Skip to main content
Erschienen in: Inflammation Research 9/2019

Open Access 21.06.2019 | Original Research Paper

ER stress abrogates the immunosuppressive effect of IL-10 on human macrophages through inhibition of STAT3 activation

verfasst von: Ivo S. Hansen, Josca M. Schoonejans, Lathees Sritharan, Johan A. van Burgsteden, Carmen A. Ambarus, Dominique L. P. Baeten, Jeroen den Dunnen

Erschienen in: Inflammation Research | Ausgabe 9/2019

Abstract

Objective and design

To determine whether ER stress affects the inhibitory pathways of the human immune system, particularly the immunosuppressive effect of IL-10 on macrophages.

Material or subjects

In vitro stimulation of human monocyte-derived macrophages.

Treatment

Cells were stimulated with TLR ligands and IL-10, while ER stress was induced using thapsigargin or tunicamycin.

Methods

mRNA expression was determined using qPCR, while cytokine protein production was measured using ELISA. Protein expression of receptors and transcription factors was determined using flow cytometry. Student’s t test was used for statistics.

Results

While under normal conditions IL-10 potently suppresses pro-inflammatory cytokine production by LPS-stimulated macrophages, we demonstrate that ER stress counteracts the immunosuppressive effects of IL-10, leading to increased pro-inflammatory cytokine production. We identified that ER stress directly interferes with IL-10R signaling by reducing STAT3 phosphorylation on Tyr705, which thereby inhibits the expression of SOCS3. Moreover, we show that ER stress also inhibits STAT3 activation induced by other receptors such as IL-6R.

Conclusions

Combined, these data uncover a new general mechanism by which ER stress promotes inflammation. Considering its potential involvement in the pathogenesis of diseases such as Crohn’s disease and spondyloarthritis, targeting of this mechanism may provide new opportunities to counteract inflammation.
Hinweise
Responsible Editor: John Di Battista.
Dominique L. P. Baeten and Jeroen den Dunnen have made equal contribution and share last authorship.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The mechanisms triggering immune activation and inflammation during normal host defense responses are currently well understood. This includes the rapid recognition of pathogens by the innate immune system, which is mediated by detection of molecular patterns expressed as structural component of microorganisms by various innate immune cells. These molecular patterns, such as lipopolysaccharide (LPS) or flagellin, are recognized by a group of receptors called pattern recognition receptors (PRR) [1]. Ligation of molecular patterns to their respective receptors on innate immune cells leads to activation of receptor-specific signaling cascades inducing mostly pro-inflammatory genes tailored towards the type of pathogen that is encountered.
Significant progress has also been made in our understanding of how this inflammatory cascade, once initiated, is regulated and ultimately resolved in order to avoid that the protective response towards the invading microorganisms results in uncontrolled and/or chronic tissue inflammation. Crucial regulators of innate immune responses are anti-inflammatory cytokines such as IL-10. Mice lacking IL-10 spontaneously develop severe enterocolitis [2, 3]. Likewise, infants with loss-of-function mutations in the IL-10R are unable to downregulate LPS-induced macrophage activation and suffer from dramatic inflammatory bowel disease [4].
The latter observation suggests that lack of appropriate regulation and resolution in inflammatory responses may not only be relevant in the context of host defense but also in chronic inflammatory disorders. This mechanism may be more relevant to so-called ‘hyper-inflammatory’ diseases, which are driven by tissue-specific abnormal innate immune responses to various types of cellular stress, rather than by classical acquired immune responses to autoantigens [5]. These hyper-inflammatory diseases include rare monogenic disorders, such as the fever syndromes due to exaggerated IL-1 production [6], as well as polygenic diseases such as Crohn’s disease and spondyloarthritis [7].
A particular form of stress that has been recently recognized to play a role in a variety of chronic inflammatory conditions is endoplasmic reticulum (ER) stress [8]. The ER plays an essential role in the highly regulated process of protein production. High protein production and/or disturbances in protein assembly can lead to an accumulation of unfolded or misfolded proteins in the ER, which induces ER stress and subsequently the so-called unfolded protein response. This is a highly conserved pathway mediated by the kinases IRE1 and PERK that causes an immediate reduction in protein synthesis as well as an increase in protein folding capacity. Whereas this physiological process happens in all cell types, it has become clear in the last years that ER stress plays an important role in shaping of immune responses. For example, immune cells have been shown to be dependent on ER stress proteins during cell differentiation [912]. In addition, ER stress has a direct effect on pro-inflammatory cytokine production by macrophages, since induction of ER stress has been shown to strongly potentiate LPS-induced pro-inflammatory cytokine transcription [13, 14]. Furthermore, in macrophages TLR ligation activates the XBP1 pathway that is necessary for a proper immunological response [15].
In chronic inflammatory conditions, ER stress has mainly been linked to Crohn’s disease and ulcerative colitis, the two main forms of inflammatory bowel disease (IBD). Of the various polymorphisms that are associated with (IBD), several are associated with ER stress components. XBP1 knock-outs show a greater susceptibility to enterocolitis in mice, which was confirmed in human IBD-patients [16, 17]. ER stress has also been linked to spondyloarthritis, where misfolding of HLA-B27 in the ER is hypothesized to potentiate the production of IL-23 [18]. Whereas these data implicate a role for ER stress in triggering inflammation in these conditions, alternatively ER stress may also affect the regulation and resolution of inflammation. Therefore, in this study we set out to investigate the effects of ER stress on the regulatory effects of the prototypical anti-inflammatory cytokine, i.e., IL-10, on the inflammatory response of myeloid cells.

Materials and methods

Ethics statement

This study was done according to the ethical guidelines of the Academic Medical Center and human material was obtained in accordance with the AMC Medical Ethics Review Committee according to the Medical Research Involving Human Subjects Act. Buffy coats obtained after blood donation (Sanquin) are not subjected to informed consent, which is according to the Medical Research Involving Human Subjects Act and the AMC Medical Ethics Review Committee. All samples were handled anonymously.

Cells

In vitro differentiated macrophages were obtained by isolation of monocytes from buffy coats (Sanquin Blood Supply) by density gradient centrifugation using Lymphoprep (Nycomed) and Percoll (Pharmacia). Macrophages were differentiated by culturing the monocytes for 6 days in IMDM (Lonza) containing 5% FBS (Biowest) and 86 µg/mL gentamicin (Gibco), supplemented with 20 ng/mL GM-CSF (Invitrogen). For dendritic cells, medium was additionally supplemented with 2 ng/mL IL-4 (Miltenyi Biotec). At day 2 or 3 half of the medium was replaced with new medium containing cytokines.

Stimulation

Macrophages were harvested by removing medium and washing the cells with PBS and adding TrypLE select (Invitrogen). Macrophages were pre-treated with 10 µM thapsigargin (Calbiochem) or 10 µg/mL tunicamycin (Sigma Aldrich) for 2 h at 37 °C to induce ER stress. Cells (30,000–50,000 per well) were stimulated in 96-well plates (Corning) with 100 ng/mL LPS (from E. coli o111:B4; Sigma Aldrich), 20 µg/mL Poly I:C (Sigma Aldrich), 10 µg/mL Pam3CSK4 (Invivogen), 10 µg/mL MDP (Invivogen), 10 µg/mL curdlan (from Alcaligenes faecalis; Sigma Aldrich), 5 ng/mL IL-10 (Miltenyi Biotec), and 25 ng/mL IL-6 (R&D systems).
For analysis of the cytokines TNF, IL-6, and IL-23, cells were stimulated for 24 h and supernatant was stored at − 20 °C until measured by ELISA. Cytokine levels were measured using antibody pairs for TNF (eBioscience), IL-6, and IL-23 (both UcyTech).

Quantitative real-time PCR

To analyze mRNA levels, cells were lysed after indicated time of stimulation and subsequently mRNA extraction was performed using RNeasy Mini Kit (Qiagen) and cDNA synthesis using RevertAid H Minus First Strand cDNA Synthesis Kit (Fermentas). Quantitative real-time PCR (StepOnePlus Real-Time PCR System; Thermo Fisher Scientific) was performed using Taqman Master Mix and Taqman primers (both from Thermo Fisher Scientific). Primers used were: GAPDH (4310884E), IL12A (Hs01073447_m1), IL12B (Hs01011518_m1), IL23A (Hs00372324_m1), IL6 (Hs00174131_m1), SOCS3 (Hs02330328_s1), and TNF (Hs00174128_m1).

Flow cytometry

Macrophages were stained after inducing ER stress for IL-10R expression using anti-IL10Rα-PE antibody (CD210, REA239, Miltenyi Biotec) in PBS containing 0.5% BSA. For intracellular staining cells were stimulated with IL-10 or IL-6 for 15 min and subsequently, fixed with 4% paraformaldehyde (Thermo Scientific) for 15 min at 37 °C washed and permeabilized using ice-cold methanol for at least 60 min at − 20 °C [19]. Staining was done using the following antibodies: anti-pSTAT3(S727)-PE (558557; BD Biosciences), anti-pSTAT3(Y705) (9145S; Cell Signaling), anti-STAT3 (12640S; Cell Signaling), anti-pSTAT3(S727) (94994S; Cell Signaling), and anti-STAT3-FITC (IC1799F; R&D Systems). Fluorescence was measured using a FACSCanto II (BD Biosciences).

Data analysis

Data were analyzed for statistical significance using paired Student’s t test with GraphPad Prism version 5.01 software (GraphPad Software).

Results

ER stress counteracts the immunosuppressive effect of IL-10 on human LPS-stimulated macrophages

To assess if ER stress impacts the regulation of inflammation, we investigated the effect of IL-10 on inflammatory macrophage responses in the presence or absence of ER stress. In line with literature, stimulation of human GM-CSF differentiated macrophages with LPS induced the production of pro-inflammatory cytokine TNF, which was potently inhibited by the addition of IL-10 (Fig. 1a). However, induction of ER stress by pre-treatment with thapsigargin impaired the suppressive effect of IL-10 on TNF production (Fig. 1a). When selectively comparing the condition of LPS + IL-10 stimulation for multiple donors, ER stress led to a strong increase in the net production of TNF by human macrophages (Fig. 1b). As a control, we verified that ER stress did not affect TNF production induced by LPS stimulation in absence of IL-10 (Fig. 1c). Quantification of the inhibition revealed a significant decrease in the capacity of IL-10 to inhibit LPS-induced production of TNF, but also other pro-inflammatory cytokines such as IL-6 and IL-23 (Fig. 1d).
To confirm and extend these findings, we first assessed whether the same effects were seen at the level of gene transcription. Analysis of mRNA expression of LPS-stimulated cells after 6 h using quantitative real-time PCR confirmed that key pro-inflammatory cytokine transcripts were strongly reduced after addition of IL-10. However, this inhibition of pro-inflammatory cytokine transcription by IL-10 was again impaired by ER stress, to a similar degree as seen on protein level (Fig. 1e).
To further confirm that the observed effects truly result from induction of ER stress, we assessed the effect of tunicamycin, another potent inducer of ER stress. Tunicamycin reduced the suppressive effect of IL-10 in a similar manner as thapsigargin, as shown by protein production of TNF (Fig. 1f) and quantification of inhibition for TNF, IL-6, and IL-23 (Fig. 1g).
Taken together, these data demonstrate that ER stress counteracts the immunosuppressive effect of IL-10 on inflammatory cytokine production by LPS-stimulated macrophages.

ER stress inhibits IL-10-induced STAT3 Tyr705 phosphorylation

Next, we set out to investigate the mechanism of reduced IL-10-mediated effects during ER stress. First, we examined if ER stress affects the expression of the IL-10 receptor (IL-10R) on human macrophages. Neither thapsigargin nor tunicamycin decreased IL-10R surface expression as assessed by flow cytometry (Fig. 2a).
A key event of IL-10R signaling is phosphorylation and subsequent dimerization of STAT3 [20, 21]. We analyzed STAT3 phosphorylation using flow cytometry, which enabled us to quantify phosphorylation differences, but also to determine whether STAT3 phosphorylation occurs in all cells or only in particular subpopulations. To specifically study the impact of ER stress on IL-10-mediated signaling we stimulated the cells with IL-10 alone, i.e., in the absence of LPS. In agreement with the literature [22], stimulation of macrophages with IL-10 led to the universal phosphorylation of STAT3 on tyrosine 705 (Y705), while it did not induce phosphorylation of STAT3 on serine 727 (S727) (Fig. 2b). Whereas neither thapsigargin nor tunicamycin had any effect on the expression of total STAT3 (Fig. 2c), both ER stress inducers reduced IL-10-induced STAT3 Y705 phosphorylation (Fig. 2d, e). Since SOCS3 is one of the main regulators of IL-10R signaling [2325], we next set out to investigate the induction of SOCS3. Indeed, thapsigargin and tunicamycin strongly suppressed IL-10-induced mRNA expression of SOCS3 (Fig. 2f), one of the main downstream effector molecules of IL-10R signaling that is dependent on STAT3-induced gene transcription [23, 24].
Combined, these data indicate that ER stress interferes with IL-10 signaling by suppressing phosphorylation of STAT3 on Y705, leading to strongly reduced levels of SOCS3.

ER stress-induced suppression of STAT3 phosphorylation is not restricted to IL-10 signaling

STAT3 is not only involved in IL-10 signaling, but also in signaling of receptors for several other cytokines, such as IL-6, IL-22 and OSM [26]. To determine whether ER stress specifically suppresses STAT3 activation induced by IL-10R signaling, or whether it also suppresses STAT3 activation induced by other cytokine receptors, we assessed the effect of thapsigargin and tunicamycin on STAT3 phosphorylation after stimulation with IL-6. Similar to IL-10, stimulation of macrophages with IL-6 induced STAT3 phosphorylation at Y705 (Fig. 3a), with only discrete impact on phosphorylation of STAT3 on S727 (Fig. 3b). Importantly, induction of ER stress by both thapsigargin and tunicamycin strongly reduced IL-6-induced phosphorylation of STAT3 at Y705 (Fig. 3c, d). Downstream of STAT3, IL-6-mediated SOCS3 gene transcription was also suppressed after treatment with thapsigargin or tunicamycin (Fig. 3e). These data demonstrate that the inhibition of STAT3 activation by ER stress is not specific for IL-10R, but also suppresses STAT3 activation induced by other cytokine receptors.

ER stress-induced suppression of IL-10 immunoregulation is not restricted to TLR4 or to macrophages

To determine whether ER stress only counteracts the immunosuppressive effect of IL-10 induced by TLR4 signaling, or whether it also impairs IL-10-mediated effects induced by other PRRs, we assessed the effect of several different PRR ligands. As shown in Fig. 4a, ER stress counteracted IL-10-induced suppression of TNF production after stimulation of various different PRRs, including TLR2 (using Pam3CSK4), TLR3 [using Poly (I:C)], NOD2 (using MDP), and Dectin-1 (using curdlan). Next, we examined whether the effect of ER stress was specific for macrophages, or whether it was also functional in other human immune cells. As shown in Fig. 4b, ER stress also reduced the suppressive effect of IL-10 on human dendritic cells (DC). These data demonstrate that ER stress counteracts the immunosuppressive effect of IL-10 upon stimulation of various different families of PRRs and in different cell types.

Discussion

Regulation of inflammatory responses via key anti-inflammatory pathways such as IL-10 is essential to prevent uncontrolled and/or chronic tissue inflammation. Importantly, here we show that macrophages that are undergoing ER stress are less susceptible to the immunosuppressive effects of IL-10. Consequently, this reduced inhibitory effect of IL-10 leads to strongly increased production of pro-inflammatory cytokines such as TNF, IL-6 and IL-23. Furthermore, we identified that ER stress directly interferes with IL-10R signaling by reducing the phosphorylation of STAT3 on Tyr705, which thereby prevents expression of SOCS3 (see Fig. 5 for model).
IL-10 is one of the main cytokines responsible for inducing anti-inflammatory responses, which affects both innate and adaptive immune cells by limiting proliferation, cytokine production and costimulatory molecule expression [27, 28]. Mice deficient in either IL-10 or IL-10R spontaneous develop colitis [2], but only after colonization with microorganisms [3], showing the essential role that IL-10 plays after activation of immune cells by PRRs. Furthermore, IL-10R deficiency that was restricted to the macrophage population in the intestine was shown to induce spontaneous colitis [29]. In this study, we show that ER stress makes immune cells less susceptible to IL-10-induced inhibition of different families of PRRs. Since ER stress has been implicated in several chronic inflammatory disorders, the inability of immune cells to be inhibited during inflammation could play an important role in the pathogeneses of these diseases.
ER stress is strongly associated with pro-inflammatory cytokine production in various immune cells, but particularly in macrophages [30]. Components of the ER stress response such as XBP1 are known to be directly involved in signaling by TLRs, and lack of XBP1 results in impaired TNF, IL-6 and interferon β cytokine production [15]. In addition, ER stress has been linked to production of CXCL1/CXCL2 and IL-1β production through interaction with RIPK1 and GSK-3β, respectively [13, 14]. Recently also Notch has been described to be involved in ER stress [31], indicating that ER stress activates multiple pathways to enhance PRR activation. In addition to directly amplifying PRR-induced responses, we here identified the suppression of the inhibitory pathways of the immune system as a new mechanism by which ER stress promotes inflammation.
STAT3 controls immune regulation in a variety of immune cells. In neutrophils, knocking out STAT3 results in hyper-responsivity to stimulation [32]. Regulatory T cell function is also critically dependent on STAT3, since ablation of STAT3 in regulatory T cells leads to fatal intestinal inflammation mediated by Th17 cells [33]. In addition, STAT3 is a negative regulator of pro-inflammatory DC function, since specific knock-out of STAT3 in the DC population leads to general inflammatory conditions, most notably in the intestine of the animals [34]. Since in all these cell types ER stress is likely to occur during (chronic) immune activation, our finding that ER stress-induced STAT3 inhibition promotes inflammation may be applicable to a large variety of different immune cells. Interestingly, ER stress has previously been identified to suppress leptin-induced STAT3 activation in HEK293 cells, suggesting that this effect is not restricted to immune cells [35].
STAT3 is not only involved in IL-10R signaling, but also in signaling of receptors for several other cytokines. Intriguingly, our data show that ER stress does not only inhibit STAT3 phosphorylation after stimulation with IL-10, but also after stimulation with IL-6. These data indicate that ER stress-induced STAT3 inhibition may have broad implications, by affecting the signaling induced by multiple cytokines, including IL-10, IL-6, IL-22 and OSM [26, 36, 37]. Interestingly, while all the receptors of these cytokines signal via STAT3, the immunological effects of these cytokines are rather diverse, ranging from anti-inflammatory and wound-healing effects of IL-10 and IL-22 [38, 39] to more pro-inflammatory responses by OSM [40]. Therefore, the effect of ER stress-induced STAT3 inhibition will most likely be context-specific, depending on cytokine, cell type, and tissue involved. Interestingly, ER stress has previously also been shown to suppress the phosphorylation of STAT1 [41], suggesting that that inhibition of tyrosine phosphorylation by ER stress may be general feature for all STAT family members.
In this study, we have used SOCS3 as a read-out of STAT3 activation, but SOCS3 is also known to bind gp130 (which is part of the IL-6R complex) and prevent subsequent activation of STAT3, which functions as a negative feedback loop [42]. As such, SOCS3 could potentially play a role in ER stress-induced inhibition of STAT3 phosphorylation. However, since SOCS3 does not regulate IL-10R signaling [4244], and we show that ER stress inhibits both IL-6 and IL-10 signaling, SOCS3 is less likely to play a role in ER stress-induced STAT3 inhibition. Alternatively, numerous other factors have been described to regulate STAT3 activity. For example, protein tyrosine phosphatase 1B (PTP1B), which is upregulated under ER stress conditions [45], has been described as a negative regulator of IL-10-induced STAT3 phosphorylation [46]. In addition, ER stress inhibits the activation of focal adhesion kinase (FAK), leading to reduced STAT3 phosphorylation at S727 and Y705 [47, 48]. Recently, it has also been identified that STAT3 can be phosphorylated at S754, which suppresses STAT3 activity [49]. Other options include TC45, a protein tyrosine phosphatase that regulates STAT3 by de-phosphorylation [50]. However, TC45 seems to require phosphorylation at S727 to mediate inhibition of STAT3 [51]. Interestingly, we only observed phosphorylation of STAT3 at S727 upon stimulation with IL-6, but not upon stimulation with IL-10. Therefore, TC45 activity may provide an explanation for the stronger ER stress-induced inhibition of STAT3 phosphorylation at Y705 that we observed upon stimulation with IL-6 compared to IL-10 (compare Fig. 2d and Fig. 3c). For future research it may also be valuable to assess the activation of the proximal kinases of STAT3, i.e., JAK1 and TYK2 [26], since impaired functioning of these kinases will also lead to suppressed phosphorylation of STAT3.
Albeit beyond the scope of the present study, the mechanisms described here could contribute to chronicity and/or exacerbation of tissue inflammation in a number of immune-mediated inflammatory diseases. First, there is clear genetic and functional evidence for a role of ER stress in IBD, such as the findings that loss of XBP1 is known to result in colonic inflammation [16, 17]. Interestingly, several findings suggest that the mechanism of ER stress-induced inflammation that we identified here, i.e., the suppression of inhibitory pathways, is also involved in chronic intestinal inflammation. Our data that ER stress reduces the sensitivity to IL-10 nicely corresponds with previous findings that show that lack of IL-10 induces colitis [2, 29]. Additionally, the reduced IL-10 sensitivity induced by ER stress particularly amplified the production of TNF and IL-23, two pro-inflammatory cytokines that have strong genetic associations with Crohn’s disease [52] and are effective therapeutic targets in IBD [53, 54]. Second, our findings could be relevant in the context of spondyloarthritis. Spondyloarthritis is strongly linked to expression of HLA-B27 [55], which is prone to misfolding during processing, leading to ER stress. Spondyloarthritis shares several genetic risk factors with Crohn’s disease, including TNF family and IL23R genes [55], and patients have an increased risk of developing colitis. While transgenic rats expressing human HLA-B27 show increased pro-inflammatory cytokine production by macrophages [18, 56], reduction of ER stress by reducing HLA-B27 misfolding through additional trans-gene expression of beta2-microglobulin in rats prevents intestinal inflammation in these animals. Interestingly, these rats still develop axial and peripheral arthritis [57], which highlights the specific importance of ER stress in controlling immune regulation in the intestine.
Taken together, we identified that ER stress abrogates the immunosuppressive effect of IL-10, which is mediated by inhibition of STAT3 phosphorylation. These data uncover a new mechanism by which ER stress promotes inflammation by multiple cell types and cytokines. Considering its potential involvement in the pathogenesis of diseases such as Crohn’s disease and spondyloarthritis, targeting of this mechanism may provide new opportunities to counteract inflammation in these diseases.

Acknowledgements

This work was supported by Grants from the Dutch Digestive Foundation (MLDS; Career Development Grant), Amsterdam UMC (AMC fellowship 2015), Netherlands Scientific Organization (NWO; VICI Grant), and the European Research Council (ERC; Consolidator Grant). MLDS and AMC Grants were awarded to J.d.D. NWO and ERC Grants were awarded to D.L.P.B.

Compliance with ethical standards

Conflict of interest

D.L.P.B. is also an employee of UCB.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Dermatologie

Kombi-Abonnement

Mit e.Med Dermatologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Dermatologie, den Premium-Inhalten der dermatologischen Fachzeitschriften, inklusive einer gedruckten dermatologischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–20.CrossRefPubMed Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–20.CrossRefPubMed
2.
Zurück zum Zitat Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell. 1993;75:263–74.CrossRefPubMed Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell. 1993;75:263–74.CrossRefPubMed
3.
Zurück zum Zitat Sellon RK, Tonkonogy S, Schultz M, Dieleman LA, Grenther W, Balish E, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66:5224–31.PubMedPubMedCentral Sellon RK, Tonkonogy S, Schultz M, Dieleman LA, Grenther W, Balish E, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66:5224–31.PubMedPubMedCentral
4.
Zurück zum Zitat Glocker EO, Kotlarz D, Boztug K, Gertz EM, Schaffer AA, Noyan F, et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med. 2009;361:2033–45.CrossRefPubMedPubMedCentral Glocker EO, Kotlarz D, Boztug K, Gertz EM, Schaffer AA, Noyan F, et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med. 2009;361:2033–45.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Ambarus C, Yeremenko N, Tak PP, Baeten D. Pathogenesis of spondyloarthritis: autoimmune or autoinflammatory? Curr Opin Rheumatol. 2012;24:351–8.CrossRefPubMed Ambarus C, Yeremenko N, Tak PP, Baeten D. Pathogenesis of spondyloarthritis: autoimmune or autoinflammatory? Curr Opin Rheumatol. 2012;24:351–8.CrossRefPubMed
9.
Zurück zum Zitat Reimold AM, Iwakoshi NN, Manis J, Vallabhajosyula P, Szomolanyi-Tsuda E, Gravallese EM, et al. Plasma cell differentiation requires the transcription factor XBP-1. Nature. 2001;412:300–7.CrossRefPubMed Reimold AM, Iwakoshi NN, Manis J, Vallabhajosyula P, Szomolanyi-Tsuda E, Gravallese EM, et al. Plasma cell differentiation requires the transcription factor XBP-1. Nature. 2001;412:300–7.CrossRefPubMed
10.
Zurück zum Zitat Kamimura D, Bevan MJ. Endoplasmic reticulum stress regulator XBP-1 contributes to effector CD8+ T cell differentiation during acute infection. J Immunol. 2008;181:5433–41.CrossRefPubMed Kamimura D, Bevan MJ. Endoplasmic reticulum stress regulator XBP-1 contributes to effector CD8+ T cell differentiation during acute infection. J Immunol. 2008;181:5433–41.CrossRefPubMed
11.
Zurück zum Zitat Iwakoshi NN, Pypaert M, Glimcher LH. The transcription factor XBP-1 is essential for the development and survival of dendritic cells. J Exp Med. 2007;204:2267–75.CrossRefPubMedPubMedCentral Iwakoshi NN, Pypaert M, Glimcher LH. The transcription factor XBP-1 is essential for the development and survival of dendritic cells. J Exp Med. 2007;204:2267–75.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Osorio F, Tavernier SJ, Hoffmann E, Saeys Y, Martens L, Vetters J, et al. The unfolded-protein-response sensor IRE-1alpha regulates the function of CD8alpha + dendritic cells. Nat Immunol. 2014;15:248–57.CrossRefPubMed Osorio F, Tavernier SJ, Hoffmann E, Saeys Y, Martens L, Vetters J, et al. The unfolded-protein-response sensor IRE-1alpha regulates the function of CD8alpha + dendritic cells. Nat Immunol. 2014;15:248–57.CrossRefPubMed
13.
Zurück zum Zitat Zhao C, Pavicic PG Jr, Datta S, Sun D, Novotny M, Hamilton TA. Cellular stress amplifies TLR3/4-induced CXCL1/2 gene transcription in mononuclear phagocytes via RIPK1. J Immunol. 2014;193:879–88.CrossRefPubMed Zhao C, Pavicic PG Jr, Datta S, Sun D, Novotny M, Hamilton TA. Cellular stress amplifies TLR3/4-induced CXCL1/2 gene transcription in mononuclear phagocytes via RIPK1. J Immunol. 2014;193:879–88.CrossRefPubMed
14.
Zurück zum Zitat Kim S, Joe Y, Kim HJ, Kim YS, Jeong SO, Pae HO, et al. Endoplasmic reticulum stress-induced IRE1alpha activation mediates cross-talk of GSK-3beta and XBP-1 to regulate inflammatory cytokine production. J Immunol. 2015;194:4498–506.CrossRefPubMedPubMedCentral Kim S, Joe Y, Kim HJ, Kim YS, Jeong SO, Pae HO, et al. Endoplasmic reticulum stress-induced IRE1alpha activation mediates cross-talk of GSK-3beta and XBP-1 to regulate inflammatory cytokine production. J Immunol. 2015;194:4498–506.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Martinon F, Chen X, Lee AH, Glimcher LH. TLR activation of the transcription factor XBP1 regulates innate immune responses in macrophages. Nat Immunol. 2010;11:411–8.CrossRefPubMedPubMedCentral Martinon F, Chen X, Lee AH, Glimcher LH. TLR activation of the transcription factor XBP1 regulates innate immune responses in macrophages. Nat Immunol. 2010;11:411–8.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Kaser A, Lee AH, Franke A, Glickman JN, Zeissig S, Tilg H, et al. XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease. Cell. 2008;134:743–56.CrossRefPubMedPubMedCentral Kaser A, Lee AH, Franke A, Glickman JN, Zeissig S, Tilg H, et al. XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease. Cell. 2008;134:743–56.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Adolph TE, Tomczak MF, Niederreiter L, Ko HJ, Bock J, Martinez-Naves E, et al. Paneth cells as a site of origin for intestinal inflammation. Nature. 2013;503:272–6.CrossRefPubMedPubMedCentral Adolph TE, Tomczak MF, Niederreiter L, Ko HJ, Bock J, Martinez-Naves E, et al. Paneth cells as a site of origin for intestinal inflammation. Nature. 2013;503:272–6.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat DeLay ML, Turner MJ, Klenk EI, Smith JA, Sowders DP, Colbert RA. HLA-B27 misfolding and the unfolded protein response augment interleukin-23 production and are associated with Th17 activation in transgenic rats. Arthritis Rheum. 2009;60:2633–43.CrossRefPubMedPubMedCentral DeLay ML, Turner MJ, Klenk EI, Smith JA, Sowders DP, Colbert RA. HLA-B27 misfolding and the unfolded protein response augment interleukin-23 production and are associated with Th17 activation in transgenic rats. Arthritis Rheum. 2009;60:2633–43.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Hoepel W, Newling M, Vogelpoel LTC, Sritharan L, Hansen IS, Kapsenberg ML, et al. FcγR-TLR cross-talk enhances TNF production by human monocyte-derived DCs via IRF5-dependent gene transcription and glycolytic reprogramming. Front Immunol. 2019;10:739.CrossRefPubMedPubMedCentral Hoepel W, Newling M, Vogelpoel LTC, Sritharan L, Hansen IS, Kapsenberg ML, et al. FcγR-TLR cross-talk enhances TNF production by human monocyte-derived DCs via IRF5-dependent gene transcription and glycolytic reprogramming. Front Immunol. 2019;10:739.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Williams L, Bradley L, Smith A, Foxwell B. Signal transducer and activator of transcription 3 is the dominant mediator of the anti-inflammatory effects of IL-10 in human macrophages. J Immunol. 2004;172:567–76.CrossRefPubMed Williams L, Bradley L, Smith A, Foxwell B. Signal transducer and activator of transcription 3 is the dominant mediator of the anti-inflammatory effects of IL-10 in human macrophages. J Immunol. 2004;172:567–76.CrossRefPubMed
21.
Zurück zum Zitat Yu CL, Meyer DJ, Campbell GS, Larner AC, Carter-Su C, Schwartz J, et al. Enhanced DNA-binding activity of a Stat3-related protein in cells transformed by the Src oncoprotein. Science. 1995;269:81–3.CrossRefPubMed Yu CL, Meyer DJ, Campbell GS, Larner AC, Carter-Su C, Schwartz J, et al. Enhanced DNA-binding activity of a Stat3-related protein in cells transformed by the Src oncoprotein. Science. 1995;269:81–3.CrossRefPubMed
22.
Zurück zum Zitat Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune system. Nat Rev Immunol. 2003;3:900–11.CrossRefPubMed Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune system. Nat Rev Immunol. 2003;3:900–11.CrossRefPubMed
23.
Zurück zum Zitat Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7:454–65.CrossRefPubMed Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7:454–65.CrossRefPubMed
25.
Zurück zum Zitat Gao Y, Zhao H, Wang P, Wang J, Zou L. The roles of SOCS3 and STAT3 in bacterial infection and inflammatory diseases. Scand J Immunol. 2018;88:e12727.CrossRefPubMed Gao Y, Zhao H, Wang P, Wang J, Zou L. The roles of SOCS3 and STAT3 in bacterial infection and inflammatory diseases. Scand J Immunol. 2018;88:e12727.CrossRefPubMed
26.
Zurück zum Zitat Murray PJ. The JAK-STAT signaling pathway: input and output integration. J Immunol. 2007;178:2623–9.CrossRefPubMed Murray PJ. The JAK-STAT signaling pathway: input and output integration. J Immunol. 2007;178:2623–9.CrossRefPubMed
27.
Zurück zum Zitat Saraiva M, O’Garra A. The regulation of IL-10 production by immune cells. Nat Rev Immunol. 2010;10:170–81.CrossRefPubMed Saraiva M, O’Garra A. The regulation of IL-10 production by immune cells. Nat Rev Immunol. 2010;10:170–81.CrossRefPubMed
28.
29.
Zurück zum Zitat Zigmond E, Bernshtein B, Friedlander G, Walker CR, Yona S, Kim KW, et al. Macrophage-restricted interleukin-10 receptor deficiency, but not IL-10 deficiency, causes severe spontaneous colitis. Immunity. 2014;40:720–33.CrossRefPubMed Zigmond E, Bernshtein B, Friedlander G, Walker CR, Yona S, Kim KW, et al. Macrophage-restricted interleukin-10 receptor deficiency, but not IL-10 deficiency, causes severe spontaneous colitis. Immunity. 2014;40:720–33.CrossRefPubMed
30.
31.
Zurück zum Zitat Nolin E, Gans S, Llamas L, Bandyopadhyay S, Brittain SM, Bernasconi-Elias P, et al. Discovery of a ZIP7 inhibitor from a Notch pathway screen. Nat Chem Biol. 2019;15:179–88.CrossRefPubMedPubMedCentral Nolin E, Gans S, Llamas L, Bandyopadhyay S, Brittain SM, Bernasconi-Elias P, et al. Discovery of a ZIP7 inhibitor from a Notch pathway screen. Nat Chem Biol. 2019;15:179–88.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Lee CK, Raz R, Gimeno R, Gertner R, Wistinghausen B, Takeshita K, et al. STAT3 is a negative regulator of granulopoiesis but is not required for G-CSF-dependent differentiation. Immunity. 2002;17:63–72.CrossRefPubMed Lee CK, Raz R, Gimeno R, Gertner R, Wistinghausen B, Takeshita K, et al. STAT3 is a negative regulator of granulopoiesis but is not required for G-CSF-dependent differentiation. Immunity. 2002;17:63–72.CrossRefPubMed
33.
Zurück zum Zitat Chaudhry A, Rudra D, Treuting P, Samstein RM, Liang Y, Kas A, et al. CD4 + regulatory T cells control TH17 responses in a Stat3-dependent manner. Science. 2009;326:986–91.CrossRefPubMedPubMedCentral Chaudhry A, Rudra D, Treuting P, Samstein RM, Liang Y, Kas A, et al. CD4 + regulatory T cells control TH17 responses in a Stat3-dependent manner. Science. 2009;326:986–91.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Melillo JA, Song L, Bhagat G, Blazquez AB, Plumlee CR, Lee C, et al. Dendritic cell (DC)-specific targeting reveals Stat3 as a negative regulator of DC function. J Immunol. 2010;184:2638–45.CrossRefPubMed Melillo JA, Song L, Bhagat G, Blazquez AB, Plumlee CR, Lee C, et al. Dendritic cell (DC)-specific targeting reveals Stat3 as a negative regulator of DC function. J Immunol. 2010;184:2638–45.CrossRefPubMed
35.
Zurück zum Zitat Hosoi T, Sasaki M, Miyahara T, Hashimoto C, Matsuo S, Yoshii M, et al. Endoplasmic reticulum stress induces leptin resistance. Mol Pharmacol. 2008;74:1610–9.CrossRefPubMed Hosoi T, Sasaki M, Miyahara T, Hashimoto C, Matsuo S, Yoshii M, et al. Endoplasmic reticulum stress induces leptin resistance. Mol Pharmacol. 2008;74:1610–9.CrossRefPubMed
36.
Zurück zum Zitat Auguste P, Guillet C, Fourcin M, Olivier C, Veziers J, Pouplard-Barthelaix A, et al. Signaling of type II oncostatin M receptor. J Biol Chem. 1997;272:15760–4.CrossRefPubMed Auguste P, Guillet C, Fourcin M, Olivier C, Veziers J, Pouplard-Barthelaix A, et al. Signaling of type II oncostatin M receptor. J Biol Chem. 1997;272:15760–4.CrossRefPubMed
37.
Zurück zum Zitat Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 2011;1813:878–88.CrossRefPubMed Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 2011;1813:878–88.CrossRefPubMed
38.
Zurück zum Zitat Mizoguchi A. Healing of intestinal inflammation by IL-22. Inflamm Bowel Dis. 2012;18:1777–84.CrossRefPubMed Mizoguchi A. Healing of intestinal inflammation by IL-22. Inflamm Bowel Dis. 2012;18:1777–84.CrossRefPubMed
39.
Zurück zum Zitat King A, Balaji S, Le LD, Crombleholme TM, Keswani SG. Regenerative wound healing: the role of interleukin-10. Adv Wound Care (New Rochelle). 2014;3:315–23.CrossRef King A, Balaji S, Le LD, Crombleholme TM, Keswani SG. Regenerative wound healing: the role of interleukin-10. Adv Wound Care (New Rochelle). 2014;3:315–23.CrossRef
40.
Zurück zum Zitat West NR, Hegazy AN, Owens BMJ, Bullers SJ, Linggi B, Buonocore S, et al. Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor-neutralizing therapy in patients with inflammatory bowel disease. Nat Med. 2017;23:579–89.CrossRefPubMedPubMedCentral West NR, Hegazy AN, Owens BMJ, Bullers SJ, Linggi B, Buonocore S, et al. Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor-neutralizing therapy in patients with inflammatory bowel disease. Nat Med. 2017;23:579–89.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Ho HJ, Huang DY, Ho FM, Lee LT, Lin WW. Inhibition of lipopolysaccharide-induced inducible nitric oxide synthase expression by endoplasmic reticulum stress. Cell Signal. 2012;24:2166–78.CrossRefPubMed Ho HJ, Huang DY, Ho FM, Lee LT, Lin WW. Inhibition of lipopolysaccharide-induced inducible nitric oxide synthase expression by endoplasmic reticulum stress. Cell Signal. 2012;24:2166–78.CrossRefPubMed
42.
Zurück zum Zitat Yasukawa H, Ohishi M, Mori H, Murakami M, Chinen T, Aki D, et al. IL-6 induces an anti-inflammatory response in the absence of SOCS3 in macrophages. Nat Immunol. 2003;4:551–6.CrossRefPubMed Yasukawa H, Ohishi M, Mori H, Murakami M, Chinen T, Aki D, et al. IL-6 induces an anti-inflammatory response in the absence of SOCS3 in macrophages. Nat Immunol. 2003;4:551–6.CrossRefPubMed
43.
Zurück zum Zitat Niemand C, Nimmesgern A, Haan S, Fischer P, Schaper F, Rossaint R, et al. Activation of STAT3 by IL-6 and IL-10 in primary human macrophages is differentially modulated by suppressor of cytokine signaling 3. J Immunol. 2003;170:3263–72.CrossRefPubMed Niemand C, Nimmesgern A, Haan S, Fischer P, Schaper F, Rossaint R, et al. Activation of STAT3 by IL-6 and IL-10 in primary human macrophages is differentially modulated by suppressor of cytokine signaling 3. J Immunol. 2003;170:3263–72.CrossRefPubMed
45.
Zurück zum Zitat Panzhinskiy E, Hua Y, Culver B, Ren J, Nair S. Endoplasmic reticulum stress upregulates protein tyrosine phosphatase 1B and impairs glucose uptake in cultured myotubes. Diabetologia. 2013;56:598–607.CrossRefPubMed Panzhinskiy E, Hua Y, Culver B, Ren J, Nair S. Endoplasmic reticulum stress upregulates protein tyrosine phosphatase 1B and impairs glucose uptake in cultured myotubes. Diabetologia. 2013;56:598–607.CrossRefPubMed
46.
Zurück zum Zitat Pike KA, Hutchins AP, Vinette V, Theberge JF, Sabbagh L, Tremblay ML, et al. Protein tyrosine phosphatase 1B is a regulator of the interleukin-10-induced transcriptional program in macrophages. Sci Signal. 2014;7:ra43.CrossRefPubMed Pike KA, Hutchins AP, Vinette V, Theberge JF, Sabbagh L, Tremblay ML, et al. Protein tyrosine phosphatase 1B is a regulator of the interleukin-10-induced transcriptional program in macrophages. Sci Signal. 2014;7:ra43.CrossRefPubMed
47.
Zurück zum Zitat Banerjee K, Keasey MP, Razskazovskiy V, Visavadiya NP, Jia C, Hagg T. Reduced FAK-STAT3 signaling contributes to ER stress-induced mitochondrial dysfunction and death in endothelial cells. Cell Signal. 2017;36:154–62.CrossRefPubMedPubMedCentral Banerjee K, Keasey MP, Razskazovskiy V, Visavadiya NP, Jia C, Hagg T. Reduced FAK-STAT3 signaling contributes to ER stress-induced mitochondrial dysfunction and death in endothelial cells. Cell Signal. 2017;36:154–62.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Pei G, Lan Y, Chen D, Ji L, Hua ZC. FAK regulates E-cadherin expression via p-SrcY416/p-ERK1/2/p-Stat3Y705 and PPARgamma/miR-125b/Stat3 signaling pathway in B16F10 melanoma cells. Oncotarget. 2017;8:13898–908.PubMedPubMedCentral Pei G, Lan Y, Chen D, Ji L, Hua ZC. FAK regulates E-cadherin expression via p-SrcY416/p-ERK1/2/p-Stat3Y705 and PPARgamma/miR-125b/Stat3 signaling pathway in B16F10 melanoma cells. Oncotarget. 2017;8:13898–908.PubMedPubMedCentral
49.
Zurück zum Zitat Hsia HC, Hutti JE, Baldwin AS. Cytosolic DNA promotes signal transducer and activator of transcription 3 (STAT3) phosphorylation by TANK-binding kinase 1 (TBK1) to restrain STAT3 activity. J Biol Chem. 2017;292:5405–17.CrossRefPubMedPubMedCentral Hsia HC, Hutti JE, Baldwin AS. Cytosolic DNA promotes signal transducer and activator of transcription 3 (STAT3) phosphorylation by TANK-binding kinase 1 (TBK1) to restrain STAT3 activity. J Biol Chem. 2017;292:5405–17.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Lam MH, Michell BJ, Fodero-Tavoletti MT, Kemp BE, Tonks NK, Tiganis T. Cellular stress regulates the nucleocytoplasmic distribution of the protein-tyrosine phosphatase TCPTP. J Biol Chem. 2001;276:37700–7.CrossRefPubMed Lam MH, Michell BJ, Fodero-Tavoletti MT, Kemp BE, Tonks NK, Tiganis T. Cellular stress regulates the nucleocytoplasmic distribution of the protein-tyrosine phosphatase TCPTP. J Biol Chem. 2001;276:37700–7.CrossRefPubMed
51.
Zurück zum Zitat Wakahara R, Kunimoto H, Tanino K, Kojima H, Inoue A, Shintaku H, et al. Phospho-Ser727 of STAT3 regulates STAT3 activity by enhancing dephosphorylation of phospho-Tyr705 largely through TC45. Genes Cells. 2012;17:132–45.CrossRefPubMed Wakahara R, Kunimoto H, Tanino K, Kojima H, Inoue A, Shintaku H, et al. Phospho-Ser727 of STAT3 regulates STAT3 activity by enhancing dephosphorylation of phospho-Tyr705 largely through TC45. Genes Cells. 2012;17:132–45.CrossRefPubMed
53.
Zurück zum Zitat Ungar B, Kopylov U. Advances in the development of new biologics in inflammatory bowel disease. Ann Gastroenterol. 2016;29:243–8.PubMedPubMedCentral Ungar B, Kopylov U. Advances in the development of new biologics in inflammatory bowel disease. Ann Gastroenterol. 2016;29:243–8.PubMedPubMedCentral
54.
Zurück zum Zitat Pugliese D, Felice C, Papa A, Gasbarrini A, Rapaccini GL, Guidi L, et al. Anti TNF-alpha therapy for ulcerative colitis: current status and prospects for the future. Expert Rev Clin Immunol. 2017;13:223–33.CrossRefPubMed Pugliese D, Felice C, Papa A, Gasbarrini A, Rapaccini GL, Guidi L, et al. Anti TNF-alpha therapy for ulcerative colitis: current status and prospects for the future. Expert Rev Clin Immunol. 2017;13:223–33.CrossRefPubMed
55.
Zurück zum Zitat Brown MA, Kenna T, Wordsworth BP. Genetics of ankylosing spondylitis–insights into pathogenesis. Nat Rev Rheumatol. 2016;12:81–91.CrossRefPubMed Brown MA, Kenna T, Wordsworth BP. Genetics of ankylosing spondylitis–insights into pathogenesis. Nat Rev Rheumatol. 2016;12:81–91.CrossRefPubMed
56.
Zurück zum Zitat Smith JA, Turner MJ, DeLay ML, Klenk EI, Sowders DP, Colbert RA. Endoplasmic reticulum stress and the unfolded protein response are linked to synergistic IFN-beta induction via X-box binding protein 1. Eur J Immunol. 2008;38:1194–203.CrossRefPubMedPubMedCentral Smith JA, Turner MJ, DeLay ML, Klenk EI, Sowders DP, Colbert RA. Endoplasmic reticulum stress and the unfolded protein response are linked to synergistic IFN-beta induction via X-box binding protein 1. Eur J Immunol. 2008;38:1194–203.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Tran TM, Dorris ML, Satumtira N, Richardson JA, Hammer RE, Shang J, et al. Additional human beta2-microglobulin curbs HLA-B27 misfolding and promotes arthritis and spondylitis without colitis in male HLA-B27-transgenic rats. Arthritis Rheum. 2006;54:1317–27.CrossRefPubMed Tran TM, Dorris ML, Satumtira N, Richardson JA, Hammer RE, Shang J, et al. Additional human beta2-microglobulin curbs HLA-B27 misfolding and promotes arthritis and spondylitis without colitis in male HLA-B27-transgenic rats. Arthritis Rheum. 2006;54:1317–27.CrossRefPubMed
Metadaten
Titel
ER stress abrogates the immunosuppressive effect of IL-10 on human macrophages through inhibition of STAT3 activation
verfasst von
Ivo S. Hansen
Josca M. Schoonejans
Lathees Sritharan
Johan A. van Burgsteden
Carmen A. Ambarus
Dominique L. P. Baeten
Jeroen den Dunnen
Publikationsdatum
21.06.2019
Verlag
Springer International Publishing
Erschienen in
Inflammation Research / Ausgabe 9/2019
Print ISSN: 1023-3830
Elektronische ISSN: 1420-908X
DOI
https://doi.org/10.1007/s00011-019-01261-9

Weitere Artikel der Ausgabe 9/2019

Inflammation Research 9/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.