201
|
Cool CD, Kuebler WM, Bogaard HJ, Spiekerkoetter E, Nicolls MR, Voelkel NF. The hallmarks of severe pulmonary arterial hypertension: the cancer hypothesis-ten years later. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1115-L1130. [PMID: 32023082 PMCID: PMC9847334 DOI: 10.1152/ajplung.00476.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 01/25/2023] Open
Abstract
Severe forms of pulmonary arterial hypertension (PAH) are most frequently the consequence of a lumen-obliterating angiopathy. One pathobiological model is that the initial pulmonary vascular endothelial cell injury and apoptosis is followed by the evolution of phenotypically altered, apoptosis-resistant, proliferating cells and an inflammatory vascular immune response. Although there may be a vasoconstrictive disease component, the increased pulmonary vascular shear stress in established PAH is caused largely by the vascular wall pathology. In this review, we revisit the "quasi-malignancy concept" of severe PAH and examine to what extent the hallmarks of PAH can be compared with the hallmarks of cancer. The cancer model of severe PAH, based on the growth of abnormal vascular and bone marrow-derived cells, may enable the emergence of novel cell-based PAH treatment strategies.
Collapse
Affiliation(s)
- Carlyne D Cool
- Department of Pathology, University of Colorado, Anschuetz Campus, Aurora, Colorado
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitaetsmedizin, Berlin, Germany
| | - Harm Jan Bogaard
- Amsterdam University Medical Centers, Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical Care Medicine, Stanford University, Palo Alto, California
| | - Mark R Nicolls
- Division of Pulmonary and Critical Care Medicine, Stanford University, Palo Alto, California
| | - Norbert F Voelkel
- Amsterdam University Medical Centers, Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
202
|
Moon S, Lee S, Caesar JA, Pruchenko S, Leask A, Knowles JA, Sinon J, Chaqour B. A CTGF-YAP Regulatory Pathway Is Essential for Angiogenesis and Barriergenesis in the Retina. iScience 2020; 23:101184. [PMID: 32502964 PMCID: PMC7270711 DOI: 10.1016/j.isci.2020.101184] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/18/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022] Open
Abstract
Connective tissue growth factor (CTGF) or cellular communication network 2 (CCN2) is a matricellular protein essential for normal embryonic development and tissue repair. CTGF exhibits cell- and context-dependent activities, but CTGF function in vascular development and barrier function is unknown. We show that endothelial cells (ECs) are one of the major cellular sources of CTGF in the developing and adult retinal vasculature. Mice lacking CTGF expression either globally or specifically in ECs exhibit impaired vascular cell growth and morphogenesis and blood barrier breakdown. The global molecular signature of CTGF includes cytoskeletal and extracellular matrix protein, growth factor, and transcriptional co-regulator genes such as yes-associated protein (YAP). YAP, itself a transcriptional activator of CTGF, mediates several CTGF-controlled angiogenic and barriergenic transcriptional programs. Re-expression of YAP rescues, at least partially, angiogenesis and barriergenesis in CTGF mutant mouse retinas. Thus, the CTGF-YAP regulatory loop is integral to retinal vascular development and barrier function. CTGF has a strong and persistent expression in the retinal vasculature Mice lacking CTGF exhibit defects in angiogenesis and blood barrier integrity CTGF-targeted genes include matrix, growth, and transcription co-factors like YAP YAP re-expression partly rescues angiogenic and barriergenic defects of CTGF loss
Collapse
Affiliation(s)
- Sohyun Moon
- State University of New York, Downstate Health Science University, Department of Cell Biology, 450 Clarkson Avenue, MSC 5, Brooklyn, NY 11203, USA
| | - Sangmi Lee
- State University of New York, Downstate Health Science University, Department of Cell Biology, 450 Clarkson Avenue, MSC 5, Brooklyn, NY 11203, USA
| | - Joy Ann Caesar
- State University of New York, Downstate Health Science University, Department of Cell Biology, 450 Clarkson Avenue, MSC 5, Brooklyn, NY 11203, USA
| | - Sarah Pruchenko
- State University of New York, Downstate Health Science University, Department of Cell Biology, 450 Clarkson Avenue, MSC 5, Brooklyn, NY 11203, USA
| | - Andrew Leask
- University of Saskatchewan, College of Dentistry, E3338 HS - 105 Wiggins Road, Saskatoon, SK S7N 5E4, Canada
| | - James A Knowles
- State University of New York, Downstate Health Science University, Department of Cell Biology, 450 Clarkson Avenue, MSC 5, Brooklyn, NY 11203, USA
| | - Jose Sinon
- State University of New York, Downstate Health Science University, Department of Cell Biology, 450 Clarkson Avenue, MSC 5, Brooklyn, NY 11203, USA
| | - Brahim Chaqour
- State University of New York, Downstate Health Science University, Department of Cell Biology, 450 Clarkson Avenue, MSC 5, Brooklyn, NY 11203, USA; State University of New York, Downstate Health Science University, Department of Ophthalmology, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; SUNY Eye Institute, SUNY Downstate Health Science University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA.
| |
Collapse
|
203
|
Sun X, Li S, Gan X, Chen K, Yang D, Yang Y. NF2 deficiency accelerates neointima hyperplasia following vascular injury via promoting YAP-TEAD1 interaction in vascular smooth muscle cells. Aging (Albany NY) 2020; 12:9726-9744. [PMID: 32422606 PMCID: PMC7288949 DOI: 10.18632/aging.103240] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 03/31/2020] [Indexed: 01/12/2023]
Abstract
Neurofibromin 2 (NF2), a potent tumor suppressor, is reported to inhibit proliferation in several cell types. The role of NF2 in neointima hyperplasia after vascular injury is unknown. We explored the role of NF2 in proliferation, migration of vascular smooth muscle cell (VSMC) and neointima hyperplasia after vascular injury. NF2 phosphorylation was elevated in VSMC subjected to platelet-derived growth factor (PDGF)-BB and in artery subjected to vascular injury. Mice deficient for Nf2 in VSMC showed enhanced neointima hyperplasia after injury, increased proliferation and migration of VSMC after PDGF-BB treatment. Mechanistically, we observed increased nuclear p-NF2, declined p-Yes-Associated Protein (YAP), nuclear translocation of YAP after PDGF-BB treatment or injury. NF2 knockdown or YAP overexpression showed similar phenotype in VSMC proliferation, migration and neointima hyperplasia. YAP inhibition abolished the above effects mediated by NF2 knockdown. Finally, NF2 knockdown further promoted YAP-TEA Domain Transcription Factor 1 (TEAD1) interaction after PDGF-BB treatment. Inhibition of TEAD1 blocked PDGF-BB-induced VSMC proliferation and migration, which were not reversed by either NF2 knockdown or YAP overexpression. In conclusion, NF2 knockdown promotes VSMC proliferation, migration and neointima hyperplasia after vascular injury via inducing YAP-TEAD1 interaction.
Collapse
Affiliation(s)
- Xiongshan Sun
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Shuang Li
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Xueqing Gan
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Ken Chen
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Dachun Yang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yongjian Yang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| |
Collapse
|
204
|
Pullamsetti SS, Nayakanti S, Chelladurai P, Mamazhakypov A, Mansouri S, Savai R, Seeger W. Cancer and pulmonary hypertension: Learning lessons and real-life interplay. Glob Cardiol Sci Pract 2020; 2020:e202010. [PMID: 33150154 PMCID: PMC7590929 DOI: 10.21542/gcsp.2020.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
This article reviews the scientific reasons that support the intriguing vision of pulmonary hypertension (PH) as a disease with a cancer-like nature and to understand whether this point of view may have fruitful consequences for the overall management of PH. This review compares cancer and PH in view of Hanahan and Weinberg’s principles (i.e., hallmarks of cancer) with an emphasis on hyperproliferative, metabolic, and immune/inflammatory aspects of the disease. In addition, this review provides a perspective on the role of transcription factors and chromatin and epigenetic aberrations, besides genetics, as “common driving mechanisms” of PH hallmarks and the foreseeable use of transcription factor/epigenome targeting as multitarget approach against the hallmarks of PH. Thus, recognition of the widespread applicability and analogy of these concepts will increasingly affect the development of new means of PH treatment.
Collapse
Affiliation(s)
- Soni Savai Pullamsetti
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, 61231, Germany.,Department of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, Giessen, 35392, Germany
| | - Sreenath Nayakanti
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, 61231, Germany
| | - Prakash Chelladurai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, 61231, Germany
| | - Argen Mamazhakypov
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, 61231, Germany
| | - Siavash Mansouri
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, 61231, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, 61231, Germany.,Department of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, Giessen, 35392, Germany.,Institute for Lung Health (ILH), Member of the DZL, Justus Liebig University, Giessen, 35392, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, 61231, Germany.,Department of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, Giessen, 35392, Germany.,Institute for Lung Health (ILH), Member of the DZL, Justus Liebig University, Giessen, 35392, Germany
| |
Collapse
|
205
|
Thompson AAR, Wilkins MR, Wild JM, Kiely DG, Lawrie A. Editorial: Pulmonary Hypertension: Mechanisms and Management, History and Future. Front Med (Lausanne) 2020; 7:125. [PMID: 32373616 PMCID: PMC7186409 DOI: 10.3389/fmed.2020.00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/20/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- A. A. Roger Thompson
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Martin R. Wilkins
- Department of Medicine and National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jim M. Wild
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
- POLARIS, Academic Unit of Radiology, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for in silico Medicine, The University of Sheffield, Sheffield, United Kingdom
| | - David G. Kiely
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- INSIGNEO Institute for in silico Medicine, The University of Sheffield, Sheffield, United Kingdom
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for in silico Medicine, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
206
|
Pradhan S, Banda OA, Farino CJ, Sperduto JL, Keller KA, Taitano R, Slater JH. Biofabrication Strategies and Engineered In Vitro Systems for Vascular Mechanobiology. Adv Healthc Mater 2020; 9:e1901255. [PMID: 32100473 PMCID: PMC8579513 DOI: 10.1002/adhm.201901255] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/24/2020] [Indexed: 12/17/2022]
Abstract
The vascular system is integral for maintaining organ-specific functions and homeostasis. Dysregulation in vascular architecture and function can lead to various chronic or acute disorders. Investigation of the role of the vascular system in health and disease has been accelerated through the development of tissue-engineered constructs and microphysiological on-chip platforms. These in vitro systems permit studies of biochemical regulation of vascular networks and parenchymal tissue and provide mechanistic insights into the biophysical and hemodynamic forces acting in organ-specific niches. Detailed understanding of these forces and the mechanotransductory pathways involved is necessary to develop preventative and therapeutic strategies targeting the vascular system. This review describes vascular structure and function, the role of hemodynamic forces in maintaining vascular homeostasis, and measurement approaches for cell and tissue level mechanical properties influencing vascular phenomena. State-of-the-art techniques for fabricating in vitro microvascular systems, with varying degrees of biological and engineering complexity, are summarized. Finally, the role of vascular mechanobiology in organ-specific niches and pathophysiological states, and efforts to recapitulate these events using in vitro microphysiological systems, are explored. It is hoped that this review will help readers appreciate the important, but understudied, role of vascular-parenchymal mechanotransduction in health and disease toward developing mechanotherapeutics for treatment strategies.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Omar A. Banda
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Cindy J. Farino
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John L. Sperduto
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Keely A. Keller
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Ryan Taitano
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John H. Slater
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE 19716, USA
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, DE 19711, USA
| |
Collapse
|
207
|
Isobe S, Kataoka M, Endo J, Moriyama H, Okazaki S, Tsuchihashi K, Katsumata Y, Yamamoto T, Shirakawa K, Yoshida N, Shimoda M, Chiba T, Masuko T, Hakamata Y, Kobayashi E, Saya H, Fukuda K, Sano M. Endothelial-Mesenchymal Transition Drives Expression of CD44 Variant and xCT in Pulmonary Hypertension. Am J Respir Cell Mol Biol 2020; 61:367-379. [PMID: 30897333 DOI: 10.1165/rcmb.2018-0231oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) pathogenesis shares similarities with carcinogenesis. One CD44 variant (CD44v) isoform, CD44v8-10, binds to and stabilizes the cystine transporter subunit (xCT), producing reduced glutathione and thereby enhancing the antioxidant defense of cancer stem cells. Pharmacological inhibition of xCT by sulfasalazine suppresses tumor growth, survival, and resistance to chemotherapy. We investigated whether the CD44v-xCT axis contributes to PAH pathogenesis. CD44v was predominantly expressed on endothelial-to-mesenchymal transition (EndMT)-like cells in the neointimal layer of PAH affected pulmonary arterioles. In vitro, CD44 standard form and CD44v were induced as a result of EndMT. Among human pulmonary artery endothelial cells that have undergone EndMT, CD44v+ cells showed high levels of xCT expression on their cell surfaces and high concentrations of glutathione for survival. This made CD44v+ cells the most vulnerable target for sulfasalazine. CD44v+xCThi cells showed the highest expression levels of proinflammatory cytokines, antioxidant enzymes, antiapoptotic molecules, and cyclin-dependent kinase inhibitors. In the Sugen5416/hypoxia mouse model, CD44v+ cells were present in the thickened pulmonary vascular wall. The administration of sulfasalazine started either at the same time as "Sugen5416" administration (a prevention model) or after the development of pulmonary hypertension (a reversal model) attenuated the muscularization of the pulmonary vessels, decreased the expression of markers of inflammation, and reduced the right ventricular systolic pressure, while reducing CD44v+ cells. In conclusion, CD44v+xCThi cells appear during EndMT and in pulmonary hypertension tissues. Sulfasalazine is expected to be a novel therapeutic agent for PAH, most likely targeting EndMT-derived CD44v+xCThi cells.
Collapse
Affiliation(s)
| | | | | | | | - Shogo Okazaki
- Division of Gene Regulation, Institute for Advanced Medical Research
| | - Kenji Tsuchihashi
- Division of Gene Regulation, Institute for Advanced Medical Research
| | | | | | | | - Naohiro Yoshida
- Department of Cardiology.,Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | | | - Tomohiro Chiba
- Department of Pathology, Kyorin University School of Medicine, Tokyo, Japan
| | - Takashi Masuko
- Department of Pharmaceutical Sciences, Cell Biology Laboratory, Faculty of Pharmacy, Kindai University, Osaka, Japan; and
| | - Yoji Hakamata
- Department of Basic Science, School of Veterinary Nursing and Technology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Eiji Kobayashi
- Department of Organ Fabrication, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research
| | | | | |
Collapse
|
208
|
Girard CA, Lecacheur M, Ben Jouira R, Berestjuk I, Diazzi S, Prod'homme V, Mallavialle A, Larbret F, Gesson M, Schaub S, Pisano S, Audebert S, Mari B, Gaggioli C, Leucci E, Marine JC, Deckert M, Tartare-Deckert S. A Feed-Forward Mechanosignaling Loop Confers Resistance to Therapies Targeting the MAPK Pathway in BRAF-Mutant Melanoma. Cancer Res 2020; 80:1927-1941. [PMID: 32179513 DOI: 10.1158/0008-5472.can-19-2914] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/15/2020] [Accepted: 03/06/2020] [Indexed: 11/16/2022]
Abstract
Aberrant extracellular matrix (ECM) deposition and stiffening is a physical hallmark of several solid cancers and is associated with therapy failure. BRAF-mutant melanomas treated with BRAF and MEK inhibitors almost invariably develop resistance that is frequently associated with transcriptional reprogramming and a de-differentiated cell state. Melanoma cells secrete their own ECM proteins, an event that is promoted by oncogenic BRAF inhibition. Yet, the contribution of cancer cell-derived ECM and tumor mechanics to drug adaptation and therapy resistance remains poorly understood. Here, we show that melanoma cells can adapt to targeted therapies through a mechanosignaling loop involving the autocrine remodeling of a drug-protective ECM. Analyses revealed that therapy-resistant cells associated with a mesenchymal dedifferentiated state displayed elevated responsiveness to collagen stiffening and force-mediated ECM remodeling through activation of actin-dependent mechanosensors Yes-associated protein (YAP) and myocardin-related transcription factor (MRTF). Short-term inhibition of MAPK pathway also induced mechanosignaling associated with deposition and remodeling of an aligned fibrillar matrix. This provided a favored ECM reorganization that promoted tolerance to BRAF inhibition in a YAP- and MRTF-dependent manner. Matrix remodeling and tumor stiffening were also observed in vivo upon exposure of BRAF-mutant melanoma cell lines or patient-derived xenograft models to MAPK pathway inhibition. Importantly, pharmacologic targeting of YAP reversed treatment-induced excessive collagen deposition, leading to enhancement of BRAF inhibitor efficacy. We conclude that MAPK pathway targeting therapies mechanically reprogram melanoma cells to confer a drug-protective matrix environment. Preventing melanoma cell mechanical reprogramming might be a promising therapeutic strategy for patients on targeted therapies. SIGNIFICANCE: These findings reveal a biomechanical adaptation of melanoma cells to oncogenic BRAF pathway inhibition, which fuels a YAP/MRTF-dependent feed-forward loop associated with tumor stiffening, mechanosensing, and therapy resistance. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/10/1927/F1.large.jpg.
Collapse
Affiliation(s)
- Christophe A Girard
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Margaux Lecacheur
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Rania Ben Jouira
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Ilona Berestjuk
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Serena Diazzi
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Virginie Prod'homme
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Aude Mallavialle
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Frédéric Larbret
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Maéva Gesson
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | | | - Sabrina Pisano
- Université Côte d'Azur, CNRS, INSERM, IRCAN, Nice, France
| | - Stéphane Audebert
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Bernard Mari
- Université Côte d'Azur, CNRS, IPMC, Sophia Antipolis, France
| | | | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium.,TRACE, LKI Leuven Cancer Institute, KU Leuven
| | - Jean-Christophe Marine
- Laboratory For Molecular Cancer Biology, VIB Center for Cancer Biology, VIB, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Marcel Deckert
- Université Côte d'Azur, INSERM, C3M, Nice, France. .,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Sophie Tartare-Deckert
- Université Côte d'Azur, INSERM, C3M, Nice, France. .,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| |
Collapse
|
209
|
Dasgupta A, Wu D, Tian L, Xiong PY, Dunham-Snary KJ, Chen KH, Alizadeh E, Motamed M, Potus F, Hindmarch CCT, Archer SL. Mitochondria in the Pulmonary Vasculature in Health and Disease: Oxygen-Sensing, Metabolism, and Dynamics. Compr Physiol 2020; 10:713-765. [PMID: 32163206 DOI: 10.1002/cphy.c190027] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In lung vascular cells, mitochondria serve a canonical metabolic role, governing energy homeostasis. In addition, mitochondria exist in dynamic networks, which serve noncanonical functions, including regulation of redox signaling, cell cycle, apoptosis, and mitochondrial quality control. Mitochondria in pulmonary artery smooth muscle cells (PASMC) are oxygen sensors and initiate hypoxic pulmonary vasoconstriction. Acquired dysfunction of mitochondrial metabolism and dynamics contribute to a cancer-like phenotype in pulmonary arterial hypertension (PAH). Acquired mitochondrial abnormalities, such as increased pyruvate dehydrogenase kinase (PDK) and pyruvate kinase muscle isoform 2 (PKM2) expression, which increase uncoupled glycolysis (the Warburg phenomenon), are implicated in PAH. Warburg metabolism sustains energy homeostasis by the inhibition of oxidative metabolism that reduces mitochondrial apoptosis, allowing unchecked cell accumulation. Warburg metabolism is initiated by the induction of a pseudohypoxic state, in which DNA methyltransferase (DNMT)-mediated changes in redox signaling cause normoxic activation of HIF-1α and increase PDK expression. Furthermore, mitochondrial division is coordinated with nuclear division through a process called mitotic fission. Increased mitotic fission in PAH, driven by increased fission and reduced fusion favors rapid cell cycle progression and apoptosis resistance. Downregulation of the mitochondrial calcium uniporter complex (MCUC) occurs in PAH and is one potential unifying mechanism linking Warburg metabolism and mitochondrial fission. Mitochondrial metabolic and dynamic disorders combine to promote the hyperproliferative, apoptosis-resistant, phenotype in PAH PASMC, endothelial cells, and fibroblasts. Understanding the molecular mechanism regulating mitochondrial metabolism and dynamics has permitted identification of new biomarkers, nuclear and CT imaging modalities, and new therapeutic targets for PAH. © 2020 American Physiological Society. Compr Physiol 10:713-765, 2020.
Collapse
Affiliation(s)
- Asish Dasgupta
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Danchen Wu
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Lian Tian
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Ping Yu Xiong
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - Kuang-Hueih Chen
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Elahe Alizadeh
- Department of Medicine, Queen's Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Queen's University, Kingston, Ontario, Canada
| | - Mehras Motamed
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - François Potus
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Charles C T Hindmarch
- Department of Medicine, Queen's Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada.,Kingston Health Sciences Centre, Kingston, Ontario, Canada.,Providence Care Hospital, Kingston, Ontario, Canada
| |
Collapse
|
210
|
Nazemi M, Rainero E. Cross-Talk Between the Tumor Microenvironment, Extracellular Matrix, and Cell Metabolism in Cancer. Front Oncol 2020; 10:239. [PMID: 32175281 PMCID: PMC7054479 DOI: 10.3389/fonc.2020.00239] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/12/2020] [Indexed: 12/22/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network of secreted proteins which provides support for tissues and organs. Additionally, the ECM controls a plethora of cell functions, including cell polarity, migration, proliferation, and oncogenic transformation. One of the hallmarks of cancer is altered cell metabolism, which is currently being exploited to develop anti-cancer therapies. Several pieces of evidence indicate that the tumor microenvironment and the ECM impinge on tumor cell metabolism. Therefore, it is essential to understand the contribution of the complex 3D microenvironment in controlling metabolic plasticity and responsiveness to therapies targeting cell metabolism. In this mini-review, we will describe how the tumor microenvironment and cancer-associated fibroblasts dictate cancer cell metabolism, resulting in increased tumor progression. Moreover, we will define the cross-talk between nutrient signaling and the trafficking of the ECM receptors of the integrin family. Finally, we will present recent data highlighting the contribution of nutrient scavenging from the microenvironment to support cancer cells growth under nutrient starvation conditions.
Collapse
Affiliation(s)
- Mona Nazemi
- Biomedical Science Department, The University of Sheffield, Sheffield, United Kingdom
| | - Elena Rainero
- Biomedical Science Department, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
211
|
Liu QP, Luo Q, Deng B, Ju Y, Song GB. Stiffer Matrix Accelerates Migration of Hepatocellular Carcinoma Cells through Enhanced Aerobic Glycolysis Via the MAPK-YAP Signaling. Cancers (Basel) 2020; 12:E490. [PMID: 32093118 PMCID: PMC7072284 DOI: 10.3390/cancers12020490] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 12/24/2022] Open
Abstract
Increased extracellular matrix (ECM) stiffness and metabolic reprogramming of cancer cells are two fundamental mediators of tumor progression, including hepatocellular carcinoma (HCC). Yet, the correlation between ECM stiffness and excessive aerobic glycolysis in promoting the development of HCC remains unknown. Here, we demonstrated that stiffer ECM promotes HCC cell migration depending on their accelerated aerobic glycolysis. Our results also indicated that stiffer ECM-induced YAP activation plays a major role in promoting aerobic glycolysis of HCC cells. Moreover, we showed that JNK and p38 MAPK signaling are critical for mediating YAP activation in HCC cells. Together, our findings established that the MAPK-YAP signaling cascade that act as a mechanotransduction pathway is essential for promoting HCC cell aerobic glycolysis and migration in response to ECM stiffness.
Collapse
Affiliation(s)
- Qiu-Ping Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China; (Q.-P.L.); (Q.L.); (B.D.)
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China; (Q.-P.L.); (Q.L.); (B.D.)
| | - Bin Deng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China; (Q.-P.L.); (Q.L.); (B.D.)
| | - Yang Ju
- Department of Mechanical Science and Engineering, Nagoya University, Nagoya 464-8603, Japan;
| | - Guan-Bin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China; (Q.-P.L.); (Q.L.); (B.D.)
| |
Collapse
|
212
|
Stearman RS, Bui QM, Speyer G, Handen A, Cornelius AR, Graham BB, Kim S, Mickler EA, Tuder RM, Chan SY, Geraci MW. Systems Analysis of the Human Pulmonary Arterial Hypertension Lung Transcriptome. Am J Respir Cell Mol Biol 2020; 60:637-649. [PMID: 30562042 DOI: 10.1165/rcmb.2018-0368oc] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by increased pulmonary artery pressure and vascular resistance, typically leading to right heart failure and death. Current therapies improve quality of life of the patients but have a modest effect on long-term survival. A detailed transcriptomics and systems biology view of the PAH lung is expected to provide new testable hypotheses for exploring novel treatments. We completed transcriptomics analysis of PAH and control lung tissue to develop disease-specific and clinical data/tissue pathology gene expression classifiers from expression datasets. Gene expression data were integrated into pathway analyses. Gene expression microarray data were collected from 58 PAH and 25 control lung tissues. The strength of the dataset and its derived disease classifier was validated using multiple approaches. Pathways and upstream regulators analyses was completed with standard and novel graphical approaches. The PAH lung dataset identified expression patterns specific to PAH subtypes, clinical parameters, and lung pathology variables. Pathway analyses indicate the important global role of TNF and transforming growth factor signaling pathways. In addition, novel upstream regulators and insight into the cellular and innate immune responses driving PAH were identified. Finally, WNT-signaling pathways may be a major determinant underlying the observed sex differences in PAH. This study provides a transcriptional framework for the PAH-diseased lung, supported by previously reported findings, and will be a valuable resource to the PAH research community. Our investigation revealed novel potential targets and pathways amenable to further study in a variety of experimental systems.
Collapse
Affiliation(s)
- Robert S Stearman
- 1 Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Quan M Bui
- 1 Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Gil Speyer
- 2 Quantitative Medicine and Systems Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona.,3 Research Computing, Arizona State University, Tempe, Arizona
| | - Adam Handen
- 4 Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Amber R Cornelius
- 1 Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Brian B Graham
- 5 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado; and
| | - Seungchan Kim
- 6 Department of Electrical and Computer Engineering, Center for Computational Systems Biology, Roy G. Perry College of Engineering, Prairie View A&M University, Prairie View, Texas
| | - Elizabeth A Mickler
- 1 Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Rubin M Tuder
- 5 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado; and
| | - Stephen Y Chan
- 4 Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Mark W Geraci
- 1 Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
213
|
van der Stoel M, Schimmel L, Nawaz K, van Stalborch AM, de Haan A, Klaus-Bergmann A, Valent ET, Koenis DS, van Nieuw Amerongen GP, de Vries CJ, de Waard V, Gloerich M, van Buul JD, Huveneers S. DLC1 is a direct target of activated YAP/TAZ that drives collective migration and sprouting angiogenesis. J Cell Sci 2020; 133:jcs239947. [PMID: 31964713 DOI: 10.1242/jcs.239947] [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: 10/01/2019] [Accepted: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
Endothelial YAP/TAZ (YAP is also known as YAP1, and TAZ as WWTR1) signaling is crucial for sprouting angiogenesis and vascular homeostasis. However, the underlying molecular mechanisms that explain how YAP/TAZ control the vasculature remain unclear. This study reveals that the focal adhesion protein deleted-in-liver-cancer 1 (DLC1) is a direct transcriptional target of the activated YAP/TAZ-TEAD complex. We find that substrate stiffening and VEGF stimuli promote expression of DLC1 in endothelial cells. In turn, DLC1 expression levels are YAP and TAZ dependent, and constitutive activation of YAP is sufficient to drive DLC1 expression. DLC1 is needed to limit F-actin fiber formation, integrin-based focal adhesion lifetime and integrin-mediated traction forces. Depletion of endothelial DLC1 strongly perturbs cell polarization in directed collective migration and inhibits the formation of angiogenic sprouts. Importantly, ectopic expression of DLC1 is sufficient to restore migration and angiogenic sprouting in YAP-depleted cells. Together, these findings point towards a crucial and prominent role for DLC1 in YAP/TAZ-driven endothelial adhesion remodeling and collective migration during angiogenesis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Miesje van der Stoel
- Amsterdam UMC, University of Amsterdam, location AMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands
| | - Lilian Schimmel
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
| | - Kalim Nawaz
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
| | - Anne-Marieke van Stalborch
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
| | - Annett de Haan
- Amsterdam UMC, University of Amsterdam, location AMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands
| | - Alexandra Klaus-Bergmann
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
- DZHK (German Center for Cardiovascular Research), 10785 Berlin, Germany
| | - Erik T Valent
- Amsterdam UMC, Free University, location VUMC, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands
| | - Duco S Koenis
- Amsterdam UMC, University of Amsterdam, location AMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands
| | - Geerten P van Nieuw Amerongen
- Amsterdam UMC, Free University, location VUMC, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands
| | - Carlie J de Vries
- Amsterdam UMC, University of Amsterdam, location AMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands
| | - Vivian de Waard
- Amsterdam UMC, University of Amsterdam, location AMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands
| | - Martijn Gloerich
- University Medical Center Utrecht, Center for Molecular Medicine, Dept. Molecular Cancer Research, 3584 CX Utrecht, The Netherlands
| | - Jaap D van Buul
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, location AMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
214
|
Valuparampil Varghese M, James J, Eccles CA, Niihori M, Rafikova O, Rafikov R. Inhibition of Anaplerosis Attenuated Vascular Proliferation in Pulmonary Arterial Hypertension. J Clin Med 2020; 9:jcm9020443. [PMID: 32041182 PMCID: PMC7074087 DOI: 10.3390/jcm9020443] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 01/09/2023] Open
Abstract
Vascular remodeling is considered a key event in the pathogenesis of pulmonary arterial hypertension (PAH). However, mechanisms of gaining the proliferative phenotype by pulmonary vascular cells are still unresolved. Due to well-established pyruvate dehydrogenase (PDH) deficiency in PAH pathogenesis, we hypothesized that the activation of another branch of pyruvate metabolism, anaplerosis, via pyruvate carboxylase (PC) could be a key contributor to the metabolic reprogramming of the vasculature. In sugen/hypoxic PAH rats, vascular proliferation was found to be accompanied by increased activation of Akt signaling, which upregulated membrane Glut4 translocation and caused upregulation of hexokinase and pyruvate kinase-2, and an overall increase in the glycolytic flux. Decreased PDH activity and upregulation of PC shuttled more pyruvate to oxaloacetate. This results in the anaplerotic reprogramming of lung vascular cells and their subsequent proliferation. Treatment of sugen/hypoxia rats with the PC inhibitor, phenylacetic acid 20 mg/kg, starting after one week from disease induction, significantly attenuated right ventricular systolic pressure, Fulton index, and pulmonary vascular cell proliferation. PC inhibition reduced the glycolytic shift by attenuating Akt-signaling, glycolysis, and restored mitochondrial pyruvate oxidation. Our findings suggest that targeting PC mediated anaplerosis is a potential therapeutic intervention for the resolution of vascular remodeling in PAH.
Collapse
Affiliation(s)
| | | | | | | | - Olga Rafikova
- Correspondence: (O.R.); (R.R.); Tel.: +1-520-626-1303 (O.R.); +1-520-626-6092 (R.R.)
| | - Ruslan Rafikov
- Correspondence: (O.R.); (R.R.); Tel.: +1-520-626-1303 (O.R.); +1-520-626-6092 (R.R.)
| |
Collapse
|
215
|
Hansmann G, Calvier L, Risbano MG, Chan SY. Activation of the Metabolic Master Regulator PPARγ: A Potential PIOneering Therapy for Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2020; 62:143-156. [PMID: 31577451 PMCID: PMC6993553 DOI: 10.1165/rcmb.2019-0226ps] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/02/2019] [Indexed: 12/24/2022] Open
Abstract
Translational research is essential to the development of reverse-remodeling strategies for the treatment of pulmonary vascular disease, pulmonary hypertension, and heart failure via mechanistic in vivo studies using animal models resembling human pulmonary arterial hypertension (PAH), cardiovascular remodeling, and progressive right heart failure. Since 2007, peroxisome proliferator-activated receptor γ (PPARγ) agonists have emerged as promising novel, antiproliferative, antiinflammatory, insulin-sensitizing, efficient medications for the treatment of PAH. However, early diabetes study results, their subsequent misinterpretations, errors in published review articles, and rumors regarding potential adverse effects in the literature have dampened enthusiasm for considering pharmacological PPARγ activation for the treatment of cardiovascular diseases, including PAH. Most recently, the thiazolidinedione class PPARγ agonist pioglitazone underwent a clinical revival, especially based on the IRIS (Insulin Resistance Intervention After Stroke) study, a randomized controlled trial in 3,876 patients without diabetes status post-transient ischemic attack/ischemic stroke who were clinically followed for 4.8 years. We discuss preclinical basic translational findings and randomized controlled trials related to the beneficial and adverse effects of PPARγ agonists of the thiazolidinedione class, with a particular focus on the last 5 years. The objective is a data-driven approach to set the preclinical and clinical study record straight. The convincing recent clinical trial data on the lack of significant toxicity in high-risk populations justify the timely conduct of clinical studies to achieve "repurposing" or "repositioning" of pioglitazone for the treatment of clinical PAH.
Collapse
Affiliation(s)
- Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany; and
| | - Laurent Calvier
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany; and
| | - Michael G. Risbano
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, and
| | - Stephen Y. Chan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, and
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
216
|
Antioxidant-Conjugated Peptide Attenuated Metabolic Reprogramming in Pulmonary Hypertension. Antioxidants (Basel) 2020; 9:antiox9020104. [PMID: 31991719 PMCID: PMC7071131 DOI: 10.3390/antiox9020104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/20/2020] [Accepted: 01/23/2020] [Indexed: 01/11/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic cardiopulmonary disorder instigated by pulmonary vascular cell proliferation. Activation of Akt was previously reported to promote vascular remodeling. Also, the irreversible nitration of Y350 residue in Akt results in its activation. NitroAkt was increased in PAH patients and the SU5416/Hypoxia (SU/Hx) PAH model. This study investigated whether the prevention of Akt nitration in PAH by Akt targeted nitroxide-conjugated peptide (NP) could reverse vascular remodeling and metabolic reprogramming. Treatment of the SU/Hx model with NP significantly decreased nitration of Akt in lungs, attenuated right ventricle (RV) hypertrophy, and reduced RV systolic pressure. In the PAH model, Akt-nitration induces glycolysis by activation of the glucose transporter Glut4 and lactate dehydrogenase-A (LDHA). Decreased G6PD and increased GSK3β in SU/Hx additionally shunted intracellular glucose via glycolysis. The increased glycolytic rate upregulated anaplerosis due to activation of pyruvate carboxylase in a nitroAkt-dependent manner. NP treatment resolved glycolytic switch and activated collateral pentose phosphate and glycogenesis pathways. Prevention of Akt-nitration significantly controlled pyruvate in oxidative phosphorylation by decreasing lactate and increasing pyruvate dehydrogenases activities. Histopathological studies showed significantly reduced pulmonary vascular proliferation. Based on our current observation, preventing Akt-nitration by using an Akt-targeted nitroxide-conjugated peptide could be a useful treatment option for controlling vascular proliferation in PAH.
Collapse
|
217
|
Stable isotope metabolomics of pulmonary artery smooth muscle and endothelial cells in pulmonary hypertension and with TGF-beta treatment. Sci Rep 2020; 10:413. [PMID: 31942023 PMCID: PMC6962446 DOI: 10.1038/s41598-019-57200-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Altered metabolism in pulmonary artery smooth muscle cells (PASMCs) and endothelial cells (PAECs) contributes to the pathology of pulmonary hypertension (PH), but changes in substrate uptake and how substrates are utilized have not been fully characterized. We hypothesized stable isotope metabolomics would identify increased glucose, glutamine and fatty acid uptake and utilization in human PASMCs and PAECs from PH versus control specimens, and that TGF-β treatment would phenocopy these metabolic changes. We used 13C-labeled glucose, glutamine or a long-chain fatty acid mixture added to cell culture media, and mass spectrometry-based metabolomics to detect and quantify 13C-labeled metabolites. We found PH PASMCs had increased glucose uptake and utilization by glycolysis and the pentose shunt, but no changes in glutamine or fatty acid uptake or utilization. Diseased PAECs had increased proximate glycolysis pathway intermediates, less pentose shunt flux, increased anaplerosis from glutamine, and decreased fatty acid β-oxidation. TGF-β treatment increased glycolysis in PASMCs, but did not recapitulate the PAEC disease phenotype. In TGF-β-treated PASMCs, glucose, glutamine and fatty acids all contributed carbons to the TCA cycle. In conclusion, PASMCs and PAECs collected from PH subjects have significant changes in metabolite uptake and utilization, partially recapitulated by TGF-β treatment.
Collapse
|
218
|
White SM, Avantaggiati ML, Nemazanyy I, Di Poto C, Yang Y, Pende M, Gibney GT, Ressom HW, Field J, Atkins MB, Yi C. YAP/TAZ Inhibition Induces Metabolic and Signaling Rewiring Resulting in Targetable Vulnerabilities in NF2-Deficient Tumor Cells. Dev Cell 2020; 49:425-443.e9. [PMID: 31063758 DOI: 10.1016/j.devcel.2019.04.014] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 02/04/2019] [Accepted: 04/08/2019] [Indexed: 02/09/2023]
Abstract
Merlin/NF2 is a bona fide tumor suppressor whose mutations underlie inherited tumor syndrome neurofibromatosis type 2 (NF2), as well as various sporadic cancers including kidney cancer. Multiple Merlin/NF2 effector pathways including the Hippo-YAP/TAZ pathway have been identified. However, the molecular mechanisms underpinning the growth and survival of NF2-mutant tumors remain poorly understood. Using an inducible orthotopic kidney tumor model, we demonstrate that YAP/TAZ silencing is sufficient to induce regression of pre-established NF2-deficient tumors. Mechanistically, YAP/TAZ depletion diminishes glycolysis-dependent growth and increases mitochondrial respiration and reactive oxygen species (ROS) buildup, resulting in oxidative-stress-induced cell death when challenged by nutrient stress. Furthermore, we identify lysosome-mediated cAMP-PKA/EPAC-dependent activation of RAF-MEK-ERK signaling as a resistance mechanism to YAP/TAZ inhibition. Finally, unbiased analysis of TCGA primary kidney tumor transcriptomes confirms a positive correlation of a YAP/TAZ signature with glycolysis and inverse correlations with oxidative phosphorylation and lysosomal gene expression, supporting the clinical relevance of our findings.
Collapse
Affiliation(s)
- Shannon M White
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | | | - Ivan Nemazanyy
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, Institut Necker Enfants Malades, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Cristina Di Poto
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Yang Yang
- Department of Systems Pharmacology and Translational Therapeutics, Perelmen School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mario Pende
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, Institut Necker Enfants Malades, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Geoffrey T Gibney
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Habtom W Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Jeffery Field
- Department of Systems Pharmacology and Translational Therapeutics, Perelmen School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael B Atkins
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Chunling Yi
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA.
| |
Collapse
|
219
|
Prins KW, Thenappan T, Weir EK, Kalra R, Pritzker M, Archer SL. Repurposing Medications for Treatment of Pulmonary Arterial Hypertension: What's Old Is New Again. J Am Heart Assoc 2020; 8:e011343. [PMID: 30590974 PMCID: PMC6405714 DOI: 10.1161/jaha.118.011343] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kurt W Prins
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Thenappan Thenappan
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - E Kenneth Weir
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Rajat Kalra
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Marc Pritzker
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | | |
Collapse
|
220
|
Durante W. Amino Acids in Circulatory Function and Health. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1265:39-56. [PMID: 32761569 DOI: 10.1007/978-3-030-45328-2_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is the major cause of global mortality and disability. Abundant evidence indicates that amino acids play a fundamental role in cardiovascular physiology and pathology. Decades of research established the importance of L-arginine in promoting vascular health through the generation of the gas nitric oxide. More recently, L-glutamine, L-tryptophan, and L-cysteine have also been shown to modulate vascular function via the formation of a myriad of metabolites, including a number of gases (ammonia, carbon monoxide, hydrogen sulfide, and sulfur dioxide). These amino acids and their metabolites preserve vascular homeostasis by regulating critical cellular processes including proliferation, migration, differentiation, apoptosis, contractility, and senescence. Furthermore, they exert potent anti-inflammatory and antioxidant effects in the circulation, and block the accumulation of lipids within the arterial wall. They also mitigate known risk factors for cardiovascular disease, including hypertension, hyperlipidemia, obesity, and diabetes. However, in some instances, the metabolism of these amino acids through discrete pathways yields compounds that fosters vascular disease. While supplementation with amino acid monotherapy targeting the deficiency has ameliorated arterial disease in many animal models, this approach has been less successful in the clinic. A more robust approach combining amino acid supplementation with antioxidants, anti-inflammatory agents, and/or specific amino acid enzymatic pathway inhibitors may prove more successful. Alternatively, supplementation with amino acid-derived metabolites rather than the parent molecule may elicit beneficial effects while bypassing potentially harmful pathways of metabolism. Finally, there is an emerging recognition that circulating levels of multiple amino acids are perturbed in vascular disease and that a more holistic approach that targets all these amino acid derangements is required to restore circulatory function in diseased blood vessels.
Collapse
Affiliation(s)
- William Durante
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
221
|
Arvidsson M, Ahmed A, Bouzina H, Rådegran G. Matrix metalloproteinase 7 in diagnosis and differentiation of pulmonary arterial hypertension. Pulm Circ 2019; 9:2045894019895414. [PMID: 31908766 PMCID: PMC6935882 DOI: 10.1177/2045894019895414] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 11/19/2019] [Indexed: 01/15/2023] Open
Abstract
Pulmonary arterial hypertension is a severe disease for which diagnosis often is delayed. Matrix metalloproteinases have been suggested to play a role in vascular remodeling and pulmonary hypertension development. Our aim was therefore to investigate the potential role of matrix metalloproteinases as biomarkers in diagnosis and differentiation of pulmonary arterial hypertension in relation to various causes of dyspnea and pulmonary hypertension. Using proximity extension assays, 10 matrix metalloproteinases and associated proteins were analyzed in venous plasma from healthy controls (n = 20), as well as patients diagnosed with pulmonary arterial hypertension (n = 48), chronic thromboembolic pulmonary hypertension (n = 20), pulmonary hypertension due to heart failure with preserved (n = 33) or reduced (n = 36) ejection fraction, and heart failure with reduced ejection fraction and heart failure with preserved ejection fraction without pulmonary hypertension (n = 15). Plasma levels of matrix metalloproteinase-2, -7, -9, -12 and TIMP-4 were elevated (p < 0.01) in pulmonary arterial hypertension compared to controls. Plasma levels of matrix metalloproteinase-7 were furthermore lower (p < 0.0081) in pulmonary arterial hypertension than in all the other disease groups, but higher compared to controls (p < 0.0001). Receiver operating characteristic analysis of matrix metalloproteinase-7 resulted in sensitivity of 58.7% and a specificity of 83.3% for detecting pulmonary arterial hypertension among the other disease groups. Plasma matrix metalloproteinase-7 may provide a potential new diagnostic tool to differentiate pulmonary arterial hypertension from other causes of dyspnea, including heart failure with or without pulmonary hypertension and healthy controls. Matrix metalloproteinase-7 may furthermore be involved in the development of pulmonary hypertension and pulmonary arterial hypertension. Future studies investigating the clinical usefulness of matrix metalloproteinase-7 in the differentiation and earlier diagnosis of pulmonary arterial hypertension, as well as its relationship to pulmonary arterial hypertension pathogenesis, are encouraged.
Collapse
Affiliation(s)
- Mattias Arvidsson
- Department of Clinical Sciences Lund, Cardiology, Faculty of Medicine, Lund University, Lund, Sweden.,The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Abdulla Ahmed
- Department of Clinical Sciences Lund, Cardiology, Faculty of Medicine, Lund University, Lund, Sweden.,The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Habib Bouzina
- Department of Clinical Sciences Lund, Cardiology, Faculty of Medicine, Lund University, Lund, Sweden.,The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Göran Rådegran
- Department of Clinical Sciences Lund, Cardiology, Faculty of Medicine, Lund University, Lund, Sweden.,The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
222
|
Abstract
The Hippo pathway was initially discovered in Drosophila melanogaster as a key regulator of tissue growth. It is an evolutionarily conserved signaling cascade regulating numerous biological processes, including cell growth and fate decision, organ size control, and regeneration. The core of the Hippo pathway in mammals consists of a kinase cascade, MST1/2 and LATS1/2, as well as downstream effectors, transcriptional coactivators YAP and TAZ. These core components of the Hippo pathway control transcriptional programs involved in cell proliferation, survival, mobility, stemness, and differentiation. The Hippo pathway is tightly regulated by both intrinsic and extrinsic signals, such as mechanical force, cell-cell contact, polarity, energy status, stress, and many diffusible hormonal factors, the majority of which act through G protein-coupled receptors. Here, we review the current understanding of molecular mechanisms by which signals regulate the Hippo pathway with an emphasis on mechanotransduction and the effects of this pathway on basic biology and human diseases.
Collapse
Affiliation(s)
- Shenghong Ma
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| | - Zhipeng Meng
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| | - Rui Chen
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| |
Collapse
|
223
|
Xu W, Comhair SAA, Chen R, Hu B, Hou Y, Zhou Y, Mavrakis LA, Janocha AJ, Li L, Zhang D, Willard BB, Asosingh K, Cheng F, Erzurum SC. Integrative proteomics and phosphoproteomics in pulmonary arterial hypertension. Sci Rep 2019; 9:18623. [PMID: 31819116 PMCID: PMC6901481 DOI: 10.1038/s41598-019-55053-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial endothelial cells (PAEC) are mechanistically linked to origins of pulmonary arterial hypertension (PAH). Here, global proteomics and phosphoproteomics of PAEC from PAH (n = 4) and healthy lungs (n = 5) were performed using LC-MS/MS to confirm known pathways and identify new areas of investigation in PAH. Among PAH and control cells, 170 proteins and 240 phosphopeptides were differentially expressed; of these, 45 proteins and 18 phosphopeptides were located in the mitochondria. Pathologic pathways were identified with integrative bioinformatics and human protein-protein interactome network analyses, then confirmed with targeted proteomics in PAH PAEC and non-targeted metabolomics and targeted high-performance liquid chromatography of metabolites in plasma from PAH patients (n = 30) and healthy controls (n = 12). Dysregulated pathways in PAH include accelerated one carbon metabolism, abnormal tricarboxylic acid (TCA) cycle flux and glutamate metabolism, dysfunctional arginine and nitric oxide pathways, and increased oxidative stress. Functional studies in cells confirmed abnormalities in glucose metabolism, mitochondrial oxygen consumption, and production of reactive oxygen species in PAH. Altogether, the findings indicate that PAH is typified by changes in metabolic pathways that are primarily found in mitochondria.
Collapse
Affiliation(s)
- Weiling Xu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America.
| | - Suzy A A Comhair
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ruoying Chen
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Bo Hu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Yuan Hou
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Yadi Zhou
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Lori A Mavrakis
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Allison J Janocha
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ling Li
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Dongmei Zhang
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Belinda B Willard
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Kewal Asosingh
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Feixiong Cheng
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Serpil C Erzurum
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America. .,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, United States of America.
| |
Collapse
|
224
|
Bastounis EE, Yeh YT, Theriot JA. Subendothelial stiffness alters endothelial cell traction force generation while exerting a minimal effect on the transcriptome. Sci Rep 2019; 9:18209. [PMID: 31796790 PMCID: PMC6890669 DOI: 10.1038/s41598-019-54336-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/01/2019] [Indexed: 02/07/2023] Open
Abstract
Endothelial cells respond to changes in subendothelial stiffness by altering their migration and mechanics, but whether those responses are due to transcriptional reprogramming remains largely unknown. We measured traction force generation and also performed gene expression profiling for two endothelial cell types grown in monolayers on soft or stiff matrices: primary human umbilical vein endothelial cells (HUVEC) and immortalized human microvascular endothelial cells (HMEC-1). Both cell types respond to changes in subendothelial stiffness by increasing the traction stresses they exert on stiffer as compared to softer matrices, and exhibit a range of altered protein phosphorylation or protein conformational changes previously implicated in mechanotransduction. However, the transcriptome has only a minimal role in this conserved biomechanical response. Only few genes were differentially expressed in each cell type in a stiffness-dependent manner, and none were shared between them. In contrast, thousands of genes were differentially regulated in HUVEC as compared to HMEC-1. HUVEC (but not HMEC-1) upregulate expression of TGF-β2 on stiffer matrices, and also respond to application of exogenous TGF-β2 by enhancing their endogenous TGF-β2 expression and their cell-matrix traction stresses. Altogether, these findings provide insights into the relationship between subendothelial stiffness, endothelial mechanics and variation of the endothelial cell transcriptome, and reveal that subendothelial stiffness, while critically altering endothelial cells’ mechanical behavior, minimally affects their transcriptome.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195-1800, USA
| | - Yi-Ting Yeh
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195-1800, USA.
| |
Collapse
|
225
|
Hao S, Matsui Y, Lai ZC, Paulson RF. Yap1 promotes proliferation of transiently amplifying stress erythroid progenitors during erythroid regeneration. Exp Hematol 2019; 80:42-54.e4. [PMID: 31756359 DOI: 10.1016/j.exphem.2019.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 11/18/2022]
Abstract
In contrast to steady-state erythropoiesis, which generates new erythrocytes at a constant rate, stress erythropoiesis rapidly produces a large bolus of new erythrocytes in response to anemic stress. In this study, we illustrate that Yes-associated protein (Yap1) promotes the rapid expansion of a transit-amplifying population of stress erythroid progenitors in vivo and in vitro. Yap1-mutated erythroid progenitors failed to proliferate in the spleen after transplantation into lethally irradiated recipient mice. Additionally, loss of Yap1 impaired the growth of actively proliferating erythroid progenitors in vitro. This role in proliferation is supported by gene expression profiles showing that transiently amplifying stress erythroid progenitors express high levels of genes associated with Yap1 activity and genes induced by Yap1. Furthermore, Yap1 promotes the proliferation of stress erythroid progenitors in part by regulating the expression of key glutamine-metabolizing enzymes. Thus, Yap1 acts as an erythroid regulator that coordinates the metabolic status with the proliferation of erythroid progenitors to promote stress erythropoiesis.
Collapse
Affiliation(s)
- Siyang Hao
- Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA; Center for Molecular Immunology and Infectious Disease at Penn State University, University Park, PA
| | - Yurika Matsui
- Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA
| | - Zhi-Chun Lai
- Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA; Graduate Program in Biochemistry, Microbiology and Molecular Biology, Penn State University, University Park, PA; Department of Biology, Penn State University, University Park, PA
| | - Robert F Paulson
- Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA; Center for Molecular Immunology and Infectious Disease at Penn State University, University Park, PA; Graduate Program in Biochemistry, Microbiology and Molecular Biology, Penn State University, University Park, PA; Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA.
| |
Collapse
|
226
|
Rafikova O, Al Ghouleh I, Rafikov R. Focus on Early Events: Pathogenesis of Pulmonary Arterial Hypertension Development. Antioxid Redox Signal 2019; 31:933-953. [PMID: 31169021 PMCID: PMC6765063 DOI: 10.1089/ars.2018.7673] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/17/2022]
Abstract
Significance: Pulmonary arterial hypertension (PAH) is a progressive disease of the lung vasculature characterized by the proliferation of all vascular wall cell types, including endothelial, smooth muscle, and fibroblasts. The disease rapidly advances into a form with extensive pulmonary vascular remodeling, leading to a rapid increase in pulmonary vascular resistance, which results in right heart failure. Recent Advances: Most current research in the PAH field has been focused on the late stage of the disease, largely due to an urgent need for patient treatment options in clinics. Further, the pathobiology of PAH is multifaceted in the advanced disease, and there has been promising recent progress in identifying various pathological pathways related to the late clinical picture. Critical Issues: Early stage PAH still requires additional attention from the scientific community, and although the survival of patients with early diagnosis is comparatively higher, the disease develops in patients asymptomatically, making it difficult to identify and treat early. Future Directions: There are several reasons to focus on the early stage of PAH. First, the complexity of late stage disease, owing to multiple pathways being activated in a complex system with intra- and intercellular signaling, leads to an unclear picture of the key contributors to the pathobiology. Second, an understanding of early pathophysiological events can increase the ability to identify PAH patients earlier than what is currently possible. Third, the prompt diagnosis of PAH would allow for the therapy to start earlier, which has proved to be a more successful strategy, and it ensures better survival in PAH patients.
Collapse
Affiliation(s)
- Olga Rafikova
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona
| | - Imad Al Ghouleh
- Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ruslan Rafikov
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
227
|
Kumar V, Dong Y, Kumar V, Almawash S, Mahato RI. The use of micelles to deliver potential hedgehog pathway inhibitor for the treatment of liver fibrosis. Am J Cancer Res 2019; 9:7537-7555. [PMID: 31695785 PMCID: PMC6831471 DOI: 10.7150/thno.38913] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/28/2019] [Indexed: 12/11/2022] Open
Abstract
Rationale: Hedgehog (Hh) pathway plays an essential role in liver fibrosis by promoting the proliferation of hepatic stellate cells (HSCs) by enhancing their metabolism via yes-associated protein 1 (YAP1). Despite the presence of several inhibitors, Hh signaling cannot be controlled exclusively due to their poor efficacy and the lack of a suitable delivery system to the injury site. Therefore, it is rationale to develop new potent Hh inhibitors and suitable delivery carriers. Methods: Based on the structure and activity of Hh inhibitor GDC-0449, we replaced its sulfonamide group with two methylpyridine-2yl at amide nitrogen to synthesize MDB5. We compared the Hh pathway inhibition and anti-fibrotic effect of MDB5 with GDC-0449 in vitro. Next, we developed MDB5 loaded micelles using our methoxy poly(ethylene glycol)-blockpoly(2-methyl-2-carboxyl-propylene carbonate-graft-dodecanol (PEG-PCC-g-DC) copolymer and characterized for physicochemical properties. We evaluated the therapeutic efficacy of MDB5 loaded micelles in common bile duct ligation (CBDL) induced liver fibrosis, mouse model. We also determined the intrahepatic distribution of fluorescently labeled micelles after MDB5 treatment. Results: Our results show that MDB5 was more potent in inhibiting Hh pathway components and HSC proliferation in vitro. We successfully developed MDB5 loaded micelles with particle size of 40 ± 10 nm and drug loading up to 10% w/w. MDB5 loaded micelles at the dose of 10 mg/kg were well tolerated by mice, without visible sign of toxicity. The serum enzyme activities elevated by CBDL was significantly decreased by MDB5 loaded micelles compared to GDC-0449 loaded micelles. MDB5 loaded micelles further decreased collagen deposition, HSC activation, and Hh activity and its target genes in the liver. MDB5 loaded micelles also prevented liver sinusoidal endothelial capillarization (LSEC) and therefore restored perfusion between blood and liver cells. Conclusions: Our study provides evidence that MDB5 was more potent in inhibiting Hh pathway in HSC-T6 cells and showed better hepatoprotection in CBDL mice compared to GDC-0449.
Collapse
|
228
|
Spiekerkoetter E, Goncharova EA, Guignabert C, Stenmark K, Kwapiszewska G, Rabinovitch M, Voelkel N, Bogaard HJ, Graham B, Pullamsetti SS, Kuebler WM. Hot topics in the mechanisms of pulmonary arterial hypertension disease: cancer-like pathobiology, the role of the adventitia, systemic involvement, and right ventricular failure. Pulm Circ 2019; 9:2045894019889775. [PMID: 31798835 PMCID: PMC6868582 DOI: 10.1177/2045894019889775] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023] Open
Abstract
In order to intervene appropriately and develop disease-modifying therapeutics for pulmonary arterial hypertension, it is crucial to understand the mechanisms of disease pathogenesis and progression. We herein discuss four topics of disease mechanisms that are currently highly debated, yet still unsolved, in the field of pulmonary arterial hypertension. Is pulmonary arterial hypertension a cancer-like disease? Does the adventitia play an important role in the initiation of pulmonary vascular remodeling? Is pulmonary arterial hypertension a systemic disease? Does capillary loss drive right ventricular failure? While pulmonary arterial hypertension does not replicate all features of cancer, anti-proliferative cancer therapeutics might still be beneficial in pulmonary arterial hypertension if monitored for safety and tolerability. It was recognized that the adventitia as a cell-rich compartment is important in the disease pathogenesis of pulmonary arterial hypertension and should be a therapeutic target, albeit the data are inconclusive as to whether the adventitia is involved in the initiation of neointima formation. There was agreement that systemic diseases can lead to pulmonary arterial hypertension and that pulmonary arterial hypertension can have systemic effects related to the advanced lung pathology, yet there was less agreement on whether idiopathic pulmonary arterial hypertension is a systemic disease per se. Despite acknowledging the limitations of exactly assessing vascular density in the right ventricle, it was recognized that the failing right ventricle may show inadequate vascular adaptation resulting in inadequate delivery of oxygen and other metabolites. Although the debate was not meant to result in a definite resolution of the specific arguments, it sparked ideas about how we might resolve the discrepancies by improving our disease modeling (rodent models, large-animal studies, studies of human cells, tissues, and organs) as well as standardization of the models. Novel experimental approaches, such as lineage tracing and better three-dimensional imaging of experimental as well as human lung and heart tissues, might unravel how different cells contribute to the disease pathology.
Collapse
Affiliation(s)
- Edda Spiekerkoetter
- Division of Pulmonary and Critical Care Medicine, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Elena A. Goncharova
- Pittsburgh Heart, Blood and Vascular Medicine Institute, Pulmonary, Allergy & Critical Care Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christophe Guignabert
- INSERM UMR_S 999, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Kurt Stenmark
- Department of Pediatrics, School of Medicine, University of Colorado, Denver, CO, USA
- Cardio Vascular Pulmonary Research Lab, University of Colorado, Denver, CO, USA
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute, Lung Vascular Research, Medical University of Graz, Graz, Austria
| | - Marlene Rabinovitch
- Division of Pediatric Cardiology, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Norbert Voelkel
- Department of Pulmonary Medicine, Vrije Universiteit MC, Amsterdam, The Netherlands
| | - Harm J. Bogaard
- Department of Pulmonary Medicine, Vrije Universiteit MC, Amsterdam, The Netherlands
| | - Brian Graham
- Pulmonary Sciences and Critical Care, School of Medicine, University of Colorado, Denver, CO, USA
| | - Soni S. Pullamsetti
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité – Universitaetsmedizin Berlin, Berlin, Germany
- The Keenan Research Centre for Biomedical Science at St. Michael's, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
229
|
Bloodworth NC, Clark CR, West JD, Snider JC, Gaskill C, Shay S, Scott C, Bastarache J, Gladson S, Moore C, D'Amico R, Brittain EL, Tanjore H, Blackwell TS, Majka SM, Merryman WD. Bone Marrow-Derived Proangiogenic Cells Mediate Pulmonary Arteriole Stiffening via Serotonin 2B Receptor Dependent Mechanism. Circ Res 2019; 123:e51-e64. [PMID: 30566041 DOI: 10.1161/circresaha.118.313397] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RATIONALE Pulmonary arterial hypertension is a deadly disease of the pulmonary vasculature for which no disease-modifying therapies exist. Small-vessel stiffening and remodeling are fundamental pathological features of pulmonary arterial hypertension that occur early and drive further endovascular cell dysfunction. Bone marrow (BM)-derived proangiogenic cells (PACs), a specialized heterogeneous subpopulation of myeloid lineage cells, are thought to play an important role in pathogenesis. OBJECTIVE To determine whether BM-derived PACs directly contributed to experimental pulmonary hypertension (PH) by promoting small-vessel stiffening through 5-HT2B (serotonin 2B receptor)-mediated signaling. METHODS AND RESULTS We performed BM transplants using transgenic donor animals expressing diphtheria toxin secondary to activation of an endothelial-specific tamoxifen-inducible Cre and induced experimental PH using hypoxia with SU5416 to enhance endovascular injury and ablated BM-derived PACs, after which we measured right ventricular systolic pressures in a closed-chest procedure. BM-derived PAC lineage tracing was accomplished by transplanting BM from transgenic donor animals with fluorescently labeled hematopoietic cells and treating mice with a 5-HT2B antagonist. BM-derived PAC ablation both prevented and reversed experimental PH with SU5416-enhanced endovascular injury, reducing the number of muscularized pulmonary arterioles and normalizing arteriole stiffness as measured by atomic force microscopy. Similarly, treatment with a pharmacological antagonist of 5-HT2B also prevented experimental PH, reducing the number and stiffness of muscularized pulmonary arterioles. PACs accelerated pulmonary microvascular endothelial cell injury response in vitro, and the presence of BM-derived PACs significantly correlated with stiffer pulmonary arterioles in pulmonary arterial hypertension patients and mice with experimental PH. RNA sequencing of BM-derived PACs showed that 5-HT2B antagonism significantly altered biologic pathways regulating cell proliferation, locomotion and migration, and cytokine production and response to cytokine stimulus. CONCLUSIONS Together, our findings illustrate that BM-derived PACs directly contribute to experimental PH with SU5416-enhanced endovascular injury by mediating small-vessel stiffening and remodeling in a 5-HT2B signaling-dependent manner.
Collapse
Affiliation(s)
- Nathaniel C Bloodworth
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Cynthia R Clark
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| | - James D West
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - J Caleb Snider
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Christa Gaskill
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Sheila Shay
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Christine Scott
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Julie Bastarache
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Santhi Gladson
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Christy Moore
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Reid D'Amico
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Evan L Brittain
- Division of Cardiovascular Medicine, Department of Medicine (E.L.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Harikrishna Tanjore
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN.,Department of Veterans Affairs Medical Center, Nashville, TN (T.S.B.)
| | - Susan M Majka
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - W David Merryman
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
230
|
DelNero P, Hopkins BD, Cantley LC, Fischbach C. Cancer metabolism gets physical. Sci Transl Med 2019; 10:10/442/eaaq1011. [PMID: 29794058 DOI: 10.1126/scitranslmed.aaq1011] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 02/22/2018] [Accepted: 04/20/2018] [Indexed: 12/13/2022]
Abstract
Patient-derived culture models enable assessment of drug sensitivity and can connect personalized genomics with therapeutic options. However, their clinical translation is constrained by limited fidelity. We outline how the physical microenvironment regulates cell metabolism and describe how engineered culture systems could enhance the predictive power for precision medicine.
Collapse
Affiliation(s)
- Peter DelNero
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Benjamin D Hopkins
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Lewis C Cantley
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA. .,Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
231
|
Durante W. The Emerging Role of l-Glutamine in Cardiovascular Health and Disease. Nutrients 2019; 11:nu11092092. [PMID: 31487814 PMCID: PMC6769761 DOI: 10.3390/nu11092092] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 08/29/2019] [Accepted: 08/30/2019] [Indexed: 12/29/2022] Open
Abstract
Emerging evidence indicates that l-glutamine (Gln) plays a fundamental role in cardiovascular physiology and pathology. By serving as a substrate for the synthesis of DNA, ATP, proteins, and lipids, Gln drives critical processes in vascular cells, including proliferation, migration, apoptosis, senescence, and extracellular matrix deposition. Furthermore, Gln exerts potent antioxidant and anti-inflammatory effects in the circulation by inducing the expression of heme oxygenase-1, heat shock proteins, and glutathione. Gln also promotes cardiovascular health by serving as an l-arginine precursor to optimize nitric oxide synthesis. Importantly, Gln mitigates numerous risk factors for cardiovascular disease, such as hypertension, hyperlipidemia, glucose intolerance, obesity, and diabetes. Many studies demonstrate that Gln supplementation protects against cardiometabolic disease, ischemia-reperfusion injury, sickle cell disease, cardiac injury by inimical stimuli, and may be beneficial in patients with heart failure. However, excessive shunting of Gln to the Krebs cycle can precipitate aberrant angiogenic responses and the development of pulmonary arterial hypertension. In these instances, therapeutic targeting of the enzymes involved in glutaminolysis such as glutaminase-1, Gln synthetase, glutamate dehydrogenase, and amino acid transaminase has shown promise in preclinical models. Future translation studies employing Gln delivery approaches and/or glutaminolysis inhibitors will determine the success of targeting Gln in cardiovascular disease.
Collapse
Affiliation(s)
- William Durante
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
232
|
Li X, Sun X, Carmeliet P. Hallmarks of Endothelial Cell Metabolism in Health and Disease. Cell Metab 2019; 30:414-433. [PMID: 31484054 DOI: 10.1016/j.cmet.2019.08.011] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 01/13/2023]
Abstract
In 2009, it was postulated that endothelial cells (ECs) would only be able to execute the orders of growth factors if these cells would accordingly adapt their metabolism. Ten years later, it has become clear that ECs, often differently from other cell types, rely on distinct metabolic pathways to survive and form new blood vessels; that manipulation of EC metabolic pathways alone (even without changing angiogenic signaling) suffices to alter vessel sprouting; and that perturbations of these metabolic pathways can underlie excess formation of new blood vessels (angiogenesis) in cancer and ocular diseases. Initial proof of evidence has been provided that targeting (normalizing) these metabolic perturbations in diseased ECs and delivery of metabolites deserve increasing attention as novel therapeutic approaches for inhibiting or stimulating vessel growth in multiple disorders.
Collapse
Affiliation(s)
- Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China.
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Peter Carmeliet
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven B-3000, Belgium.
| |
Collapse
|
233
|
Brittain EL, Thennapan T, Maron BA, Chan SY, Austin ED, Spiekerkoetter E, Bogaard HJ, Guignabert C, Paulin R, Machado RF, Yu PB. Update in Pulmonary Vascular Disease 2016 and 2017. Am J Respir Crit Care Med 2019. [PMID: 29533671 DOI: 10.1164/rccm.201801-0062up] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Evan L Brittain
- 1 Division of Cardiovascular Medicine, Department of Medicine.,2 Vanderbilt Translational and Clinical Cardiovascular Research Center.,3 Pulmonary Vascular Center, Department of Medicine, and
| | | | - Bradley A Maron
- 5 Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,6 Department of Cardiology, Boston VA Healthcare System, Boston, Massachusetts
| | - Stephen Y Chan
- 7 Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Eric D Austin
- 3 Pulmonary Vascular Center, Department of Medicine, and.,8 Pediatric Pulmonary Hypertension Program, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Edda Spiekerkoetter
- 9 Division of Pulmonary and Critical Care Medicine, Department of Medicine, and.,10 Vera Moulton Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University, Stanford, California
| | - Harm J Bogaard
- 11 Pulmonary Hypertension Expert Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Christophe Guignabert
- 12 INSERM UMR-S 999, Le Plessis-Robinson, France.,13 Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Roxane Paulin
- 14 Quebec Heart and Lung Institute, Laval University, Quebec, Quebec, Canada; and
| | - Roberto F Machado
- 15 Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Paul B Yu
- 5 Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
234
|
Mercurio V, Bianco A, Campi G, Cuomo A, Diab N, Mancini A, Parrella P, Petretta M, Hassoun PM, Bonaduce D. New Drugs, Therapeutic Strategies, and Future Direction for the Treatment of Pulmonary Arterial Hypertension. Curr Med Chem 2019; 26:2844-2864. [DOI: 10.2174/0929867325666180201095743] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/21/2017] [Accepted: 12/21/2017] [Indexed: 12/20/2022]
Abstract
Despite recent advances in Pulmonary Arterial Hypertension (PAH) treatment, this condition is still characterized by an extremely poor prognosis. In this review, we discuss the use of newly-approved drugs for PAH treatment with already known mechanisms of action (macitentan), innovative targets (riociguat and selexipag), and novel therapeutic approaches with initial up-front combination therapy. Secondly, we describe new potential signaling pathways and investigational drugs with promising role in the treatment of PAH.
Collapse
Affiliation(s)
- Valentina Mercurio
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Anna Bianco
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Giacomo Campi
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Alessandra Cuomo
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Nermin Diab
- University of Ottawa, Department of Medicine, Ottawa, ON, Canada
| | - Angela Mancini
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Paolo Parrella
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Mario Petretta
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Paul M. Hassoun
- Johns Hopkins University, Division of Pulmonary and Critical Care Medicine, Baltimore, MD, United States
| | - Domenico Bonaduce
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| |
Collapse
|
235
|
Zheng Y, Pan D. The Hippo Signaling Pathway in Development and Disease. Dev Cell 2019; 50:264-282. [PMID: 31386861 PMCID: PMC6748048 DOI: 10.1016/j.devcel.2019.06.003] [Citation(s) in RCA: 527] [Impact Index Per Article: 105.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/23/2019] [Accepted: 06/09/2019] [Indexed: 12/13/2022]
Abstract
The Hippo signaling pathway regulates diverse physiological processes, and its dysfunction has been implicated in an increasing number of human diseases, including cancer. Here, we provide an updated review of the Hippo pathway; discuss its roles in development, homeostasis, regeneration, and diseases; and highlight outstanding questions for future investigation and opportunities for Hippo-targeted therapies.
Collapse
Affiliation(s)
- Yonggang Zheng
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA.
| |
Collapse
|
236
|
Wu D, Birukov K. Endothelial Cell Mechano-Metabolomic Coupling to Disease States in the Lung Microvasculature. Front Bioeng Biotechnol 2019; 7:172. [PMID: 31380363 PMCID: PMC6658821 DOI: 10.3389/fbioe.2019.00172] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/03/2019] [Indexed: 12/15/2022] Open
Abstract
Lungs are the most vascular part of humans, accepting the totality of cardiac output in a volume much smaller than the body itself. Due to this cardiac output, the lung microvasculature is subject to mechanical forces including shear stress and cyclic stretch that vary with the cardiac and breathing cycle. Vessels are surrounded by extracellular matrix which dictates the stiffness which endothelial cells also sense and respond to. Shear stress, stiffness, and cyclic stretch are known to influence endothelial cell state. At high shear stress, endothelial cells exhibit cell quiescence marked by low inflammatory markers and high nitric oxide synthesis, whereas at low shear stress, endothelial cells are thought to "activate" into a pro-inflammatory state and have low nitric oxide. Shear stress' profound effect on vascular phenotype is most apparent in the arterial vasculature and in the pathophysiology of vascular inflammation. To conduct the flow of blood from the right heart, the lung microvasculature must be rigid yet compliant. It turns out that excessive substrate rigidity or stiffness is important in the development of pulmonary hypertension and chronic fibrosing lung diseases via excessive cell proliferation or the endothelial-mesenchymal transition. Recently, a new body of literature has evolved that couples mechanical sensing to endothelial phenotypic changes through metabolic signaling in clinically relevant contexts such as pulmonary hypertension, lung injury syndromes, as well as fibrosis, which is the focus of this review. Stretch, like flow, has profound effect on endothelial phenotype; metabolism studies due to stretch are in their infancy.
Collapse
Affiliation(s)
- David Wu
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Konstantin Birukov
- Department of Anesthesia, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
237
|
Lu X, Gong J, Dennery PA, Yao H. Endothelial-to-mesenchymal transition: Pathogenesis and therapeutic targets for chronic pulmonary and vascular diseases. Biochem Pharmacol 2019; 168:100-107. [PMID: 31251941 DOI: 10.1016/j.bcp.2019.06.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/20/2019] [Indexed: 02/07/2023]
Abstract
Endothelial-to-mesenchymal transition (EndoMT) is a process of transdifferentiation where endothelial cells gradually adopt the phenotypic characteristics of mesenchymal cells. This phenomenon was first discovered in embryonic heart development. The mechanisms underlying EndoMT are due to the activation of transforming growth factor-β, bone morphogenetic protein, Wingless/Integrated, or Notch signaling pathways. The EndoMT can be modulated by pathological processes, including inflammation, disturbed shear stress, vascular stiffness, and metabolic dysregulation. Recent studies have shown that EndoMT is implicated in the pathogenesis of chronic lung diseases, including pulmonary hypertension and lung fibrosis. Lung pathology of bronchopulmonary dysplasia can be mimicked in rodents exposed to hyperoxia as neonates. Although hyperoxic exposure reduces an endothelial cell marker platelet and endothelial cell adhesion molecule but increases a mesenchymal cell biomarker α-smooth muscle actin in vitro in human pulmonary endothelial cells, there is no direct evidence showing EndoMT in the development of bronchopulmonary dysplasia. Both pulmonary hypertension and lung fibrosis occur in long-term survivors with bronchopulmonary dysplasia. In this review, we discuss the EndoMT and its modulation by pathological processes. We then focus on the role of EndoMT in the pathogenesis of these chronic lung diseases, and discuss therapeutic approaches targeting the EndoMT using its negative regulators.
Collapse
Affiliation(s)
- Xuexin Lu
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, United States
| | - Jiannan Gong
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, United States; Department of Respiratory and Critical Medicine, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Phyllis A Dennery
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, United States; Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Hongwei Yao
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, United States.
| |
Collapse
|
238
|
Bourgeois A, Bonnet S, Breuils-Bonnet S, Habbout K, Paradis R, Tremblay E, Lampron MC, Orcholski ME, Potus F, Bertero T, Peterlini T, Chan SY, Norris KA, Paulin R, Provencher S, Boucherat O. Inhibition of CHK 1 (Checkpoint Kinase 1) Elicits Therapeutic Effects in Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2019; 39:1667-1681. [PMID: 31092016 DOI: 10.1161/atvbaha.119.312537] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Pulmonary arterial hypertension (PAH) is a debilitating disease associated with progressive vascular remodeling of distal pulmonary arteries leading to elevation of pulmonary artery pressure, right ventricular hypertrophy, and death. Although presenting high levels of DNA damage that normally jeopardize their viability, pulmonary artery smooth muscle cells (PASMCs) from patients with PAH exhibit a cancer-like proproliferative and apoptosis-resistant phenotype accounting for vascular lumen obliteration. In cancer cells, overexpression of the serine/threonine-protein kinase CHK1 (checkpoint kinase 1) is exploited to counteract the excess of DNA damage insults they are exposed to. This study aimed to determine whether PAH-PASMCs have developed an orchestrated response mediated by CHK1 to overcome DNA damage, allowing cell survival and proliferation. Approach and Results: We demonstrated that CHK1 expression is markedly increased in isolated PASMCs and distal PAs from patients with PAH compared with controls, as well as in multiple complementary animal models recapitulating the disease, including monocrotaline rats and the simian immunodeficiency virus-infected macaques. Using a pharmacological and molecular loss of function approach, we showed that CHK1 promotes PAH-PASMCs proliferation and resistance to apoptosis. In addition, we found that inhibition of CHK1 induces downregulation of the DNA repair protein RAD 51 and severe DNA damage. In vivo, we provided evidence that pharmacological inhibition of CHK1 significantly reduces vascular remodeling and improves hemodynamic parameters in 2 experimental rat models of PAH. CONCLUSIONS Our results show that CHK1 exerts a proproliferative function in PAH-PASMCs by mitigating DNA damage and suggest that CHK1 inhibition may, therefore, represent an attractive therapeutic option for patients with PAH.
Collapse
Affiliation(s)
- Alice Bourgeois
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.)
| | - Sébastien Bonnet
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.).,Department of Medicine, Université Laval, QC, Canada (S.P., O.B., S.B.)
| | - Sandra Breuils-Bonnet
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.)
| | - Karima Habbout
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.)
| | - Renée Paradis
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.)
| | - Eve Tremblay
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.)
| | - Marie-Claude Lampron
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.)
| | - Mark E Orcholski
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.)
| | - Francois Potus
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.)
| | - Thomas Bertero
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging Nice (IRCAN), University Côte d'Azur, France (T.B.)
| | - Thibaut Peterlini
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.)
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA (S.Y.C.)
| | - Karen A Norris
- Center for Vaccines and Immunology, University of Georgia, Athens (K.A.N.)
| | - Roxane Paulin
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.)
| | - Steeve Provencher
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.).,Department of Medicine, Université Laval, QC, Canada (S.P., O.B., S.B.)
| | - Olivier Boucherat
- From the Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada (A.B., S.B., S.B-B., K.H., R.P., E.T., M.C.L., M.E.O., F.P., T.P., R.P., S.P., O.B.).,Department of Medicine, Université Laval, QC, Canada (S.P., O.B., S.B.)
| |
Collapse
|
239
|
Bertero T, Perk D, Chan SY. The molecular rationale for therapeutic targeting of glutamine metabolism in pulmonary hypertension. Expert Opin Ther Targets 2019; 23:511-524. [PMID: 31055988 DOI: 10.1080/14728222.2019.1615438] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Pulmonary hypertension (PH) is a deadly enigmatic disease with increasing prevalence. Cellular pathologic hallmarks of PH are driven at least partly by metabolic rewiring, but details are just emerging. The discovery that vascular matrix stiffening can mechanically activate the glutaminase (GLS) enzyme and serve as a pathogenic mechanism of PH has advanced our understanding of the complex role of glutamine in PH. It has also offered a novel therapeutic target for development as a next-generation drug for this disease. Area covered: This review discusses the cellular contribution of glutamine metabolism to PH together with the possible therapeutic application of pharmacologic GLS inhibitors in this disease. Expert opinion: Despite advances in our understanding of glutamine metabolism in PH, questions remain unanswered regarding the development of therapies targeting glutamine in PH. The comprehensive mechanisms by which glutamine metabolism rewiring influences pulmonary vascular cell behavior to drive PH are incompletely understood. Because glutamine metabolism exhibits a variety of functions in organ repair and homeostasis, a better understanding of the overall risk-benefit ratio of these strategies with long-term follow-up is needed. This knowledge should pave the way for the design of new strategies to prevent and hopefully even regress PH.
Collapse
Affiliation(s)
- Thomas Bertero
- a Institute of Molecular and Cellular Pharmacology , Université Côte d'Azur , Valbonne , France
| | - Dror Perk
- b Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine , University of Pittsburgh Medical Center , Pittsburgh , PA , USA
| | - Stephen Y Chan
- b Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine , University of Pittsburgh Medical Center , Pittsburgh , PA , USA
| |
Collapse
|
240
|
Wu D, Woods PS, Duong HT, Mutlu GM. Role of Cellular Metabolism in Pulmonary Diseases. Am J Respir Cell Mol Biol 2019; 59:127-129. [PMID: 29634283 DOI: 10.1165/rcmb.2018-0103ro] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- David Wu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois
| | - Parker S Woods
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois
| | - Heng T Duong
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois
| |
Collapse
|
241
|
McGraw MD, Sherlock LG, Bailey KL, Abman SH. Developmental Origins of Chronic Lung Diseases. Mechanical Stretch, Micro-RNAs, and Hydrogels. Am J Respir Cell Mol Biol 2019; 59:267-270. [PMID: 29641225 DOI: 10.1165/rcmb.2018-0092ro] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Matthew D McGraw
- 1 Breathing Institute and Pediatric Heart-Lung Center, Division of Pediatrics, and
| | - Laura G Sherlock
- 2 Division of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado; and
| | - Kolene L Bailey
- 3 Division of Pulmonary and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Steven H Abman
- 1 Breathing Institute and Pediatric Heart-Lung Center, Division of Pediatrics, and
| |
Collapse
|
242
|
Boopathy GTK, Hong W. Role of Hippo Pathway-YAP/TAZ Signaling in Angiogenesis. Front Cell Dev Biol 2019; 7:49. [PMID: 31024911 PMCID: PMC6468149 DOI: 10.3389/fcell.2019.00049] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a highly coordinated process of formation of new blood vessels from pre-existing blood vessels. The process of development of the proper vascular network is a complex process that is crucial for the vertebrate development. Several studies have defined essential roles of Hippo pathway-YAP/TAZ in organ size control, tissue regeneration, and self-renewal. Thus Hippo pathway is one of the central components in tissue homeostasis. There are mounting evidences on the eminence of Hippo pathway-YAP/TAZ in angiogenesis in multiple model organisms. Hippo pathway-YAP/TAZ is now demonstrated to regulate endothelial cell proliferation, migration and survival; subsequently regulating vascular sprouting, vascular barrier formation, and vascular remodeling. Major intracellular signaling programs that regulate angiogenesis concomitantly activate YAP/TAZ to regulate key events in angiogenesis. In this review, we provide a brief overview of the recent findings in the Hippo pathway and YAP/TAZ signaling in angiogenesis.
Collapse
Affiliation(s)
- Gandhi T K Boopathy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
243
|
Ren X, Johns RA, Gao WD. EXPRESS: Right Heart in Pulmonary Hypertension: From Adaptation to Failure. Pulm Circ 2019; 9:2045894019845611. [PMID: 30942134 PMCID: PMC6681271 DOI: 10.1177/2045894019845611] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/27/2019] [Indexed: 01/24/2023] Open
Abstract
Right ventricular (RV) failure (RVF) has garnered significant attention in recent years because of its negative impact on clinical outcomes in patients with pulmonary hypertension (PH). PH triggers a series of events, including activation of several signaling pathways that regulate cell growth, metabolism, extracellular matrix remodeling, and energy production. These processes render the RV adaptive to PH. However, RVF develops when PH persists, accompanied by RV ischemia, alterations in substrate and mitochondrial energy metabolism, increased free oxygen radicals, increased cell loss, downregulation of adrenergic receptors, increased inflammation and fibrosis, and pathologic microRNAs. Diastolic dysfunction is also an integral part of RVF. Emerging non-invasive technologies such as molecular or metallic imaging, cardiac MRI, and ultrafast Doppler coronary flow mapping will be valuable tools to monitor RVF, especially the transition to RVF. Most PH therapies cannot treat RVF once it has occurred. A variety of therapies are available to treat acute and chronic RVF, but they are mainly supportive, and no effective therapy directly targets the failing RV. Therapies that target cell growth, cellular metabolism, oxidative stress, and myocyte regeneration are being tested preclinically. Future research should include establishing novel RVF models based on existing models, increasing use of human samples, creating human stem cell-based in vitro models, and characterizing alterations in cardiac excitation–contraction coupling during transition from adaptive RV to RVF. More successful strategies to manage RVF will likely be developed as we learn more about the transition from adaptive remodeling to maladaptive RVF in the future.
Collapse
Affiliation(s)
- Xianfeng Ren
- Department of Anesthesiology,
China-Japan
Friendship Hospital, Beijing, China
| | - Roger A. Johns
- Department of Anesthesiology and
Critical Care Medicine,
Johns
Hopkins University School of Medicine,
Baltimore, MD, USA
| | - Wei Dong Gao
- Department of Anesthesiology and
Critical Care Medicine,
Johns
Hopkins University School of Medicine,
Baltimore, MD, USA
| |
Collapse
|
244
|
Bertero T, Gaggioli C. Mechanical forces rewire metabolism in the tumor niche. Mol Cell Oncol 2019; 6:1592945. [PMID: 31131310 DOI: 10.1080/23723556.2019.1592945] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 10/27/2022]
Abstract
Tumor niche extracellular matrix stiffening and tumor cell metabolic reprogramming are two fundamental mediators of tumor progression. We recently elucidated a mechanistic interconnection between mechanotransduction and tumor metabolic rewiring in cancer. We demonstrated a stiffness-dependent amino acid crosstalk between stromal and cancer cells that fuels tumor progression and metastatic spreading.
Collapse
Affiliation(s)
- Thomas Bertero
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis, France
| | | |
Collapse
|
245
|
Yan D, Li G, Zhang Y, Liu Y. Angiotensin-converting enzyme 2 activation suppresses pulmonary vascular remodeling by inducing apoptosis through the Hippo signaling pathway in rats with pulmonary arterial hypertension. Clin Exp Hypertens 2019; 41:589-598. [PMID: 30806090 DOI: 10.1080/10641963.2019.1583247] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objective: To investigate the effects of angiotensin-converting enzyme 2 (ACE2) activation on pulmonary arterial cell apoptosis during pulmonary vascular remodeling associated with pulmonary arterial hypertension (PAH) and to elucidate potential mechanisms related to Hippo signaling. Methods: PAH model was developed by injecting monocrotaline combined with left pneumonectomy using Sprague-Dawley rat. Then, resorcinolnaphthalein (Res; ACE2 activator), MLN-4760 (ACE2 inhibitor), A-779 (Mas inhibitor), and 4-((5,10-dimethyl-6-oxo-6,10-dihydro-5H-pyrimido[5,4-b]thieno[3,2-e][1,4]diazepin-2-yl)amino) benzenesulfonamide (XMU-MP-1; MST1/2 inhibitor) were administered via continuous subcutaneous or intraperitoneal injection for 3 weeks. Animals were randomly divided into six groups: control, PAH, PAH+Res, PAH+Res+MLN-4760, PAH+Res+A-779, and PAH+Res+XMU-MP-1. On 21 day, hemodynamics and pathologic lesions were evaluated. Apoptosis and apoptosis-associated proteins were detected by TUNEL and western blotting. ACE2 activity and Hippo pathway components including large tumor suppressor 1 (LATS1), Yes-associated protein (Yap), and phosphorylated Yap (p-Yap) were investigated by fluorogenic peptide assays and western blotting. Results: In the PAH models, the mean pulmonary arterial pressure, right ventricular hypertrophy index, pulmonary vascular remodeling, anti-apoptotic protein Bcl-2 and Yap were all increased but the pulmonary arterial cell apoptosis, pro-apoptotic proteins caspase-3 and Bax were lower. ACE2 activation significantly ameliorated pulmonary arterial remodeling, this action was related to increased apoptosis and up-regulation of LATS1 and p-Yap. These protective effects were mitigated by the co-administration of A779 or MLN-4760. Moreover, inhibiting the Hippo/LATS1/Yap pathway with XMU-MP-1 blocked apoptosis in pulmonary vascular cells induced by ACE2 activation during the prevention of PAH. Conclusions: Our findings suggest that ACE2 activation attenuates pulmonary vascular remodeling by inducing pulmonary arterial cell apoptosis via Hippo/Yap signaling during the development of PAH.
Collapse
Affiliation(s)
- Daole Yan
- a Pediatric Cardiac Center , Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| | - Gang Li
- a Pediatric Cardiac Center , Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| | - Yaozhong Zhang
- a Pediatric Cardiac Center , Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| | - Yinglong Liu
- a Pediatric Cardiac Center , Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| |
Collapse
|
246
|
Torrino S, Roustan FR, Kaminski L, Bertero T, Pisano S, Ambrosetti D, Dufies M, Uhler JP, Lemichez E, Mettouchi A, Gesson M, Laurent K, Gaggioli C, Michiels JF, Lamaze C, Bost F, Clavel S. UBTD1 is a mechano-regulator controlling cancer aggressiveness. EMBO Rep 2019; 20:embr.201846570. [PMID: 30804013 DOI: 10.15252/embr.201846570] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 01/18/2019] [Accepted: 01/29/2019] [Indexed: 12/18/2022] Open
Abstract
Ubiquitin domain-containing protein 1 (UBTD1) is highly evolutionary conserved and has been described to interact with E2 enzymes of the ubiquitin-proteasome system. However, its biological role and the functional significance of this interaction remain largely unknown. Here, we demonstrate that depletion of UBTD1 drastically affects the mechanical properties of epithelial cancer cells via RhoA activation and strongly promotes their aggressiveness. On a stiff matrix, UBTD1 expression is regulated by cell-cell contacts, and the protein is associated with β-catenin at cell junctions. Yes-associated protein (YAP) is a major cell mechano-transducer, and we show that UBTD1 is associated with components of the YAP degradation complex. Interestingly, UBTD1 promotes the interaction of YAP with its E3 ubiquitin ligase β-TrCP Consequently, in cancer cells, UBTD1 depletion decreases YAP ubiquitylation and triggers robust ROCK2-dependent YAP activation and downstream signaling. Data from lung and prostate cancer patients further corroborate the in cellulo results, confirming that low levels of UBTD1 are associated with poor patient survival, suggesting that biological functions of UBTD1 could be beneficial in limiting cancer progression.
Collapse
Affiliation(s)
- Stéphanie Torrino
- Centre Méditerranéen de Médecine Moléculaire (C3M), Inserm U1065, Nice Cedex 3, France
| | - François-René Roustan
- Centre Méditerranéen de Médecine Moléculaire (C3M), Inserm U1065, Nice Cedex 3, France
| | - Lisa Kaminski
- Centre Méditerranéen de Médecine Moléculaire (C3M), Inserm U1065, Nice Cedex 3, France
| | - Thomas Bertero
- Institute for Research on Cancer and Aging, Nice (IRCAN), CNRS, UMR7284/INSERM U1081, Université Côte d'Azur, Nice, France
| | - Sabrina Pisano
- Institute for Research on Cancer and Aging, Nice (IRCAN), CNRS, UMR7284/INSERM U1081, Université Côte d'Azur, Nice, France
| | - Damien Ambrosetti
- Department of Pathology, Nice University Hospital University of Nice Sophia Antipolis, Nice, France
| | - Maeva Dufies
- Institute for Research on Cancer and Aging, Nice (IRCAN), CNRS, UMR7284/INSERM U1081, Université Côte d'Azur, Nice, France
| | - Jay P Uhler
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Emmanuel Lemichez
- Département de Microbiologie, Institut Pasteur, Unité des Toxines Bactériennes, Université Paris Descartes, Paris, France
| | - Amel Mettouchi
- Département de Microbiologie, Institut Pasteur, Unité des Toxines Bactériennes, Université Paris Descartes, Paris, France
| | - Maeva Gesson
- Centre Méditerranéen de Médecine Moléculaire (C3M), Inserm U1065, Nice Cedex 3, France
| | - Kathiane Laurent
- Centre Méditerranéen de Médecine Moléculaire (C3M), Inserm U1065, Nice Cedex 3, France
| | - Cedric Gaggioli
- Institute for Research on Cancer and Aging, Nice (IRCAN), CNRS, UMR7284/INSERM U1081, Université Côte d'Azur, Nice, France
| | - Jean-Francois Michiels
- Department of Pathology, Nice University Hospital University of Nice Sophia Antipolis, Nice, France
| | - Christophe Lamaze
- CNRS UMR3666, INSERM U1143, Membrane Dynamics and Mechanics of Intracellular Signaling Laboratory, Institut Curie - Centre de Recherche, PSL Research University, Paris, France
| | - Frédéric Bost
- Centre Méditerranéen de Médecine Moléculaire (C3M), Inserm U1065, Nice Cedex 3, France
| | - Stéphan Clavel
- Centre Méditerranéen de Médecine Moléculaire (C3M), Inserm U1065, Nice Cedex 3, France
| |
Collapse
|
247
|
Charbonier FW, Zamani M, Huang NF. Endothelial Cell Mechanotransduction in the Dynamic Vascular Environment. ADVANCED BIOSYSTEMS 2019; 3:e1800252. [PMID: 31328152 PMCID: PMC6640152 DOI: 10.1002/adbi.201800252] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Indexed: 12/11/2022]
Abstract
The vascular endothelial cells (ECs) that line the inner layer of blood vessels are responsible for maintaining vascular homeostasis under physiological conditions. In the presence of disease or injury, ECs can become dysfunctional and contribute to a progressive decline in vascular health. ECs are constantly exposed to a variety of dynamic mechanical stimuli, including hemodynamic shear stress, pulsatile stretch, and passive signaling cues derived from the extracellular matrix. This review describes the molecular mechanisms by which ECs perceive and interpret these mechanical signals. The translational applications of mechanosensing are then discussed in the context of endothelial-to-mesenchymal transition and engineering of vascular grafts.
Collapse
Affiliation(s)
- Frank W. Charbonier
- Department of Mechanical Engineering, Stanford University, Stanford, CA, 94305
| | - Maedeh Zamani
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305
| | - Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305
- Stanford University, 300 Pasteur Drive, MC 5407, Stanford, CA 94305-5407, USA
| |
Collapse
|
248
|
Humbert M, Guignabert C, Bonnet S, Dorfmüller P, Klinger JR, Nicolls MR, Olschewski AJ, Pullamsetti SS, Schermuly RT, Stenmark KR, Rabinovitch M. Pathology and pathobiology of pulmonary hypertension: state of the art and research perspectives. Eur Respir J 2019; 53:13993003.01887-2018. [PMID: 30545970 PMCID: PMC6351340 DOI: 10.1183/13993003.01887-2018] [Citation(s) in RCA: 762] [Impact Index Per Article: 152.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 12/21/2022]
Abstract
Clinical and translational research has played a major role in advancing our understanding of pulmonary hypertension (PH), including pulmonary arterial hypertension and other forms of PH with severe vascular remodelling (e.g. chronic thromboembolic PH and pulmonary veno-occlusive disease). However, PH remains an incurable condition with a high mortality rate, underscoring the need for a better transfer of novel scientific knowledge into healthcare interventions. Herein, we review recent findings in pathology (with the questioning of the strict morphological categorisation of various forms of PH into pre- or post-capillary involvement of pulmonary vessels) and cellular mechanisms contributing to the onset and progression of pulmonary vascular remodelling associated with various forms of PH. We also discuss ways to improve management and to support and optimise drug development in this research field.
Collapse
Affiliation(s)
- Marc Humbert
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Le Plessis-Robinson, France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Le Plessis-Robinson, France
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut de Cardiologie et de Pneumologie de Quebec, Quebec City, QC, Canada.,Dept of Medicine, Université Laval, Quebec City, QC, Canada
| | - Peter Dorfmüller
- Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999, Le Plessis-Robinson, France.,Pathology Dept, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - James R Klinger
- Division of Pulmonary, Critical Care and Sleep Medicine, Dept of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Mark R Nicolls
- Cardiovascular Institute, Dept of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.,Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Stanford University School of Medicine/VA Palo Alto, Palo Alto, CA, USA.,The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford, CA, USA
| | - Andrea J Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Soni S Pullamsetti
- Max Planck Institute for Heart and Lung Research Bad Nauheim, Bad Nauheim, Germany.,Justus-Liebig University Giessen, Excellence Cluster Cardio Pulmonary Institute (CPI), Giessen, Germany
| | - Ralph T Schermuly
- University of Giessen and Marburg Lung Centre (UGMLC), Justus-Liebig University Giessen and Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio Pulmonary Institute (CPI), Giessen, Germany
| | - Kurt R Stenmark
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado, Denver, CO, USA
| | - Marlene Rabinovitch
- Cardiovascular Institute, Dept of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.,Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Stanford University School of Medicine/VA Palo Alto, Palo Alto, CA, USA.,The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford, CA, USA
| |
Collapse
|
249
|
Tumor-Stroma Mechanics Coordinate Amino Acid Availability to Sustain Tumor Growth and Malignancy. Cell Metab 2019; 29:124-140.e10. [PMID: 30293773 PMCID: PMC6432652 DOI: 10.1016/j.cmet.2018.09.012] [Citation(s) in RCA: 254] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 05/30/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022]
Abstract
Dysregulation of extracellular matrix (ECM) deposition and cellular metabolism promotes tumor aggressiveness by sustaining the activity of key growth, invasion, and survival pathways. Yet mechanisms by which biophysical properties of ECM relate to metabolic processes and tumor progression remain undefined. In both cancer cells and carcinoma-associated fibroblasts (CAFs), we found that ECM stiffening mechanoactivates glycolysis and glutamine metabolism and thus coordinates non-essential amino acid flux within the tumor niche. Specifically, we demonstrate a metabolic crosstalk between CAF and cancer cells in which CAF-derived aspartate sustains cancer cell proliferation, while cancer cell-derived glutamate balances the redox state of CAFs to promote ECM remodeling. Collectively, our findings link mechanical stimuli to dysregulated tumor metabolism and thereby highlight a new metabolic network within tumors in which diverse fuel sources are used to promote growth and aggressiveness. Furthermore, this study identifies potential metabolic drug targets for therapeutic development in cancer.
Collapse
|
250
|
Komatsu N, Kajiya M, Motoike S, Takewaki M, Horikoshi S, Iwata T, Ouhara K, Takeda K, Matsuda S, Fujita T, Kurihara H. Type I collagen deposition via osteoinduction ameliorates YAP/TAZ activity in 3D floating culture clumps of mesenchymal stem cell/extracellular matrix complexes. Stem Cell Res Ther 2018; 9:342. [PMID: 30526677 PMCID: PMC6286508 DOI: 10.1186/s13287-018-1085-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/10/2018] [Accepted: 11/20/2018] [Indexed: 01/30/2023] Open
Abstract
Background Three-dimensional (3D) floating culture clumps of mesenchymal stem cell (MSC)/extracellular matrix (ECM) complexes (C-MSCs) consist of cells and self-produced ECM. Previous studies have demonstrated that C-MSCs can be transplanted into bony lesions without an artificial scaffold to induce bone regeneration. Moreover, osteoinductive medium (OIM)-treated C-MSCs (OIM-C-MSCs) have shown rapid and increased new bone formation in vivo. To apply OIM-C-MSCs for novel bone regenerative cell therapy, their cellular properties at the molecular level must be elucidated. The transcriptional co-activators yes-associated protein/transcriptional co-activator with PDZ-binding motif (YAP/TAZ) have been recognized as key players in the mechanotransduction cascade, controlling cell lineage commitment in MSCs. It is plausible that 3D C-MSCs/OIM-C-MSCs cultured in floating conditions could provide distinct microenvironments compared to conventional 2D culture systems and thereby induce unique mechanotransduction cascades. Therefore, this study investigated the YAP/TAZ activity in 3D-cultured C-MSCs/OIM-C-MSCs in floating conditions. Methods Human bone marrow-derived MSCs were cultured in growth medium supplemented with ascorbic acid. To obtain C-MSCs, confluent cells that had formed on the cellular sheet were scratched using a micropipette tip and were then torn off. The sheet was rolled to make round clumps of cells. Then, YAP/TAZ activity, filamentous actin (F-actin) integrity, collagen type I (COL1) production, and the differentiation potency in 3D floating culture C-MSCs/OIM-C-MSCs were analyzed. Results C-MSCs cultured in floating conditions lost their actin cytoskeleton to downregulate YAP/TAZ activity, which directed cells to undergo adipogenesis/chondrogenesis. OIM treatment induced abundant COL1 deposition, which facilitated Intβ1-dependent actin fiber formation and YAP/TAZ activity to elevate the expression levels of osteogenic master transcriptional factor runt-related transcription factor 2 (RUNX2) mRNA in C-MSCs. Importantly, elevation of YAP/TAZ activity via OIM was associated with COL1 deposition and F-actin integrity, suggesting a positive feedback loop in OIM-C-MSCs. Conclusion These findings suggest that OIM-C-MSCs, which form a unique microenvironment that maintains high YAP/TAZ activity, can serve as better candidates for bone regenerative cell therapy than C-MSCs. Electronic supplementary material The online version of this article (10.1186/s13287-018-1085-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nao Komatsu
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan.
| | - Souta Motoike
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan
| | - Manabu Takewaki
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan
| | - Susumu Horikoshi
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan
| | - Tomoyuki Iwata
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan
| | - Katsuhiro Takeda
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan
| | - Shinji Matsuda
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan
| | - Tsuyoshi Fujita
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan
| | - Hidemi Kurihara
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, Hiroshima, 734-8553, Japan
| |
Collapse
|