101
|
de Souza LEB, Malta TM, Kashima Haddad S, Covas DT. Mesenchymal Stem Cells and Pericytes: To What Extent Are They Related? Stem Cells Dev 2016; 25:1843-1852. [PMID: 27702398 DOI: 10.1089/scd.2016.0109] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Mesenchymal stem cells (MSCs) were initially identified as progenitors of skeletal tissues within mammalian bone marrow and cells with similar properties were also obtained from other tissues such as adipose and dental pulp. Although MSCs have been extensively investigated, their native behavior and in vivo identity remain poorly defined. Uncovering the in vivo identity of MSCs has been challenging due to the lack of exclusive cell markers, cellular alterations caused by culture methods, and extensive focus on in vitro properties for characterization. Although MSC site of origin influences their functional properties, these mesenchymal progenitors can be found in the perivascular space in virtually all organs from where they were obtained. However, the precise identity of MSCs within the vascular wall is highly controversial. The recurrent concept that MSCs correspond to pericytes in vivo has been supported mainly by their perivascular localization and expression of some molecular markers. However, this view has been a subject of controversy, in part, due to the application of loose criteria to define pericytes and due to the lack of a marker able to unequivocally identify these cells. Furthermore, recent evidences indicate that subpopulations of MSCs can be found at extravascular sites such as the endosteum. In this opinion review, we bring together the advances and pitfalls on the search for the in vivo identity of MSCs and highlight the recent evidences that suggest that perivascular MSCs are adventitial cells, acting as precursors of pericytes and other stromal cells during tissue homeostasis.
Collapse
Affiliation(s)
- Lucas Eduardo Botelho de Souza
- 1 Department of Clinical Medicine, Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto, Brazil .,2 National Institute of Science and Technology for Stem Cell and Cell Therapy , Ribeirão Preto, Brazil
| | - Tathiane Maistro Malta
- 2 National Institute of Science and Technology for Stem Cell and Cell Therapy , Ribeirão Preto, Brazil .,3 Department of Genetics, Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto, Brazil
| | - Simone Kashima Haddad
- 2 National Institute of Science and Technology for Stem Cell and Cell Therapy , Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- 1 Department of Clinical Medicine, Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto, Brazil .,2 National Institute of Science and Technology for Stem Cell and Cell Therapy , Ribeirão Preto, Brazil
| |
Collapse
|
102
|
PDGF-A and PDGF-B induces cardiac fibrosis in transgenic mice. Exp Cell Res 2016; 349:282-290. [PMID: 27816607 DOI: 10.1016/j.yexcr.2016.10.022] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 10/26/2016] [Accepted: 10/27/2016] [Indexed: 02/06/2023]
Abstract
Platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) contribute to normal heart development. Deficient or abnormal expression of Pdgf and Pdgfr genes have a negative impact on cardiac development and function. The cellular effects of PDGFs in the hearts of Pdgf/Pdgfr mutants and the pathogenesis of the resulting abnormalities are poorly understood, but different PDGF isoforms induce varying effects. Here, we generated three new transgenic mouse types which complete a set of studies, where all different PDGF ligands have been expressed under the same heart specific alpha-myosin heavy chain promoter. Transgenic expression of the natural isoforms of Pdgfa and Pdgfb resulted in isoform specific fibrotic reactions and cardiac hypertrophy. Pdgfa overexpression resulted in a severe fibrotic reaction with up to 8-fold increase in cardiac size, leading to lethal cardiac failure within a few weeks after birth. In contrast, Pdgfb overexpression led to focal fibrosis and moderate cardiac hypertrophy. As PDGF-A and PDGF-B have different affinity for the two PDGF receptors, we analyzed the expression of the receptors and the histology of the fibrotic hearts. Our data suggest that the stronger fibrotic effect generated by Pdgfa overexpression was mediated by Pdgfrα in cardiac interstitial mesenchymal cells, i.e. the likely source of extracellular matrix depostion and fibrotic reaction. The apparent sensitivity of the heart to ectopic PDGFRα agonists supports a role for endogenous PDGFRα agonists in the pathogenesis of cardiac fibrosis.
Collapse
|
103
|
Harbi S, Wang R, Gregory M, Hanson N, Kobylarz K, Ryan K, Deng Y, Lopez P, Chiriboga L, Mignatti P. Infantile Hemangioma Originates From A Dysregulated But Not Fully Transformed Multipotent Stem Cell. Sci Rep 2016; 6:35811. [PMID: 27786256 PMCID: PMC5081534 DOI: 10.1038/srep35811] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 10/05/2016] [Indexed: 12/11/2022] Open
Abstract
Infantile hemangioma (IH) is the most common tumor of infancy. Its cellular origin and biological signals for uncontrolled growth are poorly understood, and specific pharmacological treatment is unavailable. To understand the process of hemangioma-genesis we characterized the progenitor hemangioma-derived stem cell (HemSC) and its lineage and non-lineage derivatives. For this purpose we performed a high-throughput (HT) phenotypic and gene expression analysis of HemSCs, and analyzed HemSC-derived tumorspheres. We found that IH is characterized by high expression of genes involved in vasculogenesis, angiogenesis, tumorigenesis and associated signaling pathways. These results show that IH derives from a dysregulated stem cell that remains in an immature, arrested stage of development. The potential biomarkers we identified can afford the development of diagnostic tools and precision-medicine therapies to "rewire" or redirect cellular transitions at an early stage, such as signaling pathways or immune response modifiers.
Collapse
Affiliation(s)
- Shaghayegh Harbi
- Department of Medicine, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
- VasculoTox Inc., New York, NY 10001, USA
| | - Rong Wang
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Michael Gregory
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Nicole Hanson
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Keith Kobylarz
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
- Pfizer Inc., Pearl River, NY 10965, USA
| | - Kamilah Ryan
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Yan Deng
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Peter Lopez
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Luis Chiriboga
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Paolo Mignatti
- Department of Medicine, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
- Department of Cell Biology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| |
Collapse
|
104
|
Rivera-Gonzalez GC, Shook BA, Andrae J, Holtrup B, Bollag K, Betsholtz C, Rodeheffer MS, Horsley V. Skin Adipocyte Stem Cell Self-Renewal Is Regulated by a PDGFA/AKT-Signaling Axis. Cell Stem Cell 2016; 19:738-751. [PMID: 27746098 DOI: 10.1016/j.stem.2016.09.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/14/2016] [Accepted: 09/11/2016] [Indexed: 12/15/2022]
Abstract
Tissue growth and maintenance requires stem cell populations that self-renew, proliferate, and differentiate. Maintenance of white adipose tissue (WAT) requires the proliferation and differentiation of adipocyte stem cells (ASCs) to form postmitotic, lipid-filled mature adipocytes. Here we use the dynamic adipogenic program that occurs during hair growth to uncover an unrecognized regulator of ASC self-renewal and proliferation, PDGFA, which activates AKT signaling to drive and maintain the adipogenic program in the skin. Pdgfa expression is reduced in aged ASCs and is required for ASC proliferation and maintenance in the dermis, but not in other WATs. Our molecular and genetic studies uncover PI3K/AKT2 as a direct PDGFA target that is activated in ASCs during WAT hyperplasia and is functionally required for dermal ASC proliferation. Our data therefore reveal active mechanisms that regulate ASC self-renewal in the skin and show that distinct regulatory mechanisms operate in different WAT depots.
Collapse
Affiliation(s)
| | - Brett A Shook
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Johanna Andrae
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Brandon Holtrup
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Katherine Bollag
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Matthew S Rodeheffer
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University, New Haven, CT 06520, USA
| | - Valerie Horsley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA; Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
105
|
Cho JS, Fang TC, Reynolds TL, Sofia DJ, Hamann S, Burkly LC. PDGF-BB Promotes Type I IFN-Dependent Vascular Alterations and Monocyte Recruitment in a Model of Dermal Fibrosis. PLoS One 2016; 11:e0162758. [PMID: 27618690 PMCID: PMC5019454 DOI: 10.1371/journal.pone.0162758] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/29/2016] [Indexed: 12/15/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune disorder that can result in extensive tissue damage in the skin and, in advanced cases, internal organs. Vasculopathy, aberrant immune activation, and tissue fibrosis are three hallmarks of the disease that have been identified, with vasculopathy and aberrant immunity being amongst the earliest events. However, a mechanistic link between these processes has not been established. Here, we have identified a novel role of platelet derived growth factor-BB (PDGF-BB)/PDGFRβ activation in combination with dermal injury induced by bleomycin as a driver of early, aberrant expression of interferon stimulatory genes (ISGs) and inflammatory monocyte infiltration. Activation of PDGFRβ in combination with bleomycin-induced dermal injury resulted in increased dermal thickness, vascular density, monocyte/macrophage infiltration, and exacerbation of tissue injury. Many of these features were dependent on IFNAR-signaling, and an increase in the number of interferon-beta (IFN-β) producing monocytes cells was found in the skin lesions. Taken together, these results identify a novel link between PDGFRβ activation, and Type I IFN-driven vascular maintenance and monocyte/macrophage cell recruitment, and provide a potential explanation linking key features of SSc that were previously thought to be unrelated.
Collapse
Affiliation(s)
- John S. Cho
- Immunology Research, Biogen, Cambridge, MA, United States of America
- * E-mail: (JSC); (LCB)
| | - Terry C. Fang
- Immunology Research, Biogen, Cambridge, MA, United States of America
| | - Taylor L. Reynolds
- Translational Sciences - Pathology, Biogen, Cambridge, MA, United States of America
| | - Daniel J. Sofia
- Translational Sciences - Pathology, Biogen, Cambridge, MA, United States of America
| | - Stefan Hamann
- Translational Sciences - Pathology, Biogen, Cambridge, MA, United States of America
| | - Linda C. Burkly
- Immunology Research, Biogen, Cambridge, MA, United States of America
- * E-mail: (JSC); (LCB)
| |
Collapse
|
106
|
Tang Y, Qian SW, Wu MY, Wang J, Lu P, Li X, Huang HY, Guo L, Sun X, Xu CJ, Tang QQ. BMP4 mediates the interplay between adipogenesis and angiogenesis during expansion of subcutaneous white adipose tissue. J Mol Cell Biol 2016; 8:302-12. [DOI: 10.1093/jmcb/mjw019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 12/25/2015] [Indexed: 11/14/2022] Open
|
107
|
Gallini R, Huusko J, Ylä-Herttuala S, Betsholtz C, Andrae J. Isoform-Specific Modulation of Inflammation Induced by Adenoviral Mediated Delivery of Platelet-Derived Growth Factors in the Adult Mouse Heart. PLoS One 2016; 11:e0160930. [PMID: 27513343 PMCID: PMC4981378 DOI: 10.1371/journal.pone.0160930] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 07/27/2016] [Indexed: 02/06/2023] Open
Abstract
Platelet-derived growth factors (PDGFs) are key regulators of mesenchymal cells in vertebrate development. To what extent PDGFs also exert beneficial homeostatic or reparative roles in adult organs, as opposed to adverse fibrogenic responses in pathology, are unclear. PDGF signaling plays critical roles during heart development, during which forced overexpression of PDGFs induces detrimental cardiac fibrosis; other studies have implicated PDGF signaling in post-infarct myocardial repair. Different PDGFs may exert different effects mediated through the two PDGF receptors (PDGFRα and PDGFRβ) in different cell types. Here, we assessed responses induced by five known PDGF isoforms in the adult mouse heart in the context of adenovirus vector-mediated inflammation. Our results show that different PDGFs have different, in some cases even opposing, effects. Strikingly, whereas the major PDGFRα agonists (PDGF-A and -C) decreased the amount of scar tissue and increased the numbers of PDGFRα-positive fibroblasts, PDGFRβ agonists either induced large scars with extensive inflammation (PDGF-B) or dampened the adenovirus-induced inflammation and produced a small and dense scar (PDGF-D). These results provide evidence for PDGF isoform-specific inflammation-modulating functions that may have therapeutic implications. They also illustrate a surprising complexity in the PDGF-mediated pathophysiological responses.
Collapse
Affiliation(s)
- Radiosa Gallini
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Jenni Huusko
- Department of Biotechnology and Molecular Medicine, AI Virtanen Institute for Molecular Sciences, University of Kuopio, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, AI Virtanen Institute for Molecular Sciences, University of Kuopio, Kuopio, Finland
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Johanna Andrae
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
108
|
Ando K, Fukuhara S, Izumi N, Nakajima H, Fukui H, Kelsh RN, Mochizuki N. Clarification of mural cell coverage of vascular endothelial cells by live imaging of zebrafish. Development 2016; 143:1328-39. [PMID: 26952986 PMCID: PMC4852519 DOI: 10.1242/dev.132654] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/24/2016] [Indexed: 12/16/2022]
Abstract
Mural cells (MCs) consisting of vascular smooth muscle cells and pericytes cover the endothelial cells (ECs) to regulate vascular stability and homeostasis. Here, we clarified the mechanism by which MCs develop and cover ECs by generating transgenic zebrafish lines that allow live imaging of MCs and by lineage tracing in vivo To cover cranial vessels, MCs derived from either neural crest cells or mesoderm emerged around the preformed EC tubes, proliferated and migrated along EC tubes. During their migration, the MCs moved forward by extending their processes along the inter-EC junctions, suggesting a role for inter-EC junctions as a scaffold for MC migration. In the trunk vasculature, MCs derived from mesoderm covered the ventral side of the dorsal aorta (DA), but not the posterior cardinal vein. Furthermore, the MCs migrating from the DA or emerging around intersegmental vessels (ISVs) preferentially covered arterial ISVs rather than venous ISVs, indicating that MCs mostly cover arteries during vascular development. Thus, live imaging and lineage tracing enabled us to clarify precisely how MCs cover the EC tubes and to identify the origins of MCs.
Collapse
Affiliation(s)
- Koji Ando
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Shigetomo Fukuhara
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Nanae Izumi
- Frontier Research Laboratories, R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Robert N Kelsh
- Centre for Regenerative Medicine, Developmental Biology Programme, Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan AMED-CREST, Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1, Suita, Osaka 565-8565, Japan
| |
Collapse
|
109
|
Hegner B, Schaub T, Catar R, Kusch A, Wagner P, Essin K, Lange C, Riemekasten G, Dragun D. Intrinsic Deregulation of Vascular Smooth Muscle and Myofibroblast Differentiation in Mesenchymal Stromal Cells from Patients with Systemic Sclerosis. PLoS One 2016; 11:e0153101. [PMID: 27054717 PMCID: PMC4824407 DOI: 10.1371/journal.pone.0153101] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/23/2016] [Indexed: 11/18/2022] Open
Abstract
Introduction Obliterative vasculopathy and fibrosis are hallmarks of systemic sclerosis (SSc), a severe systemic autoimmune disease. Bone marrow-derived mesenchymal stromal cells (MSCs) from SSc patients may harbor disease-specific abnormalities. We hypothesized disturbed vascular smooth muscle cell (VSMC) differentiation with increased propensity towards myofibroblast differentiation in response to SSc-microenvironment defining growth factors and determined responsible mechanisms. Methods We studied responses of multipotent MSCs from SSc-patients (SSc-MSCs) and healthy controls (H-MSCs) to long-term exposure to CTGF, b-FGF, PDGF-BB or TGF-β1. Differentiation towards VSMC and myofibroblast lineages was analyzed on phenotypic, biochemical, and functional levels. Intracellular signaling studies included analysis of TGF-β receptor regulation, SMAD, AKT, ERK1/2 and autocrine loops. Results VSMC differentiation towards both, contractile and synthetic VSMC phenotypes in response to CTGF and b-FGF was disturbed in SSc-MSCs. H-MSCs and SSc-MSCs responded equally to PDGF-BB with prototypic fibroblastic differentiation. TGF-β1 initiated myofibroblast differentiation in both cell types, yet with striking phenotypic and functional differences: In relation to H-MSC-derived myofibroblasts induced by TGF-β1, those obtained from SSc-MSCs expressed more contractile proteins, migrated towards TGF-β1, had low proliferative capacity, and secreted higher amounts of collagen paralleled by reduced MMP expression. Higher levels of TGF-β receptor 1 and enhanced canonical and noncanonical TGF-β signaling in SSc-MSCs accompanied aberrant differentiation response of SSc-MSCs in comparison to H-MSCs. Conclusions Deregulated VSMC differentiation with a shift towards myofibroblast differentiation expands the concept of disturbed endogenous regenerative capacity of MSCs from SSc patients. Disease related intrinsic hyperresponsiveness to TGF-β1 with increased collagen production may represent one responsible mechanism. Better understanding of repair barriers and harnessing beneficial differentiation processes in MSCs could widen options of autologous MSC application in SSc patients.
Collapse
MESH Headings
- Adult
- Aged
- Becaplermin
- Biomarkers/metabolism
- Cell Differentiation/drug effects
- Cell Proliferation
- Cells, Cultured
- Connective Tissue Growth Factor/pharmacology
- Female
- Fibroblast Growth Factor 2/pharmacology
- Gene Expression Regulation
- Humans
- Male
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/drug effects
- Middle Aged
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myofibroblasts/cytology
- Myofibroblasts/drug effects
- Myofibroblasts/metabolism
- Proto-Oncogene Proteins c-sis/pharmacology
- Scleroderma, Systemic/genetics
- Scleroderma, Systemic/metabolism
- Scleroderma, Systemic/pathology
- Signal Transduction/drug effects
- Transforming Growth Factor beta1/pharmacology
Collapse
Affiliation(s)
- Björn Hegner
- Clinic for Nephrology and Intensive Care Medicine, Charité University Hospital, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Berlin, Germany
- Center for Cardiovascular Research (CCR), Charitè University Hospital, Berlin, Germany
- * E-mail:
| | - Theres Schaub
- Clinic for Nephrology and Intensive Care Medicine, Charité University Hospital, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Berlin, Germany
- Center for Cardiovascular Research (CCR), Charitè University Hospital, Berlin, Germany
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Rusan Catar
- Clinic for Nephrology and Intensive Care Medicine, Charité University Hospital, Berlin, Germany
- Center for Cardiovascular Research (CCR), Charitè University Hospital, Berlin, Germany
| | - Angelika Kusch
- Clinic for Nephrology and Intensive Care Medicine, Charité University Hospital, Berlin, Germany
- Center for Cardiovascular Research (CCR), Charitè University Hospital, Berlin, Germany
| | - Philine Wagner
- Clinic for Nephrology and Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Kirill Essin
- Experimental and Clinical Research Center, Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Claudia Lange
- Clinic for Stem Cell Transplantation, Department of Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriela Riemekasten
- Clinic for Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany
| | - Duska Dragun
- Clinic for Nephrology and Intensive Care Medicine, Charité University Hospital, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Berlin, Germany
- Center for Cardiovascular Research (CCR), Charitè University Hospital, Berlin, Germany
| |
Collapse
|
110
|
Gomez IG, Roach AM, Nakagawa N, Amatucci A, Johnson BG, Dunn K, Kelly MC, Karaca G, Zheng TS, Szak S, Peppiatt-Wildman CM, Burkly LC, Duffield JS. TWEAK-Fn14 Signaling Activates Myofibroblasts to Drive Progression of Fibrotic Kidney Disease. J Am Soc Nephrol 2016; 27:3639-3652. [PMID: 27026366 DOI: 10.1681/asn.2015111227] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/16/2016] [Indexed: 01/15/2023] Open
Abstract
The identification of the cellular origins of myofibroblasts has led to the discovery of novel pathways that potentially drive myofibroblast perpetuation in disease. Here, we further investigated the role of innate immune signaling pathways in this process. In mice, renal injury-induced activation of pericytes, which are myofibroblast precursors attached to endothelial cells, led to upregulated expression of TNF receptor superfamily member 12a, also known as fibroblast growth factor-inducible 14 (Fn14), by these cells. In live rat kidney slices, administration of the Fn14 ligand, TNF-related weak inducer of apoptosis (TWEAK), promoted pericyte-dependent vasoconstriction followed by pericyte detachment from capillaries. In vitro, administration of TWEAK activated and differentiated pericytes into cytokine-producing myofibroblasts, and further activated established myofibroblasts in a manner requiring canonical and noncanonical NF-κB signaling pathways. Deficiency of Fn14 protected mouse kidneys from fibrogenesis, inflammation, and associated vascular instability after in vivo injury, and was associated with loss of NF-κB signaling. In a genetic model of spontaneous CKD, therapeutic delivery of anti-TWEAK blocking antibodies attenuated disease progression, preserved organ function, and increased survival. These results identify the TWEAK-Fn14 signaling pathway as an important factor in myofibroblast perpetuation, fibrogenesis, and chronic disease progression.
Collapse
Affiliation(s)
- Ivan G Gomez
- Research & Development, Biogen, Cambridge, Massachusetts.,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Allie M Roach
- Research & Development, Biogen, Cambridge, Massachusetts.,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Naoki Nakagawa
- Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Aldo Amatucci
- Research & Development, Biogen, Cambridge, Massachusetts
| | - Bryce G Johnson
- Research & Development, Biogen, Cambridge, Massachusetts.,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Kadeshia Dunn
- Medway School of Pharmacy, University of Kent, Chatham, Kent, United Kingdom
| | - Mark C Kelly
- Medway School of Pharmacy, University of Kent, Chatham, Kent, United Kingdom
| | - Gamze Karaca
- Research & Development, Biogen, Cambridge, Massachusetts
| | | | - Suzanne Szak
- Research & Development, Biogen, Cambridge, Massachusetts
| | | | - Linda C Burkly
- Research & Development, Biogen, Cambridge, Massachusetts;
| | - Jeremy S Duffield
- Research & Development, Biogen, Cambridge, Massachusetts; .,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| |
Collapse
|
111
|
Tremolada C, Ricordi C, Caplan AI, Ventura C. Mesenchymal Stem Cells in Lipogems, a Reverse Story: from Clinical Practice to Basic Science. Methods Mol Biol 2016; 1416:109-122. [PMID: 27236668 DOI: 10.1007/978-1-4939-3584-0_6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The idea that basic science should be the starting point for modern clinical approaches has been consolidated over the years, and emerged as the cornerstone of Molecular Medicine. Nevertheless, there is increasing concern over the low efficiency and inherent costs related to the translation of achievements from the bench to the bedside. These burdens are also perceived with respect to the effectiveness of translating basic discoveries in stem cell biology to the newly developing field of advanced cell therapy or Regenerative Medicine. As an alternative paradigm, past and recent history in Medical Science provides remarkable reverse stories in which clinical observations at the patient's bedside have fed major advances in basic research which, in turn, led to consistent progression in clinical practice. Within this context, we discuss our recently developed method and device, which forms the core of a system (Lipogems) for processing of human adipose tissue solely with the aid of mild mechanical forces to yield a microfractured tissue product.
Collapse
Affiliation(s)
| | - Camillo Ricordi
- Cell Transplant Program and Diabetes Research Institute, University of Miami, Miami, FL, USA
| | - Arnold I Caplan
- Skeletal Research Center, Case Western Reserve University, Cleveland, OH, USA
| | - Carlo Ventura
- SWITH (Stem Wave Institute for Tissue Healing), Gruppo Villa Maria (GVM) and Ettore Sansavini Health Science Foundation - ONLUS, Lugo (Ravenna), Italy.
- National Institute of Biostructures and Biosystems (NIBB) at the S. Orsola - Malpighi Hospital, Institute of Cardiology, University of Bologna, Pavilion 21, Via Massarenti N. 9, 40138, Bologna, Italy.
| |
Collapse
|
112
|
Turley SJ, Cremasco V, Astarita JL. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol 2015; 15:669-82. [PMID: 26471778 DOI: 10.1038/nri3902] [Citation(s) in RCA: 801] [Impact Index Per Article: 80.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A dynamic and mutualistic interaction between tumour cells and the surrounding stroma promotes the initiation, progression, metastasis and chemoresistance of solid tumours. Far less understood is the relationship between the stroma and tumour-infiltrating leukocytes; however, emerging evidence suggests that the stromal compartment can shape antitumour immunity and responsiveness to immunotherapy. Thus, there is growing interest in elucidating the immunomodulatory roles of the stroma that evolve within the tumour microenvironment. In this Review, we discuss the evidence that stromal determinants interact with leukocytes and influence antitumour immunity, with emphasis on the immunological attributes of stromal cells that may foster their protumorigenic function.
Collapse
Affiliation(s)
- Shannon J Turley
- Department of Cancer Immunology, Genentech, 1 DNA Way, South San Francisco, California 94080, USA
| | - Viviana Cremasco
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA.,Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Jillian L Astarita
- Department of Cancer Immunology, Genentech, 1 DNA Way, South San Francisco, California 94080, USA
| |
Collapse
|
113
|
Peng T, Frank DB, Kadzik RS, Morley MP, Rathi KS, Wang T, Zhou S, Cheng L, Lu MM, Morrisey EE. Hedgehog actively maintains adult lung quiescence and regulates repair and regeneration. Nature 2015; 526:578-82. [PMID: 26436454 PMCID: PMC4713039 DOI: 10.1038/nature14984] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 07/28/2015] [Indexed: 12/23/2022]
Abstract
Postnatal tissue quiescence is thought to be a default state in the absence of a proliferative stimulus such as injury. Previous studies have demonstrated that certain embryonic development programs are reactivated aberrantly in adult organs to drive repair and regeneration1–3, it is not well understood how quiescence is maintained in organs such as the lung which displays a remarkably low level of cellular turnover4,5. We now demonstrate that quiescence in the adult lung is an actively maintained state and is regulated by hedgehog signaling. Epithelial-specific deletion of sonic hedgehog during postnatal homeostasis in the lung results in a proliferative expansion of the adjacent lung mesenchyme. Hedgehog signaling is initially down-regulated during the acute phase of epithelial injury as the mesenchyme proliferates in response, but returns to baseline during injury resolution as quiescence is restored. Activation of hedgehog during acute epithelial injury attenuates the proliferative expansion of the lung mesenchyme, whereas inactivation of hedgehog signaling prevents the restoration of quiescence during injury resolution. Finally, we show that hedgehog also regulates epithelial quiescence and regeneration in response to injury via a mesenchymal feedback mechanism. These results demonstrate that epithelial-mesenchymal interactions coordinated by hedgehog actively maintains postnatal tissue homeostasis, and deregulation of hedgehog during injury leads to aberrant repair and regeneration in the lung.
Collapse
Affiliation(s)
- Tien Peng
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - David B Frank
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Rachel S Kadzik
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Michael P Morley
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Komal S Rathi
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Tao Wang
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Su Zhou
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Lan Cheng
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Min Min Lu
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Edward E Morrisey
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
114
|
Hong J, Tobin NP, Rundqvist H, Li T, Lavergne M, García-Ibáñez Y, Qin H, Paulsson J, Zeitelhofer M, Adzemovic MZ, Nilsson I, Roswall P, Hartman J, Johnson RS, Östman A, Bergh J, Poljakovic M, Genové G. Role of Tumor Pericytes in the Recruitment of Myeloid-Derived Suppressor Cells. J Natl Cancer Inst 2015; 107:djv209. [PMID: 26296362 DOI: 10.1093/jnci/djv209] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/06/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Pericytes are members of the tumor stroma; however, little is known about their origin, function, or interaction with other tumor components. Emerging evidence suggest that pericytes may regulate leukocyte transmigration. Myeloid-derived suppressor cells (MDSC) are immature myeloid cells with powerful inhibitory effects on T-cell-mediated antitumor reactivity. METHODS We generated subcutaneous tumors in a genetic mouse model of pericyte deficiency (the pdgfb (ret/ret) mouse) and littermate control mice (n = 6-25). Gene expression profiles from 253 breast cancer patients (stage I-III) were evaluated for clinic-pathological parameters and survival using Cox proportional hazard ratios (HRs) and 95% confidence intervals (CIs) based on a two-sided Wald test. RESULTS We report that pericyte deficiency leads to increased transmigration of Gr1(+)/CD11b(+) cells in experimentally induced tumors. Pericyte deficiency produced defective tumor vasculature, resulting in a more hypoxic microenvironment promoting IL-6 upregulation in the malignant cells. Silencing IL-6 expression in tumor cells attenuated the observed differences in MDSC transmigration. Restoring the pericyte coverage in tumors abrogated the increased MDSC trafficking to pericyte-deficient tumors. MDSC accumulation in tumors led to increases in tumor growth and in circulating malignant cells. Finally, gene expression analysis from human breast cancer patients revealed increased expression of the human MDSC markers CD33 and S100A9 with concomitant decreased expression of pericyte genes and was associated with poor prognosis (HR = 1.88, 95% CI = 1.08 to 3.25, P = .03). CONCLUSIONS Our data uncovers a novel paracrine interaction between tumor pericytes and inflammatory cells and delineates the cellular events resulting in the recruitment of MDSC to tumors. Furthermore, we propose for the first time a role for tumor pericytes in modulating the expression of immune mediators in malignant cells by promoting a hypoxic microenvironment.
Collapse
Affiliation(s)
- JongWook Hong
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Nicholas P Tobin
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Helene Rundqvist
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Tian Li
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Marion Lavergne
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Yaiza García-Ibáñez
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Hanyu Qin
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Janna Paulsson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Manuel Zeitelhofer
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Milena Z Adzemovic
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Ingrid Nilsson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Pernilla Roswall
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Johan Hartman
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Randall S Johnson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Arne Östman
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Jonas Bergh
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Mirjana Poljakovic
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ)
| | - Guillem Genové
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden (JWH, TL, ML, YGI, HQ, MZ, MZA, IN, PR, GG); Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska University Hospital, Solna, Sweden (NPT, JP, JH, AÖ, JB); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (HR, RSJ); Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK (RSJ); Department of Molecular Medicine and Surgery, Urology Laboratory, Karolinska Institute, Stockholm, Sweden (MP); Department of Urology, Karolinska University Hospital, Solna, Sweden (MP).Current affiliation: Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland (HQ).
| |
Collapse
|
115
|
PDGFRβ(+) cells in human and experimental neuro-vascular dysplasia and seizures. Neuroscience 2015; 306:18-27. [PMID: 26283024 DOI: 10.1016/j.neuroscience.2015.07.090] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/19/2015] [Accepted: 07/23/2015] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Neuro-vascular rearrangement occurs in brain disorders, including epilepsy. Platelet-derived growth factor receptor beta (PDGFRβ) is used as a marker of perivascular pericytes. Whether PDGFRβ(+) cell reorganization occurs in regions of neuro-vascular dysplasia associated with seizures is unknown. METHODS We used brain specimens derived from epileptic subjects affected by intractable seizures associated with focal cortical dysplasia (FCD) or temporal lobe epilepsy with hippocampal sclerosis (TLE-HS). Tissues from cryptogenic epilepsy, non-sclerotic hippocampi or peritumoral were used for comparison. An in vivo rat model of neuro-vascular dysplasia was obtained by pre-natal exposure to methyl-axozy methanoic acid (MAM). Status epilepticus (SE) was induced in adult MAM rats by intraperitoneal pilocarpine. MAM tissues were also used to establish organotypic hippocampal cultures (OHC) to further assess pericytes positioning at the dysplastic microvasculature. PDGFRβ and its colocalization with RECA-1 or CD34 were used to segregate perivascular pericytes. PDGFRβ and NG2 or IBA1 colocalization were performed. Rat cortices and hippocampi were used for PDGFRβ western blot analysis. RESULTS Human FCD displayed the highest perivascular PDGFRβ immunoreactivity, indicating pericytes, and presence of ramified PDGFRβ(+) cells in the parenchyma and proximal to microvessels. Tissues deriving from human cryptogenic epilepsy displayed a similar pattern of immunoreactivity, although to a lesser extent compared to FCD. In TLE-HS, CD34 vascular proliferation was paralleled by increased perivascular PDGFRβ(+) pericytes, as compared to non-HS. Parenchymal PDGFRβ immunoreactivity co-localized with NG2 but was distinct from IBA1(+) microglia. In MAM rats, we found pericyte-vascular changes in regions characterized by neuronal heterotopias. PDGFRβ immunoreactivity was differentially distributed in the heterotopic and adjacent normal CA1 region. The use of MAM OHC revealed microvascular-pericyte dysplasia at the capillary tree lining the dentate gyrus (DG) molecular layer as compared to control OHC. Severe SE induced PDGFRβ(+) immunoreactivity mostly in the CA1 region of MAM rats. CONCLUSION Our descriptive study points to microvascular-pericyte changes in the epileptic pathology. The possible link between PDGFRβ(+) cells, neuro-vascular dysplasia and remodeling during seizures is discussed.
Collapse
|
116
|
Fernández-Klett F, Priller J. The fibrotic scar in neurological disorders. Brain Pathol 2015; 24:404-13. [PMID: 24946078 DOI: 10.1111/bpa.12162] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 05/26/2014] [Indexed: 01/18/2023] Open
Abstract
Tissue fibrosis, or scar formation, is a common response to damage in most organs of the body. The central nervous system (CNS) is special in that fibrogenic cells are restricted to vascular and meningeal niches. However, disruption of the blood-brain barrier and inflammation can unleash stromal cells and trigger scar formation. Astroglia segregate from the inflammatory lesion core, and the so-called "glial scar" composed of hypertrophic astrocytes seals off the intact neural tissue from damage. In the lesion core, a second type of "fibrotic scar" develops, which is sensitive to inflammatory mediators. Genetic fate mapping studies suggest that pericytes and perivascular fibroblasts are activated, but other precursor cells may also be involved in generating a transient fibrous extracellular matrix in the CNS. The stromal cells sense inflammation and attract immune cells, which in turn drive myofibroblast transdifferentiation. We believe that the fibrotic scar represents a major barrier to CNS regeneration. Targeting of fibrosis may therefore prove to be a valuable therapeutic strategy for neurological disorders such as stroke, spinal cord injury and multiple sclerosis.
Collapse
Affiliation(s)
- Francisco Fernández-Klett
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
117
|
Betsholtz C, Keller A. PDGF, pericytes and the pathogenesis of idiopathic basal ganglia calcification (IBGC). Brain Pathol 2015; 24:387-95. [PMID: 24946076 DOI: 10.1111/bpa.12158] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 05/13/2014] [Indexed: 01/09/2023] Open
Abstract
Platelet-derived growth factors (PDGFs) are important mitogens for various types of mesenchymal cells, and as such, they exert critical functions during organogenesis in mammalian embryonic and early postnatal development. Increased or ectopic PDGF activity may also cause or contribute to diseases such as cancer and tissue fibrosis. Until recently, no loss-of-function (LOF) mutations in PDGF or PDGF receptor genes were reported as causally linked to a human disease. This changed in 2013 when reports appeared on presumed LOF mutations in the genes encoding PDGF-B and its receptor PDGF receptor-beta (PDGF-Rβ) in familial idiopathic basal ganglia calcification (IBGC), a brain disease characterized by anatomically localized calcifications in or near the blood microvessels. Here, we review PDGF-B and PDGF-Rβ biology with special reference to their functions in brain-blood vessel development, pericyte recruitment and the regulation of the blood-brain barrier. We also discuss various scenarios for IBGC pathogenesis suggested by observations in patients and genetically engineered animal models of the disease.
Collapse
Affiliation(s)
- Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
118
|
PDGFRβ signalling regulates local inflammation and synergizes with hypercholesterolaemia to promote atherosclerosis. Nat Commun 2015; 6:7770. [PMID: 26183159 PMCID: PMC4507293 DOI: 10.1038/ncomms8770] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 06/05/2015] [Indexed: 02/07/2023] Open
Abstract
Platelet-derived growth factor (PDGF) is a mitogen and chemoattractant for vascular smooth muscle cells (VSMCs). However, the direct effects of PDGF receptor β (PDGFRβ) activation on VSMCs have not been studied in the context of atherosclerosis. Here, we present a new mouse model of atherosclerosis with an activating mutation in PDGFRβ. Increased PDGFRβ signaling induces chemokine secretion and leads to leukocyte accumulation in the adventitia and media of the aorta. Furthermore, PDGFRβD849V amplifies and accelerates atherosclerosis in hypercholesterolemic ApoE−/− or Ldlr−/− mice. Intriguingly, increased PDGFRβ signaling promotes advanced plaque formation at novel sites in the thoracic aorta and coronary arteries. However, deletion of the PDGFRβ-activated transcription factor STAT1 in VSMCs alleviates inflammation of the arterial wall and reduces plaque burden. These results demonstrate that PDGFRβ pathway activation has a profound effect on vascular disease and support the conclusion that inflammation in the outer arterial layers is a driving process for atherosclerosis.
Collapse
|
119
|
Hurtado R, Zewdu R, Mtui J, Liang C, Aho R, Kurylo C, Selleri L, Herzlinger D. Pbx1-dependent control of VMC differentiation kinetics underlies gross renal vascular patterning. Development 2015; 142:2653-64. [PMID: 26138478 DOI: 10.1242/dev.124776] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/09/2015] [Indexed: 12/29/2022]
Abstract
The architecture of an organ's vascular bed subserves its physiological function and metabolic demands. However, the mechanisms underlying gross vascular patterning remain elusive. Using intravital dye labeling and 3D imaging, we discovered that systems-level vascular patterning in the kidney is dependent on the kinetics of vascular mural cell (VMC) differentiation. Conditional ablation of the TALE transcription factor Pbx1 in renal VMC progenitors in the mouse led to the premature upregulation of PDGFRβ, a master initiator of VMC-blood vessel association. This precocious VMC differentiation resulted in nonproductive angiogenesis, abnormal renal arterial tree patterning and neonatal death consistent with kidney dysfunction. Notably, we establish that Pbx1 directly represses Pdgfrb, and demonstrate that decreased Pdgfrb dosage in conditional Pbx1 mutants substantially rescues vascular patterning defects and neonatal survival. These findings identify, for the first time, an in vivo transcriptional regulator of PDGFRβ, and reveal a previously unappreciated role for VMCs in systems-level vascular patterning.
Collapse
Affiliation(s)
- Romulo Hurtado
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Rediet Zewdu
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - James Mtui
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Cindy Liang
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Robert Aho
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Chad Kurylo
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Licia Selleri
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Doris Herzlinger
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
120
|
Luzina IG, Todd NW, Sundararajan S, Atamas SP. The cytokines of pulmonary fibrosis: Much learned, much more to learn. Cytokine 2015; 74:88-100. [DOI: 10.1016/j.cyto.2014.11.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Revised: 11/09/2014] [Accepted: 11/10/2014] [Indexed: 02/07/2023]
|
121
|
Kocabayoglu P, Lade A, Lee YA, Dragomir AC, Sun X, Fiel MI, Thung S, Aloman C, Soriano P, Hoshida Y, Friedman SL. β-PDGF receptor expressed by hepatic stellate cells regulates fibrosis in murine liver injury, but not carcinogenesis. J Hepatol 2015; 63:141-7. [PMID: 25678385 PMCID: PMC4475471 DOI: 10.1016/j.jhep.2015.01.036] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 01/09/2015] [Accepted: 01/27/2015] [Indexed: 01/27/2023]
Abstract
BACKGROUND & AIMS Rapid induction of β-PDGF receptor (β-PDGFR) is a core feature of hepatic stellate cell activation, but its cellular impact in vivo is not well characterized. We explored the contribution of β-PDGFR-mediated pathway activation to hepatic stellate cell responses in liver injury, fibrogenesis, and carcinogenesis in vivo using genetic models with divergent β-PDGFR activity, and assessed its prognostic implications in human cirrhosis. METHODS The impact of either loss or constitutive activation of β-PDGFR in stellate cells on fibrosis was assessed following carbon tetrachloride (CCl4) or bile duct ligation. Hepatocarcinogenesis in fibrotic liver was tracked after a single dose of diethylnitrosamine (DEN) followed by repeated injections of CCl4. Genome-wide expression profiling was performed from isolated stellate cells that expressed or lacked β-PDGFR to determine deregulated pathways and evaluate their association with prognostic gene signatures in human cirrhosis. RESULTS Depletion of β-PDGFR in hepatic stellate cells decreased injury and fibrosis in vivo, while its auto-activation accelerated fibrosis. However, there was no difference in development of DEN-induced pre-neoplastic foci. Genomic profiling revealed ERK, AKT, and NF-κB pathways and a subset of a previously identified 186-gene prognostic signature in hepatitis C virus (HCV)-related cirrhosis as downstream of β-PDGFR in stellate cells. In the human cohort, the β-PDGFR signature was not associated with HCC development, but was significantly associated with a poorer outcome in HCV cirrhosis. CONCLUSIONS β-PDGFR is a key mediator of hepatic injury and fibrogenesis in vivo and contributes to the poor prognosis of human cirrhosis, but not by increasing HCC development.
Collapse
Affiliation(s)
- Peri Kocabayoglu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of General, Visceral and Transplant Surgery, University Hospital Essen, Germany
| | - Abigale Lade
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Youngmin A. Lee
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ana-Cristina Dragomir
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xiaochen Sun
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M. Isabel Fiel
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Swan Thung
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Costica Aloman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philippe Soriano
- Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Scott L. Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
122
|
Abstract
Chronic rejection of transplanted organs remains the main obstacle in the long-term success of organ transplantation. Thus, there is a persistent quest for development of antichronic rejection therapies and identification of novel molecular and cellular targets. One of the potential targets is the pericytes, the mural cells of microvessels, which regulate microvascular permeability, development, and maturation by controlling endothelial cell functions and regulating tissue fibrosis and inflammatory response. In this review, we discuss the potential of targeting pericytes in the development of microvasular dysfunction and the molecular pathways involved in regulation of pericyte activities for antichronic rejection intervention.
Collapse
|
123
|
Iwayama T, Steele C, Yao L, Dozmorov MG, Karamichos D, Wren JD, Olson LE. PDGFRα signaling drives adipose tissue fibrosis by targeting progenitor cell plasticity. Genes Dev 2015; 29:1106-19. [PMID: 26019175 PMCID: PMC4470280 DOI: 10.1101/gad.260554.115] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/07/2015] [Indexed: 12/19/2022]
Abstract
Adipose tissue fibrosis occurs during obesity and is associated with metabolic dysfunction. Iwayama et al. identify perivascular cells as fibro/adipogenic progenitors in white adipose tissue and show that PDGFRα targets progenitor cell plasticity as a profibrotic mechanism. Fibrosis is a common disease process in which profibrotic cells disturb organ function by secreting disorganized extracellular matrix (ECM). Adipose tissue fibrosis occurs during obesity and is associated with metabolic dysfunction, but how profibrotic cells originate is still being elucidated. Here, we use a developmental model to investigate perivascular cells in white adipose tissue (WAT) and their potential to cause organ fibrosis. We show that a Nestin-Cre transgene targets perivascular cells (adventitial cells and pericyte-like cells) in WAT, and Nestin-GFP specifically labels pericyte-like cells. Activation of PDGFRα signaling in perivascular cells causes them to transition into ECM-synthesizing profibrotic cells. Before this transition occurs, PDGFRα signaling up-regulates mTOR signaling and ribosome biogenesis pathways and perturbs the expression of a network of epigenetically imprinted genes that have been implicated in cell growth and tissue homeostasis. Isolated Nestin-GFP+ cells differentiate into adipocytes ex vivo and form WAT when transplanted into recipient mice. However, PDGFRα signaling opposes adipogenesis and generates profibrotic cells instead, which leads to fibrotic WAT in transplant experiments. These results identify perivascular cells as fibro/adipogenic progenitors in WAT and show that PDGFRα targets progenitor cell plasticity as a profibrotic mechanism.
Collapse
Affiliation(s)
- Tomoaki Iwayama
- Immunobiology and Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Cameron Steele
- Immunobiology and Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Longbiao Yao
- Immunobiology and Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Mikhail G Dozmorov
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA; Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - Dimitris Karamichos
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA; Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Lorin E Olson
- Immunobiology and Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA;
| |
Collapse
|
124
|
Combined effects of pericytes in the tumor microenvironment. Stem Cells Int 2015; 2015:868475. [PMID: 26000022 PMCID: PMC4427118 DOI: 10.1155/2015/868475] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/28/2015] [Indexed: 12/25/2022] Open
Abstract
Pericytes are multipotent perivascular cells whose involvement in vasculature development is well established. Evidences in the literature also suggest that pericytes display immune properties and that these cells may serve as an in vivo reservoir of stem cells, contributing to the regeneration of diverse tissues. Pericytes are also capable of tumor homing and are important cellular components of the tumor microenvironment (TME). In this review, we highlight the contribution of pericytes to some classical hallmarks of cancer, namely, tumor angiogenesis, growth, metastasis, and evasion of immune destruction, and discuss how collectively these hallmarks could be tackled by therapies targeting pericytes, providing a rationale for cancer drugs aiming at the TME.
Collapse
|
125
|
Grimminger F, Günther A, Vancheri C. The role of tyrosine kinases in the pathogenesis of idiopathic pulmonary fibrosis. Eur Respir J 2015; 45:1426-33. [PMID: 25745048 DOI: 10.1183/09031936.00149614] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 12/06/2014] [Indexed: 12/23/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with a median survival time from diagnosis of 2-3 years. Although the pathogenic pathways have not been fully elucidated, IPF is believed to be caused by persistent epithelial injury in genetically susceptible individuals. Tyrosine kinases are involved in a range of signalling pathways that are essential for cellular homeostasis. However, there is substantial evidence from in vitro studies and animal models that receptor tyrosine kinases, such as the platelet-derived growth factor receptor, vascular endothelial growth factor receptor and fibroblast growth factor receptor, and non-receptor tyrosine kinases, such as the Src family, play critical roles in the pathogenesis of pulmonary fibrosis. For example, the expression and release of tyrosine kinases are altered in patients with IPF, while specific tyrosine kinases stimulate the proliferation of lung fibroblasts in vitro. Agents that inhibit tyrosine kinases have shown anti-fibrotic and anti-inflammatory effects in animal models of pulmonary fibrosis. Recently, the tyrosine kinase inhibitor nintedanib has shown positive results in two phase III trials in patients with IPF. Here, we summarise the evidence for involvement of specific tyrosine kinases in the pathogenesis of IPF and the development of tyrosine kinase inhibitors as treatments for IPF.
Collapse
Affiliation(s)
- Friedrich Grimminger
- Dept of Hematology/Oncology, University Hospital of Giessen and Marburg, Marburg, Germany
| | - Andreas Günther
- Dept of Pulmonary and Critical Care Medicine, ILD Program, University Hospital of Giessen and Marburg, Marburg, Germany
| | - Carlo Vancheri
- "Regional Centre for Rare Lung Diseases", Dept of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
126
|
Experimentally-derived fibroblast gene signatures identify molecular pathways associated with distinct subsets of systemic sclerosis patients in three independent cohorts. PLoS One 2015; 10:e0114017. [PMID: 25607805 PMCID: PMC4301872 DOI: 10.1371/journal.pone.0114017] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 11/04/2014] [Indexed: 12/19/2022] Open
Abstract
Genome-wide expression profiling in systemic sclerosis (SSc) has identified four ‘intrinsic’ subsets of disease (fibroproliferative, inflammatory, limited, and normal-like), each of which shows deregulation of distinct signaling pathways; however, the full set of pathways contributing to this differential gene expression has not been fully elucidated. Here we examine experimentally derived gene expression signatures in dermal fibroblasts for thirteen different signaling pathways implicated in SSc pathogenesis. These data show distinct and overlapping sets of genes induced by each pathway, allowing for a better understanding of the molecular relationship between profibrotic and immune signaling networks. Pathway-specific gene signatures were analyzed across a compendium of microarray datasets consisting of skin biopsies from three independent cohorts representing 80 SSc patients, 4 morphea, and 26 controls. IFNα signaling showed a strong association with early disease, while TGFβ signaling spanned the fibroproliferative and inflammatory subsets, was associated with worse MRSS, and was higher in lesional than non-lesional skin. The fibroproliferative subset was most strongly associated with PDGF signaling, while the inflammatory subset demonstrated strong activation of innate immune pathways including TLR signaling upstream of NF-κB. The limited and normal-like subsets did not show associations with fibrotic and inflammatory mediators such as TGFβ and TNFα. The normal-like subset showed high expression of genes associated with lipid signaling, which was absent in the inflammatory and limited subsets. Together, these data suggest a model by which IFNα is involved in early disease pathology, and disease severity is associated with active TGFβ signaling.
Collapse
|
127
|
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: 177] [Impact Index Per Article: 17.7] [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
|
128
|
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: 211] [Impact Index Per Article: 19.2] [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
|
129
|
Chung CG, James AW, Asatrian G, Chang L, Nguyen A, Le K, Bayani G, Lee R, Stoker D, Zhang X, Ting K, Péault B, Soo C. Human perivascular stem cell-based bone graft substitute induces rat spinal fusion. Stem Cells Transl Med 2014; 3:1231-41. [PMID: 25154782 DOI: 10.5966/sctm.2014-0027] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Adipose tissue is an attractive source of mesenchymal stem cells (MSCs) because of its abundance and accessibility. We have previously defined a population of native MSCs termed perivascular stem cells (PSCs), purified from diverse human tissues, including adipose tissue. Human PSCs (hPSCs) are a bipartite cell population composed of pericytes (CD146+CD34-CD45-) and adventitial cells (CD146-CD34+CD45-), isolated by fluorescence-activated cell sorting and with properties identical to those of culture identified MSCs. Our previous studies showed that hPSCs exhibit improved bone formation compared with a sample-matched unpurified population (termed stromal vascular fraction); however, it is not known whether hPSCs would be efficacious in a spinal fusion model. To investigate, we evaluated the osteogenic potential of freshly sorted hPSCs without culture expansion and differentiation in a rat model of posterolateral lumbar spinal fusion. We compared increasing dosages of implanted hPSCs to assess for dose-dependent efficacy. All hPSC treatment groups induced successful spinal fusion, assessed by manual palpation and microcomputed tomography. Computerized biomechanical simulation (finite element analysis) further demonstrated bone fusion with hPSC treatment. Histological analyses showed robust endochondral ossification in hPSC-treated samples. Finally, we confirmed that implanted hPSCs indeed differentiated into osteoblasts and osteocytes; however, the majority of the new bone formation was of host origin. These results suggest that implanted hPSCs positively regulate bone formation via direct and paracrine mechanisms. In summary, hPSCs are a readily available MSC population that effectively forms bone without requirements for culture or predifferentiation. Thus, hPSC-based products show promise for future efforts in clinical bone regeneration and repair.
Collapse
Affiliation(s)
- Choon G Chung
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Greg Asatrian
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Le Chang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Alan Nguyen
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Khoi Le
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Georgina Bayani
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert Lee
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - David Stoker
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Kang Ting
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bruno Péault
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Chia Soo
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
130
|
Hudak CS, Gulyaeva O, Wang Y, Park SM, Lee L, Kang C, Sul HS. Pref-1 marks very early mesenchymal precursors required for adipose tissue development and expansion. Cell Rep 2014; 8:678-87. [PMID: 25088414 DOI: 10.1016/j.celrep.2014.06.060] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 05/07/2014] [Accepted: 06/26/2014] [Indexed: 11/19/2022] Open
Abstract
Pref-1 is an EGF-repeat-containing protein that inhibits adipocyte differentiation. To better understand the origin and development of white adipose tissue (WAT), we generated transgenic mouse models for transient or permanent fluorescent labeling of cells using the Pref-1 promoter, facilitating inducible ablation. We show that Pref-1-marked cells retain proliferative capacity and are very early adipose precursors, prior to expression of Zfp423 or PPARγ. In addition, the Pref-1-marked cells establish that adipose precursors are mesenchymal, but not endothelial or pericytal, in origin. During embryogenesis, Pref-1-marked cells first appear in the dorsal mesenteric region as early as embryonic day 10.5 (E10.5). These cells become lipid-laden adipocytes at E17.5 in the subcutaneous region, whereas visceral WAT develops after birth. Finally, ablation of Pref-1-marked cells prevents not only embryonic WAT development but also later adult adipose expansion upon high-fat feeding, demonstrating the requirement of Pref-1 cells for adipogenesis.
Collapse
Affiliation(s)
- Carolyn S Hudak
- Department of Nutritional Sciences & Toxicology, 119 Morgan Hall, University of California, Berkeley, CA 94720, USA
| | - Olga Gulyaeva
- Endocrinology Program, 299 LSA, University of California, Berkeley, CA 94720, USA
| | - Yuhui Wang
- Department of Nutritional Sciences & Toxicology, 119 Morgan Hall, University of California, Berkeley, CA 94720, USA
| | - Seung-Min Park
- Department of Bioengineering, 306 Stanley Hall, University of California, Berkeley, CA 94720 USA; Berkeley Sensor and Actuator Center, 403 Cory Hall, University of California, Berkeley, CA 94720, USA; Department of Radiology, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Luke Lee
- Department of Bioengineering, 306 Stanley Hall, University of California, Berkeley, CA 94720 USA
| | - Chulho Kang
- Department of Molecular & Cell Biology, 142 LSA, University of California, Berkeley, CA 94720, USA
| | - Hei Sook Sul
- Department of Nutritional Sciences & Toxicology, 119 Morgan Hall, University of California, Berkeley, CA 94720, USA; Endocrinology Program, 299 LSA, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
131
|
Sui Y, Park SH, Xu J, Monette S, Helsley RN, Han SS, Zhou C. IKKβ links vascular inflammation to obesity and atherosclerosis. ACTA ACUST UNITED AC 2014; 211:869-86. [PMID: 24799533 PMCID: PMC4010900 DOI: 10.1084/jem.20131281] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
IKKβ functions in smooth muscle cells to regulate vascular inflammatory responses and atherosclerosis development. IκB kinase β (IKKβ), a central coordinator of inflammatory responses through activation of NF-κB, has been implicated in vascular pathologies, but its role in atherogenesis remains elusive. Here, we demonstrate that IKKβ functions in smooth muscle cells (SMCs) to regulate vascular inflammatory responses and atherosclerosis development. IKKβ deficiency in SMCs driven by a SM22Cre-IKKβ-flox system rendered low density lipoprotein receptor-null mice resistant to vascular inflammation and atherosclerosis induced by high-fat feeding. Unexpectedly, IKKβ-deficient mice were also resistant to diet-induced obesity and metabolic disorders. Cell lineage analysis revealed that SM22Cre is active in primary adipose stromal vascular cells and deficiency of IKKβ diminished the ability of these cells to differentiate, leading to accumulation of adipocyte precursor cells in adipose tissue. Mechanistically, reduction of IKKβ expression or pharmacological inhibition of IKKβ inhibited proteasome-mediated β-catenin ubiquitination and degradation in murine preadipocytes, resulting in elevated β-catenin levels and impaired adipocyte differentiation. Further, chronic treatment of mice with a potent IKKβ inhibitor decreased adipogenesis and ameliorated diet-induced obesity. Our findings demonstrate a pivotal role of IKKβ in linking vascular inflammation to atherosclerosis and adipose tissue development, and provide evidence for using appropriate IKKβ inhibitors in the treatment of obesity and metabolic disorders.
Collapse
Affiliation(s)
- Yipeng Sui
- Graduate Center for Nutritional Sciences, 2 Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536
| | | | | | | | | | | | | |
Collapse
|
132
|
Seidelmann SB, Lighthouse JK, Greif DM. Development and pathologies of the arterial wall. Cell Mol Life Sci 2014; 71:1977-99. [PMID: 24071897 PMCID: PMC11113178 DOI: 10.1007/s00018-013-1478-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 01/13/2023]
Abstract
Arteries consist of an inner single layer of endothelial cells surrounded by layers of smooth muscle and an outer adventitia. The majority of vascular developmental studies focus on the construction of endothelial networks through the process of angiogenesis. Although many devastating vascular diseases involve abnormalities in components of the smooth muscle and adventitia (i.e., the vascular wall), the morphogenesis of these layers has received relatively less attention. Here, we briefly review key elements underlying endothelial layer formation and then focus on vascular wall development, specifically on smooth muscle cell origins and differentiation, patterning of the vascular wall, and the role of extracellular matrix and adventitial progenitor cells. Finally, we discuss select human diseases characterized by marked vascular wall abnormalities. We propose that continuing to apply approaches from developmental biology to the study of vascular disease will stimulate important advancements in elucidating disease mechanism and devising novel therapeutic strategies.
Collapse
MESH Headings
- Angiogenic Proteins/genetics
- Angiogenic Proteins/metabolism
- Animals
- Arteries/growth & development
- Arteries/metabolism
- Arteries/pathology
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Cell Differentiation
- Cell Lineage/genetics
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelium, Vascular/growth & development
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Gene Expression Regulation, Developmental
- Humans
- Morphogenesis/genetics
- Muscle, Smooth, Vascular/growth & development
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neovascularization, Pathologic
- Neovascularization, Physiologic
Collapse
Affiliation(s)
- Sara B. Seidelmann
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| | - Janet K. Lighthouse
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| | - Daniel M. Greif
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| |
Collapse
|
133
|
Murray IR, West CC, Hardy WR, James AW, Park TS, Nguyen A, Tawonsawatruk T, Lazzari L, Soo C, Péault B. Natural history of mesenchymal stem cells, from vessel walls to culture vessels. Cell Mol Life Sci 2014; 71:1353-74. [PMID: 24158496 PMCID: PMC11113613 DOI: 10.1007/s00018-013-1462-6] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 08/17/2013] [Accepted: 08/23/2013] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) can regenerate tissues by direct differentiation or indirectly by stimulating angiogenesis, limiting inflammation, and recruiting tissue-specific progenitor cells. MSCs emerge and multiply in long-term cultures of total cells from the bone marrow or multiple other organs. Such a derivation in vitro is simple and convenient, hence popular, but has long precluded understanding of the native identity, tissue distribution, frequency, and natural role of MSCs, which have been defined and validated exclusively in terms of surface marker expression and developmental potential in culture into bone, cartilage, and fat. Such simple, widely accepted criteria uniformly typify MSCs, even though some differences in potential exist, depending on tissue sources. Combined immunohistochemistry, flow cytometry, and cell culture have allowed tracking the artifactual cultured mesenchymal stem/stromal cells back to perivascular anatomical regions. Presently, both pericytes enveloping microvessels and adventitial cells surrounding larger arteries and veins have been described as possible MSC forerunners. While such a vascular association would explain why MSCs have been isolated from virtually all tissues tested, the origin of the MSCs grown from umbilical cord blood remains unknown. In fact, most aspects of the biology of perivascular MSCs are still obscure, from the emergence of these cells in the embryo to the molecular control of their activity in adult tissues. Such dark areas have not compromised intents to use these cells in clinical settings though, in which purified perivascular cells already exhibit decisive advantages over conventional MSCs, including purity, thorough characterization and, principally, total independence from in vitro culture. A growing body of experimental data is currently paving the way to the medical usage of autologous sorted perivascular cells for indications in which MSCs have been previously contemplated or actually used, such as bone regeneration and cardiovascular tissue repair.
Collapse
Affiliation(s)
- Iain R. Murray
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Christopher C. West
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Winters R. Hardy
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
- Indiana Center for Vascular Biology and Medicine, Indianapolis, USA
| | - Aaron W. James
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Tea Soon Park
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore, USA
| | - Alan Nguyen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Tulyapruek Tawonsawatruk
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Lorenza Lazzari
- Cell Factory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Departments of Surgery and Orthopedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Bruno Péault
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
| |
Collapse
|
134
|
Hayes BJ, Riehle KJ, Shimizu-Albergine M, Bauer RL, Hudkins KL, Johansson F, Yeh MM, Mahoney WM, Yeung RS, Campbell JS. Activation of platelet-derived growth factor receptor alpha contributes to liver fibrosis. PLoS One 2014; 9:e92925. [PMID: 24667490 PMCID: PMC3965491 DOI: 10.1371/journal.pone.0092925] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 02/27/2014] [Indexed: 01/27/2023] Open
Abstract
Chronic liver injury leads to fibrosis, cirrhosis, and loss of liver function. Liver cirrhosis is the 12th leading cause of death in the United States, and it is the primary risk factor for developing liver cancer. Fibrosis and cirrhosis result from activation of hepatic stellate cells (HSCs), which are the primary collagen producing cell type in the liver. Here, we show that platelet-derived growth factor receptor α (PDGFRα) is expressed by human HSCs, and PDGFRα expression is elevated in human liver disease. Using a green fluorescent protein (GFP) reporter mouse strain, we evaluated the role of PDGFRα in liver disease in mice and found that mouse HSCs express PDGFRα and expression is upregulated during carbon tetrachloride (CCl4) induced liver injury and fibrosis injection. This fibrotic response is reduced in Pdgfrα heterozygous mice, consistent with the hypothesis that liver fibrosis requires upregulation and activation of PDGFRα. These results indicate that Pdgfrα expression is important in the fibrotic response to liver injury in humans and mice, and suggest that blocking PDGFRα–specific signaling pathways in HSCs may provide therapeutic benefit for patients with chronic liver disease.
Collapse
Affiliation(s)
- Brian J. Hayes
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Kimberly J. Riehle
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| | - Masami Shimizu-Albergine
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Renay L. Bauer
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Kelly L. Hudkins
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Fredrik Johansson
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Matthew M. Yeh
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - William M. Mahoney
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Raymond S. Yeung
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| | - Jean S. Campbell
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
135
|
Sanchez-Gurmaches J, Guertin DA. Adipocyte lineages: tracing back the origins of fat. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:340-51. [PMID: 23747579 PMCID: PMC3805734 DOI: 10.1016/j.bbadis.2013.05.027] [Citation(s) in RCA: 188] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 12/25/2022]
Abstract
The obesity epidemic has intensified efforts to understand the mechanisms controlling adipose tissue development. Adipose tissue is generally classified as white adipose tissue (WAT), the major energy storing tissue, or brown adipose tissue (BAT), which mediates non-shivering thermogenesis. It is hypothesized that brite adipocytes (brown in white) may represent a third adipocyte class. The recent realization that brown fat exist in adult humans suggests increasing brown fat energy expenditure could be a therapeutic strategy to combat obesity. To understand adipose tissue development, several groups are tracing the origins of mature adipocytes back to their adult precursor and embryonic ancestors. From these studies emerged a model that brown adipocytes originate from a precursor shared with skeletal muscle that expresses Myf5-Cre, while all white adipocytes originate from a Myf5-negative precursors. While this provided a rational explanation to why BAT is more metabolically favorable than WAT, recent work indicates the situation is more complex because subsets of white adipocytes also arise from Myf5-Cre expressing precursors. Lineage tracing studies further suggest that the vasculature may provide a niche supporting both brown and white adipocyte progenitors; however, the identity of the adipocyte progenitor cell is under debate. Differences in origin between adipocytes could explain metabolic heterogeneity between depots and/or influence body fat patterning particularly in lipodystrophy disorders. Here, we discuss recent insights into adipose tissue origins highlighting lineage-tracing studies in mice, how variations in metabolism or signaling between lineages could affect body fat distribution, and the questions that remain unresolved. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Joan Sanchez-Gurmaches
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
136
|
Lee YH, Jung YS, Choi D. Recent advance in brown adipose physiology and its therapeutic potential. Exp Mol Med 2014; 46:e78. [PMID: 24556827 PMCID: PMC3944445 DOI: 10.1038/emm.2013.163] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/08/2013] [Accepted: 12/09/2013] [Indexed: 12/18/2022] Open
Abstract
Brown adipose tissue (BAT) is a specialized thermoregulatory organ that has a critical role in the regulation of energy metabolism. Specifically, energy expenditure can be enhanced by the activation of BAT function and the induction of a BAT-like catabolic phenotype in white adipose tissue (WAT). Since the recent recognition of metabolically active BAT in adult humans, BAT has been extensively studied as one of the most promising targets identified for treating obesity and its related disorders. In this review, we summarize information on the developmental origin of BAT and the progenitors of brown adipocytes in WAT. We explore the transcriptional control of brown adipocyte differentiation during classical BAT development and in WAT browning. We also discuss the neuronal control of BAT activity and summarize the recently identified non-canonical stimulators of BAT that can act independently of β-adrenergic stimulation. Finally, we review new findings on the beneficial effects of BAT activation and development with respect to improving metabolic profiles. We highlight the therapeutic potential of BAT and its future prospects, including pharmacological intervention and cell-based therapies designed to enhance BAT activity and development.
Collapse
Affiliation(s)
- Yun-Hee Lee
- Center for Integrative and Metabolic Endocrine Research, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Dalwoong Choi
- 1] Department of Environmental Health, College of Health Sciences, Korea University, Seoul, Republic of Korea [2] BK21+ Program, Department of Public Health Science, Graduate School, Korea University, Seoul, Republic of Korea
| |
Collapse
|
137
|
Nakayama A, Nakayama M, Turner CJ, Höing S, Lepore JJ, Adams RH. Ephrin-B2 controls PDGFRβ internalization and signaling. Genes Dev 2014; 27:2576-89. [PMID: 24298057 PMCID: PMC3861671 DOI: 10.1101/gad.224089.113] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ephrin-B2 is essential for supporting mural cells; namely, pericytes and vascular smooth muscle cells (VSMCs). Nakayama et al. find that ephrin-B2 controls platelet-derived growth factor receptor β (PDGFRβ) distribution in the VSMC plasma membrane, endocytosis, and signaling. VSMCs lacking ephrin-B2 exhibited a redistribution of PDGFRβ from caveolin-positive to clathrin-associated membrane fractions and enhanced PDGF-B-induced PDGFRβ internalization. Mice lacking ephrin-B2 in vascular smooth muscle developed vessel wall defects and aortic aneurysms. These results suggest that ephrin-B2 is an important regulator of PDGFRβ endocytosis in mural cells. B-class ephrins, ligands for EphB receptor tyrosine kinases, are critical regulators of growth and patterning processes in many organs and species. In the endothelium of the developing vasculature, ephrin-B2 controls endothelial sprouting and proliferation, which has been linked to vascular endothelial growth factor (VEGF) receptor endocytosis and signaling. Ephrin-B2 also has essential roles in supporting mural cells (namely, pericytes and vascular smooth muscle cells [VSMCs]), but the underlying mechanism is not understood. Here, we show that ephrin-B2 controls platelet-derived growth factor receptor β (PDGFRβ) distribution in the VSMC plasma membrane, endocytosis, and signaling in a fashion that is highly distinct from its role in the endothelium. Absence of ephrin-B2 in cultured VSMCs led to the redistribution of PDGFRβ from caveolin-positive to clathrin-associated membrane fractions, enhanced PDGF-B-induced PDGFRβ internalization, and augmented downstream mitogen-activated protein (MAP) kinase and c-Jun N-terminal kinase (JNK) activation but impaired Tiam1–Rac1 signaling and proliferation. Accordingly, mutant mice lacking ephrin-B2 expression in vascular smooth muscle developed vessel wall defects and aortic aneurysms, which were associated with impaired Tiam1 expression and excessive activation of MAP kinase and JNK. Our results establish that ephrin-B2 is an important regulator of PDGFRβ endocytosis and thereby acts as a molecular switch controlling the downstream signaling activity of this receptor in mural cells.
Collapse
Affiliation(s)
- Akiko Nakayama
- Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, D-48149 Münster, Germany
| | | | | | | | | | | |
Collapse
|
138
|
Wang Y, Pan L, Moens CB, Appel B. Notch3 establishes brain vascular integrity by regulating pericyte number. Development 2013; 141:307-17. [PMID: 24306108 DOI: 10.1242/dev.096107] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Brain pericytes are important regulators of brain vascular integrity, permeability and blood flow. Deficiencies of brain pericytes are associated with neonatal intracranial hemorrhage in human fetuses, as well as stroke and neurodegeneration in adults. Despite the important functions of brain pericytes, the mechanisms underlying their development are not well understood and little is known about how pericyte density is regulated across the brain. The Notch signaling pathway has been implicated in pericyte development, but its exact roles remain ill defined. Here, we report an investigation of the Notch3 receptor using zebrafish as a model system. We show that zebrafish brain pericytes express notch3 and that notch3 mutant zebrafish have a deficit of brain pericytes and impaired blood-brain barrier function. Conditional loss- and gain-of-function experiments provide evidence that Notch3 signaling positively regulates brain pericyte proliferation. These findings establish a new role for Notch signaling in brain vascular development whereby Notch3 signaling promotes expansion of the brain pericyte population.
Collapse
Affiliation(s)
- Yuying Wang
- Departments of Pediatrics and Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|
139
|
Hung C, Linn G, Chow YH, Kobayashi A, Mittelsteadt K, Altemeier WA, Gharib SA, Schnapp LM, Duffield JS. Role of lung pericytes and resident fibroblasts in the pathogenesis of pulmonary fibrosis. Am J Respir Crit Care Med 2013; 188:820-30. [PMID: 23924232 DOI: 10.1164/rccm.201212-2297oc] [Citation(s) in RCA: 285] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
RATIONALE The origin of cells that make pathologic fibrillar collagen matrix in lung disease has been controversial. Recent studies suggest mesenchymal cells may contribute directly to fibrosis. OBJECTIVES To characterize discrete populations of mesenchymal cells in the normal mouse lung and to map their fate after bleomycin-induced lung injury. METHODS We mapped the fate of Foxd1-expressing embryonic progenitors and their progeny during lung development, adult homeostasis, and after fibrosing injury in Foxd1-Cre; Rs26-tdTomato-R mice. We studied collagen-I(α)1-producing cells in normal and diseased lungs using Coll-GFP(Tg) mice. MEASUREMENTS AND MAIN RESULTS Foxd1-expressing embryonic progenitors enter lung buds before 13.5 days post-conception, expand, and form an extensive lineage of mesenchymal cells that have characteristics of pericytes. A collagen-I(α)1-expressing mesenchymal population of distinct lineage is also found in adult lung, with features of a resident fibroblast. In contrast to resident fibroblasts, Foxd1 progenitor-derived pericytes are enriched in transcripts for innate immunity, vascular development, WNT signaling pathway, and cell migration. Foxd1 progenitor-derived pericytes expand after bleomycin lung injury, and activate expression of collagen-I(α)1 and the myofibroblast marker αSMA in fibrotic foci. In addition, our studies suggest a distinct lineage of collagen-I(α)1-expressing resident fibroblasts that also expands after lung injury is a second major source of myofibroblasts. CONCLUSIONS We conclude that the lung contains an extensive population of Foxd1 progenitor-derived pericytes that are an important lung myofibroblast precursor population.
Collapse
Affiliation(s)
- Chi Hung
- 1 Center for Lung Biology, Pulmonary and Critical Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Radiation-induced senescence in securin-deficient cancer cells promotes cell invasion involving the IL-6/STAT3 and PDGF-BB/PDGFR pathways. Sci Rep 2013; 3:1675. [PMID: 23591770 PMCID: PMC3628221 DOI: 10.1038/srep01675] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 04/02/2013] [Indexed: 12/14/2022] Open
Abstract
Securin overexpression correlates with poor prognosis in various tumours. We have previously shown that securin depletion promotes radiation-induced senescence and enhances radiosensitivity in human cancer cells. However, the underlying molecular mechanisms and the paracrine effects remain unknown. In this study, we showed that radiation induced senescence in securin-deficient human breast cancer cells involving the ATM/Chk2 and p38 pathways. Conditioned medium (CM) from senescent cells promoted the invasion and migration of non-irradiated cancer and endothelial cells. Cytokine assay analysis showed the up-regulation of various senescence-associated secretory phenotypes (SASPs). The IL-6/STAT3 signalling loop and platelet-derived growth factor-BB (PDGF-BB)/PDGF receptor (PDGFR) pathway were important for CM-induced cell migration and invasion. Furthermore, CM promoted angiogenesis in the chicken chorioallantoic membrane though the induction of IL-6/STAT3- and PDGF-BB/PDGFR-dependent endothelial cell invasion. Taken together, our results provide the molecular mechanisms for radiation-induced senescence in securin-deficient human breast cancer cells and for the SASP responses.
Collapse
|
141
|
Cao Y. Angiogenesis and vascular functions in modulation of obesity, adipose metabolism, and insulin sensitivity. Cell Metab 2013; 18:478-89. [PMID: 24035587 DOI: 10.1016/j.cmet.2013.08.008] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
White and brown adipose tissues are hypervascularized and the adipose vasculature displays phenotypic and functional plasticity to coordinate with metabolic demands of adipocytes. Blood vessels not only supply nutrients and oxygen to nourish adipocytes, they also serve as a cellular reservoir to provide adipose precursor and stem cells that control adipose tissue mass and function. Multiple signaling molecules modulate the complex interplay between the vascular system and the adipocytes. Understanding fundamental mechanisms by which angiogenesis and vasculatures modulate adipocyte functions may provide new therapeutic options for treatment of obesity and metabolic disorders by targeting the adipose vasculature.
Collapse
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden; Department of Medicine and Health Sciences, Linköping University, 581 85 Linköping, Sweden.
| |
Collapse
|
142
|
Zouani OF, Lei Y, Durrieu MC. Pericytes, stem-cell-like cells, but not mesenchymal stem cells are recruited to support microvascular tube stabilization. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:3070-3075. [PMID: 23625793 DOI: 10.1002/smll.201300124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 02/20/2013] [Indexed: 06/02/2023]
Abstract
An experimental model is introduced for the induction of endothelial cell (EC) tubulogenesis after 24 h of incubation on micropatterned polymer surfaces. Pericytes or mesenchymal stem cells are added separately to this system to evaluate their effect on tubular stabilization. In the absence of additional cells, the tubular structures are lost after 36 h. Addition of only pericytes, however, stabilizes the EC vasculogenic tubes.
Collapse
Affiliation(s)
- Omar F Zouani
- Université Bordeaux 1-CNRS, UMR5248, Institut Européen de Chimie et Biologie, 2, rue Robert Escarpit, F-33607 Pessac, France.
| | | | | |
Collapse
|
143
|
Iwayama T, Olson LE. Involvement of PDGF in fibrosis and scleroderma: recent insights from animal models and potential therapeutic opportunities. Curr Rheumatol Rep 2013; 15:304. [PMID: 23307576 DOI: 10.1007/s11926-012-0304-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Fibrosis is the principal characteristic of the autoimmune disease known as scleroderma or systemic sclerosis (SSc). Studies published within the last three years suggest central involvement of platelet-derived growth factors (PDGFs) in SSc-associated fibrosis. PDGFs may also be involved in SSc-associated autoimmunity and vasculopathy. The PDGF signaling pathway is well understood and PDGF receptors are expressed on collagen-secreting fibroblasts and on mesenchymal stem and/or progenitor cells that may affect SSc in profound and unexpected ways. Although much work remains before we fully understand how PDGFs are involved in SSc, there is much interest in using PDGF inhibitors as a therapeutic approach to SSc.
Collapse
Affiliation(s)
- Tomoaki Iwayama
- Immunobiology and Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | | |
Collapse
|
144
|
Heldin CH. Targeting the PDGF signaling pathway in the treatment of non-malignant diseases. J Neuroimmune Pharmacol 2013; 9:69-79. [PMID: 23793451 DOI: 10.1007/s11481-013-9484-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 06/05/2013] [Indexed: 12/13/2022]
Abstract
Platelet-derived growth factor (PDGF) is a family of mesenchymal mitogens with important functions during the embryonal development and in the control of tissue homeostasis in the adult. The PDGF isoforms exert their effects by binding to α-and β-tyrosine kinase receptors. Overactivity of PDGF signaling has been linked to the development of certain malignant and non-malignant diseases, including atherosclerosis and various fibrotic diseases. Different types of PDGF antagonists have been developed, including inhibitory monoclonal antibodies and DNA aptamers against PDGF isoforms and receptors, and receptor tyrosine kinase inhibitors. Beneficial effects have been recorded using such inhibitors in preclinical models and in patients with certain malignant as well as non-malignant diseases. The present communication summarizes the use of PDGF antagonists in the treatment of non-malignant diseases.
Collapse
Affiliation(s)
- Carl-Henrik Heldin
- Ludwig Institute for Cancer Research Ltd, Science for Life Laboratory, Uppsala University, Box 595, SE-75124, Uppsala, Sweden,
| |
Collapse
|
145
|
Lee YH, Mottillo EP, Granneman JG. Adipose tissue plasticity from WAT to BAT and in between. Biochim Biophys Acta Mol Basis Dis 2013; 1842:358-69. [PMID: 23688783 DOI: 10.1016/j.bbadis.2013.05.011] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/21/2013] [Accepted: 05/06/2013] [Indexed: 01/09/2023]
Abstract
Adipose tissue plays an essential role in regulating energy balance through its metabolic, cellular and endocrine functions. Adipose tissue has been historically classified into anabolic white adipose tissue and catabolic brown adipose tissue. An explosion of new data, however, points to the remarkable heterogeneity among the cells types that can become adipocytes, as well as the inherent metabolic plasticity of mature cells. These data indicate that targeting cellular and metabolic plasticity of adipose tissue might provide new avenues for treatment of obesity-related diseases. This review will discuss the developmental origins of adipose tissue, the cellular complexity of adipose tissues, and the identification of progenitors that contribute to adipogenesis throughout development. We will touch upon the pathological remodeling of adipose tissue and discuss how our understanding of adipose tissue remodeling can uncover new therapeutic targets. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Yun-Hee Lee
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Emilio P Mottillo
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - James G Granneman
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
146
|
Z. Adzemovic M, Zeitelhofer M, Eriksson U, Olsson T, Nilsson I. Imatinib ameliorates neuroinflammation in a rat model of multiple sclerosis by enhancing blood-brain barrier integrity and by modulating the peripheral immune response. PLoS One 2013; 8:e56586. [PMID: 23437178 PMCID: PMC3577871 DOI: 10.1371/journal.pone.0056586] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 01/11/2013] [Indexed: 01/24/2023] Open
Abstract
Central nervous system (CNS) disorders such as ischemic stroke, multiple sclerosis (MS) or Alzheimeŕs disease are characterized by the loss of blood-brain barrier (BBB) integrity. Here we demonstrate that the small tyrosine kinase inhibitor imatinib enhances BBB integrity in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis (MS). Treatment was accompanied by decreased CNS inflammation and demyelination and especially reduced T-cell recruitment. This was supported by downregulation of the chemokine receptor (CCR) 2 in CNS and lymph nodes, and by modulation of the peripheral immune response towards an anti-inflammatory phenotype. Interestingly, imatinib ameliorated neuroinflammation, even when the treatment was initiated after the clinical manifestation of the disease. We have previously shown that imatinib reduces BBB disruption and stroke volume after experimentally induced ischemic stroke by targeting platelet-derived growth factor receptor -α (PDGFR-α) signaling. Here we demonstrate that PDGFR-α signaling is a central regulator of BBB integrity during neuroinflammation and therefore imatinib should be considered as a potentially effective treatment for MS.
Collapse
MESH Headings
- Animals
- Benzamides/administration & dosage
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/metabolism
- Brain/immunology
- Brain/pathology
- Central Nervous System/drug effects
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Expression Regulation
- Imatinib Mesylate
- Inflammation/drug therapy
- Inflammation/immunology
- Inflammation/metabolism
- Multiple Sclerosis/genetics
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/pathology
- Neurons/drug effects
- Neurons/immunology
- Neurons/pathology
- Peptides/administration & dosage
- Peptides/toxicity
- Piperazines/administration & dosage
- Pyrimidines/administration & dosage
- Rats
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Receptors, CCR2/metabolism
- Signal Transduction
- Spinal Cord/immunology
- Spinal Cord/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Milena Z. Adzemovic
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Zeitelhofer
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Eriksson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (IN); (TO)
| | - Ingrid Nilsson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (IN); (TO)
| |
Collapse
|
147
|
Campanholle G, Ligresti G, Gharib SA, Duffield JS. Cellular mechanisms of tissue fibrosis. 3. Novel mechanisms of kidney fibrosis. Am J Physiol Cell Physiol 2013; 304:C591-603. [PMID: 23325411 DOI: 10.1152/ajpcell.00414.2012] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease, defined as loss of kidney function for more than three months, is characterized pathologically by glomerulosclerosis, interstitial fibrosis, tubular atrophy, peritubular capillary rarefaction, and inflammation. Recent studies have identified a previously poorly appreciated, yet extensive population of mesenchymal cells, called either pericytes when attached to peritubular capillaries or resident fibroblasts when embedded in matrix, as the progenitors of scar-forming cells known as myofibroblasts. In response to sustained kidney injury, pericytes detach from the vasculature and differentiate into myofibroblasts, a process not only causing fibrosis, but also directly contributing to capillary rarefaction and inflammation. The interrelationship of these three detrimental processes makes myofibroblasts and their pericyte progenitors an attractive target in chronic kidney disease. In this review, we describe current understanding of the mechanisms of pericyte-to-myofibroblast differentiation during chronic kidney disease, draw parallels with disease processes in the glomerulus, and highlight promising new therapeutic strategies that target pericytes or myofibroblasts. In addition, we describe the critical paracrine roles of epithelial, endothelial, and innate immune cells in the fibrogenic process.
Collapse
Affiliation(s)
- Gabriela Campanholle
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|
148
|
Boor P, Floege J. The renal (myo-)fibroblast: a heterogeneous group of cells. Nephrol Dial Transplant 2013; 27:3027-36. [PMID: 22851626 DOI: 10.1093/ndt/gfs296] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Several studies have demonstrated that mesenchymal stem cells have the capacity to reverse acute and chronic kidney injury in different experimental models by paracrine mechanisms. This paracrine action may be accounted for, at least in part, by microvesicles (MVs) released from mesenchymal stem cells, resulting in a horizontal transfer of mRNA, microRNA and proteins. MVs, released as exosomes from the endosomal compartment, or as shedding vesicles from the cell surface, are now recognized as being an integral component of the intercellular microenvironment. By acting as vehicles for information transfer, MVs play a pivotal role in cell-to-cell communication. This exchange of information between the injured cells and stem cells has the potential to be bi-directional. Thus, MVs may either transfer transcripts from injured cells to stem cells, resulting in reprogramming of their phenotype to acquire specific features of the tissue, or conversely, transcripts could be transferred from stem cells to injured cells, restraining tissue injury and inducing cell cycle re-entry of resident cells, leading to tissue self-repair. Upon administration with a therapeutic regimen, MVs mimic the effect of mesenchymal stem cells in various experimental models by inhibiting apoptosis and stimulating cell proliferation. In this review, we discuss whether MVs released from mesenchymal stem cells have the potential to be exploited in novel therapeutic approaches in regenerative medicine to repair damaged tissues, as an alternative to stem cell-based therapy.
Collapse
|
149
|
Greif DM, Kumar M, Lighthouse JK, Hum J, An A, Ding L, Red-Horse K, Espinoza FH, Olson L, Offermanns S, Krasnow MA. Radial construction of an arterial wall. Dev Cell 2013; 23:482-93. [PMID: 22975322 DOI: 10.1016/j.devcel.2012.07.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 05/24/2012] [Accepted: 07/16/2012] [Indexed: 01/04/2023]
Abstract
Some of the most serious diseases involve altered size and structure of the arterial wall. Elucidating how arterial walls are built could aid understanding of these diseases, but little is known about how concentric layers of muscle cells and the outer adventitial layer are assembled and patterned around endothelial tubes. Using histochemical, clonal, and genetic analysis in mice, here we show that the pulmonary artery wall is constructed radially, from the inside out, by two separate but coordinated processes. One is sequential induction of successive cell layers from surrounding mesenchyme. The other is controlled invasion of outer layers by inner layer cells through developmentally regulated cell reorientation and radial migration. We propose that a radial signal gradient controls these processes and provide evidence that PDGF-B and at least one other signal contribute. Modulation of such radial signaling pathways may underlie vessel-specific differences and pathological changes in arterial wall size and structure.
Collapse
Affiliation(s)
- Daniel M Greif
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA 94305, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Boyd NL, Nunes SS, Krishnan L, Jokinen JD, Ramakrishnan VM, Bugg AR, Hoying JB. Dissecting the role of human embryonic stem cell-derived mesenchymal cells in human umbilical vein endothelial cell network stabilization in three-dimensional environments. Tissue Eng Part A 2013; 19:211-223. [PMID: 22971005 PMCID: PMC3530951 DOI: 10.1089/ten.tea.2011.0408] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 07/30/2012] [Indexed: 10/27/2022] Open
Abstract
The microvasculature is principally composed of two cell types: endothelium and mural support cells. Multiple sources are available for human endothelial cells (ECs) but sources for human microvascular mural cells (MCs) are limited. We derived multipotent mesenchymal progenitor cells from human embryonic stem cells (hES-MC) that can function as an MC and stabilize human EC networks in three-dimensional (3D) collagen-fibronectin culture by paracrine mechanisms. Here, we have investigated the basis for hES-MC-mediated stabilization and identified the pleiotropic growth factor hepatocyte growth factor/scatter factor (HGF/SF) as a putative hES-MC-derived regulator of EC network stabilization in 3D in vitro culture. Pharmacological inhibition of the HGF receptor (Met) (1 μm SU11274) inhibits EC network formation in the presence of hES-MC. hES-MC produce and release HGF while human umbilical vein endothelial cells (HUVEC) do not. When HUVEC are cultured alone the networks collapse, but in the presence of recombinant human HGF or conditioned media from human HGF-transduced cells significantly more networks persist. In addition, HUVEC transduced to constitutively express human HGF also form stable networks by autocrine mechanisms. By enzyme-linked immunosorbent assay, the coculture media were enriched in both angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2), but at significantly different levels (Ang1=159±15 pg/mL vs. Ang2=30,867±2685 pg/mL) contributed by hES-MC and HUVEC, respectively. Although the coculture cells formed stabile network architectures, their morphology suggests the assembly of an immature plexus. When HUVEC and hES-MC were implanted subcutaneously in immune compromised Rag1 mice, hES-MC increased their contact with HUVEC along the axis of the vessel. This data suggests that HUVEC and hES-MC form an immature plexus mediated in part by HGF and angiopoietins that is capable of maturation under the correct environmental conditions (e.g., in vivo). Therefore, hES-MC can function as microvascular MCs and may be a useful cell source for testing EC-MC interactions.
Collapse
Affiliation(s)
- Nolan L Boyd
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA.
| | | | | | | | | | | | | |
Collapse
|