Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(24): e2403054121, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38838017

RESUMEN

Chronic Toxoplasma gondii infection induces brain-resident CD8+ T cells (bTr), but the protective functions and differentiation cues of these cells remain undefined. Here, we used a mouse model of latent infection by T. gondii leading to effective CD8+ T cell-mediated parasite control. Thanks to antibody depletion approaches, we found that peripheral circulating CD8+ T cells are dispensable for brain parasite control during chronic stage, indicating that CD8+ bTr are able to prevent brain parasite reactivation. We observed that the retention markers CD69, CD49a, and CD103 are sequentially acquired by brain parasite-specific CD8+ T cells throughout infection and that a majority of CD69/CD49a/CD103 triple-positive (TP) CD8+ T cells also express Hobit, a transcription factor associated with tissue residency. This TP subset develops in a CD4+ T cell-dependent manner and is associated with effective parasite control during chronic stage. Conditional invalidation of Transporter associated with Antigen Processing (TAP)-mediated major histocompatibility complex (MHC) class I presentation showed that presentation of parasite antigens by glutamatergic neurons and microglia regulates the differentiation of CD8+ bTr into TP cells. Single-cell transcriptomic analyses revealed that resistance to encephalitis is associated with the expansion of stem-like subsets of CD8+ bTr. In summary, parasite-specific brain-resident CD8+ T cells are a functionally heterogeneous compartment which autonomously ensure parasite control during T. gondii latent infection and which differentiation is shaped by neuronal and microglial MHC I presentation. A more detailed understanding of local T cell-mediated immune surveillance of this common parasite is needed for harnessing brain-resident CD8+ T cells in order to enhance control of chronic brain infections.


Asunto(s)
Encéfalo , Linfocitos T CD8-positivos , Diferenciación Celular , Toxoplasma , Toxoplasmosis , Animales , Linfocitos T CD8-positivos/inmunología , Toxoplasma/inmunología , Ratones , Encéfalo/inmunología , Encéfalo/parasitología , Diferenciación Celular/inmunología , Toxoplasmosis/inmunología , Toxoplasmosis/parasitología , Infección Latente/inmunología , Infección Latente/parasitología , Antígenos CD/metabolismo , Antígenos CD/inmunología , Antígenos CD/genética , Ratones Endogámicos C57BL , Femenino
2.
Curr Opin Immunol ; 77: 102211, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35644112

RESUMEN

Tissue-resident memory T cells (TRM) represent a subset of antigen-experienced T cells that are constantly retained in a given tissue with limited trafficking through the circulation. These cells are characterized by expression of molecules enabling their tissue anchoring and downregulation of molecules promoting tissue egress. They reside at sites of previous antigen encounter and their number increases with age. TRM have been shown to provide rapid and efficient protection against tissue reinfection and TRM density correlates with efficient antitumor responses. Intriguingly, the density of CD8 TRM is increased in the central nervous system (CNS) of patients with neuroinflammatory diseases such as multiple sclerosis, or suffering from neurodegenerative diseases. In this review, we discuss current knowledge regarding the diversity of CNS-resident CD8 T cells and their role in CNS autoimmunity. Given their likely contribution to the protracted course of several inflammatory diseases of the CNS, their therapeutic targeting becomes an important challenge.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Antígenos/metabolismo , Sistema Nervioso Central , Crimen , Humanos
3.
Brain ; 145(6): 2018-2030, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35552381

RESUMEN

Narcolepsy with cataplexy or narcolepsy type 1 is a disabling chronic sleep disorder resulting from the destruction of orexinergic neurons in the hypothalamus. The tight association of narcolepsy with HLA-DQB1*06:02 strongly suggest an autoimmune origin to this disease. Furthermore, converging epidemiological studies have identified an increased incidence for narcolepsy in Europe following Pandemrix® vaccination against the 2009-2010 pandemic 'influenza' virus strain. The potential immunological link between the Pandemrix® vaccination and narcolepsy remains, however, unknown. Deciphering these mechanisms may reveal pathways potentially at play in most cases of narcolepsy. Here, we developed a mouse model allowing to track and study the T-cell response against 'influenza' virus haemagglutinin, which was selectively expressed in the orexinergic neurons as a new self-antigen. Pandemrix® vaccination in this mouse model resulted in hypothalamic inflammation and selective destruction of orexin-producing neurons. Further investigations on the relative contribution of T-cell subsets in this process revealed that haemagglutinin-specific CD4 T cells were necessary for the development of hypothalamic inflammation, but insufficient for killing orexinergic neurons. Conversely, haemagglutinin-specific CD8 T cells could not initiate inflammation but were the effectors of the destruction of orexinergic neurons. Additional studies revealed pathways potentially involved in the disease process. Notably, the interferon-γ pathway was proven essential, as interferon-γ-deficient CD8 T cells were unable to elicit the loss of orexinergic neurons. Our work demonstrates that an immunopathological process mimicking narcolepsy can be elicited by immune cross-reactivity between a vaccine antigen and a neuronal self-antigen. This process relies on a synergy between autoreactive CD4 and CD8 T cells for disease development. This work furthers our understanding of the mechanisms and pathways potentially involved in the development of a neurological side effect due to a vaccine and, likely, to narcolepsy in general.


Asunto(s)
Autoinmunidad , Vacunas contra la Influenza , Narcolepsia , Animales , Autoantígenos , Hemaglutininas , Inflamación/complicaciones , Vacunas contra la Influenza/efectos adversos , Interferón gamma , Ratones , Narcolepsia/inducido químicamente , Neuronas , Orexinas , Linfocitos T/inmunología , Vacunación/efectos adversos
4.
Sci Transl Med ; 14(640): eabl6157, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35417189

RESUMEN

The mechanisms underlying the chronicity of autoimmune diseases of the central nervous system (CNS) are largely unknown. In particular, it is unclear whether tissue-resident memory T cells (TRM) contribute to lesion pathogenesis during chronic CNS autoimmunity. Here, we observed that a high frequency of brain-infiltrating CD8+ T cells exhibit a TRM-like phenotype in human autoimmune encephalitis. Using mouse models of neuronal autoimmunity and a combination of T single-cell transcriptomics, high-dimensional flow cytometry, and histopathology, we found that pathogenic CD8+ T cells behind the blood-brain barrier adopt a characteristic TRM differentiation program, and we revealed their phenotypic and functional heterogeneity. In the diseased CNS, autoreactive tissue-resident CD8+ T cells sustained focal neuroinflammation and progressive loss of neurons, independently of recirculating CD8+ T cells. Consistently, a large fraction of autoreactive tissue-resident CD8+ T cells exhibited proliferative potential as well as proinflammatory and cytotoxic properties. Persistence of tissue-resident CD8+ T cells in the CNS and their functional output, but not their initial differentiation, were crucially dependent on CD4+ T cells. Collectively, our results point to tissue-resident CD8+ T cells as essential drivers of chronic CNS autoimmunity and suggest that therapies targeting this compartmentalized autoreactive T cell subset might be effective for treating CNS autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Linfocitos T CD8-positivos , Animales , Enfermedades Autoinmunes/patología , Sistema Nervioso Central , Memoria Inmunológica , Ratones , Neuronas
5.
Cancer Immunol Res ; 9(7): 735-747, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33906864

RESUMEN

IL11 is a member of the IL6 family of cytokines and signals through its cognate receptor subunits, IL11RA and glycoprotein 130 (GP130), to elicit biological responses via the JAK/STAT signaling pathway. IL11 contributes to cancer progression by promoting the survival and proliferation of cancer cells, but the potential immunomodulatory properties of IL11 signaling during tumor development have thus far remained unexplored. Here, we have characterized a role for IL11 in regulating CD4+ T cell-mediated antitumor responses. Absence of IL11 signaling impaired tumor growth in a sporadic mouse model of colon cancer and syngeneic allograft models of colon cancer. Adoptive bone marrow transfer experiments and in vivo depletion studies demonstrated that the tumor-promoting activity of IL11 was mediated through its suppressive effect on host CD4+ T cells in the tumor microenvironment. Indeed, when compared with Il11ra-proficient CD4+ T cells associated with MC38 tumors, their Il11ra-deficient counterparts displayed elevated expression of mRNA encoding the antitumor mediators IFNγ and TNFα. Likewise, IL11 potently suppressed the production of proinflammatory cytokines (IFNγ, TNFα, IL6, and IL12p70) by CD4+ T cells in vitro, which we corroborated by RNAscope analysis of human colorectal cancers, where IL11RAhigh tumors showed less IFNG and CD4 expression than IL11RAlow tumors. Therefore, our results ascribe a tumor cell-extrinsic immunomodulatory role to IL11 during colon cancer development that could be amenable to an anticytokine-based therapy.See related Spotlight by van der Burg, p. 724.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Neoplasias del Colon/inmunología , Subunidad alfa del Receptor de Interleucina-11/metabolismo , Interleucina-11/metabolismo , Animales , Antígenos CD4/análisis , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Colon/inmunología , Colon/patología , Neoplasias del Colon/patología , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Interferón gamma/análisis , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-11/análisis , Subunidad alfa del Receptor de Interleucina-11/genética , Ratones , Ratones Noqueados , Neoplasias de Tejido Óseo , Receptores de Interleucina-11/metabolismo , Microambiente Tumoral/inmunología
6.
Nat Commun ; 10(1): 2735, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31227713

RESUMEN

The contribution of mast cells in the microenvironment of solid malignancies remains controversial. Here we functionally assess the impact of tumor-adjacent, submucosal mast cell accumulation in murine and human intestinal-type gastric cancer. We find that genetic ablation or therapeutic inactivation of mast cells suppresses accumulation of tumor-associated macrophages, reduces tumor cell proliferation and angiogenesis, and diminishes tumor burden. Mast cells are activated by interleukin (IL)-33, an alarmin produced by the tumor epithelium in response to the inflammatory cytokine IL-11, which is required for the growth of gastric cancers in mice. Accordingly, ablation of the cognate IL-33 receptor St2 limits tumor growth, and reduces mast cell-dependent production and release of the macrophage-attracting factors Csf2, Ccl3, and Il6. Conversely, genetic or therapeutic macrophage depletion reduces tumor burden without affecting mast cell abundance. Therefore, tumor-derived IL-33 sustains a mast cell and macrophage-dependent signaling cascade that is amenable for the treatment of gastric cancer.


Asunto(s)
Interleucina-33/inmunología , Macrófagos/inmunología , Mastocitos/inmunología , Neoplasias Gástricas/inmunología , Aminopiridinas/administración & dosificación , Animales , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Cromolin Sódico/administración & dosificación , Modelos Animales de Enfermedad , Epitelio/inmunología , Epitelio/patología , Femenino , Mucosa Gástrica/citología , Mucosa Gástrica/inmunología , Mucosa Gástrica/patología , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Masculino , Ratones , Ratones Transgénicos , Pirroles/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Análisis de Matrices Tisulares , Microambiente Tumoral/inmunología
7.
Cancer Immunol Res ; 6(4): 409-421, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29463593

RESUMEN

Interleukin 33 (IL33) is an inflammatory cytokine released during necrotic cell death. The epithelium and stroma of the intestine express large amounts of IL33 and its receptor St2. IL33 is therefore continuously released during homeostatic turnover of the intestinal mucosa. Although IL33 can prevent colon cancer associated with inflammatory colitis, the contribution of IL33 signaling to sporadic colon cancer remains unknown. Here, we utilized a mouse model of sporadic colon cancer to investigate the contribution of IL33 signaling to tumorigenesis in the absence of preexisting inflammation. We demonstrated that genetic ablation of St2 enhanced colon tumor development. Conversely, administration of recombinant IL33 reduced growth of colon cancer cell allografts. In reciprocal bone marrow chimeras, the concurrent loss of IL33 signaling within radioresistant nonhematopoietic, and the radiosensitive hematopoietic, compartments was associated with increased tumor burden. We detected St2 expression within the radioresistant mesenchymal cell compartment of the colon whose stimulation with IL33 induced expression of bona fide NF-κB target genes. Mechanistically, we discovered that St2 deficiency within the nonhematopoietic compartment coincided with increased abundance of regulatory T cells and suppression of an IFNγ gene expression signature, whereas IL33 administration triggered IFNγ expression by tumor allograft-infiltrating T cells. The decrease of this IFNγ gene expression signature was associated with more aggressive disease in human colon cancer patients, suggesting that lack of IL33 signaling impaired the generation of a potent IFNγ-mediated antitumor immune response. Collectively, our data reveal that IL33 functions as a tumor suppressor in sporadic colon cancer. Cancer Immunol Res; 6(4); 409-21. ©2018 AACR.


Asunto(s)
Neoplasias del Colon/metabolismo , Interferón gamma/metabolismo , Interleucina-33/metabolismo , Transducción de Señal , Aloinjertos , Animales , Biomarcadores , Biopsia , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Interferón gamma/genética , Interleucina-33/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Ratones , FN-kappa B/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Transcriptoma
8.
Cancer Cell ; 31(4): 563-575.e5, 2017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28399411

RESUMEN

Aberrant activation of the SRC family kinase hematopoietic cell kinase (HCK) triggers hematological malignancies as a tumor cell-intrinsic oncogene. Here we find that high HCK levels correlate with reduced survival of colorectal cancer patients. Likewise, increased Hck activity in mice promotes the growth of endogenous colonic malignancies and of human colorectal cancer cell xenografts. Furthermore, tumor-associated macrophages of the corresponding tumors show a pronounced alternatively activated endotype, which occurs independently of mature lymphocytes or of Stat6-dependent Th2 cytokine signaling. Accordingly, pharmacological inhibition or genetic reduction of Hck activity suppresses alternative activation of tumor-associated macrophages and the growth of colon cancer xenografts. Thus, Hck may serve as a promising therapeutic target for solid malignancies.


Asunto(s)
Neoplasias del Colon/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/mortalidad , Proteínas Proto-Oncogénicas c-hck/metabolismo , Animales , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Activación de Macrófagos/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-hck/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-hck/genética , Pirimidinas/farmacología , Pirroles/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Células del Estroma/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Cell ; 30(1): 59-74, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27374225

RESUMEN

E proteins and their antagonists, the Id proteins, are transcriptional regulators important for normal hematopoiesis. We found that Id2 acts as a key regulator of leukemia stem cell (LSC) potential in MLL-rearranged acute myeloid leukemia (AML). Low endogenous Id2 expression is associated with LSC enrichment while Id2 overexpression impairs MLL-AF9-leukemia initiation and growth. Importantly, MLL-AF9 itself controls the E-protein pathway by suppressing Id2 while directly activating E2-2 expression, and E2-2 depletion phenocopies Id2 overexpression in MLL-AF9-AML cells. Remarkably, Id2 tumor-suppressive function is conserved in t(8;21) AML. Low expression of Id2 and its associated gene signature are associated with poor prognosis in MLL-rearranged and t(8;21) AML patients, identifying the Id2/E-protein axis as a promising new therapeutic target in AML.


Asunto(s)
Proteína 2 Inhibidora de la Diferenciación/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica/genética , Proteína 2 Similar al Factor de Transcripción 7/genética , Translocación Genética , Animales , Proliferación Celular , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Regulación Leucémica de la Expresión Génica , Humanos , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Leucemia Mieloide Aguda/metabolismo , Ratones , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Neoplasias Experimentales , Proteínas de Fusión Oncogénica/metabolismo , Pronóstico , Células Madre/citología , Células Madre/metabolismo , Análisis de Supervivencia , Proteína 2 Similar al Factor de Transcripción 7/metabolismo
10.
J Exp Med ; 213(6): 1095-111, 2016 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-27217539

RESUMEN

The generation of high-affinity antibodies requires germinal center (GC) development and differentiation of long-lived plasma cells in a multilayered process that is tightly controlled by the activity of multiple transcription factors. Here, we reveal a new layer of complexity by demonstrating that dynamic changes in Id3 and E-protein activity govern both GC and plasma cell differentiation. We show that down-regulation of Id3 in B cells is essential for releasing E2A and E2-2, which in a redundant manner are required for antigen-induced B cell differentiation. We demonstrate that this pathway controls the expression of multiple key factors, including Blimp1, Xbp1, and CXCR4, and is therefore critical for establishing the transcriptional network that controls GC B cell and plasma cell differentiation.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Diferenciación Celular/inmunología , Centro Germinal/inmunología , Proteínas Inhibidoras de la Diferenciación/inmunología , Células Plasmáticas/inmunología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Proteínas Inhibidoras de la Diferenciación/genética , Ratones , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Factor de Transcripción 4 , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/inmunología
11.
Nat Immunol ; 17(4): 422-32, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26950239

RESUMEN

T cell responses are guided by cytokines that induce transcriptional regulators, which ultimately control differentiation of effector and memory T cells. However, it is unknown how the activities of these molecular regulators are coordinated and integrated during the differentiation process. Using genetic approaches and transcriptional profiling of antigen-specific CD8(+) T cells, we reveal a common program of effector differentiation that is regulated by IL-2 and IL-12 signaling and the combined activities of the transcriptional regulators Blimp-1 and T-bet. The loss of both T-bet and Blimp-1 leads to abrogated cytotoxic function and ectopic IL-17 production in CD8(+) T cells. Overall, our data reveal two major overlapping pathways of effector differentiation governed by the availability of Blimp-1 and T-bet and suggest a model for cytokine-induced transcriptional changes that combine, quantitatively and qualitatively, to promote robust effector CD8(+) T cell differentiation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Interleucina-12/inmunología , Interleucina-2/inmunología , Proteínas de Dominio T Box/inmunología , Factores de Transcripción/inmunología , Animales , Infecciones por Arenaviridae/inmunología , Inmunoprecipitación de Cromatina , Citocinas/inmunología , Citometría de Flujo , Perfilación de la Expresión Génica , Subtipo H1N1 del Virus de la Influenza A , Interleucina-17/inmunología , Virus de la Coriomeningitis Linfocítica , Ratones , Infecciones por Orthomyxoviridae/inmunología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT4/inmunología , Factor de Transcripción STAT5/inmunología , Análisis de Secuencia de ARN , Transducción de Señal
12.
Immunity ; 43(6): 1101-11, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26682984

RESUMEN

Tissue-resident memory T (Trm) cells contribute to local immune protection in non-lymphoid tissues such as skin and mucosa, but little is known about their transcriptional regulation. Here we showed that CD8(+)CD103(+) Trm cells, independent of circulating memory T cells, were sufficient for protection against infection and described molecular elements that were crucial for their development in skin and lung. We demonstrated that the T-box transcription factors (TFs) Eomes and T-bet combined to control CD8(+)CD103(+) Trm cell formation, such that their coordinate downregulation was crucial for TGF-ß cytokine signaling. TGF-ß signaling, in turn, resulted in reciprocal T-box TF downregulation. However, whereas extinguishment of Eomes was necessary for CD8(+)CD103(+) Trm cell development, residual T-bet expression maintained cell surface interleukin-15 (IL-15) receptor ß-chain (CD122) expression and thus IL-15 responsiveness. These findings indicate that the T-box TFs control the two cytokines, TGF-ß and IL-15, which are pivotal for CD8(+)CD103(+) Trm cell development and survival.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Interleucina-15/inmunología , Proteínas de Dominio T Box/inmunología , Factor de Crecimiento Transformador beta/inmunología , Traslado Adoptivo , Animales , Regulación hacia Abajo , Citometría de Flujo , Regulación de la Expresión Génica/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Subgrupos de Linfocitos T/inmunología
13.
Dis Model Mech ; 8(8): 805-15, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26035389

RESUMEN

The cells of the intestinal epithelium provide a selectively permeable barrier between the external environment and internal tissues. The integrity of this barrier is maintained by tight junctions, specialised cell-cell contacts that permit the absorption of water and nutrients while excluding microbes, toxins and dietary antigens. Impairment of intestinal barrier function contributes to multiple gastrointestinal disorders, including food hypersensitivity, inflammatory bowel disease (IBD) and colitis-associated cancer (CAC). Glycoprotein A33 (GPA33) is an intestinal epithelium-specific cell surface marker and member of the CTX group of transmembrane proteins. Roles in cell-cell adhesion have been demonstrated for multiple CTX family members, suggesting a similar function for GPA33 within the gastrointestinal tract. To test a potential requirement for GPA33 in intestinal barrier function, we generated Gpa33(-/-) mice and subjected them to experimental regimens designed to produce food hypersensitivity, colitis and CAC. Gpa33(-/-) mice exhibited impaired intestinal barrier function. This was shown by elevated steady-state immunosurveillance in the colonic mucosa and leakiness to oral TRITC-labelled dextran after short-term exposure to dextran sodium sulphate (DSS) to injure the intestinal epithelium. Gpa33(-/-) mice also exhibited rapid onset and reduced resolution of DSS-induced colitis, and a striking increase in the number of colitis-associated tumours produced by treatment with the colon-specific mutagen azoxymethane (AOM) followed by two cycles of DSS. In contrast, Gpa33(-/-) mice treated with AOM alone showed no increase in sporadic tumour formation, indicating that their increased tumour susceptibility is dependent on inflammatory stimuli. Finally, Gpa33(-/-) mice displayed hypersensitivity to food allergens, a common co-morbidity in humans with IBD. We propose that Gpa33(-/-) mice provide a valuable model to study the mechanisms linking intestinal permeability and multiple inflammatory pathologies. Moreover, this model could facilitate preclinical studies aimed at identifying drugs that restore barrier function.


Asunto(s)
Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Glicoproteínas de Membrana/deficiencia , Animales , Antígenos/inmunología , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/patología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Alimentos , Tolerancia Inmunológica/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL
14.
Blood ; 123(22): 3420-8, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24723679

RESUMEN

Interleukin-10 (IL-10) is a key immunoregulatory cytokine that functions to prevent inflammatory and autoimmune diseases. Despite the critical role for IL-10 produced by effector CD8(+) T cells during pathogen infection and autoimmunity, the mechanisms regulating its production are poorly understood. We show that loss of the inhibitor of DNA binding 2 (Id2) in T cells resulted in aberrant IL-10 expression in vitro and in vivo during influenza virus infection and in a model of acute graft-versus-host disease (GVHD). Furthermore, IL-10 overproduction substantially reduced the immunopathology associated with GVHD. We demonstrate that Id2 acts to repress the E2A-mediated trans-activation of the Il10 locus. Collectively, our findings uncover a key regulatory role of Id2 during effector T cell differentiation necessary to limit IL-10 production by activated T cells and minimize their suppressive activity during the effector phase of disease control.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Interleucina-10/genética , Subgrupos de Linfocitos T/metabolismo , Activación Transcripcional , Animales , Médula Ósea/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Epigénesis Genética , Regulación de la Expresión Génica , Sitios Genéticos , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/mortalidad , Proteína 2 Inhibidora de la Diferenciación/deficiencia , Proteína 2 Inhibidora de la Diferenciación/genética , Interleucina-10/metabolismo , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/mortalidad
15.
Nat Med ; 20(3): 283-90, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24487434

RESUMEN

Loss of function of the tumor suppressor gene PRDM1 (also known as BLIMP1) or deregulated expression of the oncogene BCL6 occurs in a large proportion of diffuse large B cell lymphoma (DLBCL) cases. However, targeted mutation of either gene in mice leads to only slow and infrequent development of malignant lymphoma, and despite frequent mutation of BCL6 in activated B cells of healthy individuals, lymphoma development is rare. Here we show that T cells prevent the development of overt lymphoma in mice caused by Blimp1 deficiency or overexpression of Bcl6 in the B cell lineage. Impairment of T cell control results in rapid development of DLBCL-like disease, which can be eradicated by polyclonal CD8(+) T cells in a T cell receptor-, CD28- and Fas ligand-dependent manner. Thus, malignant transformation of mature B cells requires mutations that impair intrinsic differentiation processes and permit escape from T cell-mediated tumor surveillance.


Asunto(s)
Linfocitos B/patología , Proteína Ligando Fas/metabolismo , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/inmunología , Linfocitos T/inmunología , Factores de Transcripción/genética , Animales , Linfocitos T CD8-positivos/citología , Separación Celular , Transformación Celular Neoplásica , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Genes Supresores de Tumor , Vigilancia Inmunológica/genética , Ratones , Ratones Transgénicos , Mutación , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Proto-Oncogénicas c-bcl-6 , Factores de Transcripción/fisiología
16.
J Immunol ; 191(8): 4383-91, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24038093

RESUMEN

Innate lymphocyte populations play a central role in conferring protective immunity at the mucosal frontier. In this study, we demonstrate that T cell factor 1 (TCF-1; encoded by Tcf7), a transcription factor also important for NK and T cell differentiation, is expressed by multiple innate lymphoid cell (ILC) subsets, including GATA3(+) nuocytes (ILC2) and NKp46(+) ILCs (ILC3), which confer protection against lung and intestinal inflammation. TCF-1 was intrinsically required for the differentiation of both ILC2 and NKp46(+) ILC3. Loss of TCF-1 expression impaired the capacity of these ILC subsets to produce IL-5, IL-13, and IL-22 and resulted in crippled responses to intestinal infection with Citrobacter rodentium. Furthermore, a reduction in T-bet expression required for Notch-2-dependent development of NKp46(+) ILC3 showed a dose-dependent reduction in TCF-1 expression. Collectively, our findings demonstrate an essential requirement for TCF-1 in ILC2 differentiation and reveal a link among Tcf7, Notch, and Tbx21 in NKp46(+) ILC3 development.


Asunto(s)
Intestinos/inmunología , Células Asesinas Naturales/metabolismo , Factor 1 de Transcripción de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Animales , Antígenos Ly/metabolismo , Diferenciación Celular/inmunología , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Factor de Transcripción GATA3/metabolismo , Factor Nuclear 1-alfa del Hepatocito , Inflamación/inmunología , Inflamación/microbiología , Interleucina-13/biosíntesis , Interleucina-5/biosíntesis , Interleucinas/biosíntesis , Intestinos/microbiología , Activación de Linfocitos , Ratones , Ratones Noqueados , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptor Notch2/metabolismo , Factor 1 de Transcripción de Linfocitos T/genética , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/inmunología , Interleucina-22
17.
J Immunol ; 190(9): 4585-94, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23536629

RESUMEN

The transcription factor inhibitor of DNA binding (Id)2 modulates T cell fate decisions, but the molecular mechanism underpinning this regulation is unclear. In this study we show that loss of Id2 cripples effector differentiation and instead programs CD8(+) T cells to adopt a memory fate with increased Eomesodermin and Tcf7 expression. We demonstrate that Id2 restrains CD8(+) T cell memory differentiation by inhibiting E2A-mediated direct activation of Tcf7 and that Id2 expression level mirrors T cell memory recall capacity. As a result of the defective effector differentiation, Id2-deficient CD8(+) T cells fail to induce sufficient Tbx21 expression to generate short-lived effector CD8(+) T cells. Our findings reveal that the Id2/E2A axis orchestrates T cell differentiation through the induction or repression of downstream transcription factors essential for effector and memory T cell differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Memoria Inmunológica/inmunología , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Expresión Génica/inmunología , Factor Nuclear 1-alfa del Hepatocito , Memoria Inmunológica/genética , Proteína 2 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/inmunología , Ratones , Ratones Endogámicos C57BL , Factor 1 de Transcripción de Linfocitos T/genética , Factor 1 de Transcripción de Linfocitos T/inmunología , Factor 1 de Transcripción de Linfocitos T/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología , Proteínas de Dominio T Box/metabolismo
18.
EMBO J ; 30(13): 2690-704, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21587207

RESUMEN

Dendritic cells (DCs) have critical roles in the induction of the adaptive immune response. The transcription factors Id2, Batf3 and Irf-8 are required for many aspects of murine DC differentiation including development of CD8α(+) and CD103(+) DCs. How they regulate DC subset specification is not completely understood. Using an Id2-GFP reporter system, we show that Id2 is broadly expressed in all cDC subsets with the highest expression in CD103(+) and CD8α(+) lineages. Notably, CD103(+) DCs were the only DC able to constitutively cross-present cell-associated antigens in vitro. Irf-8 deficiency affected loss of development of virtually all conventional DCs (cDCs) while Batf3 deficiency resulted in the development of Sirp-α(-) DCs that had impaired survival. Exposure to GM-CSF during differentiation induced expression of CD103 in Id2-GFP(+) DCs. It did not restore cross-presenting capacity to Batf3(-/-) or CD103(-)Sirp-α(-)DCs in vitro. Thus, Irf-8 and Batf3 regulate distinct stages in DC differentiation during the development of cDCs. Genetic mapping DC subset differentiation using Id2-GFP may have broad implications in understanding the interplay of DC subsets during protective and pathological immune responses.


Asunto(s)
Antígenos CD/metabolismo , Antígenos CD8/metabolismo , Linaje de la Célula/genética , Células Dendríticas/fisiología , Proteína 2 Inhibidora de la Diferenciación/genética , Cadenas alfa de Integrinas/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Células Dendríticas/metabolismo , Expresión Génica/fisiología , Genes cdc/fisiología , Proteína 2 Inhibidora de la Diferenciación/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos
19.
Nat Immunol ; 12(4): 304-11, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21378976

RESUMEN

Regulatory T cells (T(reg) cells) are required for peripheral tolerance. Evidence indicates that T(reg) cells can adopt specialized differentiation programs in the periphery that are controlled by transcription factors usually associated with helper T cell differentiation. Here we demonstrate that expression of the transcription factor Blimp-1 defined a population of T(reg) cells that localized mainly to mucosal sites and produced IL-10. Blimp-1 was required for IL-10 production by these cells and for their tissue homeostasis. We provide evidence that the transcription factor IRF4, but not the transcription factor T-bet, was essential for Blimp-1 expression and for the differentiation of all effector T(reg) cells. Thus, our study defines a differentiation pathway that leads to the acquisition of T(reg) cell effector functions and requires both IRF4 and Blimp-1.


Asunto(s)
Diferenciación Celular/genética , Factores Reguladores del Interferón/genética , Linfocitos T Reguladores/metabolismo , Factores de Transcripción/genética , Animales , Secuencia de Bases , Sitios de Unión/genética , Células Cultivadas , Citometría de Flujo , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Factores Reguladores del Interferón/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Linfocitos T Reguladores/citología , Factores de Transcripción/metabolismo
20.
J Immunol ; 186(7): 4059-66, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21339359

RESUMEN

Upon Ag encounter, naive T cells undergo extensive Ag-driven proliferation and can differentiate into effector cells. Up to 95% of these cells die leaving a small residual population of T cells that provide protective memory. In this study, we investigated the contribution of the BH3-only family protein Bid in the shutdown of T cell responses after acute and persistent infection. Influenza virus pathogenicity has been proposed to be mediated by a peptide encoded in the basic polymerase (PB1-RF2) acting through Bid. In our experiments, we found that after acute infection with influenza virus, mice lacking Bid had normal expansion and contraction of Ag-specific CD8(+) T cells. However, in chronic γ-herpesvirus infection, Bid-deficient virus-specific CD8(+) T cells expanded normally but failed to contract fully and were maintained at ∼2-fold higher levels. Previously, we have demonstrated that Bim plays a prominent role in T cell shutdown in persistent infection by cooperating with the death receptor Fas, which regulates apoptosis in response to repeated TCR signaling. Bid lies at the nexus of these two signaling pathways, thus we reasoned that Bid and Bim might cooperate in regulation of T cell shutdown in persistent infection. In this study, we observed that the combined loss of Bid and Bim synergistically enhanced the persistence of CD8(+) T cells during γ-herpesvirus infection. Thus, these data uncover a role for Bid in coordinating apoptotic signaling pathways to ensure appropriate shutdown of T cell immune responses in the setting of persistent Ag exposure.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis/inmunología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/fisiología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Cooperación Linfocítica/inmunología , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas/fisiología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Animales , Proteína 11 Similar a Bcl2 , Comunicación Celular/inmunología , Muerte Celular/inmunología , Infecciones por Herpesviridae/metabolismo , Subtipo H3N2 del Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Rhadinovirus/inmunología , Subgrupos de Linfocitos T/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/patología , Carga Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...