Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Front Immunol ; 13: 988862, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36189261

RESUMO

The studies described here provide an analysis of the pathogenesis of Blau syndrome and thereby the function of NOD2 as seen through the lens of its dysfunction resulting from Blau-associated NOD2 mutations in its nucleotide-binding domain (NBD). As such, this analysis also sheds light on the role of NOD2 risk polymorphisms in the LRR domain occurring in Crohn's disease. The main finding was that Blau NOD2 mutations precipitate a loss of canonical NOD2 signaling via RIPK2 and that this loss has two consequences: first, it results in defective NOD2 ligand (MDP)-mediated NF-κB activation and second, it disrupts NOD2-mediated cross-regulation whereby NOD2 downregulates concomitant innate (TLR) responses. Strong evidence is also presented favoring the view that NOD2-mediated cross-regulation is under mechanistic control by IRF4 and that failure to up-regulate this factor because of faulty NOD2 signaling is the proximal cause of defective cross-regulation and the latter's effect on Blau syndrome inflammation. Overall, these studies highlight the role of NOD2 as a regulatory factor and thus provide additional insight into its function in inflammatory disease. Mutations in the nucleotide binding domain of the CARD15 (NOD2) gene underlie the granulomatous inflammation characterizing Blau syndrome (BS). In studies probing the mechanism of this inflammation we show here that NOD2 plasmids expressing various Blau mutations in HEK293 cells result in reduced NOD2 activation of RIPK2 and correspondingly reduced NOD2 activation of NF-κB. These in vitro studies of NOD2 signaling were accompanied by in vivo studies showing that BS-NOD2 also exhibit defects in cross-regulation of innate responses underlying inflammation. Thus, whereas over-expressed intact NOD2 suppresses TNBS-colitis, over-expressed BS-NOD2 does not; in addition, whereas administration of NOD2 ligand (muramyl dipeptide, MDP) suppresses DSS-colitis in Wild Type (WT) mice it fails to do so in homozygous or heterozygous mice bearing a NOD2 Blau mutation. Similarly, mice bearing a Blau mutation exhibit enhanced anti-collagen antibody-induced arthritis. The basis of such cross-regulatory failure was revealed in studies showing that MDP-stimulated cells bearing BS-NOD2 exhibit a reduced capacity to signal via RIPK2 as well as a reduced capacity to up-regulate IRF4, a factor shown previously to mediate NOD2 suppression of NF-κB activation. Indeed, TLR-stimulated cells bearing a Blau mutation exhibited enhanced in vitro cytokine responses that are quieted by lentivirus transduction of IRF4. In addition, enhanced anti-collagen-induced joint inflammation in mice bearing a Blau mutation was accompanied by reduced IRF4 expression in inflamed joint tissue and IRF4 expression was reduced in MDP-stimulated cells from BS patients. Thus, inflammation characterizing Blau syndrome are caused, at least in part, by faulty canonical signaling and reduce IRF4-mediated cross-regulation.


Assuntos
Artrite , Colite , Proteína Adaptadora de Sinalização NOD2/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Animais , Artrite/genética , Colite/induzido quimicamente , Citocinas/metabolismo , Células HEK293 , Humanos , Inflamação/genética , Ligantes , Camundongos , Mutação , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD2/genética , Nucleotídeos/metabolismo , Sarcoidose , Sinovite , Uveíte
2.
J Clin Invest ; 130(4): 1793-1807, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31895698

RESUMO

Bruton tyrosine kinase (BTK) is present in a wide variety of cells and may thus have important non-B cell functions. Here, we explored the function of this kinase in macrophages with studies of its regulation of the NLR family, pyrin domain-containing 3 (NLRP3) inflammasome. We found that bone marrow-derived macrophages (BMDMs) from BTK-deficient mice or monocytes from patients with X-linked agammaglobulinemia (XLA) exhibited increased NLRP3 inflammasome activity; this was also the case for BMDMs exposed to low doses of BTK inhibitors such as ibrutinib and for monocytes from patients with chronic lymphocytic leukemia being treated with ibrutinib. In mechanistic studies, we found that BTK bound to NLRP3 during the priming phase of inflammasome activation and, in doing so, inhibited LPS- and nigericin-induced assembly of the NLRP3 inflammasome during the activation phase of inflammasome activation. This inhibitory effect was caused by BTK inhibition of protein phosphatase 2A-mediated (PP2A-mediated) dephosphorylation of Ser5 in the pyrin domain of NLRP3. Finally, we show that BTK-deficient mice were subject to severe experimental colitis and that such colitis was normalized by administration of anti-IL-ß or anakinra, an inhibitor of IL-1ß signaling. Together, these studies strongly suggest that BTK functions as a physiologic inhibitor of NLRP3 inflammasome activation and explain why patients with XLA are prone to develop Crohn's disease.


Assuntos
Tirosina Quinase da Agamaglobulinemia/deficiência , Doença de Crohn , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Adolescente , Adulto , Tirosina Quinase da Agamaglobulinemia/metabolismo , Agamaglobulinemia/enzimologia , Agamaglobulinemia/genética , Agamaglobulinemia/patologia , Idoso , Idoso de 80 Anos ou mais , Animais , Criança , Doença de Crohn/enzimologia , Doença de Crohn/genética , Doença de Crohn/patologia , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/enzimologia , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Humanos , Inflamassomos/genética , Interleucina-1beta/genética , Masculino , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética
3.
Int Immunol ; 31(10): 669-683, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31132297

RESUMO

Previous studies have shown that inhibition of receptor-interacting serine/threonine kinase (RICK) (also known as RIP2) results in amelioration of experimental colitis. This role has largely been attributed to nucleotide-binding oligomerization domain 2 (NOD2) signaling since the latter is considered a major inducer of RICK activation. In this study, we explored the molecular mechanisms accounting for RICK-mediated inhibition of inflammatory bowel disease (IBD). In an initial series of studies focused on trinitrobenzene sulfonic acid (TNBS)-colitis and dextran sodium sulfate (DSS)-colitis we showed that down-regulation of intestinal RICK expression in NOD2-intact mice by intra-rectal administration of a plasmid expressing RICK-specific siRNA was accompanied by down-regulation of pro-inflammatory cytokine responses in the colon and protection of the mice from experimental colitis. Somewhat surprisingly, intra-rectal administration of RICK-siRNA also inhibited TNBS-colitis and DSS-colitis in NOD2-deficient and in NOD1/NOD2-double deficient mice. In complementary studies of humans with IBD we found that expression of RICK, cellular inhibitor of apoptosis protein 2 (cIAP2) and downstream signaling partners were markedly increased in inflamed tissue of IBD compared to controls without marked elevations of NOD1 or NOD2 expression. In addition, the increase in RICK expression correlated with disease activity and pro-inflammatory cytokine responses. These studies thus suggest that NOD1- or NOD2-independenent activation of RICK plays a major role in both murine experimental colitis and human IBD.


Assuntos
Inflamação/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Animais , Humanos , Inflamação/imunologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/imunologia , Transdução de Sinais/imunologia
4.
Front Immunol ; 9: 2566, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30455704

RESUMO

It is logical to assume that a major pro-inflammatory mechanism, i.e., the NLRP3 inflammasome would play a prominent role in the pathogenesis of the Inflammatory Bowel Disease (IBD) in humans. However, while both studies of murine models of gut disease and patients provide data that the main cytokine product generated by this inflammasome, IL-1ß, does in fact contribute to inflammation in IBD, there is no evidence that IL-1ß plays a decisive or prominent role in "ordinary" patients with IBD (Crohn's disease). On the other hand, there are several definable point mutations that result in over-active NLRP3 inflammasome activity and in these cases, the gut inflammation is driven by IL-1ß and is treatable by biologic agents that block the effects of this cytokine.


Assuntos
Doença de Crohn/patologia , Trato Gastrointestinal/patologia , Interleucina-1beta/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Humanos , Inflamação/patologia , Interleucina-18/metabolismo , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteínas de Neoplasias/genética , Transdução de Sinais/imunologia
5.
Exp Ther Med ; 16(5): 4277-4282, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30344701

RESUMO

Administration of bone marrow-derived mesenchymal stem cells (MSCs) is a possible treatment for graft-versus-host disease (GVHD) following allogeneic hematopoietic stem cell transplantation and other inflammatory conditions. To address the mechanism of immunosuppression by MSCs, in particular those derived from adipose tissue (AMSCs), AMSCs were isolated from three different mouse strains, and the suppressive capacity of the AMSCs thus obtained to suppress interferon (IFN)-γ generation in mixed lymphocyte reaction cultures serving as an in vitro model of GVHD were assessed. It was revealed that the AMSCs had a potent capacity to suppress IFN-γ production regardless of their strain of origin and that such suppression was not associated with production of interleukin-10. In addition, the results demonstrated that ß2-microglobulin (ß2m)-deficient AMSCs from ß2m-/- mice were also potent suppressor cells, verifying the fact that the mechanism underlying the suppression by AMSCs is independent of major histocompatibility complex (MHC) class I expression or MHC compatibility. As AMSCs appear to have immunosuppressive properties, AMSCs may be a useful source of biological suppressor cells for the control of GVHD in humans.

6.
Sci Transl Med ; 10(444)2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29875204

RESUMO

The LRRK2/MUC19 gene region constitutes a high-risk genetic locus for the occurrence of both inflammatory bowel diseases (IBDs) and Parkinson's disease. We show that dendritic cells (DCs) from patients with Crohn's disease (CD) and lymphoblastoid cell lines derived from patients without CD but bearing a high-risk allele (rs11564258) at this locus as heterozygotes exhibited increased LRRK2 expression in vitro. To investigate the immunological consequences of this increased LRRK2 expression, we conducted studies in transgenic mice overexpressing Lrrk2 and showed that these mice exhibited more severe colitis induced by dextran sodium sulfate (DSS) than did littermate control animals. This increase in colitis severity was associated with lamina propria DCs that showed increased Dectin-1-induced NF-κB activation and proinflammatory cytokine secretion. Colitis severity was driven by LRRK2 activation of NF-κB pathway components including the TAK1 complex and TRAF6. Next, we found that membrane-associated LRRK2 (in association with TAB2) caused inactivation of Beclin-1 and inhibition of autophagy. HCT116 colon epithelial cells lacking Beclin-1 exhibited increased LRRK2 expression compared to wild-type cells, suggesting that inhibition of autophagy potentially could augment LRRK2 proinflammatory signaling. We then showed that LRRK2 inhibitors decreased Dectin-1-induced TNF-α production by mouse DCs and ameliorated DSS-induced colitis, both in control and Lrrk2 transgenic animals. Finally, we demonstrated that LRRK2 inhibitors blocked TNF-α production by cultured DCs from patients with CD. Our findings suggest that normalization of LRRK2 activation could be a therapeutic approach for treating IBD, regardless of whether a LRRK2 risk allele is involved.


Assuntos
Autofagia , Colite/imunologia , Colite/patologia , Imunidade , Lectinas Tipo C/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Animais , Proteína Beclina-1/metabolismo , Células da Medula Óssea/metabolismo , Colo/patologia , Doença de Crohn/enzimologia , Doença de Crohn/patologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Sulfato de Dextrana , Modelos Animais de Doenças , Humanos , Inflamação/patologia , Camundongos Transgênicos , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Proteólise , Transdução de Sinais , Transcrição Gênica
7.
J Clin Invest ; 128(5): 1793-1806, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29408806

RESUMO

In these studies, we evaluated the contribution of the NLRP3 inflammasome to Crohn's disease (CD) in a kindred containing individuals having a missense mutation in CARD8, a protein known to inhibit this inflammasome. Whole exome sequencing and PCR studies identified the affected individuals as having a V44I mutation in a single allele of the T60 isoform of CARD8. The serum levels of IL-1ß in the affected individuals were increased compared with those in healthy controls, and their peripheral monocytes produced increased amounts of IL-1ß when stimulated by NLRP3 activators. Immunoblot studies probing the basis of these findings showed that mutated T60 CARD8 failed to downregulate the NLRP3 inflammasome because it did not bind to NLRP3 and inhibit its oligomerization. In addition, these studies showed that mutated T60 CARD8 exerted a dominant-negative effect by its capacity to bind to and form oligomers with unmutated T60 or T48 CARD8 that impeded their binding to NLRP3. Finally, inflammasome activation studies revealed that intact but not mutated CARD8 prevented NLRP3 deubiquitination and serine dephosphorylation. CD due to a CARD8 mutation was not effectively treated by anti-TNF-α, but did respond to IL-1ß inhibitors. Thus, patients with anti-TNF-α-resistant CD may respond to this treatment option.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/imunologia , Doença de Crohn/imunologia , Inflamassomos/imunologia , Mutação com Perda de Função , Monócitos/imunologia , Mutação de Sentido Incorreto , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Proteínas de Neoplasias/imunologia , Substituição de Aminoácidos , Proteínas Adaptadoras de Sinalização CARD/genética , Doença de Crohn/genética , Doença de Crohn/patologia , Feminino , Células HEK293 , Humanos , Inflamassomos/genética , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Masculino , Monócitos/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteínas de Neoplasias/genética , Fosforilação/genética , Fosforilação/imunologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Ubiquitinação/genética , Ubiquitinação/imunologia , Sequenciamento Completo do Genoma
8.
Oncol Rep ; 38(1): 449-455, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28535011

RESUMO

To evaluate systemic immunity associated with tumor growth limited to a subcutaneous site versus growth proceeding at multiple tumor sites, we established syngeneic mouse subcutaneous and pulmonary tumor models by local subcutaneous and intravenous injection of colon carcinoma CT26 cells. We found that splenic myeloid-derived suppressor cell (MDSC) levels were significantly increased in the subcutaneous tumor model but not in the pulmonary tumor model. Furthermore, both CD4+ and CD8+ T cells as well as CD4+ Foxp3+ T cells were significantly decreased in the subcutaneous tumor model and were largely unchanged in the pulmonary tumor model. In addition, the subcutaneous model, but not the pulmonary model, displayed a Th1 polarization bias. This bias was characterized by decreased IL-4, IL-9, and IL-10 production, whereas the pulmonary model displayed increased production of IL-10. These results suggest that the mode of tumor development has differential effects on systemic immunity that may, in turn, influence approaches to treatment of cancer patients.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/imunologia , Neoplasias Pulmonares/imunologia , Células Supressoras Mieloides/imunologia , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/mortalidade , Neoplasias do Colo/patologia , Feminino , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Interleucina-9/metabolismo , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/secundário , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias/métodos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Tela Subcutânea/imunologia , Tela Subcutânea/patologia , Células Th1/imunologia , Transplante Isogênico/métodos
9.
Appl Biochem Biotechnol ; 180(8): 1559-1573, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27406037

RESUMO

Transplantation of hematopoietic stem and progenitor cells (HSCs) i.e., self-renewing cells that retain multipotentiality, is now a widely performed therapy for many hematopoietic diseases. However, these cells are present in low number and are subject to replicative senescence after extraction; thus, the acquisition of sufficient numbers of cells for transplantation requires donors able to provide repetitive blood samples and/or methods of expanding cell numbers without disturbing cell multipotentiality. Previous studies have shown that HSCs maintain their multipotentiality and self-renewal activity if TCF3 transcription function is blocked under B cell differentiating conditions. Taking advantage of this finding to devise a new approach to HSC expansion in vitro, we constructed an episomal expression vector that specifically targets and transiently represses the TCF3 gene. This consisted of a vector encoding a transcription activator-like effector (TALE) fused to a Krüppel-associated box (KRAB) repressor. We showed that this TALE-KRAB vector repressed expression of an exogenous reporter gene in HEK293 and COS-7 cell lines and, more importantly, efficiently repressed endogenous TCF3 in a human B lymphoma cell line. These findings suggest that this vector can be used to maintain multipotentiality in HSC being subjected to a long-term expansion regimen prior to transplantation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Marcação de Genes , Proteínas Repressoras/metabolismo , Efetores Semelhantes a Ativadores de Transcrição/metabolismo , Animais , Células COS , Chlorocebus aethiops , Deleção de Genes , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Luciferases/metabolismo , Proteínas Luminescentes/metabolismo , Plasmídeos/metabolismo , Transfecção , Proteína Vermelha Fluorescente
10.
Immunol Rev ; 260(1): 249-60, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24942694

RESUMO

The discovery that polymorphisms in the NOD2 (nucleotide-binding oligomerization domain containing 2) gene are associated with a greatly increased risk for the development of Crohn's disease has provided a means to achieve a deeper understanding of the dysregulation of mucosal immune responses to the commensal intestinal organisms that is thought to underlie this disease. NOD2 is a NOD-like receptor (NLR) family member that senses and responds to bacterial wall peptides; thus, the most widely held view of the relation of the NOD2 polymorphisms with Crohn's disease is that these polymorphisms lead to deficient immune responses to gut bacteria, and these, in turn, lead to quantitative or qualitative changes in the bacterial population in the gut lumen or lamina propria that cause inflammation at this site. Initially, this view was based mainly on the observation that defective NOD2 function can result in reduced α-defensin production by intestinal Paneth cells and that such impairment leads to loss of host defense against gut bacteria. In this review, we reconsider this possibility and marshal evidence that it is not in fact likely to be a prime element of Crohn's disease causation. More recently, evidence has been accumulating that the NOD2 dysfunction leads to Crohn's inflammation by inducing changes in the gut microbiome that influence immune effector or regulatory function. We review the strengths and weaknesses of this emerging hypothesis. Finally, we consider the possibility that NOD2 dysfunction can lead to inflammation because of a second and somewhat overlooked aspect of its function, that as an immunoregulator of innate immune responses. In particular, we review the body of evidence that NOD2 stimulation activates a cross-tolerance response that downregulates and thus prevents excessive TLR responses that cause Crohn's inflammation.


Assuntos
Doença de Crohn/genética , Doença de Crohn/imunologia , Predisposição Genética para Doença , Proteína Adaptadora de Sinalização NOD2/genética , Polimorfismo Genético , Animais , Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Defensinas/biossíntese , Estudos de Associação Genética , Humanos , Imunidade Inata , Imunomodulação , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/microbiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Microbiota , Proteína Adaptadora de Sinalização NOD2/deficiência , Proteína Adaptadora de Sinalização NOD2/metabolismo , Celulas de Paneth/metabolismo
11.
J Immunol ; 192(8): 3969-80, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24634488

RESUMO

Spontaneous amelioration of inflammation (often accompanied by fibrosis) is a well-known, but poorly understood, outcome of many chronic inflammatory processes. We studied this phenomenon in a chronic trinitrobenzene sulfonic acid-induced colitis model, an experimental colitis in mice that we showed to ultimately undergo spontaneous resolution, despite continued trinitrobenzene sulfonic acid stimulation. Analysis of the mechanism of this resolution revealed that it was critically dependent on IL-13 activation of STAT6, followed by phosphorylation (inactivation) of glycogen synthase kinase-3ß, at least in part via STAT6 induction of p38 MAPK. Such glycogen synthase kinase-3ß inactivation causes changes in CREB and p65 DNA-binding activity that favors decreased proinflammatory IL-17 production and increased anti-inflammatory IL-10 production. Thus, in this case, IL-13 acts as a molecular switch that leads to resolution of inflammation.


Assuntos
Colite/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Interleucina-13/metabolismo , Animais , Doença Crônica , Colite/induzido quimicamente , Colite/genética , Colite/imunologia , Colite/patologia , Citocinas/biossíntese , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Interleucina-13/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fosforilação , Transdução de Sinais , Ácido Trinitrobenzenossulfônico/efeitos adversos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Gut Microbes ; 2(1): 61-5, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21637021

RESUMO

Infection of gastric epithelial cells with Helicobacter pylori (H. pylori) induces a complex array of host protective immune responses. The best known are the adaptive T helper type 1 and type 17 responses that are induced in the gastric lamina propria by antigen-presenting cells via presentation of H. pylori antigens to CD4(+) T cells. Recently, it has become apparent that innate immune responses are also induced by H.pylori infection, both in epithelial cells and in underlying antigen-presenting cells. One important component of these innate responses involves the activity of NOD1, an intra-cellular sensor of peptides derived from the peptidoglycan component of the bacterial cell wall. In this review, we discuss our recent work showing that the signaling pathway utilized by NOD1 results in the generation of type I interferon and that this cytokine mediates both chemokine and cytokine responses that regulate the severity of gastric H. pylori infection.


Assuntos
Mucosa Gástrica/imunologia , Infecções por Helicobacter/fisiopatologia , Helicobacter pylori/imunologia , Interferon Tipo I/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Transdução de Sinais , Animais , Mucosa Gástrica/microbiologia , Mucosa Gástrica/fisiopatologia , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Humanos , Inflamação/imunologia , Interferon Tipo I/genética , Camundongos , Proteína Adaptadora de Sinalização NOD1/genética
13.
Int J Inflam ; 2010: 476482, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21152124

RESUMO

Infection of the stomach with Helicobacter pylori is an important risk factor for gastritis, peptic ulcer, and gastric carcinoma. Although it has been well established that persistent colonization by H. pylori is associated with adaptive Th1 responses, the innate immune responses leading to these Th1 responses are poorly defined. Recent studies have shown that the activation of nucleotide-binding oligomerization domain 1 (NOD1) in gastric epithelial cells plays an important role in innate immune responses against H. pylori. The detection of H. pylori-derived ligands by cytosolic NOD1 induces several host defense factors, including antimicrobial peptides, cytokines, and chemokines. In this paper, we review the molecular mechanisms by which NOD1 contributes to mucosal host defense against H. pylori infection of the stomach.

14.
Curr Protoc Immunol ; Chapter 10: Unit 10.17E.1-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21053301

RESUMO

This unit describes a method for in vivo delivery of oligonucleotides or plasmids using the hemagglutinating virus of Japan envelope (HVJ-E), an inactivated Sendai virus particle, as a delivery system. Viral transfection methods generally show a higher transfection efficiency than nonviral methods for the delivery of genes to cells. However, in using these methods one must bear in mind that the introduction of a virus particle into a host carries a risk for leukemia induction and for creation of disturbances in immune function due to cytotoxicity.


Assuntos
Oligonucleotídeos/genética , Plasmídeos/genética , Vírus Sendai/genética , Transfecção/métodos , Proteínas do Envelope Viral/genética , Animais , Vetores Genéticos/efeitos adversos , Vetores Genéticos/genética , Humanos , Leucemia/virologia , Camundongos , Oligonucleotídeos/efeitos adversos , Plasmídeos/efeitos adversos
15.
Immunity ; 33(3): 313-25, 2010 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-20870174

RESUMO

The molecular mechanisms underlying retinoic acid (RA) augmentation of T cell receptor (TCR) and transforming growth factor-ß (TGF-ß)-induced Foxp3 transcription and inhibition of the latter by cytokines such as IL-27 were here shown to be related processes involving modifications of baseline (TGF-ß-induced) phosphorylated Smad3 (pSmad3) binding to a conserved enhancer region (enhancer I). RA augmentation involved the binding of retinoic acid receptor (RAR) and retinoid X receptor (RXR) to a dominant site in enhancer I and a subordinate site in the promoter. This led to increased histone acetylation in the region of the Smad3 binding site and increased binding of pSmad3. Cytokine (IL-27) inhibition involved binding of pStat3 to a gene silencer in a second conserved enhancer region (enhancer II) downstream from enhancer I; this led to loss of pSmad3 binding to enhancer I. Thus, control of accessibility and binding of pSmad3 provides a common framework for positive and negative regulation of TGF-ß-induced Foxp3 transcription.


Assuntos
Elementos Facilitadores Genéticos , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Proteína Smad3/metabolismo , Animais , Sítios de Ligação , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/fisiologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Receptores X de Retinoides/fisiologia , Fator de Transcrição AP-1/fisiologia , Transcrição Gênica , Fator de Crescimento Transformador beta/fisiologia , Tretinoína/farmacologia
16.
Curr Opin Gastroenterol ; 26(4): 310-7, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20473158

RESUMO

PURPOSE OF REVIEW: To encapsulate our current understanding of the proinflammatory cytokines responsible for the inflammation underlying Crohn's disease and the prospect of using this information to devise therapy for this condition based on inhibition of these cytokines. RECENT FINDINGS: Current research is shedding new light on the role of both T helper cell (Th)1 and Th17 responses in the pathogenesis of Crohn's disease. Initial studies conducted a decade ago highlighted the view that Crohn's disease inflammation is caused by an interleukin-12-driven Th1 response, which resulted in the generation of interferon-gamma, which then served as the main inflammatory mediator. In recent years, however, this view has been largely eclipsed by studies, conducted mainly in murine models, showing that a Th17 response is the main cause of Crohn's disease inflammation through the production of interleukin-17. Now, a somewhat more balanced view is emerging, which holds that interferon-gamma is still a major proinflammatory cytokine in Crohn's disease, although it may arise from both the Th1 and Th17-mediated responses at different phases of the inflammatory process. SUMMARY: The new findings continue to support the idea that anti-interleukin-12p40, an antibody that inhibits both the Th1 and Th17 response, is logically the most potent anticytokine for the treatment of Crohn's disease.


Assuntos
Doença de Crohn/imunologia , Citocinas/imunologia , Animais , Citocinas/antagonistas & inibidores , Citocinas/fisiologia , Modelos Animais de Doenças , Humanos , Interleucina-12/antagonistas & inibidores , Interleucina-12/imunologia , Interleucina-12/fisiologia , Interleucina-17/antagonistas & inibidores , Interleucina-17/imunologia , Interleucina-17/fisiologia , Interleucina-23/antagonistas & inibidores , Interleucina-23/imunologia , Interleucina-23/fisiologia , Interleucinas/antagonistas & inibidores , Interleucinas/imunologia , Interleucinas/fisiologia , Linfotoxina-alfa/imunologia , Células Th1/imunologia , Interleucina 22
17.
J Clin Invest ; 120(5): 1645-62, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20389019

RESUMO

Nucleotide-binding oligomerization domain 1 (NOD1) is an intracellular epithelial cell protein known to play a role in host defense at mucosal surfaces. Here we show that a ligand specific for NOD1, a peptide derived from peptidoglycan, initiates an unexpected signaling pathway in human epithelial cell lines that results in the production of type I IFN. Detailed analysis revealed the components of the signaling pathway. NOD1 binding to its ligand triggered activation of the serine-threonine kinase RICK, which was then able to bind TNF receptor-associated factor 3 (TRAF3). This in turn led to activation of TANK-binding kinase 1 (TBK1) and IkappaB kinase epsilon (IKKepsilon) and the subsequent activation of IFN regulatory factor 7 (IRF7). IRF7 induced IFN-beta production, which led to activation of a heterotrimeric transcription factor complex known as IFN-stimulated gene factor 3 (ISGF3) and the subsequent production of CXCL10 and additional type I IFN. In vivo studies showed that mice lacking the receptor for IFN-beta or subjected to gene silencing of the ISGF3 component Stat1 exhibited decreased CXCL10 responses and increased susceptibility to Helicobacter pylori infection, phenotypes observed in NOD1-deficient mice. These studies thus establish that NOD1 can activate the ISGF3 signaling pathway that is usually associated with protection against viral infection to provide mice with robust type I IFN-mediated protection from H. pylori and possibly other mucosal infections.


Assuntos
Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Interferon Tipo I/metabolismo , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Fator Gênico 3 Estimulado por Interferon/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Transdução de Sinais , Animais , Quimiocinas/metabolismo , Feminino , Humanos , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/química , Ligação Proteica , Células Th1/citologia
18.
Curr Mol Med ; 9(6): 740-50, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19689301

RESUMO

The IL-13Ralpha2 receptor is a high affinity receptor for IL-13 that is used only by IL-13 and is quite distinct from the well known IL-13Ralpha1 receptor that IL-13 shares with IL-4. It was widely considered to be a secreted receptor that is devoid of signaling activity and functional only as a decoy receptor that retarded signaling via IL-13Ralpha1. In recent studies, however, it was shown to be capable of robust signaling that results in production of TGF-beta1 and through the latter cytokine, the induction of fibrosis occuring in various experimental inflammatory states. Thus, in initial studies, IL-13 signaling via IL-13Ralpha2 was shown to play an important role in the fibrosis developing in both oxazolone colitis and bleomycin-induced pulmonary fibrosis; later, it was also shown to be critical to the development of fibrosis in a model of chronic colitis induced by trinitrobenzene sulphonic acid (TNBS). These studies suggest that blockade of IL-13 or IL-13Ralpha2 signaling might be an excellent target for the prevention of inflammation-associated fibrosis. A second role of IL-13 signaling via IL-13Ralpha2 is in tumor immune surveillance. Thus, in the relevant studies it was shown that NKT cells stimulated by tumor antigens produce IL-13 that then acts on Gr-1 cells to induce TGF-beta1; the latter then inhibits CD8+ T cells engaged in tumor immune surveillance; in effect, then, receptor signaling favors tumor growth. In addition to its signaling function and the induction of TGF-beta1, IL-13Ralpha2 also influences IL-13Ralpha1 signaling in complex ways; thus, IL-13Ralpha2 emerges as a important component of IL-13 signaling, not only in its own right but also in its possible effect on its companion receptor.


Assuntos
Trato Gastrointestinal/patologia , Vigilância Imunológica/imunologia , Subunidade alfa2 de Receptor de Interleucina-13/imunologia , Neoplasias/imunologia , Transdução de Sinais/imunologia , Animais , Fibrose , Humanos
19.
Immunity ; 30(6): 860-74, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19501001

RESUMO

Missense mutations of the gene encoding NLRP3 are associated with autoinflammatory disorders characterized with excessive production of interleukin-1beta (IL-1beta). Here we analyzed the immune responses of gene-targeted mice carrying a mutation in the Nlrp3 gene equivalent to the human mutation associated with Muckle-Wells Syndrome. We found that antigen-presenting cells (APCs) from such mice produced massive amounts of IL-1beta upon stimulation with microbial stimuli in the absence of ATP. This was likely due to a diminished inflammasome activation threshold that allowed a response to the small amount of agonist. Moreover, the Nlrp3 gene-targeted mice exhibited skin inflammation characterized by neutrophil infiltration and a Th17 cytokine-dominant response, which originated from hematopoietic cells. The inflammation of Nlrp3 gene-targeted mice resulted from excess IL-1beta production from APCs, which augmented Th17 cell differentiation. These results demonstrate that the NLRP3 mutation leads to inflammasome hyperactivation and consequently Th17 cell-dominant immunopathology in autoinflammation.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Proteínas de Transporte/metabolismo , Inflamação/genética , Interleucina-1beta/imunologia , Macrófagos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Trifosfato de Adenosina/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Células Apresentadoras de Antígenos/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Diferenciação Celular/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Inflamação/imunologia , Interleucina-18/biossíntese , Interleucina-18/imunologia , Interleucina-1beta/biossíntese , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Mutação/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Pele/imunologia , Pele/metabolismo , Pele/patologia , Linfócitos T Auxiliares-Indutores/metabolismo
20.
Gastroenterology ; 135(6): 2003-13, 2013.e1-7, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18938165

RESUMO

BACKGROUND & AIMS: Previous studies have shown that fibrosis developing in chronic experimental colitis is driven by interleukin (IL)-13 signaling via IL-13R alpha(2) and the production of transforming growth factor (TGF)-beta1. In the present study, we sought to determine the fibrogenic downstream events set in motion by such signaling. METHODS: Experimental colitis with late-onset intestinal fibrosis was induced by weekly intrarectal administration of trinitrobenzene sulfonic acid (TNBS) to BALB/c mice. Blockade of IL-13 signaling via IL-13R alpha(2) and TGF-beta1 signaling was achieved by the administration of small interfering RNA or decoy oligonucleotides that target promoter sequences of signaling components of these receptors. Effects of blockade were determined by enzyme-linked immunosorbent assay or Western blotting detecting specific key fibrogenic factors and by measurement of collagen production. RESULTS: Initially, we showed that abrogation of IL-13 activity via blockade of IL-13R alpha(2) and TGF-beta1 signaling results in severe inhibition of expression of colonic insulin-like growth factor (IGF)-I and early growth response gene (Egr)-1, factors known to initiate and sustain fibrosis. We then showed that Egr-1 was necessary early in the fibrotic process for caspase-mediated apoptosis of myofibroblasts and the production of urokinase plasminogen activator, a protein that enhances TGF-beta1 activation. Finally, we showed that IGF-I (together with TGF-beta1) acts later in the process to stimulate myofibroblasts to deposit collagen in the colon. CONCLUSIONS: These studies establish that IL-13 signaling via the IL-13R alpha(2) is a key initiation point for a complex fibrotic program in the colon consisting of TGF-beta1 activation, IGF-I and Egr-1 expression, myofibroblast apoptosis, and myofibroblast production of collagen.


Assuntos
Colite/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/metabolismo , Interleucina-13/metabolismo , Animais , Apoptose , Western Blotting , Células Cultivadas , Colite/induzido quimicamente , Colite/patologia , Colo/metabolismo , Colo/patologia , Modelos Animais de Doenças , Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Proteína 1 de Resposta de Crescimento Precoce/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose/metabolismo , Fibrose/patologia , Regulação da Expressão Gênica , Fator de Crescimento Insulin-Like I/biossíntese , Fator de Crescimento Insulin-Like I/genética , Subunidade alfa2 de Receptor de Interleucina-13/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase , RNA/genética , Transdução de Sinais , Fator de Crescimento Transformador beta1/biossíntese , Fator de Crescimento Transformador beta1/genética , Ácido Trinitrobenzenossulfônico/toxicidade , Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...