Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 21(9): 1058-1069, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32719520

RESUMEN

Innate T cells, including invariant natural killer T (iNKT) and mucosal-associated innate T (MAIT) cells, are a heterogeneous T lymphocyte population with effector properties preprogrammed during their thymic differentiation. How this program is initiated is currently unclear. Here, we show that the transcription factor BCL-6 was transiently expressed in iNKT cells upon exit from positive selection and was required for their proper development beyond stage 0. Notably, development of MAIT cells was also impaired in the absence of Bcl6. BCL-6-deficient iNKT cells had reduced expression of genes that were associated with the innate T cell lineage, including Zbtb16, which encodes PLZF, and PLZF-targeted genes. BCL-6 contributed to a chromatin accessibility landscape that was permissive for the expression of development-related genes and inhibitory for genes associated with naive T cell programs. Our results revealed new functions for BCL-6 and illuminated how this transcription factor controls early iNKT cell development.


Asunto(s)
Cromatina/metabolismo , Células T Invariantes Asociadas a Mucosa/inmunología , Células T Asesinas Naturales/inmunología , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Selección Clonal Mediada por Antígenos , Regulación del Desarrollo de la Expresión Génica , Inmunidad Innata , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Proteínas Proto-Oncogénicas c-bcl-6/genética
2.
Nat Immunol ; 17(10): 1187-96, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27487330

RESUMEN

During unresolved infections, some viruses escape immunological control and establish a persistant reservoir in certain cell types, such as human immunodeficiency virus (HIV), which persists in follicular helper T cells (TFH cells), and Epstein-Barr virus (EBV), which persists in B cells. Here we identified a specialized group of cytotoxic T cells (TC cells) that expressed the chemokine receptor CXCR5, selectively entered B cell follicles and eradicated infected TFH cells and B cells. The differentiation of these cells, which we have called 'follicular cytotoxic T cells' (TFC cells), required the transcription factors Bcl6, E2A and TCF-1 but was inhibited by the transcriptional regulators Blimp1, Id2 and Id3. Blimp1 and E2A directly regulated Cxcr5 expression and, together with Bcl6 and TCF-1, formed a transcriptional circuit that guided TFC cell development. The identification of TFC cells has far-reaching implications for the development of strategies to control infections that target B cells and TFH cells and to treat B cell-derived malignancies.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Linfocitos B/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , VIH/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Células Cultivadas , Regulación de la Expresión Génica , Centro Germinal/patología , Centro Germinal/virología , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Proc Natl Acad Sci U S A ; 121(5): e2304020121, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38261619

RESUMEN

Follicular regulatory T cells (Tfr) can play opposite roles in the regulation of germinal center (GC) responses. Depending on the studies, Tfr suppress or support GC and B cell affinity maturation. However, which factors determine positive vs. negative effects of Tfr on the GC B cell is unclear. In this study, we show that GC centrocytes that express MYC up-regulate expression of CCL3 chemokine that is needed for both the positive and negative regulation of GC B cells by Tfr. B cell-intrinsic expression of CCL3 contributes to Tfr-dependent positive selection of foreign Ag-specific GC B cells. At the same time, expression of CCL3 is critical for direct Tfr-mediated suppression of GC B cells that acquire cognate to Tfr nuclear proteins. Our study suggests that CCR5 and CCR1 receptors promote Tfr migration to CCL3 and highlights Ccr5 expression on the Tfr subset that expresses Il10. Based on our findings and previous studies, we suggest a model of chemotactically targeted checkpoint control of B cells undergoing positive selection in GCs by Tfr, where Tfr directly probe and license foreign antigen-specific B cells to complete their positive selection in GCs but, at the same time, suppress GC B cells that present self-antigens cognate to Tfr.


Asunto(s)
Linfocitos B , Linfocitos T Reguladores , Centro Germinal , Autoantígenos , Quimiocina CCL3
4.
PLoS Pathog ; 19(9): e1011672, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37721965

RESUMEN

Brucellosis, caused by facultative, intracellular Brucella spp., often results in chronic and/or lifelong infection. Therefore, Brucella must employ mechanisms to subvert adaptive immunity to cause chronic infection. B lymphocytes enhance susceptibility to infection with Brucella spp. though the mechanisms remain unclear. Here we investigated the role of antibody secretion, B cell receptor (BCR) specificity, and B cell antigen presentation on susceptibility to B. melitensis. We report that mice unable to secrete antibody do not display altered resistance to Brucella. However, animals with B cells that are unable to recognize Brucella through their BCR are resistant to infection. In addition, B cell MHCII expression enhances susceptibility to infection in a CD4+ T cell-dependent manner, and we found that follicular B cells are sufficient to inhibit CD4+ T cell-mediated immunity against Brucella. B cells promote development of T follicular helper (TFH) and T follicular regulatory (TFR) cells during Brucella infection. Inhibition of B cell and CD4+ T cell interaction via CD40L blockade enhances resistance to Brucella in a B cell dependent manner concomitant with suppression of TFH and TFR differentiation. Conversely, PD-1 blockade increases Brucella burdens in a B and CD4+ T cell dependent manner while augmenting T regulatory (TReg) and TFR responses. Intriguingly, TFR deficiency enhances resistance to Brucella via a B cell dependent, but antibody independent mechanism. Collectively, these results demonstrate B cells support TFR responses that promote susceptibility to Brucella infection independent of the antibody response.

5.
J Immunol ; 211(1): 15-22, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37339403

RESUMEN

Development of high-affinity Abs in the germinal center (GC) is dependent on a specialized subset of T cells called "T follicular helper" (TFH) cells that help select Ag-specific B cells. A second T cell subset, T follicular regulatory (TFR) cells, can act as repressors of the GC and Ab response but can also provide a helper function for GC B cells in some contexts. Recent studies showed that, apart from their traditional helper role, TFH cells can also act as repressors of the Ab response, particularly for IgE responses. We review how both TFH and TFR cells express helper and repressor factors that coordinately regulate the Ab response and how the line between these two subsets is less clear than initially thought. Thus, TFH and TFR cells are interconnected and have "nonbinary" functions. However, many questions remain about how these critical cells control the Ab response.


Asunto(s)
Células T Auxiliares Foliculares , Linfocitos T Colaboradores-Inductores , Linfocitos T Colaboradores-Inductores/metabolismo , Centro Germinal , Linfocitos B , Linfocitos T Reguladores
6.
J Immunol ; 210(7): 905-915, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36779803

RESUMEN

High-affinity allergen-specific IgE is essential for the severe allergic anaphylaxis response. High-affinity Abs are formed by successive rounds of selection of Ag-specific B cells in the germinal center (GC); however, several studies have shown that IgE+ GC B cells are impaired in their ability to undergo selection in the GC. A pathway, known as the "indirect switching pathway" for IgE, has been described whereby Ag-specific B cells initially switch to the IgG1 isotype and undergo affinity selection in the GC, with a secondary switch to the IgE isotype after affinity selection. In previous work, using a food allergy model in mice, we investigated how high-affinity IgE develops in the GC, but we did not test the indirect switching model. In this study, we analyzed the importance of the indirect switching pathway by constructing IgG1-cre Bcl6-fl/fl mice. In these mice, once B cells switch to IgG1, they delete Bcl6 and thus cannot enter or persist in the GC. When we tested IgG1-cre Bcl6-fl/fl mice with our food allergy model, we found that, as expected, IgG1 Abs had decreased affinity, but unexpectedly, the affinity of IgE for allergen was unchanged. IgG1-cre Bcl6-fl/fl mice underwent anaphylaxis in response to allergen, consistent with the formation of high-affinity IgE. Thus, in a food allergy response, high-affinity IgE can be efficiently formed in the absence of indirect switching to IgG1, either by direct selection of IgE+ GC B cells or indirect selection of IgM+ GC B cells that later switch to IgE.


Asunto(s)
Hipersensibilidad a los Alimentos , Centro Germinal , Inmunoglobulina E , Animales , Ratones , Inmunoglobulina E/biosíntesis , Inmunoglobulina E/inmunología , Inmunoglobulina G , Centro Germinal/inmunología , Hipersensibilidad a los Alimentos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Cambio de Clase de Inmunoglobulina
7.
Immunity ; 43(3): 541-53, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26320660

RESUMEN

Immunoglobulin A (IgA) is prominently secreted at mucosal surfaces and coats a fraction of the intestinal microbiota. However, the commensal bacteria bound by IgA are poorly characterized and the type of humoral immunity they elicit remains elusive. We used bacterial flow cytometry coupled with 16S rRNA gene sequencing (IgA-Seq) in murine models of immunodeficiency to identify IgA-bound bacteria and elucidate mechanisms of commensal IgA targeting. We found that residence in the small intestine, rather than bacterial identity, dictated induction of specific IgA. Most commensals elicited strong T-independent (TI) responses that originated from the orphan B1b lineage and from B2 cells, but excluded natural antibacterial B1a specificities. Atypical commensals including segmented filamentous bacteria and Mucispirillum evaded TI responses but elicited T-dependent IgA. These data demonstrate exquisite targeting of distinct commensal bacteria by multiple layers of humoral immunity and reveal a specialized function of the B1b lineage in TI mucosal IgA responses.


Asunto(s)
Inmunidad Adaptativa/inmunología , Bacterias/inmunología , Inmunidad Humoral/inmunología , Inmunidad Innata/inmunología , Inmunoglobulina A/inmunología , Intestino Delgado/inmunología , Inmunidad Adaptativa/genética , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Bacterias/clasificación , Bacterias/genética , Colon/inmunología , Colon/metabolismo , Colon/microbiología , Citometría de Flujo , Variación Genética/inmunología , Humanos , Inmunidad Humoral/genética , Inmunidad Innata/genética , Inmunoglobulina A/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Ribosómico 16S/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo
8.
Mol Ther ; 30(3): 1149-1162, 2022 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-34793974

RESUMEN

STAT3 signaling has been shown to regulate cellular function and cytokine production in the tumor microenvironment (TME). Within the head and neck squamous cell carcinoma (HNSCC) TME, we previously showed that therapeutic targeting of STAT3 in combination with radiation resulted in improved tumor growth delay. However, given the independent regulatory effects STAT3 has on anti-tumor immunity, we aimed to decipher the effects of individually targeting STAT3 in the cancer cell, regulatory T cells (Tregs), and natural killer (NK) cell compartments in driving tumor growth and resistance to therapy in HNSCCs. We utilized a CRISPR knockout system for genetic deletion of STAT3 within the cancer cell as well as two genetic knockout mouse models, FoxP3-Cre/STAT3 fl and NKp46-Cre/STAT3 fl, for Tregs and NK cell targeting, respectively. Our data revealed differences in development of resistance to treatment with STAT3 CRISPR knockout in the cancer cell, driven by differential recruitment of immune cells. Knockout of STAT3 in Tregs overcomes this resistance and results in Treg reprogramming and recruitment and activation of antigen-presenting cells. In contrast, knockout of STAT3 in the NK cell compartment results in NK cell inactivation and acceleration of tumor growth. These data underscore the complex interplay between the cancer cell and the immune TME and carry significant implications for drug targeting and design of combination approaches in HNSCCs.


Asunto(s)
Neoplasias de Cabeza y Cuello , Factor de Transcripción STAT3/metabolismo , Animales , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/terapia , Ratones , Ratones Noqueados , Factor de Transcripción STAT3/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia , Linfocitos T Reguladores , Microambiente Tumoral/genética
10.
Hepatology ; 74(3): 1234-1250, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33710653

RESUMEN

BACKGROUND AND AIMS: Chronic alcohol drinking is a major risk factor for alcohol-associated liver disease (ALD). FK506-binding protein 51 (FKBP5), a cochaperone protein, is involved in many key regulatory pathways. It is known to be involved in stress-related disorders, but there are no reports regarding its role in ALD. This present study aimed to examine the molecular mechanism of FKBP5 in ALD. APPROACH AND RESULTS: We found a significant increase in hepatic FKBP5 transcripts and protein expression in patients with ALD and mice fed with chronic-plus-single binge ethanol. Loss of Fkbp5 in mice protected against alcohol-induced hepatic steatosis and inflammation. Transcriptomic analysis revealed a significant reduction of Transcriptional enhancer factor TEF-1 (TEA) domain transcription factor 1 (Tead1) and chemokine (C-X-C motif) ligand 1 (Cxcl1) mRNA in ethanol-fed Fkbp5-/- mice. Ethanol-induced Fkbp5 expression was secondary to down-regulation of methylation level at its 5' untranslated promoter region. The increase in Fkbp5 expression led to induction in transcription factor TEAD1 through Hippo signaling pathway. Fkbp5 can interact with yes-associated protein (YAP) upstream kinase, mammalian Ste20-like kinase 1 (MST1), affecting its ability to phosphorylate YAP and the inhibitory effect of hepatic YAP phosphorylation by ethanol leading to YAP nuclear translocation and TEAD1 activation. Activation of TEAD1 led to increased expression of its target, CXCL1, a chemokine-mediated neutrophil recruitment, causing hepatic inflammation and neutrophil infiltration in our mouse model. CONCLUSIONS: We identified an FKBP5-YAP-TEAD1-CXCL1 axis in the pathogenesis of ALD. Loss of FKBP5 ameliorates alcohol-induced liver injury through the Hippo pathway and CXCL1 signaling, suggesting its potential role as a target for the treatment of ALD.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Quimiocina CXCL1/metabolismo , Etanol/farmacología , Vía de Señalización Hippo/genética , Hepatopatías Alcohólicas/genética , Proteínas de Unión a Tacrolimus/genética , Animales , Metilación de ADN , Perfilación de la Expresión Génica , Humanos , Inflamación , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/patología , Ratones , Ratones Noqueados , Infiltración Neutrófila/genética , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , Factores de Transcripción de Dominio TEA , Proteínas de Unión a Tacrolimus/metabolismo , Proteínas Señalizadoras YAP/metabolismo
11.
Proc Natl Acad Sci U S A ; 116(24): 11888-11893, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31138703

RESUMEN

Neutrophils are vital for antimicrobial defense; however, their role during viral infection is less clear. Furthermore, the molecular regulation of neutrophil fate and function at the viral infected sites is largely elusive. Here we report that BCL6 deficiency in myeloid cells exhibited drastically enhanced host resistance to severe influenza A virus (IAV) infection. In contrast to the notion that BCL6 functions to suppress innate inflammation, we find that myeloid BCL6 deficiency diminished lung inflammation without affecting viral loads. Using a series of Cre-transgenic, reporter, and knockout mouse lines, we demonstrate that BCL6 deficiency in neutrophils, but not in monocytes or lung macrophages, attenuated host inflammation and morbidity following IAV infection. Mechanistically, BCL6 bound to the neutrophil gene loci involved in cellular apoptosis in cells specifically at the site of infection. As such, BCL6 disruption resulted in increased expression of apoptotic genes in neutrophils in the respiratory tract, but not in the circulation or bone marrow. Consequently, BCL6 deficiency promoted tissue neutrophil apoptosis. Partial neutrophil depletion led to diminished pulmonary inflammation and decreased host morbidity. Our results reveal a previously unappreciated role of BCL6 in modulating neutrophil apoptosis at the site of infection for the regulation of host disease development following viral infection. Furthermore, our studies indicate that tissue-specific regulation of neutrophil survival modulates host inflammation and tissue immunopathology during acute respiratory viral infection.


Asunto(s)
Virus de la Influenza A/patogenicidad , Neutrófilos/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Neumonía/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Animales , Apoptosis/fisiología , Interacciones Huésped-Patógeno/fisiología , Pulmón/metabolismo , Pulmón/virología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/fisiología , Neutrófilos/virología , Neumonía/virología , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/virología
12.
Eur J Immunol ; 50(7): 1067-1077, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32133634

RESUMEN

Stearoyl-CoA desaturases (SCD) are endoplasmic reticulum (ER)-associated enzymes that catalyze the synthesis of the monounsaturated fatty acids (MUFAs). As such, SCD play important roles in maintaining the intracellular balance between saturated fatty acid (SFAs) and MUFAs. The roles of SCD in CD4+ T-helper cell responses are currently unexplored. Here, we have found that murine and human follicular helper T (TFH ) cells express higher levels of SCD compared to non-TFH cells. Further, the expression of SCD in TFH cells is dependent on the TFH lineage-specification transcription factor BCL6. We found that the inhibition of SCD impaired TFH cell maintenance and shifted the balance between TFH and follicular regulatory T (TFR ) cells in the spleen. Consequently, SCD inhibition dampened germinal center B-cell responses following influenza immunization. Mechanistically, we found that SCD inhibition led to increased ER stress and enhanced TFH cell apoptosis in vitro and in vivo. These results reveal a possible link between fatty acid metabolism and cellular and humoral responses induced by immunization or potentially, autoimmunity.


Asunto(s)
Apoptosis/inmunología , Linfocitos B/inmunología , Centro Germinal/inmunología , Bazo/inmunología , Estearoil-CoA Desaturasa/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos B/citología , Centro Germinal/citología , Humanos , Ratones , Ratones Noqueados , Bazo/citología , Estearoil-CoA Desaturasa/genética , Linfocitos T Reguladores/citología
13.
J Allergy Clin Immunol ; 146(5): 1121-1136.e9, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32179158

RESUMEN

BACKGROUND: Bcl6 is required for the development of T follicular helper cells and T follicular regulatory (Tfr) cells that regulate germinal center responses. Bcl6 also affects the function of regulatory T (Treg) cells. OBJECTIVE: The goal of this study was to define the functions of Bcl6 in Treg cells, including Tfr cells, in the context of allergic airway inflammation. METHODS: We used a model of house dust mite sensitization to challenge wild-type, Bcl6fl/fl Foxp3-Cre, and Prdm1 (Blimp1)fl/fl Foxp3-Cre mice to study the reciprocal roles of Bcl6 and Blimp1 in allergic airway inflammation. RESULTS: In the house dust mite model, Tfr cells repress the production of IgE and Bcl6+ Treg cells suppress the generation of type 2 cytokine-producing cells in the lungs. In mice with Bcl6-deficient Treg cells, twice as many ST2+ (IL-33R+) Treg cells develop as are observed in wild-type mice. ST2+ Treg cells in the context of allergic airway inflammation are Blimp1 dependent, express type 2 cytokines, and share features of visceral adipose tissue Treg cells. Bcl6-deficient Treg cells are more susceptible, and Blimp1-deficient Treg cells are resistant, to acquiring the ST2+ Treg-cell phenotype in vitro and in vivo in response to IL-33. Bcl6-deficient ST2+ Treg cells, but not Bcl6-deficient ST2+ conventional T cells, strongly promote allergic airway inflammation when transferred into recipient mice. Lastly, ST2 is required for the exacerbated allergic airway inflammation in Bcl6fl/fl Foxp3-Cre mice. CONCLUSIONS: During allergic airway inflammation, Bcl6 and Blimp1 play dual roles in regulating Tfr-cell activity in the germinal center and in the development of ST2+ Treg cells that promote type 2 cytokine responses.


Asunto(s)
Centro Germinal/inmunología , Hipersensibilidad/inmunología , Neumonía/inmunología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Antígenos Dermatofagoides/inmunología , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Proteínas Proto-Oncogénicas c-bcl-6/genética , Pyroglyphidae
14.
Blood ; 132(10): 1027-1038, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30042096

RESUMEN

We hypothesized that megakaryocyte (MK) phosphoinositide signaling mediated by phosphatidylinositol transfer proteins (PITPs) contributes to hematopoietic stem cell (HSC) and hematopoietic progenitor cell (HPC) regulation. Conditional knockout mice lacking PITPs specifically in MKs and platelets (pitpα-/- and pitpα-/-/ß-/-) bone marrow (BM) manifested decreased numbers of HSCs, MK-erythrocyte progenitors, and cycling HPCs. Further, pitpα-/-/ß-/- BM had significantly reduced engrafting capability in competitive transplantation and limiting dilution analysis. Conditioned media (CM) from cultured pitpα-/- and pitpα-/-/ß-/- BM MKs contained higher levels of transforming growth factor ß1 (TGF-ß1) and interleukin-4 (IL-4), among other myelosuppressive cytokines, than wild-type BM MKs. Correspondingly, BM flush fluid from pitpα-/- and pitpα-/-/ß-/- mice had higher concentrations of TGF-ß1. CM from pitpα-/- and pitpα-/-/ß-/- MKs significantly suppressed HPC colony formation, which was completely extinguished in vitro by neutralizing anti-TGF-ß antibody, and treatment of pitpα-/-/ß-/- mice in vivo with anti-TGF-ß antibodies completely reverted their defects in BM HSC and HPC numbers. TGF-ß and IL-4 synergized to inhibit HPC colony formation in vitro. Electron microscopy analysis of pitpα-/-/ß-/- MKs revealed ultrastructural defects with depleted α-granules and large, misshaped multivesicular bodies. Von Willebrand factor and thrombospondin-1, like TGF-ß, are stored in MK α-granules and were also elevated in CM of cultured pitpα-/-/ß-/- MKs. Altogether, these data show that ablating PITPs in MKs indirectly dysregulates hematopoiesis in the BM by disrupting α-granule physiology and secretion of TGF-ß1.


Asunto(s)
Médula Ósea/metabolismo , Hematopoyesis/fisiología , Megacariocitos/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Animales , Interleucina-4/genética , Interleucina-4/metabolismo , Megacariocitos/citología , Ratones , Ratones Noqueados , Proteínas de Transferencia de Fosfolípidos/genética , Trombospondina 1/genética , Trombospondina 1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
17.
J Immunol ; 198(11): 4352-4359, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28468971

RESUMEN

IL-2 is a pleiotropic cytokine that promotes the differentiation of Th cell subsets, including Th1, Th2, and Th9 cells, but it impairs the development of Th17 and T follicular helper cells. Although IL-2 is produced by all polarized Th subsets to some level, how it impacts cytokine production when effector T cells are restimulated is unknown. We show in this article that Golgi transport inhibitors (GTIs) blocked IL-9 production. Mechanistically, GTIs blocked secretion of IL-2 that normally feeds back in a paracrine manner to promote STAT5 activation and IL-9 production. IL-2 feedback had no effect on Th1- or Th17-signature cytokine production, but it promoted Th2- and Th9-associated cytokine expression. These data suggest that the use of GTIs results in an underestimation of the presence of type 2 cytokine-secreting cells and highlight IL-2 as a critical component in optimal cytokine production by Th2 and Th9 cells in vitro and in vivo.


Asunto(s)
Citocinas/biosíntesis , Interleucina-2/metabolismo , Interleucina-9/biosíntesis , Comunicación Paracrina , Células Th2/inmunología , Animales , Brefeldino A/farmacología , Diferenciación Celular , Citocinas/inmunología , Interleucina-9/antagonistas & inhibidores , Interleucina-9/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Monensina/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Ionóforos de Protónes/farmacología , Factor de Transcripción STAT5/metabolismo , Células TH1/inmunología , Células Th17/inmunología
18.
J Allergy Clin Immunol ; 142(4): 1144-1158.e8, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29247716

RESUMEN

BACKGROUND: Little is currently known regarding the immunologic mechanism(s) that initiate peanut allergy. Notably, peanut proteins have been detected in house dust, and their levels correlate with peanut allergy prevalence. OBJECTIVE: This study aimed to develop a new mouse model for peanut allergy and to investigate the immunologic mechanisms involved in peanut allergen sensitization. METHODS: To mimic environmental exposure, naive mice were exposed to peanut flour by inhalation for up to 4 weeks. We then analyzed serum levels of IgE antibody and challenged mice with peanut proteins. Immunological mechanisms involved in sensitization were analyzed using cytokine reporter mice, an adoptive cell transfer model, and gene knockout mice. RESULTS: When exposed to peanut flour by inhalation, both BALB/c and C57BL/6 mice developed peanut allergy, as demonstrated by the presence of peanut-specific IgE antibodies and manifestation of acute anaphylaxis on challenge. A large number of follicular helper T (Tfh) cells were also detected in draining lymph nodes of allergic mice. These cells produced IL-4 and IL-21, and they more robustly promoted peanut-specific IgE production than Th2 cells did. Genetic depletion of Tfh cells decreased IgE antibody levels and protected mice from anaphylaxis, without affecting Th2 cells. Furthermore, peanut flour exposure increased lung levels of IL-1α and IL-1ß, and mice deficient in the receptor for these cytokines showed a significant decrease in Tfh cells compared with in wild-type mice. CONCLUSIONS: Tfh cells play a key role in peanut allergy, and the IL-1 pathway is involved in the Tfh response to peanut allergen exposure.


Asunto(s)
Citocinas/inmunología , Modelos Animales de Enfermedad , Hipersensibilidad al Cacahuete/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Administración por Inhalación , Alérgenos/inmunología , Animales , Arachis/inmunología , Femenino , Inmunoglobulina E/inmunología , Pulmón/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
19.
Eur J Immunol ; 47(7): 1136-1141, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28586108

RESUMEN

The transcription factors Bcl6 and Blimp1 have opposing roles in the development of the follicular helper T (TFH ) cells: Bcl6 promotes and Blimp1 inhibits TFH -cell differentiation. Similarly, Bcl6 activates, while Blimp1 represses, expression of the TFH -cell marker PD-1. However, Bcl6 and Blimp1 repress each other's expression, complicating the interpretation of the regulatory network. Here we sought to clarify the extent to which Bcl6 and Blimp1 independently control TFH -cell differentiation by generating mice with T-cell specific deletion of both Bcl6 and Blimp1 (double conditional KO [dcKO] mice). Our data indicate that Blimp1 does not control TFH -cell differentiation independently of Bcl6. However, a population of T follicular regulatory (TFR ) cells developed independently of Bcl6 in dcKO mice. We have also analyzed regulation of IL-10 and PD-1, two genes controlled by both Bcl6 and Blimp1, and observed that Bcl6 regulates both genes independently of Blimp1. We found that Bcl6 positively regulates PD-1 expression by inhibiting the ability of T-bet/Tbx21 to repress Pdcd1 transcription. Our data provide a novel mechanism for positive control of gene expression by Bcl6, and illuminate how Bcl6 and Blimp1 control TFH -cell differentiation.


Asunto(s)
Diferenciación Celular , Receptor de Muerte Celular Programada 1/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Transcripción/metabolismo , Animales , Expresión Génica , Regulación de la Expresión Génica , Centro Germinal/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Activación de Linfocitos , Ratones , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Receptor de Muerte Celular Programada 1/inmunología , Proteínas Proto-Oncogénicas c-bcl-6/deficiencia , Proteínas Proto-Oncogénicas c-bcl-6/genética , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología , Proteínas de Dominio T Box/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
20.
J Immunol ; 196(8): 3297-304, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26976954

RESUMEN

Th cell subsets develop in response to multiple activating signals, including the cytokine environment. IL-9-secreting T cells develop in response to the combination of IL-4 and TGF-ß, although they clearly require other cytokine signals, leading to the activation of transcription factors including STAT5. In Th17 cells, there is a molecular antagonism of STAT5 with STAT3 signaling, although whether this paradigm exists in other Th subsets is not clear. In this paper, we demonstrate that STAT3 attenuates the ability of STAT5 to promote the development of IL-9-secreting T cells. We demonstrate that production of IL-9 is increased in the absence of STAT3 and cytokines that result in a sustained activation of STAT3, including IL-6, have the greatest potency in repressing IL-9 production in a STAT3-dependent manner. Increased IL-9 production in the absence of STAT3 correlates with increased endogenous IL-2 production and STAT5 activation, and blocking IL-2 responses eliminates the difference in IL-9 production between wild-type and STAT3-deficient T cells. Moreover, transduction of developing Th9 cells with a constitutively active STAT5 eliminates the ability of IL-6 to reduce IL-9 production. Thus, STAT3 functions as a negative regulator of IL-9 production through attenuation of STAT5 activation and function.


Asunto(s)
Interleucina-6/metabolismo , Interleucina-9/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/antagonistas & inhibidores , Células Th17/inmunología , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Interleucina-2/biosíntesis , Interleucina-2/inmunología , Interleucina-2/metabolismo , Interleucina-4/inmunología , Interleucina-4/metabolismo , Interleucina-6/inmunología , Interleucina-9/biosíntesis , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Células Th17/citología , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA