51
|
Cornwell JA, Nordon RE, Harvey RP. Analysis of cardiac stem cell self-renewal dynamics in serum-free medium by single cell lineage tracking. Stem Cell Res 2018; 28:115-124. [PMID: 29455006 DOI: 10.1016/j.scr.2018.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 02/03/2023] Open
Abstract
Cardiac colony forming unit-fibroblasts (cCFU-F) are a population of stromal cells residing within the SCA1+/PDGFRα+/CD31- fraction of adult mouse hearts, and which have functional characteristics akin to bone marrow mesenchymal stem cells. We hypothesise that they participate in cardiac homeostasis and repair through their actions as lineage progenitors and paracrine signaling hubs. However, cCFU-F are rare and there are no specific markers for these cells, making them challenging to study. cCFU can self-renew in vitro, although the common use of serum has made it difficult to identify cytokines that maintain lineage identity and self-renewal ability. Cell heterogeneity is an additional confounder as cCFU-F cultures are metastable. Here, we address these limitations by identifying serum-free medium (SFM) for growth, and by using cCFU-F isolated from PdgfraGFP/+ mice to record fate outcomes, morphology and PDGFRα expression for hundreds of single cells over time. We show that SFM supplemented with basic fibroblast growth factor, transforming growth factor-β and platelet-derived growth factor, enhanced cCFU-F colony formation and long-term self-renewal, while maintaining cCFU-F potency. cCFU-F cultured in SFM maintained a higher proportion of PDGFRα+ cells, a marker of self-renewing cCFU-F, by increasing Pdgfra-GFP+ divisions and reducing the probability of spontaneous myofibroblast differentiation.
Collapse
Affiliation(s)
- J A Cornwell
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia; Australian Research Council Special Research Initiative in Stem Cell Science - Stem Cells, Australia; Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia; Department of Life Sciences, Faculty of Dentistry, University of Sydney, Westmead Centre for Oral Health, Westmead Hospital, Westmead 2145, Australia
| | - R E Nordon
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia; Australian Research Council Special Research Initiative in Stem Cell Science - Stem Cells, Australia.
| | - R P Harvey
- Australian Research Council Special Research Initiative in Stem Cell Science - Stem Cells, Australia; Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia; School of Biological and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
52
|
Abstract
Major cardiovascular events including myocardial infarction (MI) continue to dominate morbidity rates in the developed world. Although multiple device therapies and various pharmacological agents have been shown to improve patient care and reduce mortality rates, clinicians and researchers alike still lack a true panacea to regenerate damaged cardiac tissue. Over the previous two to three decades, cardiovascular stem cell therapies have held great promise. Several stem cell-based approaches have now been shown to improve ventricular function and are documented in preclinical animal models as well as phase I and phase II clinical trials. More recently, the cardiac progenitor cell has begun to gain momentum as an ideal candidate for stem cell therapy in heart disease. Here, we will highlight the most recent advances in cardiac stem/progenitor cell biology in regard to both the basics and applied settings.
Collapse
|
53
|
Platelet-Derived Growth Factor Receptor-Alpha Expressing Cardiac Progenitor Cells Can Be Derived from Previously Cryopreserved Human Heart Samples. Stem Cells Dev 2018; 27:184-198. [DOI: 10.1089/scd.2017.0082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
54
|
Marotta P, Cianflone E, Aquila I, Vicinanza C, Scalise M, Marino F, Mancuso T, Torella M, Indolfi C, Torella D. Combining cell and gene therapy to advance cardiac regeneration. Expert Opin Biol Ther 2018; 18:409-423. [PMID: 29347847 DOI: 10.1080/14712598.2018.1430762] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The characterization of multipotent endogenous cardiac stem cells (eCSCs) and the breakthroughs of somatic cell reprogramming to boost cardiomyocyte replacement have fostered the prospect of achieving functional heart repair/regeneration. AREAS COVERED Allogeneic CSC therapy through its paracrine stimulation of the endogenous resident reparative/regenerative process produces functional meaningful myocardial regeneration in pre-clinical porcine myocardial infarction models and is currently tested in the first-in-man human trial. The in vivo test of somatic reprogramming and cardioregenerative non-coding RNAs revived the interest in gene therapy for myocardial regeneration. The latter, together with the advent of genome editing, has prompted most recent efforts to produce genetically-modified allogeneic CSCs that secrete cardioregenerative factors to optimize effective myocardial repair. EXPERT OPINION The current war against heart failure epidemics in western countries seeks to find effective treatments to set back the failing hearts prolonging human lifespan. Off-the-shelf allogeneic-genetically-modified CSCs producing regenerative agents are a novel and evolving therapy set to be affordable, safe, effective and available at all times for myocardial regeneration to either prevent or treat heart failure.
Collapse
Affiliation(s)
- Pina Marotta
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Eleonora Cianflone
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Iolanda Aquila
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Carla Vicinanza
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Mariangela Scalise
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Fabiola Marino
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Teresa Mancuso
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Michele Torella
- b Department of Cardiothoracic Sciences , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Ciro Indolfi
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Daniele Torella
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| |
Collapse
|
55
|
Abstract
Fibrosis is part of a tissue repair response to injury, defined as increased deposition of extracellular matrix. In some instances, fibrosis is beneficial; however, in the majority of diseases fibrosis is detrimental. Virtually all chronic progressive diseases are associated with fibrosis, representing a huge number of patients worldwide. Fibrosis occurs in all organs and tissues, becomes irreversible with time and further drives loss of tissue function. Various cells types initiate and perpetuate pathological fibrosis by paracrine activation of the principal cellular executors of fibrosis, i.e. stromal mesenchymal cells like fibroblasts, pericytes and myofibroblasts. Multiple pathways are involved in fibrosis, platelet-derived growth factor (PDGF)-signaling being one of the central mediators. Stromal mesenchymal cells express both PDGF receptors (PDGFR) α and β, activation of which drives proliferation, migration and production of extracellular matrix, i.e. the principal processes of fibrosis. Here, we review the role of PDGF signaling in organ fibrosis, with particular focus on the more recently described ligands PDGF-C and -D. We discuss the potential challenges, opportunities and open questions in using PDGF as a potential target for anti-fibrotic therapies.
Collapse
Affiliation(s)
| | - Jürgen Floege
- Division of Nephrology, RWTH University of Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH University of Aachen, Germany; Division of Nephrology, RWTH University of Aachen, Germany.
| |
Collapse
|
56
|
PDGFRα Regulated by miR-34a and FoxO1 Promotes Adipogenesis in Porcine Intramuscular Preadipocytes through Erk Signaling Pathway. Int J Mol Sci 2017; 18:ijms18112424. [PMID: 29140299 PMCID: PMC5713392 DOI: 10.3390/ijms18112424] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 11/04/2017] [Accepted: 11/11/2017] [Indexed: 12/27/2022] Open
Abstract
Suitable intramuscular fat (IMF) content improves porcine meat quality. The vital genes regulating IMF deposition are necessary for the selection and breeding of an IMF trait. However, the effect and mechanism of PDGFRα on IMF deposition are still unclear. Here, PDGFRα is moderately expressed in porcine longissimus dorsi muscle (LD), whereas it highly expressed in white adipose tissue (WAT). Moreover, PDGFRα-positive cells were located in the gaps of LD fibers which there were IMF adipocytes. Compared with 180-day-old and lean-type pigs, the levels of PDGFRα were much higher in one-day-old and fat-type pigs. Meanwhile the levels of PDGFRα gradually decreased during IMF preadipocyte differentiation. Furthermore, PDGFRα promoted adipogenic differentiation through activating Erk signaling pathway. Based on PDGFRα upstream regulation analysis, we found that the knockdown of FoxO1 repressed lipogenesis by downregulating PDGFRα, and miR-34a inhibited adipogenesis through targeting PDGFRα. Collectively, PDGFRα is a positive regulator of IMF deposition. Therefore, we suggest that PDGFRα is a possible target to improve meat quality.
Collapse
|
57
|
Platelet-derived growth factor-C and -D in the cardiovascular system and diseases. Mol Aspects Med 2017; 62:12-21. [PMID: 28965749 DOI: 10.1016/j.mam.2017.09.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 09/26/2017] [Indexed: 12/31/2022]
Abstract
The cardiovascular system is among the first organs formed during development and is pivotal for the formation and function of the rest of the organs and tissues. Therefore, the function and homeostasis of the cardiovascular system are finely regulated by many important molecules. Extensive studies have shown that platelet-derived growth factors (PDGFs) and their receptors are critical regulators of the cardiovascular system. Even though PDGF-C and PDGF-D are relatively new members of the PDGF family, their critical roles in the cardiovascular system as angiogenic and survival factors have been amply demonstrated. Understanding the functions of PDGF-C and PDGF-D and the signaling pathways involved may provide novel insights into both basic biomedical research and new therapeutic possibilities for the treatment of cardiovascular diseases.
Collapse
|
58
|
Savi M, Frati C, Cavalli S, Graiani G, Galati S, Buschini A, Madeddu D, Falco A, Prezioso L, Mazzaschi G, Galaverna F, Lagrasta CAM, Corradini E, De Angelis A, Cappetta D, Berrino L, Aversa F, Quaini F, Urbanek K. Imatinib mesylate-induced cardiomyopathy involves resident cardiac progenitors. Pharmacol Res 2017; 127:15-25. [PMID: 28964914 DOI: 10.1016/j.phrs.2017.09.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/05/2017] [Accepted: 09/26/2017] [Indexed: 02/06/2023]
Abstract
Cardiovascular complications are included among the systemic effects of tyrosine kinase inhibitor (TKI)-based therapeutic strategies. To test the hypothesis that inhibition of Kit tyrosine kinase that promotes cardiac progenitor cell (CPC) survival and function may be one of the triggering mechanisms of imatinib mesylate (IM)-related cardiovascular effects, the anatomical, structural and ultrastructural changes in the heart of IM-treated rats were evaluated. Cardiac anatomy in IM-exposed rats showed a dose-dependent, restrictive type of remodeling and depressed hemodynamic performance in the absence of remarkable myocardial fibrosis. The effects of IM on rat and human CPCs were also assessed. IM induced rat CPC depletion, reduced growth and increased cell death. Similar effects were observed in CPCs isolated from human hearts. These results extend the notion that cardiovascular side effects are driven by multiple actions of IM. The identification of cellular mechanisms responsible for cardiovascular complications due to TKIs will enable future strategies aimed at preserving concomitantly cardiac integrity and anti-tumor activity of advanced cancer treatment.
Collapse
Affiliation(s)
- Monia Savi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Stefano Cavalli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gallia Graiani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Serena Galati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Annamaria Buschini
- Department of Genetics, Biology of Microorganisms, Anthropology, Evolution, University of Parma, Parma, Italy
| | - Denise Madeddu
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Angela Falco
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Lucia Prezioso
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giulia Mazzaschi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | | | - Emilia Corradini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Franco Aversa
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
59
|
Guo Y, Gupte M, Umbarkar P, Singh AP, Sui JY, Force T, Lal H. Entanglement of GSK-3β, β-catenin and TGF-β1 signaling network to regulate myocardial fibrosis. J Mol Cell Cardiol 2017; 110:109-120. [PMID: 28756206 DOI: 10.1016/j.yjmcc.2017.07.011] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 07/20/2017] [Accepted: 07/24/2017] [Indexed: 12/31/2022]
Abstract
Nearly every form of the heart disease is associated with myocardial fibrosis, which is characterized by the accumulation of activated cardiac fibroblasts (CFs) and excess deposition of extracellular matrix (ECM). Although, CFs are the primary mediators of myocardial fibrosis in a diseased heart, in the traditional view, activated CFs (myofibroblasts) and resulting fibrosis were simply considered the secondary consequence of the disease, not the cause. Recent studies from our lab and others have challenged this concept by demonstrating that fibroblast activation and fibrosis are not simply the secondary consequence of a diseased heart, but are crucial for mediating various myocardial disease processes. In regards to the mechanism, the vast majority of literature is focused on the direct role of canonical SMAD-2/3-mediated TGF-β signaling to govern the fibrogenic process. Herein, we will discuss the emerging role of the GSK-3β, β-catenin and TGF-β1-SMAD-3 signaling network as a critical regulator of myocardial fibrosis in the diseased heart. The underlying molecular interactions and cross-talk among signaling pathways will be discussed. We will primarily focus on recent in vivo reports demonstrating that CF-specific genetic manipulation can lead to aberrant myocardial fibrosis and sturdy cardiac phenotype. This will allow for a better understanding of the driving role of CFs in the myocardial disease process. We will also review the specificity and limitations of the currently available genetic tools used to study myocardial fibrosis and its associated mechanisms. A better understanding of the GSK-3β, β-catenin and SMAD-3 signaling network may provide a novel therapeutic target for the management of myocardial fibrosis in the diseased heart.
Collapse
Affiliation(s)
- Yuanjun Guo
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB, Suite#348, Nashville, TN 37232, United States
| | - Manisha Gupte
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB, Suite#348, Nashville, TN 37232, United States
| | - Prachi Umbarkar
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB, Suite#348, Nashville, TN 37232, United States
| | - Anand Prakash Singh
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB, Suite#348, Nashville, TN 37232, United States
| | - Jennifer Y Sui
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB, Suite#348, Nashville, TN 37232, United States
| | - Thomas Force
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB, Suite#348, Nashville, TN 37232, United States
| | - Hind Lal
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB, Suite#348, Nashville, TN 37232, United States.
| |
Collapse
|
60
|
The epicardium as a source of multipotent adult cardiac progenitor cells: Their origin, role and fate. Pharmacol Res 2017; 127:129-140. [PMID: 28751220 DOI: 10.1016/j.phrs.2017.07.020] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/12/2017] [Accepted: 07/21/2017] [Indexed: 12/23/2022]
Abstract
Since the regenerative capacity of the adult mammalian heart is limited, cardiac injury leads to the formation of scar tissue and thereby increases the risk of developing compensatory heart failure. Stem cell therapy is a promising therapeutic approach but is facing problems with engraftment and clinical feasibility. Targeting an endogenous stem cell population could circumvent these limitations. The epicardium, a membranous layer covering the outside of the myocardium, is an accessible cell population which plays a key role in the developing heart. Epicardial cells undergo epithelial to mesenchymal transition (EMT), thus providing epicardial derived cells (EPDCs) that migrate into the myocardium and cooperate in myocardial vascularisation and compaction. In the adult heart, injury activates the epicardium, and an embryonic-like response is observed which includes EMT and differentiation of the EPDCs into cardiac cell types. Furthermore, paracrine communication between the epicardium and myocardium improves the regenerative response. The significant role of the epicardium has been shown in both the developing and the regenerating heart. Interestingly, the epicardial contribution to cardiac repair can be improved in several ways. In this review, an overview of the epicardial origin and fate will be given and potential therapeutic approaches will be discussed.
Collapse
|
61
|
Garreta E, Prado P, Izpisua Belmonte JC, Montserrat N. Non-coding microRNAs for cardiac regeneration: Exploring novel alternatives to induce heart healing. Noncoding RNA Res 2017; 2:93-99. [PMID: 30159426 PMCID: PMC6096419 DOI: 10.1016/j.ncrna.2017.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 01/06/2023] Open
Abstract
In recent years, different studies have revealed that adult mammalian cardiomyocytes have the capacity to self-renew under homeostatic conditions and after myocardial injury. Interestingly, data from animal models capable of regeneration, such as the adult zebrafish and neonatal mice, have identified different non-coding RNAs (ncRNAs) as functional RNA molecules driving cardiac regeneration and repair. In this review, we summarize the current knowledge of the roles that a specific subset of ncRNAs, namely microRNAs (miRNA), plays in these animal models. We also emphasize the importance of characterizing and manipulating miRNAs as a novel approach to awaken the dormant regenerative potential of the adult mammalian heart by the administration of miRNA mimics or inhibitors. Overall, the use of these strategies alone or in combination with current cardiac therapies may represent new avenues to pursue for cardiac regeneration.
Collapse
Affiliation(s)
- Elena Garreta
- Pluripotent Stem Cells and Activation of Endogenous Tissue Programs for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - Patricia Prado
- Pluripotent Stem Cells and Activation of Endogenous Tissue Programs for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | | | - Nuria Montserrat
- Pluripotent Stem Cells and Activation of Endogenous Tissue Programs for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| |
Collapse
|
62
|
Cimini M, Cannatá A, Pasquinelli G, Rota M, Goichberg P. Phenotypically heterogeneous podoplanin-expressing cell populations are associated with the lymphatic vessel growth and fibrogenic responses in the acutely and chronically infarcted myocardium. PLoS One 2017; 12:e0173927. [PMID: 28333941 PMCID: PMC5363820 DOI: 10.1371/journal.pone.0173927] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 02/28/2017] [Indexed: 01/08/2023] Open
Abstract
Cardiac lymphatic vasculature undergoes substantial expansion in response to myocardial infarction (MI). However, there is limited information on the cellular mechanisms mediating post-MI lymphangiogenesis and accompanying fibrosis in the infarcted adult heart. Using a mouse model of permanent coronary artery ligation, we examined spatiotemporal changes in the expression of lymphendothelial and mesenchymal markers in the acutely and chronically infarcted myocardium. We found that at the time of wound granulation, a three-fold increase in the frequency of podoplanin-labeled cells occurred in the infarcted hearts compared to non-operated and sham-operated counterparts. Podoplanin immunoreactivity detected LYVE-1-positive lymphatic vessels, as well as masses of LYVE-1-negative cells dispersed between myocytes, predominantly in the vicinity of the infarcted region. Podoplanin-carrying populations displayed a mesenchymal progenitor marker PDGFRα, and intermittently expressed Prox-1, a master regulator of the lymphatic endothelial fate. At the stages of scar formation and maturation, concomitantly with the enlargement of lymphatic network in the injured myocardium, the podoplanin-rich LYVE-1-negative multicellular assemblies were apparent in the fibrotic area, aligned with extracellular matrix deposits, or located in immediate proximity to activated blood vessels with high VEGFR-2 content. Of note, these podoplanin-containing cells acquired the expression of PDGFRβ or a hematoendothelial epitope CD34. Although Prox-1 labeling was abundant in the area affected by MI, the podoplanin-presenting cells were not consistently Prox-1-positive. The concordance of podoplanin with VEGFR-3 similarly varied. Thus, our data reveal previously unknown phenotypic and structural heterogeneity within the podoplanin-positive cell compartment in the infarcted heart, and suggest an alternate ability of podoplanin-presenting cardiac cells to generate lymphatic endothelium and pro-fibrotic cells, contributing to scar development.
Collapse
Affiliation(s)
- Maria Cimini
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Antonio Cannatá
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gianandrea Pasquinelli
- Unit of Surgical Pathology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Marcello Rota
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Polina Goichberg
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
63
|
Doppler SA, Carvalho C, Lahm H, Deutsch MA, Dreßen M, Puluca N, Lange R, Krane M. Cardiac fibroblasts: more than mechanical support. J Thorac Dis 2017; 9:S36-S51. [PMID: 28446967 DOI: 10.21037/jtd.2017.03.122] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Fibroblasts are cells with a structural function, synthesizing components of the extracellular matrix. They are accordingly associated with various forms of connective tissue. During cardiac development fibroblasts originate from different sources. Most derive from the epicardium, some derive from the endocardium, and a small population derives from the neural crest. Cardiac fibroblasts have important functions during development, homeostasis, and disease. However, since fibroblasts are a very heterogeneous cell population no truly specific markers exist. Therefore, studying them in detail is difficult. Nevertheless, several lineage tracing models have been widely used. In this review, we describe the developmental origins of cardiac fibroblasts, comment on fibroblast markers and related lineage tracing approaches, and discuss the cardiac cell composition, which has recently been revised, especially in terms of non-myocyte cells.
Collapse
Affiliation(s)
- Stefanie A Doppler
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany
| | - Catarina Carvalho
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany
| | - Harald Lahm
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany
| | - Marcus-André Deutsch
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Martina Dreßen
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany
| | - Nazan Puluca
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany
| | - Rüdiger Lange
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Markus Krane
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
64
|
Brunner R, Lai HL, Deliu Z, Melman E, Geenen DL, Wang QT. Asxl2 -/- Mice Exhibit De Novo Cardiomyocyte Production during Adulthood. J Dev Biol 2016; 4:jdb4040032. [PMID: 29615595 PMCID: PMC5831801 DOI: 10.3390/jdb4040032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/26/2016] [Accepted: 10/27/2016] [Indexed: 12/20/2022] Open
Abstract
Heart attacks affect more than seven million people worldwide each year. A heart attack, or myocardial infarction, may result in the death of a billion cardiomyocytes within hours. The adult mammalian heart does not have an effective mechanism to replace lost cardiomyocytes. Instead, lost muscle is replaced with scar tissue, which decreases blood pumping ability and leads to heart failure over time. Here, we report that the loss of the chromatin factor ASXL2 results in spontaneous proliferation and cardiogenic differentiation of a subset of interstitial non-cardiomyocytes. The adult Asxl2-/- heart displays spontaneous overgrowth without cardiomyocyte hypertrophy. Thymidine analog labeling and Ki67 staining of 12-week-old hearts revealed 3- and 5-fold increases of proliferation rate for vimentin⁺ non-cardiomyocytes in Asxl2-/- over age- and sex-matched wildtype controls, respectively. Approximately 10% of proliferating non-cardiomyocytes in the Asxl2-/- heart express the cardiogenic marker NKX2-5, a frequency that is ~7-fold higher than that observed in the wildtype. EdU lineage tracing experiments showed that ~6% of pulsed-labeled non-cardiomyocytes in Asxl2-/- hearts differentiate into mature cardiomyocytes after a four-week chase, a phenomenon not observed for similarly pulse-chased wildtype controls. Taken together, these data indicate de novo cardiomyocyte production in the Asxl2-/- heart due to activation of a population of proliferative cardiogenic non-cardiomyocytes. Our study suggests the existence of an epigenetic barrier to cardiogenicity in the adult heart and raises the intriguing possibility of unlocking regenerative potential via transient modulation of epigenetic activity.
Collapse
Affiliation(s)
- Rachel Brunner
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
| | - Hsiao-Lei Lai
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
- PTM Biolabs Inc., Chicago, IL 60612, USA.
| | - Zane Deliu
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
| | - Elan Melman
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
- The School of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Champaign, IL 61801, USA.
| | - David L Geenen
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA.
- Physician Assistant Studies, Grand Valley State University, Grand Rapids, MI 49503, USA.
| | - Q Tian Wang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
- Congressionally Directed Medical Research Programs, Frederick, MD 21702, USA.
| |
Collapse
|
65
|
PDGF-A and PDGF-B induces cardiac fibrosis in transgenic mice. Exp Cell Res 2016; 349:282-290. [PMID: 27816607 DOI: 10.1016/j.yexcr.2016.10.022] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 10/26/2016] [Accepted: 10/27/2016] [Indexed: 02/06/2023]
Abstract
Platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) contribute to normal heart development. Deficient or abnormal expression of Pdgf and Pdgfr genes have a negative impact on cardiac development and function. The cellular effects of PDGFs in the hearts of Pdgf/Pdgfr mutants and the pathogenesis of the resulting abnormalities are poorly understood, but different PDGF isoforms induce varying effects. Here, we generated three new transgenic mouse types which complete a set of studies, where all different PDGF ligands have been expressed under the same heart specific alpha-myosin heavy chain promoter. Transgenic expression of the natural isoforms of Pdgfa and Pdgfb resulted in isoform specific fibrotic reactions and cardiac hypertrophy. Pdgfa overexpression resulted in a severe fibrotic reaction with up to 8-fold increase in cardiac size, leading to lethal cardiac failure within a few weeks after birth. In contrast, Pdgfb overexpression led to focal fibrosis and moderate cardiac hypertrophy. As PDGF-A and PDGF-B have different affinity for the two PDGF receptors, we analyzed the expression of the receptors and the histology of the fibrotic hearts. Our data suggest that the stronger fibrotic effect generated by Pdgfa overexpression was mediated by Pdgfrα in cardiac interstitial mesenchymal cells, i.e. the likely source of extracellular matrix depostion and fibrotic reaction. The apparent sensitivity of the heart to ectopic PDGFRα agonists supports a role for endogenous PDGFRα agonists in the pathogenesis of cardiac fibrosis.
Collapse
|
66
|
O’Rourke M, Cullen CL, Auderset L, Pitman KA, Achatz D, Gasperini R, Young KM. Evaluating Tissue-Specific Recombination in a Pdgfrα-CreERT2 Transgenic Mouse Line. PLoS One 2016; 11:e0162858. [PMID: 27626928 PMCID: PMC5023134 DOI: 10.1371/journal.pone.0162858] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/30/2016] [Indexed: 11/18/2022] Open
Abstract
In the central nervous system (CNS) platelet derived growth factor receptor alpha (PDGFRα) is expressed exclusively by oligodendrocyte progenitor cells (OPCs), making the Pdgfrα promoter an ideal tool for directing transgene expression in this cell type. Two Pdgfrα-CreERT2 mouse lines have been generated for this purpose which, when crossed with cre-sensitive reporter mice, allow the temporally restricted labelling of OPCs for lineage-tracing studies. These mice have also been used to achieve the deletion of CNS-specific genes from OPCs. However the ability of Pdgfrα-CreERT2 mice to induce cre-mediated recombination in PDGFRα+ cell populations located outside of the CNS has not been examined. Herein we quantify the proportion of PDGFRα+ cells that become YFP-labelled following Tamoxifen administration to adult Pdgfrα-CreERT2::Rosa26-YFP transgenic mice. We report that the vast majority (>90%) of PDGFRα+ OPCs in the CNS, and a significant proportion of PDGFRα+ stromal cells within the bone marrow (~38%) undergo recombination and become YFP-labelled. However, only a small proportion of the PDGFRα+ cell populations found in the sciatic nerve, adrenal gland, pituitary gland, heart, gastrocnemius muscle, kidney, lung, liver or intestine become YFP-labelled. These data suggest that Pdgfrα-CreERT2 transgenic mice can be used to achieve robust recombination in OPCs, while having a minimal effect on most PDGFRα+ cell populations outside of the CNS.
Collapse
Affiliation(s)
- Megan O’Rourke
- Menzies Institute for Medical Research, University of Tasmania, Liverpool Street, Hobart, Tasmania 7000, Australia
| | - Carlie L. Cullen
- Menzies Institute for Medical Research, University of Tasmania, Liverpool Street, Hobart, Tasmania 7000, Australia
| | - Loic Auderset
- Menzies Institute for Medical Research, University of Tasmania, Liverpool Street, Hobart, Tasmania 7000, Australia
| | - Kimberley A. Pitman
- Menzies Institute for Medical Research, University of Tasmania, Liverpool Street, Hobart, Tasmania 7000, Australia
| | - Daniela Achatz
- Menzies Institute for Medical Research, University of Tasmania, Liverpool Street, Hobart, Tasmania 7000, Australia
| | - Robert Gasperini
- Menzies Institute for Medical Research, University of Tasmania, Liverpool Street, Hobart, Tasmania 7000, Australia
- School of Medicine, University of Tasmania, Liverpool Street, Hobart, Tasmania 7000, Australia
| | - Kaylene M. Young
- Menzies Institute for Medical Research, University of Tasmania, Liverpool Street, Hobart, Tasmania 7000, Australia
- * E-mail:
| |
Collapse
|
67
|
Nelson DO, Lalit PA, Biermann M, Markandeya YS, Capes DL, Addesso L, Patel G, Han T, John MC, Powers PA, Downs KM, Kamp TJ, Lyons GE. Irx4 Marks a Multipotent, Ventricular-Specific Progenitor Cell. Stem Cells 2016; 34:2875-2888. [PMID: 27570947 DOI: 10.1002/stem.2486] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/18/2016] [Indexed: 12/11/2022]
Abstract
While much progress has been made in the resolution of the cellular hierarchy underlying cardiogenesis, our understanding of chamber-specific myocardium differentiation remains incomplete. To better understand ventricular myocardium differentiation, we targeted the ventricle-specific gene, Irx4, in mouse embryonic stem cells to generate a reporter cell line. Using an antibiotic-selection approach, we purified Irx4+ cells in vitro from differentiating embryoid bodies. The isolated Irx4+ cells proved to be highly proliferative and presented Cxcr4, Pdgfr-alpha, Flk1, and Flt1 on the cell surface. Single Irx4+ ventricular progenitor cells (VPCs) exhibited cardiovascular potency, generating endothelial cells, smooth muscle cells, and ventricular myocytes in vitro. The ventricular specificity of the Irx4+ population was further demonstrated in vivo as VPCs injected into the cardiac crescent subsequently produced Mlc2v+ myocytes that exclusively contributed to the nascent ventricle at E9.5. These findings support the existence of a newly identified ventricular myocardial progenitor. This is the first report of a multipotent cardiac progenitor that contributes progeny specific to the ventricular myocardium. Stem Cells 2016;34:2875-2888.
Collapse
Affiliation(s)
- Daryl O Nelson
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Pratik A Lalit
- Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mitch Biermann
- Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yogananda S Markandeya
- Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Deborah L Capes
- Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Luke Addesso
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Gina Patel
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Tianxiao Han
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Manorama C John
- University of Wisconsin Biotechnology Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Patricia A Powers
- University of Wisconsin Biotechnology Center, University of Wisconsin, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Karen M Downs
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Timothy J Kamp
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Gary E Lyons
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
68
|
Santini MP, Forte E, Harvey RP, Kovacic JC. Developmental origin and lineage plasticity of endogenous cardiac stem cells. Development 2016; 143:1242-58. [PMID: 27095490 DOI: 10.1242/dev.111591] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Over the past two decades, several populations of cardiac stem cells have been described in the adult mammalian heart. For the most part, however, their lineage origins and in vivo functions remain largely unexplored. This Review summarizes what is known about different populations of embryonic and adult cardiac stem cells, including KIT(+), PDGFRα(+), ISL1(+)and SCA1(+)cells, side population cells, cardiospheres and epicardial cells. We discuss their developmental origins and defining characteristics, and consider their possible contribution to heart organogenesis and regeneration. We also summarize the origin and plasticity of cardiac fibroblasts and circulating endothelial progenitor cells, and consider what role these cells have in contributing to cardiac repair.
Collapse
Affiliation(s)
- Maria Paola Santini
- Cardiovascular Research Centre, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Elvira Forte
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst 2010, Australia St Vincent's Clinical School, University of New South Wales, Kensington 2052, Australia Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst 2010, Australia St Vincent's Clinical School, University of New South Wales, Kensington 2052, Australia Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington 2052, Australia
| | - Jason C Kovacic
- Cardiovascular Research Centre, Icahn School of Medicine at Mount Sinai, New York City, NY, USA Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
69
|
Faiella W, Atoui R. Therapeutic use of stem cells for cardiovascular disease. Clin Transl Med 2016; 5:34. [PMID: 27539581 PMCID: PMC4990528 DOI: 10.1186/s40169-016-0116-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 08/10/2016] [Indexed: 12/21/2022] Open
Abstract
Stem cell treatments are a desirable therapeutic option to regenerate myocardium and improve cardiac function after myocardial infarction. Several different types of cells have been explored, each with their own benefits and limitations. Induced pluripotent stem cells possess an embryonic-like state and therefore have a high proliferative capacity, but they also pose a risk of teratoma formation. Mesenchymal stem cells have been investigated from both bone marrow and adipose tissue. Their immunomodulatory characteristics may permit the use of allogeneic cells as universal donor cells in the future. Lastly, studies have consistently shown that cardiac stem cells are better able to express markers of cardiogenesis compared to other cell types, as well improve cardiac function. The ideal source of stem cells depends on multiple factors such as the ease of extraction/isolation, effectiveness of engraftment, ability to differentiate into cardiac lineages and effect on cardiac function. Although multiple studies highlight the benefits and limitations of each cell type and reinforce the successful potential use of these cells to regenerate damaged myocardium, more studies are needed to directly compare cells from various sources. It is interesting to note that research using stem cell therapies is also expanding to treat other cardiovascular diseases including non-ischemic cardiomyopathies.
Collapse
Affiliation(s)
- Whitney Faiella
- Division of Cardiac Surgery, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada
| | - Rony Atoui
- Division of Cardiac Surgery, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada.
| |
Collapse
|
70
|
Kurose H, Mangmool S. Myofibroblasts and inflammatory cells as players of cardiac fibrosis. Arch Pharm Res 2016; 39:1100-13. [PMID: 27515051 DOI: 10.1007/s12272-016-0809-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/01/2016] [Indexed: 01/18/2023]
Abstract
On myocardial infarction, many cells are injured or died owing to arterial occlusion. Intracellular molecules released from injured or dead cells initiate inflammatory responses that play important roles in cardiac remodeling including fibrosis. Fibrosis is an excess accumulation of extracellular collagen. Currently, drugs used to treat cardiac fibrosis are not commercially available. Myofibroblasts are responsible for the production and secretion of collagen. Infiltrating inflammatory cells interact with fibroblasts or other cells and promote myofibroblast formation. Inflammatory cells also modulate the activities of myofibroblasts. Regulation of collagen production is critical for modulating the progression of fibrosis. Hence, the manipulation of activities of inflammatory cells and myofibroblasts will provide promising therapeutic targets for treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Hitoshi Kurose
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
71
|
Le T, Chong J. Cardiac progenitor cells for heart repair. Cell Death Discov 2016; 2:16052. [PMID: 27551540 PMCID: PMC4979410 DOI: 10.1038/cddiscovery.2016.52] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 05/25/2016] [Indexed: 02/06/2023] Open
Abstract
Stem cell therapy is being investigated as an innovative and promising strategy to restore cardiac function in patients with heart failure. Several stem cell populations, including adult (multipotent) stem cells from developed organs and tissues, have been tested for cardiac repair with encouraging clinical and pre-clinical results. The heart has been traditionally considered a post-mitotic organ, however, this view has recently changed with the identification of stem/progenitor cells residing within the adult heart. Given their cardiac developmental origins, these endogenous cardiac progenitor cells (CPCs) may represent better candidates for cardiac cell therapy compared with stem cells from other organs such as the bone marrow and adipose tissue. This brief review will outline current research into CPC populations and their cardiac repair/regenerative potential.
Collapse
Affiliation(s)
- Tyl Le
- Centre of Heart Research, Westmead Institute for Medical Research, The University of Sydney, Hawkesbury Road, Westmead, Sydney, New South Wale 2145, Australia; Department of Cardiology, Westmead Hospital, Hawkesbury Road, Westmead, Sydney, New South Wale 2145, Australia
| | - Jjh Chong
- Centre of Heart Research, Westmead Institute for Medical Research, The University of Sydney, Hawkesbury Road, Westmead, Sydney, New South Wale 2145, Australia; Department of Cardiology, Westmead Hospital, Hawkesbury Road, Westmead, Sydney, New South Wale 2145, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wale 2006, Australia
| |
Collapse
|
72
|
Furtado MB, Costa MW, Rosenthal NA. The cardiac fibroblast: Origin, identity and role in homeostasis and disease. Differentiation 2016; 92:93-101. [PMID: 27421610 DOI: 10.1016/j.diff.2016.06.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 06/24/2016] [Indexed: 12/22/2022]
Abstract
The mammalian heart is responsible for supplying blood to two separate circulation circuits in a parallel manner. This design provides efficient oxygenation and nutrients to the whole body through the left-sided pump, while the right-sided pump delivers blood to the pulmonary circulation for re-oxygenation. In order to achieve this demanding job, the mammalian heart evolved into a highly specialised organ comprised of working contractile cells or cardiomyocytes, a directional and insulated conduction system, capable of independently generating and conducting electric impulses that synchronises chamber contraction, valves that allow the generation of high pressure and directional blood flow into the circulation, coronary circulation, that supplies oxygenated blood for the heart muscle high metabolically active pumping role and inlet/outlet routes, as the venae cavae and pulmonary veins, aorta and pulmonary trunk. This organization highlights the complexity and compartmentalization of the heart. This review will focus on the cardiac fibroblast, a cell type until recently ignored, but that profoundly influences heart function in its various compartments. We will discuss current advances on definitions, molecular markers and function of cardiac fibroblasts in heart homeostasis and disease.
Collapse
Affiliation(s)
- Milena B Furtado
- The Jackson Laboratory, Bar Harbor, ME, USA; Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.
| | - Mauro W Costa
- The Jackson Laboratory, Bar Harbor, ME, USA; Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Nadia A Rosenthal
- The Jackson Laboratory, Bar Harbor, ME, USA; Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia; National Heart and Lung Institute, Imperial College London, UK
| |
Collapse
|
73
|
Furtado MB, Nim HT, Boyd SE, Rosenthal NA. View from the heart: cardiac fibroblasts in development, scarring and regeneration. Development 2016; 143:387-97. [PMID: 26839342 DOI: 10.1242/dev.120576] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the adult, tissue repair after injury is generally compromised by fibrosis, which maintains tissue integrity with scar formation but does not restore normal architecture and function. The process of regeneration is necessary to replace the scar and rebuild normal functioning tissue. Here, we address this problem in the context of heart disease, and discuss the origins and characteristics of cardiac fibroblasts, as well as the crucial role that they play in cardiac development and disease. We discuss the dual nature of cardiac fibroblasts, which can lead to scarring, pathological remodelling and functional deficit, but can also promote heart function in some contexts. Finally, we review current and proposed approaches whereby regeneration could be fostered by interventions that limit scar formation.
Collapse
Affiliation(s)
- Milena B Furtado
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Hieu T Nim
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia Systems Biology Institute (SBI) Australia, Monash University, Clayton, Victoria 3800, Australia
| | - Sarah E Boyd
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia Systems Biology Institute (SBI) Australia, Monash University, Clayton, Victoria 3800, Australia
| | - Nadia A Rosenthal
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia Systems Biology Institute (SBI) Australia, Monash University, Clayton, Victoria 3800, Australia National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK The Jackson Laboratory, Bar Harbor, ME 04609, USA
| |
Collapse
|
74
|
Månsson-Broberg A, Rodin S, Bulatovic I, Ibarra C, Löfling M, Genead R, Wärdell E, Felldin U, Granath C, Alici E, Le Blanc K, Smith CIE, Salašová A, Westgren M, Sundström E, Uhlén P, Arenas E, Sylvén C, Tryggvason K, Corbascio M, Simonson OE, Österholm C, Grinnemo KH. Wnt/β-Catenin Stimulation and Laminins Support Cardiovascular Cell Progenitor Expansion from Human Fetal Cardiac Mesenchymal Stromal Cells. Stem Cell Reports 2016; 6:607-617. [PMID: 27052314 PMCID: PMC4834052 DOI: 10.1016/j.stemcr.2016.02.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 02/19/2016] [Accepted: 02/22/2016] [Indexed: 11/29/2022] Open
Abstract
The intrinsic regenerative capacity of human fetal cardiac mesenchymal stromal cells (MSCs) has not been fully characterized. Here we demonstrate that we can expand cells with characteristics of cardiovascular progenitor cells from the MSC population of human fetal hearts. Cells cultured on cardiac muscle laminin (LN)-based substrata in combination with stimulation of the canonical Wnt/β-catenin pathway showed increased gene expression of ISL1, OCT4, KDR, and NKX2.5. The majority of cells stained positive for PDGFR-α, ISL1, and NKX2.5, and subpopulations also expressed the progenitor markers TBX18, KDR, c-KIT, and SSEA-1. Upon culture of the cardiac MSCs in differentiation media and on relevant LNs, portions of the cells differentiated into spontaneously beating cardiomyocytes, and endothelial and smooth muscle-like cells. Our protocol for large-scale culture of human fetal cardiac MSCs enables future exploration of the regenerative functions of these cells in the context of myocardial injury in vitro and in vivo. Cells with progenitor properties can be expanded from human fetal cardiac MSCs Specific LNs support expansion and differentiation of cardiac MSCs The fetal cardiac MSCs express ISL1, PDGFR-α, and NKX2.5 Subpopulations express the progenitor markers KDR, SSEA-1, c-KIT, and TBX18
Collapse
Affiliation(s)
- Agneta Månsson-Broberg
- Division of Cardiology, Department of Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Sergey Rodin
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ivana Bulatovic
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Cristián Ibarra
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Cardiovascular & Metabolic Diseases, Innovative Medicines and Early Development, AstraZeneca R&D, 43150 Mölndal, Sweden
| | - Marie Löfling
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Rami Genead
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Eva Wärdell
- Division of Cardiology, Department of Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ulrika Felldin
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Carl Granath
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Evren Alici
- Division of Hematology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden; Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Katarina Le Blanc
- Division of Hematology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden; Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - C I Edvard Smith
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Alena Salašová
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Magnus Westgren
- CLINTEC, Division of Obstetrics and Gynecology, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Erik Sundström
- Division of Neurodegeneration, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Per Uhlén
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ernest Arenas
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Christer Sylvén
- Division of Cardiology, Department of Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Karl Tryggvason
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Duke-NUS Graduate Medical School, Durham, NC 27710, USA
| | - Matthias Corbascio
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Oscar E Simonson
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Cecilia Österholm
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden; Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Karl-Henrik Grinnemo
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden; Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL 33314, USA.
| |
Collapse
|
75
|
Abstract
Myocardial fibrosis is a significant global health problem associated with nearly all forms of heart disease. Cardiac fibroblasts comprise an essential cell type in the heart that is responsible for the homeostasis of the extracellular matrix; however, upon injury, these cells transform to a myofibroblast phenotype and contribute to cardiac fibrosis. This remodeling involves pathological changes that include chamber dilation, cardiomyocyte hypertrophy and apoptosis, and ultimately leads to the progression to heart failure. Despite the critical importance of fibrosis in cardiovascular disease, our limited understanding of the cardiac fibroblast impedes the development of potential therapies that effectively target this cell type and its pathological contribution to disease progression. This review summarizes current knowledge regarding the origins and roles of fibroblasts, mediators and signaling pathways known to influence fibroblast function after myocardial injury, as well as novel therapeutic strategies under investigation to attenuate cardiac fibrosis.
Collapse
Affiliation(s)
- Joshua G Travers
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Fadia A Kamal
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Jeffrey Robbins
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Katherine E Yutzey
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Burns C Blaxall
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH.
| |
Collapse
|
76
|
Harnessing the secretome of cardiac stem cells as therapy for ischemic heart disease. Biochem Pharmacol 2016; 113:1-11. [PMID: 26903387 DOI: 10.1016/j.bcp.2016.02.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/18/2016] [Indexed: 12/22/2022]
Abstract
Adult stem cells continue to promise opportunities to repair damaged cardiac tissue. However, precisely how adult stem cells accomplish cardiac repair, especially after ischemic damage, remains controversial. It has been postulated that the clinical benefit of adult stem cells for cardiovascular disease results from the release of cytokines and growth factors by the transplanted cells. Studies in animal models of myocardial infarction have reported that such paracrine factors released from transplanted adult stem cells contribute to improved cardiac function by several processes. These include promoting neovascularization of damaged tissue, reducing inflammation, reducing fibrosis and scar formation, as well as protecting cardiomyocytes from apoptosis. In addition, these factors might also stimulate endogenous repair by activating cardiac stem cells. Interestingly, stem cells discovered to be resident in the heart appear to be functionally superior to extra-cardiac adult stem cells when transplanted for cardiac repair and regeneration. In this review, we discuss the therapeutic potential of cardiac stem cells and how the proteins secreted from these cells might be harnessed to promote repair and regeneration of damaged cardiac tissue. We also highlight how recent controversies about the efficacy of adult stem cells in clinical trials of ischemic heart disease have not dampened enthusiasm for the application of cardiac stem cells and their paracrine factors for cardiac repair: the latter have proved superior to the mesenchymal stem cells used in most clinical trials in the past, some of which appear to have been conducted with sub-optimal rigor.
Collapse
|
77
|
Dierick F, Héry T, Hoareau-Coudert B, Mougenot N, Monceau V, Claude C, Crisan M, Besson V, Dorfmüller P, Marodon G, Fadel E, Humbert M, Yaniz-Galende E, Hulot JS, Marazzi G, Sassoon D, Soubrier F, Nadaud S. Resident PW1+ Progenitor Cells Participate in Vascular Remodeling During Pulmonary Arterial Hypertension. Circ Res 2016; 118:822-33. [PMID: 26838788 DOI: 10.1161/circresaha.115.307035] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 01/12/2016] [Indexed: 12/20/2022]
Abstract
RATIONALE Pulmonary arterial hypertension is characterized by vascular remodeling and neomuscularization. PW1(+) progenitor cells can differentiate into smooth muscle cells (SMCs) in vitro. OBJECTIVE To determine the role of pulmonary PW1(+) progenitor cells in vascular remodeling characteristic of pulmonary arterial hypertension. METHODS AND RESULTS We investigated their contribution during chronic hypoxia-induced vascular remodeling in Pw1(nLacZ+/-) mouse expressing β-galactosidase in PW1(+) cells and in differentiated cells derived from PW1(+) cells. PW1(+) progenitor cells are present in the perivascular zone in rodent and human control lungs. Using progenitor markers, 3 distinct myogenic PW1(+) cell populations were isolated from the mouse lung of which 2 were significantly increased after 4 days of chronic hypoxia. The number of proliferating pulmonary PW1(+) cells and the proportion of β-gal(+) vascular SMC were increased, indicating a recruitment of PW1(+) cells and their differentiation into vascular SMC during early chronic hypoxia-induced neomuscularization. CXCR4 inhibition using AMD3100 prevented PW1(+) cells differentiation into SMC but did not inhibit their proliferation. Bone marrow transplantation experiments showed that the newly formed β-gal(+) SMC were not derived from circulating bone marrow-derived PW1(+) progenitor cells, confirming a resident origin of the recruited PW1(+) cells. The number of pulmonary PW1(+) cells was also increased in rats after monocrotaline injection. In lung from pulmonary arterial hypertension patients, PW1-expressing cells were observed in large numbers in remodeled vascular structures. CONCLUSIONS These results demonstrate the existence of a novel population of resident SMC progenitor cells expressing PW1 and participating in pulmonary hypertension-associated vascular remodeling.
Collapse
Affiliation(s)
- France Dierick
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Tiphaine Héry
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Bénédicte Hoareau-Coudert
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Nathalie Mougenot
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Virginie Monceau
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Caroline Claude
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Mihaela Crisan
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Vanessa Besson
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Peter Dorfmüller
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Gilles Marodon
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Elie Fadel
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Marc Humbert
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Elisa Yaniz-Galende
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Jean-Sébastien Hulot
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Giovanna Marazzi
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - David Sassoon
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Florent Soubrier
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Sophie Nadaud
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.).
| |
Collapse
|
78
|
Abstract
Defective vascular and cardiomyocyte function are implicated in the development and progression of both heart failure with reduced and preserved ejection fraction. Any treatment option that augments these myocardial processes may therefore be of significant value. The platelet-derived growth factor (PDGF) family is involved in a wide range of growth processes and plays a key role in both regulating angiogenesis and mesenchymal cell development. Thus, PDGF may serve as a potent therapy for heart failure. While numerous animal studies have demonstrated beneficial cardiovascular effects of growth factor therapy, promising laboratory data has not yet translated to effective therapies. In this review, we outline the biological role of PDGF and summarize previous studies that have focused on the cardiovascular effects of normal PDGF signaling, administration of PDGF, and the effects of PDGF on stem cell therapy.
Collapse
Affiliation(s)
- John Medamana
- School of Medicine, Stony Brook University, Stony Brook, NY, 11794-8165, USA
| | - Richard A Clark
- Department of Dermatology, Health Science Center T16-060, Stony Brook University, Stony Brook, NY, 11794-8165, USA.
| | - Javed Butler
- Division of Cardiology, Health Science Center T16-080, Stony Brook University, Stony Brook, NY, 11794-8165, USA.
| |
Collapse
|
79
|
Alvarez R, Lee HL, Hong C, Wang CY. Single CD271 marker isolates mesenchymal stem cells from human dental pulp. Int J Oral Sci 2015; 7:205-12. [PMID: 26674422 PMCID: PMC5153594 DOI: 10.1038/ijos.2015.29] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2015] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a promising tool in regenerative medicine due to their capacity to differentiate into multiple lineages. In addition to MSCs isolated from bone marrow (BMSCs), adult MSCs are isolated from craniofacial tissues including dental pulp tissues (DPs) using various stem cell surface markers. However, there has been a lack of consensus on a set of surface makers that are reproducibly effective at isolating putative multipotent dental mesenchymal stem cells (DMSCs). In this study, we used different combinations of surface markers (CD51/CD140α, CD271, and STRO-1/CD146) to isolate homogeneous populations of DMSCs from heterogeneous dental pulp cells (DPCs) obtained from DP and compared their capacity to undergo multilineage differentiation. Fluorescence-activated cell sorting revealed that 27.3% of DPCs were CD51+/CD140α+, 10.6% were CD271+, and 0.3% were STRO-1+/CD146+. Under odontogenic conditions, all three subsets of isolated DMSCs exhibited differentiation capacity into odontogenic lineages. Among these isolated subsets of DMSCs, CD271+ DMSCs demonstrated the greatest odontogenic potential. While all three combinations of surface markers in this study successfully isolated DMSCs from DPCs, the single CD271 marker presents the most effective stem cell surface marker for identification of DMSCs with high odontogenic potential. Isolated CD271+ DMSCs could potentially be utilized for future clinical applications in dentistry and regenerative medicine.
Collapse
Affiliation(s)
- Ruth Alvarez
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA
| | - Hye-Lim Lee
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA
| | - Christine Hong
- Section of Orthodontics, School of Dentistry, University of California at Los Angeles, Los Angeles, USA
| | - Cun-Yu Wang
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California at Los Angeles, Los Angeles, USA
| |
Collapse
|
80
|
Alvarez R, Lee HL, Wang CY, Hong C. Characterization of the osteogenic potential of mesenchymal stem cells from human periodontal ligament based on cell surface markers. Int J Oral Sci 2015; 7:213-9. [PMID: 26674423 PMCID: PMC5153597 DOI: 10.1038/ijos.2015.42] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2015] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cell (MSC)-mediated therapy has been shown to be clinically effective in regenerating tissue defects. For improved regenerative therapy, it is critical to isolate homogenous populations of MSCs with high capacity to differentiate into appropriate tissues. The utilization of stem cell surface antigens provides a means to identify MSCs from various tissues. However, few surface markers that consistently isolate highly regenerative MSCs have been validated, making it challenging for routine clinical applications and making it all the more imperative to identify reliable surface markers. In this study, we used three surface marker combinations: CD51/CD140α, CD271, and STRO-1/CD146 for the isolation of homogenous populations of dental mesenchymal stem cells (DMSCs) from heterogeneous periodontal ligament cells (PDLCs). Fluorescence-activated cell sorting analysis revealed that 24% of PDLCs were CD51+/CD140α+, 0.8% were CD271+, and 2.4% were STRO-1+/CD146+. Sorted cell populations were further assessed for their multipotent properties by inducing osteogenic and chondrogenic differentiation. All three subsets of isolated DMSCs exhibited differentiation capacity into osteogenic and chondrogenic lineages but with varying degrees. CD271+ DMSCs demonstrated the greatest osteogenic potential with strong induction of osteogenic markers such as DLX5, RUNX2, and BGLAP. Our study provides evidence that surface marker combinations used in this study are sufficient markers for the isolation of DMSCs from PDLCs. These results provide important insight into using specific surface markers for identifying homogenous populations of DMSCs for their improved utilization in regenerative medicine.
Collapse
Affiliation(s)
- Ruth Alvarez
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA
| | - Hye-Lim Lee
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA
| | - Cun-Yu Wang
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA
| | - Christine Hong
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA.,Section of Orthodontics, School of Dentistry, University of California at Los Angeles, Los Angeles, USA
| |
Collapse
|
81
|
van Putten S, Shafieyan Y, Hinz B. Mechanical control of cardiac myofibroblasts. J Mol Cell Cardiol 2015; 93:133-42. [PMID: 26620422 DOI: 10.1016/j.yjmcc.2015.11.025] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 11/20/2015] [Accepted: 11/23/2015] [Indexed: 12/17/2022]
Abstract
Fibroblasts produce and turn over collagenous extracellular matrix as part of the normal adaptive response to increased mechanical load in the heart, e.g. during prolonged exercise. However, chronic overload as a consequence of hypertension or myocardial injury trigger a repair program that culminates in the formation of myofibroblasts. Myofibroblasts are opportunistically activated from various precursor cells that all acquire a phenotype promoting excessive collagen secretion and contraction of the neo-matrix into stiff scar tissue. Stiff fibrotic tissue reduces heart distensibility, impedes pumping and valve function, contributes to diastolic and systolic dysfunction, and affects myocardial electrical transmission, potentially leading to arrhythmia and heart failure. Here, we discuss how mechanical factors, such as matrix stiffness and strain, are feeding back and cooperate with cytokine signals to drive myofibroblast activation. We elaborate on the importance of considering the mechanical boundary conditions in the heart to generate better cell culture models for mechanistic studies of cardiac fibroblast function. Elements of the force transmission and mechanoperception apparatus acting in myofibroblasts are presented as potential therapeutic targets to treat fibrosis.
Collapse
Affiliation(s)
- Sander van Putten
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Yousef Shafieyan
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, ON M5S 3E2, Canada.
| |
Collapse
|
82
|
Chen WCW, Baily JE, Corselli M, Díaz ME, Sun B, Xiang G, Gray GA, Huard J, Péault B. Human myocardial pericytes: multipotent mesodermal precursors exhibiting cardiac specificity. Stem Cells 2015; 33:557-73. [PMID: 25336400 DOI: 10.1002/stem.1868] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 09/08/2014] [Accepted: 09/29/2014] [Indexed: 12/20/2022]
Abstract
Perivascular mesenchymal precursor cells (i.e., pericytes) reside in skeletal muscle where they contribute to myofiber regeneration; however, the existence of similar microvessel-associated regenerative precursor cells in cardiac muscle has not yet been documented. We tested whether microvascular pericytes within human myocardium exhibit phenotypes and multipotency similar to their anatomically and developmentally distinct counterparts. Fetal and adult human heart pericytes (hHPs) express canonical pericyte markers in situ, including CD146, NG2, platelet-derived growth factor receptor (PDGFR) β, PDGFRα, alpha-smooth muscle actin, and smooth muscle myosin heavy chain, but not CD117, CD133, and desmin, nor endothelial cell (EC) markers. hHPs were prospectively purified to homogeneity from ventricular myocardium by flow cytometry, based on a combination of positive- (CD146) and negative-selection (CD34, CD45, CD56, and CD117) cell lineage markers. Purified hHPs expanded in vitro were phenotypically similar to human skeletal muscle-derived pericytes (hSkMPs). hHPs express mesenchymal stem/stromal cell markers in situ and exhibited osteo-, chondro-, and adipogenic potentials but, importantly, no ability for skeletal myogenesis, diverging from pericytes of all other origins. hHPs supported network formation with/without ECs in Matrigel cultures; hHPs further stimulated angiogenic responses under hypoxia, markedly different from hSkMPs. The cardiomyogenic potential of hHPs was examined following 5-azacytidine treatment and neonatal cardiomyocyte coculture in vitro, and intramyocardial transplantation in vivo. Results indicated cardiomyocytic differentiation in a small fraction of hHPs. In conclusion, human myocardial pericytes share certain phenotypic and developmental similarities with their skeletal muscle homologs, yet exhibit different antigenic, myogenic, and angiogenic properties. This is the first example of an anatomical restriction in the developmental potential of pericytes as native mesenchymal stem cells.
Collapse
Affiliation(s)
- William C W Chen
- Department of Bioengineering, University of Pittsburgh, Pennsylvania, USA; Department of Orthopedic Surgery, University of Pittsburgh, Pennsylvania, USA; Stem Cell Research Centre, University of Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Abstract
Despite the increasing use of stem cells for regenerative-based cardiac therapy, the optimal stem cell population(s) remains in a cloud of uncertainty. In the past decade, the field has witnessed a surge of researchers discovering stem cell populations reported to directly and/or indirectly contribute to cardiac regeneration through processes of cardiomyogenic commitment and/or release of cardioprotective paracrine factors. This review centers upon defining basic biological characteristics of stem cells used for sustaining cardiac integrity during disease and maintenance of communication between the cardiac environment and stem cells. Given the limited successes achieved so far in regenerative therapy, the future requires development of unprecedented concepts involving combinatorial approaches to create and deliver the optimal stem cell(s) that will enhance myocardial healing.
Collapse
Affiliation(s)
- Pearl Quijada
- Integrated Regenerative Research Institute, Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | | |
Collapse
|
84
|
Zhang Y, Sivakumaran P, Newcomb AE, Hernandez D, Harris N, Khanabdali R, Liu GS, Kelly DJ, Pébay A, Hewitt AW, Boyle A, Harvey R, Morrison WA, Elliott DA, Dusting GJ, Lim SY. Cardiac Repair With a Novel Population of Mesenchymal Stem Cells Resident in the Human Heart. Stem Cells 2015; 33:3100-13. [PMID: 26184084 DOI: 10.1002/stem.2101] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 05/26/2015] [Accepted: 06/14/2015] [Indexed: 01/20/2023]
Abstract
Cardiac resident stem cells (CRSCs) hold much promise to treat heart disease but this remains a controversial field. Here, we describe a novel population of CRSCs, which are positive for W8B2 antigen and were obtained from adult human atrial appendages. W8B2(+) CRSCs exhibit a spindle-shaped morphology, are clonogenic and capable of self-renewal. W8B2(+) CRSCs show high expression of mesenchymal but not hematopoietic nor endothelial markers. W8B2(+) CRSCs expressed GATA4, HAND2, and TBX5, but not C-KIT, SCA-1, NKX2.5, PDGFRα, ISL1, or WT1. W8B2(+) CRSCs can differentiate into cardiovascular lineages and secrete a range of cytokines implicated in angiogenesis, chemotaxis, inflammation, extracellular matrix remodeling, cell growth, and survival. In vitro, conditioned medium collected from W8B2(+) CRSCs displayed prosurvival, proangiogenic, and promigratory effects on endothelial cells, superior to that of other adult stem cells tested, and additionally promoted survival and proliferation of neonatal rat cardiomyocytes. Intramyocardial transplantation of human W8B2(+) CRSCs into immunocompromised rats 1 week after myocardial infarction markedly improved cardiac function (∼40% improvement in ejection fraction) and reduced fibrotic scar tissue 4 weeks after infarction. Hearts treated with W8B2(+) CRSCs showed less adverse remodeling of the left ventricle, a greater number of proliferating cardiomyocytes (Ki67(+) cTnT(+) cells) in the remote region, higher myocardial vascular density, and greater infiltration of CD163(+) cells (a marker for M2 macrophages) into the border zone and scar regions. In summary, W8B2(+) CRSCs are distinct from currently known CRSCs found in human hearts, and as such may be an ideal cell source to repair myocardial damage after infarction.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Andrew E Newcomb
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia.,Department of Cardiothoracic Surgery, St. Vincent's Hospital, Melbourne, Victoria, Australia.,Vascular and Cardiac Surgery, The Cardiovascular Research Centre (CvRC), Australian Catholic University, Fitzroy, Victoria, Australia
| | - Damián Hernandez
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia.,O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Nicole Harris
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Ramin Khanabdali
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia.,O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Guei-Sheung Liu
- Department of Ophthalmology, University of Melbourne, Melbourne, Victoria, Australia.,Centre for Eye Research Australia & Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Darren J Kelly
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Alice Pébay
- Department of Ophthalmology, University of Melbourne, Melbourne, Victoria, Australia.,Centre for Eye Research Australia & Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Alex W Hewitt
- Department of Ophthalmology, University of Melbourne, Melbourne, Victoria, Australia.,Centre for Eye Research Australia & Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Andrew Boyle
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Richard Harvey
- Developmental and Stem Cell Biology, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Wayne A Morrison
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia.,O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,AORTEC, Faculty of Health Sciences, Australian Catholic University, Fitzroy, Victoria, Australia
| | - David A Elliott
- Cardiac Development, Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Gregory J Dusting
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia.,Department of Ophthalmology, University of Melbourne, Melbourne, Victoria, Australia.,O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Centre for Eye Research Australia & Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Shiang Y Lim
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia.,O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| |
Collapse
|
85
|
Abstract
Myofibroblasts are activated in response to tissue injury with the primary task to repair lost or damaged extracellular matrix. Enhanced collagen secretion and subsequent contraction - scarring - are part of the normal wound healing response and crucial to restore tissue integrity. Due to myofibroblasts ability to repair but not regenerate, accumulation of scar tissue is always associated with reduced organ performance. This is a fair price to pay by the body for not falling apart. Whereas myofibroblasts typically vanish after successful repair, dysregulation of the normal repair process can lead to persistent myofibroblast activation, for instance by chronic inflammation or mechanical stress in the tissue. Excessive repair leads to the accumulation of stiff collagenous ECM contractures - fibrosis - with dramatic consequences for organ function. The clinical need to terminate detrimental myofibroblast activities has stimulated researchers to answer a number of essential questions: where do myofibroblasts come from, what are the factors leading to their activation, how do we discriminate myofibroblasts from other cells, what is the molecular basis for their contractile activity, and how can we stop or at least control them? This article reviews the current state of the myofibroblast literature by emphasizing their role in ocular repair and fibrosis. It appears that although the eye is quite an extraordinary organ, ocular myofibroblasts behave or misbehave just like their siblings in other organs.
Collapse
Affiliation(s)
- Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, 150 College Street, FitzGerald Building, Room 234, Toronto, M5S 3E2 Ontario, Canada.
| |
Collapse
|
86
|
Platelet-Derived Growth Factor Receptor Alpha as a Marker of Mesenchymal Stem Cells in Development and Stem Cell Biology. Stem Cells Int 2015; 2015:362753. [PMID: 26257789 PMCID: PMC4519552 DOI: 10.1155/2015/362753] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 05/24/2015] [Accepted: 06/17/2015] [Indexed: 11/17/2022] Open
Abstract
Three decades on, the mesenchymal stem cells (MSCs) have been intensively researched on the bench top and used clinically. However, ambiguity still exists in regard to their anatomical locations, identities, functions, and extent of their differentiative abilities. One of the major impediments in the quest of the MSC research has been lack of appropriate in vivo markers. In recent years, this obstacle has been resolved to some degree as PDGFRα emerges as an important mesenchymal stem cell marker. Accumulating lines of evidence are showing that the PDGFRα (+) cells reside in the perivascular locations of many adult interstitium and fulfil the classic concepts of MSCs in vitro and in vivo. PDGFRα has long been recognised for its roles in the mesoderm formation and connective tissue development during the embryogenesis. Current review describes the lines of evidence regarding the role of PDGFRα in morphogenesis and differentiation and its implications for MSC biology.
Collapse
|
87
|
Cardiac fibroblasts: from development to heart failure. J Mol Med (Berl) 2015; 93:823-30. [PMID: 26169532 DOI: 10.1007/s00109-015-1314-y] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/22/2015] [Accepted: 06/24/2015] [Indexed: 01/18/2023]
Abstract
Cardiac fibroblasts are a major cell population of the heart and are characterized by their capacity to produce extracellular matrix (ECM). In hearts subjected to pressure overload, excessive fibroblast accumulation is responsible for fibrosis of the myocardium, a major clinical issue. Hence, understanding mechanisms generating fibroblasts in this context has become a key question in the cardiovascular field. Recent studies now point to the activation of resident fibroblasts as the underlying cause of fibrosis. However, de novo generation of fibroblasts from endothelium and circulating hematopoietic cells has also been proposed to significantly contribute to fibrosis. Here, we discuss the latest findings on fibroblast origins, with a particular emphasis on the pressure overload model, and the implication of these findings for the development of anti-fibrotic therapies that are currently lacking.
Collapse
|
88
|
Noseda M, Harada M, McSweeney S, Leja T, Belian E, Stuckey DJ, Abreu Paiva MS, Habib J, Macaulay I, de Smith AJ, al-Beidh F, Sampson R, Lumbers RT, Rao P, Harding SE, Blakemore AIF, Eirik Jacobsen S, Barahona M, Schneider MD. PDGFRα demarcates the cardiogenic clonogenic Sca1+ stem/progenitor cell in adult murine myocardium. Nat Commun 2015; 6:6930. [PMID: 25980517 PMCID: PMC4479024 DOI: 10.1038/ncomms7930] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/16/2015] [Indexed: 12/24/2022] Open
Abstract
Cardiac progenitor/stem cells in adult hearts represent an attractive therapeutic target for heart regeneration, though (inter)-relationships among reported cells remain obscure. Using single-cell qRT-PCR and clonal analyses, here we define four subpopulations of cardiac progenitor/stem cells in adult mouse myocardium all sharing stem cell antigen-1 (Sca1), based on side population (SP) phenotype, PECAM-1 (CD31) and platelet-derived growth factor receptor-α (PDGFRα) expression. SP status predicts clonogenicity and cardiogenic gene expression (Gata4/6, Hand2 and Tbx5/20), properties segregating more specifically to PDGFRα(+) cells. Clonal progeny of single Sca1(+) SP cells show cardiomyocyte, endothelial and smooth muscle lineage potential after cardiac grafting, augmenting cardiac function although durable engraftment is rare. PDGFRα(-) cells are characterized by Kdr/Flk1, Cdh5, CD31 and lack of clonogenicity. PDGFRα(+)/CD31(-) cells derive from cells formerly expressing Mesp1, Nkx2-5, Isl1, Gata5 and Wt1, distinct from PDGFRα(-)/CD31(+) cells (Gata5 low; Flk1 and Tie2 high). Thus, PDGFRα demarcates the clonogenic cardiogenic Sca1(+) stem/progenitor cell.
Collapse
Affiliation(s)
- Michela Noseda
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Mutsuo Harada
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Sara McSweeney
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Thomas Leja
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Elisa Belian
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Daniel J. Stuckey
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
- Centre for Advanced Biomedical Imaging (CABI), University College London, London WC1E 6DD, UK
| | - Marta S. Abreu Paiva
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Josef Habib
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
- Department of Biomedical Engineering, King's College London, London SE1 7EH, UK
| | - Iain Macaulay
- Haematopoietic Stem Cell Biology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Adam J. de Smith
- Department of Medicine, Imperial College London, London SW7 2AZ, UK
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California 94143, USA
| | - Farah al-Beidh
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Robert Sampson
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - R. Thomas Lumbers
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Pulivarthi Rao
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Sian E. Harding
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | | | - Sten Eirik Jacobsen
- Haematopoietic Stem Cell Biology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Mauricio Barahona
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- Department of Mathematics, Imperial College London, London SW7 2AZ, UK
| | - Michael D. Schneider
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
89
|
Pessina P, Kharraz Y, Jardí M, Fukada SI, Serrano AL, Perdiguero E, Muñoz-Cánoves P. Fibrogenic Cell Plasticity Blunts Tissue Regeneration and Aggravates Muscular Dystrophy. Stem Cell Reports 2015; 4:1046-60. [PMID: 25981413 PMCID: PMC4472037 DOI: 10.1016/j.stemcr.2015.04.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 04/17/2015] [Accepted: 04/17/2015] [Indexed: 01/26/2023] Open
Abstract
Preservation of cell identity is necessary for homeostasis of most adult tissues. This process is challenged every time a tissue undergoes regeneration after stress or injury. In the lethal Duchenne muscular dystrophy (DMD), skeletal muscle regenerative capacity declines gradually as fibrosis increases. Using genetically engineered tracing mice, we demonstrate that, in dystrophic muscle, specialized cells of muscular, endothelial, and hematopoietic origins gain plasticity toward a fibrogenic fate via a TGFβ-mediated pathway. This results in loss of cellular identity and normal function, with deleterious consequences for regeneration. Furthermore, this fibrogenic process involves acquisition of a mesenchymal progenitor multipotent status, illustrating a link between fibrogenesis and gain of progenitor cell functions. As this plasticity also was observed in DMD patients, we propose that mesenchymal transitions impair regeneration and worsen diseases with a fibrotic component.
Collapse
Affiliation(s)
- Patrizia Pessina
- Cell Biology Group, Department of Experimental and Health Sciences (DCEXS), Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Yacine Kharraz
- Cell Biology Group, Department of Experimental and Health Sciences (DCEXS), Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Mercè Jardí
- Cell Biology Group, Department of Experimental and Health Sciences (DCEXS), Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - So-ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Antonio L Serrano
- Cell Biology Group, Department of Experimental and Health Sciences (DCEXS), Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Eusebio Perdiguero
- Cell Biology Group, Department of Experimental and Health Sciences (DCEXS), Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Cell Biology Group, Department of Experimental and Health Sciences (DCEXS), Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Dr. Aiguader, 88, 08003 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys, 23, 08010 Barcelona, Spain.
| |
Collapse
|
90
|
Let-7 family of microRNA is required for maturation and adult-like metabolism in stem cell-derived cardiomyocytes. Proc Natl Acad Sci U S A 2015; 112:E2785-94. [PMID: 25964336 DOI: 10.1073/pnas.1424042112] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In metazoans, transition from fetal to adult heart is accompanied by a switch in energy metabolism-glycolysis to fatty acid oxidation. The molecular factors regulating this metabolic switch remain largely unexplored. We first demonstrate that the molecular signatures in 1-year (y) matured human embryonic stem cell-derived cardiomyocytes (hESC-CMs) are similar to those seen in in vivo-derived mature cardiac tissues, thus making them an excellent model to study human cardiac maturation. We further show that let-7 is the most highly up-regulated microRNA (miRNA) family during in vitro human cardiac maturation. Gain- and loss-of-function analyses of let-7g in hESC-CMs demonstrate it is both required and sufficient for maturation, but not for early differentiation of CMs. Overexpression of let-7 family members in hESC-CMs enhances cell size, sarcomere length, force of contraction, and respiratory capacity. Interestingly, large-scale expression data, target analysis, and metabolic flux assays suggest this let-7-driven CM maturation could be a result of down-regulation of the phosphoinositide 3 kinase (PI3K)/AKT protein kinase/insulin pathway and an up-regulation of fatty acid metabolism. These results indicate let-7 is an important mediator in augmenting metabolic energetics in maturing CMs. Promoting maturation of hESC-CMs with let-7 overexpression will be highly significant for basic and applied research.
Collapse
|
91
|
Dixit P, Katare R. Challenges in identifying the best source of stem cells for cardiac regeneration therapy. Stem Cell Res Ther 2015; 6:26. [PMID: 25886612 PMCID: PMC4357059 DOI: 10.1186/s13287-015-0010-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 02/17/2015] [Indexed: 12/14/2022] Open
Abstract
The overall clinical cardiac regeneration experience suggests that stem cell therapy can be safely performed, but it also underlines the need for reproducible results for their effective use in a real-world scenario. One of the significant challenges is the identification and selection of the best suited stem cell type for regeneration therapy. Bone marrow mononuclear cells, bone marrow-derived mesenchymal stem cells, resident or endogenous cardiac stem cells, endothelial progenitor cells and induced pluripotent stem cells are some of the stem cell types which have been extensively tested for their ability to regenerate the lost myocardium. While most of these cell types are being evaluated in clinical trials for their safety and efficacy, results show significant heterogeneity in terms of efficacy. The enthusiasm surrounding regenerative medicine in the heart has been dampened by the reports of poor survival, proliferation, engraftment, and differentiation of the transplanted cells. Therefore, the primary challenge is to create clearcut evidence on what actually drives the improvement of cardiac function after the administration of stem cells. In this review, we provide an overview of different types of stem cells currently being considered for cardiac regeneration and discuss why associated factors such as practicality and difficulty in cell collection should also be considered when selecting the stem cells for transplantation. Next, we discuss how the experimental variables (type of disease, marker-based selection and use of different isolation techniques) can influence the study outcome. Finally, we provide an outline of the molecular and genetic approaches to increase the functional ability of stem cells before and after transplantation.
Collapse
Affiliation(s)
- Parul Dixit
- Department of Physiology, HeartOtago, Otago School of Medical Sciences, University of Otago, Dunedin, 9010, New Zealand.
| | - Rajesh Katare
- Department of Physiology, HeartOtago, Otago School of Medical Sciences, University of Otago, Dunedin, 9010, New Zealand.
| |
Collapse
|
92
|
Affiliation(s)
- Michela Noseda
- National Heart and Lung Institute, Imperial College London
| | | | | |
Collapse
|
93
|
Duerrschmid C, Trial J, Wang Y, Entman ML, Haudek SB. Tumor necrosis factor: a mechanistic link between angiotensin-II-induced cardiac inflammation and fibrosis. Circ Heart Fail 2014; 8:352-61. [PMID: 25550440 DOI: 10.1161/circheartfailure.114.001893] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Continuous angiotensin-II infusion induced the uptake of monocytic fibroblast precursors that initiated the development of cardiac fibrosis; these cells and concurrent fibrosis were absent in mice lacking tumor necrosis factor receptor 1 (TNFR1). We now investigated their cellular origin and temporal uptake and the involvement of TNFR1 in monocyte-to-fibroblast differentiation. METHODS AND RESULTS Within a day, angiotensin-II induced a proinflammatory environment characterized by production of inflammatory chemokines, cytokines, and TH1-interleukins and uptake of bone marrow-derived M1 cells. After a week, the cardiac environment changed to profibrotic with growth factor and TH2-interleukin synthesis, uptake of bone marrow-derived M2 cells, and the presence of M2-related fibroblasts. TNFR1 signaling was not necessary for early M1 uptake, but its absence diminished the amount of M2 cells. TNFR1-knockout hearts also showed reduced levels of cytokine expression, but not of TH-related lymphokines. Reconstitution of wild-type bone marrow into TNFR1-knockout mice was sufficient to restore M2 uptake, upregulation of proinflammatory and profibrotic genes, and development of fibrosis in response to angiotensin-II. We also developed an in vitro mouse monocyte-to-fibroblast maturation assay that confirmed the essential role of TNFR1 in the sequential progression of monocyte activation and fibroblast formation. CONCLUSIONS Development of cardiac fibrosis in response to angiotensin-II was mediated by myeloid precursors and consisted of 2 stages. A primary M1 inflammatory response was followed by a subsequent M2 fibrotic response. Although the first phase seemed to be independent of TNFR1 signaling, the later phase (and development of fibrosis) was abrogated by deletion of TNFR1.
Collapse
Affiliation(s)
- Clemens Duerrschmid
- From the Division of Cardiovascular Sciences (C.D., J.T., M.L.E., S.B.H.) and Division of Nephrology (Y.W.), Department of Medicine, Baylor College of Medicine, Houston, TX
| | - JoAnn Trial
- From the Division of Cardiovascular Sciences (C.D., J.T., M.L.E., S.B.H.) and Division of Nephrology (Y.W.), Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Yanlin Wang
- From the Division of Cardiovascular Sciences (C.D., J.T., M.L.E., S.B.H.) and Division of Nephrology (Y.W.), Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Mark L Entman
- From the Division of Cardiovascular Sciences (C.D., J.T., M.L.E., S.B.H.) and Division of Nephrology (Y.W.), Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Sandra B Haudek
- From the Division of Cardiovascular Sciences (C.D., J.T., M.L.E., S.B.H.) and Division of Nephrology (Y.W.), Department of Medicine, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
94
|
Ge Z, Lal S, Le TYL, Dos Remedios C, Chong JJH. Cardiac stem cells: translation to human studies. Biophys Rev 2014; 7:127-139. [PMID: 28509972 DOI: 10.1007/s12551-014-0148-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/13/2014] [Indexed: 02/08/2023] Open
Abstract
The discovery of multiple classes of cardiac progenitor cells in the adult mammalian heart has generated hope for their use as a therapeutic in heart failure. However, successful results from animal models have not always yielded similar findings in human studies. Recent Phase I/II trials of c-Kit (SCIPIO) and cardiosphere-based (CADUCEUS) cardiac progenitor cells have demonstrated safety and some therapeutic efficacy. Gaps remain in our understanding of the origins, function and relationships between the different progenitor cell families, many of which are heterogeneous populations with overlapping definitions. Another challenge lies in the limitations of small animal models in replicating the human heart. Cryopreserved human cardiac tissue provides a readily available source of cardiac progenitor cells and may help address these questions. We review important findings and relative unknowns of the main classes of cardiac progenitor cells, highlighting differences between animal and human studies.
Collapse
Affiliation(s)
- Zijun Ge
- Bosch Institute, The University of Sydney, Sydney, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Sean Lal
- Bosch Institute, The University of Sydney, Sydney, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Thi Y L Le
- Department of Cardiology Westmead Hospital, Sydney, NSW, Australia.,Centre for Heart Research, Westmead Millennium Institute for Medical Research, 176 Hawkesbury Road, Westmead, Sydney, NSW, Australia, 2145
| | | | - James J H Chong
- Department of Cardiology Westmead Hospital, Sydney, NSW, Australia. .,Sydney Medical School, University of Sydney, Sydney, NSW, Australia. .,Centre for Heart Research, Westmead Millennium Institute for Medical Research, 176 Hawkesbury Road, Westmead, Sydney, NSW, Australia, 2145.
| |
Collapse
|
95
|
Chong JJ, Forte E, Harvey RP. Developmental origins and lineage descendants of endogenous adult cardiac progenitor cells. Stem Cell Res 2014; 13:592-614. [DOI: 10.1016/j.scr.2014.09.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 09/24/2014] [Accepted: 09/26/2014] [Indexed: 12/30/2022] Open
|
96
|
Matar AA, Chong JJ. Stem cell therapy for cardiac dysfunction. SPRINGERPLUS 2014; 3:440. [PMID: 25191634 PMCID: PMC4153875 DOI: 10.1186/2193-1801-3-440] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/11/2014] [Indexed: 01/05/2023]
Abstract
Following significant injury, the heart undergoes induced compensation and gradually deteriorates towards impending heart failure. Current therapy slows but does not halt the resultant adverse remodeling. Stem cell therapy, however, has the potential to regenerate or repair infarcted heart tissue and therefore is a promising therapeutic strategy undergoing intensive investigation. Due to the wide range of stem cells investigated, it is difficult to navigate this field. This review aims to summarize the main types of stem cells (both of cardiac and extra-cardiac origin) that possess promising therapeutic potential. Particular focus is placed on clinical trials supporting this therapeutic strategy.
Collapse
Affiliation(s)
- Amer A Matar
- Sydney Medical School, University of Sydney, Sydney, NSW Australia
| | - James Jh Chong
- Sydney Medical School, University of Sydney, Sydney, NSW Australia ; Department of Cardiology, Westmead Hospital, Sydney, NSW Australia ; Centre for Heart Research, Westmead Millennium Institute, Sydney, NSW Australia
| |
Collapse
|
97
|
Kaur K, Yang J, Eisenberg CA, Eisenberg LM. 5-azacytidine promotes the transdifferentiation of cardiac cells to skeletal myocytes. Cell Reprogram 2014; 16:324-30. [PMID: 25090621 DOI: 10.1089/cell.2014.0021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The DNA methylation inhibitor 5-azacytidine is widely used to stimulate the cardiac differentiation of stem cells. However, 5-azacytidine has long been employed as a tool for stimulating skeletal myogenesis. Yet, it is unclear whether the ability of 5-azacytidine to promote both cardiac and skeletal myogenesis is dependent strictly on the native potential of the starting cell population or if this drug is a transdifferentiation agent. To address this issue, we examined the effect of 5-azacytidine on cultures of adult mouse atrial tissue, which contains cardiac but not skeletal muscle progenitors. Exposure to 5-azacytidine caused atrial cells to elongate and increased the presence of fat globules within the cultures. 5-Azacytidine also induced expression of the skeletal myogenic transcription factors MyoD and myogenin. 5-Azacytidine pretreatments allowed atrial cells to undergo adipogenesis or skeletal myogenesis when subsequently cultured with either insulin and dexamethasone or low-serum media, respectively. The presence of skeletal myocytes in atrial cultures was indicated by dual staining for myogenin and sarcomeric α-actin. These data demonstrate that 5-azacytidine converts cardiac cells to noncardiac cell types and suggests that this drug has a compromised efficacy as a cardiac differentiation factor.
Collapse
Affiliation(s)
- Keerat Kaur
- New York Medical College/Westchester Medical Center Stem Cell Laboratory, Departments of Physiology and Medicine, New York Medical College , Valhalla, NY, 10595
| | | | | | | |
Collapse
|
98
|
Valente M, Nascimento DS, Cumano A, Pinto-do-Ó P. Sca-1+ cardiac progenitor cells and heart-making: a critical synopsis. Stem Cells Dev 2014; 23:2263-73. [PMID: 24926741 DOI: 10.1089/scd.2014.0197] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The identification, in the adult, of cardiomyocyte turnover events and of cardiac progenitor cells (CPCs) has revolutionized the field of cardiovascular medicine. However, the low rate of CPCs differentiation events reported both in vitro and in vivo, even after injury, raised concerns on the biological significance of these subsets. In this Comprehensive Review, we discuss the current understanding of cardiac Lin(-)Sca-1(+) cells in light of what is also known for cellular compartments with similar phenotypes in other organs. The Lin(-)Sca-1(+) heart subset is heterogeneous and displays a mesenchymal profile, characterized by a limited ability to generate cardiomyocytes in vitro and in vivo, even after injury. There is no evidence for Sca-1 expression in embryonic cardiovascular progenitors. In other organs, Sca-1 expression is mainly observed on mesoderm-derived cells, although it is not restricted to stem/progenitor cell populations. It is urgent to determine, at a single cell level, to which extent cardiac Lin(-)Sca-1(+) cells overlap with the fibroblast compartment.
Collapse
Affiliation(s)
- Mariana Valente
- 1 Stem-Cell Microenvironments in Repair/Regeneration Team, Microenvironments for NewTherapies Group, INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto , Porto, Portugal
| | | | | | | |
Collapse
|
99
|
Chong JJH, Yang X, Don CW, Minami E, Liu YW, Weyers JJ, Mahoney WM, Van Biber B, Cook SM, Palpant NJ, Gantz JA, Fugate JA, Muskheli V, Gough GM, Vogel KW, Astley CA, Hotchkiss CE, Baldessari A, Pabon L, Reinecke H, Gill EA, Nelson V, Kiem HP, Laflamme MA, Murry CE. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 2014; 510:273-7. [PMID: 24776797 PMCID: PMC4154594 DOI: 10.1038/nature13233] [Citation(s) in RCA: 1025] [Impact Index Per Article: 93.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 03/06/2014] [Indexed: 12/16/2022]
Abstract
Pluripotent stem cells provide a potential solution to current epidemic rates of heart failure by providing human cardiomyocytes to support heart regeneration. Studies of human embryonic-stem-cell-derived cardiomyocytes (hESC-CMs) in small-animal models have shown favourable effects of this treatment. However, it remains unknown whether clinical-scale hESC-CM transplantation is feasible, safe or can provide sufficient myocardial regeneration. Here we show that hESC-CMs can be produced at a clinical scale (more than one billion cells per batch) and cryopreserved with good viability. Using a non-human primate model of myocardial ischaemia followed by reperfusion, we show that cryopreservation and intra-myocardial delivery of one billion hESC-CMs generates extensive remuscularization of the infarcted heart. The hESC-CMs showed progressive but incomplete maturation over a 3-month period. Grafts were perfused by host vasculature, and electromechanical junctions between graft and host myocytes were present within 2 weeks of engraftment. Importantly, grafts showed regular calcium transients that were synchronized to the host electrocardiogram, indicating electromechanical coupling. In contrast to small-animal models, non-fatal ventricular arrhythmias were observed in hESC-CM-engrafted primates. Thus, hESC-CMs can remuscularize substantial amounts of the infarcted monkey heart. Comparable remuscularization of a human heart should be possible, but potential arrhythmic complications need to be overcome.
Collapse
Affiliation(s)
- James J H Chong
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Cardiology Westmead Hospital, Westmead, New South Wales 2145, Australia [4] School of Medicine, University of Sydney, Sydney, New South Wales 2006, Australia [5] Department of Pathology, University of Washington, Seattle, Washington 98195, USA [6] University of Sydney School of Medicine, Sydney, New South Wales 2006, Australia and Westmead Millennium Institute and Westmead Hospital, Westmead, New South Wales 2145, Australia
| | - Xiulan Yang
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - Creighton W Don
- Department of Medicine/Cardiology, University of Washington, Seattle, Washington 98195, USA
| | - Elina Minami
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA [4] Department of Medicine/Cardiology, University of Washington, Seattle, Washington 98195, USA
| | - Yen-Wen Liu
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - Jill J Weyers
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - William M Mahoney
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - Benjamin Van Biber
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - Savannah M Cook
- Department of Comparative Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA
| | - Nathan J Palpant
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - Jay A Gantz
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA [4] Department of Bioengineering, University of Washington, Seattle, Washington 98195, USA
| | - James A Fugate
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - Veronica Muskheli
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - G Michael Gough
- Washington National Primate Research Center, University of Washington, Seattle, Washington 98195, USA
| | - Keith W Vogel
- Washington National Primate Research Center, University of Washington, Seattle, Washington 98195, USA
| | - Cliff A Astley
- Washington National Primate Research Center, University of Washington, Seattle, Washington 98195, USA
| | - Charlotte E Hotchkiss
- Washington National Primate Research Center, University of Washington, Seattle, Washington 98195, USA
| | - Audrey Baldessari
- Washington National Primate Research Center, University of Washington, Seattle, Washington 98195, USA
| | - Lil Pabon
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - Hans Reinecke
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - Edward A Gill
- Department of Medicine/Cardiology, University of Washington, Seattle, Washington 98195, USA
| | - Veronica Nelson
- Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Hans-Peter Kiem
- 1] Department of Pathology, University of Washington, Seattle, Washington 98195, USA [2] Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Michael A Laflamme
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | - Charles E Murry
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, USA [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA [3] Department of Pathology, University of Washington, Seattle, Washington 98195, USA [4] Department of Medicine/Cardiology, University of Washington, Seattle, Washington 98195, USA [5] Department of Bioengineering, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
100
|
Hayes BJ, Riehle KJ, Shimizu-Albergine M, Bauer RL, Hudkins KL, Johansson F, Yeh MM, Mahoney WM, Yeung RS, Campbell JS. Activation of platelet-derived growth factor receptor alpha contributes to liver fibrosis. PLoS One 2014; 9:e92925. [PMID: 24667490 PMCID: PMC3965491 DOI: 10.1371/journal.pone.0092925] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 02/27/2014] [Indexed: 01/27/2023] Open
Abstract
Chronic liver injury leads to fibrosis, cirrhosis, and loss of liver function. Liver cirrhosis is the 12th leading cause of death in the United States, and it is the primary risk factor for developing liver cancer. Fibrosis and cirrhosis result from activation of hepatic stellate cells (HSCs), which are the primary collagen producing cell type in the liver. Here, we show that platelet-derived growth factor receptor α (PDGFRα) is expressed by human HSCs, and PDGFRα expression is elevated in human liver disease. Using a green fluorescent protein (GFP) reporter mouse strain, we evaluated the role of PDGFRα in liver disease in mice and found that mouse HSCs express PDGFRα and expression is upregulated during carbon tetrachloride (CCl4) induced liver injury and fibrosis injection. This fibrotic response is reduced in Pdgfrα heterozygous mice, consistent with the hypothesis that liver fibrosis requires upregulation and activation of PDGFRα. These results indicate that Pdgfrα expression is important in the fibrotic response to liver injury in humans and mice, and suggest that blocking PDGFRα–specific signaling pathways in HSCs may provide therapeutic benefit for patients with chronic liver disease.
Collapse
Affiliation(s)
- Brian J. Hayes
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Kimberly J. Riehle
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| | - Masami Shimizu-Albergine
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Renay L. Bauer
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Kelly L. Hudkins
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Fredrik Johansson
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Matthew M. Yeh
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - William M. Mahoney
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Raymond S. Yeung
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| | - Jean S. Campbell
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|