SAT0294 IL33 ACTIVATES FIBROBLASTS AND INDUCES SKIN FIBROSIS IN SYSTEMIC SCLEROSIS

2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 1091.1-1092
Author(s):  
X. Wu ◽  
B. Ming ◽  
L. Dong

Background:Systemic sclerosis (SSc) is a chronic immune-mediated autoimmune disease that is characterized by fibrotic changes of the skin and internal organs, which in turn leads to distortion of tissue structure and gradual loss of organ function. So far, there is still no treatment allows full recovery from this severe disorder. Therefore, it is of great social significance to study the pathogenesis of this disease and find new targets for treatment. Interleukin 33 (IL-33), which is a potent inducer of type 2 immune response, has been confirmed to be involved in the development and progression of multiple fibrotic diseases. However, the role and mechanism of IL-33 in SSc-related fibrosis remains unclear.Objectives:To clarify the role of interleukin 33 (IL-33) and its receptor Suppression of tumorigenicity 2 (ST2) in the skin fibrosis of SSc, so to provides a new target for the treatment of fibrosis in patients with SSc.Methods:The levels of IL-33 and ST2 was analysed in human samples, murine models of SSc and in cultured fibroblasts by immunohistochemistry and immunofluorescence. The functional role of IL-33 was evaluated by detecting changes in proliferation, migration, and activation of fibroblasts stimulated with recombinant IL-33 protein. MAPK and NF-κB signallings of fibroblasts were assessed by western blotting and analyses of target genes. The role of IL-33 in skin fibrosis was analysed in IL-33 deficient mice (il33−/−) and wild-type controls injected with bleomycin or NaCl.Results:The expression of IL-33 and its receptor ST2 were up-regulated in skin lesions of SSc patients (Fig 1 A-C) and bleomycin-treated mice(Fig1 D-F). Compared to the healthy skin, the skin from SSc patients expressed more ST2 on fibroblasts membrane(Fig 1 B-C). IL33 induces MAPK and IκBα activation in human dermal fibroblast(Fig 2 A), and promote proliferation, migration and production of collagen of human dermal fibroblasts, but not the release of inflammatory factors(IL-6, MCP-1)(Fig2 B-G). Mice deficient for IL33 are protected from bleomycin-induced dermal fibrosis (Fig3).Fig 1.Increased expression of IL33, ST2 in SSc patients and bleomycin-treated mice.Fig 2.IL33 induces MAPK and IκBα activation in human dermal fibroblast, and and promote proliferation, migration and production of collagen of human dermal fibroblasts.Fig 3.Mice deficient for IL33 are protected from bleomycin-induced dermal fibrosis.Conclusion:IL33 promotes skin fibrosis by activating fibroblasts, and IL33/ST2 may be an important target for the treatment of fibrosis in patients with SSc.References:[1]Ingegnoli F, Ughi N, Mihai C. Update on the epidemiology, risk factors, and disease outcomes of systemic sclerosis. Best practice & research. Clinical rheumatology. 2018;32(2):223-240.[2]Schmitz J, Owyang A, Oldham E, et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity. 2005;23(5):479-490.[3]Molofsky AB, Savage AK, Locksley RM. Interleukin-33 in Tissue Homeostasis, Injury, and Inflammation.Immunity.2015;42(6):1005-1019.Disclosure of Interests:None declared

2003 ◽  
Vol 10 (3) ◽  
pp. 473-475 ◽  
Author(s):  
Philipp H. Reichel ◽  
Christine Seemann ◽  
Elena Csernok ◽  
Jens-M. Schröder ◽  
Antje Müller ◽  
...  

ABSTRACT The bactericidal/permeability-increasing protein (BPI) is an antibiotic- and endotoxin-neutralizing protein of granulocytes and epithelial cells. Constitutive expression of BPI, which increases upon interleukin 4 stimulation, by human dermal fibroblast was demonstrated, suggesting an important role of BPI in gram-negative bacterial clearance and a dampened response to endotoxin in the skin.


2019 ◽  
Vol 78 (11) ◽  
pp. 1583-1591 ◽  
Author(s):  
Minghua Wu ◽  
Brian Skaug ◽  
Xiongjie Bi ◽  
Tingting Mills ◽  
Gloria Salazar ◽  
...  

ObjectivesThere is considerable evidence that implicates dysregulation of type I interferon signalling (or type I IFN signature) in the pathogenesis of systemic sclerosis (SSc). Interferon regulatory factor 7 (IRF7) has been recognised as a master regulator of type I IFN signalling. The objective of this study was to elucidate the role of IRF7 in dermal fibrosis and SSc pathogenesis.MethodsSSc and healthy control skin biopsies were investigated to determine IRF7 expression and activation. The role of IRF7 in fibrosis was investigated using IRF7 knockout (KO) mice in the bleomycin-induced and TSK/+mouse models. In vitro experiments with dermal fibroblasts from patients with SSc and healthy controls were performed.ResultsIRF7 expression was significantly upregulated and activated in SSc skin tissue and explanted SSc dermal fibroblasts compared with unaffected, matched controls. Moreover, IRF7 expression was stimulated by IFN-α in dermal fibroblasts. Importantly, IRF7 co-immunoprecipitated with Smad3, a key mediator of transforming growth factor (TGF)-β signalling, and IRF7 knockdown reduced profibrotic factors in SSc fibroblasts. IRF7 KO mice demonstrated attenuated dermal fibrosis and inflammation compared with wild-type mice in response to bleomycin. Specifically, hydroxyproline content, dermal thickness as well as Col1a2, ACTA2 and interleukin-6 mRNA levels were significantly attenuated in IRF7 KO mice skin tissue. Furthermore, IRF7 KO in TSK/+mice attenuated hydroxyproline content, subcutaneous hypodermal thickness, Col1a2 mRNA as well as α-smooth muscle actin and fibronectin expression.ConclusionsIRF7 is upregulated in SSc skin, interacts with Smad3 and potentiates TGF-β-mediated fibrosis, and therefore may represent a promising therapeutic target in SSc.


2008 ◽  
Vol 20 (06) ◽  
pp. 337-343
Author(s):  
Yuan-Haun Lee ◽  
Bor-Yann Chen ◽  
Feng-Huei Lin ◽  
Kun-Yu Lin ◽  
King-Fu Lin

This first-attempt study tended to inspect the cytotoxic effects of montmorillonite (MMT) or 0.01 N phosphoric acid treated MMT supplemented with L-ascorbic acid (LAA) upon human dermal fibroblasts for possible applications. Light micrographs of human dermal fibroblast cell cultures revealed that more dense black spots in larger sizes were observed when higher levels of MMT were supplemented into the fibroblast culture, indicating that more dermal fibroblasts were covered by MMT particles. Compared with the supplementation of LAA alone, this study selected mitochondrial dehydrogenase activity (MTT) assay as an indicator bioreaction to show possible cytotoxic (or allergic) responses upon human dermal fibroblasts in vitro when LAA/acid-treated MMT composites were added. Statistical analysis showed that LAA augmented with either MMT or 0.01 N phosphoric-acid-treated MMT provoked insignificant cytotoxic responses to human dermal fibroblasts. Thus, an augmentation of MMT or 0.01 N phosphoric-acid-treated MMT to LAA should be biologically feasible for possible skin applications according to this human dermal fibroblasts model.


2009 ◽  
Vol 69 (01) ◽  
pp. 249-254 ◽  
Author(s):  
Y Hou ◽  
B J Rabquer ◽  
M L Gerber ◽  
F Del Galdo ◽  
S A Jimenez ◽  
...  

Objective:To investigate the role of junctional adhesion molecule-A (JAM-A) in the pathogenesis of systemic sclerosis (SSc).Methods:Biopsy specimens from proximal and distal arm skin and serum were obtained from patients with SSc and normal volunteers. To determine the expression of JAM-A on SSc dermal fibroblasts and in SSc skin, cell surface ELISAs and immunohistology were performed. An ELISA was designed to determine the amount of soluble JAM-A (sJAM-A) in serum. Myeloid U937 cell–SSc dermal fibroblast and skin adhesion assays were performed to determine the role of JAM-A in myeloid cell adhesion.Results:The stratum granulosum and dermal endothelial cells (ECs) from distal arm SSc skin exhibited significantly decreased expression of JAM-A in comparison with normal volunteers. However, sJAM-A was increased in the serum of patients with SSc compared with normal volunteers. Conversely, JAM-A was increased on the surface of SSc compared with normal dermal fibroblasts. JAM-A accounted for a significant portion of U937 binding to SSc dermal fibroblasts. In addition, JAM-A contributed to U937 adhesion to both distal and proximal SSc skin.Conclusions:JAM-A expression is dysregulated in SSc skin. Decreased expression of JAM-A on SSc ECs may result in a reduced response to proangiogenic basic fibroblast growth factor. Increased JAM-A expression on SSc fibroblasts may serve to retain myeloid cells, which in turn secrete angiogenic factors.


2021 ◽  
Vol 80 (Suppl 1) ◽  
pp. 441.2-441
Author(s):  
C. Wasson ◽  
R. Ross ◽  
J. Bryon ◽  
F. Del Galdo

Background:Skin fibrosis is the hallmark fibrotic manifestation of systemic sclerosis (SSc). Despite a key role of tissue fibroblasts, skin changes extend to the keratinocyte layer, which contribute to the loss of skin function. RNA seq. analysis of SSc patient forearm skin showed that palmoplantar specific Keratin 9 (CK9) was highly expressed (1). SSc affected skin shares several features with palmoplantar skin including increased keratinocyte layer thickness and lack of hair. Seminal work of last decade has shown that long noncoding RNA in the HOX loci play a crucial role in skin keratinocyte differentiation (2), with the lncRNA HOTAIR being one of the HOX lncRNA mostly expressed in the palmoplantar region.Objectives:Following recent data suggesting a role of HOTAIR in the profibrotic phenotype of dermal fibroblasts in SSc (3), here we set out to determine if HOTAIR expressed in SSc dermal fibroblasts was a contributing factor to the high levels of CK9 found in SSc patient skinMethods:Full-thickness skin biopsies were surgically obtained from the forearms of patients with SSc of recent onset. Fibroblasts were isolated and cultured in monolayers. HOX transcript antisense RNA (HOTAIR) was expressed in healthy dermal fibroblasts by lentiviral induction employing a vector containing the specific sequence. Exosomes were isolated from dermal fibroblast media using the Total exosome isolation reagent (Thermo Fisher). Enhancer of zeste 2 (EZH2) was blocked with GSK126 inhibitor. Skin equivalents were created using scramble and HOTAIR expressing fibroblasts with primary keratinocytesResults:Media from both SSc patient fibroblasts and HOTAIR expressing fibroblasts induced CK9 expression in healthy keratinocytes in vitro. In addition, HOTAIR expressing fibroblasts induces CK9 expression in keratinocytes in 3D skin equivalent models. Media fractionation studies indicated that HOTAIR was present in fibroblasts exosomes and found at a higher concentration (2.7 fold p=0.01) in exosomes from SSc fibroblasts. Importantly, transfection of Exosomal RNAs from SSc fibroblasts could reproduce the increase in CK9 in keratinocytes. Mechanistically, CK9 induction was mediated by changes to the histone methylation profile in the keratinocytes through EZH2.Conclusion:Pro-fibrotic dermal fibroblasts in systemic sclerosis contribute to the overall skin loss of function by inducing CK9 in adjacent keratinocytes through transfer of the long non-coding RNA HOTAIR. Unraveling the crosstalk of activated fibroblasts with adjacent cells may lead to identify therapeutic targets to re-establish tissue homeostasis and function during fibrosis.References:[1]Assassi S et.al Arthritis and Rheumatology 2015[2]Rinn JL et.al Cell 2007[3]Wasson CW et.al Annals of Rheumatic Disease 2020Disclosure of Interests:Chris Wasson: None declared, rebecca ross: None declared, Jessica Bryon: None declared, Francesco Del Galdo Speakers bureau: Speakers bureau: Astra-Zeneca, Boehringer Ingelheim, Actelion, Consultant of: Astra-Zeneca, Mitsubishi-Tanabe, Capella Biosciences, Chemomab, Actelion, Boehringer-Ingelheim, Grant/research support from: Grant/research support from: Capella Biosciences, Chemomab, Kymab, Mitsubishi-Tanabe


2021 ◽  
Vol 80 (Suppl 1) ◽  
pp. 391.1-391
Author(s):  
X. Xu ◽  
Y. N. Li ◽  
C. W. Chen ◽  
T. Trinh-Minh ◽  
G. Schett ◽  
...  

Background:The aldehyde dehydrogenase (ALDH) superfamily composes a group of 20 enzymes that catalyze aldehyde oxidation. Within this enzyme family, ALDH3A2 stands out for its central role in the oxidation of long-chain aldehydes. Of particular interest, the substrates of ALDH3A2 include also profibrotic lipid mediators such as sphingosine 1-phosphate or leukotrienes, which have been reported to be deregulated in the context of SSc.Objectives:We aimed to investigate the role of ALDH3A2 in fibrotic tissue remodeling in SSc.Methods:Fibroblast-to-myofibroblast transition was analyzed by quantification of ACTA2/αSMA, by assessment of stress fiber formation and mRNA and protein levels of type I collagens. ALDH3A2/Aldh3a2 siRNAs were employed to specifically knockdown ALDH3A2 in dermal fibroblasts both in vitro and in vivo. Overexpression of ALDH3A2 was achieved by ALDH3A2-pcDNA transfection. The role of ALDH3A2 was investigated in three different mouse models: Bleomycin- and cGvHD-induced dermal fibrosis as well as fibrosis induced by overexpression of a constitutively active TGFβ receptor I (TBRICA). Target genes of ALDH3A2 in fibroblasts were identified by RNA sequencing.Results:The expression of ALDH3A2 was modestly reduced in dermal fibroblasts of SSc skin as compared to matched healthy controls. This reduction in ALDH3A2 expression was phenocopied by activation of TGFβ signaling, whereas selective inhibition of TGFβ signaling prevented the downregulation of ALDH3A2 in experimental fibrosis. ALDH3A2 overexpression promoted fibroblast-to-myofibroblast transition with increased levels of αSMA, enhanced formation of stress fibers and reduced collagen release. In contrast, knockdown of ALDH3A2 in dermal fibroblasts inhibited fibroblast activation and collagen release. Moreover, in vivo knockdown of ALDH3A2 in the skin of mice ameliorated dermal thickening, myofibroblast differentiation and collagen deposition in three different murine models of skin fibrosis: Bleomycin-induced skin fibrosis and sclerodermatous GvHD-as models of inflammatory stages of SSc and TBRICA-induced fibrosis as an inflammation-independent model of SSc. RNA sequencing of ALDH3A2-knockdown fibroblasts demonstrated that ALDH3A2 regulates the activity of a network of profibrotic developmental pathways including TGFβ, Wnt, Notch, and Hedgehog signaling.Conclusion:We demonstrate that ALDH3A2 regulates a network of profibrotic pathways to control fibroblast activation and tissue fibrosis. ALDH3A2 is modestly downregulated in SSc fibroblasts as result of an endogenous, TGFβ-driven feedback loop. Although this modest downregulation is not sufficient to counterbalance the aberrant fibroblast activation in SSc, augmentation of this endogenous regulation by knockdown of ALDH3A2 demonstrates potent antifibrotic potential in experimental dermal fibrosis, thereby providing first evidence for ALDH3A2 as a target for antifibrotic therapies.Disclosure of Interests:Xiaohan Xu: None declared, Yi-Nan Li: None declared, Chih-Wei Chen: None declared, Thuong Trinh-Minh: None declared, Georg Schett: None declared, Jörg H.W. Distler Consultant of: Actelion, Active Biotech, Anamar, ARXX, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, JB Therapeutics, Medac, Pfizer, RuiYi and UCB, Grant/research support from: Anamar, Active Biotech, Array Biopharma, ARXX, aTyr, BMS, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, Novartis, Sanofi-Aventis, RedX, UCB


2007 ◽  
Vol 342-343 ◽  
pp. 377-380 ◽  
Author(s):  
Sang Gil Lee ◽  
Jung Bok Lee ◽  
Jong Chul Park ◽  
Young Il Yang ◽  
Jeong Koo Kim

The effect of β-glucan-reinforced PLGA scaffold on cell proliferation was investigated. The PLGA scaffolds were prepared by salt-leaching method. The prepared scaffolds were grafted with (1→3) (1→6)-β-glucan in various ratios after plasma treatment on the surface. The surface of the scaffold was characterized by scanning electron microscope (SEM). The HDFs (Human dermal fibroblasts, 1105 cells/scaffold) were used to evaluate the cell proliferation on PLGA scaffold before and after plasma/β-glucan treatment. In results, in the β-glucan treated scaffolds, the pores seemed to become narrower and even looked like closed form. The result of cell proliferation showed that the plasma/β-glucan treated scaffolds had narrower pores because the β-glucan was attached in the pores that would not be allowed the cells to penetrate into the inner areas. Consequently, cell proliferation was not effective in the plasma/β-glucan treated scaffolds in this study.


Author(s):  
Sophia Letsiou ◽  
Manuel Manchado ◽  
Mariela Zografaki ◽  
Sofia Marka ◽  
Liliana Anjos ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document