Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nat Immunol ; 14(9): 901-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23872679

RESUMO

Type I interferons are important in regulating immune responses to pathogens and tumors. All interferons are considered to signal via the heterodimeric IFNAR1-IFNAR2 complex, yet some subtypes such as interferon-ß (IFN-ß) can exhibit distinct functional properties, although the molecular basis of this is unclear. Here we demonstrate IFN-ß can uniquely and specifically ligate to IFNAR1 in an IFNAR2-independent manner, and we provide the structural basis of the IFNAR1-IFN-ß interaction. The IFNAR1-IFN-ß complex transduced signals that modulated expression of a distinct set of genes independently of Jak-STAT pathways. Lipopolysaccharide-induced sepsis was ameliorated in Ifnar1(-/-) mice but not Ifnar2(-/-) mice, suggesting that IFNAR1-IFN-ß signaling is pathologically relevant. Thus, we provide a molecular basis for understanding specific functions of IFN-ß.


Assuntos
Interferon beta/química , Interferon beta/metabolismo , Receptor de Interferon alfa e beta/química , Receptor de Interferon alfa e beta/metabolismo , Transdução de Sinais , Animais , Modelos Animais de Doenças , Feminino , Lipopolissacarídeos/efeitos adversos , Camundongos , Camundongos Knockout , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Receptor de Interferon alfa e beta/genética , Choque Séptico/induzido quimicamente , Choque Séptico/genética , Choque Séptico/metabolismo , Choque Séptico/mortalidade
2.
Cytokine ; 149: 155750, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34689057

RESUMO

Interleukin-11 (IL-11) is a cytokine that has been strongly implicated in the pathogenesis of fibrotic diseases and solid malignancies. Elevated IL-11 expression is also associated with several non-malignant inflammatory diseases where its function remains less well-characterized. Here, we summarize current literature surrounding the contribution of IL-11 to the pathogenesis of autoimmune inflammatory diseases, including rheumatoid arthritis, multiple sclerosis, diabetes and systemic sclerosis, as well as other chronic inflammatory conditions such as periodontitis, asthma, chronic obstructive pulmonary disease, psoriasis and colitis.


Assuntos
Inflamação/metabolismo , Interleucina-11/metabolismo , Animais , Doenças Autoimunes/metabolismo , Humanos
3.
Adv Exp Med Biol ; 1240: 59-72, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32060888

RESUMO

Interleukin (IL)-18, a member of the IL-1 family of cytokines, has emerged as a key regulator of mucosal homeostasis within the gastrointestinal tract. Like other members of this family, IL-18 is secreted as an inactive protein and is processed into its active form by caspase-1, although other contributors to precursor processing are emerging.Numerous studies have evaluated the role of IL-18 within the gastrointestinal tract using genetic or complementary pharmacological tools and have revealed multiple roles in tumorigenesis. Most striking among these are the divergent roles for IL-18 in colon and gastric cancers. Here, we review our current understanding of IL-18 biology and how this applies to colorectal and gastric cancers.


Assuntos
Neoplasias Colorretais/patologia , Interleucina-18/metabolismo , Neoplasias Gástricas/patologia , Microambiente Tumoral , Animais , Caspase 1/metabolismo , Humanos
4.
J Biol Chem ; 293(9): 3168-3179, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29187603

RESUMO

The type I interferons (IFNs) are a family of cytokines with diverse biological activities, including antiviral, antiproliferative, and immunoregulatory functions. The discovery of the hormonally regulated, constitutively expressed IFNϵ has suggested a function for IFNs in reproductive tract homeostasis and protection from infections, but its intrinsic activities are untested. We report here the expression, purification, and functional characterization of murine IFNϵ (mIFNϵ). Recombinant mIFNϵ (rmIFNϵ) exhibited an α-helical fold characteristic of type I IFNs and bound to IFNα/ß receptor 1 (IFNAR1) and IFNAR2, but, unusually, it had a preference for IFNAR1. Nevertheless, rmIFNϵ induced typical type I IFN signaling activity, including STAT1 phosphorylation and activation of canonical type I IFN signaling reporters, demonstrating that it uses the JAK-STAT signaling pathway. We also found that rmIFNϵ induces the activation of T, B, and NK cells and exhibits antiviral, antiproliferative, and antibacterial activities typical of type I IFNs, albeit with 100-1000-fold reduced potency compared with rmIFNα1 and rmIFNß. Surprisingly, although the type I IFNs generally do not display cross-species activities, rmIFNϵ exhibited high antiviral activity on human cells, suppressing HIV replication and inducing the expression of known HIV restriction factors in human lymphocytes. Our findings define the intrinsic properties of murine IFNϵ, indicating that it distinctly interacts with IFNAR and elicits pathogen-suppressing activity with a potency enabling host defense but with limited toxicity, appropriate for a protein expressed constitutively in a sensitive mucosal site, such as the reproductive tract.


Assuntos
Interferon Tipo I/química , Interferon Tipo I/metabolismo , Animais , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Antivirais/química , Antivirais/metabolismo , Antivirais/farmacologia , Proliferação de Células/efeitos dos fármacos , Chlamydia/efeitos dos fármacos , Feminino , Humanos , Imunidade nas Mucosas , Interferon Tipo I/farmacologia , Camundongos , Fosforilação , Conformação Proteica em alfa-Hélice , Células RAW 264.7 , Receptores de Interferon/metabolismo , Reprodução , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
5.
PLoS Pathog ; 12(6): e1005691, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27300652

RESUMO

Legionella pneumophila is the causative agent of Legionnaires' disease, a potentially fatal lung infection. Alveolar macrophages support intracellular replication of L. pneumophila, however the contributions of other immune cell types to bacterial killing during infection are unclear. Here, we used recently described methods to characterise the major inflammatory cells in lung after acute respiratory infection of mice with L. pneumophila. We observed that the numbers of alveolar macrophages rapidly decreased after infection coincident with a rapid infiltration of the lung by monocyte-derived cells (MC), which, together with neutrophils, became the dominant inflammatory cells associated with the bacteria. Using mice in which the ability of MC to infiltrate tissues is impaired it was found that MC were required for bacterial clearance and were the major source of IL12. IL12 was needed to induce IFNγ production by lymphoid cells including NK cells, memory T cells, NKT cells and γδ T cells. Memory T cells that produced IFNγ appeared to be circulating effector/memory T cells that infiltrated the lung after infection. IFNγ production by memory T cells was stimulated in an antigen-independent fashion and could effectively clear bacteria from the lung indicating that memory T cells are an important contributor to innate bacterial defence. We also determined that a major function of IFNγ was to stimulate bactericidal activity of MC. On the other hand, neutrophils did not require IFNγ to kill bacteria and alveolar macrophages remained poorly bactericidal even in the presence of IFNγ. This work has revealed a cooperative innate immune circuit between lymphoid cells and MC that combats acute L. pneumophila infection and defines a specific role for IFNγ in anti-bacterial immunity.


Assuntos
Interferon gama/imunologia , Doença dos Legionários/imunologia , Linfócitos/imunologia , Monócitos/imunologia , Transferência Adotiva , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Imunidade Inata/imunologia , Legionella pneumophila/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Reação em Cadeia da Polimerase
6.
Infect Immun ; 85(12)2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28947652

RESUMO

Kingella kingae is a common etiological agent of pediatric osteoarticular infections. While current research has expanded our understanding of K. kingae pathogenesis, there is a paucity of knowledge about host-pathogen interactions and virulence gene regulation. Many host-adapted bacterial pathogens contain phase variable DNA methyltransferases (mod genes), which can control expression of a regulon of genes (phasevarion) through differential methylation of the genome. Here, we identify a phase variable type III mod gene in K. kingae, suggesting that phasevarions operate in this pathogen. Phylogenetic studies revealed that there are two active modK alleles in K. kingae Proteomic analysis of secreted and surface-associated proteins, quantitative PCR, and a heat shock assay comparing the wild-type modK1 ON (i.e., in frame for expression) strain to a modK1 OFF (i.e., out of frame) strain revealed three virulence-associated genes under ModK1 control. These include the K. kingae toxin rtxA and the heat shock genes groEL and dnaK Cytokine expression analysis showed that the interleukin-8 (IL-8), IL-1ß, and tumor necrosis factor responses of THP-1 macrophages were lower in the modK1 ON strain than in the modK1::kan mutant. This suggests that the ModK1 phasevarion influences the host inflammatory response and provides the first evidence of this phase variable epigenetic mechanism of gene regulation in K. kingae.


Assuntos
Metilases de Modificação do DNA/metabolismo , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Kingella kingae/crescimento & desenvolvimento , Metilases de Modificação do DNA/genética , Perfilação da Expressão Gênica , Humanos , Kingella kingae/enzimologia , Kingella kingae/genética , Filogenia , Proteoma/análise , Proteômica , Reação em Cadeia da Polimerase em Tempo Real , Regulon , Células THP-1/microbiologia , Virulência , Fatores de Virulência/biossíntese
7.
Infect Immun ; 84(5): 1346-1360, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26883593

RESUMO

Enteropathogenic Escherichia coli (EPEC) interferes with host cell signaling by injecting virulence effector proteins into enterocytes via a type III secretion system (T3SS). NleB1 is a novel T3SS glycosyltransferase effector from EPEC that transfers a single N-acetylglucosamine (GlcNAc) moiety in an N-glycosidic linkage to Arg(117) of the Fas-associated death domain protein (FADD). GlcNAcylation of FADD prevents the assembly of the canonical death-inducing signaling complex and inhibits Fas ligand (FasL)-induced cell death. Apart from the DXD catalytic motif of NleB1, little is known about other functional sites in the enzyme. In the present study, members of a library of 22 random transposon-based, in-frame, linker insertion mutants of NleB1 were tested for their ability to block caspase-8 activation in response to FasL during EPEC infection. Immunoblot analysis of caspase-8 cleavage showed that 17 mutant derivatives of NleB1, including the catalytic DXD mutant, did not inhibit caspase-8 activation. Regions of interest around the insertion sites with multiple or single amino acid substitutions were examined further. Coimmunoprecipitation studies of 34 site-directed mutants showed that the NleB1 derivatives with the E253A, Y219A, and PILN(63-66)AAAA (in which the PILN motif from residues 63 to 66 was changed to AAAA) mutations bound to but did not GlcNAcylate FADD. A further mutant derivative, the PDG(236-238)AAA mutant, did not bind to or GlcNAcylate FADD. Infection of mice with the EPEC-like mouse pathogen Citrobacter rodentium expressing NleBE253A and NleBY219A showed that these strains were attenuated, indicating the importance of residues E253 and Y219 in NleB1 virulence in vivo In summary, we identified new amino acid residues critical for NleB1 activity and confirmed that these are required for the virulence function of NleB1.


Assuntos
Análise Mutacional de DNA , Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Animais , Apoptose , Arginina/metabolismo , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidade , Elementos de DNA Transponíveis , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Proteína Ligante Fas/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Feminino , Humanos , Camundongos Endogâmicos C57BL , Mutagênese Insercional , Mutagênese Sítio-Dirigida , Processamento de Proteína Pós-Traducional , Virulência
8.
Mol Cell Biol ; 44(8): 334-343, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38975675

RESUMO

Interferon epsilon (IFNε) is a unique type I interferon (IFN) that shows distinct constitutive expression in reproductive tract epithelium. Understanding how IFNε expression is regulated is critical for the mechanism of action in protecting the mucosa from infection. Combined computational and experimental investigation of the promoter of IFNε predicted transcription factor binding sites for the ETS family of transcription factors. We demonstrate here that Ifnε is regulated by Elf3, an epithelial restricted member of the ETS family. It is co-expressed with IFNε at the epithelium of uterus, lung and intestine, and we focused on regulation of IFNε expression in the uterus. Promoter reporter studies demonstrated that Elf3 was a strong driver of Ifnε expression; knockdown of Elf3 reduced expression levels of IFNε; Elf3 regulated Ifnε expression and chromatin immunoprecipitation (ChIP) confirmed the direct binding of Elf3 to the IFNε promoter. These data show that Elf3 is important in regulating protective mucosal immunity by driving constitutive expression of IFNε to protect mucosal tissues from infection in at least three organ systems.


Assuntos
Proteínas de Ligação a DNA , Regiões Promotoras Genéticas , Fatores de Transcrição , Animais , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Regiões Promotoras Genéticas/genética , Feminino , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Camundongos , Humanos , Regulação da Expressão Gênica , Útero/metabolismo , Útero/imunologia , Mucosa/metabolismo , Mucosa/imunologia , Sítios de Ligação , Interferon Tipo I/metabolismo , Camundongos Endogâmicos C57BL , Epitélio/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Proto-Oncogênicas c-ets/genética , Pulmão/metabolismo , Pulmão/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/imunologia
9.
Methods Mol Biol ; 2823: 77-93, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39052215

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a lethal solid malignancy with many patients succumbing to the disease within 6 months of diagnosis. The mechanisms that underlie PDAC initiation and progression are poorly understood. Current treatment options are primarily limited to chemotherapy, which is often provided with palliative intent. Unfortunately, there are no robust biomarkers to guide treatment selection or monitor treatment response. This is concerning given the increasing incidence of this cancer. We and others have generated organoid models to explore the biology underlying PDAC with the goal of identifying new therapeutic targets. Here we provide protocols to generate a preclinical PDAC organoid model and methods to use these to define the proteomic landscape of this cancer.


Assuntos
Carcinoma Ductal Pancreático , Organoides , Neoplasias Pancreáticas , Proteômica , Organoides/metabolismo , Proteômica/métodos , Humanos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Animais , Proteoma , Biomarcadores Tumorais/metabolismo , Camundongos
10.
Methods Mol Biol ; 2691: 257-262, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37355552

RESUMO

The utilization of preclinical murine models of colorectal cancer (CRC) has been essential to our understanding of the onset and progression of disease. As the genetic complexity of these models evolves to better recapitulate emerging CRC subtypes, our ability to utilize these models to discover and validate novel therapeutic targets will also improve. This will be aided, in part, by the development of live animal imaging techniques, including confocal endomicroscopy for mice. Here in this chapter, we describe the combined use of standard white light endoscopy and confocal endomicroscopy thereby providing a method to rapidly image and assess changes in the colon of an individual live mouse in real time. These methods permit the generation of high-resolution cross-sectional images of the tumor microenvironment for immediate visualization of cells of interest, avoiding the need for euthanasia and tissue collection across multiple cohorts of mice.


Assuntos
Colo , Neoplasias , Animais , Camundongos , Colo/patologia , Endoscopia/métodos , Neoplasias/patologia , Microscopia Confocal/métodos , Microambiente Tumoral
11.
Nat Commun ; 14(1): 7543, 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37985757

RESUMO

Interleukin (IL-)11, an IL-6 family cytokine, has pivotal roles in autoimmune diseases, fibrotic complications, and solid cancers. Despite intense therapeutic targeting efforts, structural understanding of IL-11 signalling and mechanistic insights into current inhibitors are lacking. Here we present cryo-EM and crystal structures of the human IL-11 signalling complex, including the complex containing the complete extracellular domains of the shared IL-6 family ß-receptor, gp130. We show that complex formation requires conformational reorganisation of IL-11 and that the membrane-proximal domains of gp130 are dynamic. We demonstrate that the cytokine mutant, IL-11 Mutein, competitively inhibits signalling in human cell lines. Structural shifts in IL-11 Mutein underlie inhibition by altering cytokine binding interactions at all three receptor-engaging sites and abrogating the final gp130 binding step. Our results reveal the structural basis of IL-11 signalling, define the molecular mechanisms of an inhibitor, and advance understanding of gp130-containing receptor complexes, with potential applications in therapeutic development.


Assuntos
Citocinas , Interleucina-11 , Humanos , Interleucina-11/genética , Receptor gp130 de Citocina/genética , Interleucina-6/metabolismo , Antígenos CD/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Interleucina-6/metabolismo
12.
Cell Death Differ ; 30(5): 1155-1165, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36828915

RESUMO

Epithelial-mesenchymal transition (EMT) is a continuum that includes epithelial, partial EMT, and mesenchymal states, each of which is associated with cancer progression, invasive capabilities, and ultimately, metastasis. We used a lineage-traced sporadic model of pancreatic cancer to generate a murine organoid biobank from primary and secondary tumors, including sublines that underwent partial EMT and complete EMT. Using an unbiased proteomics approach, we found that organoid morphology predicts the EMT state, and the solid organoids are associated with a partial EMT signature. We also observed that exogenous TGFß1 induces solid organoid morphology that is associated with changes in the S100 family, complete EMT, and the formation of high-grade tumors. S100A4 may be a useful biomarker for predicting EMT state, disease progression, and outcome in patients with pancreatic cancer.


Assuntos
Neoplasias Pancreáticas , Proteínas S100 , Humanos , Animais , Camundongos , Proteínas S100/genética , Proteínas S100/metabolismo , Transição Epitelial-Mesenquimal , Neoplasias Pancreáticas/patologia , Linhagem Celular Tumoral , Neoplasias Pancreáticas
13.
Immunol Cell Biol ; 90(5): 510-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22430250

RESUMO

The mucosal system is the first line of defense against many pathogens. It is continuously exposed to dietary and microbial antigens, and thus the host must maintain a homeostatic environment between commensal microbiota and pathogenic infections. Following infections and inflammatory events, a rapid innate immune response is evoked to dampen the inflammatory processes. Type I interferons, a family of pleiotropic cytokines and major products of the innate immune response, have a key role in these early immune events at the mucosa, as reviewed here. With the emergence of new discoveries of immune cell types in mucosal tissues and their reactions to commensal and pathogenic organisms, we also review the opportunities for exciting research in this field.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Imunidade nas Mucosas , Interferon Tipo I/imunologia , Animais , Homeostase/imunologia , Humanos , Imunomodulação , Metagenoma/imunologia , Mucosa/imunologia
14.
Pathogens ; 11(4)2022 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-35456132

RESUMO

Alteration of the gut virome has been associated with colorectal cancer (CRC); however, when and how the alteration takes place has not been studied. Here, we employ a longitudinal study in mice to characterize the gut virome alteration in azoxymethane (AOM)-induced colorectal neoplasia and identify important viruses associated with tumor growth. The number and size of the tumors increased as the mice aged in the AOM treated group, as compared to the control group. Tumors were first observed in the AOM group at week 12. We observed a significantly lower alpha diversity and shift in viral profile when tumors first appeared. In addition, we identified novel viruses from the genera Brunovirus, Hpunavirus that are positively associated with tumor growth and enriched at a late time point in AOM group, whereas members from Lubbockvirus show a negative correlation with tumor growth. Moreover, network analysis revealed two clusters of viruses in the AOM virome, a group that is positively correlated with tumor growth and another that is negatively correlated with tumor growth, all of which are bacteriophages. Our findings suggest that the gut virome changes along with tumor formation and provides strong evidence of a potential role for bacteriophage in the development of colorectal neoplasia.

15.
Cell Death Differ ; 28(5): 1466-1476, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33230260

RESUMO

Chronic inflammation of the large intestine is associated with an increased risk of developing colorectal cancer (CRC), the second most common cause of cancer-related deaths worldwide. Necroptosis has emerged as a form of lytic programmed cell death that, distinct from apoptosis, triggers an inflammatory response. Dysregulation of necroptosis has been linked to multiple chronic inflammatory diseases, including inflammatory bowel disease and cancer. Here, we used murine models of acute colitis, colitis-associated CRC, sporadic CRC, and spontaneous intestinal tumorigenesis to investigate the role of necroptosis in these gastrointestinal pathologies. In the Dextran Sodium Sulfate-induced acute colitis model, in some experiments, mice lacking the terminal necroptosis effector protein, MLKL, or its activator RIPK3, exhibited greater weight loss compared to wild-type mice, consistent with some earlier reports. However, the magnitude of weight loss and accompanying inflammatory pathology upon Mlkl deletion varied substantially between independent repeats. Such variation provides a possible explanation for conflicting literature reports. Furthermore, contrary to earlier reports, we observed that genetic deletion of MLKL had no impact on colon cancer development using several mouse models. Collectively, these data do not support an obligate role for necroptosis in inflammation or cancer within the gastrointestinal tract.


Assuntos
Neoplasias do Colo/genética , Inflamação/genética , Necroptose/genética , Animais , Modelos Animais de Doenças , Camundongos
16.
Cell Death Differ ; 27(2): 742-757, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31296963

RESUMO

Gastrointestinal epithelial cells provide a selective barrier that segregates the host immune system from luminal microorganisms, thereby contributing directly to the regulation of homeostasis. We have shown that from early embryonic development Bcl-G, a Bcl-2 protein family member with unknown function, was highly expressed in gastrointestinal epithelial cells. While Bcl-G was dispensable for normal growth and development in mice, the loss of Bcl-G resulted in accelerated progression of colitis-associated cancer. A label-free quantitative proteomics approach revealed that Bcl-G may contribute to the stability of a mucin network, which when disrupted, is linked to colon tumorigenesis. Consistent with this, we observed a significant reduction in Bcl-G expression in human colorectal tumors. Our study identifies an unappreciated role for Bcl-G in colon cancer.


Assuntos
Neoplasias Colorretais/metabolismo , Inflamação/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Colite/metabolismo , Colite/patologia , Neoplasias Colorretais/patologia , Humanos , Inflamação/patologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-2/deficiência , Proteínas Proto-Oncogênicas c-bcl-2/genética
17.
Mol Immunol ; 110: 48-56, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-29174430

RESUMO

Innate lymphoid cells (ILCs) contribute to the regulation of gastrointestinal (GI) homeostasis. Over the past 15 years, there has been a large effort to dissect the mechanisms required for GI homeostasis, with a major focus on different immune cell populations and the cytokines that they produce. In contrast to T-helper (Th) cells, ILCs respond rapidly to cytokines in their microenvironment in the absence of specific antigens; however, once activated both cell populations have similar effector functions. Two effector cytokines produced by both ILC3 and Th17 cell populations, Interleukin (IL)-17 and IL-22, have taken center stage for their ability to signal directly to GI epithelial cells and promote epithelial cell survival. In this review, we outline our current understanding of ILCs in the GI tract, and focus on GI cancers associated with aberrant production of IL-17 and IL-22. We highlight evidence from both mouse and patient-based analyses and discuss how tumor cells may hijack the potential evolutionary redundancy of these two cell populations.


Assuntos
Neoplasias Gastrointestinais/imunologia , Imunidade Inata , Linfócitos/fisiologia , Linfócitos T/fisiologia , Animais , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/patologia , Neoplasias Gastrointestinais/terapia , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Camundongos , Evasão Tumoral/genética , Evasão Tumoral/imunologia , Microambiente Tumoral/imunologia
18.
Front Immunol ; 10: 608, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31024525

RESUMO

We have examined the influence of depleting plasmacytoid dendritic cells (pDC) in mice on the immune response to the gut pathogen Citrobacter rodentium, an organism that is a model for human attaching effacing pathogens such as enterohaemorraghic E. coli. A significantly higher number of C. rodentium were found in mice depleted of pDC from 7 days after infection and pDC depleted mice showed increased gut pathology and higher levels of mRNA encoding inflammatory cytokines in the colon upon infection. pDC-depletion led to a compromising of the gut mucosal barrier that may have contributed to increased numbers of C. rodentium in systemic organs. pDC-depleted mice infected with C. rodentium suffered substantial weight loss necessitating euthanasia. A number of observations suggested that this was not simply the result of dysregulation of immunity in the colon as pDC-depleted mice infected intravenously with C. rodentium also exhibited exacerbated weight loss, arguing that pDC influence systemic immune responses. Overall, these data indicate that pDC contribute at multiple levels to immunity to C. rodentium including control of bacterial numbers in the colon, maintenance of colon barrier function and regulation of immune responses to disseminated bacteria.


Assuntos
Citrobacter rodentium/imunologia , Colite/imunologia , Células Dendríticas/imunologia , Infecções por Enterobacteriaceae/imunologia , Animais , Colite/microbiologia , Colo/imunologia , Colo/microbiologia , Citocinas/imunologia , Infecções por Enterobacteriaceae/microbiologia , Inflamação/imunologia , Inflamação/microbiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL
19.
Methods Mol Biol ; 1725: 3-13, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29322404

RESUMO

A single layer of epithelial cells separates luminal antigens from the host immune system throughout the gastrointestinal tract. A breakdown in the integrity of the epithelial barrier can lead to chronic inflammation, which is associated with numerous complications including cancer. Here we describe three experimental protocols to chemically induce acute and chronic inflammatory bowel disease (IBD) symptoms and colitis-associated colorectal cancer (CRC) progression. These in vivo mouse models are based on the induction of damage to the colonic epithelium, resulting in an inflammatory and wound healing response. In addition, we outline colonoscopy procedures to monitor the onset of disease in individual live mice.


Assuntos
Colite/complicações , Neoplasias do Colo/etiologia , Modelos Animais de Doenças , Doenças Inflamatórias Intestinais/complicações , Animais , Neoplasias do Colo/patologia , Feminino , Humanos , Camundongos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa