A single layer of polarized epithelial cells lining the colonic mucosa create a semipermeable barrier indispensable for gut homeostasis. The role of intestinal epithelial cell (IEC) polarization in the maintenance of the epithelial homeostasis and in the development of inflammatory bowel diseases is not fully understood. In this review, now we report that IEC polarization plays an essential role in the regulation of IL-6/STAT3 signaling in the colonic mucosa. Our results demonstrate that autocrine STAT3 activation in IECs is mediated by the apical secretion of IL-6 in response to the basolateral stimulation with IFN-γ. This process relies on the presence of functional, IFN-γ−producing CD4+ T cells. In the absence of basolateral IFN-γ, the compartmentalization of the IL-6/STAT3 signaling is disrupted, and STAT3 is activated mainly in macrophages. Thus, in this study, we show that during inflammation, IFN-γ regulates IL-6/STAT3 signaling in IEC in the colonic mucosa.

Diarrhea, rectal bleeding, abdominal pain, and body weight loss are hallmarks of inflammatory bowel diseases (IBD) (1). IBD encompasses two main pathologies: Crohn disease and ulcerative colitis. The pathogenesis of both disorders is not well understood, but increasing evidence suggests that locally produced cytokines (2, 3) and chemokines (4, 5) play an important role not only in the establishment, but also in the progression of the pathologies.

STAT3 is a transcription factor that regulates genes involved in programed cell death, proliferation, differentiation, migration, and survival downstream of a variety of cytokines and growth factors (68). In IBD, STAT3 is constitutively active in intestinal epithelial cells (IECs) and in immune cells located at the intestinal mucosa (911). Furthermore, spontaneous colitis has been observed in mice lacking STAT3 in macrophages (12, 13), but T cell–specific deletion of STAT3 has a protective effect in autoimmune disease models (14, 15). In IECs at the colonic mucosa, STAT3 displays a proliferative and prosurvival function important for tumor initiation in colitis-associated cancer, a major complication associated with IBD (10). It is clear, therefore, that tissue-specific functions of STAT3 are necessary for maintaining intestinal homeostasis and understanding the mechanisms controlling the activation of STAT3 in the colonic mucosa during the course of inflammation is highly relevant.

In this report, by using two models of colonic inflammation, we describe that IEC polarization plays an important role in the regulation of IL-6/STAT3 signaling in the intestinal epithelium. We observed that several cell types secrete IL-6 in the colonic mucosa, including macrophages and IECs. However, in the context in which inflammation is triggered and an intact epithelium remains, only IECs are able to release IL-6 intraluminally to induce the activation of STAT3 in neighbored IECs. Activation of STAT3 in such conditions occurs mainly in the epithelial cells lining the apical surface of the crypts and happens in response to high levels of basolateral IFN-γ. We also observed that basal levels of IFN-γ produced by CD4+ cells play an important role in the apical polarization of the IL-6Rα. In the context of epithelial damage, such as that observed in ulcerated areas during colitis, STAT3 activation occurs in epithelial cells along the whole crypt axis, maybe because of the disruption in the compartmentalization of the IL-6 signal established by the epithelial barrier. Thus, epithelial barrier loss during colitis is an important factor that directly influences the activation of STAT3 via IL-6 in epithelial cells. In conclusion, polarization of the intestinal epithelium by IFN-γ could be essential for priming/conditioning the inflammatory response mediated by STAT3 in mucosal tissue.

All procedures were reviewed and approved by the Cinvestav Institutional Committee for Care and Use of Laboratory Animals. Six- to eight-week-old C57BL/6J and CRTAM-deficient (kindly donated by Genentech, San Francisco, CA) mice were bred at the Center for Research and Advanced Studies of Cinvestav. Animals were housed in a standard day and night cycle with free access to food and water.

Recombinant human and murine IFN-γ and IL-6 were obtained from PeproTech (Rocky Hill, NJ) and used at 100 U/ml and 20 ng/ml for in vitro treatments and 2 and 10 μg/kg for in vivo treatments. Abs were used as manufacturer suggested. p-Hist3 (sc-56745), STAT1 (sc-346), STAT3 (sc-482), GAPDH (sc-32233), Muc-2 (sc-15334), E-cadh (sc-59778), and IL-6R (BE0047) were obtained from Santa Cruz Biotechnology (Santa Cruz, CA) and Bio X Cell. Claudin-1 (ab15098) and F4/80 (ab6640) were obtained from Abcam. p-STAT1 (9167S), p-STAT3 (9134P), AKT1 (2938), p-AKT473 (4060), PARP (9541), PCNA (13110), Ki67 (9129), and IL-6 (CST 12912) were purchased from Cell Signaling and Claudin-2 (516100) from Invitrogen. HRP-conjugated secondary Abs were purchased from Jackson ImmunoResearch Laboratories (West Grove, PA).

Colonic epithelial cell lines SW480 and Caco-2 were obtained from American Type Culture Collection and were grown in Transwells with DMEM and high glucose medium with 10% FCS and antibiotics. Cells were maintained in a humidified incubator with 5% of CO2.

Proximal colon was identified, and a 1–2-cm segment was removed. The viscus was opened and then rinsed in cold PBS 1×. The colon was incubated in a Ca2+ chelation solution for 30 min at room temperature (RT) (966 mM NaCl, 1.5 mM KCl, 10 mM HEPES, 10 mM Tris, 27 mM Na EDTA or Na EGTA, 45 mM sorbitol, 28 mM sucrose, and 0.1% BSA). After chelation, the tissue was manually shaken to liberate the crypts. The tissue was removed, and the solution was centrifuged at 200 rpm for 2 min. The supernatant was removed, and the pellet was lysed in radioimmunoprecipitation assay buffer for Western blot or F12 medium for cytokine treatment (16, 17).

Colonic epithelial cell lines SW480 and Caco-2 were stimulated with recombinant human IFN-γ (no. 300-02) or IL-6 (no. 200-06). Control cells were treated with BSA. For colon explants and colonic isolated crypt treatment, a colon segment or crypts were incubated at RT for 1 h with recombinant murine cytokines (IFN-γ no. 315-05 or IL-6 no. 216-16) dissolved in F12 medium. The tissue or crypts were collected for analysis.

For cytokine administration, mice were randomly divided, and recombinant murine IFN-γ (no. 315-05) or IL-6 (no. 216-16) was administered i.p. Intraluminal administration of recombinant murine IFN-γ and IL-6 dissolved in 100 μl of sterile PBS was carried out using a 20GX1.5′′ probe (Cadence Science, Ref. 9920). Thirty minutes after intrarectal administration, the animals were euthanized, and the colon was collected and processed for analysis. Control mice were administrated with mouse seric albumin (MSA).

Cytokine inhibition in SW480 and Caco-2 cells was performed with human anti–IL-6R Ab (AF-227-NA) or human anti–IL-6 Ab (ab6672). IECs were treated with IFN-γ or IL-6 in presence of neutralizing Abs or isotype controls. The Abs were used according to the manufacturer’s instructions.

Mice received 2.5% (w/v) of dextran sulphate sodium (DSS) (molecular mass 40 kDa, Carbosynth, CA) dissolved in tap water. Mice were sacrificed, and colons were assessed for weight and length and processed for histology. Colon was processed as previously reported by us for immunofluorescence and Western blot analysis (18).

Cytokines were quantified from supernatants of culture media of stimulated cells by using the OptEiA Human IL-6 ELISA Set (RUO-555220; BD Biosciences), according to the manufacturer’s instructions. Briefly, well plates were coated overnight with the capture Ab for cytokine using carbonate buffer (pH 9.5) at 4°C, blocked with 1× PBS supplemented with 10% FBS (1 h). Supernatant dilutions were made using blocking buffer, added to the wells of the plate, and incubated for 2 h at RT. Detection Abs and streptavidin/HRP were added together and incubated for 1 h at RT. The o-phenylenediamine substrate solution was added, and after 15-min incubation at 37°C, reaction was halted with 2 N of H2SO4. Absorbance at 450 nm was read using an absorbance microplate reader (Tecan Sunrise, Salzburg, Austria).

Colon samples in radioimmunoprecipitation assay buffer (150 mM NaCl, 1% NP-40, 0.5% deoxycholic acid, 0.1% SDS, and 50 mM Tris; pH 8) were homogenized using Tissue Master using Omni International (Kennesaw, GA) and sonicated for 10-s maximal output. Protein concentration was determined using the BCA Protein Assay (Pierce, Thermo Fisher Scientific) and 20–25 μg of protein were loaded in denaturing SDS-PAGE. GAPDH and actin were used as loading controls.

Colon specimens were cryopreserved in O.C.T. (Sakura Finetek, Torrance, CA) and sectioned in 20 μm using a cryostat (Leica, Nussloch, Germany). Then, they were fixed with paraformaldehyde (20 min; RT) and permeabilized with 100% methanol (20 min; −20°C). Nonspecific binding interactions were prevented by blocking the tissue with 2% BSA (BSA, Sigma-Aldrich, St. Louis, MO) for 1 h at RT, and primary Abs were incubated at 4°C overnight. Alexa Fluor–labeled secondary Abs were incubated then for 1 h, and nuclei were counterstained with DAPI. Representative images were taken on an LSM 510 Confocal Microscope (Zeiss, Jena, Germany) with 20×, 40×, and 60×.

Statistical significance was assumed at p < 0.05, p < 0.01, or p < 0.001. All results are displayed as mean ± SEM.

STAT3 regulates several biological processes in the gut, including colorectal cancer development, IBD establishment, and epithelial regeneration (6, 7, 19). We therefore investigated the presence of active STAT3 in the colonic mucosa of wild-type (WT) mice treated with DSS from 1–6 d to mimic different degrees of injury in the colonic mucosa. As shown in Fig. 1A, increased levels of STAT3 phosphorylated at Y705, from now on referred to as p-STAT3, were observed starting 4 d after DSS treatment. In colitic animals, p-STAT3 was enriched at nucleus of colonic IECs located along the whole crypt axis (Fig. 1B, 1C, arrows). No nuclear p-STAT3 staining was detected in IECs of healthy control animals. However, p-STAT3 was detected at nuclei of nonepithelial cells that were bordering the epithelial crypts in both control and colitic animals (Fig. 1C, asterisk). Activation of STAT3 in SW480 cells and in isolated crypts exposed to IL-6 strongly suggested a direct implication of the cytokine in the generation of p-STAT3 in IECs during colitis (Fig. 1D, 1E). Therefore, we analyzed IL-6 secretion in the mucosa of colitic mice exposed to DSS for 1–4 d. As shown in Fig. 1F, IL-6 secretion augments in mucosal explants of colitic mice starting at day 3 of treatment, a process that strongly increased on day 4, when STAT3 activation was clearly noticed (Fig. 1A). Thus, our findings suggest that IL-6 could be inducing activation of STAT3 in IECs of colitic mice.

FIGURE 1.

IL-6 activates STAT3 in colonic IECs. (A) Phospho-STAT3 (Y705) and STAT3 were analyzed by Western blotting colonic cell lysates of C57BL/6J mice treated with DSS for 1–6 d. A total of 2.5% DSS was dissolved in tap water. GAPDH was used as loading control (n = 3). (B) p-STAT3 and STAT3 were evaluated in cell lysates obtained from isolated crypts, and whole mucosal tissue was collected from control and colitic mice. C57BL/6J mice were treated with 2.5% DSS for 5 d. GAPDH was used as loading control (n = 3). (C) Immunofluorescence staining for the p-STAT3 (red) in mucosal tissue of C57BL/6J. A total of 2.5% DSS in tap water was administered for 5 d. Discontinuous white line surrounds epithelial cells located at the crypt. White asterisk indicates nonepithelial cells that are positive for p-STAT3. White arrow indicates IECs that are positive for p-STAT3. E-cadherin, green; nuclei, blue. Scale bar, 20 μm. Quantification of p-STAT3–positive IECs per crypt is shown in the graph (n = 6). ***p < 0.001 versus control (ANOVA two-way test). (D and E) p-STAT3 and STAT3 were assessed in cell lysates obtained from SW480 cells or colonic isolated crypts of C57BL/6J mice. IL-6 treatment using recombinant proteins was carried out for 2 h. GAPDH was used as loading control. Graph represents a densitometric analysis of p-STAT3 in colonic isolated crypts. ***p < 0.001 versus control (ANOVA two-way test; n = 3). (F) IL-6 secretion was analyzed in colonic explants obtained from C57BL/6J mice that were treated with 2.5% DSS for 1–4 d (n = 3). Data in graph are represented as mean ± SEM. ***p < 0.001 versus control (ANOVA two-way test).

FIGURE 1.

IL-6 activates STAT3 in colonic IECs. (A) Phospho-STAT3 (Y705) and STAT3 were analyzed by Western blotting colonic cell lysates of C57BL/6J mice treated with DSS for 1–6 d. A total of 2.5% DSS was dissolved in tap water. GAPDH was used as loading control (n = 3). (B) p-STAT3 and STAT3 were evaluated in cell lysates obtained from isolated crypts, and whole mucosal tissue was collected from control and colitic mice. C57BL/6J mice were treated with 2.5% DSS for 5 d. GAPDH was used as loading control (n = 3). (C) Immunofluorescence staining for the p-STAT3 (red) in mucosal tissue of C57BL/6J. A total of 2.5% DSS in tap water was administered for 5 d. Discontinuous white line surrounds epithelial cells located at the crypt. White asterisk indicates nonepithelial cells that are positive for p-STAT3. White arrow indicates IECs that are positive for p-STAT3. E-cadherin, green; nuclei, blue. Scale bar, 20 μm. Quantification of p-STAT3–positive IECs per crypt is shown in the graph (n = 6). ***p < 0.001 versus control (ANOVA two-way test). (D and E) p-STAT3 and STAT3 were assessed in cell lysates obtained from SW480 cells or colonic isolated crypts of C57BL/6J mice. IL-6 treatment using recombinant proteins was carried out for 2 h. GAPDH was used as loading control. Graph represents a densitometric analysis of p-STAT3 in colonic isolated crypts. ***p < 0.001 versus control (ANOVA two-way test; n = 3). (F) IL-6 secretion was analyzed in colonic explants obtained from C57BL/6J mice that were treated with 2.5% DSS for 1–4 d (n = 3). Data in graph are represented as mean ± SEM. ***p < 0.001 versus control (ANOVA two-way test).

Close modal

IL-6 is produced mainly by immune cells underneath the intestinal epithelium, such cells face the basolateral side of epithelial cells [Fig. 2A, arrow and (7, 20)]. We therefore investigated whether IL-6 administered basolaterally was able to activate STAT3 in IECs in vivo. To this end, C57BL/6J mice were injected i.p. with IL-6, and STAT3 activation was analyzed in the colon 2 h postinjection. As shown in Fig. 2B, increased levels of p-STAT3 were detected in the colonic mucosa of IL-6–treated animals. No changes in p-STAT1, total STAT1, or total STAT3 were observed. However, p-STAT3 analysis by Western blotting isolated colonic crypts or by performing immunofluorescence staining revealed that basolateral administration of IL-6 induces nuclear accumulation of STAT3 in nonepithelial cells. Those cells were always located in close proximity with the IECs, forming the crypts (Fig. 2B, 2C, asterisk). Nonepithelial cells displaying nuclear p-STAT3 staining were F4/80 positive, suggesting that basolateral administration of IL-6 activates STAT3 in macrophages (Fig. 2D, arrowhead; Supplemental Fig. 1A, asterisk). Interestingly, i.p. administration of IL-6 induces sporadic accumulation of p-STAT1 at nuclei of IECs located at the crypt surface (Supplemental Fig. 1B, arrow).

FIGURE 2.

The i.p. administration of IL-6 does not activate STAT3 in IECs present at the colonic mucosa. (A) Immunolocalization of IL-6 (red)/E-cadherin (green) in colonic cryosections obtained from C57BL/6J. Discontinuous white line surrounds epithelial cells located at the crypt (L, luminal; I, interstitium). White arrow indicates IL-6–positive nonepithelial cells, and white asterisk marks IECs with cytosolic IL-6. Nuclei, blue. Scale bar, 20 μm (n = 6). (B) Phospho-STAT1 (Y701), phospho-STAT3 (S705), STAT1, and STAT3 were analyzed in cell lysates of colonic mucosa and colonic isolated crypts obtained from C57BL/6J mice injected i.p. with MSA, IL-6 (10 μg/kg), or IFN-γ (2.5 μg/kg). Mice were euthanized 2 h postinjection. GAPDH was used as control (n = 6). (C and D) Immunolocalization for the p-STAT3 (red)/E-cadherin (green) and F4/80 (red)/p-STAT3 (green) in colonic cryosections obtained from C57BL/6J injected i.p. with MSA, IL-6, or IFN-γ. Discontinuous white line surrounds epithelial cells located at the crypt. White asterisk indicates nonepithelial cells that are positive for active STAT3. White arrow indicates IECs that are positive for nuclear p-STAT3. Nuclei, blue. Scale bar, 20 μm. Original magnification ×40, zoom ×100. Quantification of colonocytes and p-STAT3–positive F4/80 cells per crypt is shown in the graphs (n = 7). ***p < 0.001, **p < 0.01 versus control (ANOVA two-way test).

FIGURE 2.

The i.p. administration of IL-6 does not activate STAT3 in IECs present at the colonic mucosa. (A) Immunolocalization of IL-6 (red)/E-cadherin (green) in colonic cryosections obtained from C57BL/6J. Discontinuous white line surrounds epithelial cells located at the crypt (L, luminal; I, interstitium). White arrow indicates IL-6–positive nonepithelial cells, and white asterisk marks IECs with cytosolic IL-6. Nuclei, blue. Scale bar, 20 μm (n = 6). (B) Phospho-STAT1 (Y701), phospho-STAT3 (S705), STAT1, and STAT3 were analyzed in cell lysates of colonic mucosa and colonic isolated crypts obtained from C57BL/6J mice injected i.p. with MSA, IL-6 (10 μg/kg), or IFN-γ (2.5 μg/kg). Mice were euthanized 2 h postinjection. GAPDH was used as control (n = 6). (C and D) Immunolocalization for the p-STAT3 (red)/E-cadherin (green) and F4/80 (red)/p-STAT3 (green) in colonic cryosections obtained from C57BL/6J injected i.p. with MSA, IL-6, or IFN-γ. Discontinuous white line surrounds epithelial cells located at the crypt. White asterisk indicates nonepithelial cells that are positive for active STAT3. White arrow indicates IECs that are positive for nuclear p-STAT3. Nuclei, blue. Scale bar, 20 μm. Original magnification ×40, zoom ×100. Quantification of colonocytes and p-STAT3–positive F4/80 cells per crypt is shown in the graphs (n = 7). ***p < 0.001, **p < 0.01 versus control (ANOVA two-way test).

Close modal

Next, we sought to identify the alternative mechanism involved in the activation of STAT3 in IECs. In fibroblasts, STAT3 activation is mediated by IFN-γ (21), a proinflammatory cytokine increased at the basolateral side of IECs during colitis (4, 22, 23). Thus, we analyzed p-STAT3 levels in colonic cell lysates and colonic isolated crypts of mice administered i.p. with IFN-γ for 2 h. Similar to the findings with IL-6, basolateral administration of IFN-γ strongly increased p-STAT3 in the colonic mucosa of C57BL/6J animals, and, as expected, IFN-γ also induced STAT1 activation. No changes in total STAT1 or total STAT3 were perceived (Fig. 2B). Interestingly, we observed STAT3 activation after basolateral stimulation with IFN-γ and nuclear accumulation of p-STAT3 in both epithelial (Fig. 2B, 2C, arrow) and nonepithelial cells (Fig. 2D, Supplemental Fig. 1A, asterisk). p-STAT3–positive IECs were scattered along the whole crypt axis; however, they were clearly enriched at the apical surface of the crypts (Fig. 2C, 2D, white arrows). Furthermore, p-STAT3–positive nonepithelial cells that were induced after basolateral administration of IFN-γ were positive for F4/80 and were observed in close apposition to the IECs, as shown in Fig. 2D and Supplemental Fig. 1A (white asterisk). Contrary to p-STAT3, a more uniform distribution of p-STAT1 in IECs along the crypt axis was induced by IFN-γ (Supplemental Fig. 1B). Furthermore, a more detailed analysis for STAT1 and STAT3 activation revealed that IFN-γ induced a fast activation of STAT1 in IECs, such as the process started at the crypt surface. However, over time, nuclear staining of p-STAT3 was detected in the same cells following a similar pattern of activation as the one displayed by p-STAT1 (Supplemental Fig. 1C). Thus, taken together, these results strongly suggest that IFN-γ actively contributes to the activation of STAT3 in differentiated IECs.

To understand the mechanism by which IFN-γ activates STAT3 in IECs, we stimulated SW480 with IFN-γ for 0.5–6 h. As shown in Fig. 3A, STAT3 phosphorylation was strongly induced starting 30 min after cytokine treatment, but its presence declined rapidly. Furthermore, p-STAT3 was not detected in cell lysates of SW480 cells treated with IFN-γ for 24 h, but it was present in cells exposed to IL-6 or the mixture IL-6/IFN-γ for the same period of time (Supplemental Fig. 2A). Conversely, IFN-γ presence strongly induced phosphorylation of STAT1 for up to 24 h even in the presence of IL-6 (Fig. 3A, Supplemental Fig. 2A). No changes in STAT1 or STAT3 were observed during the course of the experiment. Thus, these results suggest that IFN-γ transiently activates STAT3 in IECs, but in contrast, STAT3 activation by IL-6 or IL-6/IFN-γ results in a long-term effect.

FIGURE 3.

IFN-γ enhances IL-6 secretion in IEC during colitis. (A) p-STAT1, p-STAT3, STAT1, and STAT3 were analyzed in cell lysates of SW480 cells treated with IFN-γ. Cells plated in six-well plates were collected at 0.5, 1, 2, 4, and 6 h after cytokine stimulation. GAPDH was used as loading control (n = 5). (B) p-STAT3 and STAT3 were analyzed in cell lysates of SW480 cells treated with IFN-γ. SW480 cells were plated in six-well plates and incubated with cytokines for 30 min, and then media was removed and replaced with fresh media. Cells were collected at 0.5, 1, 2, and 4 h after new media was added. GAPDH was used as loading control (n = 3). (C) p-STAT3 and STAT3 were analyzed in cell lysates of SW480 cells stimulated with conditioned media obtained from SW480 cells stimulated with IFN-γ in (B). GAPDH was used as loading control (n = 3). (D) p-STAT1, p-STAT3, STAT1, and STAT3 were analyzed in cell lysates of SW480 cells treated with IL-6 or IFN-γ. Secretion of epithelial cells was blocked using Bafilomycin A. BfA was administered 30 min prior to cytokine stimulation. GAPDH was used as loading control (n = 3). (E) IL-6 secretion was analyzed in SW480 cells treated with IFN-γ or IL-6 in presence or absence of BfA. BfA was administered 30 min prior to cytokine stimulation. Cytokine stimulation was carried out for 2 h (n = 6). Data in graph are represented as mean ± SEM. ***p < 0.001 (ANOVA two-way test). (F) Stimulation of IL-6 secretion by IL-6 and IFN-γ was investigated in colonic isolated crypts of mice. Cytokine stimulation was carried out for 1 h (n = 5). Data in graph are represented as mean ± SEM. ****p < 0.0001, **p < 0.05 (ANOVA two-way test). (G) Schematic model and analysis by Western blot of p-STAT3 and STAT3 in cell lysates of Caco-2 cells treated basolaterally with IFN-γ and apically with anti–IL-6 Ab or isotype control. GAPDH was used as loading control (n = 3).

FIGURE 3.

IFN-γ enhances IL-6 secretion in IEC during colitis. (A) p-STAT1, p-STAT3, STAT1, and STAT3 were analyzed in cell lysates of SW480 cells treated with IFN-γ. Cells plated in six-well plates were collected at 0.5, 1, 2, 4, and 6 h after cytokine stimulation. GAPDH was used as loading control (n = 5). (B) p-STAT3 and STAT3 were analyzed in cell lysates of SW480 cells treated with IFN-γ. SW480 cells were plated in six-well plates and incubated with cytokines for 30 min, and then media was removed and replaced with fresh media. Cells were collected at 0.5, 1, 2, and 4 h after new media was added. GAPDH was used as loading control (n = 3). (C) p-STAT3 and STAT3 were analyzed in cell lysates of SW480 cells stimulated with conditioned media obtained from SW480 cells stimulated with IFN-γ in (B). GAPDH was used as loading control (n = 3). (D) p-STAT1, p-STAT3, STAT1, and STAT3 were analyzed in cell lysates of SW480 cells treated with IL-6 or IFN-γ. Secretion of epithelial cells was blocked using Bafilomycin A. BfA was administered 30 min prior to cytokine stimulation. GAPDH was used as loading control (n = 3). (E) IL-6 secretion was analyzed in SW480 cells treated with IFN-γ or IL-6 in presence or absence of BfA. BfA was administered 30 min prior to cytokine stimulation. Cytokine stimulation was carried out for 2 h (n = 6). Data in graph are represented as mean ± SEM. ***p < 0.001 (ANOVA two-way test). (F) Stimulation of IL-6 secretion by IL-6 and IFN-γ was investigated in colonic isolated crypts of mice. Cytokine stimulation was carried out for 1 h (n = 5). Data in graph are represented as mean ± SEM. ****p < 0.0001, **p < 0.05 (ANOVA two-way test). (G) Schematic model and analysis by Western blot of p-STAT3 and STAT3 in cell lysates of Caco-2 cells treated basolaterally with IFN-γ and apically with anti–IL-6 Ab or isotype control. GAPDH was used as loading control (n = 3).

Close modal

Indirect activation of STAT3 signaling by secreted factors could be triggered in an autocrine or paracrine way (24). Thus, we investigated whether STAT3 activation downstream of IFN-γR was mediated by a secreted factor. We therefore analyzed the phosphorylation of STAT3 as a function of time in SW480 cells exposed to IFN-γ for 30 min, followed by washout (Fig. 3B, Supplemental Fig. 2B). Phosphorylation of STAT3 after IFN-γ removal lasted for several hours (Fig. 3B), and the conditioned medium obtained from IFN-γ–treated cells induced STAT3 phosphorylation in freshly plated SW480 cells (Fig. 3C). No changes for STAT3 were detected during the course of the experiment (Fig. 3B, 3C). Following a similar protocol, no changes in p-STAT3, p-STAT1, STAT1, or STAT3 were detected in SW480 stimulated with BSA (carrier) alone (data not shown). Next, we analyzed STAT3 activation in IEC pretreated with brefeldin A (BfA), a potent transport inhibitor used to prevent cytokine release (25). As shown in Fig. 3D, the activation of STAT3 induced by IFN-γ was strongly reduced in the presence of BfA, but no changes in p-STAT1 were noticed. In agreement with previous published results, STAT3 activation induced by IL-6 was also suppressed by BfA. These results suggest that in IECs, long-term activation of STAT3 by proinflammatory cytokines requires the activation of the secretory pathway.

Because of our results in Fig. 1, we next investigated whether IFN-γ induces the secretion of IL-6 in IECs. As shown in Fig. 3E, IL-6 was enriched in the supernatant of nonpolarized SW480 cells exposed to IFN-γ. Furthermore, as reported previously, IL-6 treatment induced a strong release of IL-6 by IECs. No synergistic effect was noticed when SW480 cells were treated with both cytokine mixtures, IL-6/IFN-γ (data not show). Furthermore, treating SW480 cells with BfA before IFN-γ or IL-6 stimulation prevented the release of IL-6 into the media (Fig. 3E). Also, IFN-γ and IL-6 treatment induced IL-6 secretion in freshly isolated colonic crypts obtained from C57BL/6J mice (Fig. 3F). Interestingly, IFN-γ induced IL-6 release in several cell lines, including IECs, fibroblasts, and macrophages (Fig. 3E, 3F, Supplemental Fig. 2C, 2D). However, the amount of IL-6 secreted by those cells after IFN-γ stimulation was considerably lower than the one induced by IL-6 (fibroblasts) or LPS (macrophages). In fibroblasts, IL-6 release induced by TGF-β treatment was observed at similar extent as the one induced by IFN-γ, corroborating the viability of our assay (26). Interestingly, IFN-γ treatment induced only a transient secretion of IL-6 in IECs, suggesting that it does not affect IL-6 expression (Supplemental Fig. 2E). To analyze if IFN-γ was inducing STAT3 activation via autocrine stimulation of IL-6/STAT3 signaling, we evaluated p-STAT3 in SW480 and Caco-2 cells treated with IFN-γ in the presence of neutralizing Abs against IL-6 signaling. As expected, anti–IL-6R or anti–IL-6 Ab strongly reduced the activation of STAT3 IFN-γ−mediated in comparison with cells treated with the cytokine plus an isotype control (Fig. 3G, Supplemental Fig. 2F). Thus, taken together, these results strongly suggest that in IECs, IFN-γ induces STAT3 activation by stimulating IL-6 secretion.

Next, we evaluated the secretion of IL-6 by the IECs lining the colonic mucosa of colitic mice. Thus, we investigated the presence of IL-6 in the mucosa of C57BL/6J animals in basal conditions. As shown by immunofluorescence studies, IL-6 staining was detected in epithelial cells and nonepithelial cells at the colonic mucosa (Fig. 2A). IL-6–positive IECs were observed along the crypt axis, including the surface (arrows). Nonepithelial, IL-6–positive cells expressed the macrophage marker F4/80 and were detected surrounding the crypt base (Fig. 4A, white asterisk; Fig. 4B). However, increased staining for IL-6 was clearly noticed in epithelial cells present at the mucosa of colitic mice when compared with their nontreated counterparts (Fig. 4A, arrows). Furthermore, the number of double-positive cells expressing F4/80 and IL-6 was also augmented (Fig. 4B). It is noteworthy to mention that patchy areas enriched with double-positive cells (F4/80/IL-6) were frequently observed in the inflamed mucosa of colitic mice. In agreement with our findings, the induction in IL-6 secretion was enhanced in both mucosal explants and isolated crypts obtained from colitic mice (Fig. 4C). However, as expected, IL-6 secretion was more pronounced in the colonic explants when compared with isolated crypts. Thus, all these results strongly suggest that IEC present in the colon constantly produce and secrete IL-6, and this process is enhanced during colitis.

FIGURE 4.

IL-6 is produced and secreted by IECs during colitis. (A) Immunolocalization of IL-6 (green) in the colonic mucosa of control (Ctl) and colitic mice. C57BL/6J mice were treated with 2.5% DSS for 5 d. Discontinuous white line surrounds epithelial cells in the crypt. White asterisk indicates nonepithelial cells producing IL-6. White arrow indicates IECs that are positive for nuclear IL-6. Nuclei, blue. Scale bar, 20 μm (n = 5). (B) Immunolocalization of IL-6 (green) and F4/80 (red) in the colonic mucosa of Ctl and colitic mice. C57BL/6J mice were treated with 2.5% DSS for 5 d. White arrow indicates double-positive cells. Nuclei, blue. Scale bar, 10 μm (n = 3). (C) IL-6 secretion was analyzed by ELISA in isolated crypts and colonic explants obtained from control or colitic C57BL/6J mice. Mice were treated with 2.5% DSS for 5 d (n = 4). Data in graph are represented as mean ± SEM. ***p < 0.001, **p < 0.05 versus control (Student t test).

FIGURE 4.

IL-6 is produced and secreted by IECs during colitis. (A) Immunolocalization of IL-6 (green) in the colonic mucosa of control (Ctl) and colitic mice. C57BL/6J mice were treated with 2.5% DSS for 5 d. Discontinuous white line surrounds epithelial cells in the crypt. White asterisk indicates nonepithelial cells producing IL-6. White arrow indicates IECs that are positive for nuclear IL-6. Nuclei, blue. Scale bar, 20 μm (n = 5). (B) Immunolocalization of IL-6 (green) and F4/80 (red) in the colonic mucosa of Ctl and colitic mice. C57BL/6J mice were treated with 2.5% DSS for 5 d. White arrow indicates double-positive cells. Nuclei, blue. Scale bar, 10 μm (n = 3). (C) IL-6 secretion was analyzed by ELISA in isolated crypts and colonic explants obtained from control or colitic C57BL/6J mice. Mice were treated with 2.5% DSS for 5 d (n = 4). Data in graph are represented as mean ± SEM. ***p < 0.001, **p < 0.05 versus control (Student t test).

Close modal

IECs elicit a complex response to cytokines depending on several factors such as cytokine concentration, receptor availability, and stage of differentiation (2730). Therefore, we examined STAT3 activation and IL-6 secretion in fully differentiated Caco-2 cells that were exposed in a polarized way to proinflammatory stimuli. To this end, Caco-2 cells were plated in Transwell filters, and the stimulus was added at the apical or basolateral side. As shown in Fig. 5A, no activation of STAT3 was observed in Caco-2 cells stimulated apically with IL-6 or IFN-γ. However, as expected, STAT3 activation was induced once IL-6 or IFN-γ were administered basolaterally. No synergistic effect in the activation of STAT3 was obtained in monolayers stimulated with IL-6 and IFN-γ at the basolateral side. Importantly, TGF-β and LPS did not affect STAT3 phosphorylation status when administered apically or basolaterally. No significant changes in STAT3 were noticed with any of the treatments. Surprisingly, IL-6 was detected at the apical compartment of Caco-2 monolayers that were treated with IFN-γ or IL-6 basolaterally, and the administration of the mixture IL-6/IFN-γ potentiated such effect (Fig. 5B), but this process was not observed in Caco-2 cells stimulated with IFN-γ at the apical side. Also, we detected a strong release of IL-6 at the basolateral after stimulating Caco-2 cells with IFN-γ at the basolateral side. The basolateral administration of TGF-β or LPS has no influence in the presence of IL-6 either at the apical or basolateral side. However, the apical stimulation of Caco-2 cells with IL-6 triggered the basolateral release of IL-6, an effect that was not observed in monolayers stimulated with IFN-γ at the apical side. Thus, these results strongly suggest that activation of STAT3 in IECs stimulated with IFN-γ is mediated by the endogenous IL-6 that is secreted at the apical side.

FIGURE 5.

IFN-γ induces polarization and compartmentalization of IL-6/STAT3 signal in IECs. (A) p-STAT3 and STAT3 were analyzed in cell lysates of Caco-2 cells stimulated with IL-6, IFN-γ, LPS, TGF-β, and IFN-γ/IL-6. Cells were stimulated for 2 h apically (Api) or basolaterally (BL). GAPDH was used as loading control (n = 4). (B) IL-6 secreted after BL or Api stimulation was analyzed by ELISA in supernatant obtained from Caco-2 cells stimulated with IL-6, IFN-γ, LPS, TGF-β, and IFN-γ/IL-6. Cells were stimulated for 2 h Api or BL. Supernatant was collected at the Api compartment of the Transwell (n = 3). Data in graph are represented as mean ± SEM. ***p < 0.001, **p < 0.05 versus control (ANOVA two-way test). (C) IL-6Rα and gp130 were analyzed in cell lysates of Caco-2 cells stimulated with IFN-γ for 0, 3, 6, 9, and 12 h. GAPDH was used as loading control. Densitometric analysis for IL-6Rα is presented (n = 5). Data in graph are represented as mean ± SEM. *p < 0.05, **p < 0.01 versus control (ANOVA two-way test). (D) Immunolocalization of IL-6Rα (red) and E-cadherin (green) in Caco-2 cells stimulated BL with IFN-γ. The orientation of microscopic cross-sections is indicated on the right. Scale bar, 10 μm (xz); 30 μm (xy) (n = 3). (E) Immunolocalization of IL-6R (red) and β-catenin (green) was carried out in cryosections of colonic mucosa of control and colitic mice. Discontinuous white line surrounds epithelial cells located at the crypt. White arrows mark positive staining for murine IL-6R (mIL-6R); nuclei, blue. Scale bar, 10 μm (n = 3). (F) p-STAT3 and STAT3 were analyzed in colonic cell lysates obtained from C57BL/6J mice stimulated with IL-6. Intraluminal administration of IL-6 was carried out for 30 min. GAPDH was used as loading control (n = 3). (G) Immunolocalization of p-STAT3 (green) in the colonic mucosa of C57BL/6J mice stimulated intraluminally with IL-6 or IFN-γ for 30 min. Discontinuous white line surrounds epithelial cells located at the crypt. White arrows mark IECs that are positive for STAT. E-cadherin, red; nuclei, blue. Scale bar, 20 μm (n = 3).

FIGURE 5.

IFN-γ induces polarization and compartmentalization of IL-6/STAT3 signal in IECs. (A) p-STAT3 and STAT3 were analyzed in cell lysates of Caco-2 cells stimulated with IL-6, IFN-γ, LPS, TGF-β, and IFN-γ/IL-6. Cells were stimulated for 2 h apically (Api) or basolaterally (BL). GAPDH was used as loading control (n = 4). (B) IL-6 secreted after BL or Api stimulation was analyzed by ELISA in supernatant obtained from Caco-2 cells stimulated with IL-6, IFN-γ, LPS, TGF-β, and IFN-γ/IL-6. Cells were stimulated for 2 h Api or BL. Supernatant was collected at the Api compartment of the Transwell (n = 3). Data in graph are represented as mean ± SEM. ***p < 0.001, **p < 0.05 versus control (ANOVA two-way test). (C) IL-6Rα and gp130 were analyzed in cell lysates of Caco-2 cells stimulated with IFN-γ for 0, 3, 6, 9, and 12 h. GAPDH was used as loading control. Densitometric analysis for IL-6Rα is presented (n = 5). Data in graph are represented as mean ± SEM. *p < 0.05, **p < 0.01 versus control (ANOVA two-way test). (D) Immunolocalization of IL-6Rα (red) and E-cadherin (green) in Caco-2 cells stimulated BL with IFN-γ. The orientation of microscopic cross-sections is indicated on the right. Scale bar, 10 μm (xz); 30 μm (xy) (n = 3). (E) Immunolocalization of IL-6R (red) and β-catenin (green) was carried out in cryosections of colonic mucosa of control and colitic mice. Discontinuous white line surrounds epithelial cells located at the crypt. White arrows mark positive staining for murine IL-6R (mIL-6R); nuclei, blue. Scale bar, 10 μm (n = 3). (F) p-STAT3 and STAT3 were analyzed in colonic cell lysates obtained from C57BL/6J mice stimulated with IL-6. Intraluminal administration of IL-6 was carried out for 30 min. GAPDH was used as loading control (n = 3). (G) Immunolocalization of p-STAT3 (green) in the colonic mucosa of C57BL/6J mice stimulated intraluminally with IL-6 or IFN-γ for 30 min. Discontinuous white line surrounds epithelial cells located at the crypt. White arrows mark IECs that are positive for STAT. E-cadherin, red; nuclei, blue. Scale bar, 20 μm (n = 3).

Close modal

IL-6R and IFN-γR are located at the basolateral side of epithelial cells (31, 32); however, STAT3 activation in Caco-2 cells stimulated with cytokines was only observed in monolayers secreting IL-6 at the apical side, suggesting that other components were involved in the process. Thus, we speculated that IL-6R expression and location in IECs could also be affected by IFN-γ, similar to the observed with IL-10R (33). Therefore, we next evaluated the presence of IL-6Rα and the accessory coreceptor, the gp130, in IECs exposed to IFN-γ. Interestingly, IFN-γ treatment rapidly increased IL-6Rα and gp130 protein levels in Caco-2 cells (Fig. 5C). Furthermore, immunofluorescence staining demonstrated that IL-6Rα was also relocated to the apical surface in Caco-2 cells treated with the cytokine (Fig. 5D). Next, we evaluated the distribution of IL-6Rα in the colonic mucosa of C57BL/6J mice. IL-6Rα was highly enriched in nonepithelial cells surrounding the colonic crypt in both control and DSS-treated animals (Supplemental Fig. 2G). Also, as reported previously, positive staining for IL-6Rα at the basolateral side of IECs located at the crypt base was observed [Supplemental Fig. 2H and (31)]. Those cells were in close contact with nonepithelial, IL-6Rα–positive cells (white asterisk). However, and unexpectedly, we noticed the presence of IL-6Rα at the apical cell surface of IECs located in the bottom of the crypt (Supplemental Fig. 2H, arrowheads). The apical staining for IL-6Rα was more noticeable in IECs present at the surface of the crypt, and the presence of the receptor in those cells was strongly enhanced after colitis induction (Fig. 5E). To investigate the functionality of the IL-6Rα detected at the apical membrane in IECs of the colonic mucosa, we next administered C57BL/6J mice intraluminally with carrier alone (MSA), IL-6, or IFN-γ for 30 min, and STAT3 and STAT1 activation was evaluated by Western blot (Fig. 5F). As expected, activation of STAT3 was only observed in the animals that received IL-6, but no changes were detected after IFN-γ or MSA administration. Immunofluorescence staining demonstrated activation of STAT3 in epithelial cells located at the apical surface of the colonic crypt (Fig. 5G). Thus, we conclude that during inflammation, IFN-γ regulates the activation of STAT3 in IECs by polarizing both the receptor and the secretion of the cytokine.

Apical administration of IL-6 failed to induce STAT3 activation in Caco-2 cells but evoked a strong phosphorylation of the molecule in colonocytes in vivo (Fig. 5A, 5F). Thus, the discrepancy between our in vitro and in vivo results prompted us to speculate that basal release of IFN-γ induced by colonic microbiota (34) was essential for IL-6Rα polarization. Therefore, IL-6/STAT3 signaling in the mucosa of Crtam-deficient mice was analyzed. As shown in Supplemental Fig. 3A, in the absence of Crtam, the production of IFN-γ induced by the resident microbiota is greatly reduced because of functional impairment of CD4+ T cells (35). However, no changes in IL-23, an unrelated cytokine, were noticed. Low levels of IFN-γ were likely a consequence in the reduction of colonic CD4+ T cells because no differences in the number of IFN-γ–producing macrophages were detected between Crtam knockout (KO) and WT animals (Supplemental Fig. 3B). Similar to that observed in WT animals, i.p. administration of IFN-γ induced activation of STAT1 and STAT3 in the colonic mucosa of Crtam KO mice without affecting STAT1 or STAT3 levels (Fig. 6A). However, STAT3 activation was greatly reduced in Crtam-deficient mice when compared with WT animals (Supplemental Fig. 3C). Activation of STAT1 arose in IECs along the whole-crypt axis (Fig. 6B, white arrows) and in nonepithelial cells present in the mucosa (Fig. 6B, white asterisk). However, p-STAT3 staining was noticed preferentially in nonepithelial cells (Fig. 6C, white asterisk) and was almost absent in epithelial cells. Nonetheless, some epithelial cells at the bottom of the crypt displayed nuclear staining for p-STAT3 (white arrows). Furthermore, the presence of IL-6Rα at the luminal side of the enterocytes was greatly reduced in the animals lacking Crtam (Fig. 6D). Interestingly, despite in the differential response observed in STAT3 activation, no morphological changes were noticed in the colonic mucosa of Crtam-deficient mice when compared with WT animals (Supplemental Fig. 3D). However, the amount of Claudin-1, an epithelial protein induced by STAT3 (36) to strength barrier function in the gut, was strongly decreased in the colon in the absence of Crtam (Fig. 6E). Furthermore, Claudin-1 and -2 were present in the cellular junctions in IECs. However, the distribution of both molecules along the crypt axis displayed a slightly different pattern in the mucosa of Crtam-deficient mice when compared with WT animals (Fig. 6F, Supplemental Fig. 3E). In addition, mucin-2, another molecule that is regulated by STAT3 in epithelial cells, was strongly reduced in the absence of Crtam (Fig. 6F), and the protein was mostly accumulated in the cytosol of goblet cells (Supplemental Fig. 3F). The i.p. administration of Crtam KO mice with IFN-γ for 72 h partially rescued those phenotypes (data not shown).

FIGURE 6.

IL-6/STAT3 signaling is not fully polarized in the mucosa of Crtam-deficient mice. (A) p-STAT3, STAT3, p-STAT1, and STAT1 were analyzed in colonic cell lysates obtained from Crtam-deficient mice stimulated with MSA or IFN-γ. The i.p. administration of MSA and IFN-γ was carried out for 2 h (n = 3). (B) Immunolocalization of p-STAT1 (red) in the colonic mucosa of C57BL/6J mice and Crtam KO mice stimulated i.p. with IFN-γ for 2 h. Discontinuous white line surrounds epithelial cells located at the crypt. White star denotes nonepithelial cells that are positive for active STAT1. White arrows mark IEC-positive cells that are positive for STAT. E-cadherin, green; nuclei, blue. Scale bar, 10 μm (n = 3). (C) Immunolocalization of p-STAT3 (red) in the colonic mucosa of C57BL/6J mice and Crtam KO mice stimulated i.p. with IFN-γ for 2 h. Discontinuous white line surrounds epithelial cells located at the crypt. White asterisk denotes nonepithelial cells that are positive for active STAT3. White arrows mark IECs positive for p-STAT3. E-cadherin, green; nuclei, blue. Scale bar, 20 μm (n = 3). (D) Immunolocalization of IL-6R (green) and E-cadherin (red) was carried out in cryosections of colonic mucosa of WT and Crtam KO mice. Discontinuous white line surrounds epithelial cells located at the crypt. Nuclei, blue. Scale bar, 50 μm (n = 3). (E) Claudin-1 was analyzed in colonic lysates obtained from WT and Crtam-deficient mice. GAPDH was used as loading control (n = 3). (F) Immunolocalization of claudin-1 and mucin-2 (red) in the colonic mucosa of C57BL/6J and Crtam-deficient mice. E-cadherin, green; nuclei, blue. Discontinuous white line surrounds epithelial cells located at the crypt. Scale bar, 50 μm (n = 3).

FIGURE 6.

IL-6/STAT3 signaling is not fully polarized in the mucosa of Crtam-deficient mice. (A) p-STAT3, STAT3, p-STAT1, and STAT1 were analyzed in colonic cell lysates obtained from Crtam-deficient mice stimulated with MSA or IFN-γ. The i.p. administration of MSA and IFN-γ was carried out for 2 h (n = 3). (B) Immunolocalization of p-STAT1 (red) in the colonic mucosa of C57BL/6J mice and Crtam KO mice stimulated i.p. with IFN-γ for 2 h. Discontinuous white line surrounds epithelial cells located at the crypt. White star denotes nonepithelial cells that are positive for active STAT1. White arrows mark IEC-positive cells that are positive for STAT. E-cadherin, green; nuclei, blue. Scale bar, 10 μm (n = 3). (C) Immunolocalization of p-STAT3 (red) in the colonic mucosa of C57BL/6J mice and Crtam KO mice stimulated i.p. with IFN-γ for 2 h. Discontinuous white line surrounds epithelial cells located at the crypt. White asterisk denotes nonepithelial cells that are positive for active STAT3. White arrows mark IECs positive for p-STAT3. E-cadherin, green; nuclei, blue. Scale bar, 20 μm (n = 3). (D) Immunolocalization of IL-6R (green) and E-cadherin (red) was carried out in cryosections of colonic mucosa of WT and Crtam KO mice. Discontinuous white line surrounds epithelial cells located at the crypt. Nuclei, blue. Scale bar, 50 μm (n = 3). (E) Claudin-1 was analyzed in colonic lysates obtained from WT and Crtam-deficient mice. GAPDH was used as loading control (n = 3). (F) Immunolocalization of claudin-1 and mucin-2 (red) in the colonic mucosa of C57BL/6J and Crtam-deficient mice. E-cadherin, green; nuclei, blue. Discontinuous white line surrounds epithelial cells located at the crypt. Scale bar, 50 μm (n = 3).

Close modal

Given our results, we then analyzed the role of STAT3 activation in the mucosa of Crtam-deficient mice that were induced to colitis. Disease Activity Index analysis showed a similar evolution in the development of the disease in Crtam KO and WT mice (Fig. 7A). Also, analysis of weight loss and colon length in colitic WT and Crtam-deficient mice reported similar changes in both strains (Supplemental Fig. 4A, 4B). Furthermore, we also noticed that in Crtam-deficient mice, the activation and response of STAT3 in the colonic mucosa of colitic mice followed a similar pattern to the one observed in WT animals (Fig. 7B, Supplemental Fig. 4C). However, the secretion of IL-6 was greatly reduced in the mucosa of Crtam-deficient mice (Fig. 7C), and p-STAT3 staining was observed mainly at the nuclei of nonepithelial cells (Fig. 7D, asterisk). Furthermore, analysis of colonic isolated crypts showed a decrease in p-STAT3 in Crtam-deficient mice when compared with WT animals (Fig. 7E). Microscopic inspection of the colonic mucosa of colitic mice demonstrated less infiltration in the colon of Crtam KO. However, it was noticeable that in the absence of Crtam, a more widespread damage occurs in the epithelium (Fig. 7F, arrow). Consequently, histopathological evaluation of colonic samples according to the parameters established by Rayudu et al., demonstrated no differences between both strains (Supplemental Fig. 4D). Thus, these results suggest that IL-6/STAT3 signaling is altered in the mucosa of Crtam-deficient mice.

FIGURE 7.

Crtam confers resistance to colitis development. (A) Disease Activity Index changes among WT and Crtam-deficient mice after 5 d of DSS treatment. Results are expressed as mean ± SE (n = 3). (B) phospho-STAT3 and STAT3 were analyzed by Western blotting colonic cell lysates of C57BL/6J and Crtam-deficient mice treated with DSS for 1–5 d. A total of 2.5% DSS was dissolved in tap water. GAPDH was used as loading control (n = 3). (C) IL-6 secretion was analyzed in colonic explants obtained from C57BL/6J mice and Crtam KO mice that were treated with 2.5% DSS for 1–4 d (n = 3). Data in graph are represented as mean ± SEM. **p < 0.01 (ANOVA two-way test). (D) Immunolocalization of p-STAT3 (green) and E-cadherin (red) in the colonic mucosa of C57BL/6J mice and Crtam KO mice after 4 d of DSS treatment. Discontinuous white line surrounds epithelial cells located at the crypt. E-cadherin, red; nuclei, blue. Scale bar, 50 μm (left); 30 μm (right) (n = 4). (E) p-STAT3 was analyzed in colonic isolated crypts obtained from C57BL/6J and Crtam KO mice treated with DSS for 4 d. GAPDH was used as loading control. Densitometric analysis of p-STAT3 is presented (n = 3). Data in graph are represented as mean ± SEM. ***p < 0.001 versus control (ANOVA two-way test). (F) H&E staining from colon C57BL/6J and Crtam-deficient mice after 4 d of DSS treatment (n = 3). Original magnification ×1; zoom ×2 and ×10.

FIGURE 7.

Crtam confers resistance to colitis development. (A) Disease Activity Index changes among WT and Crtam-deficient mice after 5 d of DSS treatment. Results are expressed as mean ± SE (n = 3). (B) phospho-STAT3 and STAT3 were analyzed by Western blotting colonic cell lysates of C57BL/6J and Crtam-deficient mice treated with DSS for 1–5 d. A total of 2.5% DSS was dissolved in tap water. GAPDH was used as loading control (n = 3). (C) IL-6 secretion was analyzed in colonic explants obtained from C57BL/6J mice and Crtam KO mice that were treated with 2.5% DSS for 1–4 d (n = 3). Data in graph are represented as mean ± SEM. **p < 0.01 (ANOVA two-way test). (D) Immunolocalization of p-STAT3 (green) and E-cadherin (red) in the colonic mucosa of C57BL/6J mice and Crtam KO mice after 4 d of DSS treatment. Discontinuous white line surrounds epithelial cells located at the crypt. E-cadherin, red; nuclei, blue. Scale bar, 50 μm (left); 30 μm (right) (n = 4). (E) p-STAT3 was analyzed in colonic isolated crypts obtained from C57BL/6J and Crtam KO mice treated with DSS for 4 d. GAPDH was used as loading control. Densitometric analysis of p-STAT3 is presented (n = 3). Data in graph are represented as mean ± SEM. ***p < 0.001 versus control (ANOVA two-way test). (F) H&E staining from colon C57BL/6J and Crtam-deficient mice after 4 d of DSS treatment (n = 3). Original magnification ×1; zoom ×2 and ×10.

Close modal

IBD are chronic pathologies of unknown etiology (37). Our work in the last years has unveiled an essential role of the intestinal epithelial barrier in the development and establishment of IBD. However, the apparatus by which IECs contribute to IBD progress is not fully understood. In this study, we analyzed the mechanism triggering STAT3 signaling in the colonic mucosa during inflammation, and we demonstrated the importance of IECs polarization and barrier function in this process. Our findings uncovered an unknown role for IFN-γ in the activation of STAT3 in differentiated IECs located at the apical region of the crypts during colitis. Also, these results highlight an active role of the epithelial barrier in the compartmentalization of IL-6/STAT3 signaling. Finally, we demonstrated that impaired production of IFN-γ by CD4 T cells largely attenuated IL-6/STAT3 signaling in IECs, but not in immune cells.

Understanding the pathophysiology of IBD has been a challenge that limited the development of more efficient therapies. The use of IBD-like models has been broadly implemented as an approach to investigate the role of factors known to directly influence the outcome of the disease. However, contradictory results have been obtained using similar models (e.g., DSS) and specific factors (e.g., IL-6), mainly because we have obviated the complexity of the tissue (several cell types with specific intrinsic characteristics are affected) and the broad variety of mediators that are present in the inflamed mucosa (cytokines, growth factors, chemokines, etc). One essential component in the maintenance of the colonic mucosa that has not been fully considered in this process is the polarity of the epithelium. The epithelial barrier in the colonic mucosa is formed by a single layer of fully polarized epithelial cells with two well-defined domains, apical and basolateral. Functionally and structurally, both domains are different, and the role of such polarization in the development of IBD is not totally understood. By using a model of colitis based in epithelial injury (DSS-mediated colitis), we now demonstrate that in addition to the epithelial barrier function, the polarization of IEC is important for the development of IBD-like symptoms. Our results highlight the role of the exclusion of the cytokine receptors at apical and basolateral domains in the compartmentalization of the signaling pathways that activated in the colonic mucosa during inflammation. Such process could play an important role for the maintenance of the intestinal epithelial barrier and for the restraining of the inflammatory responses such as those described for TLR (38). Therefore, these findings could be important to explain why the same signaling (STAT3) displays protective or detrimental effects in the colonic mucosa during colitis (31, 39). In fact, according to our results, the mechanisms underlying the activation of STAT3 in the colonic mucosa imply the establishment of feedback loops between apical and basolateral receptors as the observed in the tolerizing response mediated by TLR (38).

The biological relevance of the polarized response triggered by IFN-γ in IECs could be envisioned as part of two different scenarios (Supplemental Fig. 4E). First, in the context of an intact epithelial barrier, basolateral secretion of IFN-γ at low levels will drive the apical enrichment of IL-6Rα in IECs. If a challenge induces the release of high levels of IFN-γ, mimicked in this study by the i.p. injection of the cytokine, apical secretion of IL-6 will stimulate the activation of STAT3 in crypt surface epithelial cells to strength epithelial barrier functions (36, 40). Lack of activation of STAT3 in IECs after i.p. administration of IL-6 strongly supports this theory. The context in which high amounts of IFN-γ are released in a mucosa with an intact epithelial barrier needs to be investigated but could be ranging from helminth to bacterial or even viral infections (41). Thus, activation of STAT3 in such a context could be an important component of the immune response orchestrated by the organism to fight or clear infections (40). The second scenario involves a situation in which the epithelial barrier is compromised, such as that observed during DSS-mediated colitis. In those circumstances, IL-6 produced by immune cells (e.g., macrophages) will diffuse from the interstitial compartment to the luminal region to interact with the apical receptor in differentiated epithelial cells. Activation of STAT3 in IECs in the latter scenario could be also mediated by IL-10 given that IFN-γ triggers the expression of the cytokine receptor in the epithelium (33). However, it is noteworthy to mention that activation of STAT3 in colonic mucosa of colitic mice has been linked to the presence of several other cytokines, including IL-22 and EGF (24, 42). Therefore, the feedback loops established between all those cytokine receptors could dictate the final outcome. However, giving that polarization and expression of cytokine receptors in the intestinal epithelium is regulated by IFN-γ, the compartmentalization of several signaling pathways in the gut could be subject to the amount of IFN-γ delivered by immune cells. Thus, the mechanisms and machinery responsible for controlling the polarization of the receptors needs to be investigated because such process could be indispensable for the proper development of the innate and adaptive immune responses of the colonic mucosa.

The biological function for the polarization of IL-6/STAT3 in IECs present at the crypt surface needs to be investigated; nevertheless, our results provide some clues. For example, IFN-γ induces a sequential activation of STAT1 and STAT3 in the same epithelial cell, something similar to the observed during priming of immune cells by IFN-γ (43, 44). In macrophages, for example, transient accumulation of STAT1 in the nucleus has been described during priming and commitment into specific cell lineages (45). Therefore, we can speculate that a similar function could be performed by STAT proteins in epithelial cells. Furthermore, because p-STAT3, but not p-STAT1, was detected at the nuclei of IECs of colitic mice, we believe that STAT3 plays a more prominent role as a transcription factor, and mucosal p-STAT1 could be rather implicated in the inhibition of cell death (46). Activation of STAT3 transcriptional activity in IECs after cytokine stimulation could be important for the maintenance of epithelial barrier function in the intestinal epithelium by modulating tight junction proteins or reducing cell damage (10, 40, 47). However, an immunomodulatory role for such signaling cannot be ruled out based in our findings as proposed in other contexts (48).

The mechanism by which mucosal CD4+ T cells maintain basal levels of IFN-γ necessary for inducing polarization of IL-6Rα in the IECs remains elusive. However, the tolerogenic CD4+ T cell response triggered by bacterial components could play an active role in such process. In fact, it has been shown that gut microbiota stimulates not only cell maturation, but also commitment of the immune system cells toward specific lineages or functions (49, 50). The impaired secretion of IFN-γ in the colonic mucosa of Crtam-deficient mice strongly supports this theory. Basal levels of IFN-γ in mucosal tissue during such tolerogenic responses could be essential for inducing polarization of cytokine receptors by enhancing IEC differentiation (30). Thus, a mechanism of STAT3 activation like the one described in this review could therefore be influenced/regulated by the microbiota and might be an important immunomodulatory mechanism in the tolerogenic or proinflammatory immune response.

IFN-γ is a pleiotropic cytokine that regulates polarization, commitment, and differentiation of several cell types (27, 30, 51, 52). The mechanisms behind these processes remain highly unknown. In this study, we observed that IFN-γ induced polarized secretion of IL-6 in IECs in a similar fashion to the observed in endometrial epithelial cells exposed to bacteria, LPS, or IL-1α (53). Polarized secretion of cytokines has been poorly investigated; therefore, the relevance of such process in the orchestration of the immune response can only be hypothesized. However, given that our results strongly suggest that IL-6 production by IECs is low in comparison with immune cells (e.g., macrophages), we speculate that its release could be aimed to have an autocrine, rather than a paracrine, effect with specific roles in the local innate immune response. Furthermore, because of the variety of cytokine receptors expressed in the different IECs along the intestinal crypt, STAT3 activation by cytokines could be aimed to target cells with different degrees of differentiation, and therefore, different outcomes should be expected. Investigating the mechanism of STAT3 activation in IECs along the crypt axis could, therefore, be important to understand the real function of STAT3 signaling in the development of IBD and colorectal cancer (11, 47).

In conclusion, we demonstrated a new role for IEC polarization in the compartmentalization of the IL-6/STAT3 signaling in the colonic mucosa during inflammation. This new mechanism could be important for regulating the immune response in the colonic mucosa during colitis development.

We thank Norma Trejo and Octavio López-Méndez for excellent technical assistance. The authors thank Dr. Angeles Hernandez-Cueto for histopathological evaluation of colonic samples.

This work was supported by the Premio Lisker 2016 grant (to P.N.) and Consejo Nacional de Ciencia y Tecnología Grant FC 2015-02-1204 (to V.O.-N.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

BfA

brefeldin A

DSS

dextran sulphate sodium

IBD

inflammatory bowel disease

IEC

intestinal epithelial cell

KO

knockout

MSA

mouse seric albumin

RT

room temperature

WT

wild-type.

1
Gassler
,
N.
,
C.
Rohr
,
A.
Schneider
,
J.
Kartenbeck
,
A.
Bach
,
N.
Obermüller
,
H. F.
Otto
,
F.
Autschbach
.
2001
.
Inflammatory bowel disease is associated with changes of enterocytic junctions.
Am. J. Physiol. Gastrointest. Liver Physiol.
281
:
G216
G228
.
2
Neurath
,
M. F.
2014
.
Cytokines in inflammatory bowel disease.
Nat. Rev. Immunol.
14
:
329
342
.
3
Niessner
,
M.
,
B. A.
Volk
.
1995
.
Altered Th1/Th2 cytokine profiles in the intestinal mucosa of patients with inflammatory bowel disease as assessed by quantitative reversed transcribed polymerase chain reaction (RT-PCR).
Clin. Exp. Immunol.
101
:
428
435
.
4
Autschbach
,
F.
,
T.
Giese
,
N.
Gassler
,
B.
Sido
,
G.
Heuschen
,
U.
Heuschen
,
I.
Zuna
,
P.
Schulz
,
H.
Weckauf
,
I.
Berger
,
H. F.
Otto
,
S. C.
Meuer
.
2002
.
Cytokine/chemokine messenger-RNA expression profiles in ulcerative colitis and Crohn's disease.
Virchows Arch.
441
:
500
513
.
5
Danese
,
S.
,
A.
Gasbarrini
.
2005
.
Chemokines in inflammatory bowel disease.
J. Clin. Pathol.
58
:
1025
1027
.
6
Bollrath
,
J.
,
T. J.
Phesse
,
V. A.
von Burstin
,
T.
Putoczki
,
M.
Bennecke
,
T.
Bateman
,
T.
Nebelsiek
,
T.
Lundgren-May
,
O.
Canli
,
S.
Schwitalla
, et al
.
2009
.
gp130-mediated Stat3 activation in enterocytes regulates cell survival and cell-cycle progression during colitis-associated tumorigenesis.
Cancer Cell
15
:
91
102
.
7
Chen
,
Y. Y.
,
Z. B.
Ma
,
H. Y.
Xu
,
L. J.
Shi
,
D. Y.
Li
,
L. Y.
Sun
,
X. H.
Yin
,
G. Y.
Sang
,
D.
Xu
,
Y. H.
Tang
, et al
.
2015
.
IL-6/STAT3/SOCS3 signaling pathway playing a regulatory role in ulcerative colitis carcinogenesis.
Int. J. Clin. Exp. Med.
8
:
12009
12017
.
8
Zhang
,
J.
,
J.
Wu
,
X.
Peng
,
J.
Song
,
J.
Wang
,
W.
Dong
.
2014
.
Associations between STAT3 rs744166 polymorphisms and susceptibility to ulcerative colitis and Crohn’s disease: a meta-analysis.
PLoS One
9
:
e109625
.
9
Däbritz
,
J.
,
L. M.
Judd
,
H. V.
Chalinor
,
T. R.
Menheniott
,
A. S.
Giraud
.
2016
.
Altered gp130 signalling ameliorates experimental colitis via myeloid cell-specific STAT3 activation and myeloid-derived suppressor cells.
Sci. Rep.
6
:
20584
.
10
Grivennikov
,
S.
,
E.
Karin
,
J.
Terzic
,
D.
Mucida
,
G. Y.
Yu
,
S.
Vallabhapurapu
,
J.
Scheller
,
S.
Rose-John
,
H.
Cheroutre
,
L.
Eckmann
,
M.
Karin
.
2009
.
IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer.
Cancer Cell
15
:
103
113
.
11
Han
,
J.
,
A. L.
Theiss
.
2014
.
Stat3: friend or foe in colitis and colitis-associated cancer?
Inflamm. Bowel Dis.
20
:
2405
2411
.
12
Welte
,
T.
,
S. S.
Zhang
,
T.
Wang
,
Z.
Zhang
,
D. G.
Hesslein
,
Z.
Yin
,
A.
Kano
,
Y.
Iwamoto
,
E.
Li
,
J. E.
Craft
, et al
.
2003
.
STAT3 deletion during hematopoiesis causes Crohn’s disease-like pathogenesis and lethality: a critical role of STAT3 in innate immunity.
Proc. Natl. Acad. Sci. USA
100
:
1879
1884
.
13
Alonzi
,
T.
,
I. P.
Newton
,
P. J.
Bryce
,
E.
Di Carlo
,
G.
Lattanzio
,
M.
Tripodi
,
P.
Musiani
,
V.
Poli
.
2004
.
Induced somatic inactivation of STAT3 in mice triggers the development of a fulminant form of enterocolitis.
Cytokine
26
:
45
56
.
14
Backert
,
I.
,
S. B.
Koralov
,
S.
Wirtz
,
V.
Kitowski
,
U.
Billmeier
,
E.
Martini
,
K.
Hofmann
,
K.
Hildner
,
N.
Wittkopf
,
K.
Brecht
, et al
.
2014
.
STAT3 activation in Th17 and Th22 cells controls IL-22-mediated epithelial host defense during infectious colitis.
J. Immunol.
193
:
3779
3791
.
15
Willson
,
T. A.
,
I.
Jurickova
,
M.
Collins
,
L. A.
Denson
.
2013
.
Deletion of intestinal epithelial cell STAT3 promotes T-lymphocyte STAT3 activation and chronic colitis following acute dextran sodium sulfate injury in mice.
Inflamm. Bowel Dis.
19
:
512
525
.
16
Bjerknes
,
M.
,
H.
Cheng
.
1981
.
Methods for the isolation of intact epithelium from the mouse intestine.
Anat. Rec.
199
:
565
574
.
17
Ecke
,
D.
,
M.
Bleich
,
B.
Schwartz
,
G.
Fraser
,
R.
Greger
.
1996
.
The ion conductances of colonic crypts from dexamethasone-treated rats.
Pflugers Arch.
431
:
419
426
.
18
Laukoetter
,
M. G.
,
P.
Nava
,
A.
Nusrat
.
2008
.
Role of the intestinal barrier in inflammatory bowel disease.
World J. Gastroenterol.
14
:
401
407
.
19
Li
,
Y.
,
C.
de Haar
,
M.
Chen
,
J.
Deuring
,
M. M.
Gerrits
,
R.
Smits
,
B.
Xia
,
E. J.
Kuipers
,
C. J.
van der Woude
.
2010
.
Disease-related expression of the IL6/STAT3/SOCS3 signalling pathway in ulcerative colitis and ulcerative colitis-related carcinogenesis.
Gut
59
:
227
235
.
20
Lee
,
M. J.
,
J. K.
Lee
,
J. W.
Choi
,
C. S.
Lee
,
J. H.
Sim
,
C. H.
Cho
,
K. H.
Lee
,
I. H.
Cho
,
M. H.
Chung
,
H. R.
Kim
, et al
.
2012
.
Interleukin-6 induces S100A9 expression in colonic epithelial cells through STAT3 activation in experimental ulcerative colitis.
PLoS One
7
:
e38801
.
21
Qing
,
Y.
,
G. R.
Stark
.
2004
.
Alternative activation of STAT1 and STAT3 in response to interferon-gamma.
J. Biol. Chem.
279
:
41679
41685
.
22
Dieleman
,
L. A.
,
M. J.
Palmen
,
H.
Akol
,
E.
Bloemena
,
A. S.
Peña
,
S. G.
Meuwissen
,
E. P.
Van Rees
.
1998
.
Chronic experimental colitis induced by dextran sulphate sodium (DSS) is characterized by Th1 and Th2 cytokines.
Clin. Exp. Immunol.
114
:
385
391
.
23
Ito
,
R.
,
M.
Shin-Ya
,
T.
Kishida
,
A.
Urano
,
R.
Takada
,
J.
Sakagami
,
J.
Imanishi
,
M.
Kita
,
Y.
Ueda
,
Y.
Iwakura
, et al
.
2006
.
Interferon-gamma is causatively involved in experimental inflammatory bowel disease in mice.
Clin. Exp. Immunol.
146
:
330
338
.
24
Wang
,
Y.
,
A. H.
van Boxel-Dezaire
,
H.
Cheon
,
J.
Yang
,
G. R.
Stark
.
2013
.
STAT3 activation in response to IL-6 is prolonged by the binding of IL-6 receptor to EGF receptor.
Proc. Natl. Acad. Sci. USA
110
:
16975
16980
.
25
Nebenführ
,
A.
,
C.
Ritzenthaler
,
D. G.
Robinson
.
2002
.
Brefeldin A: deciphering an enigmatic inhibitor of secretion.
Plant Physiol.
130
:
1102
1108
.
26
Luckett-Chastain
,
L. R.
,
R. M.
Gallucci
.
2009
.
Interleukin (IL)-6 modulates transforming growth factor-beta expression in skin and dermal fibroblasts from IL-6-deficient mice.
Br. J. Dermatol.
161
:
237
248
.
27
Boehm
,
U.
,
T.
Klamp
,
M.
Groot
,
J. C.
Howard
.
1997
.
Cellular responses to interferon-gamma.
Annu. Rev. Immunol.
15
:
749
795
.
28
Doğan
,
A.
,
Z. D.
Wang
,
J.
Spencer
.
1995
.
E-cadherin expression in intestinal epithelium.
J. Clin. Pathol.
48
:
143
146
.
29
Hernández-Trejo
,
J. A.
,
D.
Suárez-Pérez
,
I. Z.
Gutiérrez-Martínez
,
O. E.
Fernandez-Vargas
,
C.
Serrano
,
A. A.
Candelario-Martínez
,
M. A.
Meraz-Ríos
,
A. F.
Citalán-Madrid
,
M.
Hernández-Ruíz
,
E.
Reyes-Maldonado
, et al
.
2016
.
The pro-inflammatory cytokines IFNγ/TNFα increase chromogranin A-positive neuroendocrine cells in the colonic epithelium.
Biochem. J.
473
:
3805
3818
.
30
Madara
,
J. L.
,
J.
Stafford
.
1989
.
Interferon-gamma directly affects barrier function of cultured intestinal epithelial monolayers.
J. Clin. Invest.
83
:
724
727
.
31
Jeffery
,
V.
,
A. J.
Goldson
,
J. R.
Dainty
,
M.
Chieppa
,
A.
Sobolewski
.
2017
.
IL-6 signaling regulates small intestinal crypt homeostasis.
J. Immunol.
199
:
304
311
.
32
Schuhmann
,
D.
,
P.
Godoy
,
C.
Weiss
,
A.
Gerloff
,
M. V.
Singer
,
S.
Dooley
,
U.
Böcker
.
2011
.
Interfering with interferon-γ signalling in intestinal epithelial cells: selective inhibition of apoptosis-maintained secretion of anti-inflammatory interleukin-18 binding protein.
Clin. Exp. Immunol.
163
:
65
76
.
33
Kominsky
,
D. J.
,
E. L.
Campbell
,
S. F.
Ehrentraut
,
K. E.
Wilson
,
C. J.
Kelly
,
L. E.
Glover
,
C. B.
Collins
,
A. J.
Bayless
,
B.
Saeedi
,
E.
Dobrinskikh
, et al
.
2014
.
IFN-γ-mediated induction of an apical IL-10 receptor on polarized intestinal epithelia.
J. Immunol.
192
:
1267
1276
.
34
Fuss
,
I. J.
,
M.
Neurath
,
M.
Boirivant
,
J. S.
Klein
,
C.
de la Motte
,
S. A.
Strong
,
C.
Fiocchi
,
W.
Strober
.
1996
.
Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn’s disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5.
J. Immunol.
157
:
1261
1270
.
35
Yeh
,
J. H.
,
S. S.
Sidhu
,
A. C.
Chan
.
2008
.
Regulation of a late phase of T cell polarity and effector functions by Crtam.
Cell
132
:
846
859
.
36
Kuhn
,
K. A.
,
H. M.
Schulz
,
E. H.
Regner
,
E. L.
Severs
,
J. D.
Hendrickson
,
G.
Mehta
,
A. K.
Whitney
,
D.
Ir
,
N.
Ohri
,
C. E.
Robertson
, et al
.
2018
.
Bacteroidales recruit IL-6-producing intraepithelial lymphocytes in the colon to promote barrier integrity.
Mucosal Immunol.
11
:
357
368
.
37
Fiocchi
,
C.
2005
.
Inflammatory bowel disease pathogenesis: therapeutic implications.
Chin. J. Dig. Dis.
6
:
6
9
.
38
Lee
,
J.
,
J. M.
Gonzales-Navajas
,
E.
Raz
.
2008
.
The “polarizing-tolerizing” mechanism of intestinal epithelium: its relevance to colonic homeostasis.
Semin. Immunopathol.
30
:
3
9
.
39
Wittkopf
,
N.
,
G.
Pickert
,
U.
Billmeier
,
M.
Mahapatro
,
S.
Wirtz
,
E.
Martini
,
M.
Leppkes
,
M. F.
Neurath
,
C.
Becker
.
2015
.
Activation of intestinal epithelial Stat3 orchestrates tissue defense during gastrointestinal infection.
PLoS One
10
:
e0118401
.
40
Bao
,
S.
,
K. W.
Beagley
,
M. P.
France
,
J.
Shen
,
A. J.
Husband
.
2000
.
Interferon-gamma plays a critical role in intestinal immunity against Salmonella typhimurium infection.
Immunology
99
:
464
472
.
41
Rothfuchs
,
A. G.
,
C.
Trumstedt
,
H.
Wigzell
,
M. E.
Rottenberg
.
2004
.
Intracellular bacterial infection-induced IFN-γ is critically but not solely dependent on Toll-like receptor 4-myeloid differentiation factor 88-IFN-alpha beta-STAT1 signaling.
J. Immunol.
172
:
6345
6353
.
42
Mitra
,
A.
,
S. K.
Raychaudhuri
,
S. P.
Raychaudhuri
.
2012
.
IL-22 induced cell proliferation is regulated by PI3K/Akt/mTOR signaling cascade.
Cytokine
60
:
38
42
.
43
Günthner
,
R.
,
H. J.
Anders
.
2013
.
Interferon-regulatory factors determine macrophage phenotype polarization.
Mediators Inflamm.
2013
:
731023
.
44
Chistiakov
,
D. A.
,
V. A.
Myasoedova
,
V. V.
Revin
,
A. N.
Orekhov
,
Y. V.
Bobryshev
.
2018
.
The impact of interferon-regulatory factors to macrophage differentiation and polarization into M1 and M2.
Immunobiology
223
:
101
111
.
45
Hoeksema
,
M. A.
,
B. P.
Scicluna
,
M. C.
Boshuizen
,
S.
van der Velden
,
A. E.
Neele
,
J.
Van den Bossche
,
H. L.
Matlung
,
T. K.
van den Berg
,
P.
Goossens
,
M. P.
de Winther
.
2015
.
IFN-γ priming of macrophages represses a part of the inflammatory program and attenuates neutrophil recruitment.
J. Immunol.
194
:
3909
3916
.
46
Sisler
,
J. D.
,
M.
Morgan
,
V.
Raje
,
R. C.
Grande
,
M.
Derecka
,
J.
Meier
,
M.
Cantwell
,
K.
Szczepanek
,
W. J.
Korzun
,
E. J.
Lesnefsky
, et al
.
2015
.
The signal transducer and activator of transcription 1 (STAT1) inhibits mitochondrial biogenesis in liver and fatty acid oxidation in adipocytes.
PLoS One
10
:
e0144444
.
47
Sugimoto
,
K.
2008
.
Role of STAT3 in inflammatory bowel disease.
World J. Gastroenterol.
14
:
5110
5114
.
48
Lavelle
,
E. C.
,
C.
Murphy
,
L. A.
O’Neill
,
E. M.
Creagh
.
2010
.
The role of TLRs, NLRs, and RLRs in mucosal innate immunity and homeostasis.
Mucosal Immunol.
3
:
17
28
.
49
Belkaid
,
Y.
,
T. W.
Hand
.
2014
.
Role of the microbiota in immunity and inflammation.
Cell
157
:
121
141
.
50
Chung
,
H.
,
S. J.
Pamp
,
J. A.
Hill
,
N. K.
Surana
,
S. M.
Edelman
,
E. B.
Troy
,
N. C.
Reading
,
E. J.
Villablanca
,
S.
Wang
,
J. R.
Mora
, et al
.
2012
.
Gut immune maturation depends on colonization with a host-specific microbiota.
Cell
149
:
1578
1593
.
51
Adams
,
R. B.
,
S. M.
Planchon
,
J. K.
Roche
.
1993
.
IFN-gamma modulation of epithelial barrier function. Time course, reversibility, and site of cytokine binding.
J. Immunol.
150
:
2356
2363
.
52
Youakim
,
A.
,
M.
Ahdieh
.
1999
.
Interferon-gamma decreases barrier function in T84 cells by reducing ZO-1 levels and disrupting apical actin.
Am. J. Physiol.
276
:
G1279
G1288
.
53
Healy
,
L. L.
,
J. G.
Cronin
,
I. M.
Sheldon
.
2015
.
Polarized epithelial cells secrete interleukin 6 apically in the bovine endometrium.
Biol. Reprod.
92
:
151
.

The authors have no financial conflicts of interest.

Supplementary data