Introduction: Many cytokines, acting via Janus kinase (JAK)/tyrosine kinase 2 (TYK2)/signal transducer and activator of transcription (STAT) signalling pathways, are critical mediators of pathology in immune-mediated inflammatory diseases (IMIDs). Therefore, drugs that inhibit JAKs and TYKs (JAKis) have gained traction as effective treatment options for IMIDs. However, a common side effect of JAKis is increased risk of viral infection. Type I and III interferon (IFN) signal via JAK1 and TYK2. Type III IFNs protect mucosal membranes, including the intestinal barrier, from viral infection and type I IFNs prevent systemic viral spread. Methods: The human cell lines Caco-2 and HT-29 were used to investigate the impact of currently approved JAKis, baricitinib (JAK1/2 inhibitor) and deucravacitinib (TYK2 inhibitor), on type I and III IFN signalling and antiviral defence of intestinal epithelial cells (IECs). Results: Using concentrations commonly used for in vitro studies on IFNs, we found that attenuation of IFN-mediated antiviral defence in IECs is a mechanism that could contribute to the increased risk of viral infections associated with JAKis. However, when we examined clinically relevant drug concentrations, we found that while both drugs blocked the type I IFN response, type III IFN-induced antiviral immunity was less affected during TYK2 inhibition (deucravacitinib) compared to JAK1/2 inhibition (baricitinib). Conclusions: Our studies suggest that in treating diseases associated with excessive cytokine production (e.g., type I IFNs, interleukin (IL)-6, and IL-23), a TYK2 inhibitor may be preferable over a JAK1/2 inhibitor. This preference arises as the TYK2 inhibitor can effectively block the activity of proinflammatory and tissue-damaging cytokines (e.g., type I IFNs), while having less negative impact on the intestine’s ability to respond to type III IFN, thereby enabling it to enter an antiviral state.

Cytokines are small molecules that play a crucial role in causing inflammatory diseases. Janus kinase (JAK) and tyrosine kinase (TYK) are proteins that help these messengers communicate with cells. JAK inhibitors (JAKis) are drugs that block these proteins and are becoming more and more commonly used to treat inflammatory diseases. A common side effect of JAKis is an increased risk for viral infections. In this study, it is shown that a TYK2 inhibitor drug may not weaken our defences against viruses in our intestines as much as a JAK1/2 inhibitor. As this inhibitor blocks the disease causing cytokines, this inhibitor drug may be better than the other and be associated with lower risk of infections with viruses that enter the body through the gut.

Immune-mediated inflammatory diseases (IMIDs) include autoimmune diseases and other conditions characterized by dysregulated immune responses. IMIDs are common and increasing in incidence worldwide. Cytokines are important mediators of pathology in these diseases. For example, cytokines including interleukin (IL)-6, IL-17 and tumour necrosis factor alpha (TNFα) contribute to synovial inflammation and joint destruction in rheumatoid arthritis. Furthermore, IL-17, IL-23, and TNFα are involved in causing skin inflammation in plaque psoriasis [1]. In type 1 diabetes, production of type I interferon (IFN) in the pancreatic islets during the prediabetic period has been proposed to cause excessive expression of MHC class I which thereby facilitates CD8+ T-cell-mediated killing of pancreatic beta cells [2, 3].

Many of the cytokines implicated in the pathogenesis of IMIDs act via receptor-mediated activation of Janus kinase (JAK) and signal transducer and activator of transcription (STAT) signalling pathways (recently reviewed in [4]). There are four known JAK isoforms in humans (JAK1-3 and tyrosine kinase 2 [TYK2]). Cytokines and growth factors bind to membrane‐bound cellular receptors, resulting in the activation of associated JAKs (via phosphorylation). Activated JAKs then phosphorylate the intracellular part of the receptor to allow docking and phosphorylation and thereby activation of STATs, which translocate to the nucleus to initiate gene transcription. Type I and II cytokines, including IFNs, IL-2, IL-4, IL-5, IL-10, IL-13, IL-12, IL-21, IL-22, and IL-23, as well as endocrine- and colony-stimulating factors signal via different combinations of JAK-STATs. They have critical functions in regulating antiviral and antitumoral responses, and various systemic functions, including haematopoiesis, lipid metabolism, and granulopoiesis [4].

Much work has been devoted to developing selective inhibitors of the JAK-STAT signalling pathway as therapeutic options for IMIDs, as well as other types of disease including type I interferonopathies (a subset of autoinflammatory disorders characterized by excess IFNα/β signalling) [5, 6]. JAK inhibitors (JAKis) have demonstrated striking efficacy in the treatment of several diseases (e.g., rheumatoid arthritis, atopic dermatitis, and alopecia areata) and are currently being tested for others (e.g., psoriasis, Crohn’s disease and type 1 diabetes) [1, 7, 8]. The first JAKis to be developed target two or three different JAKs and are thus broad acting. Newer generations of JAKis have shown greater specificity for selected JAKs, although partial selectivity for other JAKs has often been observed [5]. Several adverse events (AEs) were reported in trials testing the early JAKis and safety concerns have also remained when testing more recently developed agents with higher specificity [1, 5, 7, 9‒11]. Commonly reported AEs include respiratory tract infections, nasopharyngitis, diarrhoea and headache. More severe AEs such as dyslipidaemia, certain viral and bacterial infections, anaemia, and malignancies have also been documented, occasionally resulting in failure to gain regulatory approval. The side effects may stem from a lack of selectivity, with higher drug doses associated with worse AEs, but the mechanism of action may also be implicated [5, 10]. Due to this, in part, JAKis selectively targeting TYK2 have been developed and tested in clinical trials for diseases such as plaque psoriasis and psoriatic arthritis [1, 12]. Reports from trials testing one of these TYK2 inhibitors, deucravacitinib (SOTYKTU), in moderate-to-severe psoriasis showed that the drug was well tolerated and induced a mid-high rate clinical response without causing the more severe toxicities commonly reported for pan-JAKis (e.g., dyslipidaemia) [13].

Increased risk of infections (including opportunistic infections) such as nasopharyngitis, upper respiratory tract infection, tuberculosis, gastritis, urinary tract infections and herpes zoster (shingles) has been associated with the use of JAKis [10]. This could result from the inhibitors blocking antiviral immunity [14] and leads to the hypothesis that more specific JAKis, which preserve the host antiviral defence, could lower the risk of infections. A recent meta-analysis suggested that the infection risk is similar among approved JAKi drugs [9], but how these compare to more recently developed inhibitors of TYK2 (e.g., deucravacitinib, approved September 2022 [12]) remains to be fully investigated.

IFNs play a central role in antiviral immune defence by inducing the expression of IFN-induced genes (ISGs) encoding proteins with antiviral activity (e.g., ISG15, Mx1, OAS1, and PKR) and by modulating the antiviral immune response [15, 16]. Three groups of IFNs have been described: type I (IFNα, β and related molecules), II (IFNγ), and III (IFNλ1-4, also denoted IL28A, IL28B, IL29, and IFNλ4, respectively). Type I and III IFNs play important roles in the early immune response to viral pathogens (reviewed in [16, 17]). Type II IFN (IFNγ) is instrumental in activating both innate and adaptive cell-mediated immunity. Each IFN family signals via a unique receptor, which is followed by the activation of JAK/STATs. Type I (IFNαβ) and III (IFNλ) IFNs signal via their own unique receptors and activation of JAK1/TYK2, while type II (IFNγ) receptor signalling is mediated via JAK1/JAK2 [16]. Due to these similarities and differences, JAK1 and JAK1/2 inhibitors block the effect of all three IFN families, while TYK2 specific inhibitors are expected to suppress type I and III but not type II IFN signalling. However, recent studies have questioned whether TYK2 is essential for type III IFN (IFNλ) signalling [16, 18, 19].

Many viruses that are common in humans infect via mucosal sites including the intestinal and respiratory tracts. These include, for example, entero-, noro-, rota-, and influenza viruses. Several studies have documented the paramount role of type III IFNs in protecting the intestinal barrier, while type I IFNs are important for preventing systemic viral spread [16, 17, 20‒23]. However, despite mounting evidence for the important role of type III IFNs in protecting the gut from viral infection, virtually nothing is known about whether treatment with JAKis interferes with the antiviral defence mechanisms of the human intestinal epithelium.

The aim of the present study was to investigate whether clinically approved JAKis affect type III IFN-induced antiviral responses in epithelial cells lining the gastrointestinal tract. As type III IFNs (and type I IFNs) signal via JAK1/TYK2, we focused on JAKis that inhibit these signalling molecules and not JAK3. Furthermore, we examined whether there was any difference in the effect on the antiviral response if JAK1/2 or TYK2 was inhibited. To this end, we studied the effect of the JAK1/2 inhibitor baricitinib and the TYK2 selective inhibitor deucravacitinib on human intestinal epithelial cells (IECs; Caco-2 and HT-29). MxA, a key mediator of cellular antiviral responses, served as an indicator for the IFN-induced cellular antiviral response. Drug concentrations commonly used for in vitro studies as well as those that are clinically relevant were employed.

Cell Lines

Caco-2 cells originally obtained from Dr. Aspenström Fagerlund, the Swedish National Food Agency, Stockholm, Sweden, were cultured in DMEM (Gibco, Life Technologies Fisher Scientific, Göteborg, Sweden) supplemented with 20% heat-inactivated fetal bovine serum (FBS, Sigma-Aldrich, Merck KGaA, Darmstadt, Germany) and 2 mml-glutamine (Cytiva Europe GmbH, Uppsala, Sweden). HT-29 cells (HTB-38; American Type Culture Collection, ATCC, Manassas, VA, USA) were cultured in McCoys 5 A medium (Merck KGaA, Darmstadt, Germany) supplemented with 2 mml-Glutamine and 10% heat-inactivated FBS. All cell culture media was free of antibiotics. Cells were cultured at 37°C, 5% CO2 and passaged when they reached confluency. Mycoplasma testing was performed, and cells were confirmed mycoplasma negative.

Treatment with JAK1/JAK2 and TYK2 Inhibitors

Cells were plated to reach a confluence level of 50–70% on day 2 post seeding. Cells were then exposed to either baricitinib (Aviva Systems Biology, CA, USA) or deucravacitinib (BMS-986165; MedChemExpress, NJ, USA), both dissolved in DMSO, at the concentrations indicated. Control cells were exposed to the solvent DMSO at corresponding concentrations. In some experiments, cells were later exposed to IFNs or buffer alone (see below) in the continued presence of JAK1/JAK2 or TYK2 inhibitor. Cells were then collected for mRNA extraction, FACS staining, or infected by Coxsackie B virus (CVB) virus (see below).

IFN Treatment

Cells were exposed to IFNα (IntronA, Paranova Läkemedel AB, Solna, Sweden), IFNλ1 or IFNλ2 (PeproTech, NJ, USA) or buffer alone (0.1% BSA, Sigma-Aldrich, Merck KGaA, Darmstadt, Germany). Cytokines were used at the concentrations indicated in each results section (IFNα 0–3,000 U/mL, IFNλ1 and IFNλ2 0–300 ng/mL, 0.1% BSA ≤1:500) and stimulated for the indicated length of time (6–72 h).

FACS Analysis

Upon termination of the experiment, cells were collected by trypsinization, then unspecific binding was blocked with 10% FCS. Cells were stained for viability assessment with Live/Dead Fixable Far-Red stain (ThermoFisher Scientific, MA, USA) and HLA-ABC expression (Antibody clone W6/33, BioLegend, CA, USA). Cells were acquired by the BD Accuri C6 (BD Biosciences, Stockholm, Sweden) and analysed using FlowJo v10 software (BD Biosciences, OR, USA).

Virus Stock, Infection of Cells, and Measurement of Cytopathic Effect

CVB3-V13 was originally obtained as previously described (Stone, 2021), propagated and titered in RD cells. Pilot experiments were carried out to optimize infection conditions (data not shown). Cells were plated in 96-well plates at a concentration of 5 × 103 cells/well (Caco-2) or 1 × 104 cells/well (HT-29). Infections were performed using the protocol described in [21]. In brief, at the day of infection, cells in at least two individual wells were counted to calculate the average number of cells/well. Cells were then washed in PBS followed by infection by CVB3-V13 diluted to the indicated multiplicity of infection (MOI) using serum free medium. Control cells were mock infected (using serum free medium). One hour later, the cells were washed 3 times in PBS and incubated for the indicated time in complete media. If present prior to infection, JAK/STAT and TYK2 inhibitors and/or IFNs were not included during the 1 h infection period, but after this, they were re-added to the culture medium at similar concentrations as before infection. After the indicated time periods, the media was removed, and the cells were fixed using modified Carnoy’s reagent (25% Glacial Acetic Acid in Methanol) followed by staining with 0.2% crystal violet. Optical density was measured at 595 nm using a Bio-Rad xMark Microplate Spectrophotometer (Bio-Rad Laboratories AB, Solna, Sweden). In every experiment (biological replicate), each condition was performed in at least triplicate (technical replicates) and a mean of the technical replicates was calculated.

RNA Extraction

Total RNA was extracted using the Qiagen RNeasy mini kit (Qiagen, Sollentuna, Sweden). Briefly, cells were washed in ice-cold PBS followed by the addition of Buffer RLT. Retrieved cell suspensions were homogenized using QIAshredder columns, loaded onto RNeasy spin column and on-column DNase treatment was performed. RNA was eluted in RNase free water. RNA concentration and purity were measured using a NanoDrop ND-1000 spectrophotometer (Saveen Werner AB, Limhamn, Sweden).

qRT-PCR

Total RNA (2 μg per sample) was reverse transcribed to cDNA using the High Capacity cDNA Reverse Transcription Kit from Applied Biosystems (Life Technologies Fisher Scientific, Göteborg, Sweden) according to the manufacturer’s instructions. The mRNA expression of ISG15, MX1 and GAPDH was quantified by real-time PCR using QuantiTect primers (Qiagen) (MX1: QT00090895; ISG15: QT00072814; GAPDH: QT00079247) and RT2 SYBR Green qPCR Mastermix (Qiagen). Samples were analysed with the QuantStudio 5 Real-Time PCR System (Applied Biosystems, Life Technologies Fisher Scientific, Göteborg, Sweden). The cycle detection threshold for both MX1 and ISG15 was set to a Ct value of 35, samples below the detection level was considered gene not expressed. Gene expression levels were normalized to the expression of GAPDH. The data are presented as either 2−ΔCt or ΔΔCt normalized to control sample, as indicated in each figure legend.

RT2 Profiler PCR Array

Total RNA from 2 to 6 individual experiments per setup was mixed in equal concentration to gain 3 µg total RNA. cDNA synthesis was performed using the RT2 first strand kit according to manufacturer’s instructions (Qiagen). RT2 profiler PCR arrays for human interferon and receptors (PAHS-064Z-D) (Qiagen) were performed on each cDNA using RT2 SYBR Green qPCR Master Mix in the Bio-Rad CFX96 real-time PCR system. The manual Ct threshold was set to 300 across all arrays, quality control on RTC and gDNA Ct-values pass was confirmed. Array relative quantity normalization was performed using the five housekeeping genes (ACTB, B2M, GAPDH, HPRT1, RPLP0) and normalized gene expression value was calculated for all genes compared to the housekeeping genes in each array. Analysis was performed using the online RT2 Profiler PCR Data Analysis (https://dataanalysis2.qiagen.com/pcr/analysis). Data are presented as differential gene expression compared to control cells with 2-fold primer efficiency across all genes (2DGE), or as relative quantity (2−ΔCt) compared to the geometric mean of the housekeeping genes.

Statistical Analysis

Statistical analysis was performed using the GraphPad Prism 9 software (GraphPad Software, La Jolla, CA, USA). Comparisons between two groups were performed using the Student’s t-test. Comparisons between groups in studies including two independent variables were done using two-way ANOVA with Šídák's multiple comparisons test. A p value <0.05 was considered statistically significant. Data are presented as the mean ± standard deviation (S.D.).

IFN-Induced Upregulation of ISG Expression in IECs Is Inhibited by JAK1/2 and TYK2 Inhibitors

We have previously demonstrated that IECs respond to both type I and III IFNs by expressing genes encoding ISGs (e.g., ISG15, MX1, PKR, and OAS2) [21]. By assessing the induction of ISG15 and MX1 mRNA expression following a 6 h exposure of Caco-2 cells to increasing concentrations of IFNs, we confirmed that a robust induction of MX1 mRNA expression was reached after exposure to 100 ng/mL IFNλ1 or IFNλ2, and 1,000 U/mL IFNα (online suppl. Fig. S1; for all online suppl. material, see https://doi.org/10.1159/000538571). We also noted a clear increase in ISG15 mRNA expression after stimulation with increasing concentrations of all three IFNs (online suppl. Fig. S1). In contrast to MX1, the basal expression level of ISG15 was very low (Ct-values in control cells between 35 and 37 compared to 26 and 28 for MX1) and as the accuracy of the PCR-based quantification is poor at low expression levels, ISG15 expression was not assessed in further studies. We next analysed the expression of a larger set of ISGs and related genes in Caco-2 cells treated with baricitinib, a JAK1/2 inhibitor, or deucravacitinib, a selective TYK2 inhibitor [12], at concentrations commonly used for in vitro studies on IFN signalling [2, 24, 25] followed by exposure to type III IFN (IFNλ1). Consistent with the previous studies [21], the expression of MX1, ISG15 and numerous other ISGs were induced by IFNλ1. However, this expression was reduced or completely prevented by both inhibitors (Fig. 1a; online suppl. Table S1). Furthermore, cell surface expression of MHC class I (HLA-ABC) was upregulated in both Caco-2 cells after 24 h exposure to IFNλ and this was attenuated by both baricitinib and deucravacitinib (Fig. 1b). Similar results were obtained when studying IFNλ-induced MHC class I expression in HT-29 cells (Fig. 1b). These studies confirmed that ISG expression is increased in both Caco-2 and HT-29 cells following IFN stimulation and provide new data, demonstrating that JAK1/2 and TYK2 inhibitors prevent this upregulation.

Fig. 1.

JAK1/2 and Tyk2 inhibitors block type III IFN induced antiviral gene and HLA class I expression in IECs. a Global IFN signalling pathway analysis was performed by RT2 Profiler PCR arrays on Caco-2 cells pre-treated with JAK1/2 inhibitor Bar (4 µm) or Tyk2 inhibitor Deuc (2 µm), or mock treated with buffer alone (DMSO) for 2 h before stimulated with IFNλ1 (100 ng/mL) or mock for an additional 6 h in the presence of inhibitors. Differential gene expression was calculated using housekeeping genes and buffer (DMSO) + mock-treated cell gene expression was set as baseline expression. Genes with expression above or below 1.5-fold in IFNλ1 + buffer (DMSO) treated cells are shown. Gene expression below detection is shown as grey. Two to six separate experiments were pooled for each array per treatment group. b Caco-2 and HT-29 cells were pre-treated with JAK1/2 inhibitor Bar (4 µm), Tyk2 inhibitor Deuc (2 µm), or buffer alone (DMSO) for 2 h and then stimulated with type III IFN (IFNλ1, 100 ng/mL) or buffer alone (BSA) for 24 h. Representative histograms depict modal curves of HLA-ABC cell surface expression: isotype control (grey line), mock-treated control cells (light grey filled), IFNλ1 (dark grey filled), Bar + IFNλ1 (dotted line), and Deuc + IFNλ1 (dashed line). Summary of the independent experiments are shown as bar graphs, with gMFI normalized to the DMSO + BSA controls of each experiment. Bar, baricitinib; Deuc, deucravacitinib.

Fig. 1.

JAK1/2 and Tyk2 inhibitors block type III IFN induced antiviral gene and HLA class I expression in IECs. a Global IFN signalling pathway analysis was performed by RT2 Profiler PCR arrays on Caco-2 cells pre-treated with JAK1/2 inhibitor Bar (4 µm) or Tyk2 inhibitor Deuc (2 µm), or mock treated with buffer alone (DMSO) for 2 h before stimulated with IFNλ1 (100 ng/mL) or mock for an additional 6 h in the presence of inhibitors. Differential gene expression was calculated using housekeeping genes and buffer (DMSO) + mock-treated cell gene expression was set as baseline expression. Genes with expression above or below 1.5-fold in IFNλ1 + buffer (DMSO) treated cells are shown. Gene expression below detection is shown as grey. Two to six separate experiments were pooled for each array per treatment group. b Caco-2 and HT-29 cells were pre-treated with JAK1/2 inhibitor Bar (4 µm), Tyk2 inhibitor Deuc (2 µm), or buffer alone (DMSO) for 2 h and then stimulated with type III IFN (IFNλ1, 100 ng/mL) or buffer alone (BSA) for 24 h. Representative histograms depict modal curves of HLA-ABC cell surface expression: isotype control (grey line), mock-treated control cells (light grey filled), IFNλ1 (dark grey filled), Bar + IFNλ1 (dotted line), and Deuc + IFNλ1 (dashed line). Summary of the independent experiments are shown as bar graphs, with gMFI normalized to the DMSO + BSA controls of each experiment. Bar, baricitinib; Deuc, deucravacitinib.

Close modal

JAK1/2 and TYK2 Inhibitors Reduce Basal ISG Expression Without Affecting Permissiveness of IECs to Enterovirus Infection

Treatment with JAK1/2 and TYK2 inhibitors may not have an impact on IFN-induced expression of ISGs alone but may also affect the basal expression of ISGs and other IFN-related genes in IECs. In line with this, the basal expression levels of several IFN-related genes were decreased in Caco-2 cells treated with either baricitinib or deucravacitinib alone (Fig. 2a; online suppl. Table S2). The relative mRNA expression of MX1 (shown in Fig. 2b) was reduced to around 60% compared to that in untreated control cells (Caco-2 + baricitinib: 0.66 ± 0.25; Caco-2 + deucravacitinib: 0.51 ± 0.13; HT-29 + baricitinib: 0.61 ± 0.11; HT-29 + deucravacitinib: 0.63 ± 0.19).

Fig. 2.

JAK1/2 and TYK2 inhibitors reduce basal IFN-related gene expression in IECs. a Caco-2 cells treated with Bar (4 μm) (grey bar), deucravacitinib (2 μm) (white bar), or buffer alone (DMSO) (black bar) for 8 h were analysed by RT2 Profiler PCR arrays for human IFN and receptor response gene expression. Relative expression (2-(ΔCT)) for the 15 most down-regulated genes by either Bar or deucravacitinib compared to housekeeping genes (RPLP0, ACTB, GAPDH, HPRT, B2M, HPRT1) are shown. b Caco-2 and HT-29 cells were treated with Bar (4 µm) (grey bars), deucravacitinib (2 µm) (white bars) or buffer alone (DMSO, black bars) for 8 h. The expression of the IFN-inducible gene MX1 was measured by qRT-PCR. The expression levels were normalized to GAPDH and presented as relative expression (2-(ΔCt) x 10−4) for 5–6 independent experiments. *p < 0.05, **p < 0.01, paired t test.

Fig. 2.

JAK1/2 and TYK2 inhibitors reduce basal IFN-related gene expression in IECs. a Caco-2 cells treated with Bar (4 μm) (grey bar), deucravacitinib (2 μm) (white bar), or buffer alone (DMSO) (black bar) for 8 h were analysed by RT2 Profiler PCR arrays for human IFN and receptor response gene expression. Relative expression (2-(ΔCT)) for the 15 most down-regulated genes by either Bar or deucravacitinib compared to housekeeping genes (RPLP0, ACTB, GAPDH, HPRT, B2M, HPRT1) are shown. b Caco-2 and HT-29 cells were treated with Bar (4 µm) (grey bars), deucravacitinib (2 µm) (white bars) or buffer alone (DMSO, black bars) for 8 h. The expression of the IFN-inducible gene MX1 was measured by qRT-PCR. The expression levels were normalized to GAPDH and presented as relative expression (2-(ΔCt) x 10−4) for 5–6 independent experiments. *p < 0.05, **p < 0.01, paired t test.

Close modal

Lowered basal ISG expression caused by JAKi treatment may result in a weakened antiviral defence in the gut and increase permissiveness to viruses infecting via the gastrointestinal tract. CVB belongs to the enterovirus genus and infects via the intestine. Infections cause the common cold and occasionally more severe and life-threatening illnesses (e.g., meningitis, myocarditis, and pleurodynia). Infections with this type of virus have also been associated with the development of type 1 diabetes [26]. To address if exposure to baricitinib or deucravacitinib increases the permissiveness of IECs to CVB infection, IECs pre-treated with inhibitors or mock treated were infected with CVB serotype 3 (CVB3) at increasing MOIs. Two days later, cytopathic effect (CPE) was assessed by measuring the optical density of the cell monolayer. CPE was visible in mock-treated cells following CVB infection at MOIs of 10−5 and 10−6 or higher for Caco-2 and HT-29 cells, respectively (online suppl. Fig. 2, and data not shown). The CPE was not different in cells preincubated with baricitinib or deucravacitinib. Taken together, these observations suggested that although basal ISG expression levels were reduced, permissiveness to infection per se was not increased following the blockage of either JAK1/2 or TYK2.

JAK1/2 and TYK2 Inhibitors Prevent IFN-Induced Protection from Enterovirus Infection in IECs

During early viral infection, IFNs released by parenchymal and immune cells have an essential function in acting on uninfected cells, stimulating the upregulation of genes involved in antiviral defence, thereby lowering permissiveness to infection [15, 16]. In a previous study, we demonstrated that type III IFNs protect IECs from CVB3 infection [21]. To examine whether inhibition of JAK1/2 or TYK2 signalling has a negative impact on the ability of IECs to enter an antiviral state following type III IFN stimulation, we pre-treated IECs with inhibitors prior to stimulation with IFNλ1 and then infected the cells with CVB3 at different MOIs. Cells exposed to type I IFN (IFNα) were included for comparison. Pre-treatment with either type III (IFNλ1) or type I (IFNα) IFN provided strong protection from infection (Fig. 3). Indeed, CPE was seen at MOIs of 10−5 and 10−6 or higher for untreated Caco-2 and HT-29 cells, respectively, while for the IFN treated cells, the virus dose had to be increased by one to two orders of magnitude (10–100 times higher infectious dose) to cause CPE. The protection afforded by IFN treatment was completely lost when cells were treated with baricitinib or deucravacitinib prior to IFN treatment and subsequent infection (Fig. 3), suggesting that infection risk is increased during treatment with JAK1/2 and TYK2 inhibitors.

Fig. 3.

Type I and III IFN-induced reduced permissiveness to CVB3 infection is abrogated by high concentration of JAK1/2 and TYK2 inhibitors. CPE of CVB3 infection was measured as loss of cell monolayer after 48 h of infection. Caco-2 and HT-29 cells were cultured in 96-well plates. At 50% confluency cells were treated with (a) baricitinib (4 µm), (b) deucravacitinib (2 µm) or diluted solvent (DMSO). Two hours after addition of inhibitor, IFNλ1 (100 ng/mL), IFNα (1,000 U/mL), or buffer control (BSA) were added in continuous presence of inhibitors or solvent control. After 24 h, the cells were infected with indicated MOI of CVB3-V13 or mock virus control (MOI 0). At 48 h post infection, cells were fixed to the culture plate and stained with crystal violet. CPE was measured by optical densitometry (OD) measurements (595 nm) and plotted as relative density of mock infected buffer treated control. In each experiment, 4–6 individual wells of cells were exposed to each condition and MOI after which an average of the wells was calculated. Data are presented as one experiment out of two with similar results.

Fig. 3.

Type I and III IFN-induced reduced permissiveness to CVB3 infection is abrogated by high concentration of JAK1/2 and TYK2 inhibitors. CPE of CVB3 infection was measured as loss of cell monolayer after 48 h of infection. Caco-2 and HT-29 cells were cultured in 96-well plates. At 50% confluency cells were treated with (a) baricitinib (4 µm), (b) deucravacitinib (2 µm) or diluted solvent (DMSO). Two hours after addition of inhibitor, IFNλ1 (100 ng/mL), IFNα (1,000 U/mL), or buffer control (BSA) were added in continuous presence of inhibitors or solvent control. After 24 h, the cells were infected with indicated MOI of CVB3-V13 or mock virus control (MOI 0). At 48 h post infection, cells were fixed to the culture plate and stained with crystal violet. CPE was measured by optical densitometry (OD) measurements (595 nm) and plotted as relative density of mock infected buffer treated control. In each experiment, 4–6 individual wells of cells were exposed to each condition and MOI after which an average of the wells was calculated. Data are presented as one experiment out of two with similar results.

Close modal

TYK2 Inhibition Preferentially Blocks Type I IFN-Induced Gene Expression in IECs

Given the key role of type III IFNs in protecting the intestinal tract from infection [16, 17, 20‒23], we hypothesized that a preserved type III IFN response in the intestine may be beneficial for individuals treated with JAKis. Therefore, we next asked whether the JAK1/2 or TYK2 inhibitors have any difference in impact on type III IFN-induced antiviral defence. To this end, Caco-2 and HT-29 cells were treated with increasing concentrations of either baricitinib or deucravacitinib, including those corresponding to plasma concentrations at approved treatment doses [27]. Two hours later, the cells were stimulated with type III (IFNλ1) IFN for 6 h, after which they were collected for gene expression analysis. For comparison, separate wells of cells were stimulated with type I (IFNα). Both baricitinib and deucravacitinib inhibited IFNλ1-and IFNα-induced MX1 expression in a dose-dependent manner (Fig. 4). At all concentrations tested, baricitinib (JAK1/2 inhibition) was equally efficient in blocking IFNλ1-or IFNα-induced MX1 gene expression. In contrast, deucravacitinib (TYK2 inhibition) was much less effective in inhibiting IFNλ1-than IFNα-induced MX1 gene expression at equimolar concentrations below 0.25 μm (Fig. 4), indicating that at clinically relevant concentrations, a TYK2 inhibitor better preserves type III IFN-induced host antiviral defence pathways in IECs than a more broadly acting JAK1/2 inhibitor.

Fig. 4.

TYK2 but not JAK1/2 inhibition preferentially blocks type I IFN-induced ISG expression in IECs. Caco-2 and HT-29 cells were treated with either baricitinib or deucravacitinib in increasing concentrations, spanning clinically relevant concentrations (grey box). Two hours after inhibitor was added, IFNs were added to the cell culture with inhibitors remaining at indicated concentrations (IFNλ circle black line, IFNα diamonds grey line). Control cells were buffer treated (BSA and DMSO) or treated with IFNλ (100 ng/mL) or IFNα (1000U/mL) and buffer (0.2% DMSO). At 6 h of cytokine stimuli, cells were harvested, RNA extracted, cDNA was produced and qRT-PCR for MX1 and GAPDH was performed. Gene expression of MX1 in each condition was normalized to GAPDH. Data are plotted as % of maximum MX1 expression for each cytokine without inhibitor, from three independent experiments. Significant differences between IFNλ and IFNα MX1 expression was assessed by 2-way ANOVA with Šídák’s multiple comparisons test, ** ≤0.005, *** <0.0001.

Fig. 4.

TYK2 but not JAK1/2 inhibition preferentially blocks type I IFN-induced ISG expression in IECs. Caco-2 and HT-29 cells were treated with either baricitinib or deucravacitinib in increasing concentrations, spanning clinically relevant concentrations (grey box). Two hours after inhibitor was added, IFNs were added to the cell culture with inhibitors remaining at indicated concentrations (IFNλ circle black line, IFNα diamonds grey line). Control cells were buffer treated (BSA and DMSO) or treated with IFNλ (100 ng/mL) or IFNα (1000U/mL) and buffer (0.2% DMSO). At 6 h of cytokine stimuli, cells were harvested, RNA extracted, cDNA was produced and qRT-PCR for MX1 and GAPDH was performed. Gene expression of MX1 in each condition was normalized to GAPDH. Data are plotted as % of maximum MX1 expression for each cytokine without inhibitor, from three independent experiments. Significant differences between IFNλ and IFNα MX1 expression was assessed by 2-way ANOVA with Šídák’s multiple comparisons test, ** ≤0.005, *** <0.0001.

Close modal

Type III IFNs activate intracellular signalling via JAK/STATs and have an essential role in protecting the intestinal mucosa from infection [16, 17]. In this study, we found that drugs which target and inhibit JAK1/2 or TYK2 signalling have an impact on the autonomous antiviral defence in IECs. At high concentrations, the inhibitors lowered steady-state ISG expression; however, this occurred without increasing permissiveness to a common human enteric virus, CVB3. Importantly, our study revealed a difference in the efficiency of a JAK1/2 and a TYK2 inhibitor in blocking type III IFN-induced ISG expression at clinically relevant drug concentrations. While the JAK1/2 inhibitor blocked both type I and III IFN-induced ISG expression at such concentrations, the more selective TYK2 inhibitor only showed efficient suppression of type I IFN-induced ISG expression. These data suggest that TYK2 inhibition at drug concentrations relevant to clinical practice prevents type I IFN-mediated signalling while preserving the type III IFN response, known to be critical for intestinal antiviral barrier protection.

Type I and III IFNs are produced early following viral infection [15]. They have unique and complementary activities which contribute to controlling and clearing viral infections [15, 16]. While the type I IFN receptor is ubiquitously expressed, the type III IFN receptor is mainly expressed on cells of epithelial origin and a few immune cell types. This in part explains the different actions of the two IFN families; both induce the expression of ISGs (many of which have antiviral activity), but their primary roles are in different body compartments during infection. There is extensive literature demonstrating that type III IFNs are of greater importance for the protection of mucosal sites than type I IFNs, which in turn have a more prominent role in regulating the systemic spread of infection and are also more commonly involved in excessive inflammatory responses. For example, type III IFNs but not type I IFNs are important in controlling rotavirus infection in both human IECs [28] and in the intestine of mice [29, 30]. Human IECs lacking the type III IFN receptor showed increased permissiveness for SARS-CoV-2 infection, whereas there was no difference in permissiveness to infection between control cells and cells depleted of the type I IFN receptor [31]. Furthermore, type III IFNs have a key role in limiting enterovirus replication in the intestinal epithelium [23].

There is a well-documented increased risk of infections associated with the use of JAKis [10]. Nasopharyngitis, upper respiratory tract infections and diarrhoea are commonly reported AEs, and, with several JAK1/2/3 inhibitors, reactivation of latent infections (e.g., varicella zoster and herpes simplex virus) has been described. The risk of infections varies between different JAKis (e.g., [32]) and is also dependent on the dose administered [10]. This suggests a correlation between the magnitude of inhibition and the risk of infection. The aim of the current study was to investigate and compare the effect of JAK1/2 (baricitinib) and TYK2 (deucravacitinib) inhibitors on intestinal antiviral defence mechanisms, with a particular focus on type III IFNs.

Using concentrations of baricitinib and deucravacitinib comparable to those studied previously [2, 24, 25], we observed a clear reduction in baseline ISG expression in human IECs cultured with either baricitinib or deucravacitinib. This suggests that IECs have a basal expression of proteins involved in antiviral defence which is dependent on JAK1/2 and TYK2 signalling and is in line with studies demonstrating homoeostatic IFN-stimulated gene expression in the intestinal epithelium [17, 33]. For example, Van Winkle et al. elegantly showed that bacterial microbiota stimulates homoeostatic ISG expression in mouse intestinal epithelium via immune cell derived IFNλ. Furthermore, when this type III IFN-induced antiviral response is absent, mice show dramatically increased susceptibility to rotavirus infection [33]. Our experimental in vitro model system lacks the influence of microbiota and immune cells and whether the low constitutive expression of ISGs in human IEC can be explained by other cell-intrinsic factors remains to be understood. However, we found that although expression of host antiviral defence in IECs was attenuated, the inhibitors did not increase susceptibility to infection by a common enteric virus per se.

Early during viral infection, cells at the local site of infection respond by producing and secreting type I and III IFNs. Through autocrine and paracrine signalling, IFNs induce an antiviral state that protects uninfected cells from infection and prevents unrestricted virus replication in cells that are already infected [15, 16]. As previously shown [21], treatment with IFNs reduced the permissiveness of IECs to CVB infection. This protection was abolished if the cells were treated with a high concentration of inhibitor, showing that although JAKi treatment does not increase permissiveness to infection per se, it could reduce critical IFN-induced intestinal antiviral defence leading to rampant virus replication.

Up to this point, our studies utilized drug concentrations previously employed to investigate the effects of baricitinib and deucravacitinib on IFN signalling in vitro [2, 24, 25]. However, the concentrations we used exceed both the anticipated plasma levels at approved treatment doses [27] and the IC50 for the drugs. Consequently, we conducted dose-response studies to explore more relevant drug concentrations. These analyses revealed an interesting and significant observation: JAK1/2 and TYK2 inhibitors exhibit distinct effects in blocking type III IFN-mediated antiviral defence in the intestine. At drug concentrations similar to those estimated in patients receiving treatment [27], the TYK2 inhibitor deucravacitinib did not block type III IFN-induced ISG expression as efficiently as type I IFN-induced ISG expression. This observation was made in both IEC cell lines and has several implications. First, the results suggest that in IECs, type III IFN is less dependent on TYK2 for signalling than type I IFN. To the best of our knowledge, this is the first time that such a difference has been uncovered in human IECs. These findings are in line with recent studies highlighting a low or negligible role for TYK2 in type III IFN signalling [16, 18, 34, 35] and add human IECs to the list of cell types where this has been observed. Second, our studies imply that while a JAK1/2 inhibitor affects both type I and III IFN signalling, a TYK2 inhibitor is more effective in blocking type I IFN than type III IFN signalling at clinically relevant concentrations. Given the critical importance of type III IFNs in the local antiviral defence of the gut [16, 17], these data may be of relevance for the treatment of certain IMIDs. Indeed, when JAKis are used in the treatment of diseases associated with cytokines signalling via JAK1/2- and TYK2-coupled receptors (e.g., type I IFNs, IL-6, IL-12, IL-22, and IL-23), a TYK2 inhibitor may be preferable to a JAK1/2 inhibitor, reducing risk of excessive infections by viruses infecting via the gut while still strongly blocking the activity of cytokines with proinflammatory and tissue-damaging effects. Indirect support for such a scenario comes from studies demonstrating that TYK2 variants with reduced function are linked to decreased or loss of function of type I IFN, IL-12, and IL-23 signalling and protection from autoimmune disease without, however, resulting in general immunodeficiency (e.g., [36‒39]).

It has been suggested that a few IMIDs have a viral trigger. For example, Epstein Barr virus infection has been implicated in multiple sclerosis [40]. Epstein Barr virus infects epithelial cells of the oropharynx and parotid glands and causes latent infection. Furthermore, enterovirus infections have been linked to the appearance of islet autoantibodies and subsequent development of type 1 diabetes [41, 42]. Most enteroviruses infect via the intestine and are rapidly cleared; however, continuous faecal shedding of enteroviruses was recently associated with a risk of developing islet autoimmunity [43]. Considering the heightened risk of viral infections and reactivation of latent virus infections associated with the use of JAKis [9‒11], the preservation of antiviral defence mechanisms in mucous membranes may be of importance when using JAKis to treat diseases with a suspected viral aetiology.

Both type I and II (IFNγ) IFNs are proposed to play important roles in the pathogenesis of type 1 diabetes. Type I IFN may be of importance both in the early, prediabetic stages of the disease and at clinical disease onset, while IFNγ seem to mainly contribute to pancreatic beta cell destruction mainly in the late stage (e.g., [44, 45]). Preclinical studies using JAKis for the prevention of autoimmune diabetes have demonstrated positive outcomes [46, 47]. Due to this, initiatives have been taken to investigate the efficacy of baricitinib in reducing beta cell loss after clinical disease presentation, stage 3 [8]. A new notion that IFNγ dampens the proliferation of autoreactive T cells has however called into question the appropriateness of blocking this cytokine [45]. Given that studies have shown an important role for type III IFNs in attenuating enterovirus infection of IECs (present study and [20‒23]) and in clearing chronic viral infections of the gut [48, 49], the recent approval of deucravacitinib (SOTYKTU) in the USA and Japan [12] now opens up the possibility of using a more targeted inhibition of type I IFN signalling (but not IFNγ) that may simultaneously preserve type III IFN-mediated intestinal defence against enteroviruses.

In summary, our study shows that antiviral defence mechanisms of the intestinal epithelium are affected by JAKis. We also show that at clinically relevant drug concentrations, a TYK2 inhibitor effectively block the activity of proinflammatory type I IFN but has less negative impact on critical type III IFN-induced antiviral defence mechanisms in the intestine. The information gained from these studies may help inform strategies to treat various diseases of inflammatory and autoimmune origin with greater precision and fewer side effects.

The authors thank Mrs. Selina Parvin for excellent technical support.

The cell lines used in this study were obtained from Dr. Aspenström Fagerlund, the Swedish National Food Agency, Stockholm, Sweden (Caco-2), and American Type Culture Collection, ATCC, Manassas, VA, USA (HT-29). Ethical approval for the use of these is not required in accordance with local/national guidelines.

The authors have no conflicts of interest to declare.

This work was supported by Grants from Karolinska Institutet, Sweden, including the Strategic Research Programme in Diabetes, the Swedish Child Diabetes Foundation, the Swedish Diabetes Foundation, the Swedish Heart and Lung foundation, and the Swedish Research Council. Martha A. Castro was supported by a Fulbright Fellowship. The funders had no role in the design of the study; the collection, analyses, or interpretation of data; writing the manuscript; or the decision to publish the results.

M.A.C.: experimental work, data interpretation, data curation, formal analysis, critical revision of the article, and final approval. E.E.R.: conceptualization and supervision, experimental work, data interpretation, data curation, formal analysis, critical revision of the article, and final approval. V.M.S.: supervision, experimental work, critical revision of the article, and final approval. A.P.: experimental work, final approval of article. M.F.T.: conceptualization and supervision, funding acquisition, data interpretation, drafting of the article, and final approval.

Raw data and analyses of the RT2 profiler PCR arrays are available as online supplementary Tables 1 and 22. Further enquiries can be directed to the corresponding author.

1.
Krueger
JG
,
McInnes
IB
,
Blauvelt
A
.
Tyrosine kinase 2 and Janus kinase: signal transducer and activator of transcription signaling and inhibition in plaque psoriasis
.
J Am Acad Dermatol
.
2022
;
86
(
1
):
148
57
.
2.
Colli
ML
,
Ramos-Rodriguez
M
,
Nakayasu
ES
,
Alvelos
MI
,
Lopes
M
,
Hill
JLE
, et al
.
An integrated multi-omics approach identifies the landscape of interferon-α-mediated responses of human pancreatic beta cells
.
Nat Commun
.
2020
;
11
(
1
):
2584
.
3.
Apaolaza
PS
,
Balcacean
D
,
Zapardiel-Gonzalo
J
,
Nelson
G
,
Lenchik
N
,
Akhbari
P
, et al
.
Islet expression of type I interferon response sensors is associated with immune infiltration and viral infection in type 1 diabetes
.
Sci Adv
.
2021
;
7
(
9
):
eabd6527
.
4.
Morris
R
,
Kershaw
NJ
,
Babon
JJ
.
The molecular details of cytokine signaling via the JAK/STAT pathway
.
Protein Sci
.
2018
;
27
(
12
):
1984
2009
.
5.
Spinelli
FR
,
Meylan
F
,
O'Shea
JJ
,
Gadina
M
.
JAK inhibitors: ten years after
.
Eur J Immunol
.
2021
;
51
(
7
):
1615
27
.
6.
Crow
YJ
,
Stetson
DB
.
The type I interferonopathies: 10 years on
.
Nat Rev Immunol
.
2022
;
22
(
8
):
471
83
.
7.
Nakayamada
S
,
Tanaka
Y
.
Pathological relevance and treatment perspective of JAK targeting in systemic lupus erythematosus
.
Expert Rev Clin Immunol
.
2022
;
18
(
3
):
245
52
.
8.
Waibel
M
,
Wentworth
JM
,
So
M
,
Couper
JJ
,
Cameron
FJ
,
MacIsaac
RJ
, et al
.
Baricitinib and β-cell function in patients with new-onset type 1 diabetes
.
N Engl J Med
.
2023
;
389
(
23
):
2140
50
.
9.
Bechman
K
,
Subesinghe
S
,
Norton
S
,
Atzeni
F
,
Galli
M
,
Cope
AP
, et al
.
A systematic review and meta-analysis of infection risk with small molecule JAK inhibitors in rheumatoid arthritis
.
Rheumatol
.
2019
;
58
(
10
):
1755
66
.
10.
Adas
MA
,
Alveyn
E
,
Cook
E
,
Dey
M
,
Galloway
JB
,
Bechman
K
.
The infection risks of JAK inhibition
.
Expert Rev Clin Immunol
.
2022
;
18
(
3
):
253
61
.
11.
Alves
C
,
Penedones
A
,
Mendes
D
,
Batel Marques
F
.
The safety of systemic Janus kinase inhibitors in atopic dermatitis: a systematic review and network meta-analysis
.
Eur J Clin Pharmacol
.
2022
;
78
(
12
):
1923
33
.
12.
Hoy
SM
.
Deucravacitinib: first approval
.
Drugs
.
2022
;
82
(
17
):
1671
9
.
13.
Estevinho
T
,
Le
AM
,
Torres
T
.
Deucravacitinib in the treatment of psoriasis
.
J Dermatolog Treat
.
2022
;
34
:
1
22
.
14.
Hawerkamp
HC
,
Domdey
A
,
Radau
L
,
Sewerin
P
,
Olah
P
,
Homey
B
, et al
.
Tofacitinib downregulates antiviral immune defence in keratinocytes and reduces T cell activation
.
Arthritis Res Ther
.
2021
;
23
(
1
):
144
.
15.
Lind
K
,
Huhn
MH
,
Flodstrom-Tullberg
M
.
Immunology in the clinic review series; focus on type 1 diabetes and viruses: the innate immune response to enteroviruses and its possible role in regulating type 1 diabetes
.
Clin Exp Immunol
.
2012
;
168
(
1
):
30
8
.
16.
Stanifer
ML
,
Guo
C
,
Doldan
P
,
Boulant
S
.
Importance of type I and III interferons at respiratory and intestinal barrier surfaces
.
Front Immunol
.
2020
;
11
:
608645
.
17.
Wirusanti
NI
,
Baldridge
MT
,
Harris
VC
.
Microbiota regulation of viral infections through interferon signaling
.
Trends Microbiol
.
2022
;
30
(
8
):
778
92
.
18.
Fuchs
S
,
Kaiser-Labusch
P
,
Bank
J
,
Ammann
S
,
Kolb-Kokocinski
A
,
Edelbusch
C
, et al
.
Tyrosine kinase 2 is not limiting human antiviral type III interferon responses
.
Eur J Immunol
.
2016
;
46
(
11
):
2639
49
.
19.
Schnepf
D
,
Crotta
S
,
Thamamongood
T
,
Stanifer
M
,
Polcik
L
,
Ohnemus
A
, et al
.
Selective Janus kinase inhibition preserves interferon-lambda-mediated antiviral responses
.
Sci Immunol
.
2021
;
6
(
59
):
eabd5318
.
20.
Good
C
,
Wells
AI
,
Coyne
CB
.
Type III interferon signaling restricts enterovirus 71 infection of goblet cells
.
Sci Adv
.
2019
;
5
(
3
):
eaau4255
.
21.
Stone
VM
,
Ringqvist
EE
,
Larsson
PG
,
Domsgen
E
,
Holmlund
U
,
Sverremark-Ekstrom
E
, et al
.
Inhibition of type III interferon expression in intestinal epithelial cells-A strategy used by coxsackie B virus to evade the host’s innate immune response at the primary site of infection
.
Microorganisms
.
2021
;
9
(
1
):
105
.
22.
Wells
AI
,
Coyne
CB
.
An in vivo model of echovirus-induced meningitis defines the differential roles of type I and type III interferon signaling in central nervous system infection
.
J Virol
.
2022
;
96
(
13
):
e0033022
.
23.
Wells
AI
,
Grimes
KA
,
Coyne
CB
.
Enterovirus replication and dissemination are differentially controlled by type I and III interferons in the gastrointestinal tract
.
mBio
.
2022
;
13
(
3
):
e0044322
.
24.
Coomans de Brachene
A
,
Castela
A
,
Op de Beeck
A
,
Mirmira
RG
,
Marselli
L
,
Marchetti
P
, et al
.
Preclinical evaluation of tyrosine kinase 2 inhibitors for human beta-cell protection in type 1 diabetes
.
Diabetes Obes Metab
.
2020
;
22
(
10
):
1827
36
.
25.
Aota
K
,
Yamanoi
T
,
Kani
K
,
Ono
S
,
Momota
Y
,
Azuma
M
.
Inhibition of JAK-STAT signaling by baricitinib reduces interferon-gamma-induced CXCL10 production in human salivary gland ductal cells
.
Inflammation
.
2021
;
44
(
1
):
206
16
.
26.
Marjomaki
V
,
Flodstrom-Tullberg
M
.
Coxsackie B virus
.
Trends Microbiol
.
2022
;
30
(
6
):
606
7
.
27.
Chimalakonda
A
,
Burke
J
,
Cheng
L
,
Catlett
I
,
Tagen
M
,
Zhao
Q
, et al
.
Selectivity profile of the tyrosine kinase 2 inhibitor deucravacitinib compared with Janus kinase 1/2/3 inhibitors
.
Dermatol Ther
.
2021
;
11
(
5
):
1763
76
.
28.
Doldan
P
,
Dai
J
,
Metz-Zumaran
C
,
Patton
JT
,
Stanifer
ML
,
Boulant
S
.
Type III and not type I interferons efficiently prevent the spread of rotavirus in human intestinal epithelial cells
.
J Virol
.
2022
;
96
(
17
):
e0070622
.
29.
Pott
J
,
Mahlakoiv
T
,
Mordstein
M
,
Duerr
CU
,
Michiels
T
,
Stockinger
S
, et al
.
IFN-lambda determines the intestinal epithelial antiviral host defense
.
Proc Natl Acad Sci U S A
.
2011
;
108
(
19
):
7944
9
.
30.
Hernandez
PP
,
Mahlakoiv
T
,
Yang
I
,
Schwierzeck
V
,
Nguyen
N
,
Guendel
F
, et al
.
Interferon-lambda and interleukin 22 act synergistically for the induction of interferon-stimulated genes and control of rotavirus infection
.
Nat Immunol
.
2015
;
16
(
7
):
698
707
.
31.
Stanifer
ML
,
Kee
C
,
Cortese
M
,
Zumaran
CM
,
Triana
S
,
Mukenhirn
M
, et al
.
Critical role of type III interferon in controlling SARS-CoV-2 infection in human intestinal epithelial cells
.
Cell Rep
.
2020
;
32
(
1
):
107863
.
32.
Xu
Q
,
He
L
,
Yin
Y
.
Risk of herpes zoster associated with JAK inhibitors in immune-mediated inflammatory diseases: a systematic review and network meta-analysis
.
Front Pharmacol
.
2023
;
14
:
1241954
.
33.
Van Winkle
JA
,
Peterson
ST
,
Kennedy
EA
,
Wheadon
MJ
,
Ingle
H
,
Desai
C
, et al
.
Homeostatic interferon-lambda response to bacterial microbiota stimulates preemptive antiviral defense within discrete pockets of intestinal epithelium
.
Elife
.
2022
;
11
:
11
.
34.
Karaghiosoff
M
,
Neubauer
H
,
Lassnig
C
,
Kovarik
P
,
Schindler
H
,
Pircher
H
, et al
.
Partial impairment of cytokine responses in Tyk2-deficient mice
.
Immunity
.
2000
;
13
(
4
):
549
60
.
35.
Kreins
AY
,
Ciancanelli
MJ
,
Okada
S
,
Kong
XF
,
Ramirez-Alejo
N
,
Kilic
SS
, et al
.
Human TYK2 deficiency: mycobacterial and viral infections without hyper-IgE syndrome
.
J Exp Med
.
2015
;
212
(
10
):
1641
62
.
36.
Dendrou
CA
,
Cortes
A
,
Shipman
L
,
Evans
HG
,
Attfield
KE
,
Jostins
L
, et al
.
Resolving TYK2 locus genotype-to-phenotype differences in autoimmunity
.
Sci Transl Med
.
2016
;
8
(
363
):
363ra149
.
37.
Greb
JE
,
Goldminz
AM
,
Elder
JT
,
Lebwohl
MG
,
Gladman
DD
,
Wu
JJ
, et al
.
Psoriasis
.
Nat Rev Dis Primers
.
2016
;
2
:
16082
.
38.
Gorman
JA
,
Hundhausen
C
,
Kinsman
M
,
Arkatkar
T
,
Allenspach
EJ
,
Clough
C
, et al
.
The TYK2-P1104A autoimmune protective variant limits coordinate signals required to generate specialized T cell subsets
.
Front Immunol
.
2019
;
10
:
44
.
39.
Pellenz
FM
,
Dieter
C
,
Lemos
NE
,
Bauer
AC
,
Souza
BM
,
Crispim
D
.
Association of TYK2 polymorphisms with autoimmune diseases: a comprehensive and updated systematic review with meta-analysis
.
Genet Mol Biol
.
2021
;
44
(
2
):
e20200425
.
40.
Soldan
SS
,
Lieberman
PM
.
Epstein-barr virus and multiple sclerosis
.
Nat Rev Microbiol
.
2023
;
21
(
1
):
51
64
.
41.
Rodriguez-Calvo
T
.
Enteroviral infections as a trigger for type 1 diabetes
.
Curr Diab Rep
.
2018
;
18
(
11
):
106
.
42.
Nekoua
MP
,
Alidjinou
EK
,
Hober
D
.
Persistent coxsackievirus B infection and pathogenesis of type 1 diabetes mellitus
.
Nat Rev Endocrinol
.
2022
;
18
(
8
):
503
16
.
43.
Vehik
K
,
Lynch
KF
,
Wong
MC
,
Tian
X
,
Ross
MC
,
Gibbs
RA
, et al
.
Prospective virome analyses in young children at increased genetic risk for type 1 diabetes
.
Nat Med
.
2019
;
25
(
12
):
1865
72
.
44.
Akhbari
P
,
Richardson
SJ
,
Morgan
NG
.
Type 1 diabetes: interferons and the aftermath of pancreatic beta-cell enteroviral infection
.
Microorganisms
.
2020
;
8
(
9
):
1419
.
45.
De George
DJ
,
Ge
T
,
Krishnamurthy
B
,
Kay
TWH
,
Thomas
HE
.
Inflammation versus regulation: how interferon-gamma contributes to type 1 diabetes pathogenesis
.
Front Cell Dev Biol
.
2023
;
11
:
1205590
.
46.
Trivedi
PM
,
Graham
KL
,
Scott
NA
,
Jenkins
MR
,
Majaw
S
,
Sutherland
RM
, et al
.
Repurposed JAK1/JAK2 inhibitor reverses established autoimmune insulitis in NOD mice
.
Diabetes
.
2017
;
66
(
6
):
1650
60
.
47.
Ge
T
,
Jhala
G
,
Fynch
S
,
Akazawa
S
,
Litwak
S
,
Pappas
EG
, et al
.
The JAK1 selective inhibitor ABT 317 blocks signaling through interferon-gamma and common gamma chain cytokine receptors to reverse autoimmune diabetes in NOD mice
.
Front Immunol
.
2020
;
11
:
588543
.
48.
Nice
TJ
,
Baldridge
MT
,
McCune
BT
,
Norman
JM
,
Lazear
HM
,
Artyomov
M
, et al
.
Interferon-lambda cures persistent murine norovirus infection in the absence of adaptive immunity
.
Science
.
2015
;
347
(
6219
):
269
73
.
49.
Strine
MS
,
Alfajaro
MM
,
Graziano
VR
,
Song
J
,
Hsieh
LL
,
Hill
R
, et al
.
Tuft-cell-intrinsic and: extrinsic mediators of norovirus tropism regulate viral immunity
.
Cell Rep
.
2022
;
41
(
6
):
111593
.