201
|
Lerman DA, Alotti N, Ume KL, Péault B. Cardiac Repair and Regeneration: The Value of Cell Therapies. Eur Cardiol 2015; 11:43-48. [PMID: 27499812 DOI: 10.15420/ecr.2016:8:1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ischaemic heart disease is the predominant contributor to cardiovascular morbidity and mortality; one million myocardial Infarctions occur per year in the USA, while more than five million patients suffer from chronic heart failure. Recently, heart failure has been singled out as an epidemic and is a staggering clinical and public health problem associated with significant mortality, morbidity and healthcare expenditures, particularly among those aged ≥65 years. Death rates have improved dramatically over the last four decades, but new approaches are nevertheless urgently needed for those patients who go on to develop ventricular dysfunction and chronic heart failure. Over the past decade, stem cell transplantation has emerged as a promising therapeutic strategy for acute or chronic ischaemic cardiomyopathy. Multiple candidate cell types have been used in preclinical animal models and in humans to repair or regenerate the injured heart, either directly or indirectly (through paracrine effects), including: embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), neonatal cardiomyocytes, skeletal myoblasts (SKMs), endothelial progenitor cells, bone marrow mononuclear cells (BMMNCs), mesenchymal stem cells (MSCs) and, most recently, cardiac stem cells (CSCs). Although no consensus has emerged yet, the ideal cell type for the treatment of heart disease should: (a) improve heart function; (b) create healthy and functional cardiac muscle and vasculature, integrated into the host tissue; (c) be amenable to delivery by minimally invasive clinical methods; (d) be available 'off the shelf' as a standardised reagent; (e) be tolerated by the immune system; (f) be safe oncologically, i.e. not create tumours; and (g) circumvent societal ethical concerns. At present, it is not clear whether such a 'perfect' stem cell exists; what is apparent, however, is that some cell types are more promising than others. In this brief review, we provide ongoing data on agreement and controversy arising from clinical trials and touch upon the future directions of cell therapy for heart disease.
Collapse
Affiliation(s)
- Daniel Alejandro Lerman
- Department of Cardiothoracic Surgery, Royal Infirmary Hospital of Edinburgh (NHS Lothian), University of Edinburgh, Scotland, UK; MRC Centre for Regenerative Medicine and College of Medicine and Veterinary, University of Edinburgh, Scotland, UK
| | | | | | - Bruno Péault
- MRC Centre for Regenerative Medicine and College of Medicine and Veterinary, University of Edinburgh, Scotland, UK; David Geffen School of Medicine at UCLA, Orthopaedic Hospital Research Centre, University of California at Los Angeles, USA
| |
Collapse
|
202
|
Poché RA, Hsu CW, McElwee ML, Burns AR, Dickinson ME. Macrophages engulf endothelial cell membrane particles preceding pupillary membrane capillary regression. Dev Biol 2015; 403:30-42. [PMID: 25912686 DOI: 10.1016/j.ydbio.2015.03.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 01/08/2023]
Abstract
Programmed capillary regression and remodeling are essential developmental processes. However, the cellular and molecular mechanisms that regulate vessel regression are only the beginning to be understood. Here, using in vivo, dynamic, confocal imaging of mouse transgenic reporters as well as static confocal and electron microscopy, we studied the embryonic development and postnatal regression of the transient mouse pupillary membrane (PM) vasculature. This approach allowed us to directly observe the precise temporal sequence of cellular events preceding and during the elimination of the PM from the mouse eye. Imaging of Tcf/Lef-H2B::GFP Wnt-reporter mice uncovered that, unlike the hyaloid vasculature of the posterior eye, a PM endothelial cell (EC) Wnt/β-catenin response is unlikely to be part of the regression mechanism. Live imaging of EC and macrophage dynamics revealed highly active Csf1r-GFP+ macrophages making direct contact with the Flk1-myr::mCherry+ vessel surface and with membrane protrusions or filopodia extending from the ECs. Flk1-myr::mCherry+ EC membrane particles were observed on and around ECs as well as within macrophages. Electron microscopy studies confirmed that they were in phagosomes within macrophages, indicating that the macrophages engulfed the membrane particles. Interestingly, EC plasma membrane uptake by PM macrophages did not correlate with apoptosis and was found shortly after vessel formation at mid-gestation stages in the embryo; long before vessel regression begins during postnatal development. Additionally, genetic ablation of macrophages showed that EC membrane particles were still shed in the absence of macrophages suggesting that macrophages do not induce the formation or release of EC microparticles. These studies have uncovered a novel event during programmed capillary regression in which resident macrophages scavenge endothelial cell microparticles released from the PM vessels. This finding suggests that there may be an initial disruption in vessel homeostasis embryonically as the PM forms that may underlie its ultimate regression postnatally.
Collapse
Affiliation(s)
- Ross A Poché
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States; Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, United States
| | - Chih-Wei Hsu
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States; Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, United States
| | - Melissa L McElwee
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States
| | - Alan R Burns
- College of Optometry, University of Houston, Houston, TX, United States
| | - Mary E Dickinson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States; Program in Developmental Biology, Baylor College of Medicine, Houston, TX, United States; Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, United States; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
203
|
Endogenous brain pericytes are widely activated and contribute to mouse glioma microvasculature. PLoS One 2015; 10:e0123553. [PMID: 25875288 PMCID: PMC4395339 DOI: 10.1371/journal.pone.0123553] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/05/2015] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common brain tumor in adults. It presents an extremely challenging clinical problem, and treatment very frequently fails due to the infiltrative growth, facilitated by extensive angiogenesis and neovascularization. Pericytes constitute an important part of the GBM microvasculature. The contribution of endogenous brain pericytes to the tumor vasculature in GBM is, however, unclear. In this study, we determine the site of activation and the extent of contribution of endogenous brain pericytes to the GBM vasculature. GL261 mouse glioma was orthotopically implanted in mice expressing green fluorescent protein (GFP) under the pericyte marker regulator of G protein signaling 5 (RGS5). Host pericytes were not only activated within the glioma, but also in cortical areas overlying the tumor, the ipsilateral subventricular zone and within the hemisphere contralateral to the tumor. The host-derived activated pericytes that infiltrated the glioma were mainly localized to the tumor vessel wall. Infiltrating GFP positive pericytes co-expressed the pericyte markers platelet-derived growth factor receptor-β (PDGFR-β) and neuron-glial antigen 2. Interestingly, more than half of all PDGFR-β positive pericytes within the tumor were contributed by the host brain. We did not find any evidence that RGS5 positive pericytes adopt another phenotype within glioma in this paradigm. We conclude that endogenous pericytes become activated in widespread areas of the brain in response to an orthotopic mouse glioma. Host pericytes are recruited into the tumor and constitute a major part of the tumor pericyte population.
Collapse
|
204
|
Braunger BM, Leimbeck SV, Schlecht A, Volz C, Jägle H, Tamm ER. Deletion of ocular transforming growth factor β signaling mimics essential characteristics of diabetic retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1749-68. [PMID: 25857227 DOI: 10.1016/j.ajpath.2015.02.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/22/2014] [Accepted: 02/03/2015] [Indexed: 01/16/2023]
Abstract
Diabetic retinopathy, a major cause of blindness, is characterized by a distinct phenotype. The molecular causes of the phenotype are not sufficiently clear. Here, we report that deletion of transforming growth factor β signaling in the retinal microenvironment of newborn mice induces changes that largely mimic the phenotype of nonproliferative and proliferative diabetic retinopathy in humans. Lack of transforming growth factor β signaling leads to the formation of abundant microaneurysms, leaky capillaries, and retinal hemorrhages. Retinal capillaries are not covered by differentiated pericytes, but by a coat of vascular smooth muscle-like cells and a thickened basal lamina. Reactive microglia is found in close association with retinal capillaries. In older animals, loss of endothelial cells and the formation of ghost vessels are observed, findings that correlate with the induction of angiogenic molecules and the accumulation of retinal hypoxia-inducible factor 1α, indicating hypoxia. Consequently, retinal and vitreal neovascularization occurs, a scenario that leads to retinal detachment, vitreal hemorrhages, neuronal apoptosis, and impairment of sensory function. We conclude that transforming growth factor β signaling is required for the differentiation of retinal pericytes during vascular development of the retina. Lack of differentiated pericytes initiates a scenario of structural and functional changes in the retina that mimics those of diabetic retinopathy strongly indicating a common mechanism.
Collapse
Affiliation(s)
- Barbara M Braunger
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Sarah V Leimbeck
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Anja Schlecht
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Cornelia Volz
- Department of Ophthalmology, University of Regensburg, Regensburg, Germany
| | - Herbert Jägle
- Department of Ophthalmology, University of Regensburg, Regensburg, Germany
| | - Ernst R Tamm
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
205
|
van Dijk CGM, Nieuweboer FE, Pei JY, Xu YJ, Burgisser P, van Mulligen E, el Azzouzi H, Duncker DJ, Verhaar MC, Cheng C. The complex mural cell: pericyte function in health and disease. Int J Cardiol 2015; 190:75-89. [PMID: 25918055 DOI: 10.1016/j.ijcard.2015.03.258] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 03/16/2015] [Accepted: 03/18/2015] [Indexed: 02/06/2023]
Abstract
Pericytes are perivascular cells that can be distinguished from vascular smooth muscle cells by their specific morphology and expression of distinct molecular markers. Found in the microvascular beds distributed throughout the body, they are well known for their regulation of a healthy vasculature. In this review, we examine the mechanism of pericyte support to vasomotion, and the known pathways that regulate pericyte response in angiogenesis and neovascular stabilization. We will also discuss the role of pericytes in vascular basement membrane and endothelial barrier function regulation. In contrast, recent findings have indicated that pericyte dysfunction, characterized by changes in pericyte contractility or pericyte loss of microvascular coverage, plays an important role in onset and progression of vascular-related and fibrogenic diseases. From a therapeutic point of view, pericytes have recently been identified as a putative pool of endogenous mesenchymal stem cells that could be activated in response to tissue injury to contribute to the regenerative process on multiple levels. We will discuss the mechanisms via which pericytes are involved in disease onset and development in a number of pathophysiological conditions, as well as present the evidence that supports a role for multipotent pericytes in tissue regeneration. The emerging field of pericyte research will not only contribute to the identification of new drug targets in pericyte dysfunction associated diseases, but may also boost the use of this cell type in future cell-based regenerative strategies.
Collapse
Affiliation(s)
- Christian G M van Dijk
- Laboratory of Renal and Vascular Biology, Department of Nephrology and Hypertension, University Medical Center Utrecht, The Netherlands
| | - Frederieke E Nieuweboer
- Laboratory of Renal and Vascular Biology, Department of Nephrology and Hypertension, University Medical Center Utrecht, The Netherlands
| | - Jia Yi Pei
- Laboratory of Renal and Vascular Biology, Department of Nephrology and Hypertension, University Medical Center Utrecht, The Netherlands
| | - Yan Juan Xu
- Laboratory of Renal and Vascular Biology, Department of Nephrology and Hypertension, University Medical Center Utrecht, The Netherlands
| | - Petra Burgisser
- Department of Cardiology, University Medical Center Utrecht, The Netherlands
| | - Elise van Mulligen
- Laboratory of Renal and Vascular Biology, Department of Nephrology and Hypertension, University Medical Center Utrecht, The Netherlands
| | - Hamid el Azzouzi
- Department of Cardiology, University Medical Center Utrecht, The Netherlands
| | - Dirk J Duncker
- Department of Cardiology, Cardiovascular Research School COEUR, Erasmus University Medical Center Rotterdam, The Netherlands
| | - Marianne C Verhaar
- Laboratory of Renal and Vascular Biology, Department of Nephrology and Hypertension, University Medical Center Utrecht, The Netherlands
| | - Caroline Cheng
- Laboratory of Renal and Vascular Biology, Department of Nephrology and Hypertension, University Medical Center Utrecht, The Netherlands; Department of Cardiology, Cardiovascular Research School COEUR, Erasmus University Medical Center Rotterdam, The Netherlands.
| |
Collapse
|
206
|
Kostallari E, Baba-Amer Y, Alonso-Martin S, Ngoh P, Relaix F, Lafuste P, Gherardi RK. Pericytes in the myovascular niche promote post-natal myofiber growth and satellite cell quiescence. Development 2015; 142:1242-53. [PMID: 25742797 DOI: 10.1242/dev.115386] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The satellite cells, which serve as adult muscle stem cells, are both located beneath myofiber basement membranes and closely associated with capillary endothelial cells. We observed that 90% of capillaries were associated with pericytes in adult mouse and human muscle. During post-natal growth, newly formed vessels with their neuroglial 2 proteoglycan (NG2)-positive pericytes became progressively associated with the post-natal muscle stem cells, as myofibers increased in size and satellite cells entered into quiescence. In vitro, human muscle-derived pericytes promoted myogenic cell differentiation through insulin-like growth factor 1 (IGF1) and myogenic cell quiescence through angiopoietin 1 (ANGPT1). Diphtheria toxin-induced ablation of muscle pericytes in growing mice led both to myofiber hypotrophy and to impaired establishment of stem cells quiescence. Similar effects were observed following conditional in vivo deletion of pericyte Igf1 and Angpt1 genes, respectively. Our data therefore demonstrate that, by promoting post-natal myogenesis and stem cell quiescence, pericytes play a key role in the microvascular niche of satellite cells.
Collapse
Affiliation(s)
- Enis Kostallari
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Faculté de Médecine, 8 rue du Général Sarrail, Créteil F-94010, France Université Paris-Est, 5 boulevard Descartes, Marne-la-Vallée cedex 2 F-77454, France
| | - Yasmine Baba-Amer
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Faculté de Médecine, 8 rue du Général Sarrail, Créteil F-94010, France Université Paris-Est Créteil, 62 avenue du Général de Gaulle, Créteil F-94000, France
| | - Sonia Alonso-Martin
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Faculté de Médecine, 8 rue du Général Sarrail, Créteil F-94010, France Université Paris-Est Créteil, 62 avenue du Général de Gaulle, Créteil F-94000, France
| | - Pamela Ngoh
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Faculté de Médecine, 8 rue du Général Sarrail, Créteil F-94010, France Université d'Evry-Val d'Essonne, Boulevard François Mitterrand, Evry F-91000, France
| | - Frederic Relaix
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Faculté de Médecine, 8 rue du Général Sarrail, Créteil F-94010, France Université Paris-Est Créteil, 62 avenue du Général de Gaulle, Créteil F-94000, France Etablissement Français du Sang, Créteil 94017, France Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort 94700, France Hôpital Henri Mondor, Département de Pathologie, 51 avenue du Maréchal de Lattre de Tassigny, Créteil F-94010, France
| | - Peggy Lafuste
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Faculté de Médecine, 8 rue du Général Sarrail, Créteil F-94010, France Université Paris-Est Créteil, 62 avenue du Général de Gaulle, Créteil F-94000, France
| | - Romain K Gherardi
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Faculté de Médecine, 8 rue du Général Sarrail, Créteil F-94010, France Université Paris-Est Créteil, 62 avenue du Général de Gaulle, Créteil F-94000, France Hôpital Henri Mondor, Département de Pathologie, 51 avenue du Maréchal de Lattre de Tassigny, Créteil F-94010, France
| |
Collapse
|
207
|
Canonical Wnt signalling in PDGFRα-expressing cells is a critical regulator of astrogliosis and axon regeneration following CNS injury. J Neurosci 2015; 34:16163-5. [PMID: 25471558 DOI: 10.1523/jneurosci.4052-14.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
208
|
Lai AY, Dorr A, Thomason LAM, Koletar MM, Sled JG, Stefanovic B, McLaurin J. Venular degeneration leads to vascular dysfunction in a transgenic model of Alzheimer's disease. ACTA ACUST UNITED AC 2015; 138:1046-58. [PMID: 25688079 DOI: 10.1093/brain/awv023] [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: 11/12/2022]
Abstract
Most patients with Alzheimer's disease exhibit accumulation of amyloid-β peptide on leptomeningeal and cortical arterioles, or cerebral amyloid angiopathy, which is associated with impaired vascular reactivity and accelerated cognitive decline. Despite widespread recognition of the significance of vascular dysfunction in Alzheimer's disease aetiology and progression, much uncertainty still surrounds the mechanism underlying Alzheimer's disease vascular injury. Studies to date have focused on amyloid-β-induced damage to capillaries and plaque-associated arterioles, without examining effects across the entire vascular bed. In the present study, we investigated the structural and functional impairment of the feeding arteriolar versus draining venular vessels in a transgenic murine Alzheimer's disease model, with a particular focus on the mural cell populations that dictate these vessels' contractility. Although amyloid-β deposition was restricted to arterioles, we found that vascular impairment extended to the venules, which showed significant depletion of their mural cell coverage by the mid-stage of Alzheimer's disease pathophysiology. These structural abnormalities were accompanied by an abolishment of the normal vascular network flow response to hypercapnia: this functional impairment was so severe as to result in hypercapnia-induced flow decreases in the arterioles. Further pharmacological depletion of mural cells using SU6668, a platelet-derived growth factor receptor-β antagonist, resulted in profound structural abnormalities of the cortical microvasculature, including vessel coiling and short-range looping, increased tortuosity of the venules but not of the arterioles, increased amyloid-β deposition on the arterioles, and further alterations of the microvascular network cerebral blood flow response to hypercapnia. Together, this work shows hitherto unrecognized structural alterations in penetrating venules, demonstrates their functional significance and sheds light on the complexity of the relationship between vascular network structure and function in Alzheimer's disease.
Collapse
Affiliation(s)
- Aaron Y Lai
- 1 Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada 2 Sunnybrook Research Institute, Toronto, ON, Canada
| | - Adrienne Dorr
- 2 Sunnybrook Research Institute, Toronto, ON, Canada
| | | | | | - John G Sled
- 3 Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada 4 Hospital for Sick Children, Toronto, ON, Canada
| | - Bojana Stefanovic
- 2 Sunnybrook Research Institute, Toronto, ON, Canada 3 Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - JoAnne McLaurin
- 1 Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada 2 Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
209
|
Kelly-Goss MR, Sweat RS, Stapor PC, Peirce SM, Murfee WL. Targeting pericytes for angiogenic therapies. Microcirculation 2015; 21:345-57. [PMID: 24267154 DOI: 10.1111/micc.12107] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/19/2013] [Indexed: 12/18/2022]
Abstract
In pathological scenarios, such as tumor growth and diabetic retinopathy, blocking angiogenesis would be beneficial. In others, such as myocardial infarction and hypertension, promoting angiogenesis might be desirable. Due to their putative influence on endothelial cells, vascular pericytes have become a topic of growing interest and are increasingly being evaluated as a potential target for angioregulatory therapies. The strategy of manipulating pericyte recruitment to capillaries could result in anti- or proangiogenic effects. Our current understanding of pericytes, however, is limited by knowledge gaps regarding pericyte identity and lineage. To use a music analogy, this review is a "mash-up" that attempts to integrate what we know about pericyte functionality and expression with what is beginning to be elucidated regarding their regenerative potential. We explore the lingering questions regarding pericyte phenotypic identity and lineage. The expression of different pericyte markers (e.g., SMA, Desmin, NG2, and PDGFR-β) varies for different subpopulations and tissues. Previous use of these markers to identify pericytes has suggested potential phenotypic overlaps and plasticity toward other cell phenotypes. Our review chronicles the state of the literature, identifies critical unanswered questions, and motivates future research aimed at understanding this intriguing cell type and harnessing its therapeutic potential.
Collapse
Affiliation(s)
- Molly R Kelly-Goss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | | | | | | | | |
Collapse
|
210
|
Arboleda-Velasquez JF, Valdez CN, Marko CK, D'Amore PA. From pathobiology to the targeting of pericytes for the treatment of diabetic retinopathy. Curr Diab Rep 2015; 15:573. [PMID: 25620405 PMCID: PMC5599150 DOI: 10.1007/s11892-014-0573-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pericytes, the mural cells that constitute the capillaries along with endothelial cells, have been associated with the pathobiology of diabetic retinopathy; however, therapeutic implications of this association remain largely unexplored. Pericytes appear to be highly susceptible to the metabolic challenges associated with a diabetic environment, and there is substantial evidence that their loss may contribute to microvascular instability leading to the formation of microaneurysms, microhemorrhages, acellular capillaries, and capillary nonperfusion. Since pericytes are strategically located at the interface between the vascular and neural components of the retina, they offer extraordinary opportunities for therapeutic interventions in diabetic retinopathy. Moreover, the availability of novel imaging methodologies now allows for the in vivo visualization of pericytes, enabling a new generation of clinical trials that use pericyte tracking as clinical endpoints. The recognition of multiple signaling mechanisms involved in pericyte development and survival should allow for a renewed interest in pericytes as a therapeutic target for diabetic retinopathy.
Collapse
Affiliation(s)
- Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute/Massachusetts Eye and Ear and the Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | | | | | | |
Collapse
|
211
|
Pigment epithelium-derived factor enhances tumor response to radiation through vasculature normalization in allografted lung cancer in mice. Cancer Gene Ther 2015; 22:181-7. [PMID: 25591809 DOI: 10.1038/cgt.2014.79] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/11/2014] [Accepted: 12/13/2014] [Indexed: 12/30/2022]
Abstract
This study aimed to explore the potential therapeutic effects of the combination of pigment epithelium-derived factor (PEDF) and radiation on lung cancer. The Lewis lung cancer (LLC) allografts in nude mice were treated with radiation, PEDF and PEDF combined with radiation. The morphologic changes of tumor vasculature and the hypoxic fraction of tumor tissues were evaluated. Significant inhibition of tumor growth was observed when radiation was applied between the 3rd and 7th day (the vasculature normalization window) after the initiation of PEDF treatment. During the vasculature normalization window, the tumor blood vessels in PEDF-treated mice were less tortuous and more uniform than those in the LLC allograft tumor treated with phosphate-buffered saline. Meanwhile, the thickness of the basement membrane was remarkably reduced and pericyte coverage was significantly increased with the PEDF treatment. We also found that tumor hypoxic fraction decreased during the 3rd to the 7th day after PEDF treatment, suggesting improved intratumoral oxygenation. Taken together, our results show that PEDF improved the effects of radiation therapy on LLC allografts by inducing a vascular normalization window from the 3rd to the 7th day after PEDF treatment. Our findings provide a basis for treating lung cancer with the combination of PEDF and radiation.
Collapse
|
212
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Pericytes at the intersection between tissue regeneration and pathology. Clin Sci (Lond) 2015; 128:81-93. [PMID: 25236972 PMCID: PMC4200531 DOI: 10.1042/cs20140278] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Perivascular multipotent cells, pericytes, contribute to the generation and repair of various tissues in response to injury. They are heterogeneous in their morphology, distribution, origin and markers, and elucidating their molecular and cellular differences may inform novel treatments for disorders in which tissue regeneration is either impaired or excessive. Moreover, these discoveries offer novel cellular targets for therapeutic approaches to many diseases. This review discusses recent studies that support the concept that pericyte subtypes play a distinctive role in myogenesis, neurogenesis, adipogenesis, fibrogenesis and angiogenesis.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| |
Collapse
|
213
|
Stegen S, van Gastel N, Carmeliet G. Bringing new life to damaged bone: the importance of angiogenesis in bone repair and regeneration. Bone 2015; 70:19-27. [PMID: 25263520 DOI: 10.1016/j.bone.2014.09.017] [Citation(s) in RCA: 306] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 09/14/2014] [Accepted: 09/18/2014] [Indexed: 12/26/2022]
Abstract
Bone has the unique capacity to heal without the formation of a fibrous scar, likely because several of the cellular and molecular processes governing bone healing recapitulate the events during skeletal development. A critical component in bone healing is the timely appearance of blood vessels in the fracture callus. Angiogenesis, the formation of new blood vessels from pre-existing ones, is stimulated after fracture by the local production of numerous angiogenic growth factors. The fracture vasculature not only supplies oxygen and nutrients, but also stem cells able to differentiate into osteoblasts and in a later phase also the ions necessary for mineralization. This review provides a concise report of the regulation of angiogenesis by bone cells, its importance during bone healing and its possible therapeutic applications in bone tissue engineering. This article is part of a Special Issue entitled "Stem Cells and Bone".
Collapse
Affiliation(s)
- Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Nick van Gastel
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
214
|
Stanton GB, Kohler SJ, Boklweski J, Cameron JL, Greenough WT. Cytogenesis in the adult monkey motor cortex: perivascular NG2 cells are the major adult born cell type. J Comp Neurol 2014; 523:849-68. [PMID: 25308320 PMCID: PMC4354441 DOI: 10.1002/cne.23693] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 10/08/2014] [Accepted: 10/08/2014] [Indexed: 12/11/2022]
Abstract
We used confocal microscopy and immunohistochemistry (IHC) to look for new cells in the motor cortex of adult macaque monkeys that might form the cellular bases of improved brain function from exercise. Twenty-four female Macaca fascicularis monkeys divided into groups by age (10–12 years, 15–17 years), postexercise survival periods, and controls, received 10 weekly injections of the thymidine analog, bromodeoxyuridine (BrdU) to mark new cells. Sixteen monkeys survived 15 weeks (5 weeks postexercise) and 8 monkeys survived 27 weeks (12 weeks postexercise) after initial BrdU injections. Additionally, five Macaca mulatta female monkeys (∼5.5–7 years) received single injections of BrdU and survived 2 days, 2 weeks, and 6 weeks after BrdU injections. Neural and glial antibodies were used to identify new cell phenotypes and to look for changes in proportions of these cells with respect to time and experimental conditions. No BrdU+/DCx+ cells were found but about 7.5% of new cells were calretinin-positive (Cr+). BrdU+/GABA+ (gamma-aminobutyric acid) cells were also found but no new Cr+ or GABA+ cells colabeled with a mature neuron marker, NeuN or chondroitin sulfate antibody, NG2. The proportion of new cells that were NG2+ was about 85% for short and long survival monkeys of which two, newly described perivascular phenotypes (Pldv and Elu) and a small percentage of pericytes (2.5%) comprised 44% and 51% of the new NG2+ cells, respectively. Proportions of NG2+ phenotypes were affected by post-BrdU survival periods, monkey age, and possibly a postexercise sedentary period but no direct effect of exercise was found.
Collapse
|
215
|
Trost A, Schroedl F, Marschallinger J, Rivera FJ, Bogner B, Runge C, Couillard-Despres S, Aigner L, Reitsamer HA. Characterization of dsRed2-positive cells in the doublecortin-dsRed2 transgenic adult rat retina. Histochem Cell Biol 2014; 142:601-17. [PMID: 25138677 DOI: 10.1007/s00418-014-1259-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2014] [Indexed: 10/24/2022]
Abstract
Doublecortin (DCX) is predominantly expressed in neuronal precursor cells and young immature neurons of the developing and adult brain, where it is involved in neuronal differentiation, migration and plasticity. Moreover, its expression pattern reflects neurogenesis, and transgenic DCX promoter-driven reporter models have been previously used to investigate adult neurogenesis. In this study, we characterize dsRed2 reporter protein-expressing cells in the adult retina of the transgenic DCX promoter-dsRed2 rat model, with the aim to identify cells with putative neurogenic activity. Additionally, we confirmed the expression of the dsRed2 protein in DCX-expressing cells in the adult hippocampal dentate gyrus. Adult DCX-dsRed2 rat retinas were analyzed by immunohistochemistry for expression of DCX, NF200, Brn3a, Sox2, NeuN, calbindin, calretinin, PKC-a, Otx2, ChAT, PSA-NCAM and the glial markers GFAP and CRALBP, followed by confocal laser-scanning microscopy. In addition, brain sections of transgenic rats were analyzed for dsRed2 expression and co-localization with DCX, NeuN, GFAP and Sox2 in the cortex and dentate gyrus. Endogenous DCX expression in the adult retina was confined to horizontal cells, and these cells co-expressed the DCX promoter-driven dsRed2 reporter protein. In addition, we encountered dsRed2 expression in various other cell types in the retina: retinal ganglion cells (RGCs), a subpopulation of amacrine cells, a minority of bipolar cells and in perivascular cells. Since also RGCs expressed dsRed2, the DCX-dsRed2 rat model might offer a useful tool to study RGCs in vivo under various conditions. Müller glial cells, which have previously been identified as cells with stem cell features and with neurogenic potential, did express neither endogenous DCX nor the dsRed2 reporter. However, and surprisingly, we identified a perivascular glial cell type expressing the dsRed2 reporter, enmeshed with the glia/stem cell marker GFAP and colocalizing with the neural stem cell marker Sox2. These findings suggest the so far undiscovered existence of perivascular associated cell with neural stem cell-like properties in the adult retina.
Collapse
Affiliation(s)
- A Trost
- Ophthalmology/Optometry, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020, Salzburg, Austria,
| | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Characterization of Nestin-positive stem Leydig cells as a potential source for the treatment of testicular Leydig cell dysfunction. Cell Res 2014; 24:1466-85. [PMID: 25418539 PMCID: PMC4260348 DOI: 10.1038/cr.2014.149] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 01/19/2014] [Accepted: 09/01/2014] [Indexed: 11/23/2022] Open
Abstract
The ability to identify and isolate lineage-specific stem cells from adult tissues could facilitate cell replacement therapy. Leydig cells (LCs) are the primary source of androgen in the mammalian testis, and the prospective identification of stem Leydig cells (SLCs) may offer new opportunities for treating testosterone deficiency. Here, in a transgenic mouse model expressing GFP driven by the Nestin (Nes) promoter, we observed Nes-GFP+ cells located in the testicular interstitial compartment where SLCs normally reside. We showed that these Nes-GFP+ cells expressed LIFR and PDGFR-α, but not LC lineage markers. We further observed that these cells were capable of clonogenic self-renewal and extensive proliferation in vitro and could differentiate into neural or mesenchymal cell lineages, as well as LCs, with the ability to produce testosterone, under defined conditions. Moreover, when transplanted into the testes of LC-disrupted or aging models, the Nes-GFP+ cells colonized the interstitium and partially increased testosterone production, and then accelerated meiotic and post-meiotic germ cell recovery. In addition, we further demonstrated that CD51 might be a putative cell surface marker for SLCs, similar with Nestin. Taken together, these results suggest that Nes-GFP+ cells from the testis have the characteristics of SLCs, and our study would shed new light on developing stem cell replacement therapy for testosterone deficiency.
Collapse
|
217
|
Birbrair A, Zhang T, Files DC, Mannava S, Smith T, Wang ZM, Messi ML, Mintz A, Delbono O. Type-1 pericytes accumulate after tissue injury and produce collagen in an organ-dependent manner. Stem Cell Res Ther 2014; 5:122. [PMID: 25376879 PMCID: PMC4445991 DOI: 10.1186/scrt512] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 09/30/2014] [Indexed: 02/06/2023] Open
Abstract
Introduction Fibrosis, or scar formation, is a pathological condition characterized by excessive production and accumulation of collagen, loss of tissue architecture, and organ failure in response to uncontrolled wound healing. Several cellular populations have been implicated, including bone marrow-derived circulating fibrocytes, endothelial cells, resident fibroblasts, epithelial cells, and recently, perivascular cells called pericytes. We previously demonstrated pericyte functional heterogeneity in skeletal muscle. Whether pericyte subtypes are present in other tissues and whether a specific pericyte subset contributes to organ fibrosis are unknown. Methods Here, we report the presence of two pericyte subtypes, type-1 (Nestin-GFP-/NG2-DsRed+) and type-2 (Nestin-GFP+/NG2-DsRed+), surrounding blood vessels in lungs, kidneys, heart, spinal cord, and brain. Using Nestin-GFP/NG2-DsRed transgenic mice, we induced pulmonary, renal, cardiac, spinal cord, and cortical injuries to investigate the contributions of pericyte subtypes to fibrous tissue formation in vivo. Results A fraction of the lung’s collagen-producing cells corresponds to type-1 pericytes and kidney and heart pericytes do not produce collagen in pathological fibrosis. Note that type-1, but not type-2, pericytes increase and accumulate near the fibrotic tissue in all organs analyzed. Surprisingly, after CNS injury, type-1 pericytes differ from scar-forming PDGFRβ + cells. Conclusions Pericyte subpopulations respond differentially to tissue injury, and the production of collagen by type-1 pericytes is organ-dependent. Characterization of the mechanisms underlying scar formation generates cellular targets for future anti-fibrotic therapeutics. Electronic supplementary material The online version of this article (doi:10.1186/scrt512) contains supplementary material, which is available to authorized users.
Collapse
|
218
|
Collins JJP, Thébaud B. Lung mesenchymal stromal cells in development and disease: to serve and protect? Antioxid Redox Signal 2014; 21:1849-62. [PMID: 24350665 DOI: 10.1089/ars.2013.5781] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
SIGNIFICANCE Bronchopulmonary dysplasia (BPD) is a disease of the developing lung that afflicts extreme preterm infants in the neonatal intensive care unit. Follow-up studies into adulthood show that BPD is not merely a problem of the neonatal period, as it also may predispose to early-onset emphysema and poor lung function in later life. RECENT ADVANCES The increasing promise of bone marrow- or umbilical cord-derived mesenchymal stromal cells (MSCs) to repair neonatal and adult lung diseases may for the first time offer the chance to make substantial strides in improving the outcome of extreme premature infants at risk of developing BPD. As more knowledge has been obtained on MSCs over the past decades, it has become clear that each organ has its own reservoir of endogenous MSCs, including the lung. CRITICAL ISSUES We have only barely scratched the surface on what resident lung MSCs exactly are and what their role and function in lung development may be. Moreover, what happens to these putative repair cells in BPD when alveolar development goes awry and why do their counterparts from the bone marrow and umbilical cord succeed in restoring normal alveolar development when they themselves do not? FUTURE DIRECTIONS Much work remains to be carried out to validate lung MSCs, but with the high potential of MSC-based treatment for BPD and other lung diseases, a thorough understanding of the endogenous lung MSC will be pivotal to get to the bottom of these diseases.
Collapse
Affiliation(s)
- Jennifer J P Collins
- 1 Regenerative Medicine Program, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, University of Ottawa , Ottawa, Canada
| | | |
Collapse
|
219
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Pericytes: multitasking cells in the regeneration of injured, diseased, and aged skeletal muscle. Front Aging Neurosci 2014; 6:245. [PMID: 25278877 PMCID: PMC4166895 DOI: 10.3389/fnagi.2014.00245] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/29/2014] [Indexed: 12/16/2022] Open
Abstract
Pericytes are perivascular cells that envelop and make intimate connections with adjacent capillary endothelial cells. Recent studies show that they may have a profound impact in skeletal muscle regeneration, innervation, vessel formation, fibrosis, fat accumulation, and ectopic bone formation throughout life. In this review, we summarize and evaluate recent advances in our understanding of pericytes' influence on adult skeletal muscle pathophysiology. We also discuss how further elucidating their biology may offer new approaches to the treatment of conditions characterized by muscle wasting.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA ; Neuroscience Program, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Maria L Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA ; Neuroscience Program, Wake Forest School of Medicine Winston-Salem, NC, USA
| |
Collapse
|
220
|
Dimou L, Götz M. Glial cells as progenitors and stem cells: new roles in the healthy and diseased brain. Physiol Rev 2014; 94:709-37. [PMID: 24987003 DOI: 10.1152/physrev.00036.2013] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The diverse functions of glial cells prompt the question to which extent specific subtypes may be devoted to a specific function. We discuss this by reviewing one of the most recently discovered roles of glial cells, their function as neural stem cells (NSCs) and progenitor cells. First we give an overview of glial stem and progenitor cells during development; these are the radial glial cells that act as NSCs and other glial progenitors, highlighting the distinction between the lineage of cells in vivo and their potential when exposed to a different environment, e.g., in vitro. We then proceed to the adult stage and discuss the glial cells that continue to act as NSCs across vertebrates and others that are more lineage-restricted, such as the adult NG2-glia, the most frequent progenitor type in the adult mammalian brain, that remain within the oligodendrocyte lineage. Upon certain injury conditions, a distinct subset of quiescent astrocytes reactivates proliferation and a larger potential, clearly demonstrating the concept of heterogeneity with distinct subtypes of, e.g., astrocytes or NG2-glia performing rather different roles after brain injury. These new insights not only highlight the importance of glial cells for brain repair but also their great potential in various aspects of regeneration.
Collapse
Affiliation(s)
- Leda Dimou
- Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University, Munich, Germany; Institute for Stem Cell Research, HelmholtzZentrum, Neuherberg, Germany; and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Magdalena Götz
- Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University, Munich, Germany; Institute for Stem Cell Research, HelmholtzZentrum, Neuherberg, Germany; and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
221
|
Özen I, Deierborg T, Miharada K, Padel T, Englund E, Genové G, Paul G. Brain pericytes acquire a microglial phenotype after stroke. Acta Neuropathol 2014; 128:381-96. [PMID: 24848101 PMCID: PMC4131168 DOI: 10.1007/s00401-014-1295-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/14/2014] [Accepted: 05/14/2014] [Indexed: 11/28/2022]
Abstract
Pericytes are located on the abluminal side of endothelial cells lining the microvasculature in all organs. They have been identified as multipotent progenitor cells in several tissues of the body including the human brain. New evidence suggests that pericytes contribute to tissue repair, but their role in the injured brain is largely unknown. Here, we investigate the role of pericytes in ischemic stroke. Using a pericyte-reporter mouse model, we provide unique evidence that regulator of G-protein signaling 5 expressing cells are activated pericytes that leave the blood vessel wall, proliferate and give rise to microglial cells after ischemic brain injury. Consistently, we show that activated pericytes express microglial markers in human stroke brain tissue. We demonstrate that human brain-derived pericytes adopt a microglial phenotype and upregulate mRNA specific for activated microglial cells under hypoxic conditions in vitro. Our study indicates that the vasculature is a novel source of inflammatory cells with a microglial phenotype in brain ischemia and hence identifies pericytes as an important new target for the development of future stroke therapies.
Collapse
Affiliation(s)
- Ilknur Özen
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, BMC, Lund University, 22184 Lund, Sweden
| | - Kenichi Miharada
- Department of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology and Cell Therapy, BMC, Lund University, 22184 Lund, Sweden
| | - Thomas Padel
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Elisabet Englund
- Department of Neuropathology, Scania University Hospital, 22185 Lund, Sweden
| | - Guillem Genové
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
- Department of Neurology, Scania University Hospital, 22185 Lund, Sweden
| |
Collapse
|
222
|
Ding L, Cheng R, Hu Y, Takahashi Y, Jenkins AJ, Keech AC, Humphries KM, Gu X, Elliott MH, Xia X, Ma JX. Peroxisome proliferator-activated receptor α protects capillary pericytes in the retina. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2709-20. [PMID: 25108226 DOI: 10.1016/j.ajpath.2014.06.021] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 05/15/2014] [Accepted: 06/05/2014] [Indexed: 12/25/2022]
Abstract
Pericyte degeneration is an early event in diabetic retinopathy and plays an important role in progression of diabetic retinopathy. Clinical studies have shown that fenofibrate, a peroxisome proliferator-activated receptor α (PPARα) agonist, has robust therapeutic effects on diabetic retinopathy in type 2 diabetic patients. We evaluated the protective effect of PPARα against pericyte loss in diabetic retinopathy. In streptozotocin-induced diabetic mice, fenofibrate treatment significantly ameliorated retinal acellular capillary formation and pericyte loss. In contrast, PPARα(-/-) mice with diabetes developed more severe retinal acellular capillary formation and pericyte dropout, compared with diabetic wild-type mice. Furthermore, PPARα knockout abolished the protective effect of fenofibrate against diabetes-induced retinal pericyte loss. In cultured primary human retinal capillary pericytes, activation and expression of PPARα both significantly reduced oxidative stress-induced apoptosis, decreased reactive oxygen species production, and down-regulated NAD(P)H oxidase 4 expression through blockade of NF-κB activation. Furthermore, activation and expression of PPARα both attenuated the oxidant-induced suppression of mitochondrial O2 consumption in human retinal capillary pericytes. Primary retinal pericytes from PPARα(-/-) mice displayed more apoptosis, compared with those from wild-type mice under the same oxidative stress. These findings identified a protective effect of PPARα on retinal pericytes, a novel function of endogenous PPARα in the retina.
Collapse
Affiliation(s)
- Lexi Ding
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China; Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Rui Cheng
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Yang Hu
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Yusuke Takahashi
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Alicia J Jenkins
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Anthony C Keech
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Kenneth M Humphries
- Free Radical Biology and Aging Research Program, Medical Research Foundation, Oklahoma City, Oklahoma
| | - Xiaowu Gu
- Department of Ophthalmology, Dean McGee Eye Institute, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Michael H Elliott
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Ophthalmology, Dean McGee Eye Institute, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Xiaobo Xia
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China.
| | - Jian-Xing Ma
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| |
Collapse
|
223
|
Involvement of matrix metalloproteinase-9 in amyloid-β 1-42-induced shedding of the pericyte proteoglycan NG2. J Neuropathol Exp Neurol 2014; 73:684-92. [PMID: 24918635 PMCID: PMC4072439 DOI: 10.1097/nen.0000000000000084] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Supplemental digital content is available in the text. Deposition of amyloid-β (Aβ) 1–42, the major component of senile plaques characteristic of Alzheimer disease, affects brain microvascular integrity and causes blood-brain barrier dysfunction, increased angiogenesis, and pericyte degeneration. To understand the cellular events underlying Aβ1–42 effects on microvascular alterations, we investigated whether different aggregation forms of Aβ1–42 affect shedding of the pericyte proteoglycan NG2 and whether they affect proteolytic cleavage mediated by matrix metalloproteinase (MMP)-9. We found decreased levels of soluble NG2, total MMP-9, and MMP-9 activity in pericyte culture supernatants in response to fibril-enriched preparations of Aβ1–42. Conversely, oligomer-enriched preparations of Aβ1–42 increased soluble NG2 levels in the supernatants. This increase was ablated by the MMP-9/MMP-2 inhibitor SB-3CT. There was also a trend toward increased MMP-9 activity observed after oligomeric Aβ1–42 exposure. Our results, demonstrating an Aβ1–42 aggregation-dependent effect on levels of NG2 and MMP-9, support previous studies showing an impact of Aβ1–42 on vascular integrity and thereby add to our understanding of mechanisms behind the microvascular changes commonly found in patients with Alzheimer disease.
Collapse
|
224
|
Liu G, Meng C, Pan M, Chen M, Deng R, Lin L, Zhao L, Liu X. Isolation, Purification, and Cultivation of Primary Retinal Microvascular Pericytes: A Novel Model Using Rats. Microcirculation 2014; 21:478-89. [PMID: 24495210 DOI: 10.1111/micc.12121] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 01/30/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Guanghui Liu
- School of Optometry and Ophthalmology and Eye Hospital; Wenzhou Medical University; Wenzhou China
- Department of Ophthalmology; Affiliated People's Hospital (People's Hospital of Fujian Province); Fujian University of Traditional Chinese Medicine; Fuzhou China
| | - Chun Meng
- Department of Bioengineering; College of Biological Science and Biotechnology; Fuzhou University; Fuzhou China
| | - Mingdong Pan
- Department of Ophthalmology; Affiliated People's Hospital (People's Hospital of Fujian Province); Fujian University of Traditional Chinese Medicine; Fuzhou China
| | - Meng Chen
- Department of Ophthalmology; Baylor College of Medicine; Houston Texas USA
| | - Ruzhi Deng
- School of Optometry and Ophthalmology and Eye Hospital; Wenzhou Medical University; Wenzhou China
| | - Ling Lin
- Department of Bioengineering; College of Biological Science and Biotechnology; Fuzhou University; Fuzhou China
| | - Li Zhao
- Department of Cardiology; Affiliated People's Hospital (People's Hospital of Fujian Province); Fujian University of Traditional Chinese Medicine; Fuzhou China
| | - Xiaoling Liu
- School of Optometry and Ophthalmology and Eye Hospital; Wenzhou Medical University; Wenzhou China
| |
Collapse
|
225
|
Moore-Morris T, Guimarães-Camboa N, Banerjee I, Zambon AC, Kisseleva T, Velayoudon A, Stallcup WB, Gu Y, Dalton ND, Cedenilla M, Gomez-Amaro R, Zhou B, Brenner DA, Peterson KL, Chen J, Evans SM. Resident fibroblast lineages mediate pressure overload-induced cardiac fibrosis. J Clin Invest 2014; 124:2921-34. [PMID: 24937432 DOI: 10.1172/jci74783] [Citation(s) in RCA: 494] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 04/24/2014] [Indexed: 01/22/2023] Open
Abstract
Activation and accumulation of cardiac fibroblasts, which result in excessive extracellular matrix deposition and consequent mechanical stiffness, myocyte uncoupling, and ischemia, are key contributors to heart failure progression. Recently, endothelial-to-mesenchymal transition (EndoMT) and the recruitment of circulating hematopoietic progenitors to the heart have been reported to generate substantial numbers of cardiac fibroblasts in response to pressure overload-induced injury; therefore, these processes are widely considered to be promising therapeutic targets. Here, using multiple independent murine Cre lines and a collagen1a1-GFP fusion reporter, which specifically labels fibroblasts, we found that following pressure overload, fibroblasts were not derived from hematopoietic cells, EndoMT, or epicardial epithelial-to-mesenchymal transition. Instead, pressure overload promoted comparable proliferation and activation of two resident fibroblast lineages, including a previously described epicardial population and a population of endothelial origin. Together, these data present a paradigm for the origins of cardiac fibroblasts during development and in fibrosis. Furthermore, these data indicate that therapeutic strategies for reducing pathogenic cardiac fibroblasts should shift from targeting presumptive EndoMT or infiltrating hematopoietically derived fibroblasts, toward common pathways upregulated in two endogenous fibroblast populations.
Collapse
|
226
|
Youn SW, Jung KH, Chu K, Lee JY, Lee ST, Bahn JJ, Park DK, Yu JS, Kim SY, Kim M, Lee SK, Han MH, Roh JK. Feasibility and Safety of Intra-arterial Pericyte Progenitor Cell Delivery Following Mannitol-Induced Transient Blood-Brain Barrier Opening in a Canine Model. Cell Transplant 2014; 24:1469-79. [PMID: 24932854 DOI: 10.3727/096368914x682413] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Stem cell therapy is currently being studied with a view to rescuing various neurological diseases. Such studies require not only the discovery of potent candidate cells but also the development of methods that allow optimal delivery of those candidates to the brain tissues. Given that the blood-brain barrier (BBB) precludes cells from entering the brain, the present study was designed to test whether hyperosmolar mannitol securely opens the BBB and enhances intra-arterial cell delivery. A noninjured normal canine model in which the BBB was presumed to be closed was used to evaluate the feasibility and safety of the tested protocol. Autologous adipose tissue-derived pericytes with platelet-derived growth factor receptor β positivity were utilized. Cells were administered 5 min after mannitol pretreatment using one of following techniques: (1) bolus injection of a concentrated suspension, (2) continuous infusion of a diluted suspension, or (3) bolus injection of a concentrated suspension that had been shaken by repeated syringe pumping. Animals administered a concentrated cell suspension without mannitol pretreatment served as a control group. Vital signs, blood parameters, neurologic status, and major artery patency were kept stable throughout the experiment and the 1-month posttreatment period. Although ischemic lesions were noted on magnetic resonance imaging in several mongrel dogs with concentrated cell suspension, the injection technique using repeated syringe shaking could avert this complication. The cells were detected in both ipsilateral and contralateral cortices and were more frequent at the ipsilateral and frontal locations, whereas very few cells were observed anywhere in the brain when mannitol was not preinjected. These data suggest that intra-arterial cell infusion with mannitol pretreatment is a feasible and safe therapeutic approach in stable brain diseases such as chronic stroke.
Collapse
Affiliation(s)
- Sung Won Youn
- Department of Neuroradiology, Catholic University of Daegu Medical Center, School of Medicine, Catholic University of Daegu, Daegu, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Siroky BJ, Yin H, Dixon BP, Reichert RJ, Hellmann AR, Ramkumar T, Tsuchihashi Z, Bunni M, Dillon J, Bell PD, Sampson JR, Bissler JJ. Evidence for pericyte origin of TSC-associated renal angiomyolipomas and implications for angiotensin receptor inhibition therapy. Am J Physiol Renal Physiol 2014; 307:F560-70. [PMID: 24920756 DOI: 10.1152/ajprenal.00569.2013] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nearly all patients with tuberous sclerosis complex (TSC) develop renal angiomyolipomas, although the tumor cell of origin is unknown. We observed decreased renal angiomyolipoma development in patients with TSC2- polycystic kidney disease 1 deletion syndrome and hypertension that were treated from an early age with angiotensin-converting enzyme inhibitors or angiotensin receptor blockers compared with patients who did not receive this therapy. TSC-associated renal angiomyolipomas expressed ANG II type 1 receptors, platelet-derived growth factor receptor-β, desmin, α-smooth muscle actin, and VEGF receptor 2 but did not express the adipocyte marker S100 or the endothelial marker CD31. Sera of TSC patients exhibited increased vascular mural cell-secreted peptides, such as VEGF-A, VEGF-D, soluble VEGF receptor 2, and collagen type IV. These findings suggest that angiomyolipomas may arise from renal pericytes. ANG II treatment of angiomyolipoma cells in vitro resulted in an exaggerated intracellular Ca(2+) response and increased proliferation, which were blocked by the ANG II type 2 receptor antagonist valsartan. Blockade of ANG II signaling may have preventative therapeutic potential for angiomyolipomas.
Collapse
Affiliation(s)
- Brian J Siroky
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Hong Yin
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Bradley P Dixon
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ryan J Reichert
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Anna R Hellmann
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Institute of Medical Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | | | | | - Marlene Bunni
- Department of Medicine, Division of Nephrology, Medical University of South Carolina and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Joshua Dillon
- Department of Medicine, Division of Nephrology, Medical University of South Carolina and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - P Darwin Bell
- Department of Medicine, Division of Nephrology, Medical University of South Carolina and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Julian R Sampson
- Institute of Medical Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - John J Bissler
- Tuberous Sclerosis Complex Center of Excellence, Le Bonheur Children's Hospital, University of Tennessee College of Medicine, Memphis, Tennessee
| |
Collapse
|
228
|
Stapor PC, Sweat RS, Dashti DC, Betancourt AM, Murfee WL. Pericyte dynamics during angiogenesis: new insights from new identities. J Vasc Res 2014; 51:163-74. [PMID: 24853910 DOI: 10.1159/000362276] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 03/11/2014] [Indexed: 12/13/2022] Open
Abstract
Therapies aimed at manipulating the microcirculation require the ability to control angiogenesis, defined as the sprouting of new capillaries from existing vessels. Blocking angiogenesis would be beneficial in many pathologies (e.g. cancer, retinopathies and rheumatoid arthritis). In others (e.g. myocardial infarction, stroke and hypertension), promoting angiogenesis would be desirable. We know that vascular pericytes elongate around endothelial cells (ECs) and are functionally associated with regulating vessel stabilization, vessel diameter and EC proliferation. During angiogenesis, bidirectional pericyte-EC signaling is critical for capillary sprout formation. Observations of pericytes leading capillary sprouts also implicate their role in EC guidance. As such, pericytes have recently emerged as a therapeutic target to promote or inhibit angiogenesis. Advancing our basic understanding of pericytes and developing pericyte-related therapies are challenged, like in many other fields, by questions regarding cell identity. This review article discusses what we know about pericyte phenotypes and the opportunity to advance our understanding by defining the specific pericyte cell populations involved in capillary sprouting.
Collapse
Affiliation(s)
- Peter C Stapor
- Department of Biomedical Engineering, Tulane University, Lindy Boggs Center, New Orleans, La., USA
| | | | | | | | | |
Collapse
|
229
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Olson JD, Mintz A, Delbono O. Type-2 pericytes participate in normal and tumoral angiogenesis. Am J Physiol Cell Physiol 2014; 307:C25-38. [PMID: 24788248 DOI: 10.1152/ajpcell.00084.2014] [Citation(s) in RCA: 230] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tissue growth and function depend on vascularization, and vascular insufficiency or excess exacerbates many human diseases. Identification of the biological processes involved in angiogenesis will dictate strategies to modulate reduced or excessive vessel formation. We examine the essential role of pericytes. Their heterogeneous morphology, distribution, origins, and physiology have been described. Using double-transgenic Nestin-GFP/NG2-DsRed mice, we identified two pericyte subsets. We found that Nestin-GFP(-)/NG2-DsRed(+) (type-1) and Nestin-GFP(+)/NG2-DsRed(+) (type-2) pericytes attach to the walls of small and large blood vessels in vivo; in vitro, type-2, but not type-1, pericytes spark endothelial cells to form new vessels. Matrigel assay showed that only type-2 pericytes participate in normal angiogenesis. Moreover, when cancer cells were transplanted into Nestin-GFP/NG2-DsRed mice, type-1 pericytes did not penetrate the tumor, while type-2 pericytes were recruited during its angiogenesis. As inhibition of angiogenesis is a promising strategy in cancer therapy, type-2 pericytes may provide a cellular target susceptible to signaling and pharmacological manipulation in treating malignancy. This work also reports the potential of type-2 pericytes to improve blood perfusion in ischemic hindlimbs, indicating their potential for treating ischemic illnesses.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - John D Olson
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina; and
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina; and Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina;
| |
Collapse
|
230
|
Multicellular self-assembled spheroidal model of the blood brain barrier. Sci Rep 2014; 3:1500. [PMID: 23511305 PMCID: PMC3603320 DOI: 10.1038/srep01500] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 03/06/2013] [Indexed: 12/27/2022] Open
Abstract
The blood brain barrier (BBB) has evolved unique characteristics such as dense coverage of the endothelial cells by pericytes and interactions with astrocytes through perivascular endfeet. We study BBB formation in a 3-dimensional multicellular spheroid system of human primary brain endothelial cells (hpBECs), primary pericytes (hpPs) and primary astrocytes (hpAs). We show for the first time that hpBECs, hpPs and hpAs spontaneously self-organize into a defined multicellular structure which recapitulates the complex arrangement of the individual cell types in the BBB structure. Pericytes play a crucial role mediating the interaction between hpBECs and hpAs. This process is not dependent on a scaffold support demonstrating that formation and cellular architecture of the BBB is intrinsically programmed within each specific cell type. In a matrigel setup the hpBECs, hpPs and hpAs also undergo self-arrangement to form endothelial tube-like structures tightly covered by hpPs and loosely attached hpAs mainly at the junctions.
Collapse
|
231
|
Wanjare M, Kusuma S, Gerecht S. Defining differences among perivascular cells derived from human pluripotent stem cells. Stem Cell Reports 2014; 2:561-75. [PMID: 24936446 PMCID: PMC4050491 DOI: 10.1016/j.stemcr.2014.03.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 03/12/2014] [Accepted: 03/13/2014] [Indexed: 02/06/2023] Open
Abstract
Distinguishing between perivascular cell types remains a hurdle in vascular biology due to overlapping marker expressions and similar functionalities. Clarifying and defining heterogeneities in vitro among perivascular cells could lead to improved cell-based tissue regeneration strategies and a better understanding of human developmental processes. We studied contractile vascular smooth muscle cells (vSMCs), synthetic vSMCs, and pericytes derived from a common human pluripotent stem cell source. Using in vitro cultures, we show unique cell morphology, subcellular organelle organization (namely endoplasmic reticulum, mitochondria, and stress fibers), and expression of smooth muscle myosin heavy chain and elastin for each cell type. While differences in extracellular matrix deposition and remodeling were less pronounced, the multipotency, in vivo, migratory, invasion, and contractile functionalities are distinctive for each cell type. Overall, we define a repertoire of functional phenotypes in vitro specific for each of the human perivascular cell types, enabling their study and use in basic and translational research. Contractile and synthetic vSMCs and pericytes were derived from a common hiPSC line Morphology and organelle organization differ among the perivascular derivatives SMMHC and elastin specify the mature contractile vSMC phenotype Migration, invasion, and contractility are unique for each perivascular cell type.
Collapse
Affiliation(s)
- Maureen Wanjare
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Sravanti Kusuma
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA ; Department of Biomedical Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA ; Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| |
Collapse
|
232
|
Milbreta U, von Boxberg Y, Mailly P, Nothias F, Soares S. Astrocytic and vascular remodeling in the injured adult rat spinal cord after chondroitinase ABC treatment. J Neurotrauma 2014; 31:803-18. [PMID: 24380419 DOI: 10.1089/neu.2013.3143] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Upregulation of extracellular chondroitin sulfate proteoglycans (CSPG) is a primary cause for the failure of axons to regenerate after spinal cord injury (SCI), and the beneficial effect of their degradation by chondroitinase ABC (ChABC) is widely documented. Little is known, however, about the effect of ChABC treatment on astrogliosis and revascularization, two important factors influencing axon regrowth. This was investigated in the present study. Immediately after a spinal cord hemisection at thoracic level 8-9, we injected ChABC intrathecally at the sacral level, repeated three times until 10 days post-injury. Our results show an effective cleavage of CSPG glycosaminoglycan chains and stimulation of axonal remodeling within the injury site, accompanied by an extended period of astrocyte remodeling (up to 4 weeks). Interestingly, ChABC treatment favored an orientation of astrocytic processes directed toward the injury, in close association with axons at the lesion entry zone, suggesting a correlation between axon and astrocyte remodeling. Further, during the first weeks post-injury, ChABC treatment affected the morphology of laminin-positive blood vessel basement membranes and vessel-independent laminin deposits: hypertrophied blood vessels with detached or duplicated basement membrane were more numerous than in lesioned untreated animals. In contrast, at later time points, laminin expression increased and became more directly associated with newly formed blood vessels, the size of which tended to be closer to that found in intact tissue. Our data reinforce the idea that ChABC injection in combination with other synergistic treatments is a promising therapeutic strategy for SCI repair.
Collapse
Affiliation(s)
- Ulla Milbreta
- 1 Neuroscience Paris Seine/UMR8246/U1130/UMCR18 , IBPS/UPMC Univ Paris 06, Paris, France
| | | | | | | | | |
Collapse
|
233
|
Rupp C, Scherzer M, Rudisch A, Unger C, Haslinger C, Schweifer N, Artaker M, Nivarthi H, Moriggl R, Hengstschläger M, Kerjaschki D, Sommergruber W, Dolznig H, Garin-Chesa P. IGFBP7, a novel tumor stroma marker, with growth-promoting effects in colon cancer through a paracrine tumor-stroma interaction. Oncogene 2014; 34:815-25. [PMID: 24632618 DOI: 10.1038/onc.2014.18] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 12/26/2013] [Accepted: 01/31/2014] [Indexed: 02/07/2023]
Abstract
The activated tumor stroma participates in many processes that control tumorigenesis, including tumor cell growth, invasion and metastasis. Cancer-associated fibroblasts (CAFs) represent the major cellular component of the stroma and are the main source for connective tissue components of the extracellular matrix and various classes of proteolytic enzymes. The signaling pathways involved in the interactions between tumor and stromal cells and the molecular characteristics that distinguish normal 'resting' fibroblasts from cancer-associated or '-activated' fibroblasts remain poorly defined. Recent studies emphasized the prognostic and therapeutic significance of CAF-related molecular signatures and a number of those genes have been shown to serve as putative therapeutic targets. We have used immuno-laser capture microdissection and whole-genome Affymetrix GeneChip analysis to obtain transcriptional signatures from the activated tumor stroma of colon carcinomas that were compared with normal resting colonic fibroblasts. Several members of the Wnt-signaling pathway and gene sets related to hypoxia, epithelial-to-mesenchymal transition (EMT) and transforming growth factor-β (TGFβ) pathway activation were induced in CAFs. The putative TGFβ-target IGFBP7 was identified as a tumor stroma marker of epithelial cancers and as a tumor antigen in mesenchyme-derived sarcomas. We show here that in contrast to its tumor-suppressor function in epithelial cells, IGFPB7 can promote anchorage-independent growth in malignant mesenchymal cells and in epithelial cells with an EMT phenotype when IGFBP7 is expressed by the tumor cells themselves and can induce colony formation in colon cancer cells co-cultured with IGFBP7-expressing CAFs by a paracrine tumor-stroma interaction.
Collapse
Affiliation(s)
- C Rupp
- Clinical Institute for Pathology, Medical University of Vienna, Vienna, Austria
| | - M Scherzer
- Institute of Medical Genetics, Center of Pathobiology and Genetics, Medical University of Vienna, Vienna, Austria
| | - A Rudisch
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - C Unger
- Institute of Medical Genetics, Center of Pathobiology and Genetics, Medical University of Vienna, Vienna, Austria
| | - C Haslinger
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - N Schweifer
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - M Artaker
- Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - H Nivarthi
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - R Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - M Hengstschläger
- Institute of Medical Genetics, Center of Pathobiology and Genetics, Medical University of Vienna, Vienna, Austria
| | - D Kerjaschki
- Clinical Institute for Pathology, Medical University of Vienna, Vienna, Austria
| | | | - H Dolznig
- Institute of Medical Genetics, Center of Pathobiology and Genetics, Medical University of Vienna, Vienna, Austria
| | - P Garin-Chesa
- 1] Clinical Institute for Pathology, Medical University of Vienna, Vienna, Austria [2] Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| |
Collapse
|
234
|
Ozerdem U, Wells J, Hoda SA. Hyaline globules in mammary myofibroblastoma: a case report. Int J Surg Pathol 2014; 23:89-91. [PMID: 24583838 DOI: 10.1177/1066896914525235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A 52-year-old otherwise healthy woman presented with a solitary firm mass in the right breast. Histopathological evaluation of the 1.5-cm mass showed a mammary myofibroblastoma of the conventional spindle-cell type. High-power examination of hematoxylin-eosin-stained sections showed round, eosinophilic, intracytoplasmic, as well as extracellular, hyaline globules. These 5- to 20-µm globules appeared gray with a pinkish rim on Masson's trichrome stain. Immunohistochemically, the hyaline globules were strongly reactive with smooth muscle myosin heavy chain, desmin, and caldesmon. Histologically similar inclusion bodies have been reported in phylloides tumors--including those with myoid differentiation. To our knowledge, this is the first description of hyaline globules, a peculiar histological curiosity with no known clinical significance, in mammary myofibroblastoma.
Collapse
Affiliation(s)
- Ugur Ozerdem
- Weill Cornell Medical College, New York, NY, USA
| | - Justin Wells
- Weill Cornell Medical College, New York, NY, USA
| | - Syed A Hoda
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
235
|
Crisan M, Corselli M, Chen WCW, Péault B. Perivascular cells for regenerative medicine. J Cell Mol Med 2014; 16:2851-60. [PMID: 22882758 PMCID: PMC4393715 DOI: 10.1111/j.1582-4934.2012.01617.x] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 08/02/2012] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSC) are currently the best candidate therapeutic cells for regenerative medicine related to osteoarticular, muscular, vascular and inflammatory diseases, although these cells remain heterogeneous and necessitate a better biological characterization. We and others recently described that MSC originate from two types of perivascular cells, namely pericytes and adventitial cells and contain the in situ counterpart of MSC in developing and adult human organs, which can be prospectively purified using well defined cell surface markers. Pericytes encircle endothelial cells of capillaries and microvessels and express the adhesion molecule CD146 and the PDGFRβ, but lack endothelial and haematopoietic markers such as CD34, CD31, vWF (von Willebrand factor), the ligand for Ulex europaeus 1 (UEA1) and CD45 respectively. The proteoglycan NG2 is a pericyte marker exclusively associated with the arterial system. Besides its expression in smooth muscle cells, smooth muscle actin (αSMA) is also detected in subsets of pericytes. Adventitial cells surround the largest vessels and, opposite to pericytes, are not closely associated to endothelial cells. Adventitial cells express CD34 and lack αSMA and all endothelial and haematopoietic cell markers, as for pericytes. Altogether, pericytes and adventitial perivascular cells express in situ and in culture markers of MSC and display capacities to differentiate towards osteogenic, adipogenic and chondrogenic cell lineages. Importantly, adventitial cells can differentiate into pericyte-like cells under inductive conditions in vitro. Altogether, using purified perivascular cells instead of MSC may bring higher benefits to regenerative medicine, including the possibility, for the first time, to use these cells uncultured.
Collapse
Affiliation(s)
- Mihaela Crisan
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
236
|
Yao Y, Tsirka SE. Monocyte chemoattractant protein-1 and the blood-brain barrier. Cell Mol Life Sci 2014; 71:683-97. [PMID: 24051980 PMCID: PMC3946874 DOI: 10.1007/s00018-013-1459-1] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 07/20/2013] [Accepted: 08/19/2013] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) is a dynamic structure that maintains the homeostasis of the brain and thus proper neurological functions. BBB compromise has been found in many pathological conditions, including neuroinflammation. Monocyte chemoattractant protein-1 (MCP1), a chemokine that is transiently and significantly up-regulated during inflammation, is able to disrupt the integrity of BBB and modulate the progression of various diseases, including excitotoxic injury and hemorrhage. In this review, we first introduce the biochemistry and biology of MCP1, and then summarize the effects of MCP1 on BBB integrity as well as individual BBB components.
Collapse
Affiliation(s)
- Yao Yao
- Program in Molecular and Cellular Pharmacology, Department of Pharmacological Sciences, BST8-192, Stony Brook University, Stony Brook, NY 11794-8651 USA
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065 USA
| | - Stella E. Tsirka
- Program in Molecular and Cellular Pharmacology, Department of Pharmacological Sciences, BST8-192, Stony Brook University, Stony Brook, NY 11794-8651 USA
| |
Collapse
|
237
|
Cortez E, Roswall P, Pietras K. Functional subsets of mesenchymal cell types in the tumor microenvironment. Semin Cancer Biol 2014; 25:3-9. [PMID: 24412106 DOI: 10.1016/j.semcancer.2013.12.010] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 12/19/2013] [Accepted: 12/30/2013] [Indexed: 11/26/2022]
Abstract
In the field of tumor biology, increasing attention is now focused on the complex interactions between various constituent cell types within the tumor microenvironment as being functionally important for the etiology of the disease. The detailed description of tumor-promoting properties of cancer-associated fibroblasts, endothelial cells, pericytes, and immune cells, introduces novel potential drug targets for improved cancer treatments, as well as a rationale for exploring the tumor stroma as a previously unchartered source for prognostic or predictive biomarkers. However, recent work highlights the fact that cellular identity is perhaps too broadly defined and that subdivision of each cell type may reveal functionally distinct subsets of cells. Here, we will review our current understanding of the diversity of different subsets of mesenchymal cells, i.e., cancer-associated fibroblasts and pericytes, residing within the tumor parenchyma.
Collapse
Affiliation(s)
- Eliane Cortez
- Lund University, Department of Laboratory Medicine Lund, Division of Translational Cancer Research, Medicon Village, Building 404:A3, SE-223 81 Lund, Sweden
| | - Pernilla Roswall
- Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Scheeles Väg 2, SE-171 77 Stockholm, Sweden
| | - Kristian Pietras
- Lund University, Department of Laboratory Medicine Lund, Division of Translational Cancer Research, Medicon Village, Building 404:A3, SE-223 81 Lund, Sweden; Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Scheeles Väg 2, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
238
|
Binamé F. Transduction of extracellular cues into cell polarity: the role of the transmembrane proteoglycan NG2. Mol Neurobiol 2014; 50:482-93. [PMID: 24390567 DOI: 10.1007/s12035-013-8610-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/08/2013] [Indexed: 01/23/2023]
Abstract
Resident progenitor cells expressing nerve/glial antigen 2 (NG2) such as oligodendrocyte precursor cells (OPC) and pericytes persist in the adult brain. The transmembrane proteoglycan NG2 regulates migration of both these cell types in response to growth factors or specific components of the extracellular matrix. This role of NG2 is linked to the control of cell polarity. The polarization of OPC toward an acute lesion in the brain is impaired in NG2-deficient mice, supporting this concept. A review of the signaling pathways impinged on by NG2 reveals key proteins of cell polarity: phosphatidylinositol 3-kinase, focal adhesion kinase, Rho GTPases, and polarity complex proteins. In the scope of cell migration, I discuss here how the interplay of NG2 with signaling transmitted by extracellular cues can control the establishment of cell polarity, and I propose a model to integrate the apparent opposite effects of NG2 on cellular dynamics.
Collapse
Affiliation(s)
- Fabien Binamé
- Molecular Cell Biology, Department of Biology, Johannes Gutenberg University of Mainz, Mainz, Germany,
| |
Collapse
|
239
|
Welser-Alves JV, Boroujerdi A, Tigges U, Milner R. Analysis of TNF-alpha-mediated cerebral pericyte remodeling. Methods Mol Biol 2014; 1155:81-93. [PMID: 24788175 DOI: 10.1007/978-1-4939-0669-7_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
As well as being a central regulator of inflammatory and immune-mediated events, TNF-α also influences vascular remodeling, resulting in alterations in the structure and function of blood vessels. In addition to endothelial cells, pericytes are another type of vascular cell that significantly contribute to the development, maturation, stabilization, and remodeling of blood vessels. To investigate the regulatory influence of different factors on pericyte behavior, we recently described a novel yet simple approach of isolating and culturing highly pure, high density cultures of mouse brain pericytes. In this chapter, we briefly describe this culture system, as well as methods for examining different aspects of pericyte behavior, including cell adhesion, cell migration, and cell proliferation. These assays can be used to examine the influence of TNF-α or any other factor on pericyte behavior.
Collapse
Affiliation(s)
- Jennifer V Welser-Alves
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM-132, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | | | | | | |
Collapse
|
240
|
Chintalgattu V, Rees ML, Culver JC, Goel A, Jiffar T, Zhang J, Dunner K, Pati S, Bankson JA, Pasqualini R, Arap W, Bryan NS, Taegtmeyer H, Langley RR, Yao H, Kupferman ME, Entman ML, Dickinson ME, Khakoo AY. Coronary microvascular pericytes are the cellular target of sunitinib malate-induced cardiotoxicity. Sci Transl Med 2013; 5:187ra69. [PMID: 23720580 DOI: 10.1126/scitranslmed.3005066] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sunitinib malate is a multitargeted receptor tyrosine kinase inhibitor used in the treatment of human malignancies. A substantial number of sunitinib-treated patients develop cardiac dysfunction, but the mechanism of sunitinib-induced cardiotoxicity is poorly understood. We show that mice treated with sunitinib develop cardiac and coronary microvascular dysfunction and exhibit an impaired cardiac response to stress. The physiological changes caused by treatment with sunitinib are accompanied by a substantial depletion of coronary microvascular pericytes. Pericytes are a cell type that is dependent on intact platelet-derived growth factor receptor (PDGFR) signaling but whose role in the heart is poorly defined. Sunitinib-induced pericyte depletion and coronary microvascular dysfunction are recapitulated by CP-673451, a structurally distinct PDGFR inhibitor, confirming the role of PDGFR in pericyte survival. Thalidomide, an anticancer agent that is known to exert beneficial effects on pericyte survival and function, prevents sunitinib-induced pericyte cell death in vitro and prevents sunitinib-induced cardiotoxicity in vivo in a mouse model. Our findings suggest that pericytes are the primary cellular target of sunitinib-induced cardiotoxicity and reveal the pericyte as a cell type of concern in the regulation of coronary microvascular function. Furthermore, our data provide preliminary evidence that thalidomide may prevent cardiotoxicity in sunitinib-treated cancer patients.
Collapse
|
241
|
Schallek J, Geng Y, Nguyen H, Williams DR. Morphology and topography of retinal pericytes in the living mouse retina using in vivo adaptive optics imaging and ex vivo characterization. Invest Ophthalmol Vis Sci 2013; 54:8237-50. [PMID: 24150762 DOI: 10.1167/iovs.13-12581] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To noninvasively image retinal pericytes in the living eye and characterize NG2-positive cell topography and morphology in the adult mouse retina. METHODS Transgenic mice expressing fluorescent pericytes (NG2, DsRed) were imaged using a two-channel, adaptive optics scanning laser ophthalmoscope (AOSLO). One channel imaged vascular perfusion with near infrared light. A second channel simultaneously imaged fluorescent retinal pericytes. Mice were also imaged using wide-field ophthalmoscopy. To confirm in vivo imaging, five eyes were enucleated and imaged in flat mount with conventional fluorescent microscopy. Cell topography was quantified relative to the optic disc. RESULTS We observed strong DsRed fluorescence from NG2-positive cells. AOSLO revealed fluorescent vascular mural cells enveloping all vessels in the living retina. Cells were stellate on larger venules, and showed banded morphology on arterioles. NG2-positive cells indicative of pericytes were found on the smallest capillaries of the retinal circulation. Wide-field SLO enabled quick assessment of NG2-positive distribution, but provided insufficient resolution for cell counts. Ex vivo microscopy showed relatively even topography of NG2-positive capillary pericytes at eccentricities more than 0.3 mm from the optic disc (515 ± 94 cells/mm(2) of retinal area). CONCLUSIONS We provide the first high-resolution images of retinal pericytes in the living animal. Subcellular resolution enabled morphological identification of NG2-positive cells on capillaries showing classic features and topography of retinal pericytes. This report provides foundational basis for future studies that will track and quantify pericyte topography, morphology, and function in the living retina over time, especially in the progression of microvascular disease.
Collapse
Affiliation(s)
- Jesse Schallek
- Center for Visual Science, University of Rochester, Rochester, New York
| | | | | | | |
Collapse
|
242
|
Gu X, Fliesler SJ, Zhao YY, Stallcup WB, Cohen AW, Elliott MH. Loss of caveolin-1 causes blood-retinal barrier breakdown, venous enlargement, and mural cell alteration. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:541-55. [PMID: 24326256 DOI: 10.1016/j.ajpath.2013.10.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 09/13/2013] [Accepted: 10/28/2013] [Indexed: 12/20/2022]
Abstract
Blood-retinal barrier (BRB) breakdown and related vascular changes are implicated in several ocular diseases. The molecules and mechanisms regulating BRB integrity and pathophysiology are not fully elucidated. Caveolin-1 (Cav-1) ablation results in loss of caveolae and microvascular pathologies, but the role of Cav-1 in the retina is largely unknown. We examined BRB integrity and vasculature in Cav-1 knockout mice and found a significant increase in BRB permeability, compared with wild-type controls, with branch veins being frequent sites of breakdown. Vascular hyperpermeability occurred without apparent alteration in junctional proteins. Such hyperpermeability was not rescued by inhibiting eNOS activity. Veins of Cav-1 knockout retinas exhibited additional pathological features, including i) eNOS-independent enlargement, ii) altered expression of mural cell markers (eg, down-regulation of NG2 and up-regulation of αSMA), and iii) dramatic alterations in mural cell phenotype near the optic nerve head. We observed a significant NO-dependent increase in retinal artery diameter in Cav-1 knockout mice, suggesting that Cav-1 plays a role in autoregulation of resistance vessels in the retina. These findings implicate Cav-1 in maintaining BRB integrity in retinal vasculature and suggest a previously undefined role in the retinal venous system and associated mural cells. Our results are relevant to clinically significant retinal disorders with vascular pathologies, including diabetic retinopathy, uveoretinitis, and primary open-angle glaucoma.
Collapse
Affiliation(s)
- Xiaowu Gu
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Steven J Fliesler
- Research Service, Veterans Affairs Western New York Healthcare System, Buffalo, New York; Department of Ophthalmology, University at Buffalo, State University of New York, Buffalo, New York; Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, New York; SUNY Eye Institute, University at Buffalo, State University of New York, Buffalo, New York
| | - You-Yang Zhao
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois; Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois
| | - William B Stallcup
- Tumor Microenvironment Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Alex W Cohen
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Michael H Elliott
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| |
Collapse
|
243
|
Matsusue Y, Horii-Hayashi N, Kirita T, Nishi M. Distribution of corticosteroid receptors in mature oligodendrocytes and oligodendrocyte progenitors of the adult mouse brain. J Histochem Cytochem 2013; 62:211-26. [PMID: 24309510 DOI: 10.1369/0022155413517700] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The expression of glucocorticoid receptors (GRs) was investigated immunohistochemically in two different lineages of oligodendrocytes, using carbonic anhydrase (CA) II and neuron glial antigen (NG) 2 as markers of mature oligodendrocytes and oligodendrocyte progenitors, respectively. We focused on the gray matter regions, including CA1, CA3 and the dentate gyrus of the hippocampus, the primary somatosensory cortex barrel field and the basolateral amygdala, and the white matter regions, including the corpus callosum, external capsule and fimbria of the hippocampus. More than 80% of CAII-immunoreactive (IR) cells and more than 95% of NG2-IR cells expressed GRs in various regions of the brain. In contrast, neither CAII-IR cells nor NG2-IR cells expressed mineralocorticoid receptors (MRs) in the same regions. The intensity of GR expression was drastically reduced in CA II-IR cells and NG2-IR cells in the same regions in adrenalectomized mice. Finally, steroid receptor co-activator (SRC)-1 and p300, both of which are cofactors for GR, were expressed in the gray and white matter regions in NG2-IR cells, but not in CAII-IR cells. These results suggest that the expression of GRs in oligodendrocytes and their progenitor cells mediates several functions in vivo, including differentiation and myelination, as a major target of glucocorticoids and their cofactors.
Collapse
Affiliation(s)
- Yumiko Matsusue
- Department of Oral and Maxillofacial Surgery, Nara Medical University (YM, TK)
| | | | | | | |
Collapse
|
244
|
Harlow DE, Macklin WB. Inhibitors of myelination: ECM changes, CSPGs and PTPs. Exp Neurol 2013; 251:39-46. [PMID: 24200549 DOI: 10.1016/j.expneurol.2013.10.017] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/22/2013] [Accepted: 10/26/2013] [Indexed: 01/06/2023]
Abstract
After inflammation-induced demyelination, such as in the disease multiple sclerosis, endogenous remyelination often fails. However, in animal models of demyelination induced with toxins, remyelination can be quite robust. A significant difference between inflammation-induced and toxin-induced demyelination is the response of local cells within the lesion, including astrocytes, oligodendrocytes, microglia/macrophages, and NG2+ cells, which respond to inflammatory stimuli with increased extracellular matrix (ECM) protein and chondroitin sulfate proteoglycan (CSPG) production and deposition. Here, we summarize current knowledge of ECM changes in demyelinating lesions, as well as oligodendrocyte responses to aberrant ECM proteins and CSPGs after various types of demyelinating insults. The discovery that CSPGs act through the receptor protein tyrosine phosphatase sigma (PTPσ) and the Rho-ROCK pathway to inhibit oligodendrocyte process extension and myelination, but not oligodendrocyte differentiation (Pendleton et al., Experimental Neurology (2013) vol. 247, pp. 113-121), highlights the need to better understand the ECM changes that accompany demyelination and their influence on oligodendrocytes and effective remyelination.
Collapse
Affiliation(s)
- Danielle E Harlow
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, 12801 East 17th Avenue, Research Complex 1 South, Mail Stop 8108, Aurora, CO 80045, USA; Center for NeuroScience, University of Colorado School of Medicine, 12801 East 17th Avenue, Research Complex 1 South, Mail Stop 8108, Aurora, CO 80045, USA.
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, 12801 East 17th Avenue, Research Complex 1 South, Mail Stop 8108, Aurora, CO 80045, USA; Center for NeuroScience, University of Colorado School of Medicine, 12801 East 17th Avenue, Research Complex 1 South, Mail Stop 8108, Aurora, CO 80045, USA.
| |
Collapse
|
245
|
Jo DH, Kim JH, Heo JI, Kim JH, Cho CH. Interaction between pericytes and endothelial cells leads to formation of tight junction in hyaloid vessels. Mol Cells 2013; 36:465-71. [PMID: 24213675 PMCID: PMC3887934 DOI: 10.1007/s10059-013-0228-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 09/03/2013] [Accepted: 09/09/2013] [Indexed: 11/25/2022] Open
Abstract
The hyaloid vessel is a transient vascular network that nourishes the lens and the primary vitreous in the early developmental periods. In hyaloid vessels devoid of the support of astrocytes, we demonstrate that tight junction proteins, zonula occludens-1 and occludin, are regularly expressed at the junction of endothelial cells. To figure out the factor influencing the formation of tight junctions in hyaloid vessels, we further progress to investigate the interactions between endothelial cells and pericytes, two representative constituent cells in hyaloid vessels. Interestingly, endothelial cells interact with pericytes in the early postnatal periods and the interaction between two cell types provokes the up-regulation of transforming growth factor β1. Further in vitro experiments demonstrate that transforming growth factor β1 induces the activation of Smad2 and Smad3 and the formation of tight junction proteins. Taken together, in hyaloid vessels, pericytes seem to regulate the formation of tight junctions by the interaction with endothelial cells even without the support of astrocytes. Additionally, we suggest that the hyaloid vessel is a valuable system that can be utilized for the investigation of cell-cell interaction in the formation of tight junctions in developing vasculatures.
Collapse
Affiliation(s)
- Dong Hyun Jo
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 110-744, Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-744, Korea
| | - Jin Hyoung Kim
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 110-744, Korea
| | - Jong-Ik Heo
- Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul 110-744, Korea
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul 110-744, Korea
| | - Jeong Hun Kim
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 110-744, Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-744, Korea
- Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul 110-744, Korea
- Department of Ophthalmology, College of Medicine, Seoul National University, Seoul 110-744, Korea
| | - Chung-Hyun Cho
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-744, Korea
- Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul 110-744, Korea
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul 110-744, Korea
| |
Collapse
|
246
|
Robins SC, Trudel E, Rotondi O, Liu X, Djogo T, Kryzskaya D, Bourque CW, Kokoeva MV. Evidence for NG2-glia derived, adult-born functional neurons in the hypothalamus. PLoS One 2013; 8:e78236. [PMID: 24205170 PMCID: PMC3812154 DOI: 10.1371/journal.pone.0078236] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/11/2013] [Indexed: 11/18/2022] Open
Abstract
Accumulating evidence suggests that the adult murine hypothalamus, a control site of several fundamental homeostatic processes, has neurogenic capacity. Correspondingly, the adult hypothalamus exhibits considerable cell proliferation that is ongoing even in the absence of external stimuli, and some of the newborn cells have been shown to mature into cells that express neuronal fate markers. However, the identity and characteristics of proliferating cells within the hypothalamic parenchyma have yet to be thoroughly investigated. Here we show that a subset of NG2-glia distributed throughout the mediobasal hypothalamus are proliferative and express the stem cell marker Sox2. We tracked the constitutive differentiation of hypothalamic NG2-glia by employing genetic fate mapping based on inducible Cre recombinase expression under the control of the NG2 promoter, demonstrating that adult hypothalamic NG2-glia give rise to substantial numbers of APC+ oligodendrocytes and a smaller population of HuC/D+ or NeuN+ neurons. Labelling with the cell proliferation marker BrdU confirmed that some NG2-derived neurons have proliferated shortly before differentiation. Furthermore, patch-clamp electrophysiology revealed that some NG2-derived cells display an immature neuronal phenotype and appear to receive synaptic input indicative of their electrical integration in local hypothalamic circuits. Together, our studies show that hypothalamic NG2-glia are able to take on neuronal fates and mature into functional neurons, indicating that NG2-glia contribute to the neurogenic capacity of the adult hypothalamus.
Collapse
Affiliation(s)
| | - Eric Trudel
- Centre for Research in Neuroscience, McGill University, Montreal, Canada
| | - Olivia Rotondi
- Department of Medicine, McGill University, Montreal, Canada
| | - Xiaohong Liu
- Department of Medicine, McGill University, Montreal, Canada
| | - Tina Djogo
- Department of Medicine, McGill University, Montreal, Canada
| | | | - Charles W. Bourque
- Centre for Research in Neuroscience, McGill University, Montreal, Canada
| | - Maia V. Kokoeva
- Department of Medicine, McGill University, Montreal, Canada
- * E-mail:
| |
Collapse
|
247
|
Kang HS, Moon YJ, Kim YY, Park WY, Park AK, Wang KC, Kim JE, Phi JH, Lee JY, Kim SK. Smooth-muscle progenitor cells isolated from patients with moyamoya disease: novel experimental cell model. J Neurosurg 2013; 120:415-25. [PMID: 24160477 DOI: 10.3171/2013.9.jns131000] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Moyamoya disease (MMD) is a cerebrovascular occlusive disease affecting bilateral internal carotid termini. Smooth-muscle cells are one of the major cell types involved in this disease process. The characteristics of circulating smooth-muscle progenitor cells (SPCs) in MMD are poorly understood. The authors purified SPCs from the peripheral blood of patients with MMD and sought to identify differentially expressed genes (DEGs) in SPCs from these patients. METHODS The authors cultured and isolated SPCs from the peripheral blood of patients with MMD (n = 25) and healthy control volunteers (n = 22). After confirmation of the cellular phenotype, RNA was extracted from the cells and DEGs were identified using a commercially available gene chip. Real-time quantitative reverse transcription polymerase chain reaction was performed to confirm the putative pathogenetic DEGs. RESULTS The SPC-type outgrowth cells in patients with MMD invariably showed a hill-and-valley appearance under microscopic examination, and demonstrated high α-smooth muscle actin, myosin heavy chain, and calponin expression (96.5% ± 2.1%, 42.8% ± 18.6%, and 87.1% ± 8.2%, respectively), and minimal CD31 expression (less than 1%) on fluorescence-activated cell sorter analysis. The SPCs in the MMD group tended to make more irregularly arranged and thickened tubules on the tube formation assay. In the SPCs from patients with MMD, 286 genes (124 upregulated and 162 downregulated) were differentially expressed; they were related to cell adhesion, cell migration, immune response, and vascular development. CONCLUSIONS With adequate culture conditions, SPCs could be established from the peripheral blood of patients with MMD. These cells showed specific DEGs compared with healthy control volunteers. This study provides a novel experimental cell model for further research of MMD.
Collapse
Affiliation(s)
- Hyun-Seung Kang
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul
| | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Simplified protocol for isolation of multipotential NG2 cells from postnatal mouse. J Neurosci Methods 2013; 219:252-61. [DOI: 10.1016/j.jneumeth.2013.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 11/17/2022]
|
249
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Type-1 pericytes participate in fibrous tissue deposition in aged skeletal muscle. Am J Physiol Cell Physiol 2013; 305:C1098-113. [PMID: 24067916 DOI: 10.1152/ajpcell.00171.2013] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In older adults, changes in skeletal muscle composition are associated with increased fibrosis, loss of mass, and decreased force, which can lead to dependency, morbidity, and mortality. Understanding the biological mechanisms responsible is essential to sustaining and improving their quality of life. Compared with young mice, aged mice take longer to recover from muscle injury; their tissue fibrosis is more extensive, and regenerated myofibers are smaller. Strong evidence indicates that cells called pericytes, embedded in the basement membrane of capillaries, contribute to the satellite-cell pool and muscle growth. In addition to their role in skeletal muscle repair, after tissue damage, they detach from capillaries and migrate to the interstitial space to participate in fibrosis formation. Here we distinguish two bona fide pericyte subtypes in the skeletal muscle interstitium, type-1 (Nestin-GFP(-)/NG2-DsRed(+)) and type-2 (Nestin-GFP(+)/NG2-DsRed(+)), and characterize their heretofore unknown specific roles in the aging environment. Our in vitro results show that type-1 and type-2 pericytes are either fibrogenic or myogenic, respectively. Transplantation studies in young animals indicate that type-2 pericytes are myogenic, while type-1 pericytes remain in the interstitial space. In older mice, however, the muscular regenerative capacity of type-2 pericytes is limited, and type-1 pericytes produce collagen, contributing to fibrous tissue deposition. We conclude that in injured muscles from aging mice, the pericytes involved in skeletal muscle repair differ from those associated with scar formation.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | | | | | | | | |
Collapse
|
250
|
Edwards IJ, Singh M, Morris S, Osborne L, Le Ruez T, Fuad M, Deuchars SA, Deuchars J. A simple method to fluorescently label pericytes in the CNS and skeletal muscle. Microvasc Res 2013; 89:164-8. [DOI: 10.1016/j.mvr.2013.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/23/2013] [Accepted: 05/31/2013] [Indexed: 11/16/2022]
|