1
|
Ko J, Noviani M, Chellamuthu VR, Albani S, Low AHL. The Pathogenesis of Systemic Sclerosis: The Origin of Fibrosis and Interlink with Vasculopathy and Autoimmunity. Int J Mol Sci 2023; 24:14287. [PMID: 37762589 PMCID: PMC10532389 DOI: 10.3390/ijms241814287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease associated with increased mortality and poor morbidity, impairing the quality of life in patients. Whilst we know that SSc affects multiple organs via vasculopathy, inflammation, and fibrosis, its exact pathophysiology remains elusive. Microvascular injury and vasculopathy are the initial pathological features of the disease. Clinically, the vasculopathy in SSc is manifested as Raynaud's phenomenon (reversible vasospasm in reaction to the cold or emotional stress) and digital ulcers due to ischemic injury. There are several reports that medications for vasculopathy, such as bosentan and soluble guanylate cyclase (sGC) modulators, improve not only vasculopathy but also dermal fibrosis, suggesting that vasculopathy is important in SSc. Although vasculopathy is an important initial step of the pathogenesis for SSc, it is still unclear how vasculopathy is related to inflammation and fibrosis. In this review, we focused on the clinical evidence for vasculopathy, the major cellular players for the pathogenesis, including pericytes, adipocytes, endothelial cells (ECs), and myofibroblasts, and their signaling pathway to elucidate the relationship among vasculopathy, inflammation, and fibrosis in SSc.
Collapse
Affiliation(s)
- Junsuk Ko
- Duke-National University of Singapore Medical School, Singapore 169857, Singapore; (J.K.); (M.N.); (S.A.)
| | - Maria Noviani
- Duke-National University of Singapore Medical School, Singapore 169857, Singapore; (J.K.); (M.N.); (S.A.)
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore 169608, Singapore
- Translational Immunology Institute, SingHealth Duke-National University of Singapore Academic Medical Centre, Singapore 169856, Singapore;
| | - Vasuki Ranjani Chellamuthu
- Translational Immunology Institute, SingHealth Duke-National University of Singapore Academic Medical Centre, Singapore 169856, Singapore;
| | - Salvatore Albani
- Duke-National University of Singapore Medical School, Singapore 169857, Singapore; (J.K.); (M.N.); (S.A.)
- Translational Immunology Institute, SingHealth Duke-National University of Singapore Academic Medical Centre, Singapore 169856, Singapore;
| | - Andrea Hsiu Ling Low
- Duke-National University of Singapore Medical School, Singapore 169857, Singapore; (J.K.); (M.N.); (S.A.)
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore 169608, Singapore
| |
Collapse
|
2
|
De Rossi G, Da Vitoria Lobo ME, Greenwood J, Moss SE. LRG1 as a novel therapeutic target in eye disease. Eye (Lond) 2022; 36:328-340. [PMID: 34987199 PMCID: PMC8807626 DOI: 10.1038/s41433-021-01807-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/22/2021] [Accepted: 10/01/2021] [Indexed: 02/08/2023] Open
Abstract
Retinal and choroidal diseases are major causes of blindness and visual impairment in the developed world and on the rise due to an ageing population and diabetes epidemic. Standard of care is centred around blockade of vascular endothelial growth factor (VEGF), but despite having halved the number of patients losing sight, a high rate of patient non-response and loss of efficacy over time are key challenges. Dysregulation of vascular homoeostasis, coupled with fibrosis and inflammation, are major culprits driving sight-threatening eye diseases. Improving our knowledge of these pathological processes should inform the development of new drugs to address the current clinical challenges for patients. Leucine-rich α-2 glycoprotein 1 (LRG1) is an emerging key player in vascular dysfunction, inflammation and fibrosis. Under physiological conditions, LRG1 is constitutively expressed by the liver and granulocytes, but little is known about its normal biological function. In pathological scenarios, such as diabetic retinopathy (DR) and neovascular age-related macular degeneration (nvAMD), its expression is ectopically upregulated and it acquires a much better understood pathogenic role. Context-dependent modulation of the transforming growth-factor β (TGFβ) pathway is one of the main activities of LRG1, but additional roles have recently been emerging. This review aims to highlight the clinical and pre-clinical evidence for the pathogenic contribution of LRG1 to vascular retinopathies, as well as extrapolate from other diseases, functions which may be relevant to eye disease. Finally, we will provide a current update on the development of anti-LRG1 therapies for the treatment of nvAMD.
Collapse
Affiliation(s)
- Giulia De Rossi
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | | | - John Greenwood
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Stephen E Moss
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| |
Collapse
|
3
|
Furukawa K, Kono M, Kataoka T, Hasebe Y, Jia H, Kato H. Effects of Eggshell Membrane on Keratinocyte Differentiation and Skin Aging In Vitro and In Vivo. Nutrients 2021; 13:nu13072144. [PMID: 34206704 PMCID: PMC8308305 DOI: 10.3390/nu13072144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/23/2022] Open
Abstract
Skin aging is one of the hallmarks of the aging process that causes physiological and morphological changes. Recently, several nutritional studies were conducted to delay or suppress the aging process. This study investigated whether nutritional supplementation of the eggshell membrane (ESM) has a beneficial effect on maintaining skin health and improving the skin aging process in vitro using neonatal normal human epidermal keratinocytes (NHEK-Neo) and in vivo using interleukin-10 knockout (IL-10 KO) mice. In NHEK-Neo cells, 1 mg/mL of enzymatically hydrolyzed ESM (eESM) upregulated the expression of keratinocyte differentiation markers, including keratin 1, filaggrin and involucrin, and changed the keratinocyte morphology. In IL-10 KO mice, oral supplementation of 8% powdered-ESM (pESM) upregulated the expression of growth factors, including transforming growth factor β1, platelet-derived growth factor-β and connective tissue growth factor, and suppressed skin thinning. Furthermore, voltage-gated calcium channel, transient receptor potential cation channel subfamily V members were upregulated by eESM treatment in NHEK-Neo cells and pESM supplementation in IL-10 KO mice. Collectively, these data suggest that ESM has an important role in improving skin health and aging, possibly via upregulating calcium signaling.
Collapse
Affiliation(s)
- Kyohei Furukawa
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.F.); (M.K.); (T.K.)
| | - Masaya Kono
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.F.); (M.K.); (T.K.)
| | - Tetsuro Kataoka
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.F.); (M.K.); (T.K.)
| | | | - Huijuan Jia
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.F.); (M.K.); (T.K.)
- Correspondence: (H.J.); (H.K.); Tel.: +81-3-5841-5116 (H.J.); +81-3-5841-1607 (H.K.)
| | - Hisanori Kato
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.F.); (M.K.); (T.K.)
- Correspondence: (H.J.); (H.K.); Tel.: +81-3-5841-5116 (H.J.); +81-3-5841-1607 (H.K.)
| |
Collapse
|
4
|
Chen L, Wu H, Ren C, Liu G, Zhang W, Liu W, Lu P. Inhibition of PDGF-BB reduces alkali-induced corneal neovascularization in mice. Mol Med Rep 2021; 23:238. [PMID: 33537811 PMCID: PMC7893695 DOI: 10.3892/mmr.2021.11877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The aim of the present study was to investigate the role of platelet-derived growth factor (PDGF)-BB/PDGF receptor (R)-β signaling in an experimental murine corneal neovascularization (CrNV) model. Experimental CrNV was induced by alkali injury. The intra-corneal expression of PDGF-BB was examined using immunohistochemistry. The effect of PDGF-BB on CrNV was evaluated using immunofluorescence staining. The expression levels of PDGFR-β in human retinal endothelial cells (HRECs) under normal conditions or following cobalt chloride treatment, which induced hypoxic conditions, was assessed using reverse transcription-quantitative PCR. The effect of exogenous treatment of PDGF-BB on the proliferation, migration and tube formation of HRECs under normoxic or hypoxic conditions was evaluated in vitro using Cell Counting Kit-8, wound healing and 3D Matrigel capillary tube formation assays, respectively. The results indicated that the intra-corneal expression levels of the proteins of PDGF-BB and PDGFR-β were detectable on days 2 and 7 following alkali injury. The treatment with neutralizing anti-PDGF-BB antibody resulted in significant inhibition of CrNV. The intra-corneal expression levels of vascular endothelial growth factor A, matrix metallopeptidase (MMP)-2 and MMP-9 proteins were downregulated, while the expression levels of thrombospondin (TSP)-1, TSP-2, a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-1 and ADAMTS-2 were upregulated significantly in mice treated with anti-PDGF-BB antibody. The expression levels of PDGFR-β were upregulated in HRECs under hypoxic conditions compared with those noted under normoxic conditions. Recombinant human PDGF-BB promoted the proliferation, migration and tube formation of HRECs under hypoxic conditions. The data indicated that PDGF-BB/PDGFR-β signaling was involved in CrNV and that it promoted endothelial cell proliferation, migration and tube formation. The pro-angiogenic effects of this pathway may be mediated via the induction of pro-angiogenic cytokine secretion and the suppression of anti-angiogenic cytokine secretion.
Collapse
Affiliation(s)
- Lei Chen
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongya Wu
- Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Chi Ren
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Gaoqin Liu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Wenpeng Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Weiming Liu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Peirong Lu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
5
|
PDGFA gene rs9690350 polymorphism increases biliary atresia risk in Chinese children. Biosci Rep 2020; 40:225782. [PMID: 32662506 PMCID: PMC7374268 DOI: 10.1042/bsr20200068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 06/25/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022] Open
Abstract
Biliary atresia (BA) is a genetic and severe fibro-inflammatory obliterative cholangiopathy of neonates. Platelet-derived growth factor subunit A (PDGFA), as one of participants in liver fibrosis, the overexpression of PDGFA through DNA hypomethylation may lead to the development of BA, but the pathogenesis is still unclear. We conducted a large case-control cohort to investigate the association of genetic variants in PDGFA with BA susceptibility in the Southern Chinese population (506 cases and 1473 controls). We observed that the G allele of rs9690350(G>C) in PDGFA was significantly associated with an increased risk of BA (OR = 1.24, 95% CI = 1.04-1.49, P=0.02). Additionally, the rs9690350 G allele increased the risk of non-cystic biliary atresia (OR = 1.26, 95% CI = 1.04-1.52, P=0.02) and was a genetic biomarker of severe manifestations after surgery. These findings indicate that the rs9690350 G allele is a PDGFA polymorphism associated with the risk of BA that may confer increased disease susceptibility.
Collapse
|
6
|
Hou Z, Neng L, Zhang J, Cai J, Wang X, Zhang Y, Lopez IA, Shi X. Acoustic Trauma Causes Cochlear Pericyte-to-Myofibroblast-Like Cell Transformation and Vascular Degeneration, and Transplantation of New Pericytes Prevents Vascular Atrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1943-1959. [PMID: 32562655 DOI: 10.1016/j.ajpath.2020.05.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/19/2020] [Accepted: 05/26/2020] [Indexed: 12/20/2022]
Abstract
Acoustic trauma disrupts cochlear blood flow and damages sensory hair cells. Damage and regression of capillaries after acoustic trauma have long been observed, but the underlying mechanism of pathology has not been understood. We show herein that loud sound causes change of phenotype from neural/glial antigen 2 positive/α-smooth muscle actin negative to neural/glial antigen 2 positive/α-smooth muscle actin positive in some pericytes (PCs) on strial capillaries that is strongly associated with up-regulation of transforming growth factor-β1. The acoustic trauma also reduced capillary density and increased deposition of matrix proteins, particularly in the vicinity of transformed PCs. In a newly established in vitro three-dimensional endothelial cell (EC) and PC co-culture model, transformed PCs induced thicker capillary-like branches in ECs and increased collagen IV and laminin expression. Transplantation of exogenous PCs derived from neonatal day 10 mouse cochleae to acoustic traumatized cochleae, however, significantly attenuated the decreased vascular density in the stria. Transplantation of PCs pretransfected with adeno-associated virus 1-vascular endothelial growth factor-A165 under control of a hypoxia-response element markedly promotes vascular volume and blood flow, increased proliferation of PCs and ECs, and attenuated loud sound-caused loss in endocochlear potential and hearing. Our results indicate that loud sound-triggered PC transformation contributes to capillary wall thickening and regression, and young PC transplantation effectively rehabilitates the vascular regression and improves hearing.
Collapse
Affiliation(s)
- Zhiqiang Hou
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon
| | - Lingling Neng
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon
| | - Jinhui Zhang
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon
| | - Jing Cai
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon
| | - Xiaohan Wang
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon; Center for Life Sciences, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yunpei Zhang
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon
| | - Ivan A Lopez
- Cellular and Molecular Biology of the Inner Ear Laboratory, Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Xiaorui Shi
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
7
|
Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med 2020; 217:e20190103. [PMID: 32997468 PMCID: PMC7062524 DOI: 10.1084/jem.20190103] [Citation(s) in RCA: 701] [Impact Index Per Article: 140.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 12/24/2019] [Indexed: 12/21/2022] Open
Abstract
TGF-β is extensively implicated in the pathogenesis of fibrosis. In fibrotic lesions, spatially restricted generation of bioactive TGF-β from latent stores requires the cooperation of proteases, integrins, and specialized extracellular matrix molecules. Although fibroblasts are major targets of TGF-β, some fibrogenic actions may reflect activation of other cell types, including macrophages, epithelial cells, and vascular cells. TGF-β–driven fibrosis is mediated through Smad-dependent or non-Smad pathways and is modulated by coreceptors and by interacting networks. This review discusses the role of TGF-β in fibrosis, highlighting mechanisms of TGF-β activation and signaling, the cellular targets of TGF-β actions, and the challenges of therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
8
|
Morikawa S, Iribar H, Gutiérrez-Rivera A, Ezaki T, Izeta A. Pericytes in Cutaneous Wound Healing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:1-63. [DOI: 10.1007/978-3-030-16908-4_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
9
|
Certainties and uncertainties concerning the contribution of pericytes to the pathogenesis of systemic sclerosis. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2017; 3:14-20. [DOI: 10.5301/jsrd.5000254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/09/2017] [Indexed: 02/08/2023]
Abstract
The role of pericytes in systemic sclerosis (SSc) is unclear because of the difficulty in phenotyping them. They are mainly distributed in the pre-capillary, capillary and post-capillary abluminal side of non-muscular micro-vessels, express platelet-derived growth factor receptors (PDGFRs), and preside over vascular integrity and regeneration. By establishing close contact with many endothelial cells, a single pericyte can regulate ion influx, mechanical stress, leukocyte diapedesis, and platelet activation. Moreover, under pathological conditions such as SSc, pericytes may acquire a contractile phenotype and respond to various stimuli, including endothelin, angiotensin II and reactive oxygen species. The pericytes of SSc patients share some molecular patterns with myofibroblasts or fibroblasts, including A disintegrin and metalloproteinase domain 12 (ADAM-12), α-smooth muscle actin (α-SMA), the extra domain A (ED-A) variant of fibronectin, and Thy-1. Following stimulation with PDGF-β or transforming growth factor-β (TGF-β), pericytes may acquire a myofibroblast phenotype, and produce extracellular matrix or indirectly promote fibroblast activation. They may also contribute to fibrosis by means of epigenetic regulation. The pericyte plasmalemma is particularly rich in caveolae containing caveolin-1, a deficit of which has been associated with defective vessel tone control and lung fibrosis in mice. Consequently, dysfunctional pericytes may underlie the microangiopathy and fibrosis observed in SSc patients. However, given its variability in biological behaviour and the lack of a pan-pericyte marker, the exact role of these cells in SSc warrants further investigation.
Collapse
|
10
|
Olsson PO, Gustafsson R, In 't Zandt R, Friman T, Maccarana M, Tykesson E, Oldberg Å, Rubin K, Kalamajski S. The Tyrosine Kinase Inhibitor Imatinib Augments Extracellular Fluid Exchange and Reduces Average Collagen Fibril Diameter in Experimental Carcinoma. Mol Cancer Ther 2016; 15:2455-2464. [PMID: 27474147 DOI: 10.1158/1535-7163.mct-16-0026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/23/2016] [Indexed: 11/16/2022]
Abstract
A typical obstacle to cancer therapy is the limited distribution of low molecular weight anticancer drugs within the carcinoma tissue. In experimental carcinoma, imatinib (STI571) increases efficacy of synchronized chemotherapy, reduces tumor interstitial fluid pressure, and increases interstitial fluid volume. STI571 also increases the water-perfusable fraction in metastases from human colorectal adenocarcinomas. Because the mechanism(s) behind these effects have not been fully elucidated, we investigated the hypothesis that STI571 alters specific properties of the stromal extracellular matrix. We analyzed STI571-treated human colorectal KAT-4/HT-29 experimental carcinomas, known to have a well-developed stromal compartment, for solute exchange and glycosaminoglycan content, as well as collagen content, structure, and synthesis. MRI of STI571-treated KAT-4/HT-29 experimental carcinomas showed a significantly increased efficacy in dynamic exchanges of solutes between tumor interstitium and blood. This effect was paralleled by a distinct change of the stromal collagen network architecture, manifested by a decreased average collagen fibril diameter, and increased collagen turnover. The glycosaminoglycan content was unchanged. Furthermore, the apparent effects on the stromal cellular composition were limited to a reduction in an NG2-positive stromal cell population. The current data support the hypothesis that the collagen network architecture influences the dynamic exchanges of solutes between blood and carcinoma tissue. It is conceivable that STI571 reprograms distinct nonvascular stromal cells to produce a looser extracellular matrix, ultimately improving transport characteristics for traditional chemotherapeutic agents. Mol Cancer Ther; 15(10); 2455-64. ©2016 AACR.
Collapse
Affiliation(s)
- P Olof Olsson
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Renata Gustafsson
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | | | - Tomas Friman
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Marco Maccarana
- Department of Experimental Medical Sciences, Matrixbiology, Lund University, Lund, Sweden
| | - Emil Tykesson
- Department of Experimental Medical Sciences, Matrixbiology, Lund University, Lund, Sweden
| | - Åke Oldberg
- Department of Experimental Medical Sciences, Matrixbiology, Lund University, Lund, Sweden
| | - Kristofer Rubin
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden.
| | - Sebastian Kalamajski
- Department of Medical Biochemistry and Microbiology, SciLife Laboratories, Uppsala University, Uppsala, Sweden
| |
Collapse
|
11
|
Wietecha MS, Król MJ, Michalczyk ER, Chen L, Gettins PG, DiPietro LA. Pigment epithelium-derived factor as a multifunctional regulator of wound healing. Am J Physiol Heart Circ Physiol 2015; 309:H812-26. [PMID: 26163443 DOI: 10.1152/ajpheart.00153.2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/08/2015] [Indexed: 01/12/2023]
Abstract
During dermal wound repair, hypoxia-driven proliferation results in dense but highly permeable, disorganized microvascular networks, similar to those in solid tumors. Concurrently, activated dermal fibroblasts generate an angiopermissive, provisional extracellular matrix (ECM). Unlike cancers, wounds naturally resolve via blood vessel regression and ECM maturation, which are essential for reestablishing tissue homeostasis. Mechanisms guiding wound resolution are poorly understood; one candidate regulator is pigment epithelium-derived factor (PEDF), a secreted glycoprotein. PEDF is a potent antiangiogenic in models of pathological angiogenesis and a promising cancer and cardiovascular disease therapeutic, but little is known about its physiological function. To examine the roles of PEDF in physiological wound repair, we used a reproducible model of excisional skin wound healing in BALB/c mice. We show that PEDF is abundant in unwounded and healing skin, is produced primarily by dermal fibroblasts, binds to resident microvascular endothelial cells, and accumulates in dermal ECM and epidermis. PEDF transcript and protein levels were low during the inflammatory and proliferative phases of healing but increased in quantity and colocalization with microvasculature during wound resolution. Local antibody inhibition of endogenous PEDF delayed vessel regression and collagen maturation during the remodeling phase. Treatment of wounds with intradermal injections of exogenous, recombinant PEDF inhibited nascent angiogenesis by repressing endothelial proliferation, promoted vascular integrity and function, and increased collagen maturity. These results demonstrate that PEDF contributes to the resolution of healing wounds by causing regression of immature blood vessels and stimulating maturation of the vascular microenvironment, thus promoting a return to tissue homeostasis after injury.
Collapse
Affiliation(s)
- Mateusz S Wietecha
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois; and
| | - Mateusz J Król
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois; and
| | - Elizabeth R Michalczyk
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois; and
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois; and
| | - Peter G Gettins
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois
| | - Luisa A DiPietro
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
12
|
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: 58] [Impact Index Per Article: 5.8] [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]
|
13
|
Zhang J, Chen S, Hou Z, Cai J, Dong M, Shi X. Lipopolysaccharide-induced middle ear inflammation disrupts the cochlear intra-strial fluid-blood barrier through down-regulation of tight junction proteins. PLoS One 2015; 10:e0122572. [PMID: 25815897 PMCID: PMC4376743 DOI: 10.1371/journal.pone.0122572] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/16/2015] [Indexed: 12/20/2022] Open
Abstract
Middle ear infection (or inflammation) is the most common pathological condition that causes fluid to accumulate in the middle ear, disrupting cochlear homeostasis. Lipopolysaccharide, a product of bacteriolysis, activates macrophages and causes release of inflammatory cytokines. Many studies have shown that lipopolysaccharides cause functional and structural changes in the inner ear similar to that of inflammation. However, it is specifically not known how lipopolysaccharides affect the blood-labyrinth barrier in the stria vascularis (intra-strial fluid–blood barrier), nor what the underlying mechanisms are. In this study, we used a cell culture-based in vitro model and animal-based in vivo model, combined with immunohistochemistry and a vascular leakage assay, to investigate lipopolysaccharide effects on the integrity of the mouse intra-strial fluid–blood barrier. Our results show lipopolysaccharide-induced local infection significantly affects intra-strial fluid–blood barrier component cells. Pericytes and perivascular-resident macrophage-like melanocytes are particularly affected, and the morphological and functional changes in these cells are accompanied by substantial changes in barrier integrity. Significant vascular leakage is found in the lipopolysaccharide treated-animals. Consistent with the findings from the in vivo animal model, the permeability of the endothelial cell monolayer to FITC-albumin was significantly higher in the lipopolysaccharide-treated monolayer than in an untreated endothelial cell monolayer. Further study has shown the lipopolysaccharide-induced inflammation to have a major effect on the expression of tight junctions in the blood barrier. Lipopolysaccharide was also shown to cause high frequency hearing loss, corroborated by previous reports from other laboratories. Our findings show lipopolysaccharide-evoked middle ear infection disrupts inner ear fluid balance, and its particular effects on the intra-strial fluid–blood barrier, essential for cochlear homeostasis. The barrier is degraded as the expression of tight junction-associated proteins such as zona occludens 1, occludin, and vascular endothelial cadherin are down-regulated.
Collapse
Affiliation(s)
- Jinhui Zhang
- Department of Otolaryngology/Head and Neck Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Oregon Hearing Research Center, Department of Otolaryngology/Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Songlin Chen
- Oregon Hearing Research Center, Department of Otolaryngology/Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Zhiqiang Hou
- Oregon Hearing Research Center, Department of Otolaryngology/Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jing Cai
- Oregon Hearing Research Center, Department of Otolaryngology/Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Mingmin Dong
- Department of Otolaryngology/Head and Neck Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaorui Shi
- Oregon Hearing Research Center, Department of Otolaryngology/Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
14
|
Hill J, Rom S, Ramirez SH, Persidsky Y. Emerging roles of pericytes in the regulation of the neurovascular unit in health and disease. J Neuroimmune Pharmacol 2014; 9:591-605. [PMID: 25119834 PMCID: PMC4209199 DOI: 10.1007/s11481-014-9557-x] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/10/2014] [Indexed: 12/14/2022]
Abstract
Pericytes of the central nervous system (CNS) are uniquely positioned within a multicellular structure termed the neurovascular unit (NVU) to provide crucial support to blood brain barrier (BBB) formation, maintenance, and stability. Numerous CNS diseases are associated with some aspect of BBB dysfunction. A dysfunction can manifest as one or multiple disruptions to any of the following barriers: physical, metabolic, immunological and transport barrier. A breach in the BBB can notably result in BBB hyper-permeability, endothelial activation and enhanced immune-endothelial interaction. How the BBB is regulated within this integrated unit remains largely unknown, especially as it relates to pericyte-endothelial interaction. We summarize the latest findings on pericyte origin, possible marker expression, and availability within different organ systems. We highlight pericyte-endothelial cell interactions, concentrating on extra- and intra- cellular signaling mechanisms linked to platelet derived growth factor-B, transforming growth factor -β, angiopoietins, Notch, and gap junctions. We discuss the role of pericytes in the NVU under inflammatory insult, focusing on how pericytes may indirectly affect leukocyte CNS infiltration, the direct role of pericyte-mediated basement membrane modifications, and immune responses. We review new findings of pericyte actions in CNS pathologies including Alzheimer's disease, stroke, multiple sclerosis, diabetic retinopathy, and HIV-1 infection. The uncovering of the regulatory role of pericytes on the BBB will provide key insight into how barrier integrity can be re-established during neuroinflammation.
Collapse
Affiliation(s)
- Jeremy Hill
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia PA
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia PA
| | - Slava Rom
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia PA
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia PA
| | - Servio H. Ramirez
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia PA
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia PA
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia PA
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia PA
| |
Collapse
|
15
|
Cipriani P, Di Benedetto P, Ruscitti P, Campese AF, Liakouli V, Carubbi F, Pantano I, Berardicurt O, Screpanti I, Giacomelli R. Impaired endothelium-mesenchymal stem cells cross-talk in systemic sclerosis: a link between vascular and fibrotic features. Arthritis Res Ther 2014; 16:442. [PMID: 25248297 PMCID: PMC4206764 DOI: 10.1186/s13075-014-0442-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 08/28/2014] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION To assess if an impaired cross-talk between endothelial cells (ECs) and perivascular/multipotent mesenchymal stem cells (MSCs) might induce a perturbation of vascular repair and leading to a phenotypic switch of MSC toward myofibroblast in Systemic Sclerosis (SSc). METHODS We investigated different angiogenic and profibrotic molecules in a tridimentional matrigel assay, performing co-cultures with endothelial cells (ECs) and bone marrow derived MSCs from patients and healthy controls (HC). After 48 hours of co-culture, cells were sorted and analyzed for mRNA and protein expression. RESULTS ECs-SSc showed a decreased tube formation ability which is not improved by co-cultures with different MSCs. After sorting, we showed: i. an increased production of vascular endothelial growth factor A (VEGF-A) in SSc-MSCs when co-cultured with SSc-ECs; ii. an increased level of transforming growth factor beta (TGF-β) and platelet growth factor BB (PDGF-BB) in SSc-ECs when co-cultured with both HC- and SSc-MSCs; iii. an increase of TGF-β, PDGF-R, alpha smooth muscle actin (α-SMA) and collagen 1 (Col1) in both HC- and SSc-MSCs when co-cultured with SSc-ECs. CONCLUSION We showed that during SSc, the ECs-MSCs crosstalk resulted in an altered expression of different molecules involved in the angiogenic processes, and mainly SSc-ECs seem to modulate the phenotypic switch of perivascular MSCs toward a myofibroblast population, thus supporting the fibrotic process.
Collapse
Affiliation(s)
- Paola Cipriani
- />Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Paola Di Benedetto
- />Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Piero Ruscitti
- />Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Antonio Francesco Campese
- />Department of Molecular Medicine, School of Medicine ‘Sapienza’ University of Rome, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Vasiliki Liakouli
- />Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Francesco Carubbi
- />Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Ilenia Pantano
- />Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Onorina Berardicurt
- />Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Isabella Screpanti
- />Department of Molecular Medicine, School of Medicine ‘Sapienza’ University of Rome, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Roberto Giacomelli
- />Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| |
Collapse
|