1
|
Goldman N, Ong VH, Denton CP. Pathogenesis of interstitial lung disease in systemic sclerosis. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2024; 5:141-151. [PMID: 39439973 PMCID: PMC11492824 DOI: 10.2478/rir-2024-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 10/25/2024]
Abstract
Interstitial lung disease (ILD) is a frequent important complication of systemic sclerosis (SSc). Factors relevant to aetiopathogenesis of SSc are also central to SSc-ILD. Severity of SSc-ILD is variable but it has a major impact on morbidity and mortality. Factors determining SSc-ILD susceptibility reflect the genetic architecture of SSc and are increasingly being defined. There are aspects linked to immunogenomics and non-immunological genetic factors that may be less conserved and underlie some of the geographical and racial diversity of SSc. These associations may also underlie important links between autoantibody subgroups and patient level risk of SSc-ILD. Examination of blood and tissue samples and observational clinical research together with integrated analysis of in vitro and in vivo preclinical models have elucidated pathogenic mechanisms of SSc-ILD. These have confirmed the potential importance of immune mechanisms in the innate and adaptive immune systemic as well as a significant role for profibrotic pathways especially transforming growth factor beta (TGFbeta) and its regulators and downstream mediators. Recent analysis of clinical trial cohorts as well as integrated and multilevel high dimensional analysis of bio-samples has shed further light on SSc-ILD. This is likely to underpin future advances in stratified and precision medicine for treatment of SSc.
Collapse
Affiliation(s)
- Nina Goldman
- Center for Rheumatology, University College London, London, UK
| | - Voon H Ong
- Center for Rheumatology, University College London, London, UK
| | | |
Collapse
|
2
|
Papaioannou I, Dritsoula A, Kang P, Baliga RS, Trinder SL, Cook E, Shiwen X, Hobbs AJ, Denton CP, Abraham DJ, Ponticos M. NKX2-5 regulates vessel remodeling in scleroderma-associated pulmonary arterial hypertension. JCI Insight 2024; 9:e164191. [PMID: 38652537 PMCID: PMC11141943 DOI: 10.1172/jci.insight.164191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
NKX2-5 is a member of the homeobox-containing transcription factors critical in regulating tissue differentiation in development. Here, we report a role for NKX2-5 in vascular smooth muscle cell phenotypic modulation in vitro and in vascular remodeling in vivo. NKX2-5 is upregulated in scleroderma patients with pulmonary arterial hypertension. Suppression of NKX2-5 expression in smooth muscle cells halted vascular smooth muscle proliferation and migration, enhanced contractility, and blocked the expression of extracellular matrix genes. Conversely, overexpression of NKX2-5 suppressed the expression of contractile genes (ACTA2, TAGLN, CNN1) and enhanced the expression of matrix genes (COL1) in vascular smooth muscle cells. In vivo, conditional deletion of NKX2-5 attenuated blood vessel remodeling and halted the progression to hypertension in a mouse chronic hypoxia model. This study revealed that signals related to injury such as serum and low confluence, which induce NKX2-5 expression in cultured cells, is potentiated by TGF-β and further enhanced by hypoxia. The effect of TGF-β was sensitive to ERK5 and PI3K inhibition. Our data suggest a pivotal role for NKX2-5 in the phenotypic modulation of smooth muscle cells during pathological vascular remodeling and provide proof of concept for therapeutic targeting of NKX2-5 in vasculopathies.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Middle Aged
- Cell Proliferation/genetics
- Disease Models, Animal
- Homeobox Protein Nkx-2.5/genetics
- Homeobox Protein Nkx-2.5/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/genetics
- Pulmonary Arterial Hypertension/pathology
- Pulmonary Arterial Hypertension/etiology
- Scleroderma, Systemic/pathology
- Scleroderma, Systemic/complications
- Scleroderma, Systemic/metabolism
- Scleroderma, Systemic/genetics
- Transforming Growth Factor beta/metabolism
- Vascular Remodeling
Collapse
Affiliation(s)
- Ioannis Papaioannou
- Division of Medicine, Department of Inflammation, University College London, Royal Free Campus, London, United Kingdom
| | - Athina Dritsoula
- Division of Medicine, Department of Inflammation, University College London, Royal Free Campus, London, United Kingdom
| | - Ping Kang
- Division of Medicine, Department of Inflammation, University College London, Royal Free Campus, London, United Kingdom
| | - Reshma S. Baliga
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Sarah L. Trinder
- Division of Medicine, Department of Inflammation, University College London, Royal Free Campus, London, United Kingdom
| | - Emma Cook
- Division of Medicine, Department of Inflammation, University College London, Royal Free Campus, London, United Kingdom
| | - Xu Shiwen
- Division of Medicine, Department of Inflammation, University College London, Royal Free Campus, London, United Kingdom
| | - Adrian J. Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Christopher P. Denton
- Division of Medicine, Department of Inflammation, University College London, Royal Free Campus, London, United Kingdom
| | - David J. Abraham
- Division of Medicine, Department of Inflammation, University College London, Royal Free Campus, London, United Kingdom
| | - Markella Ponticos
- Division of Medicine, Department of Inflammation, University College London, Royal Free Campus, London, United Kingdom
| |
Collapse
|
3
|
Qin L, Liu N, Bao CLM, Yang DZ, Ma GX, Yi WH, Xiao GZ, Cao HL. Mesenchymal stem cells in fibrotic diseases-the two sides of the same coin. Acta Pharmacol Sin 2023; 44:268-287. [PMID: 35896695 PMCID: PMC9326421 DOI: 10.1038/s41401-022-00952-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 06/29/2022] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is caused by extensive deposition of extracellular matrix (ECM) components, which play a crucial role in injury repair. Fibrosis attributes to ~45% of all deaths worldwide. The molecular pathology of different fibrotic diseases varies, and a number of bioactive factors are involved in the pathogenic process. Mesenchymal stem cells (MSCs) are a type of multipotent stem cells that have promising therapeutic effects in the treatment of different diseases. Current updates of fibrotic pathogenesis reveal that residential MSCs may differentiate into myofibroblasts which lead to the fibrosis development. However, preclinical and clinical trials with autologous or allogeneic MSCs infusion demonstrate that MSCs can relieve the fibrotic diseases by modulating inflammation, regenerating damaged tissues, remodeling the ECMs, and modulating the death of stressed cells after implantation. A variety of animal models were developed to study the mechanisms behind different fibrotic tissues and test the preclinical efficacy of MSC therapy in these diseases. Furthermore, MSCs have been used for treating liver cirrhosis and pulmonary fibrosis patients in several clinical trials, leading to satisfactory clinical efficacy without severe adverse events. This review discusses the two opposite roles of residential MSCs and external MSCs in fibrotic diseases, and summarizes the current perspective of therapeutic mechanism of MSCs in fibrosis, through both laboratory study and clinical trials.
Collapse
Affiliation(s)
- Lei Qin
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Nian Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Chao-le-meng Bao
- CASTD Regengeek (Shenzhen) Medical Technology Co. Ltd, Shenzhen, 518000 China
| | - Da-zhi Yang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Gui-xing Ma
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| | - Wei-hong Yi
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Guo-zhi Xiao
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| | - Hui-ling Cao
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| |
Collapse
|
4
|
Alpinetin Suppresses Effects of TGF-β1 on Stimulating the Production and Organization of Fibrotic Markers in Human Primary Dermal Fibroblasts. Cells 2022; 11:cells11172731. [PMID: 36078140 PMCID: PMC9455016 DOI: 10.3390/cells11172731] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/22/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Overgrowths of dermal fibroblasts and myofibroblast phenoconversion in response to TGF-β stimulation are the hallmarks of skin fibrosis. Constitutive activation of dermal fibroblasts by TGF-β induces the excessive production of extracellular matrix as well as certain key intracellular proteins which form a complex interaction network. Current therapies include monoclonal anti-bodies against TGF-β and surgery, but these treatments generally elicit a limited effect on certain kinds of skin fibrosis. In the current study, we investigated the effects of alpinetin (AP) on human primary dermal fibroblasts (HPDFs) stimulated with TGF-β1. Results demonstrated that AP exhibited strong inhibitory effects on TGF-β1-induced proliferation and migration of HPDFs. AP also inhibited TGF-β1-induced morphological changes of fibroblasts to myofibroblasts, and these were found to be from its effects on blocking actin stress fiber formation and organization. The expression of major fibrotic molecules including α-SMA and type I collagen upon TGF-β1 stimulation was also inhibited by AP. In addition, AP attenuated TGF-β1-induced production and organization of vimentin, β-catenin, and N-cadherin, important for the pathophysiology of skin fibrosis. In conclusion, we revealed that AP has an ability to reverse the fibrotic effects of TGF-β1 at the cellular level, and this discovery suggests the therapeutic potential of AP for skin fibrosis.
Collapse
|
5
|
Animal Models of Systemic Sclerosis: Using Nailfold Capillaroscopy as a Potential Tool to Evaluate Microcirculation and Microangiopathy: A Narrative Review. Life (Basel) 2022; 12:life12050703. [PMID: 35629370 PMCID: PMC9147447 DOI: 10.3390/life12050703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/18/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease with three pathogenic hallmarks, i.e., inflammation, vasculopathy, and fibrosis. A wide plethora of animal models have been developed to address the complex pathophysiology and for the development of possible anti-fibrotic treatments. However, no current model comprises all three pathological mechanisms of the disease. To highlight the lack of a complete model, a review of some of the most widely used animal models for SSc was performed. In addition, to date, no model has accomplished the recreation of primary or secondary Raynaud’s phenomenon, a key feature in SSc. In humans, nailfold capillaroscopy (NFC) has been used to evaluate secondary Raynaud’s phenomenon and microvasculature changes in SSc. Being a non-invasive technique, it is widely used both in clinical studies and as a tool for clinical evaluation. Because of this, its potential use in animal models has been neglected. We evaluated NFC in guinea pigs to investigate the possibility of applying this technique to study microcirculation in the nailfold of animal models and in the future, development of an animal model for Raynaud’s phenomenon. The applications are not only to elucidate the pathophysiological mechanisms of vasculopathy but can also be used in the development of novel treatment options.
Collapse
|
6
|
The Evolution of Pharmacological Activities Bouea macrophylla Griffith In Vivo and In Vitro Study: A Review. Pharmaceuticals (Basel) 2022; 15:ph15020238. [PMID: 35215350 PMCID: PMC8880147 DOI: 10.3390/ph15020238] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 02/05/2023] Open
Abstract
Bouea macrophylla Griffith (B. macrophylla) is one of the many herbal plants found in Asia, and its fruit is plum mango. This plant is rich in secondary metabolites, including flavonoids, tannins, polyphenolic compounds, and many others. Due to its bioactive components, plum mango has powerful antioxidants that have therapeutic benefits for many common ailments, including cardiovascular disease, diabetes, and cancer. This review describes the evolution of plum mango’s phytochemical properties and pharmacological activities including in vitro and in vivo studies. The pharmacological activities of B. macrophylla Griffith reviewed in this article are antioxidant, anticancer, antihyperglycemic, antimicrobial, and antiphotoaging. Each of these pharmacological activities described and studied the possible cellular and molecular mechanisms of action. Interestingly, plum mango seeds show good pharmacological activity where the seed is the part of the plant that is a waste product. This can be an advantage because of its economic value as a herbal medicine. Overall, the findings described in this review aim to allow this plant to be explored and utilized more widely, especially as a new drug discovery.
Collapse
|
7
|
Wasson CW, Caballero-Ruiz B, Gillespie J, Derrett-Smith E, Mankouri J, Denton CP, Canettieri G, Riobo-Del Galdo NA, Del Galdo F. Induction of Pro-Fibrotic CLIC4 in Dermal Fibroblasts by TGF-β/Wnt3a Is Mediated by GLI2 Upregulation. Cells 2022; 11:cells11030530. [PMID: 35159339 PMCID: PMC8834396 DOI: 10.3390/cells11030530] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 11/16/2022] Open
Abstract
Chloride intracellular channel 4 (CLIC4) is a recently discovered driver of fibroblast activation in Scleroderma (SSc) and cancer-associated fibroblasts (CAF). CLIC4 expression and activity are regulated by TGF-β signalling through the SMAD3 transcription factor. In view of the aberrant activation of canonical Wnt-3a and Hedgehog (Hh) signalling in fibrosis, we investigated their role in CLIC4 upregulation. Here, we show that TGF-β/SMAD3 co-operates with Wnt3a/β-catenin and Smoothened/GLI signalling to drive CLIC4 expression in normal dermal fibroblasts, and that the inhibition of β-catenin and GLI expression or activity abolishes TGF-β/SMAD3-dependent CLIC4 induction. We further show that the expression of the pro-fibrotic marker α-smooth muscle actin strongly correlates with CLIC4 expression in dermal fibroblasts. Further investigations revealed that the inhibition of CLIC4 reverses morphogen-dependent fibroblast activation. Our data highlights that CLIC4 is a common downstream target of TGF-β, Hh, and Wnt-3a through signalling crosstalk and we propose a potential therapeutic avenue using CLIC4 inhibitors
Collapse
Affiliation(s)
- Christopher W. Wasson
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS29JT, UK; (J.G.); (F.D.G.)
- Correspondence:
| | - Begoña Caballero-Ruiz
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS29JT, UK; (B.C.-R.); (J.M.); (N.A.R.-D.G.)
- Department of Molecular Medicine, Sapienza University of Rome, 00196 Rome, Italy;
| | - Justin Gillespie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS29JT, UK; (J.G.); (F.D.G.)
| | - Emma Derrett-Smith
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, London NW32PF, UK; (E.D.-S.); (C.P.D.)
| | - Jamel Mankouri
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS29JT, UK; (B.C.-R.); (J.M.); (N.A.R.-D.G.)
| | - Christopher P. Denton
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, London NW32PF, UK; (E.D.-S.); (C.P.D.)
| | - Gianluca Canettieri
- Department of Molecular Medicine, Sapienza University of Rome, 00196 Rome, Italy;
| | - Natalia A. Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS29JT, UK; (B.C.-R.); (J.M.); (N.A.R.-D.G.)
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds LS29JT, UK
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS29JT, UK; (J.G.); (F.D.G.)
- Scleroderma Programme, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds LS29JT, UK
| |
Collapse
|
8
|
Asano Y. Insights Into the Preclinical Models of SSc. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2021. [DOI: 10.1007/s40674-021-00187-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
9
|
Derrett-Smith E, Clark KEN, Shiwen X, Abraham DJ, Hoyles RK, Lacombe O, Broqua P, Junien JL, Konstantinova I, Ong VH, Denton CP. The pan-PPAR agonist lanifibranor reduces development of lung fibrosis and attenuates cardiorespiratory manifestations in a transgenic mouse model of systemic sclerosis. Arthritis Res Ther 2021; 23:234. [PMID: 34488870 PMCID: PMC8419933 DOI: 10.1186/s13075-021-02592-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/26/2021] [Indexed: 12/02/2022] Open
Abstract
Background The TβRII∆k-fib transgenic (TG) mouse model of scleroderma replicates key fibrotic and vasculopathic complications of systemic sclerosis through fibroblast-directed upregulation of TGFβ signalling. We have examined peroxisome proliferator-activated receptor (PPAR) pathway perturbation in this model and explored the impact of the pan-PPAR agonist lanifibranor on the cardiorespiratory phenotype. Methods PPAR pathway gene and protein expression differences from TG and WT sex-matched littermate mice were determined at baseline and following administration of one of two doses of lanifibranor (30 mg/kg or 100 mg/kg) or vehicle administered by daily oral gavage up to 4 weeks. The prevention of bleomycin-induced lung fibrosis and SU5416-induced pulmonary hypertension by lanifibranor was explored. Results Gene expression data were consistent with the downregulation of the PPAR pathway in the TβRII∆k-fib mouse model. TG mice treated with high-dose lanifibranor demonstrated significant protection from lung fibrosis after bleomycin and from right ventricular hypertrophy following induction of pulmonary hypertension by SU5416, despite no significant change in right ventricular systolic pressure. Conclusions In the TβRII∆k-fib mouse strain, treatment with 100 mg/kg lanifibranor reduces the development of lung fibrosis and right ventricular hypertrophy induced by bleomycin or SU5416, respectively. Reduced PPAR activity may contribute to the exaggerated fibroproliferative response to tissue injury in this transgenic model of scleroderma and its pulmonary complications.
Collapse
Affiliation(s)
- Emma Derrett-Smith
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK
| | - Kristina E N Clark
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK
| | - Xu Shiwen
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK
| | - David J Abraham
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK
| | | | | | | | | | | | - Voon H Ong
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK
| | - Christopher P Denton
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, Rowland Hill St., London, NW3 2PF, UK.
| |
Collapse
|
10
|
Mouse Models of Skin Fibrosis. Methods Mol Biol 2021; 2299:371-383. [PMID: 34028755 DOI: 10.1007/978-1-0716-1382-5_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Systemic sclerosis (SSc) is a rare systemic autoimmune disease associated with a high mortality. The first histopathological hallmarks are vasculopathy and inflammation, followed by fibrosis of the skin and internal organs. The molecular and cellular mechanisms are incompletely understood. Rodent models provide important insights into the pathogenesis of SSc and are a mainstay for the development of novel targeted therapies. Here we describe the mechanistic insights of inducible and genetic models, and also discuss in detail the limitations and pitfalls of the most frequently used SSc mouse models. We also describe protocols for running the established bleomycin-induced scleroderma skin fibrosis model.
Collapse
|
11
|
Zhang Y, Yang B, Davis JM, Drake MM, Younes M, Shen Q, Zhao Z, Cao Y, Ko TC. Distinct Murine Pancreatic Transcriptomic Signatures during Chronic Pancreatitis Recovery. Mediators Inflamm 2021; 2021:5595464. [PMID: 34104113 PMCID: PMC8158417 DOI: 10.1155/2021/5595464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/15/2021] [Accepted: 04/25/2021] [Indexed: 11/17/2022] Open
Abstract
We have previously demonstrated that the pancreas can recover from chronic pancreatitis (CP) lesions in the cerulein-induced mouse model. To explore how pancreatic recovery is achieved at the molecular level, we used RNA-sequencing (seq) and profiled transcriptomes during CP transition to recovery. CP was induced by intraperitoneally injecting cerulein in C57BL/6 mice. Time-matched controls (CON) were given normal saline. Pancreata were harvested from mice 4 days after the final injections (designated as CP and CON) or 4 weeks after the final injections (designated as CP recovery (CPR) and control recovery (CONR)). Pancreatic RNAs were extracted for RNA-seq and quantitative (q) PCR validation. Using RNA-seq, we identified a total of 3,600 differentially expressed genes (DEGs) in CP versus CON and 166 DEGs in CPR versus CONR. There are 132 DEGs overlapped between CP and CPR and 34 DEGs unique to CPR. A number of selected pancreatic fibrosis-relevant DEGs were validated by qPCR. The top 20 gene sets enriched from DEGs shared between CP and CPR are relevant to extracellular matrix and cancer biology, whereas the top 10 gene sets enriched from DEGs specific to CPR are pertinent to DNA methylation and specific signaling pathways. In conclusion, we identified a distinct set of DEGs in association with extracellular matrix and cancer cell activities to contrast CP and CPR. Once during ongoing CP recovery, DEGs relevant to DNA methylation and specific signaling pathways were induced to express. The DEGs shared between CP and CPR and the DEGs specific to CPR may serve as the unique transcriptomic signatures and biomarkers for determining CP recovery and monitoring potential therapeutic responses at the molecular level to reflect pancreatic histological resolution.
Collapse
Affiliation(s)
- Yinjie Zhang
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Baibing Yang
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Joy M. Davis
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Madeline M. Drake
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Mamoun Younes
- Department of Pathology & Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Qiang Shen
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yanna Cao
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Tien C. Ko
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
12
|
Tam AYY, Horwell AL, Trinder SL, Khan K, Xu S, Ong V, Denton CP, Norman JT, Holmes AM, Bou-Gharios G, Abraham DJ. Selective deletion of connective tissue growth factor attenuates experimentally-induced pulmonary fibrosis and pulmonary arterial hypertension. Int J Biochem Cell Biol 2021; 134:105961. [PMID: 33662577 PMCID: PMC8111417 DOI: 10.1016/j.biocel.2021.105961] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/17/2022]
Abstract
Connective tissue growth factor (CTGF, CCN2) is a matricellular protein which plays key roles in normal mammalian development and in tissue homeostasis and repair. In pathological conditions, dysregulated CCN2 has been associated with cancer, cardiovascular disease, and tissue fibrosis. In this study, genetic manipulation of the CCN2 gene was employed to investigate the role of CCN2 expression in vitro and in experimentally-induced models of pulmonary fibrosis and pulmonary arterial hypertension (PAH). Knocking down CCN2 using siRNA reduced expression of pro-fibrotic markers (fibronectin p < 0.01, collagen type I p < 0.05, α-SMA p < 0.0001, TIMP-1 p < 0.05 and IL-6 p < 0.05) in TGF-β-treated lung fibroblasts derived from systemic sclerosis patients. In vivo studies were performed in mice using a conditional gene deletion strategy targeting CCN2 in a fibroblast-specific and time-dependent manner in two models of lung disease. CCN2 deletion significantly reduced pulmonary interstitial scarring and fibrosis following bleomycin-instillation, as assessed by fibrotic scores (wildtype bleomycin 3.733 ± 0.2667 vs CCN2 knockout (KO) bleomycin 4.917 ± 0.3436, p < 0.05) and micro-CT. In the well-established chronic hypoxia/Sugen model of pulmonary hypertension, CCN2 gene deletion resulted in a significant decrease in pulmonary vessel remodelling, less right ventricular hypertrophy and a reduction in the haemodynamic measurements characteristic of PAH (RVSP and RV/LV + S were significantly reduced (p < 0.05) in CCN2 KO compared to WT mice in hypoxic/SU5416 conditions). These results support a prominent role for CCN2 in pulmonary fibrosis and in vessel remodelling associated with PAH. Therefore, therapeutics aimed at blocking CCN2 function are likely to benefit several forms of severe lung disease.
Collapse
Affiliation(s)
- Angela Y Y Tam
- Centre for Rheumatology and Connective Tissue Disease, Department of Inflammation, Division of Medicine, University College London, London, NW3 2PF, UK.
| | - Amy L Horwell
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - Sarah L Trinder
- Centre for Rheumatology and Connective Tissue Disease, Department of Inflammation, Division of Medicine, University College London, London, NW3 2PF, UK
| | - Korsa Khan
- Centre for Rheumatology and Connective Tissue Disease, Department of Inflammation, Division of Medicine, University College London, London, NW3 2PF, UK
| | - Shiwen Xu
- Centre for Rheumatology and Connective Tissue Disease, Department of Inflammation, Division of Medicine, University College London, London, NW3 2PF, UK
| | - Voon Ong
- Centre for Rheumatology and Connective Tissue Disease, Department of Inflammation, Division of Medicine, University College London, London, NW3 2PF, UK
| | - Christopher P Denton
- Centre for Rheumatology and Connective Tissue Disease, Department of Inflammation, Division of Medicine, University College London, London, NW3 2PF, UK
| | - Jill T Norman
- Department of Renal Medicine, Division of Medicine, University College London, London, NW3 2PF, UK
| | - Alan M Holmes
- Centre for Rheumatology and Connective Tissue Disease, Department of Inflammation, Division of Medicine, University College London, London, NW3 2PF, UK
| | - George Bou-Gharios
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - David J Abraham
- Centre for Rheumatology and Connective Tissue Disease, Department of Inflammation, Division of Medicine, University College London, London, NW3 2PF, UK
| |
Collapse
|
13
|
Kleiser S, Nyström A. Interplay between Cell-Surface Receptors and Extracellular Matrix in Skin. Biomolecules 2020; 10:E1170. [PMID: 32796709 PMCID: PMC7465455 DOI: 10.3390/biom10081170] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
Skin consists of the epidermis and dermis, which are connected by a specialized basement membrane-the epidermal basement membrane. Both the epidermal basement membrane and the underlying interstitial extracellular matrix (ECM) created by dermal fibroblasts contain distinct network-forming macromolecules. These matrices play various roles in order to maintain skin homeostasis and integrity. Within this complex interplay of cells and matrices, cell surface receptors play essential roles not only for inside-out and outside-in signaling, but also for establishing mechanical and biochemical properties of skin. Already minor modulations of this multifactorial cross-talk can lead to severe and systemic diseases. In this review, major epidermal and dermal cell surface receptors will be addressed with respect to their interactions with matrix components as well as their roles in fibrotic, inflammatory or tumorigenic skin diseases.
Collapse
Affiliation(s)
- Svenja Kleiser
- Department of Dermatology, Faculty of Medicine and Medical Center, University of Freiburg, Hauptstraße 7, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine and Medical Center, University of Freiburg, Hauptstraße 7, 79104 Freiburg, Germany
| |
Collapse
|
14
|
Nihtyanova SI, Denton CP. Pathogenesis of systemic sclerosis associated interstitial lung disease. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2020; 5:6-16. [PMID: 35382227 PMCID: PMC8922569 DOI: 10.1177/2397198320903867] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022]
Abstract
Systemic sclerosis is an autoimmune disease leading to vasculopathy and fibrosis
of skin and internal organs. Despite likely shared pathogenic mechanisms, the
patterns of skin and lung fibrosis differ. Pathogenesis of interstitial lung
disease, a major cause of death in systemic sclerosis, reflects the intrinsic
disease pathobiology and is associated with distinct clinical phenotypes and
laboratory characteristics. The commonest histological pattern of systemic
sclerosis–interstitial lung disease is non-specific interstitial pneumonia.
Systemic sclerosis–interstitial lung disease pathogenesis involves multiple
components, including susceptibility and triggering factors, which could be
genetic or environmental. The process is amplified likely through ongoing
inflammation and the link between inflammatory activity and fibrosis with IL6
emerging as a key mediator. The disease is driven by epithelial injury,
reflected by markers in the serum, such as surfactant proteins and KL-6. In
addition, mediators that are produced by epithelial cells and that regulate
inflammatory cell trafficking may be important, especially CCL2. Other factors,
such as CXCL4 and CCL18, point towards immune-mediated damage or injury
response. Monocytes and alternatively activated macrophages appear to be
important. Transforming growth factor beta appears central to pathogenesis and
regulates epithelial repair and fibroblast activation. Understanding
pathogenesis may help to unravel the stages of systemic sclerosis–interstitial
lung disease, risks of progression and determinants of outcome. With this
article, we set out to review the multiple factors, including genetic,
environmental, cellular and molecular, that may be involved in the pathogenesis
of systemic sclerosis–interstitial lung disease and the mechanisms leading to
sustained fibrosis. We propose a model for the pathogenesis of systemic
sclerosis–interstitial lung disease, based on the available literature.
Collapse
Affiliation(s)
- Svetlana I Nihtyanova
- Centre for Rheumatology and Connective Tissue Diseases, University College London, London, UK
| | - Christopher P Denton
- Centre for Rheumatology and Connective Tissue Diseases, University College London, London, UK
| |
Collapse
|
15
|
Kim YS, Li Q, Youn HY, Kim DY. Oral Administration of Chitosan Attenuates Bleomycin-induced Pulmonary Fibrosis in Rats. In Vivo 2020; 33:1455-1461. [PMID: 31471392 DOI: 10.21873/invivo.11624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM Idiopathic pulmonary fibrosis (PF) is a fatal disorder of unknown aetiology with limited treatment options. Chitosan has antibacterial, antifungal, antioxidant, antitumour, and anti-inflammatory effects. This study aimed to investigate the effects of chitosan administration on bleomycin (BLM)-induced PF in rats. MATERIALS AND METHODS A PF rat model was established by endotracheal instillation of 5 mg/kg BLM; then, chitosan was administered in drinking water for 3 weeks. Histology, cell counts, and cytokine responses in the bronchoalveolar lavage fluid (BALF) and weight measurements (body and lung) were analyzed to assess its therapeutic effects. RESULTS Chitosan administration tended to reduce transforming growth factor (TGF)-β1 and interferon (IFN)-γ levels in BALF, and histopathological examination confirmed that chitosan attenuated the degree of inflammation and fibrosis in the lung. CONCLUSION This study revealed that oral chitosan exhibits potential antifibrotic effects, as measured by decreased proinflammatory cytokine levels and histological evaluation, in a BLM-induced PF rat model.
Collapse
Affiliation(s)
- You-Seok Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,KPC Corporation, Oporo, Gwangju, Republic of Korea
| | - Qiang Li
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hwa-Young Youn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Dae Young Kim
- Department of Life Science, College of Bio-nano Technology, Gachon University, Seongnam, Republic of Korea
| |
Collapse
|
16
|
Lin J, Vora M, Kane NS, Gleason RJ, Padgett RW. Human Marfan and Marfan-like Syndrome associated mutations lead to altered trafficking of the Type II TGFβ receptor in Caenorhabditis elegans. PLoS One 2019; 14:e0216628. [PMID: 31071172 PMCID: PMC6508650 DOI: 10.1371/journal.pone.0216628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/24/2019] [Indexed: 12/14/2022] Open
Abstract
The transforming growth factor-β (TGFβ) family plays an important role in many developmental processes and when mutated often contributes to various diseases. Marfan syndrome is a genetic disease with an occurrence of approximately 1 in 5,000. The disease is caused by mutations in fibrillin, which lead to an increase in TGFβ ligand activity, resulting in abnormalities of connective tissues which can be life-threatening. Mutations in other components of TGFβ signaling (receptors, Smads, Schnurri) lead to similar diseases with attenuated phenotypes relative to Marfan syndrome. In particular, mutations in TGFβ receptors, most of which are clustered at the C-terminal end, result in Marfan-like (MFS-like) syndromes. Even though it was assumed that many of these receptor mutations would reduce or eliminate signaling, in many cases signaling is active. From our previous studies on receptor trafficking in C. elegans, we noticed that many of these receptor mutations that lead to Marfan-like syndromes overlap with mutations that cause mis-trafficking of the receptor, suggesting a link between Marfan-like syndromes and TGFβ receptor trafficking. To test this hypothesis, we introduced three of these key MFS and MFS-like mutations into the C. elegans TGFβ receptor and asked if receptor trafficking is altered. We find that in every case studied, mutated receptors mislocalize to the apical surface rather than basolateral surface of the polarized intestinal cells. Further, we find that these mutations result in longer animals, a phenotype due to over-stimulation of the nematode TGFβ pathway and, importantly, indicating that function of the receptor is not abrogated in these mutants. Our nematode models of Marfan syndrome suggest that MFS and MFS-like mutations in the type II receptor lead to mis-trafficking of the receptor and possibly provides an explanation for the disease, a phenomenon which might also occur in some cancers that possess the same mutations within the type II receptor (e.g. colon cancer).
Collapse
MESH Headings
- Amino Acid Sequence
- Amino Acid Substitution
- Animals
- Animals, Genetically Modified
- Caenorhabditis elegans/genetics
- Caenorhabditis elegans/metabolism
- Caenorhabditis elegans Proteins/chemistry
- Caenorhabditis elegans Proteins/genetics
- Caenorhabditis elegans Proteins/metabolism
- Disease Models, Animal
- Humans
- Marfan Syndrome/genetics
- Marfan Syndrome/metabolism
- Mutation, Missense
- Protein Domains
- Receptor, Transforming Growth Factor-beta Type II/chemistry
- Receptor, Transforming Growth Factor-beta Type II/genetics
- Receptor, Transforming Growth Factor-beta Type II/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Transforming Growth Factor beta/chemistry
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Sequence Homology, Amino Acid
- Species Specificity
Collapse
Affiliation(s)
- Jing Lin
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
| | - Mehul Vora
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
- * E-mail: (MV); (RWP)
| | - Nanci S. Kane
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
| | - Ryan J. Gleason
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Richard W. Padgett
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
- * E-mail: (MV); (RWP)
| |
Collapse
|
17
|
Consequences of BMPR2 Deficiency in the Pulmonary Vasculature and Beyond: Contributions to Pulmonary Arterial Hypertension. Int J Mol Sci 2018; 19:ijms19092499. [PMID: 30149506 PMCID: PMC6165502 DOI: 10.3390/ijms19092499] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/17/2018] [Accepted: 08/18/2018] [Indexed: 12/18/2022] Open
Abstract
Since its association with familial pulmonary arterial hypertension (PAH) in 2000, Bone Morphogenetic Protein Receptor II (BMPR2) and its related signaling pathway have become recognized as a key regulator of pulmonary vascular homeostasis. Herein, we define BMPR2 deficiency as either an inactivation of the receptor, decreased receptor expression, or an impairment of the receptor’s downstream signaling pathway. Although traditionally the phenotypic consequences of BMPR2 deficiency in PAH have been thought to be limited to the pulmonary vasculature, there is evidence that abnormalities in BMPR2 signaling may have consequences in many other organ systems and cellular compartments. Revisiting how BMPR2 functions throughout health and disease in cells and organs beyond the lung vasculature may provide insight into the contribution of these organ systems to PAH pathogenesis as well as the potential systemic manifestation of PAH. Here we review our knowledge of the consequences of BMPR2 deficiency across multiple organ systems.
Collapse
|
18
|
Denton CP, Wells AU, Coghlan JG. Major lung complications of systemic sclerosis. Nat Rev Rheumatol 2018; 14:511-527. [DOI: 10.1038/s41584-018-0062-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
19
|
Gillespie J, Ross RL, Corinaldesi C, Esteves F, Derrett-Smith E, McDermott MF, Doody GM, Denton CP, Emery P, Del Galdo F. Transforming Growth Factor β Activation Primes Canonical Wnt Signaling Through Down-Regulation of Axin-2. Arthritis Rheumatol 2018; 70:932-942. [PMID: 29409120 DOI: 10.1002/art.40437] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/30/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Aberrant activation of Wnt signaling has been observed in tissues from patients with systemic sclerosis (SSc). This study aimed to determine the role of transforming growth factor β (TGFβ) in driving the increased Wnt signaling, through modulation of axis inhibition protein 2 (Axin-2), a critical regulator of the Wnt canonical pathway. METHODS Canonical Wnt signaling activation was analyzed by TOPflash T cell factor/lymphoid enhancer factor promoter assays. Axin-2 was evaluated in vitro by analysis of Axin-2 primary/mature transcript expression and decay, TGFβ receptor type I (TGFβRI) blockade, small interfering RNA-mediated depletion of tristetraprolin 1, and XAV-939-mediated Axin-2 stabilization. In vivo, Axin-2 messenger RNA (mRNA) and protein expression was determined in skin and lung biopsy samples from mice that express a kinase-deficient TGFβRII specifically on fibroblasts (TβRIIΔk-fib-transgenic mice) and from littermate controls. RESULTS SSc fibroblasts displayed an increased response to canonical Wnt ligands despite basal levels of Wnt signaling that were comparable to those in healthy control fibroblasts in vitro. Notably, we showed that SSc fibroblasts had reduced basal expression of Axin-2, which was caused by an endogenous TGFβ-dependent increase in Axin-2 mRNA decay. Accordingly, we observed that TGFβ decreased Axin-2 expression both in vitro in healthy control fibroblasts and in vivo in TβRIIΔk-fib-transgenic mice. Additionally, using Axin-2 gain- and loss-of-function experiments, we demonstrated that the TGFβ-induced increased response to Wnt activation characteristic of SSc fibroblasts depended on reduced bioavailability of Axin-2. CONCLUSION This study highlights the importance of reduced bioavailability of Axin-2 in mediating the increased canonical Wnt response observed in SSc fibroblasts. This novel mechanism extends our understanding of the processes involved in Wnt/β-catenin-driven pathology and supports the rationale for targeting the TGFβ pathway to regulate the aberrant Wnt signaling observed during fibrosis.
Collapse
Affiliation(s)
- Justin Gillespie
- University of Leeds and Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | | | | | | | | | | | | | - Paul Emery
- University of Leeds and Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW We discuss recent advances in evaluating and optimizing animal models of systemic sclerosis (SSc). Such models could be of value for illuminating etiopathogenesis using hypothesis-testing experimental approaches, for developing effective disease-modifying therapies, and for uncovering clinically relevant biomarkers. RECENT FINDINGS We describe recent advances in previously reported and novel animal models of SSc. The limitations of each animal model and their ability to recapitulate the pathophysiology of recognized molecular subsets of SSc are discussed. We highlight attrition of dermal white adipose tissue as a consistent pathological feature of dermal fibrosis in mouse models, and its relevance to SSc-associated cutaneous fibrosis. SUMMARY Several animal models potentially useful for studying SSc pathogenesis have been described. Recent studies highlight particular strengths and weaknesses of selected models in recapitulating distinct features of the human disease. When used in the appropriate experimental setting, and in combination, these models singly and together provide a powerful set of in-vivo tools to define underlying mechanisms of disease and to develop and evaluate effective antifibrotic therapies.
Collapse
|
21
|
Asano Y. Recent advances in animal models of systemic sclerosis. J Dermatol 2017; 43:19-28. [PMID: 26782003 DOI: 10.1111/1346-8138.13185] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 09/16/2015] [Indexed: 12/19/2022]
Abstract
Systemic sclerosis (SSc) is a multisystem connective tissue disease characterized by the three cardinal pathological features, comprising aberrant immune activation, vasculopathy and tissue fibrosis, with unknown etiology. Although many inducible and genetic animal models mimicking the selected aspects of SSc have been well documented, the lack of models encompassing the full clinical manifestations hindered the development and preclinical testing of therapies against this disease. Under this situation, three new genetic animal models have recently been established, such as Fra2 transgenic mice, urokinase-type plasminogen activator receptor deficient mice and Klf5(+/-) ;Fli1(+/-) mice, all of which recapitulate the pathological cascade of SSc. The former two murine models demonstrate endothelial cell apoptosis and capillary loss followed by tissue fibrosis, whereas the immune systems show no remarkable abnormality. Klf5(+/-) ;Fli1(+/-) mice develop immune activation, vasculopathy and tissue fibrosis in this sequence, eventually resulting in the development of dermal fibrosis, interstitial lung disease and pulmonary vascular involvement resembling those of SSc. Because Krueppel-like factor (KLF)5 and Friend leukemia integration 1 transcription factor (Fli1) are the transcription factors epigenetically suppressed in SSc dermal fibroblasts, the reproduction of SSc manifestations in Klf5(+/-) ;Fli1(+/-) mice supports the canonical idea that environmental influences play a central role in the development of SSc in genetically predisposed individuals. These new animal models offer important clues for the better understanding of the underlying molecular mechanisms of SSc pathology and the identification of potential molecular targets for the treatment of this incurable disease.
Collapse
Affiliation(s)
- Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Li IMH, Horwell AL, Chu G, de Crombrugghe B, Bou-Gharios G. Characterization of Mesenchymal-Fibroblast Cells Using the Col1a2 Promoter/Enhancer. Methods Mol Biol 2017; 1627:139-161. [PMID: 28836200 DOI: 10.1007/978-1-4939-7113-8_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Excessive deposition of extracellular matrix (ECM) is a common hallmark of fibrotic diseases in various organs. Chiefly among this ECM are collagen types I and III, secreted by local fibroblasts, and other mesenchymal cells recruited for repair purposes. In the last two decades, the search for a fibroblast-specific promoter/enhancer has intensified in order to control the regulation of ECM in these cells and limit the scarring of the fibrotic process. In our previous work, we characterized an enhancer region 17 kb upstream of the Col1a2 gene transcription start site. This enhancer in transgenic mice is expressed mainly in mesenchymal cells during development and in adults upon injury. When driving transgenes such as beta-galactosidase or luciferase, this construct acts as an informative reporter of collagen transcription and is predictive of collagen type I deposition. In this chapter, we provide detailed protocols for identifying similar enhancers and using the sequence to generate a construct for transfection and producing transgenic animals. We also provided information on the use of luminescence in transgenic mice, tissue processing, as well as using cre/lox system to obtain conditional gain and loss of function in mice.
Collapse
Affiliation(s)
- Ian M H Li
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Amy L Horwell
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Grace Chu
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | | - George Bou-Gharios
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK.
| |
Collapse
|
23
|
Skin fibrosis: Models and mechanisms. Curr Res Transl Med 2016; 64:185-193. [PMID: 27939457 DOI: 10.1016/j.retram.2016.06.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/15/2016] [Accepted: 06/17/2016] [Indexed: 02/06/2023]
Abstract
Matrix synthesis, deposition and remodeling are complex biological processes that are critical in development, maintenance of tissue homeostasis and repair of injured tissues. Disturbances in the regulation of these processes can result in severe pathological conditions which are associated with tissue fibrosis as e.g. in Scleroderma, cutaneous Graft-versus-Host-Disease, excessive scarring after trauma or carcinogenesis. Therefore, finding efficient treatments to limit skin fibrosis is of major clinical importance. However the pathogenesis underlying the development of tissue fibrosis is still not entirely resolved. In recent years progress has been made unraveling the complex cellular and molecular mechanisms that determine fibrosis. Here we provide an overview of established and more recently developed mouse models that can be used to investigate the mechanisms of skin fibrosis and to test potential therapeutic approaches.
Collapse
|
24
|
Ikeda T, Fragiadaki M, Shi-wen X, Ponticos M, Khan K, Denton C, Garcia P, Bou-Gharios G, Yamakawa A, Morimoto C, Abraham D. Transforming growth factor- β-induced CUX1 isoforms are associated with fibrosis in systemic sclerosis lung fibroblasts. Biochem Biophys Rep 2016; 7:246-252. [PMID: 28955913 PMCID: PMC5613511 DOI: 10.1016/j.bbrep.2016.06.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 01/21/2023] Open
Abstract
In the enhancer region of the human type I collagen alpha 2 (COL1A2) gene, we identified cis-elements for the transcription factor CUX1. However, the role of CUX1 in fibrosis remains unclear. Here we investigated the role of CUX1 in the regulation of COL1 expression and delineated the mechanisms underlying the regulation of COL1A2 expression by CUX1 in systemic sclerosis (SSc) lung fibroblasts. The binding of CUX1 to the COL1A2 enhancer region was assessed using electrophoretic mobility shift assays after treatment with transforming growth factor (TGF)-β. Subsequently, the protein expression levels of CUX1 isoforms were determined using Western blotting. Finally, the expression levels of COL1 and fibrosis-related cytokines, including CTGF, ET-1, Wnt1 and β-catenin were determined. The binding of CUX1 isoforms to the COL1A2 enhancer region increased after TGF-β treatment. TGF-β also increased the protein levels of the CUX1 isoforms p200, p150, p110, p75, p30 and p28. Moreover, SSc lung fibroblasts showed higher levels of CUX1 isoforms than normal lung fibroblasts, and treatment of SSc lung fibroblasts with a cathepsin L inhibitor (IW-CHO) decreased COL1 protein expression and reduced cell size, as measured using immunocytochemistry. In SSc and diffuse alveolar damage lung tissue sections, CUX1 localised within α-smooth muscle actin-positive cells. Our results suggested that CUX1 isoforms play vital roles in connective tissue deposition during wound repair and fibrosis.
Collapse
Affiliation(s)
- Tetsurou Ikeda
- Royal Free and University College Medical School, London, UK
- Imperial College School of Medicine, London, UK
- University of Tokyo, Institute of Medical Science, Tokyo, Japan
| | | | - Xu Shi-wen
- Royal Free and University College Medical School, London, UK
| | | | - Korsa Khan
- Royal Free and University College Medical School, London, UK
| | | | - Patricia Garcia
- Royal Free and University College Medical School, London, UK
| | | | - Akio Yamakawa
- University of Tokyo, Institute of Medical Science, Tokyo, Japan
| | - Chikao Morimoto
- University of Tokyo, Institute of Medical Science, Tokyo, Japan
| | - David Abraham
- Royal Free and University College Medical School, London, UK
| |
Collapse
|
25
|
Maria ATJ, Toupet K, Bony C, Pirot N, Vozenin MC, Petit B, Roger P, Batteux F, Le Quellec A, Jorgensen C, Noël D, Guilpain P. Antifibrotic, Antioxidant, and Immunomodulatory Effects of Mesenchymal Stem Cells in HOCl-Induced Systemic Sclerosis. Arthritis Rheumatol 2016; 68:1013-25. [PMID: 26474311 DOI: 10.1002/art.39477] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 10/13/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is a rare intractable disease with unmet medical need and fibrosis-related mortality. Absence of efficient treatments has prompted the development of novel therapeutic strategies, among which mesenchymal stem cells/stromal cells (MSCs) or progenitor stromal cells appear to be one of the most attractive options. The purpose of this study was to use the murine model of hypochlorite-induced SSc to investigate the systemic effects of MSCs on the main features of the diffuse form of the disease: skin and lung fibrosis, autoimmunity, and oxidative status. METHODS We compared the effects of different doses of MSCs (2.5 × 10(5) , 5 × 10(5) , and 10(6) ) infused at different time points. Skin thickness was assessed during the experiment. At the time of euthanasia, biologic parameters were quantified in blood and tissues (by enzyme-linked immunosorbent assay, quantitative reverse transcription-polymerase chain reaction, assessment of collagen content). Assessments of histology and immunostaining were also performed. RESULTS A lower expression of markers of fibrosis (Col1, Col3, Tgfb1, and aSma) was observed in both skin and lung following MSC infusion, which was consistent with histologic improvement and was inversely proportional to the injected dose. Importantly, sera from treated mice exhibited lower levels of anti-Scl-70 autoantibodies and enhanced antioxidant capacity, confirming the systemic effect of MSCs. Of interest, MSC administration was efficient in both the preventive and the curative approach. We further provide evidence that MSCs exerted an antifibrotic role by normalizing extracellular matrix remodeling parameters as well as reducing proinflammatory cytokine levels and increasing antioxidant defenses. CONCLUSION The results of this study demonstrate the beneficial and systemic effects of MSC administration in the HOCl murine model of diffuse SSc, which is a promising finding from a clinical perspective.
Collapse
Affiliation(s)
- Alexandre T J Maria
- INSERM U1183, St. Eloi Hospital, and Montpellier University Medical School, Montpellier, France
| | - Karine Toupet
- INSERM U1183, St. Eloi Hospital, and Montpellier University Medical School, Montpellier, France
| | - Claire Bony
- INSERM U1183, St. Eloi Hospital, and Montpellier University Medical School, Montpellier, France
| | - Nelly Pirot
- INSERM U1194 and UMS BioCampus Montpellier, Montpellier, France
| | | | - Benoît Petit
- University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Pascal Roger
- Montpellier University Medical School, Montpellier, France, and Caremeau Hospital, Nîmes, France
| | | | - Alain Le Quellec
- St. Eloi Hospital and Montpellier University Medical School, Montpellier, France
| | - Christian Jorgensen
- INSERM U1183, St. Eloi Hospital, Montpellier University Medical School, and Lapeyronie Hospital, Montpellier, France
| | - Danièle Noël
- INSERM U1183, St. Eloi Hospital, and Montpellier University Medical School, Montpellier, France
| | - Philippe Guilpain
- INSERM U1183, St. Eloi Hospital, and Montpellier University Medical School, Montpellier, France
| |
Collapse
|
26
|
Abstract
Systemic sclerosis (SSc) is a connective tissue disease of unknown etiology that is characterized by fibrosis of the skin and several internal organs, vasculopathy, inflammation and autoimmunity. Animal models have improved our understanding of the pathogenesis of SSc. Many inducible and genetic animal models of SSc have been developed and characterized in the last years. All of these models have different strengths and limitations and mimic different aspects of the pathogenesis of SSc. The purpose of this review is to summarize the characteristics of the various animal models of SSc and to provide an outline of how to use these models to study certain aspects in the pathogenesis of SSc and to test the effects of potential therapeutic approaches.
Collapse
|
27
|
Abstract
Myofibroblast-derived αvβ1 integrin has emerged as a critical mediator of transforming growth factor-β activation in experimental models of tissue fibrosis (Reed et al., this issue).
Collapse
Affiliation(s)
- Harry C Dietz
- Institute of Genetic Medicine, Howard Hughes Medical Institute, and Smilow Center for Marfan Syndrome Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
28
|
Tsujino K, Sheppard D. Critical Appraisal of the Utility and Limitations of Animal Models of Scleroderma. Curr Rheumatol Rep 2015; 18:4. [DOI: 10.1007/s11926-015-0553-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
29
|
Turner NA. Inflammatory and fibrotic responses of cardiac fibroblasts to myocardial damage associated molecular patterns (DAMPs). J Mol Cell Cardiol 2015; 94:189-200. [PMID: 26542796 DOI: 10.1016/j.yjmcc.2015.11.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/30/2015] [Accepted: 11/01/2015] [Indexed: 02/07/2023]
Abstract
Cardiac fibroblasts (CF) are well-established as key regulators of extracellular matrix (ECM) turnover in the context of myocardial remodelling and fibrosis. Recently, this cell type has also been shown to act as a sensor of myocardial damage by detecting and responding to damage-associated molecular patterns (DAMPs) upregulated with cardiac injury. CF express a range of innate immunity pattern recognition receptors (TLRs, NLRs, IL-1R1, RAGE) that are stimulated by a host of different DAMPs that are evident in the injured or remodelling myocardium. These include intracellular molecules released by necrotic cells (heat shock proteins, high mobility group box 1 protein, S100 proteins), proinflammatory cytokines (interleukin-1α), specific ECM molecules up-regulated in response to tissue injury (fibronectin-EDA, tenascin-C) or molecules modified by a pathological environment (advanced glycation end product-modified proteins observed with diabetes). DAMP receptor activation on fibroblasts is coupled to altered cellular function including changes in proliferation, migration, myofibroblast transdifferentiation, ECM turnover and production of fibrotic and inflammatory paracrine factors, which directly impact on the heart's ability to respond to injury. This review gives an overview of the important role played by CF in responding to myocardial DAMPs and how the DAMP/CF axis could be exploited experimentally and therapeutically.
Collapse
Affiliation(s)
- Neil A Turner
- Division of Cardiovascular & Diabetes Research, and Multidisciplinary Cardiovascular Research Centre (MCRC), University of Leeds, Leeds, UK.
| |
Collapse
|
30
|
Asano Y. Double heterozygous mice for Klf5 and Fli1 genes: a new animal model of systemic sclerosis recapitulating its three cardinal pathological features. Med Mol Morphol 2015; 48:123-8. [DOI: 10.1007/s00795-015-0107-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/12/2015] [Indexed: 01/09/2023]
|
31
|
Gilbane AJ, Derrett-Smith E, Trinder SL, Good RB, Pearce A, Denton CP, Holmes AM. Impaired Bone Morphogenetic Protein Receptor II Signaling in a Transforming Growth Factor-β–Dependent Mouse Model of Pulmonary Hypertension and in Systemic Sclerosis. Am J Respir Crit Care Med 2015; 191:665-77. [DOI: 10.1164/rccm.201408-1464oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
32
|
Dickneite G, Herwald H, Korte W, Allanore Y, Denton CP, Matucci Cerinic M. Coagulation factor XIII: a multifunctional transglutaminase with clinical potential in a range of conditions. Thromb Haemost 2015; 113:686-97. [PMID: 25652913 DOI: 10.1160/th14-07-0625] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/27/2014] [Indexed: 12/28/2022]
Abstract
Coagulation factor XIII (FXIII), a plasma transglutaminase, is best known as the final enzyme in the coagulation cascade, where it is responsible for cross-linking of fibrin. However, a growing body of evidence has demonstrated that FXIII targets a wide range of additional substrates that have important roles in health and disease. These include antifibrinolytic proteins, with cross-linking of α2-antiplasmin to fibrin, and potentially fibrinogen, being the principal mechanism(s) whereby plasmin-mediated clot degradation is minimised. FXIII also acts on endothelial cell VEGFR-2 and αvβ3 integrin, which ultimately leads to downregulation of the antiangiogenic protein thrombospondin-1, promoting angiogenesis and neovascularisation. Under infectious disease conditions, FXIII cross-links bacterial surface proteins to fibrinogen, resulting in immobilisation and killing, while during wound healing, FXIII induces cross-linking of the provisional matrix. The latter process has been shown to influence the interaction of leukocytes with the provisional extracellular matrix and promote wound healing. Through these actions, there are good rationales for evaluating the therapeutic potential of FXIII in diseases in which tissue repair is dysregulated or perturbed, including systemic sclerosis (scleroderma), invasive bacterial infections, and tissue repair, for instance healing of venous leg ulcers or myocardial injuries. Adequate levels of FXIII are also required in patients undergoing surgery to prevent or treat perioperative bleeding, and its augmentation in patients with/at risk for perioperative bleeding may also have potential clinical benefit. While there are preclinical and/or clinical data to support the use of FXIII in a range of settings, further clinical evaluation in these underexplored applications is warranted.
Collapse
Affiliation(s)
- Gerhard Dickneite
- Prof. Dr Gerhard Dickneite, Preclinical R&D, CSL Behring, PO Box 1230, 35002 Marburg, Germany, Tel.: +49 6421 392306, Fax: +49 6421 394663, E-mail:
| | | | | | | | | | | |
Collapse
|
33
|
Hamburg-Shields E, DiNuoscio GJ, Mullin NK, Lafyatis R, Atit RP. Sustained β-catenin activity in dermal fibroblasts promotes fibrosis by up-regulating expression of extracellular matrix protein-coding genes. J Pathol 2015; 235:686-97. [PMID: 25385294 DOI: 10.1002/path.4481] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 11/02/2014] [Accepted: 11/05/2014] [Indexed: 12/11/2022]
Abstract
Fibrosis is an end-stage response to tissue injury that is associated with loss of organ function as a result of excess extracellular matrix (ECM) production by fibroblasts. In skin, pathological fibrosis is evident during keloid scar formation, systemic sclerosis (SSc) and morphea. Dermal fibroblasts in these fibrotic diseases exhibit increased Wnt/β-catenin signalling, a pathway that is sufficient to cause fibrosis in mice. However, in the context of this complex pathology, the precise pro-fibrotic consequences of Wnt/β-catenin signalling are not known. We found that expression of stabilized β-catenin in mouse dermal fibroblasts resulted in spontaneous, progressive skin fibrosis with thickened collagen fibres and altered collagen fibril morphology. The fibrotic phenotype was predominated by resident dermal fibroblasts. Genome-wide profiling of the fibrotic mouse dermis revealed elevated expression of matrix-encoding genes, and the promoter regions of these genes were enriched for Tcf/Lef family transcription factor binding sites. Additionally, we identified 32 β-catenin-responsive genes in our mouse model that are also over-expressed in human fibrotic tissues and poised for regulation by Tcf/Lef family transcription factors. Therefore, we have uncovered a matrix-regulatory role for stabilized β-catenin in fibroblasts in vivo and have defined a set of β-catenin-responsive genes with relevance to fibrotic disease.
Collapse
|
34
|
Gilbane AJ, Denton CP, Holmes AM. Scleroderma pathogenesis: a pivotal role for fibroblasts as effector cells. Arthritis Res Ther 2014; 15:215. [PMID: 23796020 PMCID: PMC4060542 DOI: 10.1186/ar4230] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Scleroderma (systemic sclerosis; SSc) is characterised by fibrosis of the skin and internal organs in the context of autoimmunity and vascular perturbation. Overproduction of extracellular matrix components and loss of specialised epithelial structures are analogous to the process of scar formation after tissue injury. Fibroblasts are the resident cells of connective tissue that become activated at sites of damage and are likely to be important effector cells in SSc. Differentiation into myofibroblasts is a hallmark process, although the mechanisms and cellular origins of this important fibroblastic cell are still unclear. This article reviews fibroblast biology in the context of SSc and highlights the potentially important place of fibroblast effector cells in fibrosis. Moreover, the heterogeneity of fibroblast properties, multiplicity of regulatory pathways and diversity of origin for myofibroblasts may underpin clinical diversity in SSc, and provide novel avenues for targeted therapy.
Collapse
|
35
|
Wang L, Sun Y, Ruan C, Liu B, Zhao L, Gu X. Angelica sinensis is effective in treating diffuse interstitial pulmonary fibrosis in rats. BIOTECHNOL BIOTEC EQ 2014; 28:923-928. [PMID: 26019579 PMCID: PMC4433952 DOI: 10.1080/13102818.2014.957487] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 06/03/2014] [Indexed: 11/05/2022] Open
Abstract
The aim of this study was to investigate the therapeutic effect of Angelica sinensis on a rat model of diffuse interstitial pulmonary fibrosis induced by bleomycin A5. The mechanism by which A. sinensis exerts its effect is also discussed. A diffuse interstitial pulmonary fibrosis model was established in 36 male Wistar rats by an endotracheal injection of bleomycin A5 (5 mg/kg). Then, these rats were randomly divided into the model group (n = 18) and the treatment group (treated with A. sinensis after modelling, n = 18). Control rats (n = 6) received an equal volume of saline. Hematoxylin–eosin (HE) staining was performed to analyse alveolitis and Masson staining, to observe pulmonary fibrosis. Collagen content was determined by hydroxyproline assay. Nuclear factor kappa B (NF-κB) activity was measured by electrophoretic mobility shift assay. Transforming growth factor-β (TGF-β) expression at mRNA level was detected by northern blotting and at protein level by enzyme-linked immunosorbent assay. The results obtained showed that the alveolitis and pulmonary fibrosis of the rats treated with A. sinensis was significantly alleviated compared with that of the rats in the model group. Treatment with A. sinensis also lowered the content of collagen, decreased NF-κB activity in alveolar macrophages and reduced the TGF-β expression at the mRNA and protein level. These results indicated that A. sinensis is effective in treating and alleviating interstitial pulmonary fibrosis, possibly by lowering collagen, inhibiting the activity of NF-κB and reducing the TGF-β expression.
Collapse
Affiliation(s)
- Lu Wang
- Department of Human Anatomy, School of Medical Sciences, Yan'an University , Yan'an , Shaanxi , P.R. China
| | - Yanmei Sun
- Department of Internal Medicine, Qinghua Hospital , Xi'an , Shaanxi , P.R. China
| | - Cailian Ruan
- Department of Human Anatomy, School of Medical Sciences, Yan'an University , Yan'an , Shaanxi , P.R. China
| | - Bofeng Liu
- Department of Human Anatomy, School of Medical Sciences, Yan'an University , Yan'an , Shaanxi , P.R. China
| | - Lin Zhao
- Department of Human Anatomy, School of Medical Sciences, Yan'an University , Yan'an , Shaanxi , P.R. China
| | - Xiujuan Gu
- Department of Inspection Division, Yan'an University Affiliated Hospital , Yan'an , Shaanxi , P.R. China
| |
Collapse
|
36
|
Abstract
Pulmonary complications are an important extra-articular feature of autoimmune rheumatic diseases and a major cause of mortality. The underlying pathogenesis probably involves multiple cellular compartments, including the epithelium, lung fibroblasts, and the innate and adaptive immune system. Heterogeneity in the extent and progression of lung fibrosis probably reflects differences in underlying pathogenic mechanisms. Growing understanding of the key pathogenic drivers of lung fibrosis might lead to the development of more effective targeted therapies to replicate the treatment advances in other aspects of these diseases. Interstitial lung disease (ILD) in connective tissue disease (CTD) is characterized using the classification of the idiopathic interstitial pneumonias. Systemic sclerosis is most frequently associated with ILD and, in most of these patients, ILD manifests as a histological pattern of nonspecific interstitial pneumonia. Conversely, in rheumatoid arthritis, the pattern of ILD is most often usual interstitial pneumonia. The key goals of clinical assessment of patients with both ILD and CTD are the detection of ILD and prognostic evaluation to determine which patients should be treated. Data from treatment trials in systemic sclerosis support the use of immunosuppressive therapy, with the treatment benefit largely relating to the prevention of progression of lung disease.
Collapse
|
37
|
Wells AU. Interstitial lung disease in systemic sclerosis. Presse Med 2014; 43:e329-43. [PMID: 25217474 DOI: 10.1016/j.lpm.2014.08.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 08/21/2014] [Indexed: 12/12/2022] Open
Abstract
Based on international collaborative data, interstitial lung disease is now the most frequent cause of death in systemic sclerosis (SSc), having supplanted renal crisis in that regard. Despite detailed explorations of candidate mediators, no primary pathway in the pathogenesis of interstitial lung disease associated with SSc (SSc-ILD) has been definitively identified and, therefore, treatment with current agents is only partially successful. However, as immunomodulatory agents do, on average, retard progression of lung disease, early identification of SSc-ILD, using thoracic high resolution computed tomography (HRCT), is highly desirable. The decision whether to introduce therapy immediately is often difficult as the balance of risk and benefit favours a strategy of careful observation when lung disease is very limited, especially in long-standing SSc. The threshold for initiating treatment is substantially reduced when lung disease is severe, systemic disease is short in duration or ongoing progression is evident, based on pulmonary function tests and symptoms. This review summarises epidemiology, pathogenesis, difficult clinical problems and management issues in SSc-ILD.
Collapse
Affiliation(s)
- Athol U Wells
- Royal Brompton hospital, interstitial lung disease unit, Sydney street, Chelsea, London SW3 6HP, United Kingdom.
| |
Collapse
|
38
|
Wellbrock J, Sheikhzadeh S, Oliveira-Ferrer L, Stamm H, Hillebrand M, Keyser B, Klokow M, Vohwinkel G, Bonk V, Otto B, Streichert T, Balabanov S, Hagel C, Rybczynski M, Bentzien F, Bokemeyer C, von Kodolitsch Y, Fiedler W. Overexpression of Gremlin-1 in patients with Loeys-Dietz syndrome: implications on pathophysiology and early disease detection. PLoS One 2014; 9:e104742. [PMID: 25116393 PMCID: PMC4130545 DOI: 10.1371/journal.pone.0104742] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/14/2014] [Indexed: 11/23/2022] Open
Abstract
Backgrounds The Loeys-Dietz syndrome (LDS) is an inherited connective tissue disorder caused by mutations in the transforming growth factor β (TGF-β) receptors TGFBR1 or TGFBR2. Most patients with LDS develop severe aortic aneurysms resulting in early need of surgical intervention. In order to gain further insight into the pathophysiology of the disorder, we investigated circulating outgrowth endothelial cells (OEC) from the peripheral blood of LDS patients from a cohort of 23 patients including 6 patients with novel TGF-β receptor mutations. Methods and Results We performed gene expression profiling of OECs using microarray analysis followed by quantitative PCR for verification of gene expression. Compared to OECs of age- and sex-matched healthy controls, OECs isolated from three LDS patients displayed altered expression of several genes belonging to the TGF-β pathway, especially those affecting bone morphogenic protein (BMP) signalling including BMP2, BMP4 and BMPR1A. Gene expression of BMP antagonist Gremlin-1 (GREM1) showed the most prominent up-regulation. This increase was confirmed at the protein level by immunoblotting of LDS-OECs. In immunohistochemistry, abundant Gremlin-1 protein expression could be verified in endothelial cells as well as smooth muscle cells within the arterial media. Furthermore, Gremlin-1 plasma levels of LDS patients were significantly elevated compared to healthy control subjects. Conclusions These findings open new avenues in the understanding of the pathogenesis of Loeys-Dietz syndrome and the development of new diagnostic serological methods for early disease detection.
Collapse
Affiliation(s)
- Jasmin Wellbrock
- Hubertus Wald University Cancer Centre, Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| | - Sara Sheikhzadeh
- Center of Cardiology and Cardiovascular Surgery, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Leticia Oliveira-Ferrer
- Hubertus Wald University Cancer Centre, Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Hauke Stamm
- Hubertus Wald University Cancer Centre, Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Mathias Hillebrand
- Center of Cardiology and Cardiovascular Surgery, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Britta Keyser
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Marianne Klokow
- Hubertus Wald University Cancer Centre, Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Gabi Vohwinkel
- Hubertus Wald University Cancer Centre, Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Veronika Bonk
- Hubertus Wald University Cancer Centre, Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Otto
- Department of Clinical Chemistry/Central Laboratories, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Streichert
- Department of Clinical Chemistry/Central Laboratories, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Balabanov
- Hubertus Wald University Cancer Centre, Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Christian Hagel
- Institute for Neuropathology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Meike Rybczynski
- Center of Cardiology and Cardiovascular Surgery, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Frank Bentzien
- Department of Transfusion Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Hubertus Wald University Cancer Centre, Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Yskert von Kodolitsch
- Center of Cardiology and Cardiovascular Surgery, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Hubertus Wald University Cancer Centre, Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
39
|
Abstract
Without doubt, animal models have provided significant insights into our understanding of the rheumatological diseases; however, no model has accurately replicated all aspects of any autoimmune disease. Recent years have seen a plethora of knockouts and transgenics that have contributed to our knowledge of the initiating events of systemic sclerosis, an autoimmune disease. In this review, the focus is on models of systemic sclerosis and how they have progressed our understanding of fibrosis and vasculopathy, and whether they are relevant to the pathogenesis of systemic sclerosis.
Collapse
Affiliation(s)
- Carol M Artlett
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
40
|
Matsushita T, Fujimoto M. Scleroderma: recent lessons from murine models and implications for future therapeutics. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17469872.2013.835924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
41
|
Wheeler JB, Ikonomidis JS, Jones JA. Connective tissue disorders and cardiovascular complications: the indomitable role of transforming growth factor-beta signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 802:107-27. [PMID: 24443024 PMCID: PMC4410689 DOI: 10.1007/978-94-007-7893-1_8] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Marfan Syndrome (MFS) and Loeys-Dietz Syndrome (LDS) represent heritable connective tissue disorders that cosegregate with a similar pattern of cardiovascular defects (thoracic aortic aneurysm, mitral valve prolapse/regurgitation, and aortic root dilatation with regurgitation). This pattern of cardiovascular defects appears to be expressed along a spectrum of severity in many heritable connective tissue disorders and raises suspicion of a relationship between the normal development of connective tissues and the cardiovascular system. Given the evidence of increased transforming growth factor-beta (TGF-β) signaling in MFS and LDS, this signaling pathway may represent the common link in this relationship. To further explore this hypothetical link, this chapter will review the TGF-β signaling pathway, heritable connective tissue syndromes related to TGF-β receptor (TGFBR) mutations, and discuss the pathogenic contribution of TGF-β to these syndromes with a primary focus on the cardiovascular system.
Collapse
MESH Headings
- Adrenergic beta-Antagonists/therapeutic use
- Angiotensin II Type 1 Receptor Blockers/therapeutic use
- Antibodies, Neutralizing/pharmacology
- Aortic Aneurysm, Thoracic/drug therapy
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/surgery
- Aortic Valve/pathology
- Aortic Valve/surgery
- Bicuspid Aortic Valve Disease
- Gene Expression Regulation
- Heart Defects, Congenital/drug therapy
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/pathology
- Heart Defects, Congenital/surgery
- Heart Valve Diseases/drug therapy
- Heart Valve Diseases/genetics
- Heart Valve Diseases/pathology
- Heart Valve Diseases/surgery
- Humans
- Loeys-Dietz Syndrome/drug therapy
- Loeys-Dietz Syndrome/genetics
- Loeys-Dietz Syndrome/pathology
- Loeys-Dietz Syndrome/surgery
- Marfan Syndrome/drug therapy
- Marfan Syndrome/genetics
- Marfan Syndrome/pathology
- Marfan Syndrome/surgery
- Mutation
- Receptors, Transforming Growth Factor beta/genetics
- Signal Transduction/genetics
- Smad Proteins/genetics
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/genetics
Collapse
Affiliation(s)
- Jason B. Wheeler
- Division of Cardiothoracic Surgery, Medical University of South Carolina
| | - John S. Ikonomidis
- Division of Cardiothoracic Surgery, Medical University of South Carolina
| | - Jeffrey A. Jones
- Division of Cardiothoracic Surgery, Medical University of South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| |
Collapse
|
42
|
Derrett-Smith EC, Dooley A, Gilbane AJ, Trinder SL, Khan K, Baliga R, Holmes AM, Hobbs AJ, Abraham D, Denton CP. Endothelial injury in a transforming growth factor β-dependent mouse model of scleroderma induces pulmonary arterial hypertension. ARTHRITIS AND RHEUMATISM 2013; 65:2928-39. [PMID: 23839959 DOI: 10.1002/art.38078] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 06/27/2013] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To delineate the constitutive pulmonary vascular phenotype of the TβRIIΔk-fib mouse model of scleroderma, and to selectively induce pulmonary endothelial cell injury using vascular endothelial growth factor (VEGF) inhibition to develop a model with features characteristic of pulmonary arterial hypertension (PAH). METHODS The TβRIIΔk-fib mouse strain expresses a kinase-deficient transforming growth factor β (TGFβ) receptor type II driven by a fibroblast-specific promoter, leading to ligand-dependent up-regulation of TGFβ signaling, and replicates key fibrotic features of scleroderma. Structural, biochemical, and functional assessments of pulmonary vessels, including in vivo hemodynamic studies, were performed before and following VEGF inhibition, which induced pulmonary endothelial cell apoptosis. These assessments included biochemical analysis of the TGFβ and VEGF signaling axes in tissue sections and explanted smooth muscle cells. RESULTS In the TβRIIΔk-fib mouse strain, a constitutive pulmonary vasculopathy with medial thickening, a perivascular proliferating chronic inflammatory cell infiltrate, and mildly elevated pulmonary artery pressure resembled the well-described chronic hypoxia model of pulmonary hypertension. Following administration of SU5416, the pulmonary vascular phenotype was more florid, with pulmonary arteriolar luminal obliteration by apoptosis-resistant proliferating endothelial cells. These changes resulted in right ventricular hypertrophy, confirming hemodynamically significant PAH. Altered expression of TGFβ and VEGF ligand and receptor was consistent with a scleroderma phenotype. CONCLUSION In this study, we replicated key features of systemic sclerosis-related PAH in a mouse model. Our results suggest that pulmonary endothelial cell injury in a genetically susceptible mouse strain triggers this complication and support the underlying role of functional interplay between TGFβ and VEGF, which provides insight into the pathogenesis of this disease.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Animals
- Disease Models, Animal
- Endothelium, Vascular/physiopathology
- Familial Primary Pulmonary Hypertension
- Female
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/physiopathology
- Hypoxia/genetics
- Hypoxia/physiopathology
- Indoles/pharmacology
- Lac Operon
- Male
- Mice
- Mice, Transgenic
- Phenotype
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Pulmonary Circulation/physiology
- Pyrroles/pharmacology
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors
- Scleroderma, Systemic/complications
- Scleroderma, Systemic/genetics
- Scleroderma, Systemic/physiopathology
- Signal Transduction/physiology
- Transforming Growth Factor beta/metabolism
Collapse
|
43
|
|
44
|
Finnson KW, Arany PR, Philip A. Transforming Growth Factor Beta Signaling in Cutaneous Wound Healing: Lessons Learned from Animal Studies. Adv Wound Care (New Rochelle) 2013; 2:225-237. [PMID: 24761336 DOI: 10.1089/wound.2012.0419] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Indexed: 12/11/2022] Open
Abstract
SIGNIFICANCE Wound healing is a complex physiological process involving a multitude of growth factors, among which transforming growth factor beta (TGF-β) has the broadest spectrum of effects. Animal studies have provided key information on the mechanisms of TGF-β action in wound healing and have guided the development of therapeutic strategies targeting the TGF-β pathway to improve wound healing and scarring outcome. RECENT ADVANCES Development of tissue-specific expression systems for overexpression or knockout of TGF-β signaling pathway components has led to novel insight into the role of TGF-β signaling in wound healing. This work has also identified molecules that might serve as molecular targets for the treatment of pathological skin conditions such as chronic wounds and excessive scarring (fibrosis). CRITICAL ISSUES Many of the mouse models with genetic alterations in the TGF-β signaling pathway develop an underlying skin abnormality, which may pose some limitations on the interpretation of wound-healing results obtained in these animals. Also, TGF-β's pleiotropic effects on many cell types throughout all phases of wound healing present a challenge in designing specific strategies for targeting the TGF-β signaling pathway to promote wound healing or reduce scarring. FUTURE DIRECTIONS Further characterization of TGF-β signaling pathway components using inducible tissue-specific overexpression or knockout technology will be needed to corroborate results obtained in mouse models that display a skin phenotype, and to better understand the role of TGF-β signaling during distinct phases of the wound-healing process. Such studies will also provide a better understanding of how TGF-β mediates its autocrine, paracrine, and double paracrine effects on cellular responses in vivo during wound healing.
Collapse
Affiliation(s)
- Kenneth W. Finnson
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal General Hospital, Montreal, Canada
| | - Praveen R. Arany
- Cell Regulation and Control Unit, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Anie Philip
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal General Hospital, Montreal, Canada
| |
Collapse
|
45
|
Experimental models of dermal fibrosis and systemic sclerosis. Joint Bone Spine 2013; 80:23-8. [DOI: 10.1016/j.jbspin.2012.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 06/15/2012] [Indexed: 11/23/2022]
|
46
|
Thoua NM, Derrett-Smith EC, Khan K, Dooley A, Shi-Wen X, Denton CP. Gut fibrosis with altered colonic contractility in a mouse model of scleroderma. Rheumatology (Oxford) 2012; 51:1989-98. [DOI: 10.1093/rheumatology/kes191] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
47
|
Pechkovsky DV, Prêle CM, Wong J, Hogaboam CM, McAnulty RJ, Laurent GJ, Zhang SSM, Selman M, Mutsaers SE, Knight DA. STAT3-Mediated Signaling Dysregulates Lung Fibroblast-Myofibroblast Activation and Differentiation in UIP/IPF. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1398-412. [DOI: 10.1016/j.ajpath.2011.12.022] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 11/28/2011] [Accepted: 12/06/2011] [Indexed: 12/20/2022]
|
48
|
Meng XM, Huang XR, Xiao J, Chen HY, Zhong X, Chung ACK, Lan HY. Diverse roles of TGF-β receptor II in renal fibrosis and inflammation in vivo and in vitro. J Pathol 2012; 227:175-88. [PMID: 22190171 DOI: 10.1002/path.3976] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 11/07/2011] [Accepted: 12/02/2011] [Indexed: 11/09/2022]
Abstract
TGF-β1 binds receptor II (TβRII) to exert its biological activities but its functional importance in kidney diseases remains largely unclear. In the present study, we hypothesized that TβRII may function to initiate the downstream TGF-β signalling and determine the diverse role of TGF-β1 in kidney injury. The hypothesis was examined in a model of unilateral ureteral obstructive (UUO) nephropathy and in kidney fibroblasts and tubular epithelial cells in which the TβRII was deleted conditionally. We found that disruption of TβRII inhibited severe tubulointerstitial fibrosis in the UUO kidney, which was associated with the impairment of TGF-β/Smad3 signalling, but not with the ERK/p38 MAP kinase pathway. In contrast, deletion of TβRII enhanced NF-κB signalling and renal inflammation including up-regulation of Il-1β and Tnfα in the UUO kidney. Similarly, in vitro disruption of TβRII from kidney fibroblasts or tubular epithelial cells inhibited TGF-β1-induced Smad signalling and fibrosis but impaired the anti-inflammatory effect of TGF-β1 on IL-1β-stimulated NF-κB activation and pro-inflammatory cytokine expression. In conclusion, TβRII plays an important but diverse role in regulating renal fibrosis and inflammation. Impaired TGF-β/Smad3, but not the non-canonical TGF-β signalling pathway, may be a key mechanism by which disruption of TβRII protects against renal fibrosis. In addition, deletion of TβRII also enhances NF-κB signalling along with up-regulation of renal pro-inflammatory cytokines, which may be associated with the impairment of anti-inflammatory properties of TGF-β1.
Collapse
Affiliation(s)
- Xiao-Ming Meng
- Li Ka Shing Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Animal models of cutaneous and hepatic fibrosis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 105:371-409. [PMID: 22137437 DOI: 10.1016/b978-0-12-394596-9.00011-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fibrosis occurs as a part of normal wound healing. However, excessive or dysregulated fibrosis can lead to severe organ dysfunction and is a feature of a variety of diseases. Due to its insidious onset, fibrosis tends to go undetected in its early stages. This is in part why these diseases remain so poorly understood. Animal models have provided a means to examine these early stages and to isolate and understand the effect of perturbations in signaling pathways, chemokines, and cytokines. Here, we summarize recent progress in the understanding of the molecular pathogenesis of fibrosis, both its initiation and its maintenance phases, from animal models of fibrosis in the skin and liver. Due to these organs' properties, modeling fibrosis in them poses unique challenges. Elegant solutions have therefore been developed for modeling fibrosis in each, and now, great potential for animal models to contribute to our understanding appears scientifically imminent.
Collapse
|
50
|
Sargent JL, Whitfield ML. Capturing the heterogeneity in systemic sclerosis with genome-wide expression profiling. Expert Rev Clin Immunol 2011; 7:463-73. [PMID: 21790289 DOI: 10.1586/eci.11.41] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heterogeneity in the clinical presentation and basic science findings of systemic sclerosis (SSc) has hindered the understanding of pathogenesis and development of effective treatments. Genome-wide profiling of SSc has measured this heterogeneity. Gene expression studies of diffuse SSc skin have shown reproducible, disease-specific gene expression signatures when compared with healthy controls and, surprisingly, disease-specific gene expression was found in both lesional and non-lesional skin. SSc-specific gene expression in peripheral blood cells and the lungs has also been demonstrated. Hypothesis-driven approaches that assess the contribution of individual pathways provide insight into the etiology of gene expression subsets.
Collapse
Affiliation(s)
- Jennifer L Sargent
- Department of Genetics, Dartmouth Medical School, Hanover, NH 03755, USA
| | | |
Collapse
|