• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 22
  • 4
  • 3
  • 2
  • 2
  • 2
  • 1
  • Tagged with
  • 42
  • 42
  • 16
  • 12
  • 10
  • 9
  • 8
  • 7
  • 7
  • 6
  • 6
  • 6
  • 6
  • 5
  • 5
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
31

Mononuclear phagocytes in intestinal homeostasis and inflammation

Mathisen, Stephanie Jane January 2015 (has links)
Changes to the composition and function of the gut mononuclear phagocyte (MNP) compartment are associated with the development of intestinal inflammation. Much work has focused on the role of MNPs in gut-associated lymphoid tissue in maintaining homeostasis, however little is known regarding the roles of MNPs during colitis. We have investigated MNPs in the large intestinal lamina propria during the steady state and inflammation. One of our primary aims was to determine the contribution of MNP subsets to intestinal pathology. For our studies of inflammation, we focused mainly on the Helicobacter hepaticus infection &plus; anti-IL-10R model, which induces inflammation of the colon and caecum (typhlocolitis). We defined the composition of the MNP compartment alongside intestinal pathology scores throughout Hh &plus; anti-IL-10R typhlocolitis. Peak pathology, 2-3 weeks after induction of colitis, coincided with peak frequencies of CX<sub>3</sub>CR1<sup>int</sup> Ly6C<sup>&plus;</sup> MNPs. Having observed the accumulation of CX<sub>3</sub>CR1<sup>int</sup> CD64<sup>&plus;</sup> monocyte/macrophage MNPs in the inflamed lamina propria, we conducted comparative whole genome microarray analysis of these cells isolated from the large intestine three weeks after Hh &plus; anti-IL-10R treatment. CX<sub>3</sub>CR1<sup>int</sup> CD64<sup>&plus;</sup> MNPs selectively expressed a variety of pro- and anti-inflammatory genes, including a number of genes which individually can both promote and negatively regulate inflammation. IL-23 is essential for Hh &plus; anti-IL-10R-induced intestinal pathology. We investigated the role of MNPs as a source of IL-23 which drives Hh &plus; anti-IL-10R colitis. Unexpectedly, our results indicate that normally hyporesponsive CX<sub>3</sub>CR1<sup>hi</sup> macrophages may act as the initial source of IL-23, which induces development of colitis. Recruitment of Ly6C<sup>&plus;</sup> MHCII<sup>&plus;</sup> MNPs to the lamina propria was IL-23-dependent, and these cells also expressed IL-23, which may establish a positive feedback loop of immune cell recruitment, activation and IL-23 production. Finally, we also examined how MNPs might be recruited to the colonic lamina propria during inflammation. Our studies support the conclusion that CCR6 is not required for accumulation of monocyte-derived populations in the inflamed intestine. We cannot rule out a role for CCR2, however preliminary data from the Hh &plus; anti-IL-10R colitis model suggest a potential role for CCR1 or its close relation CCRL2. Such pathways could represent new therapeutic targets in inflammatory bowel disease.
32

The Effects of HIV on the Regulation of IL-12 Family Cytokines, IL-12, IL-23, and IL-27 Production in Human Monocyte-derived Macrophages

O'Hara, Shifawn R.K. January 2012 (has links)
IL-12 family cytokines IL-23 and IL-27 play an important role linking innate and adaptive immunity, and regulating T-cell responses. The production of IL-12, a structurally similar cytokine, is decreased in chronic HIV infection; therefore IL-23 and IL-27 may also be influenced by HIV infection. I hypothesized that HIV inhibits LPS-induced IL-23 and IL-27 production in human MDMs by suppressing the activation of signalling pathways regulating their expression. In vitro HIV-infection of MDMs did not have any effect on basal secretion of IL-23 or IL-27; however, HIV inhibited LPS-induced production of IL-12/23 p40 and IL-23 p19, and IL-27 EBI3 and IL-27 p28 mRNA expression, and IL-23, IL-12/23 p40 and IL-27 secretion. In order to evaluate the molecular mechanisms by which HIV inhibits IL-23 and IL-27 in LPS-stimulated MDMs, the signalling pathways regulating their expression were evaluated. The PI3K, p38 MAPK, and JNK MAPK pathways were found to positively regulate LPS-induced IL-27 secretion. Interestingly, in vitro HIV infection inhibited LPS-induced p38 and JNK MAPK activation in MDMs. In summary, I have shown that HIV inhibits IL-23 and IL-27 production in LPS-stimulated MDMs and that HIV may inhibit LPS-induced IL-27 production through the inhibition of p38 and JNK MAPK activation. It is currently unknown whether PKCs regulate LPS-induced IL-23 or IL-27 in human monocytes/macrophages. I demonstrated that classical PKCs differentially regulate LPS-induced IL-23 and IL-27 secretion within THP-1 cells, primary monocytes, and MDMs. Classical PKCs were found to positively regulate LPS-induced IL-12/23 p40 and IL-27 p28 mRNA expression and IL-12/23 p40, IL-23, and IL-27 secretion in primary human monocytes. Similarly, the classical PKCs were found to positively regulate IL-27 p28 mRNA expression and IL-27 secretion in THP-1 cells. However, classical PKCs did not regulate LPS-induced IL-27 production in MDMs, or LPS-induced IL-23 production in THP-1 cells. Overall, this demonstrates that classical PKCs differentially regulate LPS-induced IL-23 and IL-27 production in different myeloid cells.
33

Beyond Th1 and Th2: A non-classical immune pathway induced by Interleukin (IL)-23 complements IL-12 in immunity to Cryptococcus neoformans infection

Kleinschek, Melanie 07 November 2006 (has links)
The interleukin (IL)-12 family of cytokines plays a key role in the orchestration of cellular immune responses, bridging innate and adaptive immunity. The founding member, IL-12, was discovered in the late 1980s as the first heterodimeric cytokine, composed of a 40 kDa (p40) and 35 kDa (p35) subunit. Years of basic and clinical research on this prototypical T helper type (Th)1 cytokine revealed its importance in immunity to intracellular non-viral infections, as well as in cancer and autoimmune diseases. Since the discovery of IL-23 as another cytokine composed of the p40 subunit of IL-12 in the year 2000, IL-23, rather than IL-12, could be shown to be the key player in rodent models of autoimmune diseases such as multiple sclerosis and rheumatoid arthritis. With accumulating evidence revealing IL-23 as the crucial regulator of a non-classical pathway of cellular immunity which is hallmarked by IL-17 producing T cells it is intriguing to gain understanding of the importance of such findings in immunity to infections. The present work describes a series of in vivo studies investigating the role of endogenous as well as exogenous IL-23 in a murine model of chronic fungal infection, cryptococcosis. To address the role of endogenous IL-23, wild-type (WT), IL-12- (IL-12p35-/-), IL-23- (IL-23p19-/-) deficient, as well as IL-12- and IL-23- double deficient (p40-deficient) mice on a C57BL/6 background were infected with Cryptococcus neoformans (C. neoformans). Following infection, p40-deficient mice demonstrated higher mortality than IL-12p35-/- mice. Reconstitution of p40-deficient mice with recombinant murine IL-23 prolonged their survival to levels similar to IL-12p35-/- mice. IL-23p19-/- mice showed a moderately reduced survival time and delayed fungal clearance in the liver. While interferon (IFN)-γ production was similar in WT and IL-23p19-/- mice, production of IL-17 was strongly impaired in the latter. IL-23p19-/- mice produced fewer hepatic granulomata relative to organ burden and showed defective recruitment of mononuclear cells to the brain. Moreover, activation of microglia cells and expression of IL-1β, IL-6, and MCP-1 in the brain was impaired. SUMMARY - 80 - The second part of the present work explores the mechanisms underlying the IL-23 effects by characterizing the role of exogenous IL-23. C. neoformans-infected C57BL/6 WT mice treated with recombinant murine IL-23 showed significantly prolonged survival time as compared to mock-treated control mice. However, complete survival throughout the observation period (100 days) was only achieved following IL-12 treatment. At day 21 post infection (p.i.) the IL-23-treated mice as well as the IL-12 group had a significantly lower fungal burden in the brain than the control mice. However, while IL-12 treatment was associated with elevated serum levels of the proinflammatory mediators IFN-γ, tumor necrosis factor (TNF)-α and nitric oxide, IL-23-treated animals, although more resistant, developed a Th2 response similar to the control group as measured by serum IgE levels. Further experiments to assess the mechanism of action were based on the finding of reduced fungal burden at the site of infection, the peritoneal cavity, at day 8 p.i. following IL-23 treatment. This microbicidal effect was also seen in p40-deficient as well as in T and B cell deficient (RAG-deficient) mice. Administration of IL-23 led to enhanced recruitment of inflammatory cells, not only of T cells but also cells of the innate immune system such as DCs, natural killer cells and granulocytes to the infected site. Although numbers of macrophages were not altered following IL-23 treatment, co-stimulatory molecules were markedly up-regulated on such cells. The chemokine/cytokine pattern induced by IL-23 treatment was hallmarked by proinflammatory mediators such as MCP-1, IL-1β, IL-6, TNF-α and IL-17, but also the Th2 associated cytokine IL-5. From these results it can be concluded that a non-classical immune pathway induced by IL-23 complements the more dominant role of IL-12 in protection against C. neoformans. This novel immune response is characterized by an enhancement of the inflammatory cell response and the production of a proinflammatory cytokine pattern hallmarked by IL-1β, IL-6, TNF-α and IL-17.
34

Identification of disease susceptibility regions on chromosome 17 in a spondyloarthritis mouse model

Irving, Jeofferey-Ann 28 February 2024 (has links)
BACKGROUND: Spondyloarthritis is a subset of inflammatory rheumatic diseases that includes ankylosing spondyloarthritis, psoriatic arthritis, undifferentiated spondyloarthritis, and arthritis related to inflammatory bowel. The IL-23 cytokine has been implicated in the pathogenesis of spondyloarthritis. B10.RIII mice hydrodynamically injected with IL-23 minicircle overexpress the IL-23 cytokine, which leads to the development of spondyloarthritis-like disease. It is important to note that B10.RIII is a major histocompatibility complex (MHC) congenic mouse strain that is susceptible to autoimmune and autoinflammatory diseases where the background strain, C57BL/10 (B10), or the MHC donor strain, RIIIS/J, is resistant. For instance, the B10.RIII strain of mice is susceptible to IL-23-induced arthritis, while the B10 background strain is not. Large contaminating RIII-derived regions outside of the selected congenic interval on chromosome 17 were identified on chromosomes 10, 14, 15, and 17. Genetic variations in these intervals may contribute to the susceptibility of the B10.RIII mice to arthritis induced by IL-23 minicircle. OBJECTIVE: This study aimed to interrogate the arthritis phenotype after IL-23 minicircle injection in Chr17 subcongenic B10.RIII mice. In addition, chromosome 17 RIII-derived Ilrun gene and its role in regulating the Interferon signaling pathway between the B10.RIII and B10 mice were investigated. METHOD: Chromosome 17 subcongenic mice were generated by crossing B10.RIII mice with B10 mice and backcrossing the offspring to the B10.RIII mice. Offspring heterozygous b/r for the Chr17 region were then intercrossed to generate B10.RIII mice that are identical to the B10.RIII mice, except at chromosome 17, where they had the genotypes Chr17b/b, Chr17b/r, or Chr17r/r. These mice were then hydrodynamically injected with IL-23 minicircle DNA, and disease development was monitored every other day for two weeks using two parameters: clinical arthritis score and paw swelling. Bone marrow-derived macrophages were differentiated in vitro from B10.RIII and B10 mice. Cells were stimulated with TLR agonists (Pam3CSK4, Poly (I:C), LPS) that induce either the Ilrun-regulated Interferon signaling pathway or the NF-kB signaling pathway. Gene expression changes of NF-kB and Interferon-induced genes were measured using real-time quantitative PCR. TNF protein concentration in the supernatant was measured by ELISA. RESULTS: Upon IL-23 minicircle injection, Chr17r/r and Chr17b/r B10.RIII mice developed arthritis while Chr17b/b B10.RIII mice did not. In addition, the disease severity increased with the number of r alleles as the Chr17r/r B10.RIII mice had a higher clinical score and paw swelling when compared to the heterozygote Chr17b/r mice. Gene expression analysis of NF-kB and Interferon response genes revealed that there was no difference in the induction of NF-kB and Interferon response genes in bone marrow-derived macrophages from B10.RIII and B10 mice. In addition, there was also no difference in the induction of the Ilrun gene in bone marrow-derived macrophages from B10.RIII and B10 mice. CONCLUSION: The B10.RIII(71NS)/Sn strain contains three major RIII/WySn-derived regions outside of the congenic MHC region. The chromosome 17 cluster appears to play a role in susceptibility to IL-23 minicircle-induced arthritis. In vitro studies with bone marrow-derived macrophages failed to show functional differences in Ilrun between the B10.RIII and B10 mice. Future studies will interrogate chromosome 17 RIII-derived regions in arthritis development in more detail and investigate the role of Ilrun in immune responses using Ilrun knock-out mice. / 2025-02-28T00:00:00Z
35

IL-23 Receptor Expression and Effects of Signaling on T Cell Encephalitogenicity

Smith, Alan Jay 12 September 2011 (has links)
No description available.
36

Von Interleukin-12 zur p40-Zytokinfamilie: Interleukin-12-unabhängige Wirkungen von p40-Zytokinen in der Infekt- und Tumorabwehr

Werner, Christoph 30 September 2003 (has links)
Interleukin-12 (IL-12) ist ein zentrales Zytokin in der Entwicklung einer protektiven, zellulären Typ 1-Immunantwort. Es ist aus einer p40- und einer p35-Untereinheit aufgebaut. Es stimuliert NK- und T-Zellen zur Ausschüttung großer Mengen IFN-gamma und setzt so eine Typ 1-Immunantwort in Gang. Die p40-Untereinheit des IL-12 kommt auch in anderen biologisch wirksamen Verbindungen wie beispielsweise als Monomer, Homodimer oder als IL-23 (in Verbindung mit einer p19-Untereinheit) vor. Während das Homodimer in der vorliegenden Arbeit als reiner Antagonist zu IL-12 zu wirken scheint, wurden für IL-23 bereits zu IL-12 agonistische Wirkungen demonstriert. Im Mittelpunkt der vorliegenden Arbeit stand die Erarbeitung p40-abhängiger Wirkungen unter Ausschluß der Effekte von IL-12, d. h. möglicherweise IL-23-abhängiger Effekte. In den vorliegenden Untersuchungen wurde mit gendeletierten Mäusen gearbeitet, so dass IL-12 in diesen Systemen keine Rolle spielen kann sondern nur die p40-Proteine außer IL-12. Im Infektionsmodell mit Salmonella Enteritidis wurden p35-gendeletierte (p35-/--) und p40-/--Mäuse verwendet. Durch eine Induktion der p40-Expression waren Unterschiede auf die Wirkung der p40-Proteine zurückzuführen. Es zeigte sich, dass p40-Proteine durch die Induktion einer IFN-gamma Produktion eine Verbesserung der Abtötung intrazellulärer Pathogene bewirkten. Dadurch gelang in den p35-/--Mäusen die Eindämmung der systemischen Infektion besser und diese Mäuse überlebten länger als die p40-/--Mäuse. In Tumormodellen mit dem Lewis-Lungenkarzinom und dem Melanom B16 wurden p35/40-/--Mäuse, welche keine p40-Proteine bilden können, mit der für p40 kodierenden DNA gentherapiert. Durch diese lokale Gentherapie kam es zu einer Reduktion des Tumorwachstums. In immunhistologischen Untersuchungen war eine Rekrutierung von Makrophagen und eine Hemmung der Angiogenese im Tumorbereich sichtbar. Lokale und systemische Proteintherapien mit dem Homodimer oder IL-23 hatten keinen Effekt auf das Wachstum des Tumors, was auf die Existenz eines weiteren noch unbekannten heterodimeren p40-Proteins hindeutet. In vitro konnte gezeigt werden, dass IL-23 die IFN-gamma-Produktion durch Splenozyten induziert und dieser Effekt durch das Homodimer antagonisiert werden kann. Interessanterweise kann es in primären Milzzellkulturen auch IL-12 antagonisieren. Eine In-vitro-Infektion führte zu einer p40-abhängigen IFN-gamma-Produktion, die auch durch das Homodimer antagonisiert werden konnte. Während die Effekte der p40-Proteine im Infektionsmodell möglicherweise auf IL-23 zurückgeführt werden können und diese Effekte auch durch In-vitro-Untersuchungen gestützt werden, muss nach den Ergebnissen im Tumormodell auf die Existenz eines weiteren, bisher unbekannten p40-Proteines, p40-x, geschlossen werden. / Interleukin-12 (IL-12) is a key cytokine in the development of a protective cellular Th1 immune response. It consists of a p40 and a p35 subunit. Following stimulation with IL-12, NK and T cells produce large amounts of IFN-gamma resulting in a type 1 immune response. The p40 subunit of IL-12 is also part of other biologically active proteins such as monomeric or homodimeric p40 or the heterodimeric IL-23 (in combination with a p19 subunit). While in this study the homodimeric p40 appears to antagonize IL-12, IL-23 was demonstrated to have agonistic effects. The aim of this study was to investigate p40-dependent effects which can be observed independently of IL-12, i.e. potential effects mediated by IL-23. For the experiments mutant mice were used so that IL-12 dependent mechanisms could not play a role but only p40-dependent proteins excluding IL-12. In a Salmonella Enteritidis infection model p35-gene deleted (p35-/-) and p40-/- mice were used. As the expression of p40 is induced by bacterial antigen, differences between the strains were caused by the p40 protein. During the infection p40 proteins induced IFN-gamma production thus improving the killing of intracellular pathogens. This resulted in a better control of the systemic infection and longer survival periods of the p35-/- mice as compared to p40-/- mice. For the experiments in the tumor model using the Lewis-Lung carcinoma and the Melanoma B16 as tumors, p35/40-/- mice which are unable to produce any p40 proteins, received gene therapy with DNA encoding for p40. This local gene therapy resulted in a reduced tumor growth. Immunohistochemical examination revealed an infiltration of the tumor tissue with macrophages and a reduced neoangiogenesis within the tumor. This effect could not be achieved by local administration of IL-23 or the p40-homodimer as a protein, indicating the existence of an as yet unknown heterodimeric p40 protein. In vitro experiments showed that IL-23 induces IFN-gamma production by splenocytes and this effect can be antagonized by the homodimer. Interestingly, IL-23 is also able to antagonize IL-12 in primary splenocyte cultures. In vitro infection with Salmonella resulted in an p40-dependent IFN-gamma production that could also be antagonized by the homodimer. The protective effects in the infection model might be caused by IL-23, which is supported by the in vitro results. On the other hand, in the tumor model IL-23 does not seem to be the player and it must be concluded that the protective effects are caused by an other as yet unknown p40-dependent protein p40-x.
37

IL-23 receptor and IL-12 receptor expression is restricted to distinct cell types in the IL-23R-GFP reporter mouse

Bellemare, Lisa 02 1900 (has links)
Les maladies inflammatoires de l'intestin (MII) sont caractérisées par des réponses immunitaires incontrôlées dans l'intestin. Des études génétiques ont associé un polymorphisme dans le gène de l'IL23R à la résistance aux MII. IL23R code pour la protéine de l’IL-23r, une sous-unité du récepteur à l’IL-23 (IL-23R). Ce récepteur appartient à la famille de l’IL-12R, contenant plusieurs récepteurs hétérodimériques. D’ailleurs, IL-12R et IL-23R partagent la sous-unité IL12Rb1. Néanmoins, ces deux récepteurs favorisent des réponses immunitaires distinctes (Th1 vs Th17). Ce mémoire caractérise les dynamiques d’expression cellulaires de l’IL-23R et l’IL-12R, afin d’élucider leurs rôles dans l’inflammation. Nous avons établi qu’IL-23R et IL-12R ne sont jamais co-exprimés, malgré qu’ils partagent la sous-unité IL-12Rβ1. Parmi les cellules de rates de souris, la protéine IL-23r est trouvée dans certaines cellules T TCRγδ ou T CD4+, quelques cellules B et des cellules Lti-like. La protéine IL-12Rβ2 est exprimée par quelques cellules B. L’analyse de l’expression de l’IL-23R et l’IL-12R dans différents organes révéla que la plus grande proportion de cellules exprimant l’IL-23R se retrouve dans la lamina propria de l'intestin grêle, alors que les cellules exprimant l’IL-12Rβ2 ont été retrouvées en proportion équivalente dans tous les organes lymphoïdes. Ces observations appuient les études génétiques suggérant un rôle prédominant de l’IL23R dans les intestins. Finalement, des cultures in vitro suggèrent que l’IL-23R ou l’IL-12R avaient des réactions croisées à l’IL-12 ou l’IL-23. L’étude de l’IL-23R dans les MII devrait donc être complémentée par l’étude de l’IL-12R, car les deux récepteurs pourraient avoir des rôles complémentaires. / Inflammatory bowel diseases (IBD) are characterised by uncontrolled immune responses in the gut. Genome-wide association studies (GWAS) have identified a protective polymorphism for IBD in the IL23R gene. IL23R codes for the IL-23r protein, one of the two subunits of IL-23R. IL-23R belongs to the IL-12R family, which contains many heterodimeric receptors. For example, both IL-12R and IL-23R share the IL-12Rβ1 subunit. Nevertheless, IL-12R and IL-23R are associated with different immune processes (Th1 vs. Th17). This thesis characterizes the cellular patterns of expression of both IL-23R and IL-12R, to further elucidate their roles in inflammation. We established that IL-23R and IL-12R were never co-expressed together, even though they share the IL-12Rβ2 subunit. Analysis of murine splenocytes revealed that IL-23R is expressed by some TCRγδ T-cells, a few B-cells, CD4+ T-cells and several Lti-like cells. IL-12R protein was found in a few B-cells. The analysis of IL-23R and IL-12R expression in different organs revealed that the lamina propria of the small intestine was the organ containing the largest proportion of IL-23r+ cells. IL-12R+ cells were found in constant numbers throughout the organs. Finally, in vitro cultures showed that IL-23R and IL-12R had crossed reaction to IL-12 and IL-23. Study of IL-23R in IBD should always be accompanied by IL-12R analysis, because both receptors could have complementary roles.
38

IL-23 receptor and IL-12 receptor expression is restricted to distinct cell types in the IL-23R-GFP reporter mouse

Bellemare, Lisa 02 1900 (has links)
Les maladies inflammatoires de l'intestin (MII) sont caractérisées par des réponses immunitaires incontrôlées dans l'intestin. Des études génétiques ont associé un polymorphisme dans le gène de l'IL23R à la résistance aux MII. IL23R code pour la protéine de l’IL-23r, une sous-unité du récepteur à l’IL-23 (IL-23R). Ce récepteur appartient à la famille de l’IL-12R, contenant plusieurs récepteurs hétérodimériques. D’ailleurs, IL-12R et IL-23R partagent la sous-unité IL12Rb1. Néanmoins, ces deux récepteurs favorisent des réponses immunitaires distinctes (Th1 vs Th17). Ce mémoire caractérise les dynamiques d’expression cellulaires de l’IL-23R et l’IL-12R, afin d’élucider leurs rôles dans l’inflammation. Nous avons établi qu’IL-23R et IL-12R ne sont jamais co-exprimés, malgré qu’ils partagent la sous-unité IL-12Rβ1. Parmi les cellules de rates de souris, la protéine IL-23r est trouvée dans certaines cellules T TCRγδ ou T CD4+, quelques cellules B et des cellules Lti-like. La protéine IL-12Rβ2 est exprimée par quelques cellules B. L’analyse de l’expression de l’IL-23R et l’IL-12R dans différents organes révéla que la plus grande proportion de cellules exprimant l’IL-23R se retrouve dans la lamina propria de l'intestin grêle, alors que les cellules exprimant l’IL-12Rβ2 ont été retrouvées en proportion équivalente dans tous les organes lymphoïdes. Ces observations appuient les études génétiques suggérant un rôle prédominant de l’IL23R dans les intestins. Finalement, des cultures in vitro suggèrent que l’IL-23R ou l’IL-12R avaient des réactions croisées à l’IL-12 ou l’IL-23. L’étude de l’IL-23R dans les MII devrait donc être complémentée par l’étude de l’IL-12R, car les deux récepteurs pourraient avoir des rôles complémentaires. / Inflammatory bowel diseases (IBD) are characterised by uncontrolled immune responses in the gut. Genome-wide association studies (GWAS) have identified a protective polymorphism for IBD in the IL23R gene. IL23R codes for the IL-23r protein, one of the two subunits of IL-23R. IL-23R belongs to the IL-12R family, which contains many heterodimeric receptors. For example, both IL-12R and IL-23R share the IL-12Rβ1 subunit. Nevertheless, IL-12R and IL-23R are associated with different immune processes (Th1 vs. Th17). This thesis characterizes the cellular patterns of expression of both IL-23R and IL-12R, to further elucidate their roles in inflammation. We established that IL-23R and IL-12R were never co-expressed together, even though they share the IL-12Rβ2 subunit. Analysis of murine splenocytes revealed that IL-23R is expressed by some TCRγδ T-cells, a few B-cells, CD4+ T-cells and several Lti-like cells. IL-12R protein was found in a few B-cells. The analysis of IL-23R and IL-12R expression in different organs revealed that the lamina propria of the small intestine was the organ containing the largest proportion of IL-23r+ cells. IL-12R+ cells were found in constant numbers throughout the organs. Finally, in vitro cultures showed that IL-23R and IL-12R had crossed reaction to IL-12 and IL-23. Study of IL-23R in IBD should always be accompanied by IL-12R analysis, because both receptors could have complementary roles.
39

Defective IL-23/IL-17 Axis Protects p47phox−/− Mice from Colon Cancer

Richter, Cornelia, Herrero San Juan, Martina, Weigmann, Benno, Bergis, Dominik, Dauber, Katrin, Muder, Michael H., Baretton, Gustavo B., Pfeilschifter, Josef Martin, Bonig, Halvard, Brenner, Sebastian, Radeke, Heinfried H. 19 July 2017 (has links) (PDF)
In the colon, a sophisticated balance between immune reaction and tolerance is absolutely required. Dysfunction may lead to pathologic phenotypes ranging from chronic inflammatory processes to cancer development. Two prominent modulators of colon inflammation are represented by the closely related cytokines interleukin (IL)-12 and IL-23, which initiate adaptive Th1 and Th17 immune responses, respectively. In this study, we investigated the impact of the NADPH oxidase protein p47phox, which negatively regulates IL-12 in dendritic cells, on colon cancer development in a colitis-associated colon cancer model. Initially, we found that IL-12−/− mice developed less severe colitis but are highly susceptible to colon cancer. By contrast, p47phox−/− mice showed lower tumor scores and fewer high grade tumors than wild-type (WT) littermates. Treatment with toll-like receptor 9 ligand CpG2216 significantly enhanced colitis in p47phox−/− mice, whereas tumor growth was simultaneously reduced. In tumor tissue of p47phox−/− mice, the IL-23/IL-17 axis was crucially hampered. IL-23p19 protein expression in tumor tissue correlated with tumor stage. Reconstitution of WT mice with IL-23p19−/− bone marrow protected these mice from colon cancer, whereas transplantation of WT hematopoiesis into IL-23p19−/− mice increased the susceptibility to tumor growth. Our study strengthens the divergent role of IL-12 and IL-23 in colon cancer development. With the characterization of p47phox as a novel modulator of both cytokines our investigation introduces a promising new target for antitumor strategies.
40

Defective IL-23/IL-17 Axis Protects p47phox−/− Mice from Colon Cancer

Richter, Cornelia, Herrero San Juan, Martina, Weigmann, Benno, Bergis, Dominik, Dauber, Katrin, Muder, Michael H., Baretton, Gustavo B., Pfeilschifter, Josef Martin, Bonig, Halvard, Brenner, Sebastian, Radeke, Heinfried H. 19 July 2017 (has links)
In the colon, a sophisticated balance between immune reaction and tolerance is absolutely required. Dysfunction may lead to pathologic phenotypes ranging from chronic inflammatory processes to cancer development. Two prominent modulators of colon inflammation are represented by the closely related cytokines interleukin (IL)-12 and IL-23, which initiate adaptive Th1 and Th17 immune responses, respectively. In this study, we investigated the impact of the NADPH oxidase protein p47phox, which negatively regulates IL-12 in dendritic cells, on colon cancer development in a colitis-associated colon cancer model. Initially, we found that IL-12−/− mice developed less severe colitis but are highly susceptible to colon cancer. By contrast, p47phox−/− mice showed lower tumor scores and fewer high grade tumors than wild-type (WT) littermates. Treatment with toll-like receptor 9 ligand CpG2216 significantly enhanced colitis in p47phox−/− mice, whereas tumor growth was simultaneously reduced. In tumor tissue of p47phox−/− mice, the IL-23/IL-17 axis was crucially hampered. IL-23p19 protein expression in tumor tissue correlated with tumor stage. Reconstitution of WT mice with IL-23p19−/− bone marrow protected these mice from colon cancer, whereas transplantation of WT hematopoiesis into IL-23p19−/− mice increased the susceptibility to tumor growth. Our study strengthens the divergent role of IL-12 and IL-23 in colon cancer development. With the characterization of p47phox as a novel modulator of both cytokines our investigation introduces a promising new target for antitumor strategies.

Page generated in 0.1775 seconds