Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Rationale

Fibrosis leads to failure of the skin, lungs, and other organs in systemic sclerosis; accounts for substantial morbidity and mortality; and lacks effective therapy. Myofibroblast activation underlies organ fibrosis, but the key extracellular cues driving persistence of the process remain incompletely characterized.

Objectives

The objectives were to evaluate activation of the IL6/JAK/STAT axis associated with fibrosis in skin and lung biopsies from systemic sclerosis patients and effects of the Food and Drug Administration-approved JAK/STAT inhibitor, tofacitinib, on skin and lung fibrosis in animal models.

Methods

Bioinformatic analysis showed that IL6/JAK/STAT3 and tofacitinib gene signatures were aberrant in biopsies from systemic sclerosis patients in four independent cohorts. The results were confirmed by JAK and STAT3 phosphorylation in both skin and lung biopsies from patients with systemic sclerosis. Furthermore, treatment of mice with the selective JAK inhibitor tofacitinib not only prevented bleomycin-induced skin and lung fibrosis but also reduced skin fibrosis in TSK1/+ mice.

Conclusion

These findings implicate the JAK/STAT pathway in systemic sclerosis skin and lung fibrosis and identify tofacitinib as a potential antifibrotic agent for the treatment of systemic sclerosis and other fibrotic diseases.

Free full text 


Logo of jsoLink to Publisher's site
J Scleroderma Relat Disord. 2020 Feb; 5(1): 40–50.
Published online 2019 Aug 7. https://doi.org/10.1177/2397198319865367
PMCID: PMC8922593
PMID: 35382402

The JAK/STAT pathway is activated in systemic sclerosis and is effectively targeted by tofacitinib

Associated Data

Supplementary Materials

Abstract

Rationale:

Fibrosis leads to failure of the skin, lungs, and other organs in systemic sclerosis; accounts for substantial morbidity and mortality; and lacks effective therapy. Myofibroblast activation underlies organ fibrosis, but the key extracellular cues driving persistence of the process remain incompletely characterized.

Objectives:

The objectives were to evaluate activation of the IL6/JAK/STAT axis associated with fibrosis in skin and lung biopsies from systemic sclerosis patients and effects of the Food and Drug Administration–approved JAK/STAT inhibitor, tofacitinib, on skin and lung fibrosis in animal models.

Methods:

Bioinformatic analysis showed that IL6/JAK/STAT3 and tofacitinib gene signatures were aberrant in biopsies from systemic sclerosis patients in four independent cohorts. The results were confirmed by JAK and STAT3 phosphorylation in both skin and lung biopsies from patients with systemic sclerosis. Furthermore, treatment of mice with the selective JAK inhibitor tofacitinib not only prevented bleomycin-induced skin and lung fibrosis but also reduced skin fibrosis in TSK1/+ mice.

Conclusion:

These findings implicate the JAK/STAT pathway in systemic sclerosis skin and lung fibrosis and identify tofacitinib as a potential antifibrotic agent for the treatment of systemic sclerosis and other fibrotic diseases.

Keywords: Tofacitinib, JAK, STAT3, fibrosis, systemic sclerosis, interleukin 6

Introduction

Fibrosis synchronously affecting skin and internal organs is the defining hallmark of systemic sclerosis (SSc).1,2 Multiple organ fibrosis in SSc has unknown pathogenesis and no effective treatments.3,4 Myofibroblasts, the key effector cells of organ fibrosis, secrete collagens and profibrotic mediators including transforming growth factor-β (TGF-β) and interleukin 6 (IL6). 3 Recent findings demonstrate prominent JAK/STAT activation in SSc fibroblasts, SSc biopsies, and in models of disease.57 Furthermore, genetic studies revealed strong association of STAT locus variants with SSc. 8 These observations suggest a critical role for pathogenic JAK/STAT signaling in SSc.

JAKs (Janus kinases) are non-receptor tyrosine kinases with central roles in cytokine and growth factor signaling.9,10 Upon binding of IL6 and related cytokines to their surface receptors, JAK kinases become activated and phosphorylate tyrosine residues in the cytoplasmic region of the receptor.9,10 STAT proteins that are also activated by JAKs dimerize and translocate into the nucleus, where they induce transcription of several target genes. The IL6/JAK/STAT signaling pathway is implicated in the pathogenesis of numerous inflammatory and autoimmune diseases, including rheumatoid arthritis, psoriasis, and inflammatory bowel disease. Furthermore, mutations in JAK and STAT genes cause a number of immunodeficiency syndromes, and polymorphisms in these genes are associated with autoimmune diseases. 9 IL6-targeted therapy in SSc appears to show modest and variable clinical benefit.11,12 Selective inhibition of intracellular receptor–associated kinases using orally available small molecules is a highly promising novel therapeutic approach for SSc and fibrosis. Tofacitinib is a synthetic kinase inhibitor that primarily targets the activity of JAK1 and JAK3 and, to a lesser extent, JAK2.13,14 Most importantly, tofacitinib is the first Jakinib approved for the treatment of autoimmune conditions, including rheumatoid arthritis and psoriasis. 15

The aim of this study was to explore the potential pathogenic role of JAK and STAT signaling in the context SSc pathology and the effect of the selective JAK inhibitor tofacitinib on experimental models of dermal and pulmonary fibrosis. Our results demonstrate evidence of increased IL6-JAK/STAT pathway activity in subsets of SSc skin and lung biopsies. Targeted pharmacological blockade of JAK/STAT signaling using tofacitinib prevented progression of experimental organ fibrosis in multiple distinct disease models. Taken together, our findings provide strong evidence supporting a pathogenic role of JAK/STAT signaling in SSc and suggest that drug repurposing using tofacitinib might be an attractive antifibrotic strategy for treating SSc.

Results

Initial studies sought to evaluate the IL6/JAK/STAT3 signaling axis in SSc skin biopsies. The Gene Set Enrichment Analysis (GSEA) method was used using software from Broad Institute (http://software.broadinstitute.org/gsea/index.jsp). The IL6/JAK/STAT3 signature consisting of 87 prior defined set of genes (http://software.broadinstitute.org/gsea/msigdb/cards/HALLMARK_IL6_JAK_STAT3_SIGNALING.html) was evaluated in a publicly available SSc transcriptome data set (GSE9285, GSE32413, GSE45485, and GSE59785). 16 Gene expression profiling showed elevated IL6/JAK/STAT3 signaling in skin biopsies from the previously defined inflammatory subset of diffuse cutaneous systemic sclerosis (dcSSc) compared to healthy controls (Figure 1(a)). The GSEA method further confirmed enrichment of the IL6/JAK/STAT3 signature in the inflammatory intrinsic subset (nominal p < 0.005, false discovery rate (FDR) q < 0.01, family-wise error rate (FWER) p < 0.05). We next examined SSc skin biopsies for the tofacitinib gene signature, generated as described in the “Materials and methods” section. Unbiased gene expression profiling showed elevated tofacitinib gene signature in skin biopsies from patients mapping to the inflammatory intrinsic dcSSc subset compared to healthy controls (Figure 1(b)). In addition, the GSEA method showed enrichment of tofacitinib signature in the inflammatory subset of dcSSc (nominal p < 0.01, FDR q < 0.01, FWER p < 0.005). Next, to examine the activation of JAK/STAT pathway in SSc, we measured levels of phosphor-JAK1, JAK2, JAK3, and STAT3 in skin biopsies from SSc patients and healthy controls. In the epidermis, we found comparable expression levels between control subjects and patients with SSc. In contrast, in the dermis, expression of pY-JAK1, pY-JAK2, pY-JAK3, and STAT3 was significantly elevated in SSc biopsies of interstitial cells compared with control biopsies (Figure 2). Patients with late-stage disease showed increased expression of p-JAK2 and p-STAT3 compared with those with early-stage disease (p-JAK2, p = 0.022; p-STAT3, p = 0.05). There was no association of JAK/STAT pathway activation with the skin score (p > 0.05).

An external file that holds a picture, illustration, etc.
Object name is 10.1177_2397198319865367-fig1.jpg

JAK/STAT signature is elevated in SSc skin biopsies. Expression microarray data sets (GSE9285, GSE32413, GSE45485, and GSE59785) were queried for IL6/JAK/STAT3 and tofacitinib signature as described in the “Materials and methods” section. (a) Left panel, IL6/JAK/STAT3 signature; right panel, Gene Set Enrichment Analysis (GSEA). (b) Left panel, tofacitinib signature; right panel, Gene Set Enrichment Analysis (GSEA).

An external file that holds a picture, illustration, etc.
Object name is 10.1177_2397198319865367-fig2.jpg

Phospho-JAK and phosphor-STAT levels elevated in SSc skin. Skin and lung tissues from patients with SSc were analyzed by immunohistochemistry (IHC) microscopy using antibodies to pY-JAK1, pY-JAK2, pY-JAK3, and pY-STAT3. Skin biopsies from patients with SSc and healthy controls were immunostained with indicated antibodies. Left panels, Representative images; dotted lines indicate the dermal–epidermal junction. Scale bar, 10 μm. Right panels, Dot plots of positive cell population. The proportion of immunopositive cells was determined from 5 hpf/section in each biopsy. The p values shown are results of the Mann–Whitney U test.

Pulmonary fibrosis is a leading complication of SSc. 17 To explore JAK/STAT activation in SSc-ILD, we measured pY-JAK1, pY-JAK2, pY-JAK3 and STAT3 in lung biopsies from SSc-ILD (n = 7) and control lung biopsies (n = 4). Non-SSc donor lungs showed only a low level of pY-JAK1, pY-JAK2, pY-JAK3, and STAT3 expressions by immunofluorescence (Figure 3). In these biopsies, pY-JAK1, pY-JAK2, pY-JAK3, and pY-STAT3 expressions were largely restricted to alveolar epithelial lining cells and scattered intra-alveolar macrophages. In marked contrast, strong intracellular pY-JAK1, pY-JAK2, pY-JAK3, and pY-STAT3 expression was noted in each SSc-ILD biopsy. JAK/STAT activation was most prominent at fibrotic loci (Figure 3).

An external file that holds a picture, illustration, etc.
Object name is 10.1177_2397198319865367-fig3.jpg

Elevated phospho-JAK/STAT3 signaling in SSc lungs. Lung biopsies from patients with SSc-ILD (n = 7) and controls (n = 4) were immunostained using antibodies to pY-JAK1, pY-JAK2, pY-JAK3, and pY-STAT3. Nuclei were identified by DAPI (blue color). Left panels, Representative confocal microscopic images; scale bar, 25 μm. Right panels, Dot plots of positive cell population. The proportion of immunopositive cells was determined from 5 hpf/section in each biopsy. The p values shown are results of the Mann–Whitney U test.

We next sought to investigate whether activated JAK/STAT signaling contributes to pathology in SSc. For this purpose, we evaluated the antifibrotic effects of tofacitinib on preclinical disease models. Eight-week-old C57BL/6J mice were given daily SC (subcutaneous) injections of bleomycin for 2 weeks (5 days/week), along with concurrent tofacitinib (30 mg/kg, bid), a dose selected based on our preliminary experiment or vehicle by daily IP (intraperitoneal) injections (5 days/week). Mice were sacrificed at day 22, and lung and lesional skin was harvested. Tofacitinib was well tolerated with no significant weight loss or other signs of toxicity noted. The thickness of the dermis and collagen deposition was markedly increased in bleomycin-treated mice (Figure 4). Both collagen and dermal thickness were markedly reduced when tofacitinib was administered concomitantly with bleomycin. Loss of the SC adipose layer accompanying dermal fibrosis was substantially attenuated in mice treated with tofacitinib (Figure 4(a)). Furthermore, increased expression of the fibrotic markers Col1a1 and Col1a2 and the number of alpha-smooth muscle actin (ASMA)-positive interstitial myofibroblasts, as well as infiltration with macrophages, were all significantly reduced in the skin in tofacitinib-treated mice (Figure 4(b) and ((c)c) and Supplemental Figure 2A). The percent of bleomycin-induced increase in F4/80-positive macrophages was significantly reduced in tofacitinib-treated mice (Supplemental Figure 2). Dermal fibrosis was associated with a sharp increase in STAT3 phosphorylation in interstitial cells, which was markedly attenuated when mice were treated with tofacitinib (Figure 4(d)). Next, to determine the effect of JAK/STAT inhibition on fibrosis regression, tofacitinib treatment was started at day 8 when dermal fibrosis is already initiated. The results indicated no significant reduction in skin fibrosis under this condition (Supplemental Figure 3A).

An external file that holds a picture, illustration, etc.
Object name is 10.1177_2397198319865367-fig4.jpg

Tofacitinib treatment prevents skin fibrosis. C57/BL6 mice received daily SC injections of PBS or bleomycin alone, or together with tofacitinib (30 mg/kg, IP) or vehicle. Mice were sacrificed at day 22, and skin was harvested for analysis. (a) Left panel, H&E stain. Representative images. Bar = 50 μm. Right panel, Dermal thickness (means ± SD of eight determinations/hpf from five mice/group). One-way analysis of variance followed by Sidak’s multiple comparison test. (b) Real-time quantitative PCR. Results, normalized with GAPDH, are means ± SD of triplicate determinations from three or four mice/group; one-way analysis of variance followed by Sidak’s multiple comparison test. Immunohistochemistry using antibodies to ASMA, F4/80, and p-STAT3. (c) Immunohistochemistry using antibodies to ASMA. Representative images. Bar = 25 μm. (d) pY-STAT3 IHC. Representative images. Bar = 50 μm.

In order to evaluate the effect of selective JAK inhibition on an inflammation-independent skin fibrosis model, we treated TSK1/+ mice with tofacitinib or vehicle in parallel starting at 5 weeks of age, when skin fibrosis is already apparent. 18 At 10 weeks, vehicle-treated TSK1/+ mice displayed a substantial increase in hypodermal thickness (Figure 5(a)). In contrast, TSK1/+ mice that received tofacitinib showed significantly attenuated hypodermal thickening and STAT3 phosphorylation (Figure 5(b)).

An external file that holds a picture, illustration, etc.
Object name is 10.1177_2397198319865367-fig5.jpg

Tofacitinib treatment prevents fibrosis in Tsk1/+ mice. Five-week-old Tsk1/+ mice received tofacitinib (IP; 30 mg/kg) daily for 5 weeks. Mice were sacrificed at 10 weeks of age, and dorsal skin was harvested for analysis. (a) Left panel, H&E stain (dotted line indicates hypodermis). Right panel, Hypodermal thickness (means ± SD of eight determinations/hpf from eight or nine mice/group). Representative images. Bar = 200 μm. Student’s t test. (b) Immunohistochemistry using antibodies to pY-STAT3. Representative images. Bar = 50 μm.

In view of the pronounced activation of the JAK/STAT pathway seen in SSc-ILD (Figure 3), subsequent experiments sought to explore the effect of tofacitinib on lung fibrosis. Chronic SC bleomycin elicited prominent architectural changes in the lungs, with an influx of inflammatory cells accompanied by appearance of fibrotic foci primarily in the subpleural area, along with sparse perivascular and interstitial fibrosis (Figure 6). These changes were associated with substantial collagen accumulation, increase in the fibrosis score, and elevated ASMA, F4/80, and CD3 expressions (Figure 6). Each of these parameters showed substantial attenuation in mice with concurrent tofacitinib treatment (Figure 6). Moreover, tofacitinib effectively reduced the sharp increase in pulmonary STAT3 phosphorylation (Figure 6(c)). We confirmed the antifibrotic effect of tofacitinib on IL6-treated explanted skin fibroblasts. Incubation with IL6 induced stimulation of type I collagen and ASMA levels, and STAT3 phosphorylation (Supplemental Figure 1). Pretreatment of the cultures with tofacitinib completely abrogated STAT3 phosphorylation at both early and late time-points and reduced the levels of type I collagen and ASMA at late time-point. However, no significant effect on regression of lung fibrosis was observed when tofacitinib treatment started at day 8 and continued until harvest at day 29 (Supplemental Figure 3B).

An external file that holds a picture, illustration, etc.
Object name is 10.1177_2397198319865367-fig6.jpg

Tofacitinib treatment prevents lung fibrosis. C57/BL6 mice received daily SC injections of PBS or bleomycin alone, or together with tofacitinib (30 mg/kg) or vehicle. Mice were sacrificed at day 22, and lungs were harvested. (a) Left panel, Masson’s Trichome stain. Representative images. Bar = 25 μm. Right panel, Ashcroft score. Results are means ± SD from 5 hpf per mice from at least three mice per experimental group. One-way analysis of variance followed by Sidak’s multiple comparison test. (b) Real-time quantitative PCR. Results, normalized with GAPDH, are means ± SD of triplicate determinations from three or four mice/group; one-way analysis of variance followed by Sidak’s multiple comparison test. (c) Upper panels, immunofluorescence using antibodies to ASMA, ProcolI, CD3, and F4/80 and immunohistochemistry using pY-STAT3. Representative images. Bar = 50 or 25 μm as indicated. For ASMA, percent of relative fluorescence intensities (means from four randomly selected fields/sample). For CD3, F4/80, and pY-STAT3, the percent of immunopositive cells was determined from 5 hpf/section. One-way analysis of variance followed by Sidak’s multiple comparison test.

Discussion

The pathogenesis of SSc remains incompletely understood, and the disease is highly variable in its clinical presentation and course.1,19 Genome-wide molecular profiling has identified four “intrinsic” molecular SSc subsets called the fibroproliferative, inflammatory, limited, and normal-like subsets. 20 Each of these molecular subsets shows deregulation of distinct signaling pathways; however, the full set of pathways contributing to this differential gene expression has not been fully elucidated. 16 Recent studies have called attention to the presence of prominent JAK/STAT activation in SSc biopsies and in models of disease.57 Our analysis of SSc transcriptomes showed that skin biopsies mapping to the inflammatory intrinsic gene subset, accounting for approximately 40% of all biopsies, have significantly elevated levels of a consensus IL6/JAK/STAT3 signature which was derived on the basis based on experimentally derived gene expression. Moreover, immunohistochemistry similarly revealed the presence of activated JAKs and of activated STAT3 in SSc skin and lung biopsies. In addition, we found evidence of a repressed tofacitinib signature in these skin biopsies. Based on these combined transcriptome and protein data, we conclude that the disease process in particular subsets of SSc patients is directly driven by activation of the JAK/STAT3 pathway, and that these patients might show a favorable response to therapeutic interventions aimed at inhibiting the pathway, with normalization of aberrant gene signatures and resolution of tissue fibrosis.

A number of other JAK inhibitors are currently under development or in phase II and III clinical trials for the treatment of a variety of autoimmune inflammatory diseases. 21 Tofacitinib, which shows some selectivity for JAK1 and JAK3, is the first JAK inhibitor approved by the Food and Drug Administration (FDA) for the treatment of rheumatoid arthritis and psoriasis.15,22,23 The efficacy of tofacitinib for the treatment of fibrotic processes remains unknown. In contrast, previous studies have provided evidence that inhibiting JAKs and STAT3 might have antifibrotic effect. In particular, previous studies have shown that JAK/STAT inhibitor mitigates fibrosis in experimental models of skin and lung fibrosis.57 Therefore, drug repurposing using FDA-approved tofacitinib to selectively target JAK/STAT signaling is an attractive therapeutic antifibrotic strategy for SSc patients demonstrating evidence of activated JAK/STAT signaling in the skin. In the present studies, treatment with tofacitinib exerted potent antifibrotic effects on both bleomycin-induced skin fibrosis that recapitulate the inflammatory stage of SSc in fibrotic skin 24 and on Tsk1/+ mice that phenocopy non-inflammatory SSc skin fibrosis. 24 The beneficial effects of tofacitinib on bleomycin-induced fibrosis were restricted to preventive, and not therapeutic, application. This suggests that JAK/STAT3 signaling is being consistent with the recent demonstration that in established fibrosis, matrix stiffness itself is sufficient to drive STAT3 nuclear localization and activation independent of exogenous soluble ligands prompted reconsideration of the potential role of STAT3 in persistent myofiboblast mechanoactivation, 25 and STAT3 activity is essential for transcription of TGF-β-induced COL1A2 in fibroblast. 26

Together, our results indicate that tofacitinib, by selectively targeting JAK/STAT3 signaling, prevented organ fibrosis in preclinical disease models and abrogated fibrotic responses in normal fibroblasts in vitro. These studies can facilitate the development of predictive biomarkers for identifying SSc patients likely to show responses to JAK/STAT3 inhibition treatment and as pharmacodynamics markers to evaluate molecular responses in sequential skin biopsies from patients undergoing inhibitor treatment.

Materials and methods

Cell culture and reagent

Skin and lung biopsies were performed after obtaining written informed consent and in accordance with protocols approved by the Institutional Review Board for Human Studies at Northwestern University, University of Pittsburgh School of Medicine. Information of patients is shown in Table 1. Primary cultures of fibroblasts were established by explantation from neonatal foreskin. 18 Low-passage fibroblasts were grown in monolayers in plastic dishes and studied at early confluence. Cultures were maintained in Dulbecco’s Modified Eagle’s medium supplemented with 10% fetal bovine serum (Gibco BRL, Grand Island, NY), 1% vitamin solutions, and 2-mM l-glutamine. All other tissue culture reagents were from Lonza (Basel, Switzerland). For experiments, cultures were placed in serum-free media containing 0.1% bovine serum albumin overnight, followed by tofacitinib (Selleck Chemical, 1 µM) for indicated periods. Tofacitinib was added 60 min prior to IL6 (R&D, 100 ng/mL).

Table 1.

Clinical characteristics of subjects.

Study codeAgeSexRaceDisease subtypeEarly/late
Subject information for p-JAK1 and p-JAK3 IHC
 SPARC_SSc_0231FWdcSScLate
 SPARC_SSc_0670MWdcSScEarly
 SPARC_SSc_0750FWdcSScEarly
 SPARC_SSc_0930FWlcSScLate
 SPARC_SSc_1041FWdcSScLate
 SPARC_SSc_1345FWdcSScLate
 SPARC_SSc_1936FWdcSScEarly
 SPARC_SSc_2046FWVEDOSSEarly
 SPARC_SSc_2151MWlcSScLate
 SPARC_SSc_2251FWlcSScLate
 SPARC_SSc_2469MWdcSScLate
 SPARC_SSc_2951FWdcSScLate
 SPARC_SSc_3048FWlcSScLate
 SPARC_SSc_3242FWlcSScLate
 SPARC_SSc_3349MWdcSScEarly
 SPARC_SSc_3464MWdcSScEarly
 SPARC_SSc_3562MWdcSScEarly
 SPARC_SSc_3649FWVEDOSSEarly
 SPARC_SSc_3851MWdcSScLate
Subject information for p-JAK2 and p-STAT3 IHC
 SPARC_SSc_0160FWdcSScLate
 SPARC_SSc_0231FWdcSScLate
 SPARC_SSc_0351FWdcSScLate
 SPARC_SSc_0560MWlcSScLate
 SPARC_SSc_0750FWdcSScEarly
 SPARC_SSc_0859FBdcSScLate
 SPARC_SSc_1040FWdcSScLate
 SPARC_SSc_1164FWdcSScEarly
 SPARC_SSc_1245FWdcSScLate
 SPARC_SSc_1344FWdcSScLate
 SPARC_SSc_1439FAdcSScEarly
 SPARC_SSc_1530MWlcSScEarly
 SPARC_SSc_1645FBdcSScLate
 SPARC_SSc_1763FWdcSScEarly
 SPARC_SSc_1936FWdcSScEarly
 SPARC_SSc_2337MWdcSScEarly
 SPARC_SSc_2469MWdcSScEarly
 SPARC_SSc_2645FWdcSScLate
 SPARC_SSc_2950FWdcSScEarly
 SPARC_SSc_3160FWdcSScEarly

JAK: Janus kinases; IHC: immunohistochemistry; SSc: systemic sclerosis; VEDOSS: very early diagnosis of systemic sclerosis; dcSSc: diffuse cutaneous systemic sclerosis; lcSSc: limited cutaneous systemic sclerosis.

Subjects providing skin biopsy samples for IHC. Early, <3 years from the first non-Raynaud disease manifestation; late, >3 years from the first non-Raynaud manifestation. Controls were healthy adults (70% female; median age: 32 years; range: 26–57 years).

Analysis of gene expression and measurement of pathway activation

IL6/JAK/STAT3 and tofacitinib signature in skin biopsies from SSc patients and healthy controls was analyzed in combined gene expression data sets (NCBI GEO database under accession numbers GSE9285, GSE32413, GSE45485, and GSE59785). 16 Forearm biopsies from healthy controls and dcSSc patients mapping to the previously defined inflammatory subsets of SSc skin biopsies were analyzed. Longitudinal data were excluded in this analysis.

HALLMARK_IL6_JAK_STAT3_SIGNALING from publicly available Molecular Signature Database (MSigDB, software.broadinstitute.org/gsea/msigdb/index.jsp) was used to generate JIL6/JAK/STAT3 signature in our current study.

To generate tofacitinib signature, gene expression data set (GSE57896) was used. 27 This data set generated tofacitinib signature from white adipocytes treated with tofacitinib (2 μM) for 24 h. Reads that map to annotated genes were modeled by negative binomial distribution, and differential expression was quantified by generalized linear models implemented in the edgeR package. The filter for significantly differentially expressed genes was set as |logFC| [gt-or-equal, slanted] 1 (logFC = log fold change) and FDR <0.05. Gene set analysis was performed with the GSEA method for both tofacitinib and IL6/JAK/STAT3 signature. 28

Experimental models of fibrosis

Animal experiments were performed according to institutionally approved protocols and in compliance with guidelines of the Northwestern University Animal Care and Use Committee. Complementary fibrosis models were used to evaluate the effect of pharmacological JAK/STAT signaling blockade using tofacitinib in vivo. First, 8-week-old female C57BL/6J (The Jackson Laboratory, Bar Harbor, ME) mice received SC injections of bleomycin (10 mg/kg/day) or phosphate-buffered saline (PBS) daily for 10 days (5 days/week), along with tofacitinib (LC Laboratories, Woburn, MA; 30 mg/kg) by twice-daily IP injections starting concurrently with bleomycin and were sacrificed on day 22. In a second group of mice, tofacitinib injections were started at day 8 and continued until sacrifice at day 29. A third group of mice received PBS, and a fourth received bleomycin alone. In a complementary non-inflammatory fibrosis model, 5-week-old male Tsk1/+ mice (C57BL/6 background, The Jackson Laboratory) received tofacitinib injections (30 mg/kg, bid) IP daily for 5 weeks (5 days/week) until sacrifice after 10 weeks.

Lung fibrosis was quantitated using the Ashcroft score determined from 5 hpf per mice.29,30 For immunofluorescence analyses, paraffin-embedded lung sections were incubated with primary antibodies against ASMA and CD3 (both from Abcam, 1:100, Cambridge, MA), procollagen I (EMD, 1:100, Burlington, MA), or F4/80 (eBioscience, 1:100, San Diego, CA) followed by Alexa Fluor–labeled rabbit secondary antibodies. Nuclei were detected using DAPI (4′,6-diamidino-2-phenylindole) staining. Sections were imaged under a Nikon A1R laser (Melville, NY) scanning confocal microscope. For immunohistochemistry, sections of paraffin-embedded skin and lungs were immunolabeled with primary rabbit antibodies against phosphor-STAT3 (Cell signaling, 1:500, Danvers, MA), followed by appropriate biotinylated secondary antibodies (Jackson Immunoresearch, 1:250, West Grove, PA), and detected using biotin complex conjugated with horseradish peroxidase (Vector Laboratories, Burlingame, CA) and DAB (3,3′-diaminobenzidine) for color development (Dako, Carpinteria, CA, looks like belongs to Fisher right now). Images were captured by Nuance Multiple Spectra CCD with Nuance 2.10 software.

Isolation and analysis of RNA

At the end of the experiments, mice were sacrificed, and total RNA was isolated from skin and lung and reverse transcribed to cDNA using Supermix (cDNA Synthesis Supermix; Quanta Biosciences, Beverly, MA) as described. 18 Amplification products (20 ng) were amplified using SYBR Green PCR Master Mix (Applied Biosytems, Foster City, CA) on an Applied Biosystems 7500 Prism Sequence Detection System. Data were normalized to GAPDH (glyceraldehyde-3-phosphate dehydrogenase) RNA, and fold change in samples was calculated.

Evaluation of phosphorylated JAK/STAT levels in SSc skin and lung biopsies

For immunohistochemistry, sections of paraffin-embedded skin were immunolabelled with primary rabbit antibodies against pY-STAT3, JAK1, JAK2, or JAK3 (all from Abcam, 1:100) and processed as described. Sections were imaged under a Nikon A1R laser scanning confocal microscope. For immunofluorescence analyses, paraffin-embedded lung sections from healthy or SSc lung sections were incubated with primary rabbit antibodies against pY-JAK1, JAK2, JAK3, or STAT3, followed by Alexa Fluor–labeled rabbit secondary antibodies as described. Slides were mounted, and immunofluorescence was evaluated under a Nikon A1R laser scanning confocal microscope.

Statistical analysis

Data are presented as means ± SD. Two-tailed Student’s t test or Mann–Whitney test was used for comparisons between two groups. Differences among the groups were examined for statistical significance using analysis of variance followed by Sidak’s correction. A p value less than 0.05 denoted the presence of statistically significant difference. Data were analyzed using GraphPad prism (GraphPad Software version 5, GraphPad Software Inc., CA).

Supplemental Material

Suppl_Fig._1 – Supplemental material for The JAK/STAT pathway is activated in systemic sclerosis and is effectively targeted by tofacitinib:

Supplemental material, Suppl_Fig._1 for The JAK/STAT pathway is activated in systemic sclerosis and is effectively targeted by tofacitinib by Wenxia Wang, Swati Bhattacharyya, Roberta Goncalves Marangoni, Mary Carns, Kathleen Dennis-Aren, Anjana Yeldandi, Jun Wei and John Varga in Journal of Scleroderma and Related Disorders

Suppl_Fig._2 – Supplemental material for The JAK/STAT pathway is activated in systemic sclerosis and is effectively targeted by tofacitinib:

Supplemental material, Suppl_Fig._2 for The JAK/STAT pathway is activated in systemic sclerosis and is effectively targeted by tofacitinib by Wenxia Wang, Swati Bhattacharyya, Roberta Goncalves Marangoni, Mary Carns, Kathleen Dennis-Aren, Anjana Yeldandi, Jun Wei and John Varga in Journal of Scleroderma and Related Disorders

Suppl_Fig._3 – Supplemental material for The JAK/STAT pathway is activated in systemic sclerosis and is effectively targeted by tofacitinib:

Supplemental material, Suppl_Fig._3 for The JAK/STAT pathway is activated in systemic sclerosis and is effectively targeted by tofacitinib by Wenxia Wang, Swati Bhattacharyya, Roberta Goncalves Marangoni, Mary Carns, Kathleen Dennis-Aren, Anjana Yeldandi, Jun Wei and John Varga in Journal of Scleroderma and Related Disorders

Acknowledgments

We are grateful to members of the Scleroderma Research Laboratory, Mouse Histology & Phenotyping Laboratory, and the Nikon Imaging Core of Northwestern University. We are extremely thankful to Dr Carol Feghali-Bostwick for her generosity to provide us the lung tissue sections.

Footnotes

Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by grants from the NIH National Institute of Arthritis and Musculoskeletal and Skin Diseases (AR42309), Pfizer, and the Scleroderma Foundation.

Impact of research: Bioinformatic analysis revealed aberrant JAK/STAT pathway in patients with systemic sclerosis (SSc). This finding was confirmed in skin and lung biopsies from patients with SSc and experimental lung fibrosis models. Furthermore, we proved that a specific JAK/STAT inhibitor, tofacitinib, attenuated bleomycin-induced lung fibrosis. These results suggest its clinical potential in fibrotic diseases.

ORCID iD: Swati Bhattacharyya An external file that holds a picture, illustration, etc.
Object name is 10.1177_2397198319865367-img1.jpg https://orcid.org/0000-0002-4005-5896

Supplemental material: Supplemental material for this article is available online.

References

1. Allanore Y, Simms R, Distler O, et al.. Systemic sclerosis. Nat Rev Dis Prim 2015; 1: 15002. [Abstract] [Google Scholar]
2. Cottin V, Brown KK. Interstitial lung disease associated with systemic sclerosis (SSc-ILD). Respir Res 2019; 20: 13. [Europe PMC free article] [Abstract] [Google Scholar]
3. Bhattacharyya S, Wei J, Varga J. Understanding fibrosis in systemic sclerosis: shifting paradigms, emerging opportunities. Nat Rev Rheumatol 2011; 8(1): 42–54. [Europe PMC free article] [Abstract] [Google Scholar]
4. Au K, Khanna D, Clements PJ, et al.. Current concepts in disease-modifying therapy for systemic sclerosis-associated interstitial lung disease: lessons from clinical trials. Curr Rheumatol Rep 2009; 11(2): 111–119. [Abstract] [Google Scholar]
5. Chakraborty D, Sumova B, Mallano T, et al.. Activation of STAT3 integrates common profibrotic pathways to promote fibroblast activation and tissue fibrosis. Nat Commun 2017; 8(1): 1130. [Europe PMC free article] [Abstract] [Google Scholar]
6. Pedroza M, Le TT, Lewis K, et al.. STAT-3 contributes to pulmonary fibrosis through epithelial injury and fibroblast-myofibroblast differentiation. FASEB J 2016; 30(1): 129–140. [Europe PMC free article] [Abstract] [Google Scholar]
7. Dees C, Tomcik M, Palumbo-Zerr K, et al.. JAK-2 as a novel mediator of the profibrotic effects of transforming growth factor beta in systemic sclerosis. Arthritis Rheum 2012; 64(9): 3006–3015. [Abstract] [Google Scholar]
8. Dieude P, Guedj M, Wipff J, et al.. STAT4 is a genetic risk factor for systemic sclerosis having additive effects with IRF5 on disease susceptibility and related pulmonary fibrosis. Arthritis Rheum 2009; 60(8): 2472–2479. [Abstract] [Google Scholar]
9. Schwartz DM, Kanno Y, Villarino A, et al.. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov 2017; 16: 843–862. [Abstract] [Google Scholar]
10. Johnson DE, O’Keefe RA, Grandis JR. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol 2018; 15(4): 234–248. [Europe PMC free article] [Abstract] [Google Scholar]
11. Khanna D, Denton CP, Lin CJF, et al.. Safety and efficacy of subcutaneous tocilizumab in systemic sclerosis: results from the open-label period of a phase II randomised controlled trial (faSScinate). Ann Rheum Dis 2018; 77(2): 212–220. [Europe PMC free article] [Abstract] [Google Scholar]
12. Denton CP, Ong VH, Xu S, et al.. Therapeutic interleukin-6 blockade reverses transforming growth factor-beta pathway activation in dermal fibroblasts: insights from the faSScinate clinical trial in systemic sclerosis. Ann Rheum Dis 2018; 77(9): 1362–1371. [Europe PMC free article] [Abstract] [Google Scholar]
13. Hodge JA, Kawabata TT, Krishnaswami S, et al.. The mechanism of action of tofacitinib–an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis. Clin Exp Rheumatol 2016; 34(2): 318–328. [Abstract] [Google Scholar]
14. Fridman JS, Scherle PA, Collins R, et al.. Selective inhibition of JAK1 and JAK2 is efficacious in rodent models of arthritis: preclinical characterization of INCB028050. J Immunol 2010; 184(9): 5298–5307. [Abstract] [Google Scholar]
15. Berekmeri A, Mahmood F, Wittmann M, et al.. Tofacitinib for the treatment of psoriasis and psoriatic arthritis. Exp Rev Clin Immunol 2018; 14: 719–730. [Abstract] [Google Scholar]
16. Johnson ME, Mahoney JM, Taroni J, et al.. Experimentally-derived fibroblast gene signatures identify molecular pathways associated with distinct subsets of systemic sclerosis patients in three independent cohorts. PLoS ONE 2015; 10(1): e0114017. [Europe PMC free article] [Abstract] [Google Scholar]
17. Herzog EL, Mathur A, Tager AM, et al.. Review: interstitial lung disease associated with systemic sclerosis and idiopathic pulmonary fibrosis: how similar and distinct? Arthritis Rheumatol 2014; 66(8): 1967–1978. [Europe PMC free article] [Abstract] [Google Scholar]
18. Bhattacharyya S, Wang W, Morales-Nebreda L, et al.. Tenascin-C drives persistence of organ fibrosis. Nat Commun 2016; 7: 11703. [Europe PMC free article] [Abstract] [Google Scholar]
19. Varga J, Hinchcliff M. Connective tissue diseases: systemic sclerosis: beyond limited and diffuse subsets? Nat Rev Rheumatol 2014; 10(4): 200–202. [Europe PMC free article] [Abstract] [Google Scholar]
20. Martyanov V, Whitfield ML. Molecular stratification and precision medicine in systemic sclerosis from genomic and proteomic data. Curr Opin Rheumatol 2016; 28(1): 83–88. [Europe PMC free article] [Abstract] [Google Scholar]
21. Fragoulis GE, McInnes IB, Siebert S. JAK-inhibitors. New players in the field of immune-mediated diseases, beyond rheumatoid arthritis. Rheumatology 2019; 58: i43–i54. [Europe PMC free article] [Abstract] [Google Scholar]
22. Winthrop KL. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol 2017; 13: 234–243. [Abstract] [Google Scholar]
23. Banerjee S, Biehl A, Gadina M, et al.. JAK-STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. Drugs 2017; 77: 521–546. [Europe PMC free article] [Abstract] [Google Scholar]
24. Sargent JL, Li Z, Aliprantis AO, et al.. Identification of optimal mouse models of systemic sclerosis by interspecies comparative genomics. Arthritis Rheumatol 2016; 68(8): 2003–2015. [Europe PMC free article] [Abstract] [Google Scholar]
25. Oh RS, Haak AJ, Smith KMJ, et al.. RNAi screening identifies a mechanosensitive ROCK-JAK2-STAT3 network central to myofibroblast activation. J Cell Sci 2018; 131(10): jcs.209932. [Europe PMC free article] [Abstract] [Google Scholar]
26. Papaioannou I, Xu S, Denton CP, et al.. STAT3 controls COL1A2 enhancer activation cooperatively with JunB, regulates type I collagen synthesis posttranscriptionally, and is essential for lung myofibroblast differentiation. Mol Biol Cell 2018; 29(2): 84–95. [Europe PMC free article] [Abstract] [Google Scholar]
27. Moisan A, Lee YK, Zhang JD, et al.. White-to-brown metabolic conversion of human adipocytes by JAK inhibition. Nat Cell Biol 2015; 17(1): 57–67. [Europe PMC free article] [Abstract] [Google Scholar]
28. Subramanian A, Tamayo P, Mootha VK, et al.. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 2005; 102(43): 15545–15550. [Europe PMC free article] [Abstract] [Google Scholar]
29. Ashcroft T, Simpson JM, Timbrell V. Simple method of estimating severity of pulmonary fibrosis on a numerical scale. J Clin Pathol 1988; 41(4): 467–470. [Europe PMC free article] [Abstract] [Google Scholar]
30. Hubner RH, Gitter W, El Mokhtari NE, et al.. Standardized quantification of pulmonary fibrosis in histological samples. Biotechniques 2008; 44(4): 507–511, 514–517. [Abstract] [Google Scholar]

Articles from Journal of Scleroderma and Related Disorders are provided here courtesy of World Scleroderma Foundation, EUSTAR, and SAGE Publications

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Article citations


Go to all (32) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Lay summaries 


Funding 


Funders who supported this work.

NIAMS NIH HHS (2)

National Institute of Arthritis and Musculoskeletal and Skin Diseases (1)

Scleroderma Foundation

    pfizer