, Rossi AG, Denvir MA. Macrophages trigger cardiomyocyte proliferation by increasing epicardial vegfaa expression during larval zebrafish heart regeneration. Dev Cell 2022; 57:1512-1528.e5. [PMID: 35688158 PMCID: PMC9616726 DOI: 10.1016/j.devcel.2022.05.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/11/2022] [Accepted: 05/18/2022] [Indexed: 12/01/2022]
Abstract
Cardiac injury leads to the loss of cardiomyocytes, which are rapidly replaced by the proliferation of the surviving cells in zebrafish, but not in mammals. In both the regenerative zebrafish and non-regenerative mammals, cardiac injury induces a sustained macrophage response. Macrophages are required for cardiomyocyte proliferation during zebrafish cardiac regeneration, but the mechanisms whereby macrophages facilitate this crucial process are fundamentally unknown. Using heartbeat-synchronized live imaging, RNA sequencing, and macrophage-null genotypes in the larval zebrafish cardiac injury model, we characterize macrophage function and reveal that these cells activate the epicardium, inducing cardiomyocyte proliferation. Mechanistically, macrophages are specifically recruited to the epicardial-myocardial niche, triggering the expansion of the epicardium, which upregulates vegfaa expression to induce cardiomyocyte proliferation. Our data suggest that epicardial Vegfaa augments a developmental cardiac growth pathway via increased endocardial notch signaling. The identification of this macrophage-dependent mechanism of cardiac regeneration highlights immunomodulation as a potential strategy for enhancing mammalian cardiac repair. Heart regeneration in larval zebrafish is characterized in detail Macrophage ablation blocks cardiomyocyte proliferation after cardiac injury Macrophages synapse with epicardial cells and promote their proliferation Epicardial Vegfaa drives cardiomyocyte proliferation during cardiac regeneration
Collapse
Affiliation(s)
- Finnius A Bruton
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| | - Aryan Kaveh
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine M Ross-Stewart
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Gianfranco Matrone
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Magdalena E M Oremek
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Emmanouil G Solomonidis
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Carl S Tucker
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - John J Mullins
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Christopher D Lucas
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Mairi Brittan
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | | - Adriano G Rossi
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Martin A Denvir
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| |
Collapse
|
53
|
Dupont S, Wickström SA. Mechanical regulation of chromatin and transcription. Nat Rev Genet 2022; 23:624-643. [DOI: 10.1038/s41576-022-00493-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 01/14/2023]
|
54
|
Abouleisa RRE, Salama ABM, Ou Q, Tang XL, Solanki M, Guo Y, Nong Y, McNally L, Lorkiewicz PK, Kassem KM, Ahern BM, Choudhary K, Thomas R, Huang Y, Juhardeen HR, Siddique A, Ifthikar Z, Hammad SK, Elbaz AS, Ivey KN, Conklin DJ, Satin J, Hill BG, Srivastava D, Bolli R, Mohamed TMA. Transient Cell Cycle Induction in Cardiomyocytes to Treat Subacute Ischemic Heart Failure. Circulation 2022; 145:1339-1355. [PMID: 35061545 PMCID: PMC9038650 DOI: 10.1161/circulationaha.121.057641] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND The regenerative capacity of the heart after myocardial infarction is limited. Our previous study showed that ectopic introduction of 4 cell cycle factors (4F; CDK1 [cyclin-dependent kinase 1], CDK4 [cyclin-dependent kinase 4], CCNB [cyclin B1], and CCND [cyclin D1]) promotes cardiomyocyte proliferation in 15% to 20% of infected cardiomyocytes in vitro and in vivo and improves cardiac function after myocardial infarction in mice. METHODS Using temporal single-cell RNA sequencing, we aimed to identify the necessary reprogramming stages during the forced cardiomyocyte proliferation with 4F on a single cell basis. Using rat and pig models of ischemic heart failure, we aimed to start the first preclinical testing to introduce 4F gene therapy as a candidate for the treatment of ischemia-induced heart failure. RESULTS Temporal bulk and single-cell RNA sequencing and further biochemical validations of mature human induced pluripotent stem cell-derived cardiomyocytes treated with either LacZ or 4F adenoviruses revealed full cell cycle reprogramming in 15% of the cardiomyocyte population at 48 hours after infection with 4F, which was associated mainly with sarcomere disassembly and metabolic reprogramming (n=3/time point/group). Transient overexpression of 4F, specifically in cardiomyocytes, was achieved using a polycistronic nonintegrating lentivirus (NIL) encoding 4F; each is driven by a TNNT2 (cardiac troponin T isoform 2) promoter (TNNT2-4Fpolycistronic-NIL). TNNT2-4Fpolycistronic-NIL or control virus was injected intramyocardially 1 week after myocardial infarction in rats (n=10/group) or pigs (n=6-7/group). Four weeks after injection, TNNT2-4Fpolycistronic-NIL-treated animals showed significant improvement in left ventricular ejection fraction and scar size compared with the control virus-treated animals. At 4 months after treatment, rats that received TNNT2-4Fpolycistronic-NIL still showed a sustained improvement in cardiac function and no obvious development of cardiac arrhythmias or systemic tumorigenesis (n=10/group). CONCLUSIONS This study provides mechanistic insights into the process of forced cardiomyocyte proliferation and advances the clinical feasibility of this approach by minimizing the oncogenic potential of the cell cycle factors owing to the use of a novel transient and cardiomyocyte-specific viral construct.
Collapse
Affiliation(s)
- Riham R. E. Abouleisa
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Abou Bakr M. Salama
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- Faculty of Medicine, Zagazig University, Egypt
| | - Qinghui Ou
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Xian-Liang Tang
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Mitesh Solanki
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Yiru Guo
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Yibing Nong
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Lindsey McNally
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, U.S.A
| | - Pawel K. Lorkiewicz
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, U.S.A
| | - Kamal M. Kassem
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | | | | | | | - Yu Huang
- Gladstone Institute, San Francisco, CA, U.S.A
| | | | - Aisha Siddique
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Zainab Ifthikar
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Sally K. Hammad
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- Department of Biochemistry Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Ayman S. Elbaz
- Department of Bioengineering, Speed School of Engineering, University of Louisville, KY, U.S.A
| | | | - Daniel J Conklin
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, U.S.A
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, KY, U.S.A
| | - Bradford G. Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, U.S.A
| | | | - Roberto Bolli
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Tamer M A Mohamed
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, U.S.A
- Department of Bioengineering, Speed School of Engineering, University of Louisville, KY, U.S.A
- Department of Pharmacology and Toxicology, University of Louisville, KY, U.S.A
- Institute of Cardiovascular Sciences, University of Manchester, U.K
| |
Collapse
|
55
|
El-Sammak H, Yang B, Guenther S, Chen W, Marín-Juez R, Stainier DY. A Vegfc-Emilin2a-Cxcl8a Signaling Axis Required for Zebrafish Cardiac Regeneration. Circ Res 2022; 130:1014-1029. [PMID: 35264012 PMCID: PMC8976759 DOI: 10.1161/circresaha.121.319929] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Ischemic heart disease following the obstruction of coronary vessels leads to the death of cardiac tissue and the formation of a fibrotic scar. In contrast to adult mammals, zebrafish can regenerate their heart after injury, enabling the study of the underlying mechanisms. One of the earliest responses following cardiac injury in adult zebrafish is coronary revascularization. Defects in this process lead to impaired cardiomyocyte repopulation and scarring. Hence, identifying and investigating factors that promote coronary revascularization holds great therapeutic potential. METHODS We used wholemount imaging, immunohistochemistry and histology to assess various aspects of zebrafish cardiac regeneration. Deep transcriptomic analysis allowed us to identify targets and potential effectors of Vegfc (vascular endothelial growth factor C) signaling. We used newly generated loss- and gain-of-function genetic tools to investigate the role of Emilin2a (elastin microfibril interfacer 2a) and Cxcl8a (chemokine (C-X-C) motif ligand 8a)-Cxcr1 (chemokine (C-X-C) motif receptor 1) signaling in cardiac regeneration. RESULTS We first show that regenerating coronary endothelial cells upregulate vegfc upon cardiac injury in adult zebrafish and that Vegfc signaling is required for their proliferation during regeneration. Notably, blocking Vegfc signaling also significantly reduces cardiomyocyte dedifferentiation and proliferation. Using transcriptomic analyses, we identified emilin2a as a target of Vegfc signaling and found that manipulation of emilin2a expression can modulate coronary revascularization as well as cardiomyocyte proliferation. Mechanistically, Emilin2a induces the expression of the chemokine gene cxcl8a in epicardium-derived cells, while cxcr1, the Cxcl8a receptor gene, is expressed in coronary endothelial cells. We further show that Cxcl8a-Cxcr1 signaling is also required for coronary endothelial cell proliferation during cardiac regeneration. CONCLUSIONS These data show that after cardiac injury, coronary endothelial cells upregulate vegfc to promote coronary network reestablishment and cardiac regeneration. Mechanistically, Vegfc signaling upregulates epicardial emilin2a and cxcl8a expression to promote cardiac regeneration. These studies aid in understanding the mechanisms underlying coronary revascularization in zebrafish, with potential therapeutic implications to enhance revascularization and regeneration in injured human hearts.
Collapse
Affiliation(s)
- Hadil El-Sammak
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Frankfurt, Germany
| | - Bingyuan Yang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Stefan Guenther
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Frankfurt, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Rubén Marín-Juez
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Current address: Centre Hospitalier Universitaire Sainte-Justine Research Center, 3175 Chemin de la Côte-Sainte-Catherine, H3T 1C5 Montréal, QC, Canada, Department of Pathology and Cell Biology, University of Montreal, Montréal, QC H3T 1J4, Canada
| | - Didier Y.R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Frankfurt, Germany
| |
Collapse
|
56
|
Ramaccini D, Pedriali G, Perrone M, Bouhamida E, Modesti L, Wieckowski MR, Giorgi C, Pinton P, Morciano G. Some Insights into the Regulation of Cardiac Physiology and Pathology by the Hippo Pathway. Biomedicines 2022; 10:biomedicines10030726. [PMID: 35327528 PMCID: PMC8945338 DOI: 10.3390/biomedicines10030726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/17/2022] [Accepted: 03/19/2022] [Indexed: 11/16/2022] Open
Abstract
The heart is one of the most fascinating organs in living beings. It beats up to 100,000 times a day throughout the lifespan, without resting. The heart undergoes profound anatomical, biochemical, and functional changes during life, from hypoxemic fetal stages to a completely differentiated four-chambered cardiac muscle. In the middle, many biological events occur after and intersect with each other to regulate development, organ size, and, in some cases, regeneration. Several studies have defined the essential roles of the Hippo pathway in heart physiology through the regulation of apoptosis, autophagy, cell proliferation, and differentiation. This molecular route is composed of multiple components, some of which were recently discovered, and is highly interconnected with multiple known prosurvival pathways. The Hippo cascade is evolutionarily conserved among species, and in addition to its regulatory roles, it is involved in disease by drastically changing the heart phenotype and its function when its components are mutated, absent, or constitutively activated. In this review, we report some insights into the regulation of cardiac physiology and pathology by the Hippo pathway.
Collapse
Affiliation(s)
- Daniela Ramaccini
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Gaia Pedriali
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Mariasole Perrone
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Esmaa Bouhamida
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Lorenzo Modesti
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland;
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
- Correspondence: (P.P.); (G.M.); Tel.: +39-0532-455-802 (P.P.); +39-0532-455-804 (G.M.)
| | - Giampaolo Morciano
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
- Correspondence: (P.P.); (G.M.); Tel.: +39-0532-455-802 (P.P.); +39-0532-455-804 (G.M.)
| |
Collapse
|
57
|
The Hippo Pathway Effectors YAP/TAZ Are Essential for Mineralized Tissue Homeostasis in the Alveolar Bone/Periodontal Complex. J Dev Biol 2022; 10:jdb10010014. [PMID: 35323233 PMCID: PMC8948986 DOI: 10.3390/jdb10010014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/11/2022] [Accepted: 02/22/2022] [Indexed: 11/18/2022] Open
Abstract
YAP and TAZ are essential transcriptional co-activators and downstream effectors of the Hippo pathway, regulating cell proliferation, organ growth, and tissue homeostasis. To ask how the Hippo pathway affects mineralized tissue homeostasis in a tissue that is highly reliant on a tight homeostatic control of mineralized deposition and resorption, we determined the effects of YAP/TAZ dysregulation on the periodontal tissues alveolar bone, root cementum, and periodontal ligament. Loss of YAP/TAZ was associated with a reduction of mineralized tissue density in cellular cementum and alveolar bone, a downregulation in collagen I, alkaline phosphatase, and RUNX2 gene expression, an increase in the resorption markers TRAP and cathepsin K, and elevated numbers of TRAP-stained osteoclasts. Cyclic strain applied to periodontal ligament cells resulted in YAP nuclear localization, an effect that was abolished after blocking YAP. The rescue of YAP signaling with the heparan sulfate proteoglycan agrin resulted in a return of the nuclear YAP signal. Illustrating the key role of YAP on mineralization gene expression, the YAP inhibition-related downregulation of mineralization-associated genes was reversed by the extracellular matrix YAP activator agrin. Application of the unopposed mouse molar model to transform the periodontal ligament into an unloaded state and facilitate the distal drift of teeth resulted in an overall increase in mineralization-associated gene expression, an effect that was 10–20% diminished in Wnt1Cre/YAP/TAZ mutant mice. The unloaded state of the unopposed molar model in Wnt1Cre/YAP/TAZ mutant mice also caused a significant three-fold increase in osteoclast numbers, a substantial increase in bone/cementum resorption, pronounced periodontal ligament hyalinization, and thickened periodontal fiber bundles. Together, these data demonstrated that YAP/TAZ signaling is essential for the microarchitectural integrity of the periodontium by regulating mineralization gene expression and preventing excessive resorption during bodily movement of the dentoalveolar complex.
Collapse
|
58
|
Huang S, Shao T, Liu H, Li T, Gui X, Zhao Q. Resident Fibroblast MKL1 Is Sufficient to Drive Pro-fibrogenic Response in Mice. Front Cell Dev Biol 2022; 9:812748. [PMID: 35178401 PMCID: PMC8844195 DOI: 10.3389/fcell.2021.812748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/23/2021] [Indexed: 11/29/2022] Open
Abstract
Fibrosis is an evolutionarily conserved pathophysiological process serving bifurcated purposes. On the one hand, fibrosis is essential for wound healing and contributes to the preservation of organ function. On the other hand, aberrant fibrogenic response may lead to tissue remodeling and precipitate organ failure. Recently lineage tracing studies have shown that resident fibroblasts are the primary mediator of fibrosis taking place in key organs such as the heart, the lungs, and the kidneys. Megakaryocytic leukemia 1 (MKL1) is transcriptional regulator involved in tissue fibrosis. Here we generated resident fibroblast conditional MKL1 knockout (CKO) mice by crossing the Mkl1f/f mice to the Col1a2-CreERT2 mice. Models of cardiac fibrosis, pulmonary fibrosis, and renal fibrosis were reproduced in the CKO mice and wild type (WT) littermates. Compared to the WT mice, the CKO mice displayed across-the-board attenuation of fibrosis in different models. Our data cement the pivotal role MKL1 plays in tissue fibrosis but point to the cellular origin from which MKL1 exerts its pro-fibrogenic effects.
Collapse
Affiliation(s)
- Shan Huang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Cardiology, Research Unit of Island Emergency Medicine of Chinese Academy of Medical Sciences, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Tinghui Shao
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Hong Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Tianfa Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Cardiology, Research Unit of Island Emergency Medicine of Chinese Academy of Medical Sciences, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xianhua Gui
- Department of Respiratory Medicine, Affiliated Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Qianwen Zhao
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Cardiology, Research Unit of Island Emergency Medicine of Chinese Academy of Medical Sciences, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
59
|
Garduño-Rosales M, Callejas-Negrete OA, Medina-Castellanos E, Bartnicki-García S, Herrera-Estrella A, Mouriño-Pérez RR. F-actin dynamics following mechanical injury of Trichoderma atroviride and Neurospora crassa hyphae. Fungal Genet Biol 2022; 159:103672. [PMID: 35150841 DOI: 10.1016/j.fgb.2022.103672] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/24/2022] [Accepted: 01/30/2022] [Indexed: 11/04/2022]
Abstract
We investigated hyphae regeneration in Trichoderma atroviride and Neurospora crassa, with particular focus on determining the role of the actin cytoskeleton after mechanical injury. Filamentous actin (F-actin) dynamics were observed by live-cell confocal microscopy in both T. atroviride and N. crassa strains expressing Lifeact-GFP. In growing hyphae of both fungi, F-actin localized in three different structural forms: patches, cables and actomyosin rings. Most patches were conspicuously arranged in a collar in the hyphal subapex. A strong F-actin signal, likely actin filaments, colocalized with the core of the Spitzenkörper. Filaments and cables of F-actin we observed along the cortex throughout hyphae. Following mechanical damage at the margin of growing mycelia of T. atroviride and N. crassa, the severed hyphae lost their cytoplasmic contents, but plugging of the septal pore by a Woronin body, the rest of the hyphal tube remained whole. In both fungi, patches of F-actin began accumulating next to the plugged septum. Regeneration was attained by the emergence of a new hyphal tube as an extension of the plugged septum wall. The septum wall was gradually remodeled into the apical wall of the emerging hypha. Whereas in T. atroviride the re-initiation of polarized growth took about ∼1 h, in N. crassa, actin patch accumulation began almost immediately, and new growing hyphae were observed ∼30 min after injury. By confocal microscopy, we found that chitin synthase 1 (CHS-1), a microvesicle (chitosome) component, accumulated next to the plugged septum in regenerating hyphae of N. crassa. We concluded that the actin cytoskeleton plays a key role in hyphal regeneration by supporting membrane remodeling, helping to facilitate transport of vesicles responsible for new wall growth and organization of the new tip-growth apparatus.
Collapse
Affiliation(s)
- Marisela Garduño-Rosales
- Departamento de Microbiología. Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada, B.C., México
| | - Olga A Callejas-Negrete
- Departamento de Microbiología. Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada, B.C., México
| | - Elizabeth Medina-Castellanos
- Departamento de Microbiología. Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada, B.C., México; Laboratorio Nacional de Genómica para la Biodiversidad, CINVESTAV-Irapuato. Irapuato, Gto., México
| | - Salomon Bartnicki-García
- Departamento de Microbiología. Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada, B.C., México
| | - Alfredo Herrera-Estrella
- Laboratorio Nacional de Genómica para la Biodiversidad, CINVESTAV-Irapuato. Irapuato, Gto., México
| | - Rosa R Mouriño-Pérez
- Departamento de Microbiología. Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada, B.C., México.
| |
Collapse
|
60
|
Berecz T, Yiu A, Vittay O, Orsolits B, Mioulane M, dos Remedios CG, Ketteler R, Merkely B, Apáti Á, Harding SE, Hellen N, Foldes G. Transcriptional co-activators YAP1-TAZ of Hippo signalling in doxorubicin-induced cardiomyopathy. ESC Heart Fail 2022; 9:224-235. [PMID: 34931757 PMCID: PMC8787991 DOI: 10.1002/ehf2.13756] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/02/2021] [Accepted: 11/24/2021] [Indexed: 11/07/2022] Open
Abstract
AIMS Hippo signalling is an evolutionarily conserved pathway that controls organ size by regulating apoptosis, cell proliferation, and stem cell self-renewal. Recently, the pathway has been shown to exert powerful growth regulatory activity in cardiomyocytes. However, the functional role of this stress-related and cell death-related pathway in the human heart and cardiomyocytes is not known. In this study, we investigated the role of the transcriptional co-activators of Hippo signalling, YAP and TAZ, in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in response to cardiotoxic agents and investigated the effects of modulating the pathway on cardiomyocyte function and survival. METHODS AND RESULTS RNA-sequencing analysis of human heart samples with doxorubicin-induced end-stage heart failure and healthy controls showed that YAP and ERBB2 (HER2) as upstream regulators of differentially expressed genes correlated with doxorubicin treatment. Thus, we tested the effects of doxorubicin on hiPSC-CMs in vitro. Using an automated high-content screen of 96 clinically relevant antineoplastic and cardiotherapeutic drugs, we showed that doxorubicin induced the highest activation of YAP/TAZ nuclear translocation in both hiPSC-CMs and control MCF7 breast cancer cells. The overexpression of YAP rescued doxorubicin-induced cell loss in hiPSC-CMs by inhibiting apoptosis and inducing proliferation. In contrast, silencing of YAP and TAZ by siRNAs resulted in elevated mitochondrial membrane potential loss in response to doxorubicin. hiPSC-CM calcium transients did not change in response to YAP/TAZ silencing. CONCLUSIONS Our results suggest that Hippo signalling is involved in clinical anthracycline-induced cardiomyopathy. Modelling with hiPSC-CMs in vitro showed similar responses to doxorubicin as adult cardiomyocytes and revealed a potential cardioprotective effect of YAP in doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Tünde Berecz
- Heart and Vascular CenterSemmelweis University68 Városmajor StreetBudapestH1122Hungary
- Institute of Enzymology, Research Centre for Natural SciencesEötvös Loránd Research NetworkBudapestHungary
| | - Angela Yiu
- Department of Surgery and CancerImperial College LondonLondonUK
| | - Orsolya Vittay
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Barbara Orsolits
- Heart and Vascular CenterSemmelweis University68 Városmajor StreetBudapestH1122Hungary
| | - Maxime Mioulane
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Cristobal G. dos Remedios
- Victor Chang Cardiac Research InstituteDarlinghurstNSWAustralia
- Bosch InstituteThe University of SydneySydneyNSWAustralia
| | - Robin Ketteler
- Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| | - Bela Merkely
- Heart and Vascular CenterSemmelweis University68 Városmajor StreetBudapestH1122Hungary
| | - Ágota Apáti
- Institute of Enzymology, Research Centre for Natural SciencesEötvös Loránd Research NetworkBudapestHungary
| | - Sian E. Harding
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Nicola Hellen
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Gabor Foldes
- Heart and Vascular CenterSemmelweis University68 Városmajor StreetBudapestH1122Hungary
- National Heart and Lung InstituteImperial College LondonLondonUK
| |
Collapse
|
61
|
Pollak U, Feinstein Y, Mannarino CN, McBride ME, Mendonca M, Keizman E, Mishaly D, van Leeuwen G, Roeleveld PP, Koers L, Klugman D. The horizon of pediatric cardiac critical care. Front Pediatr 2022; 10:863868. [PMID: 36186624 PMCID: PMC9523119 DOI: 10.3389/fped.2022.863868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/22/2022] [Indexed: 11/21/2022] Open
Abstract
Pediatric Cardiac Critical Care (PCCC) is a challenging discipline where decisions require a high degree of preparation and clinical expertise. In the modern era, outcomes of neonates and children with congenital heart defects have dramatically improved, largely by transformative technologies and an expanding collection of pharmacotherapies. Exponential advances in science and technology are occurring at a breathtaking rate, and applying these advances to the PCCC patient is essential to further advancing the science and practice of the field. In this article, we identified and elaborate on seven key elements within the PCCC that will pave the way for the future.
Collapse
Affiliation(s)
- Uri Pollak
- Section of Pediatric Critical Care, Hadassah University Medical Center, Jerusalem, Israel.,Faculty of Medicine, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yael Feinstein
- Pediatric Intensive Care Unit, Soroka University Medical Center, Be'er Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Candace N Mannarino
- Divisions of Cardiology and Critical Care Medicine, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Mary E McBride
- Divisions of Cardiology and Critical Care Medicine, Departments of Pediatrics and Medical Education, Northwestern University Feinberg School of Medicine, Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Malaika Mendonca
- Pediatric Intensive Care Unit, Children's Hospital, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Eitan Keizman
- Department of Cardiac Surgery, The Leviev Cardiothoracic and Vascular Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - David Mishaly
- Pediatric and Congenital Cardiac Surgery, Edmond J. Safra International Congenital Heart Center, The Chaim Sheba Medical Center, The Edmond and Lily Safra Children's Hospital, Tel Hashomer, Israel
| | - Grace van Leeuwen
- Pediatric Cardiac Intensive Care Unit, Sidra Medicine, Ar-Rayyan, Qatar.,Department of Pediatrics, Weill Cornell Medicine, Ar-Rayyan, Qatar
| | - Peter P Roeleveld
- Department of Pediatric Intensive Care, Leiden University Medical Center, Leiden, Netherlands
| | - Lena Koers
- Department of Pediatric Intensive Care, Leiden University Medical Center, Leiden, Netherlands
| | - Darren Klugman
- Pediatrics Cardiac Critical Care Unit, Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins Medicine, Baltimore, MD, United States
| |
Collapse
|
62
|
Li S, Ma W, Cai B. Targeting cardiomyocyte proliferation as a key approach of promoting heart repair after injury. MOLECULAR BIOMEDICINE 2021; 2:34. [PMID: 35006441 PMCID: PMC8607366 DOI: 10.1186/s43556-021-00047-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 06/21/2021] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular diseases such as myocardial infarction (MI) is a major contributor to human mortality and morbidity. The mammalian adult heart almost loses its plasticity to appreciably regenerate new cardiomyocytes after injuries, such as MI and heart failure. The neonatal heart exhibits robust proliferative capacity when exposed to varying forms of myocardial damage. The ability of the neonatal heart to repair the injury and prevent pathological left ventricular remodeling leads to preserved or improved cardiac function. Therefore, promoting cardiomyocyte proliferation after injuries to reinitiate the process of cardiomyocyte regeneration, and suppress heart failure and other serious cardiovascular problems have become the primary goal of many researchers. Here, we review recent studies in this field and summarize the factors that act upon the proliferation of cardiomyocytes and cardiac repair after injury and discuss the new possibilities for potential clinical treatment strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Shuainan Li
- Department of Pharmacy at The Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Wenya Ma
- Department of Pharmacy at The Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Benzhi Cai
- Department of Pharmacy at The Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China. .,Institute of Clinical Pharmacy, the Heilongjiang Key Laboratory of Drug Research, Harbin Medical University, Harbin, 150086, China. .,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, 150086, China.
| |
Collapse
|
63
|
Abouleisa RRE, McNally L, Salama ABM, Hammad SK, Ou Q, Wells C, Lorkiewicz PK, Bolli R, Mohamed TMA, Hill BG. Cell cycle induction in human cardiomyocytes is dependent on biosynthetic pathway activation. Redox Biol 2021; 46:102094. [PMID: 34418597 PMCID: PMC8379496 DOI: 10.1016/j.redox.2021.102094] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 01/03/2023] Open
Abstract
AIMS The coordinated gene and metabolic programs that facilitate cardiomyocyte entry and progression in the cell cycle are poorly understood. The purpose of this study was to identify the metabolic changes that influence myocyte proliferation. METHODS AND RESULTS In adult mouse cardiomyocytes and human induced pluripotent stem cell cardiomyocytes (hiPS-CMs), cell cycle initiation by ectopic expression of Cyclin B1, Cyclin D1, CDK1, and CDK4 (termed 4F) downregulated oxidative phosphorylation genes and upregulated genes that regulate ancillary biosynthetic pathways of glucose metabolism. Results from metabolic analyses and stable isotope tracing experiments indicate that 4F-mediated cell cycle induction in hiPS-CMs decreases glucose oxidation and oxidative phosphorylation and augments NAD+, glycogen, hexosamine, phospholipid, and serine biosynthetic pathway activity. Interventions that diminish NAD+ synthesis, serine synthesis, or protein O-GlcNAcylation decreased 4F-mediated cell cycle entry. In a gain of function approach, we overexpressed phosphoenolpyruvate carboxykinase 2 (PCK2), which can drive carbon from the Krebs cycle to the glycolytic intermediate pool, and found that PCK2 augments 4F-mediated cell cycle entry. CONCLUSIONS These findings suggest that a metabolic shift from catabolic to anabolic activity is a critical step for cardiomyocyte cell cycle entry and is required to facilitate proliferation.
Collapse
Affiliation(s)
- Riham R E Abouleisa
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Lindsey McNally
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Abou Bakr M Salama
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA; Department of Cardiovascular Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt; Department of Cardiac Surgery, Verona University, Verona, Italy
| | - Sally K Hammad
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA; Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Egypt
| | - Qinghui Ou
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Collin Wells
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Pawel K Lorkiewicz
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA; Department of Chemistry, University of Louisville, KY, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Tamer M A Mohamed
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville, KY, USA; Institute of Cardiovascular Sciences, University of Manchester, UK; Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Egypt.
| | - Bradford G Hill
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
64
|
Meng F, Xie B, Martin JF. Targeting the Hippo pathway in heart repair. Cardiovasc Res 2021; 118:2402-2414. [PMID: 34528077 DOI: 10.1093/cvr/cvab291] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Indexed: 12/17/2022] Open
Abstract
The Hippo pathway is an evolutionarily and functionally conserved signaling pathway that controls organ size by regulating cell proliferation, apoptosis, and differentiation. Emerging evidence has shown that the Hippo pathway plays critical roles in cardiac development, homeostasis, disease, and regeneration. Targeting the Hippo pathway has tremendous potential as a therapeutic strategy for treating intractable cardiovascular diseases such as heart failure. In this review, we summarize the function of the Hippo pathway in the heart. Particularly, we highlight the posttranslational modification of Hippo pathway components, including the core kinases LATS1/2 and their downstream effectors YAP/TAZ, in different contexts, which has provided new insights and avenues in cardiac research.
Collapse
Affiliation(s)
- Fansen Meng
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - Bing Xie
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030.,Texas Heart Institute, Houston, Texas, 77030
| |
Collapse
|
65
|
Arjmand B, Abedi M, Arabi M, Alavi-Moghadam S, Rezaei-Tavirani M, Hadavandkhani M, Tayanloo-Beik A, Kordi R, Roudsari PP, Larijani B. Regenerative Medicine for the Treatment of Ischemic Heart Disease; Status and Future Perspectives. Front Cell Dev Biol 2021; 9:704903. [PMID: 34568321 PMCID: PMC8461329 DOI: 10.3389/fcell.2021.704903] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular disease is now the leading cause of adult death in the world. According to new estimates from the World Health Organization, myocardial infarction (MI) is responsible for four out of every five deaths due to cardiovascular disease. Conventional treatments of MI are taking aspirin and nitroglycerin as intermediate treatments and injecting antithrombotic agents within the first 3 h after MI. Coronary artery bypass grafting and percutaneous coronary intervention are the most common long term treatments. Since none of these interventions will fully regenerate the infarcted myocardium, there is value in pursuing more innovative therapeutic approaches. Regenerative medicine is an innovative interdisciplinary method for rebuilding, replacing, or repairing the missed part of different organs in the body, as similar as possible to the primary structure. In recent years, regenerative medicine has been widely utilized as a treatment for ischemic heart disease (one of the most fatal factors around the world) to repair the lost part of the heart by using stem cells. Here, the development of mesenchymal stem cells causes a breakthrough in the treatment of different cardiovascular diseases. They are easily obtainable from different sources, and expanded and enriched easily, with no need for immunosuppressing agents before transplantation, and fewer possibilities of genetic abnormality accompany them through multiple passages. The production of new cardiomyocytes can result from the transplantation of different types of stem cells. Accordingly, due to its remarkable benefits, stem cell therapy has received attention in recent years as it provides a drug-free and surgical treatment for patients and encourages a more safe and feasible cardiac repair. Although different clinical trials have reported on the promising benefits of stem cell therapy, there is still uncertainty about its mechanism of action. It is important to conduct different preclinical and clinical studies to explore the exact mechanism of action of the cells. After reviewing the pathophysiology of MI, this study addresses the role of tissue regeneration using various materials, including different types of stem cells. It proves some appropriate data about the importance of ethical problems, which leads to future perspectives on this scientific method.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Abedi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Arabi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahdieh Hadavandkhani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Kordi
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyvand Parhizkar Roudsari
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
66
|
Liu S, Tang L, Zhao X, Nguyen B, Heallen TR, Li M, Wang J, Wang J, Martin JF. Yap Promotes Noncanonical Wnt Signals From Cardiomyocytes for Heart Regeneration. Circ Res 2021; 129:782-797. [PMID: 34424032 DOI: 10.1161/circresaha.121.318966] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Shijie Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (S.L., T.R.H., J.F.M.)
| | - Li Tang
- Department of Molecular Physiology and Biophysics (L.T., B.N., J.F.M.), Baylor College of Medicine, One Baylor Plaza, Houston, TX.,School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., Jianxin Wang)
| | - Xiaolei Zhao
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center, Houston (X.Z., J.W.)
| | - Bao Nguyen
- Department of Molecular Physiology and Biophysics (L.T., B.N., J.F.M.), Baylor College of Medicine, One Baylor Plaza, Houston, TX
| | - Todd R Heallen
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (S.L., T.R.H., J.F.M.)
| | | | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., Jianxin Wang)
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center, Houston (X.Z., J.W.)
| | - James F Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (S.L., T.R.H., J.F.M.).,Department of Molecular Physiology and Biophysics (L.T., B.N., J.F.M.), Baylor College of Medicine, One Baylor Plaza, Houston, TX.,Cardiovascular Research Institute (J.F.M.), Baylor College of Medicine, One Baylor Plaza, Houston, TX
| |
Collapse
|
67
|
Record J, Saeed MB, Venit T, Percipalle P, Westerberg LS. Journey to the Center of the Cell: Cytoplasmic and Nuclear Actin in Immune Cell Functions. Front Cell Dev Biol 2021; 9:682294. [PMID: 34422807 PMCID: PMC8375500 DOI: 10.3389/fcell.2021.682294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Actin cytoskeletal dynamics drive cellular shape changes, linking numerous cell functions to physiological and pathological cues. Mutations in actin regulators that are differentially expressed or enriched in immune cells cause severe human diseases known as primary immunodeficiencies underscoring the importance of efficienct actin remodeling in immune cell homeostasis. Here we discuss recent findings on how immune cells sense the mechanical properties of their environement. Moreover, while the organization and biochemical regulation of cytoplasmic actin have been extensively studied, nuclear actin reorganization is a rapidly emerging field that has only begun to be explored in immune cells. Based on the critical and multifaceted contributions of cytoplasmic actin in immune cell functionality, nuclear actin regulation is anticipated to have a large impact on our understanding of immune cell development and functionality.
Collapse
Affiliation(s)
- Julien Record
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Mezida B. Saeed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Tomas Venit
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - Piergiorgio Percipalle
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lisa S. Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
68
|
Liu S, Li K, Wagner Florencio L, Tang L, Heallen TR, Leach JP, Wang Y, Grisanti F, Willerson JT, Perin EC, Zhang S, Martin JF. Gene therapy knockdown of Hippo signaling induces cardiomyocyte renewal in pigs after myocardial infarction. Sci Transl Med 2021; 13:13/600/eabd6892. [PMID: 34193613 DOI: 10.1126/scitranslmed.abd6892] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 04/03/2021] [Accepted: 06/11/2021] [Indexed: 01/03/2023]
Abstract
Human heart failure, a leading cause of death worldwide, is a prominent example of a chronic disease that may result from poor cell renewal. The Hippo signaling pathway is an inhibitory kinase cascade that represses adult heart muscle cell (cardiomyocyte) proliferation and renewal after myocardial infarction in genetically modified mice. Here, we investigated an adeno-associated virus 9 (AAV9)-based gene therapy to locally knock down the Hippo pathway gene Salvador (Sav) in border zone cardiomyocytes in a pig model of ischemia/reperfusion-induced myocardial infarction. Two weeks after myocardial infarction, when pigs had left ventricular systolic dysfunction, we administered AAV9-Sav-short hairpin RNA (shRNA) or a control AAV9 viral vector carrying green fluorescent protein (GFP) directly into border zone cardiomyocytes via catheter-mediated subendocardial injection. Three months after injection, pig hearts treated with a high dose of AAV9-Sav-shRNA exhibited a 14.3% improvement in ejection fraction (a measure of left ventricular systolic function), evidence of cardiomyocyte division, and reduced scar sizes compared to pigs receiving AAV9-GFP. AAV9-Sav-shRNA-treated pig hearts also displayed increased capillary density and reduced cardiomyocyte ploidy. AAV9-Sav-shRNA gene therapy was well tolerated and did not induce mortality. In addition, liver and lung pathology revealed no tumor formation. Local delivery of AAV9-Sav-shRNA gene therapy to border zone cardiomyocytes in pig hearts after myocardial infarction resulted in tissue renewal and improved function and may have utility in treating heart failure.
Collapse
Affiliation(s)
| | - Ke Li
- Texas Heart Institute, Houston, TX, USA
| | | | - Li Tang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | | | - John P Leach
- Department of Medicine, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Francisco Grisanti
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Sui Zhang
- Texas Heart Institute, Houston, TX, USA
| | - James F Martin
- Texas Heart Institute, Houston, TX, USA. .,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA.,Center for Organ Repair and Renewal and Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
69
|
Maldonado-Velez G, Firulli AB. Mechanisms Underlying Cardiomyocyte Development: Can We Exploit Them to Regenerate the Heart? Curr Cardiol Rep 2021; 23:81. [PMID: 34081213 DOI: 10.1007/s11886-021-01510-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW It is well established that the adult mammalian cardiomyocytes retain a low capacity for cell cycle activity; however, it is insufficient to effectively respond to myocardial injury and facilitate cardiac regenerative repair. Lessons learned from species in which cardiomyocytes do allow for proliferative regeneration/repair have shed light into the mechanisms underlying cardiac regeneration post-injury. Importantly, many of these mechanisms are conserved across species, including mammals, and efforts to tap into these mechanisms effectively within the adult heart are currently of great interest. RECENT FINDINGS Targeting the endogenous gene regulatory networks (GRNs) shown to play roles in the cardiac regeneration of conducive species is seen as a strong approach, as delivery of a single or combination of genes has promise to effectively enhance cell cycle activity and CM proliferation in adult hearts post-myocardial infarction (MI). In situ re-induction of proliferative gene regulatory programs within existing, local, non-damaged cardiomyocytes helps overcome significant technical hurdles, such as successful engraftment of implanted cells or achieving complete cardiomyocyte differentiation from cell-based approaches. Although many obstacles currently exist and need to be overcome to successfully translate these approaches to clinical settings, the current efforts presented here show great promise.
Collapse
Affiliation(s)
- Gabriel Maldonado-Velez
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, and Medical and Molecular Genetics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202-5225, USA
| | - Anthony B Firulli
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, and Medical and Molecular Genetics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202-5225, USA.
| |
Collapse
|
70
|
Wagh K, Ishikawa M, Garcia DA, Stavreva DA, Upadhyaya A, Hager GL. Mechanical Regulation of Transcription: Recent Advances. Trends Cell Biol 2021; 31:457-472. [PMID: 33712293 PMCID: PMC8221528 DOI: 10.1016/j.tcb.2021.02.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 01/01/2023]
Abstract
Mechanotransduction is the ability of a cell to sense mechanical cues from its microenvironment and convert them into biochemical signals to elicit adaptive transcriptional and other cellular responses. Here, we describe recent advances in the field of mechanical regulation of transcription, highlight mechanical regulation of the epigenome as a key novel aspect of mechanotransduction, and describe recent technological advances that could further elucidate the link between mechanical stimuli and gene expression. In this review, we emphasize the importance of mechanotransduction as one of the governing principles of cancer progression, underscoring the need to conduct further studies of the molecular mechanisms involved in sensing mechanical cues and coordinating transcriptional responses.
Collapse
Affiliation(s)
- Kaustubh Wagh
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Physics, University of Maryland, College Park, MD 20742, USA
| | - Momoko Ishikawa
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David A Garcia
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Physics, University of Maryland, College Park, MD 20742, USA
| | - Diana A Stavreva
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Arpita Upadhyaya
- Department of Physics, University of Maryland, College Park, MD 20742, USA; Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA.
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
71
|
Kastan N, Gnedeva K, Alisch T, Petelski AA, Huggins DJ, Chiaravalli J, Aharanov A, Shakked A, Tzahor E, Nagiel A, Segil N, Hudspeth AJ. Small-molecule inhibition of Lats kinases may promote Yap-dependent proliferation in postmitotic mammalian tissues. Nat Commun 2021; 12:3100. [PMID: 34035288 PMCID: PMC8149661 DOI: 10.1038/s41467-021-23395-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/20/2021] [Indexed: 02/04/2023] Open
Abstract
Hippo signaling is an evolutionarily conserved pathway that restricts growth and regeneration predominantly by suppressing the activity of the transcriptional coactivator Yap. Using a high-throughput phenotypic screen, we identified a potent and non-toxic activator of Yap. In vitro kinase assays show that the compound acts as an ATP-competitive inhibitor of Lats kinases-the core enzymes in Hippo signaling. The substance prevents Yap phosphorylation and induces proliferation of supporting cells in the murine inner ear, murine cardiomyocytes, and human Müller glia in retinal organoids. RNA sequencing indicates that the inhibitor reversibly activates the expression of transcriptional Yap targets: upon withdrawal, a subset of supporting-cell progeny exits the cell cycle and upregulates genes characteristic of sensory hair cells. Our results suggest that the pharmacological inhibition of Lats kinases may promote initial stages of the proliferative regeneration of hair cells, a process thought to be permanently suppressed in the adult mammalian inner ear.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Cell Line
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Ependymoglial Cells/cytology
- Ependymoglial Cells/drug effects
- Ependymoglial Cells/metabolism
- HEK293 Cells
- Hair Cells, Auditory, Inner/cytology
- Hair Cells, Auditory, Inner/drug effects
- Hair Cells, Auditory, Inner/metabolism
- Humans
- Mice, Knockout
- Mice, Transgenic
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Small Molecule Libraries/pharmacology
- Tumor Suppressor Proteins/antagonists & inhibitors
- Tumor Suppressor Proteins/metabolism
- YAP-Signaling Proteins
- Mice
Collapse
Affiliation(s)
- Nathaniel Kastan
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY, USA
| | - Ksenia Gnedeva
- Tina and Rick Caruso Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angles, CA, USA.
| | - Theresa Alisch
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY, USA
| | - Aleksandra A Petelski
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY, USA
- Department of Bioengineering and Barnett Institute, Northeastern University, Boston, MA, USA
| | - David J Huggins
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Jeanne Chiaravalli
- High-Throughput Screening Resource Center, The Rockefeller University, New York, NY, USA
- Institut Pasteur, Paris, France
| | - Alla Aharanov
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Aaron Nagiel
- Department of Surgery Children's Hospital Los Angeles, Vision Center, Los Angeles, CA, USA
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Neil Segil
- Tina and Rick Caruso Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angles, CA, USA
| | - A J Hudspeth
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY, USA
| |
Collapse
|
72
|
He M, Cheng C, Tu J, Ji SS, Lou D, Bai B. Agrin expression is correlated with tumor development and poor prognosis in cholangiocarcinoma. J Int Med Res 2021; 49:3000605211009722. [PMID: 34018826 PMCID: PMC8150497 DOI: 10.1177/03000605211009722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Objective This study examined the role of agrin in the development of cholangiocarcinoma (CCA). Methods Western blotting was performed to detect the expression of target genes. The correlation between agrin expression and prognosis was analyzed using the Kaplan–Meier method. Proliferation, migration, invasion, and tumorigenesis were examined in CCA cells and tissues using the Cell Counting Kit-8 assay, cell cycle analysis, transwell migration assay, and nude mouse tumorigenicity assay in vivo, respectively. Results Agrin expression was significantly upregulated in CCA tissues compared with that in adjacent non-tumor tissues, and agrin expression was correlated with poorer tumor characteristics such as portal vein tumor thrombus, intrahepatic metastasis, and worse survival. Forced agrin expression in CCA cells apparently promoted proliferation, colony formation, migration, invasion, and cell cycle progression, but agrin depletion had the opposite effects. Furthermore, agrin-depleted CCA cells developed fewer and smaller tumors than control cells in vivo. Mechanistic analyses indicated that agrin activated the Hippo signaling pathway and induced the translocation of YAP to the nucleus. Conclusions Agrin promoted CCA progression by activating the Hippo signaling pathway, suggesting its promise as a target for CCA therapy.
Collapse
Affiliation(s)
- Meimei He
- Department of Pharmacy, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou University, Wenzhou, 325000, Zhejiang Province, China
| | - Chen Cheng
- Department of Pharmacy, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou University, Wenzhou, 325000, Zhejiang Province, China
| | - Junxue Tu
- Department of Pharmacy, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou University, Wenzhou, 325000, Zhejiang Province, China
| | - Sha-Sha Ji
- Department of Pharmacy, Shaoxing Traditional Chinese Medicine Hospital, Shaoxing, Zhejiang Province, China
| | - Dan Lou
- Department of Pharmacy, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou University, Wenzhou, 325000, Zhejiang Province, China
| | - Binglong Bai
- Department of Pharmacy, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou University, Wenzhou, 325000, Zhejiang Province, China
| |
Collapse
|
73
|
Decreased YAP activity reduces proliferative ability in human induced pluripotent stem cell of duchenne muscular dystrophy derived cardiomyocytes. Sci Rep 2021; 11:10351. [PMID: 33990626 PMCID: PMC8121946 DOI: 10.1038/s41598-021-89603-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/20/2021] [Indexed: 11/29/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive muscle degeneration accompanied by dilated cardiomyopathy. Recently, abnormality of yes-associated protein (YAP) has been reported as the pathogenesis of muscle degeneration of DMD; however YAP activity remains unclear in dystrophic heart of DMD. Herein, we investigated YAP activity using disease-specific induced pluripotent stem cell (iPSC) derived cardiomyocytes (CMs) in DMD. DMD-iPSCs were generated from DMD patient with exon 48–54 deletion in DMD, and genome-edited (Ed)-DMD-iPSCs with in-frame (Ed-DMD-iPSCs) were created using CRISPR/Cas9. Nuclear translocation of YAP [nuclear (N)/cytoplasmic (C) ratio] was significantly lower in DMD-iPSC-CMs than in Ed-DMD-iPSC-CMs. In addition, Ki67 expression, indicating proliferative ability, was significantly lower in DMD-iPSC-CMs than Ed-DMD-iPSC-CMs. Therefore, immunofluorescent staining showed that actin stress fibers associated with YAP activity by mechanotransduction were disorganized in DMD-iPSC-CMs. Lysophosphatidic acid (LPA), a known lipid mediator on induction of actin polymerization, significantly increased YAP activity and actin dynamics in DMD-iPSC-CMs using live cell imaging. These results suggested that altered YAP activity due to impaired actin dynamics reduced proliferative ability in DMD-iPSC-CMs. Hence, decreased YAP activity in dystrophic heart may contribute to DMD-cardiomyopathy pathogenesis.
Collapse
|
74
|
Min S, Choe C, Roh S. AQP3 Increases Intercellular Cohesion in NSCLC A549 Cell Spheroids through Exploratory Cell Protrusions. Int J Mol Sci 2021; 22:ijms22084287. [PMID: 33924231 PMCID: PMC8074759 DOI: 10.3390/ijms22084287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/25/2022] Open
Abstract
Tumor cell aggregation is critical for cell survival following the loss of extracellular matrix attachment and dissemination. However, the underlying mechanotransduction of clustering solitary tumor cells is poorly understood, especially in non-small cell lung cancers (NSCLC). Here, we examined whether cell surface protrusions played an important role in facilitating the physical contact between floating cells detached from a substrate. We employed poly-2-hydroxyethyl methacrylate-based 3D culture methods to mimic in vivo tumor cell cluster formation. The suprastructural analysis of human NSCLC A549 cell spheroids showed that finger-like protrusions clung together via the actin cytoskeleton. Time-lapse holotomography demonstrated that the finger-like protrusions of free-floating cells in 3D culture displayed exploratory coalescence. Global gene expression analysis demonstrated that the genes in the organic hydroxyl transport were particularly enriched in the A549 cell spheroids. Particularly, the knockdown of the water channel aquaporin 3 gene (AQP3) impaired multicellular aggregate formation in 3D culture through the rearrangement of the actomyosin cytoskeleton. Moreover, the cells with reduced levels of AQP3 decreased their transmigration. Overall, these data indicate that cell detachment-upregulated AQP3 contributes to cell surface protrusions through actomyosin cytoskeleton remodeling, causing the aggressive aggregation of free-floating cells dependent on the property of the substratum and collective metastasis.
Collapse
Affiliation(s)
- Sol Min
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea;
| | - Chungyoul Choe
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea;
- Samsung Medical Center, Samsung Biomedical Research Institute, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea
- Correspondence: (C.C.); (S.R.); Tel.: +82-221487353 (C.C.); Tel.: +82-28802333 (S.R.)
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea;
- Correspondence: (C.C.); (S.R.); Tel.: +82-221487353 (C.C.); Tel.: +82-28802333 (S.R.)
| |
Collapse
|
75
|
Pagliari S, Vinarsky V, Martino F, Perestrelo AR, Oliver De La Cruz J, Caluori G, Vrbsky J, Mozetic P, Pompeiano A, Zancla A, Ranjani SG, Skladal P, Kytyr D, Zdráhal Z, Grassi G, Sampaolesi M, Rainer A, Forte G. YAP-TEAD1 control of cytoskeleton dynamics and intracellular tension guides human pluripotent stem cell mesoderm specification. Cell Death Differ 2021; 28:1193-1207. [PMID: 33116297 PMCID: PMC8027678 DOI: 10.1038/s41418-020-00643-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/24/2020] [Accepted: 10/08/2020] [Indexed: 12/22/2022] Open
Abstract
The tight regulation of cytoskeleton dynamics is required for a number of cellular processes, including migration, division and differentiation. YAP-TEAD respond to cell-cell interaction and to substrate mechanics and, among their downstream effects, prompt focal adhesion (FA) gene transcription, thus contributing to FA-cytoskeleton stability. This activity is key to the definition of adult cell mechanical properties and function. Its regulation and role in pluripotent stem cells are poorly understood. Human PSCs display a sustained basal YAP-driven transcriptional activity despite they grow in very dense colonies, indicating these cells are insensitive to contact inhibition. PSC inability to perceive cell-cell interactions can be restored by tampering with Tankyrase enzyme, thus favouring AMOT inhibition of YAP function. YAP-TEAD complex is promptly inactivated when germ layers are specified, and this event is needed to adjust PSC mechanical properties in response to physiological substrate stiffness. By providing evidence that YAP-TEAD1 complex targets key genes encoding for proteins involved in cytoskeleton dynamics, we suggest that substrate mechanics can direct PSC specification by influencing cytoskeleton arrangement and intracellular tension. We propose an aberrant activation of YAP-TEAD1 axis alters PSC potency by inhibiting cytoskeleton dynamics, thus paralyzing the changes in shape requested for the acquisition of the given phenotype.
Collapse
Affiliation(s)
- Stefania Pagliari
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic.
| | - Vladimir Vinarsky
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, CZ-62500, Brno, Czech Republic
| | - Fabiana Martino
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
- Faculty of Medicine, Department of Biology, Masaryk University, CZ-62500, Brno, Czech Republic
| | - Ana Rubina Perestrelo
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
| | - Jorge Oliver De La Cruz
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, CZ-62500, Brno, Czech Republic
| | - Guido Caluori
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, CZ-62500, Brno, Czech Republic
| | - Jan Vrbsky
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
| | - Pamela Mozetic
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
| | - Antonio Pompeiano
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
| | | | - Sri Ganji Ranjani
- Central European Institute of Technology, Masaryk University, CZ-62500, Brno, Czech Republic
| | - Petr Skladal
- Central European Institute of Technology, Masaryk University, CZ-62500, Brno, Czech Republic
| | - Dan Kytyr
- Czech Academy of Sciences, Institute of Theoretical and Applied Mechanics, 190 00, Prague 9, Czech Republic
| | - Zbyněk Zdráhal
- Central European Institute of Technology, Masaryk University, CZ-62500, Brno, Czech Republic
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, I-34149, Trieste, Italy
| | - Maurilio Sampaolesi
- Department of Development and Regeneration, KU Leuven, 3000, Leuven, Belgium
| | - Alberto Rainer
- Università Campus Bio-Medico di Roma, Rome, Italy
- Institute of Nanotechnology (NANOTEC), National Research Council, c/o Campus EcoTekne, via Monteroni, 73100, Lecce, Italy
| | - Giancarlo Forte
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic.
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, CZ-62500, Brno, Czech Republic.
- Department of Biomaterials Science, Institute of Dentistry, University of Turku, FI-20014, Turku, Finland.
| |
Collapse
|
76
|
Bonnard C, Navaratnam N, Ghosh K, Chan PW, Tan TT, Pomp O, Ng AYJ, Tohari S, Changede R, Carling D, Venkatesh B, Altunoglu U, Kayserili H, Reversade B. A loss-of-function NUAK2 mutation in humans causes anencephaly due to impaired Hippo-YAP signaling. J Exp Med 2021; 217:152044. [PMID: 32845958 PMCID: PMC7953732 DOI: 10.1084/jem.20191561] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/21/2020] [Accepted: 05/19/2020] [Indexed: 01/18/2023] Open
Abstract
Failure of neural tube closure during embryonic development can result in anencephaly, one of the most common birth defects in humans. A family with recurrent anencephalic fetuses was investigated to understand its etiology and pathogenesis. Exome sequencing revealed a recessive germline 21-bp in-frame deletion in NUAK2 segregating with the disease. In vitro kinase assays demonstrated that the 7–amino acid truncation in NUAK2, a serine/threonine kinase, completely abrogated its catalytic activity. Patient-derived disease models including neural progenitor cells and cerebral organoids showed that loss of NUAK2 activity led to decreased Hippo signaling via cytoplasmic YAP retention. In neural tube–like structures, endogenous NUAK2 colocalized apically with the actomyosin network, which was disrupted in patient cells, causing impaired nucleokinesis and apical constriction. Our results establish NUAK2 as an indispensable kinase for brain development in humans and suggest that a NUAK2-Hippo signaling axis regulates cytoskeletal processes that govern cell shape during neural tube closure.
Collapse
Affiliation(s)
- Carine Bonnard
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Naveenan Navaratnam
- Medical Research Council London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Kakaly Ghosh
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Puck Wee Chan
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Thong Teck Tan
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Oz Pomp
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Alvin Yu Jin Ng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Sumanty Tohari
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Rishita Changede
- Mechanobiology Institute, National University of Singapore, Singapore
| | - David Carling
- Medical Research Council London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Byrappa Venkatesh
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Paediatrics, National University of Singapore, Singapore
| | - Umut Altunoglu
- Medical Genetics Department, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey.,Medical Genetics Department, Koç University School of Medicine, Istanbul, Turkey
| | - Hülya Kayserili
- Medical Genetics Department, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey.,Medical Genetics Department, Koç University School of Medicine, Istanbul, Turkey
| | - Bruno Reversade
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Paediatrics, National University of Singapore, Singapore.,Medical Genetics Department, Koç University School of Medicine, Istanbul, Turkey
| |
Collapse
|
77
|
Valera IC, Wacker AL, Hwang HS, Holmes C, Laitano O, Landstrom AP, Parvatiyar MS. Essential roles of the dystrophin-glycoprotein complex in different cardiac pathologies. Adv Med Sci 2021; 66:52-71. [PMID: 33387942 DOI: 10.1016/j.advms.2020.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 12/20/2022]
Abstract
The dystrophin-glycoprotein complex (DGC), situated at the sarcolemma dynamically remodels during cardiac disease. This review examines DGC remodeling as a common denominator in diseases affecting heart function and health. Dystrophin and the DGC serve as broad cytoskeletal integrators that are critical for maintaining stability of muscle membranes. The presence of pathogenic variants in genes encoding proteins of the DGC can cause absence of the protein and/or alterations in other complex members leading to muscular dystrophies. Targeted studies have allowed the individual functions of affected proteins to be defined. The DGC has demonstrated its dynamic function, remodeling under a number of conditions that stress the heart. Beyond genetic causes, pathogenic processes also impinge on the DGC, causing alterations in the abundance of dystrophin and associated proteins during cardiac insult such as ischemia-reperfusion injury, mechanical unloading, and myocarditis. When considering new therapeutic strategies, it is important to assess DGC remodeling as a common factor in various heart diseases. The DGC connects the internal F-actin-based cytoskeleton to laminin-211 of the extracellular space, playing an important role in the transmission of mechanical force to the extracellular matrix. The essential functions of dystrophin and the DGC have been long recognized. DGC based therapeutic approaches have been primarily focused on muscular dystrophies, however it may be a beneficial target in a number of disorders that affect the heart. This review provides an account of what we now know, and discusses how this knowledge can benefit persistent health conditions in the clinic.
Collapse
Affiliation(s)
- Isela C Valera
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Amanda L Wacker
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Hyun Seok Hwang
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Christina Holmes
- Department of Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, Tallahassee, FL, USA
| | - Orlando Laitano
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA; Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Michelle S Parvatiyar
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
78
|
Shen H, Darehzereshki A, Sucov HM, Lien CL. Apical Resection and Cryoinjury of Neonatal Mouse Heart. Methods Mol Biol 2021; 2158:23-32. [PMID: 32857362 DOI: 10.1007/978-1-0716-0668-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2024]
Abstract
Neonatal mouse hearts have a regenerative capacity similar to adult zebrafish. Different cardiac injury models have been established to investigate the regenerative capacity of neonatal mouse hearts, including ventricular amputation, cryoinjury, and ligation of a major coronary artery. While the ventricular resection model can be utilized to study how tissue forms and regenerates de novo, cryoinjury and coronary artery ligation are methods that might better mimic myocardial infarction by creating tissue damage and necrosis as opposed to the removal of healthy tissue in the ventricular amputation model. Here we describe methods of creating ventricular resection and cardiac cryoinjury in newborn mice.
Collapse
Affiliation(s)
- Hua Shen
- Department of Stem Cell Biology and Regenerative Medicine, Broad CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ali Darehzereshki
- Saban Research Institute and Heart Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Surgery, MedStar Health Baltimore, Franklin Square Medical Center, Baltimore, MD, USA
| | - Henry M Sucov
- Department of Stem Cell Biology and Regenerative Medicine, Broad CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Ching-Ling Lien
- Saban Research Institute and Heart Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Surgery, Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
79
|
Pullamsetti SS, Günther A. A FOX-like Mechanism Regulating Lung Fibroblasts: Are We Getting There? Am J Respir Cell Mol Biol 2020; 63:723-724. [PMID: 33091319 PMCID: PMC7790149 DOI: 10.1165/rcmb.2020-0441ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Soni Savai Pullamsetti
- University of Giessen and Marburg Lung Center Justus-Liebig University Giessen Giessen, Germany.,Max Planck Institute for Heart and Lung Research Bad Nauheim, Germany
| | - Andreas Günther
- Center for Interstitial and Rare Lung Diseases Justus-Liebig University Giessen Giessen, Germany.,Agaplesion Lung Clinic Waldhof-Elgershausen Greifenstein, Germany and.,European Idiopathic Pulmonary Fibrosis Registry and Biobank Gießen, Germany
| |
Collapse
|
80
|
Zhu Y, Do VD, Richards AM, Foo R. What we know about cardiomyocyte dedifferentiation. J Mol Cell Cardiol 2020; 152:80-91. [PMID: 33275936 DOI: 10.1016/j.yjmcc.2020.11.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/22/2020] [Accepted: 11/27/2020] [Indexed: 12/16/2022]
Abstract
Cardiomyocytes (CMs) lost during cardiac injury and heart failure (HF) cannot be replaced due to their limited proliferative capacity. Regenerating the failing heart by promoting CM cell-cycle re-entry is an ambitious solution, currently vigorously pursued. Some genes have been proven to promote endogenous CM proliferation, believed to be preceded by CM dedifferentiation, wherein terminally differentiated CMs are initially reversed back to the less mature state which precedes cell division. However, very little else is known about CM dedifferentiation which remains poorly defined. We lack robust molecular markers and proper understanding of the mechanisms driving dedifferentiation. Even the term dedifferentiation is debated because there is no objective evidence of pluripotency, and could rather reflect CM plasticity instead. Nonetheless, the significance of CM transition states on cardiac function, and whether they necessarily lead to CM proliferation, remains unclear. This review summarises the current state of knowledge of both natural and experimentally induced CM dedifferentiation in non-mammalian vertebrates (primarily the zebrafish) and mammals, as well as the phenotypes and molecular mechanisms involved. The significance and potential challenges of studying CM dedifferentiation are also discussed. In summary, CM dedifferentiation, essential for CM plasticity, may have an important role in heart regeneration, thereby contributing to the prevention and treatment of heart disease. More attention is needed in this field to overcome the technical limitations and knowledge gaps.
Collapse
Affiliation(s)
- Yike Zhu
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cardiovascular Disease Translational Research Programme, National University Health Systems, Singapore; Genome Institute of Singapore, Agency of Science Research and Technology, Singapore
| | - Vinh Dang Do
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cardiovascular Disease Translational Research Programme, National University Health Systems, Singapore; Genome Institute of Singapore, Agency of Science Research and Technology, Singapore
| | - A Mark Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cardiovascular Disease Translational Research Programme, National University Health Systems, Singapore
| | - Roger Foo
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cardiovascular Disease Translational Research Programme, National University Health Systems, Singapore; Genome Institute of Singapore, Agency of Science Research and Technology, Singapore.
| |
Collapse
|
81
|
Knyazeva A, Khudiakov A, Vaz R, Muravyev A, Sukhareva K, Sejersen T, Kostareva A. FLNC Expression Level Influences the Activity of TEAD-YAP/TAZ Signaling. Genes (Basel) 2020; 11:genes11111343. [PMID: 33202721 PMCID: PMC7696573 DOI: 10.3390/genes11111343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Filamin C (FLNC), being one of the major actin-binding proteins, is involved in the maintenance of key muscle cell functions. Inherited skeletal muscle and cardiac disorders linked to genetic variants in FLNC have attracted attention because of their high clinical importance and possibility of genotype-phenotype correlations. To further expand on the role of FLNC in muscle cells, we focused on detailed alterations of muscle cell properties developed after the loss of FLNC. Using the CRISPR/Cas9 method we generated a C2C12 murine myoblast cell line with stably suppressed Flnc expression. FLNC-deficient myoblasts have a significantly higher proliferation rate combined with an impaired cell migration capacity. The suppression of Flnc expression leads to inability to complete myogenic differentiation, diminished expression of Myh1 and Myh4, alteration of transcriptional dynamics of myogenic factors, such as Mymk and Myog, and deregulation of Hippo signaling pathway. Specifically, we identified elevated basal levels of Hippo activity in myoblasts with loss of FLNC, and ineffective reduction of Hippo signaling activity during myogenic differentiation. The latter was restored by Flnc overexpression. In summary, we confirmed the role of FLNC in muscle cell proliferation, migration and differentiation, and demonstrated for the first time the direct link between Flnc expression and activity of TEAD-YAP\TAZ signaling. These findings support a role of FLNC in regulation of essential muscle processes relying on mechanical as well as signaling mechanisms.
Collapse
Affiliation(s)
- Anastasia Knyazeva
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (A.K.); (A.M.); (K.S.); (A.K.)
- Correspondence:
| | - Aleksandr Khudiakov
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (A.K.); (A.M.); (K.S.); (A.K.)
| | - Raquel Vaz
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institute, 171 76 Stockholm, Sweden;
| | - Aleksey Muravyev
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (A.K.); (A.M.); (K.S.); (A.K.)
| | - Ksenia Sukhareva
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (A.K.); (A.M.); (K.S.); (A.K.)
- Graduate School of Life and Health Science, University of Verona, 10 37134 Verona, Italy
| | - Thomas Sejersen
- Department of Women’s and Children’s Health, Karolinska Institute, 171 77 Stockholm, Sweden;
| | - Anna Kostareva
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (A.K.); (A.M.); (K.S.); (A.K.)
- Department of Women’s and Children’s Health, Karolinska Institute, 171 77 Stockholm, Sweden;
| |
Collapse
|
82
|
George RM, Maldonado-Velez G, Firulli AB. The heart of the neural crest: cardiac neural crest cells in development and regeneration. Development 2020; 147:147/20/dev188706. [PMID: 33060096 DOI: 10.1242/dev.188706] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiac neural crest cells (cNCCs) are a migratory cell population that stem from the cranial portion of the neural tube. They undergo epithelial-to-mesenchymal transition and migrate through the developing embryo to give rise to portions of the outflow tract, the valves and the arteries of the heart. Recent lineage-tracing experiments in chick and zebrafish embryos have shown that cNCCs can also give rise to mature cardiomyocytes. These cNCC-derived cardiomyocytes appear to be required for the successful repair and regeneration of injured zebrafish hearts. In addition, recent work examining the response to cardiac injury in the mammalian heart has suggested that cNCC-derived cardiomyocytes are involved in the repair/regeneration mechanism. However, the molecular signature of the adult cardiomyocytes involved in this repair is unclear. In this Review, we examine the origin, migration and fates of cNCCs. We also review the contribution of cNCCs to mature cardiomyocytes in fish, chick and mice, as well as their role in the regeneration of the adult heart.
Collapse
Affiliation(s)
- Rajani M George
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | - Gabriel Maldonado-Velez
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | - Anthony B Firulli
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| |
Collapse
|
83
|
ERBB2 drives YAP activation and EMT-like processes during cardiac regeneration. Nat Cell Biol 2020; 22:1346-1356. [PMID: 33046882 DOI: 10.1038/s41556-020-00588-4] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 09/01/2020] [Indexed: 01/06/2023]
Abstract
Cardiomyocyte loss after injury results in adverse remodelling and fibrosis, inevitably leading to heart failure. The ERBB2-Neuregulin and Hippo-YAP signalling pathways are key mediators of heart regeneration, yet the crosstalk between them is unclear. We demonstrate that transient overexpression of activated ERBB2 in cardiomyocytes (OE CMs) promotes cardiac regeneration in a heart failure model. OE CMs present an epithelial-mesenchymal transition (EMT)-like regenerative response manifested by cytoskeletal remodelling, junction dissolution, migration and extracellular matrix turnover. We identified YAP as a critical mediator of ERBB2 signalling. In OE CMs, YAP interacts with nuclear-envelope and cytoskeletal components, reflecting an altered mechanical state elicited by ERBB2. We identified two YAP-activating phosphorylations on S352 and S274 in OE CMs, which peak during metaphase, that are ERK dependent and Hippo independent. Viral overexpression of YAP phospho-mutants dampened the proliferative competence of OE CMs. Together, we reveal a potent ERBB2-mediated YAP mechanotransduction signalling, involving EMT-like characteristics, resulting in robust heart regeneration.
Collapse
|
84
|
Fan X, Shan X, Jiang S, Wang S, Zhang F, Tian Q, Chen D, Ma J, Xue F, Mao S, Fan J, Wang Y, Gong Y. YAP promotes endothelial barrier repair by repressing STAT3/VEGF signaling. Life Sci 2020; 256:117884. [DOI: 10.1016/j.lfs.2020.117884] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 01/28/2023]
|
85
|
Fukuda R, Marín‐Juez R, El‐Sammak H, Beisaw A, Ramadass R, Kuenne C, Guenther S, Konzer A, Bhagwat AM, Graumann J, Stainier DYR. Stimulation of glycolysis promotes cardiomyocyte proliferation after injury in adult zebrafish. EMBO Rep 2020; 21:e49752. [PMID: 32648304 PMCID: PMC7403660 DOI: 10.15252/embr.201949752] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/11/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac metabolism plays a crucial role in producing sufficient energy to sustain cardiac function. However, the role of metabolism in different aspects of cardiomyocyte regeneration remains unclear. Working with the adult zebrafish heart regeneration model, we first find an increase in the levels of mRNAs encoding enzymes regulating glucose and pyruvate metabolism, including pyruvate kinase M1/2 (Pkm) and pyruvate dehydrogenase kinases (Pdks), especially in tissues bordering the damaged area. We further find that impaired glycolysis decreases the number of proliferating cardiomyocytes following injury. These observations are supported by analyses using loss-of-function models for the metabolic regulators Pkma2 and peroxisome proliferator-activated receptor gamma coactivator 1 alpha. Cardiomyocyte-specific loss- and gain-of-function manipulations of pyruvate metabolism using Pdk3 as well as a catalytic subunit of the pyruvate dehydrogenase complex (PDC) reveal its importance in cardiomyocyte dedifferentiation and proliferation after injury. Furthermore, we find that PDK activity can modulate cell cycle progression and protrusive activity in mammalian cardiomyocytes in culture. Our findings reveal new roles for cardiac metabolism and the PDK-PDC axis in cardiomyocyte behavior following cardiac injury.
Collapse
Affiliation(s)
- Ryuichi Fukuda
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- The German Centre for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Rubén Marín‐Juez
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- The German Centre for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Hadil El‐Sammak
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- The German Centre for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Arica Beisaw
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- The German Centre for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Radhan Ramadass
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- The German Centre for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Carsten Kuenne
- The Cardio‐Pulmonary Institute (CPI) and Deep Sequencing PlatformBad NauheimGermany
| | - Stefan Guenther
- The Cardio‐Pulmonary Institute (CPI) and Deep Sequencing PlatformBad NauheimGermany
- Bioinformatics and Deep Sequencing PlatformMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Anne Konzer
- The German Centre for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Biomolecular Mass SpectrometryMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Aditya M Bhagwat
- The German Centre for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Biomolecular Mass SpectrometryMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Johannes Graumann
- The German Centre for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Biomolecular Mass SpectrometryMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Didier YR Stainier
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- The German Centre for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| |
Collapse
|
86
|
Selig M, Lauer JC, Hart ML, Rolauffs B. Mechanotransduction and Stiffness-Sensing: Mechanisms and Opportunities to Control Multiple Molecular Aspects of Cell Phenotype as a Design Cornerstone of Cell-Instructive Biomaterials for Articular Cartilage Repair. Int J Mol Sci 2020; 21:E5399. [PMID: 32751354 PMCID: PMC7432012 DOI: 10.3390/ijms21155399] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/23/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Since material stiffness controls many cell functions, we reviewed the currently available knowledge on stiffness sensing and elucidated what is known in the context of clinical and experimental articular cartilage (AC) repair. Remarkably, no stiffness information on the various biomaterials for clinical AC repair was accessible. Using mRNA expression profiles and morphology as surrogate markers of stiffness-related effects, we deduced that the various clinically available biomaterials control chondrocyte (CH) phenotype well, but not to equal extents, and only in non-degenerative settings. Ample evidence demonstrates that multiple molecular aspects of CH and mesenchymal stromal cell (MSC) phenotype are susceptible to material stiffness, because proliferation, migration, lineage determination, shape, cytoskeletal properties, expression profiles, cell surface receptor composition, integrin subunit expression, and nuclear shape and composition of CHs and/or MSCs are stiffness-regulated. Moreover, material stiffness modulates MSC immuno-modulatory and angiogenic properties, transforming growth factor beta 1 (TGF-β1)-induced lineage determination, and CH re-differentiation/de-differentiation, collagen type II fragment production, and TGF-β1- and interleukin 1 beta (IL-1β)-induced changes in cell stiffness and traction force. We then integrated the available molecular signaling data into a stiffness-regulated CH phenotype model. Overall, we recommend using material stiffness for controlling cell phenotype, as this would be a promising design cornerstone for novel future-oriented, cell-instructive biomaterials for clinical high-quality AC repair tissue.
Collapse
Affiliation(s)
- Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Jasmin C. Lauer
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Melanie L. Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| |
Collapse
|
87
|
Common Regulatory Pathways Mediate Activity of MicroRNAs Inducing Cardiomyocyte Proliferation. Cell Rep 2020; 27:2759-2771.e5. [PMID: 31141697 PMCID: PMC6547019 DOI: 10.1016/j.celrep.2019.05.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 05/03/2018] [Accepted: 04/30/2019] [Indexed: 01/04/2023] Open
Abstract
Loss of functional cardiomyocytes is a major determinant of heart failure after myocardial infarction. Previous high throughput screening studies have identified a few microRNAs (miRNAs) that can induce cardiomyocyte proliferation and stimulate cardiac regeneration in mice. Here, we show that all of the most effective of these miRNAs activate nuclear localization of the master transcriptional cofactor Yes-associated protein (YAP) and induce expression of YAP-responsive genes. In particular, miR-199a-3p directly targets two mRNAs coding for proteins impinging on the Hippo pathway, the upstream YAP inhibitory kinase TAOK1, and the E3 ubiquitin ligase β-TrCP, which leads to YAP degradation. Several of the pro-proliferative miRNAs (including miR-199a-3p) also inhibit filamentous actin depolymerization by targeting Cofilin2, a process that by itself activates YAP nuclear translocation. Thus, activation of YAP and modulation of the actin cytoskeleton are major components of the pro-proliferative action of miR-199a-3p and other miRNAs that induce cardiomyocyte proliferation. A few microRNAs can stimulate cardiac myocyte proliferation The most effective of these microRNAs activate YAP Several pro-proliferative microRNAs also inhibit actin depolymerization miR-199a-3p directly targets TAOK1, b-TrCP, and Cofilin2 to achieve its effects
Collapse
|
88
|
Bergeron A, Brezai A, Shukr R, Villeneuve L, Allen BG, Qureshi WMS, Hentges KE, Calderone A. Filamentous nestin and nonmuscle myosin IIB are associated with a migratory phenotype in neonatal rat cardiomyocytes. J Cell Physiol 2020; 236:1281-1294. [PMID: 32654195 DOI: 10.1002/jcp.29934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/28/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022]
Abstract
Cardiomyocyte migration represents a requisite event of cardiogenesis and the regenerative response of the injured adult zebrafish and neonatal rodent heart. The present study tested the hypothesis that the appearance of the intermediate filament protein nestin in neonatal rat ventricular cardiomyocytes (NNVMs) was associated in part with the acquisition of a migratory phenotype. The cotreatment of NNVMs with phorbol 12,13-dibutyrate (PDBu) and the p38α/β mitogen-activated protein kinase inhibitor SB203580 led to the de novo synthesis of nestin. The intermediate filament protein was subsequently reorganized into a filamentous pattern and redistributed to the leading edge of elongated membrane protrusions translating to significant lengthening of NNVMs. PDBu/SB203580 treatment concomitantly promoted the reorganization of nonmuscle myosin IIB (NMIIB) located predominantly at the periphery of the plasma membrane of NNVMs to a filamentous phenotype extending to the leading edge of elongated membrane protrusions. Coimmunoprecipitation assay revealed a physical interaction between NMIIB and nestin after PDBu/SB203580 treatment of NNVMs. In wild-type and heterozygous NMIIB embryonic hearts at E11.5-E14.5 days, nestin immunoreactivity was identified in a subpopulation of cardiomyocytes elongating perpendicular to the compact myocardium, at the leading edge of nascent trabeculae and during thickening of the compact myocardium. In mutant embryonic hearts lacking NMIIB protein expression, trabeculae formation was reduced, the compact myocardium significantly thinner and nestin immunoreactivity undetectable in cardiomyocytes at E14.5 days. These data suggest that NMIIB and nestin may act in a coordinated fashion to facilitate the acquisition of a migratory phenotype in neonatal and embryonic cardiomyocytes.
Collapse
Affiliation(s)
- Alexandre Bergeron
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Andra Brezai
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Rami Shukr
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Bruce G Allen
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Wasay M S Qureshi
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Kathryn E Hentges
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Angelino Calderone
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
89
|
Targeting the Hippo pathway in cancer, fibrosis, wound healing and regenerative medicine. Nat Rev Drug Discov 2020; 19:480-494. [PMID: 32555376 DOI: 10.1038/s41573-020-0070-z] [Citation(s) in RCA: 539] [Impact Index Per Article: 107.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2020] [Indexed: 02/07/2023]
Abstract
The Hippo pathway is an evolutionarily conserved signalling pathway with key roles in organ development, epithelial homeostasis, tissue regeneration, wound healing and immune modulation. Many of these roles are mediated by the transcriptional effectors YAP and TAZ, which direct gene expression via control of the TEAD family of transcription factors. Dysregulated Hippo pathway and YAP/TAZ-TEAD activity is associated with various diseases, most notably cancer, making this pathway an attractive target for therapeutic intervention. This Review highlights the key findings from studies of Hippo pathway signalling across biological processes and diseases, and discusses new strategies and therapeutic implications of targeting this pathway.
Collapse
|
90
|
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
|
91
|
Xiao D, Chang W, Ding W, Wang Y, Fa H, Wang J. Enhanced mitophagy mediated by the YAP/Parkin pathway protects against DOX-induced cardiotoxicity. Toxicol Lett 2020; 330:96-107. [PMID: 32434049 DOI: 10.1016/j.toxlet.2020.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
The clinical usage of Doxorubicin (DOX) is limited due to its cardiotoxicity. Although the precise mechanism remains unclear, there is an increasing body of evidence that has demonstrated that mitophagy is responsible for DOX-induced cardiotoxicity. In the present study, Parkin, a key protein for mitophagy initiation, was revealed to be downregulated in mouse hearts and in H9c2 cells upon DOX treatment. Enforced expression of Parkin led to mitophagy activation and attenuated cell apoptosis in H9c2 cells. Parkin transgenic mice inhibited DOX-induced cardiotoxicity. Furthermore, Yes-associatd protein, as a transcription co-activator, regulated the gene expression of Parkin, and in turn Parkin overexpression protected against cell apoptosis induced by DOX treatment. Taken together, enhanced mitophagy mediated by YAP/Parkin pathway protects against DOX-induced cardiotoxicity in mouse heart. These studies revealed the complex pathological process of DOX-induced cardiotoxicity and provided novel insight into potential chemotherapy targets.
Collapse
Affiliation(s)
- Dandan Xiao
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China; Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China.
| | - Wenguang Chang
- Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China.
| | - Wei Ding
- Affiliated Hospital, Qingdao University, Qingdao, 266003, China.
| | - Yu Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Hongge Fa
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China; Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
92
|
Beisaw A, Kuenne C, Guenther S, Dallmann J, Wu CC, Bentsen M, Looso M, Stainier DYR. AP-1 Contributes to Chromatin Accessibility to Promote Sarcomere Disassembly and Cardiomyocyte Protrusion During Zebrafish Heart Regeneration. Circ Res 2020; 126:1760-1778. [PMID: 32312172 DOI: 10.1161/circresaha.119.316167] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
RATIONALE The adult human heart is an organ with low regenerative potential. Heart failure following acute myocardial infarction is a leading cause of death due to the inability of cardiomyocytes to proliferate and replenish lost cardiac muscle. While the zebrafish has emerged as a powerful model to study endogenous cardiac regeneration, the molecular mechanisms by which cardiomyocytes respond to damage by disassembling sarcomeres, proliferating, and repopulating the injured area remain unclear. Furthermore, we are far from understanding the regulation of the chromatin landscape and epigenetic barriers that must be overcome for cardiac regeneration to occur. OBJECTIVE To identify transcription factor regulators of the chromatin landscape, which promote cardiomyocyte regeneration in zebrafish, and investigate their function. METHODS AND RESULTS Using the Assay for Transposase-Accessible Chromatin coupled to high-throughput sequencing (ATAC-Seq), we first find that the regenerating cardiomyocyte chromatin accessibility landscape undergoes extensive changes following cryoinjury, and that activator protein-1 (AP-1) binding sites are the most highly enriched motifs in regions that gain accessibility during cardiac regeneration. Furthermore, using bioinformatic and gene expression analyses, we find that the AP-1 response in regenerating adult zebrafish cardiomyocytes is largely different from the response in adult mammalian cardiomyocytes. Using a cardiomyocyte-specific dominant negative approach, we show that blocking AP-1 function leads to defects in cardiomyocyte proliferation as well as decreased chromatin accessibility at the fbxl22 and ilk loci, which regulate sarcomere disassembly and cardiomyocyte protrusion into the injured area, respectively. We further show that overexpression of the AP-1 family members Junb and Fosl1 can promote changes in mammalian cardiomyocyte behavior in vitro. CONCLUSIONS AP-1 transcription factors play an essential role in the cardiomyocyte response to injury by regulating chromatin accessibility changes, thereby allowing the activation of gene expression programs that promote cardiomyocyte dedifferentiation, proliferation, and protrusion into the injured area.
Collapse
Affiliation(s)
- Arica Beisaw
- From the Department of Developmental Genetics (A.B., J.D., C.-C.W., D.Y.R.S.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main (A.B., S.G., D.Y.R.S.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Carsten Kuenne
- ECCPS Bioinformatics and Deep Sequencing Platform (C.K., S.G., M.B., M.L.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Guenther
- ECCPS Bioinformatics and Deep Sequencing Platform (C.K., S.G., M.B., M.L.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main (A.B., S.G., D.Y.R.S.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Julia Dallmann
- From the Department of Developmental Genetics (A.B., J.D., C.-C.W., D.Y.R.S.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Chi-Chung Wu
- From the Department of Developmental Genetics (A.B., J.D., C.-C.W., D.Y.R.S.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mette Bentsen
- ECCPS Bioinformatics and Deep Sequencing Platform (C.K., S.G., M.B., M.L.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mario Looso
- ECCPS Bioinformatics and Deep Sequencing Platform (C.K., S.G., M.B., M.L.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Didier Y R Stainier
- From the Department of Developmental Genetics (A.B., J.D., C.-C.W., D.Y.R.S.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main (A.B., S.G., D.Y.R.S.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
93
|
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
|
94
|
Heallen TR, Kadow ZA, Wang J, Martin JF. Determinants of Cardiac Growth and Size. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037150. [PMID: 31615785 DOI: 10.1101/cshperspect.a037150] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Within the realm of zoological study, the question of how an organism reaches a specific size has been largely unexplored. Recently, studies performed to understand the regulation of organ size have revealed that both cellular signals and external cues contribute toward the determination of total cell mass within each organ. The establishment of final organ size requires the precise coordination of cell growth, proliferation, and survival throughout development and postnatal life. In the mammalian heart, the regulation of size is biphasic. During development, cardiomyocyte proliferation predominantly determines cardiac growth, whereas in the adult heart, total cell mass is governed by signals that regulate cardiac hypertrophy. Here, we review the current state of knowledge regarding the extrinsic factors and intrinsic mechanisms that control heart size during development. We also discuss the metabolic switch that occurs in the heart after birth and precedes homeostatic control of postnatal heart size.
Collapse
Affiliation(s)
- Todd R Heallen
- Cardiomyocyte Renewal Lab, Texas Heart Institute, Houston, Texas 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Zachary A Kadow
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - James F Martin
- Cardiomyocyte Renewal Lab, Texas Heart Institute, Houston, Texas 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
95
|
Lai WF, Wong WT. Roles of the actin cytoskeleton in aging and age-associated diseases. Ageing Res Rev 2020; 58:101021. [PMID: 31968269 DOI: 10.1016/j.arr.2020.101021] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/06/2020] [Accepted: 01/17/2020] [Indexed: 12/12/2022]
Abstract
The integrity of the cytoskeleton is essential to diverse cellular processes such as phagocytosis and intracellular trafficking. Disruption of the organization and dynamics of the actin cytoskeleton leads to age-associated symptoms and diseases, ranging from cancer to neurodegeneration. In addition, changes in the integrity of the actin cytoskeleton disrupt the functioning of not only somatic and stem cells but also gametes, resulting in aberrant embryonic development. Strategies to preserve the integrity and dynamics of the cytoskeleton are, therefore, potentially therapeutic to age-related disorders. The objective of this article is to revisit the current understanding of the roles played by the actin cytoskeleton in aging, and to review the opportunities and challenges for the transition of basic research into intervention development. It is hoped that, with the snapshot of evidence regarding changes in actin dynamics with advanced age, insights into future research directions can be attained.
Collapse
Affiliation(s)
- Wing-Fu Lai
- School of Pharmaceutical Sciences, Shenzhen University, PR China; School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, PR China; Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, PR China.
| | - Wing-Tak Wong
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, PR China
| |
Collapse
|
96
|
Ali H, Braga L, Giacca M. Cardiac regeneration and remodelling of the cardiomyocyte cytoarchitecture. FEBS J 2020; 287:417-438. [PMID: 31743572 DOI: 10.1111/febs.15146] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/27/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Abstract
Adult mammals are unable to regenerate their hearts after cardiac injury, largely due to the incapacity of cardiomyocytes (CMs) to undergo cell division. However, mammalian embryonic and fetal CMs, similar to CMs from fish and amphibians during their entire life, exhibit robust replicative activity, which stops abruptly after birth and never significantly resumes. Converging evidence indicates that formation of the highly ordered and stable cytoarchitecture of mammalian mature CMs is coupled with loss of their proliferative potential. Here, we review the available information on the role of the cardiac cytoskeleton and sarcomere in the regulation of CM proliferation. The actin cytoskeleton, the intercalated disc, the microtubular network and the dystrophin-glycoprotein complex each sense mechanical cues from the surrounding environment. Furthermore, they participate in the regulation of CM proliferation by impinging on the yes-associated protein/transcriptional co-activator with PDZ-binding motif, β-catenin and myocardin-related transcription factor transcriptional co-activators. Mastering the molecular mechanisms regulating CM proliferation would permit the development of innovative strategies to stimulate cardiac regeneration in adult individuals, a hitherto unachieved yet fundamental therapeutic goal.
Collapse
Affiliation(s)
- Hashim Ali
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, UK.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Luca Braga
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, UK.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Mauro Giacca
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, UK.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| |
Collapse
|
97
|
Singh AP, Umbarkar P, Guo Y, Force T, Gupte M, Lal H. Inhibition of GSK-3 to induce cardiomyocyte proliferation: a recipe for in situ cardiac regeneration. Cardiovasc Res 2020; 115:20-30. [PMID: 30321309 DOI: 10.1093/cvr/cvy255] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/09/2018] [Indexed: 01/03/2023] Open
Abstract
With an estimated 38 million current patients, heart failure (HF) is a leading cause of morbidity and mortality worldwide. Although the aetiology differs, HF is largely a disease of cardiomyocyte (CM) death or dysfunction. Due to the famously limited amount of regenerative capacity of the myocardium, the only viable option for advanced HF patients is cardiac transplantation; however, donor's hearts are in very short supply. Thus, novel regenerative strategies are urgently needed to reconstitute the injured hearts. Emerging data from our lab and others have elucidated that CM-specific deletion of glycogen synthase kinase (GSK)-3 family of kinases induces CM proliferation, and the degree of proliferation is amplified in the setting of cardiac stress. If this proliferation is sufficiently robust, one could induce meaningful regeneration without the need for delivering exogenous cells to the injured myocardium (i.e. cardiac regeneration in situ). Herein, we will discuss the emerging role of the GSK-3s in CM proliferation and differentiation, including their potential implications in cardiac regeneration. The underlying molecular interactions and cross-talk among signalling pathways will be discussed. We will also review the specificity and limitations of the available small molecule inhibitors targeting GSK-3 and their potential applications to stimulate the endogenous cardiac regenerative responses to repair the injured heart.
Collapse
Affiliation(s)
- Anand Prakash Singh
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Prachi Umbarkar
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Yuanjun Guo
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Thomas Force
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Manisha Gupte
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Hind Lal
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| |
Collapse
|
98
|
Lee EJ, Han JC, Park DY, Kee C. A neuroglia-based interpretation of glaucomatous neuroretinal rim thinning in the optic nerve head. Prog Retin Eye Res 2020; 77:100840. [PMID: 31982595 DOI: 10.1016/j.preteyeres.2020.100840] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/02/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022]
Abstract
Neuroretinal rim thinning (NRR) is a characteristic glaucomatous optic disc change. However, the precise mechanism of the rim thinning has not been completely elucidated. This review focuses on the structural role of the glioarchitecture in the formation of the glaucomatous NRR thinning. The NRR is a glia-framed structure, with honeycomb geometry and mechanically reinforced astrocyte processes along the transverse plane. When neural damage selectively involves the neuron and spares the glia, the gross structure of the tissue is preserved. The disorganization and loss of the glioarchitecture are the two hallmarks of optic nerve head (ONH) remodeling in glaucoma that leads to the thinning of NRR tissue upon axonal loss. This is in contrast to most non-glaucomatous optic neuropathies with optic disc pallor where hypertrophy of the glioarchitecture is associated with the seemingly absent optic disc cupping. Arteritic anterior ischemic optic neuropathy is an exception where pan-necrosis of ONH tissue leads to NRR thinning. Milder ischemia indicates selective neuronal loss that spares glia in non-arteritic anterior ischemic optic neuropathy. The biological reason is the heterogeneous glial response determined by the site, type, and severity of the injury. The neuroglial interpretation explains how the cellular changes underlie the clinical findings. Updated understandings on glial responses illustrate the mechanical, microenvironmental, and microglial modulation of activated astrocytes in glaucoma. Findings relevant to the possible mechanism of the astrocyte death in advanced glaucoma are also emerging. Ultimately, a better understanding of glaucomatous glial response may lead to glia-targeting neuroprotection in the future.
Collapse
Affiliation(s)
- Eun Jung Lee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Jong Chul Han
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Do Young Park
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Changwon Kee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea.
| |
Collapse
|
99
|
Yatsenko AS, Kucherenko MM, Xie Y, Aweida D, Urlaub H, Scheibe RJ, Cohen S, Shcherbata HR. Profiling of the muscle-specific dystroglycan interactome reveals the role of Hippo signaling in muscular dystrophy and age-dependent muscle atrophy. BMC Med 2020; 18:8. [PMID: 31959160 PMCID: PMC6971923 DOI: 10.1186/s12916-019-1478-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dystroglycanopathies are a group of inherited disorders characterized by vast clinical and genetic heterogeneity and caused by abnormal functioning of the ECM receptor dystroglycan (Dg). Remarkably, among many cases of diagnosed dystroglycanopathies, only a small fraction can be linked directly to mutations in Dg or its regulatory enzymes, implying the involvement of other, not-yet-characterized, Dg-regulating factors. To advance disease diagnostics and develop new treatment strategies, new approaches to find dystroglycanopathy-related factors should be considered. The Dg complex is highly evolutionarily conserved; therefore, model genetic organisms provide excellent systems to address this challenge. In particular, Drosophila is amenable to experiments not feasible in any other system, allowing original insights about the functional interactors of the Dg complex. METHODS To identify new players contributing to dystroglycanopathies, we used Drosophila as a genetic muscular dystrophy model. Using mass spectrometry, we searched for muscle-specific Dg interactors. Next, in silico analyses allowed us to determine their association with diseases and pathological conditions in humans. Using immunohistochemical, biochemical, and genetic interaction approaches followed by the detailed analysis of the muscle tissue architecture, we verified Dg interaction with some of the discovered factors. Analyses of mouse muscles and myocytes were used to test if interactions are conserved in vertebrates. RESULTS The muscle-specific Dg complexome revealed novel components that influence the efficiency of Dg function in the muscles. We identified the closest human homologs for Dg-interacting partners, determined their significant enrichment in disease-associations, and verified some of the newly identified Dg interactions. We found that Dg associates with two components of the mechanosignaling Hippo pathway: the WW domain-containing proteins Kibra and Yorkie. Importantly, this conserved interaction manages adult muscle size and integrity. CONCLUSIONS The results presented in this study provide a new list of muscle-specific Dg interactors, further analysis of which could aid not only in the diagnosis of muscular dystrophies, but also in the development of new therapeutics. To regulate muscle fitness during aging and disease, Dg associates with Kibra and Yorkie and acts as a transmembrane Hippo signaling receptor that transmits extracellular information to intracellular signaling cascades, regulating muscle gene expression.
Collapse
Affiliation(s)
- Andriy S Yatsenko
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Mariya M Kucherenko
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Present Address: Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Institute of Physiology, Charité - University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Yuanbin Xie
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Present Address: University Medical Center, Centre for Anatomy, Institute of Neuroanatomy, Georg-August-University Göttingen, Kreuzbergring 36, 37075, Göttingen, Germany
| | - Dina Aweida
- Faculty of Biology, Technion, 32000, Haifa, Israel
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Research Group, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Bioanalytics Institute for Clinical Chemistry, University Medical Center Goettingen, Robert Koch Strasse 40, 37075, Göttingen, Germany
| | - Renate J Scheibe
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | | | - Halyna R Shcherbata
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany. .,Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
100
|
Lei Y, Goldblatt ZE, Billiar KL. Micromechanical Design Criteria for Tissue-Engineering Biomaterials. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00083-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|