Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Crohns Colitis ; 14(3): 406-417, 2020 Mar 13.
Article in English | MEDLINE | ID: mdl-31504344

ABSTRACT

BACKGROUND AND AIMS: Although the mechanisms underlying the formation of intestinal fibrostrictures in Crohn's disease [CD] are not fully understood, activation of fibroblasts and excessive collagen deposition are supposed to contribute to the development of such complications. Here, we investigated the role of cadherin-11 [CDH-11], a fibroblast-derived protein that induces collagen production in various organs, in intestinal fibrosis. METHODS: CDH-11 expression was evaluated in inflammatory [I] and fibrostricturing [FS] CD mucosal samples, ulcerative colitis [UC] mucosal samples, and ileal and colonic control samples, by real-time polymerase chain reaction, western blotting, and immunohistochemistry. CDH-11 expression was evaluated in normal and in CD intestinal fibroblasts stimulated with inflammatory/fibrogenic cytokines. FS CD fibroblasts were cultured either with a specific CDH-11 antisense oligonucleotide [AS], or activating CDH-11 fusion protein and activation of RhoA/ROCK, and TGF-ß pathways and collagen production were evaluated by western blotting. Finally, we assessed the susceptibility of CDH-11-knockout [KO] mice to colitis-induced intestinal fibrosis. RESULTS: CDH-11 RNA and protein expression were increased in both CD and UC as compared with controls. In CD, the greater expression of CDH-11 was seen in FS samples. Stimulation of fibroblasts with TNF-α, interleukin [IL]-6, IFN-γ, IL-13, and IL-1ß enhanced CDH-11 expression. Knockdown of CDH-11 in FS CD fibroblasts impaired RhoA/ROCK/TGF-ß signalling and reduced collagen synthesis, whereas activation of CDH-11 increased collagen secretion. CDH-11 KO mice were largely protected from intestinal fibrosis. CONCLUSIONS: Data show that CDH-11 expression is up-regulated in inflammatory bowel disease [IBD] and suggest a role for this protein in the control of intestinal fibrosis.


Subject(s)
Cadherins/metabolism , Colitis, Ulcerative , Collagen/biosynthesis , Crohn Disease , Intestinal Mucosa/metabolism , Intestines/pathology , Animals , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colitis, Ulcerative/physiopathology , Crohn Disease/metabolism , Crohn Disease/pathology , Crohn Disease/physiopathology , Cytokines/metabolism , Disease Progression , Fibrosis/metabolism , Humans , Mice , Mice, Knockout , Signal Transduction , Up-Regulation
2.
Front Immunol ; 10: 2158, 2019.
Article in English | MEDLINE | ID: mdl-31572375

ABSTRACT

In normal conditions gut homeostasis is maintained by the suppressive activity of regulatory T cells (Tregs), characterized by the expression of the transcription factor FoxP3. In human inflammatory bowel disease, which is believed to be the consequence of the loss of tolerance toward antigens normally contained in the gut lumen, Tregs have been found to be increased and functionally active, thus pointing against their possible role in the pathogenesis of this immune-mediated disease. Though, in inflammatory conditions, Tregs have been shown to upregulate the T helper (Th) type 1-related transcription factor Tbet and to express the pro-inflammatory cytokine IFNγ, thus suggesting that at a certain point of the inflammatory process, Tregs might contribute to inflammation rather than suppress it. Starting from the observation that Tregs isolated from the lamina propria of active but not inactive IBD patients or uninflamed controls express Tbet and IFNγ, we investigated the functional role of Th1-like Tregs in the dextran sulfate model of colitis. As observed in human IBD, Th1-like Tregs were upregulated in the inflamed lamina propria of treated mice and the expression of Tbet and IFNγ in Tregs preceded the accumulation of conventional Th1 cells. By using a Treg-specific Tbet conditional knockout, we demonstrated that Tbet expression in Tregs is required for the development of colitis. Indeed, Tbet knockout mice developed milder colitis and showed an impaired Th1 immune response. In these mice not only the Tbet deficient Tregs but also the Tbet proficient conventional T cells showed reduced IFNγ expression. However, Tbet deficiency did not affect the Tregs suppressive capacity in vitro and in vivo in the adoptive transfer model of colitis. In conclusion here we show that Tbet expression by Tregs sustains the early phase of the Th1-mediated inflammatory response in the gut.


Subject(s)
Colitis/immunology , Gene Expression Regulation/immunology , Inflammatory Bowel Diseases/immunology , T-Box Domain Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Colitis/pathology , Female , Humans , Inflammatory Bowel Diseases/pathology , Male , Mice , T-Lymphocytes, Regulatory/pathology , Th1 Cells/pathology
3.
Front Immunol ; 9: 1854, 2018.
Article in English | MEDLINE | ID: mdl-30147698

ABSTRACT

In inflammatory bowel disease (IBD) mucosa, there is over-expression of Smad7, an intracellular inhibitor of the suppressive cytokine transforming growth factor-ß1, due to post-transcriptional mechanisms that enhance Smad7 acetylation status thus preventing ubiquitination-mediated proteosomal degradation of the protein. IBD-related inflammation is also marked by defective expression of Sirt1, a class III NAD+-dependent deacetylase, which promotes ubiquitination-mediated proteosomal degradation of various intracellular proteins and triggers anti-inflammatory signals. The aim of our study was to determine whether, in IBD, there is a reciprocal regulation between Smad7 and Sirt1. Smad7 and Sirt1 were examined in mucosal samples of IBD patients and normal controls by Western blotting and immunohistochemistry, and Sirt1 activity was assessed by a fluorimetric assay. To determine whether Smad7 is regulated by Sirt1, normal or IBD lamina propria mononuclear cells (LPMC) were cultured with either Sirt1 inhibitor (Ex527) or activator (Cay10591), respectively. To determine whether Smad7 controls Sirt1 expression, ex vivo organ cultures of IBD mucosal explants were treated with Smad7 sense or antisense oligonucleotide. Moreover, Sirt1 expression was evaluated in LPMC isolated from Smad7-transgenic mice given dextran sulfate sodium (DSS). Upregulation of Smad7 was seen in both the epithelial and lamina propria compartments of IBD patients and this associated with reduced expression and activity of Sirt1. Activation of Sirt1 in IBD LPMC with Cay10591 reduced acetylation and enhanced ubiquitination-driven proteasomal-mediated degradation of Smad7, while inhibition of Sirt1 activation in normal LPMC with Ex527 increased Smad7 expression. Knockdown of Smad7 in IBD mucosal explants enhanced Sirt1 expression, thus suggesting a negative effect of Smad7 on Sirt1 induction. Consistently, mucosal T cells of Smad7-transgenic mice contained reduced levels of Sirt1, a defect that was amplified by induction of DSS colitis. The data suggest the existence of a reciprocal regulatory mechanism between Smad7 and Sirt1, which could contribute to amplify inflammatory signals in the gut.


Subject(s)
Colitis/metabolism , Inflammatory Bowel Diseases/metabolism , Leukocytes, Mononuclear/physiology , Mucous Membrane/immunology , Sirtuin 1/metabolism , Smad7 Protein/metabolism , Adolescent , Adult , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , Dextran Sulfate , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Inflammatory Bowel Diseases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Oligodeoxyribonucleotides, Antisense/genetics , RNA, Small Interfering/genetics , Sirtuin 1/genetics , Smad7 Protein/genetics , Ubiquitination , Young Adult
4.
Cancer Immunol Res ; 6(9): 1082-1092, 2018 09.
Article in English | MEDLINE | ID: mdl-29991500

ABSTRACT

Chronic inflammation drives colitis-associated colorectal cancer (CAC) in inflammatory bowel disease (IBD). FoxP3+ regulatory T cells (Treg) coexpressing the Th17-related transcription factor RORγt accumulate in the lamina propria of IBD patients, where they are thought to represent an intermediate stage of development toward a Th17 proinflammatory phenotype. However, the role of these cells in CAC is unknown. RORγt+FoxP3+ cells were investigated in human samples of CAC, and their phenotypic stability and function were investigated in an azoxymethane/dextran sulfate sodium model of CAC using Treg fate-mapping reporter and Treg-specific RORγt conditional knockout mice. Tumor development and the intratumoral inflammatory milieu were characterized in these mice. The functional role of CTLA-4 expressed by Tregs and FoxO3 in dendritic cells (DC) was studied in vitro and in vivo by siRNA-silencing experiments. RORγt expression identified a phenotypically stable population of tumor-infiltrating Tregs in humans and mice. Conditional RORγt knockout mice showed reduced tumor incidence, and dysplastic cells exhibited low Ki67 expression and STAT3 activation. Tumor-infiltrating DCs produced less IL6, a cytokine that triggers STAT3-dependent proliferative signals in neoplastic cells. RORγt-deficient Tregs isolated from tumors overexpressed CTLA-4 and induced DCs to have elevated expression of the transcription factor FoxO3, thus reducing IL6 expression. Finally, in vivo silencing of FoxO3 obtained by siRNA microinjection in the tumors of RORγt-deficient mice restored IL6 expression and tumor growth. These data demonstrate that RORγt expressed by tumor-infiltrating Tregs sustains tumor growth by leaving IL6 expression in DCs unchecked. Cancer Immunol Res; 6(9); 1082-92. ©2018 AACR.


Subject(s)
Colitis/complications , Colorectal Neoplasms/immunology , Dendritic Cells/immunology , Interleukin-6/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Azoxymethane , CTLA-4 Antigen/genetics , Colitis/chemically induced , Colorectal Neoplasms/etiology , Dextran Sulfate , Forkhead Box Protein O3/genetics , Gene Silencing , Humans , Inflammation , Interleukin-6/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering
5.
Inflamm Bowel Dis ; 24(6): 1213-1224, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29668937

ABSTRACT

Background: In Crohn's disease (CD), the pathogenic immune response is associated with high Smad7, an inhibitor of TGF-ß1 signaling. Smad7 knockdown with Mongersen, a specific antisense oligonucleotide-containing compound, restores TGF-ß1 activity leading to inhibition of inflammatory signals and associates with clinical benefit in CD patients. As TGF-ß1 is pro-fibrogenic, it remains unclear whether Mongersen-induced Smad7 inhibition increases the risk of intestinal fibrosis. We assessed the impact of Smad7 inhibition on the course of colitis-driven intestinal fibrosis in mice. Methods: BALB/c mice were rectally treated with increasing doses of trinitrobenzene sulfonic acid (TNBS) for 8 or 12 weeks. The effect of oral Smad7 antisense or control oligonucleotide, administered to mice starting from week 5 or week 8, respectively, on mucosal inflammation and colitis-associated colonic fibrosis was assessed. Mucosal samples were analyzed for Smad7 by immunoblotting and immunohistochemistry, TGF-ß1 by enzyme-linked immunosorbent assay, and collagen by immunohistochemistry. Results: TNBS-induced chronic colitis was associated with colonic deposition of collagen I and fibrosis, which were evident at week 8 and became more pronounced at week 12. TNBS treatment enhanced Smad7 in both colonic epithelial and lamina propria mononuclear cells. Colitic mice treated with Smad7 antisense oligonucleotide exhibited reduced signs of colitis, less collagen deposition, and diminished fibrosis. These findings were associated with diminished synthesis of TGF-ß1 and reduced p-Smad3 protein expression. Conclusion: Attenuation of colitis with Smad7 antisense oligonucleotide limits development of colonic fibrosis.


Subject(s)
Colitis/genetics , Oligonucleotides, Antisense/pharmacology , Smad7 Protein/genetics , Transforming Growth Factor beta1/metabolism , Animals , Colitis/pathology , Collagen Type I/analysis , Colon/pathology , Crohn Disease/therapy , Disease Models, Animal , Down-Regulation , Female , Fibrosis , Gene Knockdown Techniques , Mice , Mice, Inbred BALB C , Oligonucleotides/pharmacology , Signal Transduction , Smad3 Protein/metabolism , Trinitrobenzenesulfonic Acid
6.
Oncotarget ; 6(12): 9908-23, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25839161

ABSTRACT

Interleukin (IL)-21 triggers inflammatory signals that contribute to the growth of neoplastic cells in mouse models of colitis-associated colorectal cancer (CRC). Because most CRCs are sporadic and arise in the absence of overt inflammation we have investigated the role of IL-21 in these tumors in mouse and man. IL-21 was highly expressed in human sporadic CRC and produced mostly by IFN-γ-expressing T-bet/RORγt double-positive CD3+CD8- cells. Stimulation of human CRC cell lines with IL-21 did not directly activate the oncogenic transcription factors STAT3 and NF-kB and did not affect CRC cell proliferation and survival. In contrast, IL-21 modulated the production of protumorigenic factors by human tumor infiltrating T cells. IL-21 was upregulated in the neoplastic areas, as compared with non-tumor mucosa, of Apc(min/+) mice, and genetic ablation of IL-21 in such mice resulted in a marked decrease of both tumor incidence and size. IL-21 deficiency was associated with reduced STAT3/NF-kB activation in both immune cells and neoplastic cells, diminished synthesis of protumorigenic cytokines (that is, IL-17A, IL-22, TNF-α and IL-6), downregulation of COX-2/PGE2 pathway and decreased angiogenesis in the lesions of Apc(min/+) mice. Altogether, data suggest that IL-21 promotes a protumorigenic inflammatory circuit that ultimately sustains the development of sporadic CRC.


Subject(s)
Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Interleukins/immunology , Animals , Carcinogenesis , Cell Growth Processes/immunology , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Female , HT29 Cells , Humans , Inflammation/immunology , Inflammation/pathology , Interferon-alpha/immunology , Interleukins/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/immunology , Signal Transduction
7.
Carcinogenesis ; 35(7): 1536-46, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24480808

ABSTRACT

Transforming growth factor-beta (TGF-ß) is deeply involved in colorectal cancer development and the disruption of the TGF-ß signaling in dysplastic cells is required for tumor to grow. Nevertheless, tumor cells express TGF-ß to escape the immune surveillance mediated by T cells. T-cell expression of Smad7, an intracellular inhibitor of the TGF-ß signaling, protects against colitis-associated colorectal cancer. However, whether Smad7 in T cells might influence colorectal cancer growth independently of chronic inflammation and which T-cell subset is involved in this process is unknown. To address this issue, T-cell-specific Smad7 transgenic mice and wild-type (WT) littermates were subcutaneously transplanted with syngenic MC38 colon carcinoma cells. Smad7Tg mice were resistant to tumor development compared with WT mice and protection was dependent on CD4(+) T cells. Smad7 expression in T cells increased the number of tumor-infiltrating Tbet/ROR-γ-t double-positive CD4 T cells characterized by the expression of tumor necrosis factor-alpha (TNF-α) and interferon-gamma but lower IL17A. The low expression of IL17A caused by the Smad7 expression in tumor-infiltrating CD4(+) T cells enabled the TNF-α-mediated killing of cancer cells both in vitro and in vivo, thus indicating that the Smad7-mediated plastic effect on T-cell phenotype induces protection against colorectal cancer.


Subject(s)
Apoptosis/drug effects , Colorectal Neoplasms/pathology , Interleukin-17/physiology , Lymphocytes, Tumor-Infiltrating/immunology , Smad7 Protein/physiology , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Flow Cytometry , Humans , Immunoenzyme Techniques , Interferon-gamma/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Th17 Cells/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
8.
Hepatology ; 58(4): 1436-50, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23564603

ABSTRACT

UNLABELLED: Fulminant hepatitis (FH) is a disease characterized by massive destruction of hepatocytes with severe impairment of liver function. The pathogenesis of FH is not fully understood, but hyperactivity of T cells and macrophages with excessive production of cytokines are important hallmarks of the condition. In this study, we investigated the role of interleukin (IL)-25 in FH. IL-25 expression was evaluated in patients with FH and in livers of mice with FH induced by D-galactosamine (D-Gal) and lipopolysaccharide (LPS). Mice were treated with IL-25 before D-Gal/LPS-induced FH and before or after concanavalin A (ConA)-induced FH. Mononuclear cells were isolated from livers of mice treated with or without IL-25 and analyzed for GR1(+) CD11b(+) cells. CFSE-labeled T cells were cocultured with GR1(+) CD11b(+) cells and their proliferation was evaluated by flow cytometry. Mice were also treated with a depleting anti-GR1 antibody before IL-25 and D-Gal/LPS administration. IL-25 was constitutively expressed in mouse and human liver and down-regulated during FH. IL-25 prevented D-Gal/LPS-induced FH and this effect was associated with increased infiltration of the liver with cells coexpressing GR1 and CD11b. In vitro studies showed that GR1(+) CD11b(+) cells isolated from mice given IL-25 inhibited T-cell proliferation. Consistently, in vivo depletion of GR1(+) cells abrogated the protective effect of IL-25 in experimental D-Gal/LPS-induced FH. IL-25 was both preventive and therapeutic in ConA-induced FH. CONCLUSIONS: IL-25 expression is markedly reduced during human and experimental FH. IL-25 promotes liver accumulation of GR1(+) CD11b(+) cells with immunoregulatory properties.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Hepatocytes/pathology , Interleukins/therapeutic use , Myeloid Cells/pathology , T-Lymphocytes/pathology , Animals , CD11b Antigen/metabolism , Cell Proliferation/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Coculture Techniques , Concanavalin A/adverse effects , Disease Models, Animal , Down-Regulation/drug effects , Galactosamine/adverse effects , Hepatitis/metabolism , Hepatitis/pathology , Hepatocytes/metabolism , Humans , Interleukin-17/metabolism , Interleukins/metabolism , Interleukins/pharmacology , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred BALB C , Myeloid Cells/metabolism , Receptors, Chemokine/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
9.
Gastroenterology ; 143(5): 1277-1287.e4, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22819866

ABSTRACT

BACKGROUND & AIMS: Tissue inhibitor of metalloproteinases (TIMP)-3 is an inhibitor of matrix metalloproteinases, which regulates tissue inflammation, damage, and repair. We investigated the role of TIMP-3 in intestinal inflammation in human beings and mice. METHODS: We used real-time polymerase chain reaction and flow cytometry to measure levels of TIMP-3 in intestine samples from patients with Crohn's disease (CD) and those without (controls). We also analyzed TIMP-3 levels in lamina propria mononuclear cells (LPMCs) collected from biopsy samples of individuals with or without CD (controls) and then stimulated with transforming growth factor (TGF)-ß1, as well as in biopsy samples collected from patients with CD and then incubated with a Smad7 anti-sense oligonucleotide (knock down). LPMCs and biopsy samples from patients with CD were cultured with exogenous TIMP-3 and levels of inflammatory cytokines were measured. We evaluated the susceptibility of wild-type, TIMP-3-knockout (TIMP-3-KO), and transgenic (TIMP-3-Tg) mice to induction of colitis with 2, 4, 6-trinitrobenzene-sulfonic-acid (TNBS), and the course of colitis in recombinase-activating gene-1-null mice after transfer of wild-type or TIMP-3-KO T cells. RESULTS: Levels of TIMP-3 were reduced in intestine samples from patients with CD compared with controls. Incubation of control LPMCs with TGF-ß1 up-regulated TIMP-3; knockdown of Smad7, an inhibitor of TGF-ß1, in biopsy samples from patients with CD increased levels of TIMP-3. Exogenous TIMP-3 reduced levels of inflammatory cytokines in CD LPMCs and biopsy samples. TIMP-3-KO mice developed severe colitis after administration of TNBS, whereas TIMP-3-Tg mice were resistant to TNBS-induced colitis. Reconstitution of recombinase-activating gene-1-null mice with T cells from TIMP-3-KO mice increased the severity of colitis, compared with reconstitution with wild-type T cells. CONCLUSIONS: TIMP-3 is down-regulated in inflamed intestine of patients with CD. Its expression is regulated by TGF-ß1, and knock-down of Smad7 in intestinal tissues from patient with CD up-regulates TIMP-3. Loss or reduction of TIMP-3 in mice promotes development of colitis.


Subject(s)
Colitis, Ulcerative/metabolism , Crohn Disease/metabolism , Intestinal Mucosa/metabolism , RNA, Messenger/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Adult , Aged , Amyloid Precursor Protein Secretases/metabolism , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Cytokines/metabolism , Down-Regulation , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Intestinal Mucosa/drug effects , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Middle Aged , Oligonucleotides, Antisense/pharmacology , Smad7 Protein/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Transforming Growth Factor beta/pharmacology , Trinitrobenzenesulfonic Acid
10.
Inflamm Bowel Dis ; 18(3): 449-59, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21688353

ABSTRACT

BACKGROUND: Interleukin (IL)-25, a Th2-related factor, inhibits the synthesis of inflammatory cytokines by macrophages and attenuates experimental colitis in mice. The mechanism underlying the counterregulatory effect of IL-25, however, remains unknown. Since Th2-cytokines can abrogate inflammatory pathways by inducing alternatively activated macrophages (AAMs), we evaluated whether AAMs are involved in the IL-25-mediated anticolitic effect. METHODS: AAM-related markers were evaluated in peritoneal and lamina propria mononuclear cells of mice with or without 2,4,6-trinitrobenzenesulphonic acid (TNBS)-induced colitis treated with IL-25 and/or neutralizing IL-4, IL-13, and transforming growth factor beta 1 (TGF-ß1) antibodies. Peritoneal AAMs induced in vivo by injecting mice with IL-25 were transferred to mice with TNBS colitis. Finally, we assessed the in vitro effect of IL-25 on the alternative activation of peritoneal F4/80+ cells. RESULTS: IL-25 enhanced the expression of AAM-related markers in F4/80(+) cells infiltrating the peritoneum and colon of naïve and colitic mice. Peritoneal F4/80(+) cells isolated from IL-25-treated mice reduced the severity of TNBS colitis when injected intraperitoneally to recipient mice. Since IL-25 did not directly induce AAM in vitro and in vivo in mice, IL-25 administration enhanced the expression of IL-4, IL-13, and TGF-ß1, which are known to promote AAM differentiation, we finally assessed whether such cytokines were involved in the IL-25-driven AAM induction. Blockade of IL-4, IL-13, and TGF-ß1 with neutralizing antibodies in mice did not inhibit the stimulatory effect of IL-25 on AAM gene expression. CONCLUSIONS: The IL-25-mediated anticolitic effect is associated with induction of AAMs, a subset of macrophages with antiinflammatory properties.


Subject(s)
Colitis/immunology , Interleukin-17/immunology , Macrophage Activation , Macrophages, Peritoneal/immunology , Analysis of Variance , Animals , Antibodies, Neutralizing/immunology , Antigens, Differentiation/metabolism , Arginase/genetics , Arginase/metabolism , Cell Count , Colitis/chemically induced , Gene Expression , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-13/immunology , Interleukin-17/pharmacology , Interleukin-4/immunology , Intestinal Mucosa/metabolism , Lectins/genetics , Lectins/metabolism , Macrophage Activation/drug effects , Macrophages, Peritoneal/metabolism , Mice , Statistics, Nonparametric , Transforming Growth Factor beta/immunology , Trinitrobenzenesulfonic Acid , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism
11.
Cancer Res ; 71(24): 7423-32, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22028324

ABSTRACT

Patients with inflammatory bowel disease (IBD) have an increased risk of developing colorectal cancer due to chronic inflammation. In IBD, chronic inflammation relies upon a TGFß signaling blockade, but its precise mechanistic relationship to colitis-associated colorectal cancer (CAC) remains unclear. In this study, we investigated the role of the TGFß signaling inhibitor Smad7 in CAC pathogenesis. In human colonic specimens, Smad7 was downregulated in CD4(+) T cells located in the lamina propria of patients with complicated IBD compared with uncomplicated IBD. Therefore, we assessed CAC susceptibility in a transgenic mouse model where Smad7 was overexpressed specifically in T cells. In this model, Smad7 overexpression increased colitis severity, but the mice nevertheless developed fewer tumors than nontransgenic mice. Protection was associated with increased expression of IFNγ and increased accumulation of cytotoxic CD8(+) and natural killer T cells in the tumors and peritumoral areas. Moreover, genetic deficiency in IFNγ abolished the Smad7-dependent protection against CAC. Taken together, our findings defined a novel and unexpected role for Smad7 in promoting a heightened inflammatory response that protects against CAC.


Subject(s)
Colitis/metabolism , Colorectal Neoplasms/metabolism , Smad7 Protein/metabolism , T-Lymphocytes/metabolism , Adult , Aged , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Colitis/genetics , Colitis/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Cytotoxicity, Immunologic/immunology , Dextran Sulfate/immunology , Female , Flow Cytometry , Gene Expression , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Smad7 Protein/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Young Adult
12.
J Exp Med ; 208(11): 2279-90, 2011 Oct 24.
Article in English | MEDLINE | ID: mdl-21987656

ABSTRACT

Chronic inflammation is a major driving force in the development of cancer in many tissues, but the array of factors involved in this neoplastic transformation are not well understood. We have investigated the role of interleukin (IL)-21 in colitis-associated colon cancer (CAC), as this cytokine is overexpressed in the gut mucosa of patients with ulcerative colitis (UC), a chronic inflammatory disease associated with colon cancer. IL-21 was increased in the gut of patients with UC-associated colon cancer, and in mice with CAC induced by azoxymethane (AOM) and dextran sulfate sodium (DSS). After AOM+DSS treatment, IL-21 KO mice showed reduced mucosal damage, reduced infiltration of T cells, and diminished production of IL-6 and IL-17A. IL-21-deficient mice also developed fewer and smaller tumors compared with wild-type (WT) mice. Absence of IL-21 reduced signal transducer and activator of transcription 3 activation in tumor and stromal cells. Administration of a neutralizing IL-21 antibody to WT mice after the last DSS cycle decreased the colonic T cell infiltrate and the production of IL-6 and IL-17A and reduced the number of tumors. These observations indicate that IL-21 amplifies an inflammatory milieu that promotes CAC, and suggest that IL-21 blockade may be useful in reducing the risk of UC-associated colon cancer.


Subject(s)
Colitis/complications , Colitis/immunology , Colonic Neoplasms/etiology , Colonic Neoplasms/immunology , Interleukins/immunology , Animals , Azoxymethane/adverse effects , CD4-Positive T-Lymphocytes/immunology , Carcinogens/pharmacology , Cell Line, Tumor , Colitis/chemically induced , Colitis/pathology , Colon/cytology , Colon/immunology , Colon/pathology , Colonic Neoplasms/pathology , Dextran Sulfate/adverse effects , Forkhead Transcription Factors/immunology , Humans , Interleukin-17/immunology , Interleukin-6/immunology , Interleukins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT3 Transcription Factor/metabolism
13.
World J Gastroenterol ; 17(26): 3092-100, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21912451

ABSTRACT

Chronic inflammation is thought to be the leading cause of many human cancers including colorectal cancer (CRC). Accordingly, epidemiologic and clinical studies indicate that patients affected by ulcerative colitis and Crohn's disease, the two major forms of inflammatory bowel disease, have an increased risk of developing CRC. In recent years, the role of immune cells and their products have been shown to be pivotal in initiation and progression of colitis-associated CRC. On the other hand, activation of the immune system has been shown to cause dysplastic cell elimination and cancer suppression in other settings. Clinical and experimental data herein reviewed, while confirming chronic inflammation as a risk factor for colon carcinogenesis, do not completely rule out the possibility that under certain conditions the chronic activation of the mucosal immune system might protect from colonic dysplasia.


Subject(s)
Colorectal Neoplasms/etiology , Colorectal Neoplasms/immunology , Inflammation , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/immunology , Intestines/pathology , Animals , Cell Transformation, Neoplastic/immunology , Chemokines/immunology , Colorectal Neoplasms/pathology , Humans , Immune System/physiology , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Inflammatory Bowel Diseases/pathology , Interleukin-10/immunology , Interleukin-6/immunology , Intestines/immunology , Transforming Growth Factor beta/immunology , Tumor Necrosis Factor-alpha/immunology
14.
Mol Cancer Ther ; 10(10): 1969-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21817114

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis is a crucial event in the control of tumor growth. However, many cancer cells, including colon cancer cells, are resistant to TRAIL-driven cell death. We have recently shown that 2-methoxy-5-amino-N-hydroxybenzamide (herein termed 2-14), a novel derivative of mesalamine, induces endoplasmic reticulum stress in colon cancer cells. Because endoplasmic reticulum stress-induced signals regulate the expression of molecules involved in TRAIL-driven apoptosis, we examined whether 2-14 makes colon cancer cells sensitive to TRAIL. Colon cancer cells were cultured with 2-14 and/or TRAIL. Death receptor (DR) 4/DR5 were analyzed by real-time PCR and flow cytometry. TRAIL pathway-associated proteins and extracellular signal-regulated kinase (ERK) were assessed by Western blotting. The in vivo capability of 2-14 to sensitize colon cancer cells to TRAIL-induced apoptosis was evaluated in a syngenic colon cancer model in which CT26-derived grafts were induced in mice. 2-14 promoted ERK-dependent induction of DR5, thereby enhancing TRAIL-mediated caspase-8 activation and apoptosis. Analysis of TRAIL-related pro- and antiapoptotic factors and functional studies revealed that survivin is involved in the protection of colon cancer cells against TRAIL-driven apoptosis. Notably, 2-14 enhanced ubiquitination and proteasome-mediated degradation of survivin. These data were confirmed in a murine model of TRAIL-resistant colon cancer in which 2-14 upregulated DR5, reduced survivin expression, and synergized with TRAIL in inhibiting tumor growth. Similarly, intraperitoneal administration of 2-14 to mice upregulated DR5 and downregulated survivin in a model of colitis-associated colon cancer. These findings indicate that 2-14 acts as a sensitizer for TRAIL-induced apoptosis and suggest that 2-14 can be useful in the therapy for TRAIL-resistant colon cancer.


Subject(s)
Aminosalicylic Acids/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Inhibitor of Apoptosis Proteins/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Aminosalicylic Acids/administration & dosage , Animals , Apoptosis/drug effects , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/metabolism , HCT116 Cells , HT29 Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Mice , Mice, Inbred BALB C , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Survivin , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Xenograft Model Antitumor Assays
15.
Gastroenterology ; 141(1): 237-48, 248.e1, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21600206

ABSTRACT

BACKGROUND & AIMS: The pathogenesis of inflammatory bowel disease (IBD) is believed to involve an altered balance between effector and regulatory T cells. Aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor that mediates the toxicity of dioxins, controls T-cell responses. We investigated the role of AhR in inflammation and pathogenesis of IBD in humans and mouse models. METHODS: AhR expression was evaluated in intestinal tissue samples from patients with IBD and controls by real-time polymerase chain reaction (PCR) and flow cytometry. Intestinal lamina propria mononuclear cells (LPMCs) were activated in the presence or absence of the AhR agonist 6-formylindolo(3, 2-b)carbazole (Ficz). Colitis was induced in mice using trinitrobenzene sulfonic acid (TNBS), dextran sulfate sodium (DSS), or T-cell transfer. Mice were given injections of Ficz or the AhR antagonist 2-metyl-2H-pyrazole-3-carboxylic acid; some mice first received injections of a blocking antibody against interleukin (IL)-22. Cytokines were quantified by real-time PCR and flow cytometry. RESULTS: Intestine tissue from patients with IBD expressed significantly less AhR than controls. In LPMCs from patients with IBD, incubation with Ficz reduced levels of interferon gamma (IFN)-γ and up-regulated IL-22. Mice injected with Ficz were protected against TNBS-, DSS-, and T-cell transfer-induced colitis; they had marked down-regulation of inflammatory cytokines and induction of IL-22. Mice given AhR antagonist produced more inflammatory cytokines and less IL-22 and developed a severe colitis. Neutralization of endogenous IL-22 disrupted the protective effect of Ficz on TNBS-induced colitis. CONCLUSIONS: AhR is down-regulated in intestinal tissue of patients with IBD; AhR signaling, via IL-22, inhibits inflammation and colitis in the gastrointestinal tract of mice. AhR-related compounds might be developed to treat patients with IBDs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Inflammatory Bowel Diseases/prevention & control , Interleukins/metabolism , Intestines/immunology , Receptors, Aryl Hydrocarbon/metabolism , Adult , Animals , Basic Helix-Loop-Helix Transcription Factors/drug effects , Biopsy , Carbazoles/pharmacology , Case-Control Studies , Cells, Cultured , Dextran Sulfate , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Flow Cytometry , Humans , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestines/drug effects , Intestines/pathology , Mice , Mice, Inbred BALB C , Middle Aged , Polymerase Chain Reaction , Receptors, Aryl Hydrocarbon/drug effects , Severity of Illness Index , Signal Transduction , T-Lymphocytes/transplantation , Time Factors , Trinitrobenzenesulfonic Acid , Up-Regulation , Interleukin-22
16.
Inflamm Allergy Drug Targets ; 10(3): 187-91, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21428908

ABSTRACT

Although interleukin-25 (IL-25) has been traditionally considered as a cytokine involved in T helper (Th) 2 cell-associated allergic diseases and host defence against helminthic parasites, recent studies have shown that IL-25 exerts negative effects on the initiation and progression of Th1/Th17-mediated pathologies. This later function of IL-25 is particularly evident at the gut level, where IL-25 could contribute to attenuate tissue-damaging immune responses. These new and exciting pre-clinical observations suggest that therapeutic interventions aimed at enhancing IL-25 activity could be useful in the management of patients with chronic gut inflammation.


Subject(s)
Colitis, Ulcerative/immunology , Crohn Disease/immunology , Immunotherapy , Interleukin-17/immunology , Intestinal Mucosa/drug effects , Animals , Colitis, Ulcerative/therapy , Crohn Disease/therapy , Feedback, Physiological , Humans , Immunity, Mucosal/drug effects , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Interleukin-17/pharmacology , Intestinal Mucosa/pathology , Th1-Th2 Balance/drug effects , Th17 Cells/drug effects , Th17 Cells/immunology
17.
Immunology ; 132(1): 66-77, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20840631

ABSTRACT

Interleukin-25 (IL-25), a T helper type 2 (Th2) -related factor, inhibits the production of inflammatory cytokines by monocytes/macrophages. Since Th2 cytokines antagonize classically activated monocytes/macrophages by inducing alternatively activated macrophages (AAMs), we here assessed the effect of IL-25 on the alternative activation of human monocytes/macrophages. The interleukins IL-25, IL-4 and IL-13 were effective in reducing the expression of inflammatory chemokines in monocytes. This effect was paralleled by induction of AAMs in cultures added with IL-4 or IL-13 but not with IL-25, regardless of whether cells were stimulated with lipopolysaccharide or interferon-γ. Moreover, pre-incubation of cells with IL-25 did not alter the ability of both IL-4 and IL-13 to induce AAMs. Both IL-4 and IL-13 activated signal transducer and activator of transcription 6 (STAT6), and silencing of this transcription factor markedly reduced the IL-4/IL-13-driven induction of AAMs. In contrast, IL-25 failed to trigger STAT6 activation. Among Th2 cytokines, only IL-25 and IL-10 were able to activate p38 mitogen-activated protein kinase. These results collectively indicate that IL-25 fails to induce AAMs and that Th2-type cytokines suppress inflammatory responses in human monocytes by activating different intracellular signalling pathways.


Subject(s)
Interleukin-17 , Macrophages/immunology , STAT6 Transcription Factor/immunology , Cells, Cultured , Humans , Interleukin-17/immunology , Interleukin-17/metabolism , Leukocytes, Mononuclear/immunology , Macrophage Activation
18.
Gastroenterology ; 136(7): 2270-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19505427

ABSTRACT

BACKGROUND & AIMS: During the pathogenesis of Crohn's disease (CD), interleukin (IL)-12, a cytokine produced by mucosal CD14+ monocyte-like cells, promotes tissue-damaging T helper cell (Th) 1-mediated inflammation through mechanisms that are not fully understood. IL-25 promotes Th2 cell responses by activating major histocompatibility complex class II-positive non-T and non-B cells. Because Th1 and Th2 cells, and the cytokines they release, are often mutually antagonistic, we examined whether IL-25 affects IL-12 production or Th1 cell-mediated inflammation in the gut. METHODS: Studies were performed using colonic samples from patients and mice with peptidoglycan (PGN)-, 2,4,6-trinitrobenzenesulphonic acid (TNBS)-, or oxazolone-induced colitis. IL-25 receptor (IL-25R) levels were evaluated in intestinal lamina propria mononuclear cells by flow cytometry, and IL-25 levels were measured by real-time polymerase chain reaction, immunoblotting, and immunohistochemistry. Mucosal CD14+ cells from patients with CD were incubated with IL-25 and/or lipopolysaccharide or PGN. Mice were injected with IL-25, and some mice first received injections of an IL-13 blocking antibody. Cytokines were quantified by real-time polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS: CD14+ cells from the mucosa of CD patients expressed IL-25R and responded to IL-25 by decreasing the synthesis of IL-12 and IL-23. IL-25 prevented PGN-induced colitis in mice. IL-25 induced IL-13 production in the colon, but IL-13 was not required for suppression of PGN colitis. IL-25 ameliorated TNBS- and oxazolone-colitis. Patients with CD or ulcerative colitis produced significantly less IL-25 compared with controls. CONCLUSIONS: IL-25 inhibits CD14+ cell-derived cytokines and experimental colitis. IL-25 could be a useful treatment of CD and ulcerative colitis.


Subject(s)
Cytokines/metabolism , Interleukin-12/biosynthesis , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Th1 Cells/immunology , Animals , Blotting, Western , Cells, Cultured , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Crohn Disease/immunology , Crohn Disease/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Inflammation Mediators/analysis , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Probability , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Th1 Cells/physiology
19.
Gastroenterology ; 136(4): 1308-16, e1-3, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19192480

ABSTRACT

BACKGROUND & AIMS: Foxp3-expressing regulatory T cells (Tregs) play a key role in the maintenance of the gut immune homeostasis, and an intact transforming growth factor (TGF)-beta signaling is required for their function. In inflammatory bowel disease (IBD), the TGF-beta signaling is impaired because of high expression of the inhibitory molecule Smad7. Although no intrinsic defects in Tregs function have been shown in IBD, it is still unknown whether colitogenic T cells are susceptible to Treg-mediated suppression. In this study, we have investigated whether IBD mucosal CD4+ T cells are resistant to Tregs and whether Smad7 is involved in this process. METHODS: IBD lamina propria mononuclear cells (LPMC) were cultured with or without Tregs, and proliferation was assessed by flow cytometry. Proliferation of IBD LPMC was also evaluated after Smad7 antisense oligonuclotide treatment. Treg-mediated suppression of T-cell proliferation and proinflammatory cytokine expression was investigated in murine Smad7 transgenic cells. In vivo, the Smad7-dependent resistance of colitogenic naïve T cells to Tregs was studied in the adoptive transfer model of colitis. RESULTS: IBD LPMC were resistant to Treg-mediated suppression, and this phenomenon was reverted by Smad7 antisense treatment. Consistently, CD4+ T cells isolated from Smad7 transgenic mice showed high proliferation, produced considerable amount of inflammatory cytokines following activation, and induced a severe colitis when transferred in immunodeficient RAG1 knockout mice even in the presence of wild-type Tregs. CONCLUSIONS: Smad7 makes CD4+ T cells resistant to Tregs-mediated suppression thus fine-tuning their proinflammatory potential.


Subject(s)
CD4-Positive T-Lymphocytes/pathology , Cell Communication/physiology , Colitis/metabolism , Colitis/pathology , Smad7 Protein/metabolism , T-Lymphocytes, Regulatory/pathology , Animals , Cell Proliferation , Cells, Cultured , Colon/metabolism , Colon/pathology , Crohn Disease/metabolism , Crohn Disease/pathology , Disease Models, Animal , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Up-Regulation
20.
Blood ; 113(15): 3512-9, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19129540

ABSTRACT

IL-25, a member of the IL-17 cytokine family, is known to enhance Th2-like responses associated with increased serum levels of IgE, IgG1, IgA, blood eosinophilia, and eosinophilic infiltrates in various tissues. However, IL-25 also abrogates inflammatory responses driven by Th17 cells. However, the cell types that respond to IL-25 and the mechanisms by which IL-25 differentially regulates immune reactions are not well explored. To identify potential targets of IL-25, we initially examined IL-25 receptor (IL-25R) in human peripheral blood cells. IL-25R was predominantly expressed by CD14(+) cells. We next assessed the functional role of IL-25 in modulating the response of CD14(+) cells to various inflammatory signals. CD14(+) cells responded to IL-25 by down-regulating the synthesis of inflammatory cytokines induced by toll-like receptor (TLR) ligands and inflammatory cytokines. Inhibition of cytokine response by IL-25 occurred via a p38 Map kinase-driven Socs-3-dependent mechanism. In vivo, IL-25 inhibited monocyte-derived cytokines and protected against LPS-induced lethal endotoxemia in mice. These data indicate that IL-25 is a negative regulator of monocyte proinflammatory cytokine responses, which may have therapeutic implications.


Subject(s)
Interleukin-17/metabolism , Monocytes/enzymology , Receptors, Antigen/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Endotoxemia/immunology , Endotoxemia/metabolism , Flow Cytometry , Gene Expression/immunology , Humans , Interleukin-17/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Monocytes/cytology , Monocytes/immunology , STAT1 Transcription Factor/immunology , Suppressor of Cytokine Signaling 3 Protein , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...