Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nat Immunol ; 13(10): 947-53, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22922363

RESUMO

Microbiota are essential for weight gain in mouse models of diet-induced obesity (DIO), but the pathways that cause the microbiota to induce weight gain are unknown. We report that mice deficient in lymphotoxin, a key molecule in gut immunity, were resistant to DIO. Ltbr(-/-) mice had different microbial community composition compared to their heterozygous littermates, including an overgrowth of segmented filamentous bacteria (SFB). Furthermore, cecal transplantation conferred leanness to germ-free recipients. Housing Ltbr(-/-) mice with their obese siblings rescued weight gain in Ltbr(-/-) mice, demonstrating the communicability of the obese phenotype. Ltbr(-/-) mice lacked interleukin 23 (IL-23) and IL-22, which can regulate SFB. Mice deficient in these pathways also resisted DIO, demonstrating that intact mucosal immunity guides diet-induced changes to the microbiota to enable obesity.


Assuntos
Imunidade nas Mucosas , Receptor beta de Linfotoxina/fisiologia , Linfotoxina-alfa/fisiologia , Obesidade , Animais , Bactérias/crescimento & desenvolvimento , Bactérias/imunologia , Ceco/microbiologia , Ceco/transplante , Dieta , Metabolismo Energético , Vida Livre de Germes , Interleucina-23/deficiência , Interleucina-23/fisiologia , Interleucinas/deficiência , Interleucinas/fisiologia , Receptor beta de Linfotoxina/genética , Linfotoxina-alfa/deficiência , Linfotoxina-alfa/genética , Metagenoma , Camundongos , Camundongos Knockout , Obesidade/etiologia , Obesidade/imunologia , Obesidade/metabolismo , Aumento de Peso/imunologia , Interleucina 22
2.
Immunity ; 32(3): 403-13, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20226692

RESUMO

Epithelial cells provide the first line of defense against mucosal pathogens; however, their coordination with innate and adaptive immune cells is not well understood. Using mice with conditional gene deficiencies, we found that lymphotoxin (LT) from innate cells expressing transcription factor RORgammat, but not from adaptive T and B cells, was essential for the control of mucosal C. rodentium infection. We demonstrate that the LTbetaR signaling was required for the regulation of the early innate response against infection. Furthermore, we have revealed that LTbetaR signals in gut epithelial cells and hematopoietic-derived cells coordinate to protect the host from infection. We further determined that LTbetaR signaling in intestinal epithelial cells was required for recruitment of neutrophils to the infection site early during infection via production of CXCL1 and CXCL2 chemokines. These results support a model wherein LT from RORgammat(+) cells orchestrates the innate immune response against mucosal microbial infection.


Assuntos
Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Células Epiteliais/imunologia , Imunidade Inata , Mucosa Intestinal/imunologia , Receptor beta de Linfotoxina/imunologia , Transdução de Sinais , Imunidade Adaptativa , Animais , Células da Medula Óssea/imunologia , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
Cytokine ; 101: 39-47, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-27623349

RESUMO

Lymphotoxin (LT) is a member of the tumor necrosis factor (TNF) superfamily of cytokines which serves multiple functions, including the control of lymphoid organ development and maintenance, as well as regulation of inflammation and autoimmunity. Although the role of LT in organogenesis and maintenance of lymphoid organs is well established, the contribution of LT pathway to homeostasis of lymphoid organs during the immune response to pathogens is less understood. In this review, we highlight recent advances on the role of LT pathway in antiviral immune responses. We discuss the role of LT signaling in lymphoid organ integrity, type I IFN production and regulation of protection and immunopathology during viral infections. We further discuss the potential of therapeutic targeting LT pathway for controlling immunopathology and antiviral protection.


Assuntos
Antivirais/imunologia , Tecido Linfoide/fisiologia , Linfotoxina-alfa/imunologia , Viroses/imunologia , Animais , Autoimunidade , Homeostase/imunologia , Humanos , Inflamação , Interferon Tipo I/biossíntese , Interferon Tipo I/imunologia , Tecido Linfoide/imunologia , Receptor beta de Linfotoxina/imunologia , Linfotoxina-alfa/efeitos dos fármacos , Linfotoxina-alfa/genética , Camundongos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Viroses/tratamento farmacológico , Viroses/fisiopatologia
4.
PLoS Pathog ; 10(5): e1004142, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24854422

RESUMO

Septic pneumonias resulting from bacterial infections of the lung are a leading cause of human death worldwide. Little is known about the capacity of CD8 T cell-mediated immunity to combat these infections and the types of effector functions that may be most effective. Pneumonic plague is an acutely lethal septic pneumonia caused by the Gram-negative bacterium Yersinia pestis. We recently identified a dominant and protective Y. pestis antigen, YopE69-77, recognized by CD8 T cells in C57BL/6 mice. Here, we use gene-deficient mice, Ab-mediated depletion, cell transfers, and bone marrow chimeric mice to investigate the effector functions of YopE69-77-specific CD8 T cells and their relative contributions during pulmonary Y. pestis infection. We demonstrate that YopE69-77-specific CD8 T cells exhibit perforin-dependent cytotoxicity in vivo; however, perforin is dispensable for YopE69-77-mediated protection. In contrast, YopE69-77-mediated protection is severely impaired when production of TNFα and IFNγ by CD8 T cells is simultaneously ablated. Interestingly, TNFα is absolutely required at the time of challenge infection and can be provided by either T cells or non-T cells, whereas IFNγ provided by T cells prior to challenge appears to facilitate the differentiation of optimally protective CD8 T cells. We conclude that cytokine production, not cytotoxicity, is essential for CD8 T cell-mediated control of pulmonary Y. pestis infection and we suggest that assays detecting Ag-specific TNFα production in addition to antibody titers may be useful correlates of vaccine efficacy against plague and other acutely lethal septic bacterial pneumonias.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade Celular/genética , Interferon gama/fisiologia , Peste/imunologia , Pneumonia Bacteriana/imunologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Yersinia pestis/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Interferon gama/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peste/complicações , Peste/genética , Pneumonia Bacteriana/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Fator de Necrose Tumoral alfa/genética
5.
Blood ; 116(18): 3456-64, 2010 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-20634375

RESUMO

Secondary lymphoid organs provide a unique microenvironment for generation of immune responses. Using a cell type-specific conditional knockout approach, we have dissected contributions of tumor necrosis factor (TNF) produced by B cells (B-TNF) or T cells (T-TNF) to the genesis and homeostatic organization of secondary lymphoid organs. In spleen, lymph nodes and Peyer patches, the cellular source of TNF, and its molecular form (soluble versus membrane-bound) appeared distinct. In spleen, in addition to major B-TNF signal, a complementary T-TNF signal contributed to the microstructure. In contrast, B-TNF predominantly controlled the development of follicular dendritic cells and B-cell follicles in Peyer patches. In lymph nodes, cooperation between TNF expressed by B and T cells was necessary for the maintenance of microarchitecture and for generation of an efficient humoral immune response. Unexpectedly, soluble but not membrane TNF expressed by B cells was essential for the organization of the secondary lymphoid organs. Thus, the maintenance of each type of secondary lymphoid organ is orchestrated by distinct contributions of membrane-bound and soluble TNF produced by B and T lymphocytes.


Assuntos
Linfócitos B/imunologia , Linfonodos/imunologia , Nódulos Linfáticos Agregados/imunologia , Baço/imunologia , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Regulação da Expressão Gênica , Imunidade Humoral , Linfonodos/citologia , Linfonodos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nódulos Linfáticos Agregados/citologia , Baço/citologia , Baço/ultraestrutura , Fator de Necrose Tumoral alfa/genética
6.
Mucosal Immunol ; 14(3): 703-716, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33214656

RESUMO

Innate lymphoid cells (ILCs) are a heterogeneous family of immune regulators that protect against mucosal pathogens but can also promote intestinal pathology. Although the plasticity between ILCs populations has been described, the role of mucosal pathogens in inducing ILC conversion leading to intestinal pathology remains unclear. Here we demonstrate that IFNγ-producing ILCs are responsible for promoting intestinal pathology in a mouse model of enterocolitis caused by Campylobacter jejuni, a common human enteric pathogen. Phenotypic analysis revealed a distinct population of IFNγ-producing NK1.1-T-bet+ILCs that accumulated in the intestine of C. jejuni-infected mice. Adoptive transfer experiments demonstrated their capacity to promote intestinal pathology. Inactivation of T-bet in NKp46+ ILCs ameliorated disease. Transcriptome analysis and cell-fate mapping experiments revealed that IFNγ-producing NK1.1-ILCs correspond to ILC1 profile and develop from RORγt+ progenitors. Collectively, we identified a distinct population of NK1.1-ex-ILC3s that promotes intestinal pathology through IFNγ production in response to C. jejuni infection.


Assuntos
Infecções por Campylobacter/imunologia , Campylobacter jejuni/fisiologia , Colite/imunologia , Intestinos/imunologia , Linfócitos/imunologia , Animais , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Humanos , Imunidade Inata , Interferon gama/metabolismo , Interleucina-10/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Células Th1/imunologia
7.
Gastroenterology ; 136(2): 694-704.e4, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18952083

RESUMO

BACKGROUND & AIMS: The ability of the liver to regenerate hepatic mass is essential to withstanding liver injury. The process of liver regeneration is tightly regulated by distinct signaling cascades involving components of the innate immune system, cytokines, and growth factors. However, the role of the adaptive immune system in regulation of liver regeneration is not well-defined. The role of adaptive immune system in liver regeneration was investigated in lymphocyte-deficient mice and in conditional lymphotoxin-deficient mice. METHODS: A model of liver regeneration after 70% partial hepatectomy was used, followed by examination of liver pathology, survival, DNA synthesis, and cytokine expression. RESULTS: We found that mice deficient in T cells show a reduced capacity for liver regeneration following partial hepatectomy. Furthermore, surface lymphotoxin, provided by T cells, is critical for liver regeneration. Mice specifically deficient in T-cell lymphotoxin had increased liver damage and a reduced capacity to initiate DNA synthesis after partial hepatectomy. Transfer of splenocytes from wild-type but not lymphotoxin-deficient mice improved liver regeneration in T cell-deficient mice. We found that an agonistic antibody against the lymphotoxin beta receptor was able to facilitate liver regeneration by reducing liver injury, increasing interleukin-6 production, hepatocyte DNA synthesis, and survival of lymphocyte-deficient (Rag) mice after partial hepatectomy. CONCLUSIONS: The adaptive immune system directly regulates liver regeneration via a T cell-derived lymphotoxin axis, and pharmacological stimulation of lymphotoxin beta receptor might represent a novel therapeutic approach to improve liver regeneration.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Regeneração Hepática/fisiologia , Linfotoxina-alfa/metabolismo , Linfotoxina-beta/metabolismo , Animais , Hepatectomia , Sistema Imunitário/fisiologia , Interleucina-6/metabolismo , Fígado/citologia , Fígado/metabolismo , Fígado/cirurgia , Receptor beta de Linfotoxina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Baço/citologia
8.
Cancer Lett ; 255(2): 255-62, 2007 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-17562356

RESUMO

We have identified RAP80/UIMC1, the protein highly expressed in testis, as a new cancer-associated antigen. Sera from 5% to 10% of patients with different types of cancer contain specific antibodies to RAP80/UIMC1. In order to investigate the possible reasons for RAP80/UIMC1 immunogenicity, we characterized its numerous splice isoforms and mapped immunogenic regions of the protein. The majority of RAP80/UIMC1 transcripts was detected both in normal tissues and in colon tumors. There are several RAP80/UIMC1 isoforms that are predominantly expressed in testis, however we did not observe elevated expression of these transcripts in tumors from seropositive patients. We mapped the major immunogenic region of RAP80/UIMC1 to the central part of the protein encoded by exon 9 which is present in a number of ubiquitous splice forms. Thus, based on our data, autoreactivity to RAP80/UIMC1 is related to reasons other than overexpression or tumor-specific splicing.


Assuntos
Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Proteínas de Transporte/metabolismo , Neoplasias/imunologia , Proteínas Nucleares/metabolismo , Testículo/metabolismo , Processamento Alternativo , Sequência de Aminoácidos , Anticorpos Antineoplásicos/sangue , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Proteínas de Ligação a DNA , Mapeamento de Epitopos , Epitopos de Linfócito B/genética , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito B/metabolismo , Feminino , Chaperonas de Histonas , Humanos , Masculino , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia
9.
Adv Healthc Mater ; 6(14)2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28481012

RESUMO

Accumulating evidence suggests that ischemia-reperfusion-induced injury is associated with the formation of reactive oxygen species (ROS). This study demonstrates the therapeutic effectiveness of novel europium-doped cerium oxide nanoparticles (Eu-doped Ceria NPs) as ROS scavengers in a mouse model of intestinal ischemia-reperfusion-induced injury. An increased production of superoxide radicals is detected in the intestine throughout the ischemia stage and again after initiating reperfusion. These changes in superoxide radical formation are associated with the induction of inflammatory cytokines in the intestine. This study further shows that Eu-Ceria NPs exhibit superoxide scavenging activity in vitro. Importantly, administration of Eu-Ceria NPs into the intestinal lumen during the onset of ischemia effectively blocks superoxide accumulation, reduces the expression of IL-1b, and ameliorates the intestinal pathology. These results suggest that early increased production of ROS during the ischemia-reperfusion promotes intestinal pathology and that mucosal delivery of Eu-Ceria NPs may be a potential therapeutic approach to block ROS accumulation and ameliorate the severity of intestinal disease.


Assuntos
Cério , Sequestradores de Radicais Livres , Enteropatias/tratamento farmacológico , Mucosa Intestinal/metabolismo , Nanopartículas , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Cério/química , Cério/farmacologia , Európio , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/farmacologia , Interleucina-1beta/metabolismo , Enteropatias/metabolismo , Enteropatias/patologia , Intestinos/patologia , Camundongos , Nanopartículas/química , Nanopartículas/uso terapêutico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
10.
J Vis Exp ; (111)2016 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-27213580

RESUMO

Intestinal ischemia is a life-threatening condition associated with a broad range of clinical conditions including atherosclerosis, thrombosis, hypotension, necrotizing enterocolitis, bowel transplantation, trauma and chronic inflammation. Intestinal ischemia-reperfusion (IR) injury is a consequence of acute mesenteric ischemia, caused by inadequate blood flow through the mesenteric vessels, resulting in intestinal damage. Reperfusion following ischemia can further exacerbate damage of the intestine. The mechanisms of IR injury are complex and poorly understood. Therefore, experimental small animal models are critical for understanding the pathophysiology of IR injury and the development of novel therapies. Here we describe a mouse model of acute intestinal IR injury that provides reproducible injury of the small intestine without mortality. This is achieved by inducing ischemia in the region of the distal ileum by temporally occluding the peripheral and terminal collateral branches of the superior mesenteric artery for 60 min using microvascular clips. Reperfusion for 1 hr, or 2 hr after injury results in reproducible injury of the intestine examined by histological analysis. Proper position of the microvascular clips is critical for the procedure. Therefore the video clip provides a detailed visual step-by-step description of this technique. This model of intestinal IR injury can be utilized to study the cellular and molecular mechanisms of injury and regeneration.


Assuntos
Modelos Animais de Doenças , Traumatismo por Reperfusão , Animais , Íleo , Intestino Delgado , Artéria Mesentérica Superior , Camundongos
12.
Immunol Lett ; 100(1): 88-93, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16055198

RESUMO

Thymidylate synthase (TYMS), the critical enzyme for DNA synthesis and a target for chemotherapy, was recently characterized as an oncogene and a potential target for specific immunotherapy. Here we report TYMS-specific antibody response in a fraction of colon cancer patients. Humoral immune response to TYMS is induced by chemotherapy using TYMS inhibitors, such as 5-fluorouracil (5-FU), and may be associated with tumor burden. Therefore, TYMS may serve as a useful serological biomarker for monitoring the course of disease and treatment in cancer patients.


Assuntos
Anticorpos/sangue , Antimetabólitos Antineoplásicos/administração & dosagem , Biomarcadores Tumorais/sangue , Neoplasias do Colo/sangue , Fluoruracila/administração & dosagem , Timidilato Sintase/imunologia , Anticorpos/imunologia , Biomarcadores Tumorais/imunologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/imunologia , Humanos , Monitorização Fisiológica/métodos , Carga Tumoral/efeitos dos fármacos
13.
J Immunol Methods ; 421: 61-72, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25702536

RESUMO

Citrobacter rodentium is a natural mouse pathogen which reproducibly infects mice and causes intestinal disease. The C. rodentium model of infection is very useful for investigating host-pathogen immune interactions in the gut, and can also be used to understand the pathogenesis of several important human intestinal disorders, including Crohn's disease, ulcerative colitis, dysbiosis and colon tumorigenesis. Both innate and adaptive immune responses play a critical role in protection against C. rodentium. Here, we summarize the role of immune components in protection against C. rodentium and describe techniques for the analysis of innate and adaptive mucosal immune responses, including setting up the infection, analysis of colonic hyperplasia and bacterial dissemination, evaluation of antibody responses, and purification and analysis of intestinal epithelial and lymphoid cells.


Assuntos
Citrobacter rodentium/imunologia , Colite/imunologia , Infecções por Enterobacteriaceae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Mucosa Intestinal/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Colite/microbiologia , Colite/patologia , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Hiperplasia/imunologia , Hiperplasia/microbiologia , Imunidade nas Mucosas/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
Immunol Lett ; 85(1): 71-4, 2003 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-12505200

RESUMO

Screening of expression cDNA libraries derived from human neoplasms with autologous sera (SEREX) is an established method for defining antigens immunogenic in individual cancer patients. Although the majority of SEREX-derived cDNA clones encode autoantigens, some of them represent shared cancer antigens with cancer-related serological profiles. Routine evaluation of multiple SEREX-derived clones in serological assays using panels of allogeneic sera from cancer patients is an important step towards defining disease parameters of diagnostic and prognostic significance. Here we show how the seroreactivity of multiple SEREX-derived antigens can be simultaneously evaluated using a rapid semi-quantitative protocol of allogeneic screening, which we call SMARTA (serological mini-arrays of recombinant tumor antigens).


Assuntos
Formação de Anticorpos/imunologia , Antígenos de Neoplasias/análise , Antígenos de Neoplasias/imunologia , Testes Sorológicos/métodos , Anticorpos Antineoplásicos , Antígenos de Neoplasias/genética , Autoanticorpos , Clonagem Molecular , DNA Complementar , Perfilação da Expressão Gênica , Biblioteca Gênica , Humanos , Proteínas Recombinantes/imunologia
15.
PLoS One ; 8(1): e54719, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23355893

RESUMO

LIGHT/TNFSF14 is a costimulatory molecule expressed on activated T cells for activation and maintenance of T cell homeostasis. LIGHT over expressed in T cells also down regulates hepatic lipase levels in mice through lymphotoxin beta receptor (LTßR) signaling. It is unclear whether LIGHT regulates hepatic lipase directly by interacting with LTßR expressing cells in the liver or indirectly by activation of T cells, and whether Kupffer cells, a major cell populations in the liver that expresses the LTßR, are required. Here we report that LIGHT expression via an adenoviral vector (Ad-LIGHT) is sufficient to down regulate hepatic lipase expression in mice. Depletion of Kupffer cells using clodronate liposomes had no effect on LIGHT-mediated down regulation of hepatic lipase. LIGHT-mediated regulation of hepatic lipase is also independent of LIGHT expression by T cells or activation of T cells. This is demonstrated by the decreased hepatic lipase expression in the liver of Ad-LIGHT infected recombination activating gene deficient mice that lack mature T cells and by the Ad-LIGHT infection of primary hepatocytes. Hepatic lipase expression was not responsive to LIGHT when mice lacking LTßR globally or only on hepatocytes were infected with Ad-LIGHT. Therefore, our data argues that interaction of LIGHT with LTßR on hepatocytes, but not Kupffer cells, is sufficient to down regulate hepatic lipase expression and that this effect can be independent of LIGHT's costimulatory function.


Assuntos
Regulação para Baixo/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Hepatócitos/enzimologia , Lipase/biossíntese , Receptor beta de Linfotoxina/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/biossíntese , Animais , Hepatócitos/citologia , Células de Kupffer/citologia , Células de Kupffer/enzimologia , Lipase/genética , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Linfócitos T/citologia , Linfócitos T/enzimologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
16.
Cancer Immunol Res ; 1(3): 190-200, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24777681

RESUMO

A mutation in the hydin gene has been recently described as one possible mechanism leading to lethal congenital hydrocephalus in mice, and a similar defect is proposed to be involved in an autosomal recessive form of hydrocephalus in human. Here, we report for the first time on the cancer association and immunogenicity of two HYDIN variants in humans. One is a previously described sequence derived from the chromosome 1 gene copy, that is, KIAA1864. The second is encoded by a novel alternative transcript originating from the chromosome 16, which we identified by immunoscreening of a testis-derived cDNA expression library with sera of patients with colorectal cancer, and called MO-TES391. Both variants are targeted by immunoglobulin G antibodies in a significant subset of cancer patients but only rarely in healthy donors. Moreover, we identify HLA-A*0201-restricted sequences derived from MO-TES391 and KIAA1864, which are specifically recognized by human cytotoxic CD8(+) T cells. Taken together, these results suggest frequent and coordinated adaptive immune responses against HYDIN variants in patients with cancer and propose HYDIN as a novel cancer-associated antigen.


Assuntos
Imunidade Adaptativa/genética , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas dos Microfilamentos/imunologia , Processamento Alternativo , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Cromossomos Humanos Par 1 , Cromossomos Humanos Par 16 , Antígeno HLA-A2/metabolismo , Humanos , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Mutação , Neoplasias/imunologia , RNA Mensageiro/imunologia , RNA Mensageiro/metabolismo
17.
Cell Host Microbe ; 10(1): 44-53, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21767811

RESUMO

Innate lymphoid cells (ILCs) have emerged as important players, regulating the balance between protective immunity and immunopathology at mucosal surfaces. However, mechanisms that regulate ILCs' effector functions during mucosal pathogenic challenge are poorly defined. Using mice infected with the natural mouse enteric pathogen Citrobacter rodentium, we demonstrate that lymphotoxin (LT) is essential for IL-22 production by intestinal ILCs. Blocking of LTßR signaling dramatically reduced intestinal IL-22 production after C. rodentium infection. Conversely, stimulating LTßR signaling induced an IL-22 protection pathway in LT-deficient mice. Furthermore, exogenous IL-22 expression rescued LTßR-deficient mice. IL-22-producing ILCs were predominantly located in lymphoid follicles in the colon and interacted closely with dendritic cells (DCs). We find that an LT-driven positive feedback loop controls IL-22 production by RORγt(+) ILCs via LTßR signaling in DCs. Taken together, our data show that LTßR signaling in gut lymphoid follicles regulates IL-22 production by ILCs in response to mucosal pathogen challenge.


Assuntos
Citrobacter rodentium/patogenicidade , Interleucinas/metabolismo , Mucosa Intestinal/microbiologia , Linfócitos/imunologia , Linfotoxina-alfa/metabolismo , Animais , Colo/metabolismo , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/metabolismo , Imunidade Inata , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Linfócitos/metabolismo , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/imunologia , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-alfa/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Transdução de Sinais , Interleucina 22
18.
Cancer Immunol Immunother ; 57(6): 871-81, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18026951

RESUMO

PURPOSE: Tankyrases 1 and 2 are telomere-associated poly(ADP-ribose) polymerases (PARP) that can positively regulate telomere elongation and interact with multiple cellular proteins. Recent reports implicated tankyrases as tumor antigens and potential targets of anticancer treatment. We examined expression of tankyrases in colon tumors and immune response to these enzymes in patients with different types of cancer. METHODS: mRNA and protein expression was evaluated by quantitative real-time RT-PCR and Western blotting, respectively. Humoral immune response to recombinant tankyrases was investigated by modified enzyme-linked immunoassays. Cellular immune response was analysed by ELISPOT and (51)Cr release assays. RESULTS: We found that both mRNA and protein levels of tankyrase 2 (TNKL) are upregulated in colon tumors. In contrast, protein level of tankyrase 1 (TNKS) is downregulated, while mRNA level shows variable changes. More than a quarter of colon cancer patients develop humoral immune response to at least one of the two tankyrases. In this study we mapped common and unique B-cell epitopes located in different domains of the two proteins. Additionally, we present evidence for T-cell responses both to epitopes that are unique for TNKL and to those shared between TNKL and TNKS. CONCLUSION: Our study favors a biomarker usage of antibody response to tankyrases. Spontaneous CD8(+) T-cell responses to these enzymes are rare and further investigation is needed to evaluate tankyrases as potential targets for cancer immunotherapy.


Assuntos
Formação de Anticorpos , Neoplasias do Colo/enzimologia , Imunidade Celular , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Tanquirases/biossíntese , Sequência de Aminoácidos , Biomarcadores Tumorais/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Neoplasias do Colo/patologia , Mapeamento de Epitopos/métodos , Epitopos/química , Humanos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Tanquirases/metabolismo
19.
Eur J Immunol ; 35(5): 1592-600, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15832287

RESUMO

We generated a novel tumor necrosis factor (TNF) null mutation using Cre-loxP technology. Mice homozygous for this mutation differ from their "conventional" counterparts; in particular, they completely lack Peyer's patches (PP) but retain all lymph nodes. Our analysis of these novel TNF-knockout mice supports the previously disputed notion of the involvement of TNF-TNFR1 signaling in PP organogenesis. Availability of TNF-knockout strains both with and without PP enables more definitive studies concerning the roles of TNF and PP in various immune functions and disease conditions. Here, we report that systemic ablation of TNF, but not the presence of PP per se, is critical for protection against intestinal Listeria infection in mice.


Assuntos
Camundongos Knockout/imunologia , Nódulos Linfáticos Agregados/imunologia , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Animais , Citometria de Fluxo , Hematopoese/imunologia , Imuno-Histoquímica , Listeriose/imunologia , Camundongos , Camundongos Knockout/genética , Mutação , Nódulos Linfáticos Agregados/patologia , Fator de Necrose Tumoral alfa/imunologia
20.
Int J Cancer ; 117(5): 800-6, 2005 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-15981215

RESUMO

Antibodies to cancer antigens can often be detected in the sera of patients, although the mechanism of the underlying humoral immune response is poorly understood. Using immunoscreening of tumor-derived cDNA expression libraries (SEREX), we identified human histone deacetylase 3 (HDAC3) as serologically defined antigen in colon cancer. Closely related HDAC1 and HDAC2 do not elicit humoral response in colon cancer patients. We show that the C-terminal region of HDAC3 protein lacking the homology to other Class I HDAC contains at least 3 distinct B-cell epitopes that are recognized by the serum antibodies. HDAC3 in combination with other SEREX antigens may become a useful molecular biomarker with diagnostic or prognostic value for a subset of colon cancer patients.


Assuntos
Anticorpos Antineoplásicos/biossíntese , Neoplasias do Colo/enzimologia , Neoplasias do Colo/imunologia , Histona Desacetilases/química , Sequência de Aminoácidos , Linfócitos B/imunologia , Sequência de Bases , Western Blotting , Primers do DNA , Epitopos/imunologia , Histona Desacetilases/imunologia , Humanos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA