Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 34(49): 5960-70, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-25745993

RESUMO

The poor efficacy of the in vivo anti-tumor immune response has been partially attributed to ineffective T-cell responses mounted against the tumor. Fas-FasL-dependent activation-induced cell death (AICD) of T cells is believed to be a major contributor to compromised anti-tumor immunity. The molecular mechanisms of AICD are well-investigated, yet the possibility of regulating AICD for cancer therapy remains to be explored. In this study, we show that histone deacetylase inhibitors (HDACIs) can inhibit apoptosis of CD4(+) T cells within the tumor, thereby enhancing anti-tumor immune responses and suppressing melanoma growth. This inhibitory effect is specific for AICD through suppressing NFAT1-regulated FasL expression on activated CD4(+) T cells. In gld/gld mice with mutation in FasL, the beneficial effect of HDACIs on AICD of infiltrating CD4(+) T cells is not seen, confirming the critical role of FasL regulation in the anti-tumor effect of HDACIs. Importantly, we found that the co-administration of HDACIs and anti-CTLA4 could further enhance the infiltration of CD4(+) T cells and achieve a synergistic therapeutic effect on tumor. Therefore, our study demonstrates that the modulation of AICD of tumor-infiltrating CD4(+) T cells using HDACIs can enhance anti-tumor immune responses, uncovering a novel mechanism underlying the anti-tumor effect of HDACIs.


Assuntos
Anticorpos/administração & dosagem , Antineoplásicos/administração & dosagem , Linfócitos T CD4-Positivos/efeitos dos fármacos , Antígeno CTLA-4/antagonistas & inibidores , Inibidores de Histona Desacetilases/administração & dosagem , Melanoma Experimental/tratamento farmacológico , Animais , Anticorpos/uso terapêutico , Antineoplásicos/farmacologia , Morte Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteína Ligante Fas/genética , Proteína Ligante Fas/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Ácidos Hidroxâmicos/farmacologia , Melanoma Experimental/imunologia , Camundongos , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo
2.
Cell Death Dis ; 5: e1009, 2014 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-24457953

RESUMO

Mesenchymal stem cells (MSCs) are believed to exert their regenerative effects through differentiation and modulation of inflammatory responses. However, the relationship between the severity of inflammation and stem cell-mediated tissue repair has not been formally investigated. In this study, we applied different concentrations of dexamethasone (Dex) to anti-CD3-activated splenocyte cultured with or without MSCs. As expected, Dex exhibited a classical dose-dependent inhibition of T-cell proliferation. Surprisingly, although MSCs also blocked T-cell proliferation, the presence of Dex unexpectedly showed a dose-dependent reversion of T-cell proliferation. This effect of Dex was found to be exerted through interfering STAT1 phosphorylation-prompted expression of inducible nitric oxide synthase (iNOS). Interestingly, inflammation-induced chemokines in MSCs was unaffected. To test the role of inflammation severity in stem cell-mediated tissue repair, we employed mice with carbon tetrachloride-induced advanced liver fibrosis and found that although MSCs alone were effective, concurrent administration of Dex abrogated the therapeutic effects of MSCs on fibrin deposition, serum levels of bilirubin, albumin, and aminotransferases, as well as T-lymphocyte infiltration, especially IFN-γ(+)CD4(+) and IL-17A(+)CD4(+)T cells. Likewise, iNOS(-/-) MSCs, which produce chemokines but not nitric oxide under inflammatory conditions, are ineffective in treating advanced liver fibrosis. Therefore, inflammation has a critical role in MSC-mediated tissue repair. In addition, concomitant application of MSCs with steroids should be avoided.


Assuntos
Dexametasona/administração & dosagem , Imunossupressores/administração & dosagem , Cirrose Hepática/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Comunicação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiocinas/genética , Quimiocinas/imunologia , Dexametasona/efeitos adversos , Feminino , Humanos , Imunossupressores/efeitos adversos , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/imunologia , Cirrose Hepática/fisiopatologia , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
3.
Cell Death Differ ; 21(3): 388-96, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24162664

RESUMO

Mammalian mesenchymal stem cells (MSCs) have been shown to be strongly immunosuppressive in both animal disease models and human clinical trials. We have reported that the key molecule mediating immunosuppression by MSCs is species dependent: indoleamine 2,3-dioxygenase (IDO) in human and inducible nitric oxide synthase (iNOS) in mouse. In the present study, we isolated MSCs from several mammalian species, each of a different genus, and investigated the involvement of IDO and iNOS during MSC-mediated immunosuppression. The characterization of MSCs from different species was by adherence to tissue culture plastic, morphology, specific marker expression, and differentiation potential. On the basis of the inducibility of IDO and iNOS by inflammatory cytokines in MSCs, the tested mammalian species fall into two distinct groups: IDO utilizers and iNOS utilizers. MSCs from monkey, pig, and human employ IDO to suppress immune responses, whereas MSCs from mouse, rat, rabbit, and hamster utilize iNOS. Interestingly, based on the limited number of species tested, the iNOS-utilizing species all belong to the phylogenetic clade, Glires. Although the evolutionary significance of this divergence is not known, we believe that this study provides critical guidance for choosing appropriate animal models for preclinical studies of MSCs.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Células-Tronco Mesenquimais/enzimologia , Óxido Nítrico Sintase Tipo II/imunologia , Animais , Diferenciação Celular/fisiologia , Cricetinae , Modelos Animais de Doenças , Haplorrinos , Humanos , Tolerância Imunológica , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/genética , Filogenia , Coelhos , Ratos , Ratos Sprague-Dawley , Suínos
4.
Oncogene ; 33(29): 3830-8, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-23975435

RESUMO

p53 is one of the most studied genes in cancer biology, and mutations in this gene may be predictive for the development of many types of cancer in humans and in animals. However, whether p53 mutations in non-tumor stromal cells can affect tumor development has received very little attention. In this study, we show that B16F0 melanoma cells form much larger tumors in p53-deficient mice than in wild-type mice, indicating a potential role of p53 deficiency in non-tumor cells of the microenvironment. As mesenchymal stem cells (MSCs) are attracted to tumors and form a major component of the tumor microenvironment, we examined the potential role of p53 status in MSCs in tumor development. We found that larger tumors resulted when B16F0 melanoma cells were co-injected with bone marrow MSCs derived from p53-deficient mice rather than MSCs from wild-type mice. Interestingly, this tumor-promoting effect by p53-deficient MSCs was not observed in non-obese diabetic/severe combined immunodeficiency mice, indicating the immune response has a critical role. Indeed, in the presence of inflammatory cytokines, p53-deficient MSCs expressed more inducible nitric oxide synthase (iNOS) and exhibited greater immunosuppressive capacity. Importantly, tumor promotion by p53-deficient MSCs was abolished by administration of S-methylisothiourea, an iNOS inhibitor. Therefore, our data demonstrate that p53 status in tumor stromal cells has a key role in tumor development by modulating immune responses.


Assuntos
Imunomodulação , Células-Tronco Mesenquimais/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Microambiente Tumoral/imunologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Citocinas/biossíntese , Modelos Animais de Doenças , Imunomodulação/genética , Mediadores da Inflamação , Melanoma Experimental , Camundongos , Camundongos Knockout , Neoplasias/genética , Neoplasias/patologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Microambiente Tumoral/genética , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
5.
Cell Death Differ ; 20(12): 1731-41, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24076588

RESUMO

Helper T cells are crucial for maintaining proper immune responses. Yet, they have an undefined relationship with one of the most potent immune stimulatory cytokines, granulocyte macrophage-colony-stimulating factor (GM-CSF). By depleting major cytokines during the differentiation of CD4(+) T cells in vitro, we derived cells that were found to produce large amounts of GM-CSF, but little of the cytokines produced by other helper T subsets. By their secretion of GM-CSF, this novel subset of helper T cells (which we have termed ThGM cells) promoted the production of cytokines by other T-cell subtypes, including type 1 helper T cell (Th1), type 2 helper T cell (Th2), type 1 cytotoxic T cell (Tc1), type 2 cytotoxic T cell (Tc2), and naive T cells, as evidenced by the fact that antibody neutralization of GM-CSF abolished this effect. ThGM cells were found to be highly prone to activation-induced cell death (AICD). Inhibitors of TRAIL or granzymes could not block AICD in ThGM cells, whereas inhibition of FasL/Fas interaction partially rescued ThGM cells from AICD. Thus, ThGM cells are a novel subpopulation of T helper cells that produce abundant GM-CSF, exhibit exquisite susceptibility to apoptosis, and therefore play a pivotal role in the regulation of the early stages of immune responses.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Imunidade/imunologia , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Transferência Adotiva , Animais , Diferenciação Celular/imunologia , Técnicas de Cocultura , Citidina Desaminase/metabolismo , Citocinas/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia
6.
Cell Death Differ ; 19(9): 1505-13, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22421969

RESUMO

Mesenchymal stem cells (MSCs) have been employed successfully to treat various immune disorders in animal models and clinical settings. Our previous studies have shown that MSCs can become highly immunosuppressive upon stimulation by inflammatory cytokines, an effect exerted through the concerted action of chemokines and nitric oxide (NO). Here, we show that MSCs can also enhance immune responses. This immune-promoting effect occurred when proinflammatory cytokines were inadequate to elicit sufficient NO production. When inducible nitric oxide synthase (iNOS) production was inhibited or genetically ablated, MSCs strongly enhance T-cell proliferation in vitro and the delayed-type hypersensitivity response in vivo. Furthermore, iNOS(-/-) MSCs significantly inhibited melanoma growth. It is likely that in the absence of NO, chemokines act to promote immune responses. Indeed, in CCR5(-/-)CXCR3(-/-) mice, the immune-promoting effect of iNOS(-/-) MSCs is greatly diminished. Thus, NO acts as a switch in MSC-mediated immunomodulation. More importantly, the dual effect on immune reactions was also observed in human MSCs, in which indoleamine 2,3-dioxygenase (IDO) acts as a switch. This study provides novel information about the pathophysiological roles of MSCs.


Assuntos
Proliferação de Células , Tolerância Imunológica , Células-Tronco Mesenquimais/imunologia , Linfócitos T/imunologia , Animais , Quimiocinas/genética , Quimiocinas/imunologia , Humanos , Hipersensibilidade Tardia/genética , Hipersensibilidade Tardia/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Knockout , Óxido Nítrico/genética , Óxido Nítrico/imunologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , Receptores CCR5/genética , Receptores CCR5/imunologia , Receptores CXCR3/genética , Receptores CXCR3/imunologia , Linfócitos T/patologia
7.
Cell Death Differ ; 11(12): 1258-64, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15375386

RESUMO

Cell death through apoptosis plays a critical role in regulating cellular homeostasis. Whether the disposal of apoptotic cells through phagocytosis can actively induce immune tolerance in vivo, however, remains controversial. Here, we report in a rat model that without using immunosuppressants, transfusion of apoptotic splenocytes from the donor strain prior to transplant dramatically prolonged survival of heart allografts. Histological analysis verified that rejection signs were significantly ameliorated. Splenocytes from rats transfused with donor apoptotic cells showed a dramatically decreased response to donor lymphocyte stimulation. Most importantly, blockade of phagocytosis in vivo, either with gadolinium chloride to disrupt phagocyte function or with annexin V to block binding of exposed phosphotidylserine to its receptor on phagocytes, abolished the beneficial effect of transfused apoptotic cells on heart allograft survival. Our results demonstrate that donor apoptotic cells promote specific allograft acceptance and that phagocytosis of apoptotic cells in vivo plays a crucial role in maintaining immune tolerance.


Assuntos
Apoptose/imunologia , Sobrevivência de Enxerto/imunologia , Transfusão de Linfócitos/métodos , Linfócitos/imunologia , Fagocitose/imunologia , Tolerância ao Transplante/imunologia , Animais , Anexina A5/farmacologia , Gadolínio/farmacologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Transplante de Coração/imunologia , Masculino , Modelos Animais , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/metabolismo
8.
J Immunol ; 167(10): 5527-30, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11698420

RESUMO

Unlike primary T cells in lymph nodes, effector CD8(+) CTL in tissues do not express the costimulatory receptor CD28. We report that NKG2D, the receptor for stress-induced MICA and MICB molecules expressed in the intestine, serves as a potent costimulatory receptor for CTL freshly isolated from the human intestinal epithelium. Expression and function of NKG2D are selectively up-regulated by the cytokine IL-15, which is released by the inflamed intestinal epithelium. These findings identify a novel CTL costimulatory pathway regulated by IL-15 and suggest that tissues can fine-tune the activation of effector T cells based on the presence or absence of stress and inflammation. Uncontrolled secretion of IL-15 could lead to excessive induction of NKG2D and thus contribute to the development of autoimmune disease by facilitating the activation of autoreactive T cells.


Assuntos
Antígenos CD28/análise , Interleucina-15/farmacologia , Ativação Linfocitária , Receptores Imunológicos/biossíntese , Receptores Imunológicos/fisiologia , Linfócitos T Citotóxicos/imunologia , Linhagem Celular , Células Cultivadas , Testes Imunológicos de Citotoxicidade , Citotoxicidade Imunológica , Humanos , Imunidade nas Mucosas , Memória Imunológica , Interferon gama/biossíntese , Mucosa Intestinal/imunologia , Modelos Imunológicos , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores de Células Matadoras Naturais , Linfócitos T Citotóxicos/efeitos dos fármacos
9.
Dig Dis Sci ; 46(3): 661-7, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11318549

RESUMO

This study examines the in vitro response of human lamina propria lymphocytes (LPLs) to food antigens. LPLs were obtained from jejunum of healthy individuals undergoing gastric bypass operations for morbid obesity. Proliferation was assayed by [3H]thymidine incorporation and cytokine production by ELISA. LPLs proliferated in response to keyhole limpet hemocyanin, ovalbumin, lactoglobulin, and phytohemagglutinin (PHA), but not to yeast. The responses to lactoglobulin and PHA were inhibited by the CTLA4/Fc chimera and by MAbs against CD2, CD58, CD80, and CD86, indicating stimulation of CD28+ LPLs with antigen-presenting cells through activation of the CD2 pathway. Besides producing IL-2, IL-10, and TNF-alpha, LPLs synthesized large amounts of IFN-gamma (100 ng/ml) with lactoglobulin, a process dependent upon CD80/CD86, CD40/CD40L, and IL-12. After a three-day culture with lactoglobulin, ovalbumin, or PHA, LPLs developed suppressor activity that reduced proliferation of naive LPLs to these same stimuli. In summary, LPLs first respond to lactoglobulin by proliferation and IFN-gamma production, then by development of antigen nonspecific suppression.


Assuntos
Interferon gama/biossíntese , Lactoglobulinas/farmacologia , Linfócitos/imunologia , Mucosa/citologia , Linfócitos T Reguladores/imunologia , Divisão Celular/fisiologia , Citocinas/biossíntese , Humanos , Técnicas In Vitro , Fito-Hemaglutininas/farmacologia
10.
J Exp Med ; 192(5): 761-8, 2000 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-10974041

RESUMO

The immune system has evolved specialized cellular and molecular mechanisms for targeting and regulating immune responses at epithelial surfaces. Here we show that small intestinal intraepithelial lymphocytes and lamina propria lymphocytes migrate to thymus-expressed chemokine (TECK). This attraction is mediated by CC chemokine receptor (CCR)9, a chemoattractant receptor expressed at high levels by essentially all CD4(+) and CD8(+) T lymphocytes in the small intestine. Only a small subset of lymphocytes in the colon are CCR9(+), and lymphocytes from other tissues including tonsils, lung, inflamed liver, normal or inflamed skin, inflamed synovium and synovial fluid, breast milk, and seminal fluid are universally CCR9(-). TECK expression is also restricted to the small intestine: immunohistochemistry reveals that intense anti-TECK reactivity characterizes crypt epithelium in the jejunum and ileum, but not in other epithelia of the digestive tract (including stomach and colon), skin, lung, or salivary gland. These results imply a restricted role for lymphocyte CCR9 and its ligand TECK in the small intestine, and provide the first evidence for distinctive mechanisms of lymphocyte recruitment that may permit functional specialization of immune responses in different segments of the gastrointestinal tract. Selective expression of chemokines by differentiated epithelium may represent an important mechanism for targeting and specialization of immune responses.


Assuntos
Quimiocinas CC/análise , Mucosa Intestinal/imunologia , Intestino Delgado/imunologia , Receptores de Quimiocinas/análise , Animais , Quimiocinas CC/fisiologia , Humanos , Camundongos , Especificidade de Órgãos , Receptores CCR , Receptores de Quimiocinas/fisiologia , Linfócitos T/química
11.
Immunology ; 101(2): 185-90, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11012771

RESUMO

In food poisoning, Staphylococcus aureus secretes staphylococcal enterotoxin B (SEB), a superantigen that causes intense T-cell proliferation and cytotoxicity. The effects of SEB on lytic activity by human intestinal intraepithelial lymphocytes (IEL) were investigated. Jejunal IEL, from morbidly obese individuals undergoing gastric bypass operations, were tested for SEB-induced cytotoxicity against C1R B-lymphoblastoid cells, HT-29 adenocarcinoma cells, or CD1d-transfected cells using the 51Cr-release assay. Fas and Fas ligand expression were detected by immunofluorescence and flow cytometry and soluble ligand by enzyme-linked immunosorbent assay (ELISA). In the presence of SEB, IEL became potently cytotoxic against C1R cells and interferon-gamma (IFN-gamma)-precultured HT-29 cells, causing 55+/-10% and 31+/-6% lysis, respectively, greater than that by phytohaemagglutinin (PHA)-, interleukin-2 (IL-2)-, or anti-T-cell receptor (TCR)-activated IEL. SEB-stimulated peripheral blood (PB) CD8+ T cells lysed similar numbers of C1R cells but fewer HT-29 cells (53+/-13% and 8+/-5%, respectively). IEL killing of C1R cells involved interaction of major histocompatibility complex (MHC) class II with TCR, CD2 with CD58, and CD11a with CD54, and was perforin mediated. SEB-induced IEL lysis of HT-29 cells, in contrast, was caused by an unknown target cell structure, not MHC class II or CD1d, and resulted from a combination of perforin and Fas-mediated events. The potent cytotoxic activities of IEL promoted by SEB utilize two different mechanisms, depending on the surface receptors expressed by the target cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica/imunologia , Enterotoxinas/imunologia , Mucosa Intestinal/imunologia , Antígenos CD1/imunologia , Antígenos CD1d , Técnicas de Cultura de Células , Células Epiteliais/imunologia , Humanos , Jejuno/imunologia , Staphylococcus aureus/imunologia , Superantígenos/imunologia , Células Tumorais Cultivadas , Receptor fas/imunologia
12.
Curr Biol ; 10(6): 325-8, 2000 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-10744978

RESUMO

HIV particles that use the chemokine receptor CXCR4 as a coreceptor for entry into cells (X4-HIV) inefficiently transmit infection across mucosal surfaces [1], despite their presence in seminal fluid and mucosal secretions from infected individuals [2] [3] [4]. In addition, although intestinal lymphocytes are susceptible to infection with either X4-HIV particles or particles that use the chemokine receptor CCR5 for viral entry (R5-HIV) during ex vivo culture [5], only systemic inoculation of R5-chimeric simian-HIV (S-HIV) results in a rapid loss of CD4(+) intestinal lymphocytes in macaques [6]. The mechanisms underlying the inefficient capacity of X4-HIV to transmit infection across mucosal surfaces and to infect intestinal lymphocytes in vivo have remained elusive. The CCR5 ligands RANTES, MIP-1alpha and MIP-1beta suppress infection by R5-HIV-1 particles via induction of CCR5 internalization, and individuals whose peripheral blood lymphocytes produce high levels of these chemokines are relatively resistant to infection [7] [8] [9]. Here, we show that the CXCR4 ligand stromal derived factor-1 (SDF-1) is constitutively expressed by mucosal epithelial cells at sites of HIV transmission and propagation. Furthermore, CXCR4 is selectively downmodulated on intestinal lymphocytes within the setting of prominent SDF-1 expression. We postulate that mucosally derived SDF-1 continuously downmodulates CXCR4 on resident HIV target cells, thereby reducing the transmission and propagation of X4-HIV at mucosal sites. Moreover, such a mechanism could contribute to the delayed emergence of X4 isolates, which predominantly occurs during the later stages of the HIV infection.


Assuntos
Quimiocinas CXC/fisiologia , HIV/crescimento & desenvolvimento , Mucosa Intestinal/metabolismo , Quimiocina CXCL12 , Quimiocinas CXC/biossíntese , Humanos , Receptores CCR5/metabolismo , Receptores CXCR4/biossíntese , Receptores CXCR4/genética
13.
Gastroenterology ; 118(5): 867-79, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10784586

RESUMO

BACKGROUND & AIMS: Celiac disease is a gluten-induced enteropathy characterized by the presence of gliadin-specific CD4(+) T cells in the lamina propria and by a prominent intraepithelial T-cell infiltration of unknown mechanism. The aim of this study was to characterize the subset(s) of intraepithelial lymphocytes (IELs) expanding during active celiac disease to provide insights into the mechanisms involved in their expansion. METHODS: Flow-cytometric analysis of isolated IELs and/or immunohistochemical staining of frozen sections were performed in 51 celiac patients and 50 controls with a panel of monoclonal antibodies against T-cell and natural killer (NK) receptors. In addition, in vitro studies were performed to identify candidate stimuli for NK receptor expression. RESULTS: In normal intestine, different proportions of IELs, which were mainly T cells, expressed the NK receptors CD94/NKG2, NKR-P1A, KIR2D/3D, NKp46, Pen5, or CD56. During the active phase of celiac disease, the frequency of CD94(+) IELs, which were mostly alphabeta T cells, was conspicuously increased over controls. In contrast, the expression of other NK markers was not modified. Furthermore, expression of CD94 could be selectively induced in vitro by T-cell receptor activation and/or interleukin 15, a cytokine produced by intestinal epithelial cells. CONCLUSIONS: The gut epithelium favors the development of T cells that express NK receptors. In active celiac disease, there is a specific and selective increase of IELs expressing CD94, the HLA-E-specific NK receptor that may be related to T-cell receptor activation and/or interleukin 15 secretion.


Assuntos
Antígenos CD/metabolismo , Doença Celíaca/metabolismo , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Mucosa Intestinal/metabolismo , Células Matadoras Naturais/metabolismo , Lectinas Tipo C , Linfócitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Adulto , Células Cultivadas , Criança , Pré-Escolar , Citometria de Fluxo , Humanos , Lactente , Interleucina-15/farmacologia , Pessoa de Meia-Idade , Subfamília D de Receptores Semelhantes a Lectina de Células NK , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Antígenos HLA-E
14.
Eur J Immunol ; 30(3): 819-26, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10741397

RESUMO

To determine which chemokine receptors might be involved in T lymphocyte localization to the intestinal mucosa, we examined receptor expression on human intestinal lamina propria lymphocytes (LPL), intraepithelial lymphocytes (IEL) and CD45RO+beta7hi gut homing peripheral blood lymphocytes (PBL). Virtually all LPL and IEL expressed CXCR3 and CCR5, receptors that have been associated with Th1(Tc1)/Th0 lymphocytes, while CCR3 and CCR4, receptors associated with Th2 (Tc2)lymphocytes, CCR7, CXCR1 and CXCR2 were not expressed. CXCR3 and CCR5 receptors were functional, as LPL and IEL migrated to their respective ligands I-TAC and RANTES. In addition, most alphaEbeta7- LPL and IEL expressed high levels of CCR2. While the majority of CD45RO(-)beta7hi PBL also expressed CXCR3 and CCR5, a proportion of these cells were CXCR3- and/or CCR5- and some expressed CCR4 and/or CCR7, indicating that lymphocytes recruited to the intestinal mucosa represent a subset of these cells. In summary, our results show that LPL and IEL within the normal intestine express a specific and similar array of chemokine receptors whose ligands are constitutively expressed in the intestinal mucosa and whose expression is up-regulated during intestinal inflammation. These results support the view that CXCR3, CCR5 and CCR2 may play an important role in lymphocyte localization within the intestinal mucosa.


Assuntos
Cadeias beta de Integrinas , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Linfócitos/imunologia , Receptores de Quimiocinas/metabolismo , Animais , Anticorpos Monoclonais , Quimiotaxia de Leucócito , Células Epiteliais/citologia , Células Epiteliais/imunologia , Humanos , Técnicas In Vitro , Inflamação/imunologia , Inflamação/patologia , Integrinas/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Receptores CCR2 , Receptores CCR5/metabolismo , Receptores CXCR3 , Linfócitos T/imunologia
15.
J Exp Med ; 190(9): 1241-56, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10544196

RESUMO

TECK (thymus-expressed chemokine), a recently described CC chemokine expressed in thymus and small intestine, was found to mediate chemotaxis of human G protein-coupled receptor GPR-9-6/L1.2 transfectants. This activity was blocked by anti-GPR-9-6 monoclonal antibody (mAb) 3C3. GPR-9-6 is expressed on a subset of memory alpha4beta7(high) intestinal trafficking CD4 and CD8 lymphocytes. In addition, all intestinal lamina propria and intraepithelial lymphocytes express GPR-9-6. In contrast, GPR-9-6 is not displayed on cutaneous lymphocyte antigen-positive (CLA(+)) memory CD4 and CD8 lymphocytes, which traffic to skin inflammatory sites, or on other systemic alpha4beta7(-)CLA(-) memory CD4/CD8 lymphocytes. The majority of thymocytes also express GPR-9-6, but natural killer cells, monocytes, eosinophils, basophils, and neutrophils are GPR-9-6 negative. Transcripts of GPR-9-6 and TECK are present in both small intestine and thymus. Importantly, the expression profile of GPR-9-6 correlates with migration to TECK of blood T lymphocytes and thymocytes. As migration of these cells is blocked by anti-GPR-9-6 mAb 3C3, we conclude that GPR-9-6 is the principal chemokine receptor for TECK. In agreement with the nomenclature rules for chemokine receptors, we propose the designation CCR-9 for GPR-9-6. The selective expression of TECK and GPR-9-6 in thymus and small intestine implies a dual role for GPR-9-6/CCR-9, both in T cell development and the mucosal immune response.


Assuntos
Quimiocinas CC/farmacologia , Quimiotaxia/imunologia , Mucosa Intestinal/imunologia , Receptores de Quimiocinas/imunologia , Linfócitos T/metabolismo , Timo/imunologia , Anticorpos Monoclonais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Cálcio , Linhagem Celular , Quimiocinas CC/genética , Citometria de Fluxo , Regulação da Expressão Gênica/imunologia , Humanos , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos/imunologia , RNA Mensageiro/imunologia , Receptores CCR , Receptores de Quimiocinas/genética , Receptores de Retorno de Linfócitos/imunologia , Transfecção
16.
Immunology ; 97(4): 679-85, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10457223

RESUMO

Intraepithelial lymphocytes (IELs) from human intestinal epithelium are memory CD8+ T cells that bind to epithelial cells through human mycosal lymphocyte (HML)-1 and to mesenchymal cells through very late activation antigen-4 (VLA-4). Their binding of extracellular matrix proteins and the mechanism involved were tested. Activated 51Cr-labelled lymphocytes were incubated in protein-coated microwells with various additives. After washing, the adherent cells were detected by radioactivity. The percentages of activated IELs that bound to collagen types I and IV were 20 and 31%, respectively; fewer bound to fibronectin or laminin. Compared to interleukin-2-activated peripheral blood CD8+ T lymphocytes, more IELs bound collagen IV and fewer bound fibronectin. IEL adhesion to collagen (but not fibronectin or laminin) was up-regulated by antibody ligation of CD2 or by protein kinase C stimulation by phorbol ester; staurosporine reduced binding, while herbimycin, phytohaemagglutinin and CD3 ligation had no effect. Antibody-blocking of integrin VLA-1 subunits alpha1 (CD49a) and beta1 (CD18) inhibited adhesion to collagen type I by 82+/-6% and to type IV by 94+/-1% (P<0.001), implicating VLA-1 as the main collagen receptor for IELs. Cell adhesion was dependent on extracellular divalent cations, a characteristic event of VLA-1 never before shown for IELs: manganese and magnesium ions supported binding in a dose-dependent manner; calcium ions inhibited their effectiveness. Therefore, IELs bind collagen through integrin alpha1beta1 after protein kinase C activation. Adhesion is modulated by divalent cations.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Colágeno/metabolismo , Células Epiteliais/imunologia , Integrinas/metabolismo , Mucosa Intestinal/imunologia , Adesão Celular/imunologia , Técnicas de Cultura de Células , Proteínas da Matriz Extracelular/metabolismo , Humanos , Integrina alfa1beta1 , Interleucina-2/imunologia , Ativação Linfocitária/imunologia , Proteína Quinase C/metabolismo
17.
Gastroenterology ; 113(1): 144-50, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9207272

RESUMO

BACKGROUND & AIMS: Intraepithelial lymphocytes (IELs) from human intestinal mucosa proliferate minimally to T-cell stimuli. Optimal growth may depend on factors that are missing in vitro, such as accessory cells. The aim of this study was to determine whether mesenchymal cells costimulate IELs. METHODS: IELs were isolated from human jejunum and cultured with fibroblasts or smooth muscle cells (mesenchymal cell models for mucosal myofibroblasts) and various T-cell stimuli. Proliferation was determined by [3H]thymidine incorporation, and interleukin 2 (IL-2) production was measured by enzyme-linked immunosorbent assay. Surface molecules were detected by immunofluorescence and flow cytometry. RESULTS: The proliferative responses of IELs to mitogen (phytohemagglutinin), superantigen (staphylococcal enterotoxin B), or anti-CD3 antibody were increased greatly by coculture with mesenchymal cells, while only slightly by peripheral-blood monocytes, the classical antigen-presenting cells. IL-2 production and receptor expression also increased. Mesenchymal cell costimulation of IEL growth required direct contact between the two cell types and was partly dependent on the integrin alpha4beta1 (very late activation 4[VLA-4]) and major histocompatibility complex (MHC) class I, as their respective antibodies blocked the effect. The surface molecules B7 (CD80), CD2, and MHC class II were not involved. CONCLUSIONS: Optimal IEL growth depends on their contact with mesenchymal cells, an interaction that is mediated by VLA-4 and MHC class I. In mucosal immunity, basement membrane myofibroblasts likely serve this role.


Assuntos
Linfócitos T CD8-Positivos/citologia , Mucosa Intestinal/citologia , Divisão Celular , Técnicas de Cocultura , Ensaio de Imunoadsorção Enzimática , Fibroblastos/citologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Imunidade nas Mucosas , Integrina alfa4beta1 , Integrinas/fisiologia , Interleucina-2/biossíntese , Jejuno/citologia , Músculo Liso/citologia , Receptores de Retorno de Linfócitos/fisiologia , Receptores de Antígeno muito Tardio/fisiologia
18.
Gut ; 40(3): 333-8, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9135521

RESUMO

BACKGROUND: Human intraepithelial lymphocytes (IELs), predominantly T cells of the CD8+CD45RO+ phenotype that are situated between epithelial cells, have a chemotactic response to the alpha-chemokines, IL-8 and GRO, and the beta-chemokine, and the protein termed regulated on activation, normal T cell expressed and secreted (RANTES). AIM: To evaluate the specificity of the IL-8 receptor on IELs. METHODS: Specificity was determined by the degree of desensitisation of the IL-8 response caused by each chemokine and the degree of inhibition of IL-8 binding to the cell. RESULTS: IELs migrated towards two additional beta chemokines, macrophage inflammatory protein-1 and monocyte chemotactic protein (MCP). All chemokines inhibited IL-8 induced chemotaxis and calcium ion mobilisation by IELs, with IL-8 having the greatest effect and MCP the least. In addition, specific binding of radiolabelled IL-8 to IELs was reduced by each of the five chemokines in cold competition experiments, whereas only GRO and IL-8 itself displaced 125I-IL-8 from receptors on peripheral blood mononuclear cells. CONCLUSIONS: The IL-8 responsiveness of IELs is desensitised by chemokines of both the alpha and beta families, and this is likely to occur by the binding of the chemokines to common receptors.


Assuntos
Antígenos CD/metabolismo , Quimiocinas/metabolismo , Mucosa Intestinal/metabolismo , Linfócitos/metabolismo , Receptores de Interleucina/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CCL4 , Quimiotaxia de Leucócito , Epitélio/metabolismo , Humanos , Interleucina-8/metabolismo , Proteínas Inflamatórias de Macrófagos/metabolismo , Ligação Proteica , Receptores de Interleucina-8A
19.
Clin Exp Immunol ; 105(3): 556-60, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8809149

RESUMO

IL-4 is an important regulator of intestinal inflammation, yet little is known regarding its action on intestinal lymphocytes. Intestinal lymphocytes were isolated from jejunal mucosa of patients undergoing gastric bypass operations for morbid obesity. The impact of IL-4 was measured by its effects on proliferation using 3H-thymidine incorporation, IL-2 production using the CTLL assay, and IL-2 receptor generation using immunofluorescence. The production of IL-4 was measured by ELISA. IL-4 significantly inhibited the proliferation of intraepithelial lymphocytes (IEL) to a variety of stimuli as well as the development of lymphokine-activated killer (LAK) cells. In contrast, it had no effect on the proliferation of CD8+ T cells from peripheral blood. The inhibitory effect of IL-4 on IEL did not occur during activation, since IL-2 production and receptor expression were not altered. Rather, it occurred during cell cycling, since over 50% inhibition resulted whether IL-4 was added at the initiation of an IL-2-stimulated culture or after 24 or 48 h incubation. IL-4 was secreted by activated lamina propria lymphocytes (LPL) but not by IEL. IL-4, produced by activated LPL, may enter the epithelial compartment and down-regulate responsiveness of IEL.


Assuntos
Regulação para Baixo/imunologia , Interleucina-4/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Ativação Linfocitária/efeitos dos fármacos , Epitélio/efeitos dos fármacos , Epitélio/imunologia , Humanos
20.
Cell Immunol ; 171(2): 211-6, 1996 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8806789

RESUMO

A distinctive characteristic of human intestinal lymphocytes is their low responsiveness to stimuli of the CD3 pathway in vitro. This may be due to anergy resulting from CD3 stimulation without costimulation. Costimulatory molecules, such as HML-1, VLA-4, CD44, and CD28, may be lacking or unresponsive to ligation. Expression of these costimulatory markers was examined by immunofluorescent staining and flow cytometry. Their ability to costimulate intestinal lymphocytes was tested by measuring changes in proliferation, IL-2 production, and calcium ion mobilization. HML-1 was found on 87 +/- 8% of intraepithelial lymphocytes but only 52 +/- 10% of lamina propria lymphocytes; the density of expression was three times higher on the former. VLA-4 and CD44 were expressed on more than 80% of both lymphocyte types. CD28 was found on 31 +/- 26% of intraepithelial lymphocytes and 55 +/- 15% of lamina propria lymphocytes; virtually all CD4+ T cells and 15 to 26% of CD8+ T cells were CD28+. Of the four costimulatory molecules, ligation of only CD28 increased proliferation and IL-2 production; none affected calcium ion mobilization with stimulation through the CD3 pathway. The marker B7/BB1, a binding partner to CD28, was not present on lymphocytes or epithelial cells by flow cytometry. CD28 is the only one of these costimulatory molecules that enhanced CD3-induced functions of both intestinal lymphocyte types, but little ligand is available in vivo. Without coligation, CD3 activation may lead to anergy.


Assuntos
Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Linfócitos/imunologia , Transdução de Sinais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Cálcio/metabolismo , Células Cultivadas , Humanos , Jejuno/citologia , Jejuno/imunologia , Linfócitos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...