Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Metab ; 32(3): 420-436.e12, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32768387

RESUMEN

Dysregulated metabolism is a key driver of maladaptive tumor-reactive T lymphocytes within the tumor microenvironment. Actionable targets that rescue the effector activity of antitumor T cells remain elusive. Here, we report that the Sirtuin-2 (Sirt2) NAD+-dependent deacetylase inhibits T cell metabolism and impairs T cell effector functions. Remarkably, upregulation of Sirt2 in human tumor-infiltrating lymphocytes (TILs) negatively correlates with response to TIL therapy in advanced non-small-cell lung cancer. Mechanistically, Sirt2 suppresses T cell metabolism by targeting key enzymes involved in glycolysis, tricarboxylic acid-cycle, fatty acid oxidation, and glutaminolysis. Accordingly, Sirt2-deficient murine T cells exhibit increased glycolysis and oxidative phosphorylation, resulting in enhanced proliferation and effector functions and subsequently exhibiting superior antitumor activity. Importantly, pharmacologic inhibition of Sirt2 endows human TILs with these superior metabolic fitness and effector functions. Our findings unveil Sirt2 as an unexpected actionable target for reprogramming T cell metabolism to augment a broad spectrum of cancer immunotherapies.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Sirtuina 2/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Animales , Antineoplásicos/química , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/química , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Sirtuina 2/deficiencia , Sirtuina 2/metabolismo , Linfocitos T/metabolismo
2.
PLoS One ; 12(2): e0170197, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28152014

RESUMEN

Pancreatic cancer (PC) evades immune destruction by favoring the development of regulatory T cells (Tregs) that inhibit effector T cells. The transcription factor Ikaros is critical for lymphocyte development, especially T cells. We have previously shown that downregulation of Ikaros occurs as a result of its protein degradation by the ubiquitin-proteasome system in our Panc02 tumor-bearing (TB) mouse model. Mechanistically, we observed a deregulation in the balance between Casein Kinase II (CK2) and protein phosphatase 1 (PP1), which suggested that increased CK2 activity is responsible for regulating Ikaros' stability in our model. We also showed that this loss of Ikaros expression is associated with a significant decrease in CD4+ and CD8+ T cell percentages but increased CD4+CD25+ Tregs in TB mice. In this study, we evaluated the effects of the dietary flavonoid apigenin (API), on Ikaros expression and T cell immune responses. Treatment of splenocytes from naïve mice with (API) stabilized Ikaros expression and prevented Ikaros downregulation in the presence of murine Panc02 cells in vitro, similar to the proteasome inhibitor MG132. In vivo treatment of TB mice with apigenin (TB-API) improved survival, reduced tumor weights and prevented splenomegaly. API treatment also restored protein expression of some Ikaros isoforms, which may be attributed to its moderate inhibition of CK2 activity from splenocytes of TB-API mice. This partial restoration of Ikaros expression was accompanied by a significant increase in CD4+ and CD8+ T cell percentages and a reduction in Treg percentages in TB-API mice. In addition, CD8+ T cells from TB-API mice produced more IFN-γ and their splenocytes were better able to prime allogeneic CD8+ T cell responses compared to TB mice. These results provide further evidence that Ikaros is regulated by CK2 in our pancreatic cancer model. More importantly, our findings suggest that API may be a possible therapeutic agent for stabilizing Ikaros expression and function to maintain T cell homeostasis in murine PC.


Asunto(s)
Apigenina/uso terapéutico , Quinasa de la Caseína II/antagonistas & inhibidores , Factor de Transcripción Ikaros/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Linfocitos T/efectos de los fármacos , Animales , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Femenino , Homeostasis/efectos de los fármacos , Leupeptinas/uso terapéutico , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Inhibidores de Proteasoma/uso terapéutico , Proteína Fosfatasa 1/metabolismo , Linfocitos T/inmunología , Proteínas Supresoras de Tumor/metabolismo
3.
Cancer Immunol Immunother ; 62(5): 909-18, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23589106

RESUMEN

Myeloid-derived suppressor cells (MDSC) are one of the major factors limiting the efficacy of immune therapy. In a clinical trial of patients with extensive stage small cell lung cancer (SCLC), we tested the possibility that targeting MDSC can improve the induction of immune responses by a cancer vaccine. Forty-one patients with extensive stage SCLC were randomized into three arms: arm A--control, arm B--vaccination with dendritic cells transduced with wild-type p53, and arm C--vaccination in combination with MDSC targeted therapy with all-trans-retinoic acid (ATRA). Interim results of the ongoing clinical trial are presented. Pre-treatment levels of MDSC populations in patients from all three arms were similar. Vaccine alone did not affect the proportion of MDSC, whereas in patients treated with ATRA, the MDSC decreased more than twofold (p = 0.02). Before the start of treatment, no patients had detectable p53-specific responses in IFN-γ ELISPOT. Sequential measurements did not show positive p53 responses in any of the 14 patients from arm A. After immunization, only 3 out of 15 patients (20 %) from arm B developed a p53-specific response (p = 0.22). In contrast, in arm C, 5 out of 12 patients (41.7 %) had detectable p53 responses (p = 0.012). The proportion of granzyme B-positive CD8(+) T cells was increased only in patients from arm C but not in arm B. Depletion of MDSC substantially improved the immune response to vaccination, suggesting that this approach can be used to enhance the effect of immune interventions in cancer.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Neoplasias Pulmonares/inmunología , Carcinoma Pulmonar de Células Pequeñas/inmunología , Anciano , Citocinas/metabolismo , Células Dendríticas/citología , Femenino , Humanos , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Células Mieloides/citología , Fenotipo , Carcinoma Pulmonar de Células Pequeñas/terapia , Tretinoina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
4.
Biol Blood Marrow Transplant ; 18(8): 1182-1190.e1, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22698484

RESUMEN

Histone deacetylase inhibitors (HDACis) are a new class of compounds that induce acetylation of histone lysine tails in chromatin and modify gene expression. The Food & Drug Administration approved HDACi, Vorinostat, or suberoylanilide hydroxamic acid (SAHA), has been shown to inhibit tumor cell growth and the production of proinflammatory cytokines. In preclinical allogeneic transplant models, SAHA induces graft-versus-host disease (GVHD) amelioration in treated mice without impairing graft-versus-leukemia. LBH589 (Panobinostat), a structurally novel cinnamic hydroxamic acid class, is an HDACi more potent than SAHA. In the current work, we tested the hypothesis that LBH589 would be highly effective in the prevention of GVHD. Using mouse model of allogeneic bone marrow transplant (BMT), we unexpectedly found that treatment with LBH589 accelerated GVHD, in contrast to the treatment with SAHA that alleviated GVHD. Accelerated GVHD in the recipients treated with LBH589 was associated with elevated Th1 cytokines in recipient serum, enhanced CXCR3 expression on donor T cells, and T cell infiltration in the liver. The current study highlights the distinct effects of pan HDACi on allogeneic BMT and alerts that LBH589 (Panobinostat) could have an adverse effect on GVHD, and possibly on other inflammatory diseases.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Indoles/farmacología , Linfocitos T/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Femenino , Inhibidores de Histona Desacetilasas/efectos adversos , Ácidos Hidroxámicos/efectos adversos , Indoles/efectos adversos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Panobinostat , Linfocitos T/inmunología
5.
Am J Blood Res ; 2(1): 77-85, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22432091

RESUMEN

Bim, a BH3-only Bcl-2-family protein, is essential for T-cell negative selection in the thymus as well as for the death of activated T cells in the periphery. The role of Bim has been extensively studied in T-cell responses to self-antigens and viral infections. Recent findings on Bim in autoimmunity triggered our interest in investigating whether Bim may play a role in another disease with inflammatory symptoms as graft-versus-host disease (GVHD). Here we report that Bim is required for optimal T-cell responses to alloantigens in vivo and for the development of GVHD. Using murine models of allogeneic bone marrow transplantation (BMT), we found that donor T cells deficient for Bim are impaired in the induction of GVHD primarily due to a significant defect in T cell activation and expansion in vivo. Upon TCR engagement, Bim(-/-) T cells exhibited selective defects in CD69 expression and phosphorylation of PLCγ1. Our studies uncover a novel aspect of Bim function in T-cell activation with important implications in understanding the mechanisms of T-cell activation and tolerance under allogeneic transplantation.

6.
Int J Radiat Oncol Biol Phys ; 82(2): 924-32, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21398051

RESUMEN

PURPOSE: The goal of this study was to determine the effect of combination of intratumoral administration of dendritic cells (DC) and fractionated external beam radiation (EBRT) on tumor-specific immune responses in patients with soft-tissue sarcoma (STS). METHODS AND MATERIAL: Seventeen patients with large (>5 cm) high-grade STS were enrolled in the study. They were treated in the neoadjuvant setting with 5,040 cGy of EBRT, split into 28 fractions and delivered 5 days per week, combined with intratumoral injection of 10(7) DCs followed by complete resection. DCs were injected on the second, third, and fourth Friday of the treatment cycle. Clinical evaluation and immunological assessments were performed. RESULTS: The treatment was well tolerated. No patient had tumor-specific immune responses before combined EBRT/DC therapy; 9 patients (52.9%) developed tumor-specific immune responses, which lasted from 11 to 42 weeks. Twelve of 17 patients (70.6%) were progression free after 1 year. Treatment caused a dramatic accumulation of T cells in the tumor. The presence of CD4(+) T cells in the tumor positively correlated with tumor-specific immune responses that developed following combined therapy. Accumulation of myeloid-derived suppressor cells but not regulatory T cells negatively correlated with the development of tumor-specific immune responses. Experiments with (111)In labeled DCs demonstrated that these antigen presenting cells need at least 48 h to start migrating from tumor site. CONCLUSIONS: Combination of intratumoral DC administration with EBRT was safe and resulted in induction of antitumor immune responses. This suggests that this therapy is promising and needs further testing in clinical trials design to assess clinical efficacy.


Asunto(s)
Células Dendríticas/trasplante , Sarcoma/terapia , Neoplasias de los Tejidos Blandos/terapia , Adulto , Anciano , Anciano de 80 o más Años , Movimiento Celular , Terapia Combinada/métodos , Células Dendríticas/diagnóstico por imagen , Células Dendríticas/fisiología , Fraccionamiento de la Dosis de Radiación , Femenino , Humanos , Inmunidad Humoral/inmunología , Radioisótopos de Indio , Proteínas Inhibidoras de la Apoptosis/inmunología , Inyecciones Intralesiones , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Persona de Mediana Edad , Células Mieloides/inmunología , Terapia Neoadyuvante/métodos , Proteínas de Neoplasias/inmunología , Cintigrafía , Sarcoma/inmunología , Sarcoma/patología , Sarcoma/radioterapia , Neoplasias de los Tejidos Blandos/inmunología , Neoplasias de los Tejidos Blandos/patología , Neoplasias de los Tejidos Blandos/radioterapia , Survivin , Linfocitos T/inmunología , Factores de Tiempo
7.
Clin Cancer Res ; 16(6): 1812-23, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20215551

RESUMEN

PURPOSE: Myeloid-derived suppressor cells (MDSC) are one of the major factors responsible for immune suppression in cancer. Therefore, it would be important to identify effective therapeutic means to modulate these cells. EXPERIMENTAL DESIGN: We evaluated the effect of the synthetic triterpenoid C-28 methyl ester of 2-cyano-3,12-dioxooleana-1,9,-dien-28-oic acid (CDDO-Me; bardoxolone methyl) in MC38 colon carcinoma, Lewis lung carcinoma, and EL-4 thymoma mouse tumor models, as well as blood samples from patients with renal cell cancer and soft tissue sarcoma. Samples were also analyzed from patients with pancreatic cancer treated with CDDO-Me in combination with gemcitabine. RESULTS: CDDO-Me at concentrations of 25 to 100 nmol/L completely abrogated immune suppressive activity of MDSC in vitro. CDDO-Me reduced reactive oxygen species in MDSCs but did not affect their viability or the levels of nitric oxide and arginase. Treatment of tumor-bearing mice with CDDO-Me did not affect the proportion of MDSCs in the spleens but eliminated their suppressive activity. This effect was independent of antitumor activity. CDDO-Me treatment decreased tumor growth in mice. Experiments with severe combined immunodeficient-beige mice indicated that this effect was largely mediated by the immune system. CDDO-Me substantially enhanced the antitumor effect of a cancer vaccines. Treatment of pancreatic cancer patients with CDDO-Me did not affect the number of MDSCs in peripheral blood but significantly improved the immune response. CONCLUSIONS: CDDO-Me abrogated the immune suppressive effect of MDSCs and improved immune responses in tumor-bearing mice and cancer patients. It may represent an attractive therapeutic option by enhancing the effect of cancer immunotherapy.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Inmunosupresores/farmacología , Células Mieloides/inmunología , Neoplasias Experimentales/inmunología , Ácido Oleanólico/análogos & derivados , Anciano , Anciano de 80 o más Años , Animales , Arginasa/metabolismo , Ensayos Clínicos Fase I como Asunto , Células Dendríticas/patología , Células Dendríticas/trasplante , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Persona de Mediana Edad , Células Mieloides/patología , Células Mieloides/trasplante , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Óxido Nítrico/metabolismo , Ácido Oleanólico/farmacología , Pronóstico , Especies Reactivas de Oxígeno/metabolismo
8.
Biol Blood Marrow Transplant ; 16(2): 170-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19804837

RESUMEN

T helper (Th)1 cells were considered responsible for the induction of graft-versus-host disease (GVHD), but recently the concept has been challenged. Th17 cells play a critical role in mediating autoimmune diseases, but their role in the pathogenesis of GVHD remains unclear. Herein we compare the ability of in vitro generated Th1 and Th17 cells from C57BL/6 mice to induce GVHD in lethally irradiated BALB/c recipients. Allogeneic Th17 cells had superior expansion and infiltration capabilities in GVHD target organs, which correlated with their increased pathogenicity when compared with naïve or Th1 controls. Th17 cells caused no pathology in the syngeneic recipients, indicating that antigen-activation was required for their pathogenicity. Polarized Th17 cells could not maintain their phenotype in vivo as they produced a significant amount of interferon (IFN)-gamma after being transplanted into allogeneic recipients; however, IFN-gamma was not required for Th17 cell-induced GVHD. Further, we evaluated the pathogenesis of Th17 cells in GVHD by using polyclonal nonprimed CD4T cells in a clinically relevant allogeneic bone marrow transplantation (BMT) setting. We found that disruption of Th17-differentiation alone by targeting RORgammat (Th17-specific transcription factor) had no significant effect on GVHD development. We conclude that Th17 cells are sufficient but not necessary to induce GVHD.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Peso Corporal/efectos de los fármacos , Trasplante de Médula Ósea/efectos adversos , Trasplante de Médula Ósea/patología , Complejo CD3/fisiología , Células Cultivadas , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/prevención & control , Reacción Injerto-Huésped/efectos de los fármacos , Reacción Injerto-Huésped/inmunología , Interferón gamma/deficiencia , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos , Ratones Transgénicos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/deficiencia , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Subgrupos de Linfocitos T/trasplante , Linfocitos T Colaboradores-Inductores/clasificación , Linfocitos T Colaboradores-Inductores/trasplante , Células TH1/inmunología , Células TH1/trasplante , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Imagen de Cuerpo Entero
9.
J Clin Invest ; 119(12): 3774-86, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19907075

RESUMEN

When used as therapy for hematopoietic malignancies, allogeneic BM transplantation (BMT) relies on the graft-versus-leukemia (GVL) effect to eradicate residual tumor cells through immunologic mechanisms. However, graft-versus-host disease (GVHD), which is initiated by alloreactive donor T cells that recognize mismatched major and/or minor histocompatibility antigens and cause severe damage to hematopoietic and epithelial tissues, is a potentially lethal complication of allogeneic BMT. To enhance the therapeutic potential of BMT, we sought to find therapeutic targets that could inhibit GVHD while preserving GVL and immune responses to infectious agents. We show here that T cell responses triggered in mice by either Listeria monocytogenes or administration of antigen and adjuvant were relatively well preserved in the absence of PKC isoform theta (PKCtheta), a key regulator of TCR signaling. In contrast, PKCtheta was required for alloreactivity and GVHD induction. Furthermore, absence of PKCtheta raised the threshold for T cell activation, which selectively affected alloresponses. Most importantly, PKCtheta-deficient T cells retained the ability to respond to virus infection and to induce GVL effect after BMT. These findings suggest PKCtheta is a potentially unique therapeutic target required for GVHD induction but not for GVL or protective responses to infectious agents.


Asunto(s)
Enfermedad Injerto contra Huésped/enzimología , Efecto Injerto vs Leucemia/fisiología , Isoenzimas/inmunología , Leucemia Experimental/enzimología , Leucemia Experimental/inmunología , Proteína Quinasa C/inmunología , Infecciones por Retroviridae/enzimología , Infecciones por Retroviridae/inmunología , Animales , Trasplante de Médula Ósea/inmunología , Femenino , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/inmunología , Efecto Injerto vs Leucemia/inmunología , Técnicas In Vitro , Isoantígenos , Isoenzimas/deficiencia , Isoenzimas/genética , Listeria monocytogenes/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Proteína Quinasa C/deficiencia , Proteína Quinasa C/genética , Proteína Quinasa C-theta , Transducción de Señal , Linfocitos T/inmunología
10.
Blood ; 114(5): 1026-8, 2009 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-19429865

RESUMEN

Activation-induced cell death (AICD) plays an important role in peripheral T-cell tolerance. AICD in CD4 T helper (Th) cells, including Th1 and Th2 effectors, has been extensively studied. Recently, interleukin-17-producing CD4(+) T cells (Th17 cells) have been identified as a unique Th subset, but their susceptibility to AICD and the underlying molecular mechanisms have not been defined. In this study, we found that Th17 cells were significantly less susceptible to AICD than Th1 cells, and Th17 cell resistance to AICD is due to the high levels of c-Fas-associated death domain-like interleukin-1-converting enzyme inhibitory protein preventing Fas-mediated apoptosis. The resistance of Th17 cells to AICD reveals a novel mechanism to explain the high pathogenicity of Th17 cells in autoimmune diseases, and may also provide a rationale to generate tumor-specific Th17 cells for adoptive immunotherapy.


Asunto(s)
Apoptosis/fisiología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/fisiología , Tolerancia Inmunológica/fisiología , Activación de Linfocitos , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/biosíntesis , Células Cultivadas/inmunología , Proteína Ligando Fas/biosíntesis , Proteína Ligando Fas/fisiología , Genes Reporteros , Inmunización , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , ARN Interferente Pequeño/farmacología , Quimera por Radiación , Organismos Libres de Patógenos Específicos , Linfocitos T Colaboradores-Inductores/clasificación , Células TH1/inmunología , Proteína bcl-X/fisiología
11.
J Immunol ; 181(4): 2285-91, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18684917

RESUMEN

CD28 is required for the development of regulatory T cells (Tregs; CD4(+)CD25(+)Foxp3(+)) in the thymus and also contributes to their survival and homeostasis in the periphery. We studied whether and how CD28 and ICOS control the differentiation of Tregs from naive T cells. By using wild-type, CD28-, ICOS-, or CD28/ICOS-double knockout mice on C57BL/6 background as T cell sources, we found that CD28 is essential, whereas ICOS is dispensable, for the development and homeostasis of Tregs. Furthermore, the differentiation of Tregs from naive CD4(+)CD25(-) T cells in vivo also depends on CD28. The requirement of CD28 for Treg differentiation was mediated by IL-2, because neutralization of IL-2 with its specific mAb-blocked Treg differentiation from wild-type CD4(+)CD25(-) T cells and addition of IL-2 restored Treg differentiation from CD28(-/-) T cells. Other common gamma-chain cytokines, IL-4, IL-7, or IL-15, do not share such a role with IL-2. Although CD28 is required for the differentiation of Tregs from naive T cells, already generated Tregs do not depend on CD28 to exert their suppressive function. Our study reveals a new aspect of CD28 function in regulating T cell response.


Asunto(s)
Antígenos CD28/fisiología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Animales , Antígenos de Diferenciación de Linfocitos T/fisiología , Células 3T3 BALB , Antígenos CD4/biosíntesis , Células Cultivadas , Factores de Transcripción Forkhead/biosíntesis , Homeostasis/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fase de Descanso del Ciclo Celular/inmunología
12.
Arthritis Rheum ; 56(1): 43-57, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17195207

RESUMEN

OBJECTIVE: T cells deficient in CD28 expression have been implicated in the pathogenesis of rheumatoid arthritis (RA). Given that CD28-null T cells are functionally heterogeneous, we undertook this study to screen for novel receptors on these cells. METHODS: Seventy-two patients with RA (ages 35-84 years) and 53 healthy persons (32 young controls ages 19-34 years, 21 older controls ages 39-86 years) were recruited. Phenotypes and proliferative capacity of T cells from fresh leukocytes and of long-term cultures were monitored by flow cytometry. Lung biopsy specimens from patients with RA-associated interstitial pneumonitis (IP) were examined by immunohistochemistry. Receptor functionality was assessed by crosslinking bioassays. RESULTS: Chronic stimulation of CD28(+) T cells in vitro yielded progenies that lacked CD28 but that gained CD56. Ex vivo analysis of leukocytes from patients with extraarticular RA showed a higher frequency of CD56(+),CD28-null T cells than in patients with disease confined to the joints or in healthy controls. CD56(+),CD28-null T cells had nil capacity for proliferation, consistent with cellular senescence. CD56(+) T cells had skewed T cell receptor (TCR) alpha/beta-chain usage and restricted TCR third complementarity-determining region spectra. Histologic studies showed that CD56(+) T cells were components of cellular infiltrates in RA-associated IP. CD56 crosslinking on T cells sufficiently induced cytokine production, although CD56/TCR coligation induced higher production levels. CONCLUSION: Chronic activation of T cells induces counterregulation of CD28 and CD56 expression. The loss of CD28 is accompanied by the gain of CD56 that confers TCR-independent and TCR-dependent activation pathways. We propose that accumulation of CD56(+) T cells in RA contributes to maladaptive immune responses and that CD56(+) T cells are potential targets for therapy.


Asunto(s)
Envejecimiento/metabolismo , Artritis Reumatoide/metabolismo , Antígeno CD56/metabolismo , Linfocitos T/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/inmunología , Artritis Reumatoide/complicaciones , Artritis Reumatoide/inmunología , Biopsia , Técnicas de Cultivo de Célula , Femenino , Citometría de Flujo , Humanos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Enfermedades Pulmonares Intersticiales/complicaciones , Enfermedades Pulmonares Intersticiales/metabolismo , Enfermedades Pulmonares Intersticiales/patología , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Linfocitos T/inmunología
14.
Microbiol Immunol ; 48(6): 477-84, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15215622

RESUMEN

In gld mice, CD4 and 8-double-negative (DN) T cells as well as naive and memory-phenotype T cells accumulate in the peripheral lymphoid organs. Although Fas ligand (L) defect accounts for the progressive accumulation of abnormal DN T cells, the existence of other mechanisms which may be involved in the defective homeostasis in gld mice has been unclear. In this study, we analyze T-cell homeostasis in gld mice using adoptive transfer systems. It was shown that a gld, but not C57BL/6 (B6), environment led to augmented proliferation of B6 T cells transferred without up-regulation of CD69. Thus, the augmented T-cell proliferation seemed to result from mal-homeostatic proliferation even in the presence of a large number of recipient T cells. T cells from lpr mice showed no significant proliferation in the B6 environment, suggesting that the absence of Fas-Fas L interaction was not responsible for the mal-homeostatic proliferation. Although similar levels of IL-7 mRNA were detected in gld and B6 spleens, the intensity of CD127 and the proportion of CD127+ cells in the T cells were significantly lower in gld mice than in B6 mice, suggesting that IL-7 excess in a gld environment is responsible for the abnormal proliferation of transferred T cells. The administration of anti-CD127 antibody inhibited the proliferation of transferred lymphocytes. Thus, IL-7-dependent proliferation seems to be involved in the abnormal proliferation of lymphocytes in gld recipients.


Asunto(s)
Interleucina-7/fisiología , Linfocitos T/fisiología , Animales , Antígenos CD/análisis , Antígenos de Diferenciación de Linfocitos T/análisis , Antígenos CD4/análisis , Antígenos CD8/análisis , División Celular , Proteína Ligando Fas , Femenino , Homeostasis , Interleucina-7/análisis , Lectinas Tipo C , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Receptores de Interleucina-7/análisis , Linfocitos T/inmunología
15.
Transpl Immunol ; 12(1): 79-88, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14551035

RESUMEN

T cells acquire immune functions during expansion and differentiation in the thymus. Mature T cells respond to peptide antigens (Ag) derived from foreign proteins when these peptide Ag are presented on the self major histocompatibility complex (MHC) molecules but not on allo-MHC. This is termed self-MHC restriction. On the other hand, T cells do not induce aggressive responses to self Ag (self-tolerance). Self-MHC restriction and self-tolerance are not genetically determined but acquired a posteriori by positive and negative selection in the thymus in harmony with the functional maturation. Allogeneic bone marrow (BM) chimera systems have been a useful strategy to elucidate mechanisms underlying positive and negative selection. In this communication, the contribution of BM chimera systems to the investigation of the world of T-ology is discussed.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Diferenciación Celular/inmunología , Linfocitos T/inmunología , Quimera por Trasplante/inmunología , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos de Diferenciación de Linfocitos T/análisis , Supresión Clonal/inmunología , Columbidae , Citocromos c/genética , Citocromos c/inmunología , Citometría de Flujo , Reacción Injerto-Huésped/inmunología , Tolerancia Inmunológica/inmunología , Activación de Linfocitos/inmunología , Prueba de Cultivo Mixto de Linfocitos , Depleción Linfocítica , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Ratones Endogámicos AKR , Ratones Endogámicos C57BL , Modelos Inmunológicos , Péptidos/genética , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/química , Linfocitos T Citotóxicos/inmunología , Timo/citología , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...