Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
PLoS Biol ; 17(5): e3000262, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31071093

RESUMEN

Intestinal intraepithelial lymphocytes (IELs) expressing CD8αα on αß T cells (TCRαß+CD8αα+ IELs) have suppressive capabilities in enterocolitis, but the mechanism that maintains homeostasis and cell number is not fully understood. Here, we demonstrated that the number of TCRαß+CD8αα+ IELs was severely reduced in mice lacking recombination signal binding protein for immunoglobulin kappa J region (Rbpj) or Notch1 and Notch2 in T cells. Rbpj-deficient TCRαß+CD8αα+ IELs expressed low levels of Atp8a2, which encodes a protein with flippase activity that regulates phospholipid asymmetry of plasma membrane such as flipping phosphatidylserine in the inner leaflet of plasma membrane. Rbpj-deficient TCRαß+CD8αα+ IELs cannot maintain phosphatidylserine in the inner leaflet of the plasma membrane. Furthermore, depletion of intestinal macrophages restored TCRαß+CD8αα+ IELs in Rbpj-deficient mice, suggesting that exposure of phosphatidylserine on the plasma membrane in Rbpj-deficient TCRαß+CD8αα+ IELs acts as an "eat-me" signal. Together, these results revealed that Notch-Atp8a2 is a fundamental regulator for IELs and highlighted that membrane phospholipid asymmetry controlled by Notch-mediated flippase expression is a critical determinant in setting or balancing the number of TCRαß+CD8αα+ IELs.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Linfocitos Intraepiteliales/metabolismo , Fosfolípidos/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores Notch/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteínas de Transferencia de Fosfolípidos/metabolismo
2.
Nat Immunol ; 9(10): 1140-7, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18724371

RESUMEN

The acquisition of cytotoxic effector function by CD8(+) T cells is crucial for the control of intracellular infection and tumor invasion. However, it remains unclear which signaling pathways are required for the differentiation of CD8(+) cytotoxic T lymphocytes. We show here that Notch2-deficient T cells had impaired differentiation into cytotoxic T lymphocytes. In addition, dendritic cells with lower expression of the Notch ligand Delta-like 1 induced the differentiation of cytotoxic T lymphocytes less efficiently. We found that the intracellular domain of Notch2 interacted with a phosphorylated form of the transcription factor CREB1, and together these proteins bound the transcriptional coactivator p300 to form a complex on the promoter of the gene encoding granzyme B. Our results suggest that the highly regulated, dynamic control of T cell cytotoxicity depends on the integration of Notch2 and CREB1 signals.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Diferenciación Celular/inmunología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Receptor Notch2/metabolismo , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/citología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/inmunología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/inmunología , Células Dendríticas/inmunología , Femenino , Regulación de la Expresión Génica/inmunología , Granzimas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Receptor Notch2/inmunología , Linfocitos T Citotóxicos/inmunología , Transcripción Genética/inmunología , Factores de Transcripción p300-CBP/inmunología , Factores de Transcripción p300-CBP/metabolismo
3.
J Immunol ; 199(6): 1989-1997, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28779023

RESUMEN

Dendritic cells (DCs) are important for adaptive immune responses through the activation of T cells. The molecular interplay between DCs and T cells determines the magnitude of T cell responses or outcomes of functional differentiation of T cells. In this study, we demonstrated that DCs in mice that are Rbpj deficient in CD11c+ cells (Rbpj-/- mice) promoted the differentiation of IL-17A-producing Th17 cells. Rbpj-deficient DCs expressed little Aldh1a2 protein that is required for generating retinoic acid. Those DCs exhibited a reduced ability for differentiating regulatory T cells induced by TGF-ß. Rbpj protein directly regulated Aldh1a2 transcription by binding to its promoter region. The overexpression of Aldh1a2 in Rbpj-deficient DCs negated their Th17-promoting ability. Transfer of naive CD4+ T cells into Rag1-deficient Rbpj-/- mice enhanced colitis with increased Th17 and reduced induced regulatory T cells (iTreg) compared with control Rag1-deficient mice. The cotransfer of iTreg and naive CD4+ T cells into Rag1-deficient Rbpj-/- mice improved colitis compared with transfer of naive CD4+ T cell alone. Furthermore, cotransfer of DCs from Rbpj-/- mice that overexpressed Aldh1a2 or Notch-stimulated DCs together with naive CD4+ T cells into Rbpj-/-Rag1-deficient mice led to reduced colitis with increased iTreg numbers. Therefore, our studies identify Notch signaling in DCs as a crucial balancer of Th17/iTreg, which depends on the direct regulation of Aldh1a2 transcription in DCs.


Asunto(s)
Colitis/inmunología , Células Dendríticas/inmunología , Retinal-Deshidrogenasa/metabolismo , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Familia de Aldehído Deshidrogenasa 1 , Animales , Antígeno CD11c/metabolismo , Diferenciación Celular , Células Cultivadas , Regulación de la Expresión Génica , Genes RAG-1 , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Interleucina-17/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Notch/metabolismo , Retinal-Deshidrogenasa/genética , Linfocitos T Reguladores/trasplante , Tretinoina/metabolismo
4.
Proc Natl Acad Sci U S A ; 111(16): 5986-91, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24711412

RESUMEN

The gastrointestinal tract comes into direct contact with environmental agents, including bacteria, viruses, and foods. Intestine-specific subsets of immune cells maintain gut homeostasis by continuously sampling luminal antigens and maintaining immune tolerance. CD11c(+)CX3CR1(+) cells sample luminal antigens in the small intestine and contribute to the trafficking of bacteria to lymph nodes under dysbiotic conditions. The molecular mechanisms crucial for the differentiation of CD11c(+)CX3CR1(+) cells remain unclear. Here we demonstrate that the Notch1- or Notch2-Rbpj axis is essential for the development of CD11c(+)CX3CR1(+) cells. In mice in which Rbpj or Notch1 and Notch2 were deleted from CD11c(+) cells, there was a deficit of CD11c(+)CX3CR1(+) cells and an accumulation of CD11c(low)CX3CR1(+) cells. The CD11c(low)CX3CR1(+) cells could not differentiate to CD11c(+)CX3CR1(+) cells, suggesting that CD11c(low)CX3CR1(+) cells represent a lineage distinct from CD11c(+)CX3CR1(+) cells. These data indicate that Notch signaling is essential for lineage fixation of intestinal CD11c(+)CX3CR1(+) cells.


Asunto(s)
Antígeno CD11c/metabolismo , Diferenciación Celular , Intestino Delgado/citología , Receptores de Quimiocina/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Receptor 1 de Quimiocinas CX3C , Recuento de Células , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Ratones , Ratones Endogámicos C57BL
5.
Biochem Biophys Res Commun ; 444(4): 628-33, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24491544

RESUMEN

CD4(+) T cell activation is controlled by signaling through the T cell receptor in addition to various co-receptors, and is also affected by their interactions with effector and regulatory T cells in the microenvironment. Inflammatory bowel diseases (IBD) are caused by the persistent activation and expansion of auto-aggressive CD4(+) T cells that attack intestinal epithelial cells. However, the molecular basis for the persistent activation of CD4(+) T cells in IBD remains unclear. In this study, we investigated how the CD98 heavy chain (CD98hc, Slc3a2) affected the development of colitis in an experimental animal model. Transferring CD98hc-deficient CD4(+)CD25(-) T cells into Rag2(-/-) mice did not cause colitis accompanied by increasing Foxp3(+) inducible regulatory T cells. By comparison, CD98hc-deficient naturally occurring regulatory T cells (nTregs) had a decreased capability to suppress colitis induced by CD4(+)CD25(-) T cells, although CD98hc-deficient mice did not have a defect in the development of nTregs. Blocking CD98hc with an anti-CD98 blocking antibody prevented the development of colitis. Our results indicate that CD98hc regulates the expansion of autoimmune CD4(+) T cells in addition to controlling nTregs functions, which suggests the CD98hc as an important target molecule for establishing strategies for treating colitis.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Colitis/inmunología , Colitis/patología , Colon/patología , Cadena Pesada de la Proteína-1 Reguladora de Fusión/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Colitis/genética , Colon/inmunología , Colon/metabolismo , Proteínas de Unión al ADN/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Eliminación de Gen , Subunidad alfa del Receptor de Interleucina-2/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
6.
Front Immunol ; 14: 1065790, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36776885

RESUMEN

FXR is a key molecule that modulates anti-inflammatory activity in the intestinal-liver axis. Although FXR has pleiotropic functions including regulation of liver inflammation and activation of macrophages, it remains unclear whether it is involved in macrophage polarization. In this paper we demonstrated that stimulation of macrophages derived from the bone marrow using an FXR agonist activated polarization toward M2 but not M1 macrophages. The treatment of mice with chitin skewed macrophage polarization towards M2 macrophages, while co-treatment with an FXR agonist further promoted the polarization toward M2 macrophages in vivo. This skewed polarization towards M2 macrophages by an FXR agonist was accompanied by increased expression of signaling molecules related to the retinoic acid receptor. Inhibition of the retinoic acid receptor suppressed FXR agonist-mediated M2 macrophage polarization, indicating that this polarization was, at least, partly dependent on the retinoic acid receptor pathway. These data demonstrate that FXR has a role in polarization toward M2 macrophages and suggest a possible therapeutic potential of FXR agonists in M2 macrophage-related conditions.


Asunto(s)
Macrófagos , Receptores Citoplasmáticos y Nucleares , Transducción de Señal , Animales , Ratones , Antiinflamatorios/metabolismo , Macrófagos/metabolismo , Receptores de Ácido Retinoico/metabolismo , Receptores Citoplasmáticos y Nucleares/agonistas
7.
Eur J Immunol ; 41(5): 1309-20, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21469122

RESUMEN

Dendritic cells (DCs) are specialized antigen-presenting cells (APCs) required for T-cell activation and are classified into several subtypes by phenotypic and functional characteristics. However, it remains unclear if distinct transcription factors control the development of each DC subpopulation. In this report, we demonstrate that Notch signaling controls the development of a novel DC subtype that expresses Thy1 (Thy1(+) DCs). Overstimulation of bone marrow cells with the Notch ligand Delta-like 1 promoted the development of Thy1(+) DCs. Thy1(+) DCs are characterized as CD11c(+) MHC class II(+) NK1.1(-) B220(-) CD8α(+) , and are present in the thymus but not in the spleen and lymph nodes. Thymic Thy1(+) DCs are able to capture exogenous proteins and delete CD4(+) CD8(+) T cells. Transplantation experiments demonstrated that CD44(+) CD25(-) and CD44(+) CD25(+) thymocytes can differentiate into Thy1(+) DCs. Recombination signal binding protein for immunoglobulin kappa J region (RBP-J) deficiency in lineage-negative bone marrow cells, but not CD11c(+) cells, disrupted Thy1(+) DC development in the thymus. Our data indicate that Notch signaling controls the development of a novel type of Thy1-expressing DC in the thymus that possibly controls negative selection, and indicates that there may be highly regulated, differential transcriptional control of DC development. Furthermore, our findings suggest that Notch signaling regulates T-cell development not only by intrinsically inducing T-cell lineage-specific gene programs, but also by regulating negative selection through Thy1(+) DCs.


Asunto(s)
Células Dendríticas/inmunología , Receptores Notch/metabolismo , Antígenos Thy-1/genética , Timo/inmunología , Animales , Presentación de Antígeno , Western Blotting , Células de la Médula Ósea/metabolismo , Antígeno CD11c/análisis , Diferenciación Celular , Células Dendríticas/clasificación , Células Dendríticas/citología , Citometría de Flujo , Receptores de Hialuranos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Subunidad alfa del Receptor de Interleucina-2/análisis , Péptidos y Proteínas de Señalización Intracelular , Activación de Linfocitos , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Receptores Notch/genética , Transducción de Señal , Linfocitos T , Antígenos Thy-1/metabolismo , Timo/citología , Timo/embriología
8.
Biochem Biophys Res Commun ; 418(4): 701-7, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22293205

RESUMEN

B cells are classified into several subsets depending on their functions, marker expression pattern and localization. Marginal zone B (MZB) cells are a distinct lineage from follicular B cells, and regulate host defenses against blood-borne pathogens. Notch2/RBP-J signaling regulates the development of MZB cells by interacting with delta-like 1 ligand, although the target genes for Notch2 signaling remain unclear. We identified Fos as an upregulated gene in LPS-stimulated B cells that received Notch2 signaling. Fos is expressed in CD21(high)CD23(low) MZB cells at a higher level compared to CD21(Int)CD23(high) follicular B cells. Deleting the Notch2 gene in CD19(+) B cells decreased Fos expression in B cells. Overexpression of Fos in Notch2-deficient B cells or bone marrow cells partially restored MZB development. Fos promoter activity was upregulated by Notch2 signaling, indicating that Notch2 directly controls Fos transcription associated with MZB development. These data identify Fos as one of the target genes for Notch2 signaling that is crucial for MZB development.


Asunto(s)
Linfocitos B/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas Oncogénicas v-fos/genética , Receptor Notch2/metabolismo , Animales , Linfocitos B/metabolismo , Genes Reporteros , Luciferasas/genética , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Receptor Notch2/genética , Bazo/citología , Transcripción Genética
9.
Sci Adv ; 8(34): eabq0008, 2022 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-36001653

RESUMEN

Immunoglobulin class switch recombination (CSR) plays critical roles in controlling infections and inflammatory tissue injuries. Here, we show that AFF3, a candidate gene for both rheumatoid arthritis and type 1 diabetes, is a molecular facilitator of CSR with an isotype preference. Aff3-deficient mice exhibit low serum levels of immunoglobulins, predominantly immunoglobulin G2c (IgG2c) followed by IgG1 and IgG3 but not IgM. Furthermore, Aff3-deficient mice show weak resistance to Plasmodium yoelii infection, confirming that Aff3 modulates immunity to this pathogen. Mechanistically, the AFF3 protein binds to the IgM and IgG1 switch regions via a C-terminal domain, and Aff3 deficiency reduces the binding of AID to the switch regions less efficiently. One AFF3 risk allele for rheumatoid arthritis is associated with high mRNA expression of AFF3, IGHG2, and IGHA2 in human B cells. These findings demonstrate that AFF3 directly regulates CSR by facilitating the recruitment of AID to the switch regions.

10.
Proc Natl Acad Sci U S A ; 105(19): 7010-5, 2008 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-18458347

RESUMEN

Natural killer (NK) cells regulate various immune responses by exerting cytotoxic activity or secreting cytokines. The interaction of NK cells with dendritic cells (DC) contributes to NK cell-mediated antitumor or antimicrobial responses. However, the cellular and molecular mechanisms for controlling this interaction are largely unknown. Here, we show an involvement of Jagged2-Notch interaction in augmenting NK cell cytotoxicity mediated by DC. Enforced expression of Jagged2 on A20 cells (Jag2-A20 cells) suppressed their growth in vivo, which was abrogated by depleting NK cells. Moreover, Jag2-A20 cells exerted a suppression on the growth of nonmanipulated A20 cells in SCID mice in an NK-dependent manner. Consistently, coinoculation of A20 cells with DC overexpressing Jagged2 (Jag2-DC) suppressed the growth of A20 cells in mice. Stimulation of NK cells with Jagged2 directly enhanced their cytotoxicity, IFN-gamma production, and proliferation. Ligation of Notch2 on NK cells enhanced their cytotoxic activity, and Jag2-DC or CpG-treated DC-mediated NK cell cytotoxicity was suppressed by a gamma-secretase inhibitor. These results indicate that the Jagged2-Notch axis plays a crucial role in DC-mediated NK cell cytotoxicity. Furthermore, manipulation of this interaction may provide an approach to induce potent tumor immunity or to inhibit certain autoimmune diseases caused by NK cell activation.


Asunto(s)
Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Proteínas de la Membrana/inmunología , Receptores Notch/inmunología , Animales , Proliferación Celular , Trasplante de Células , Citotoxicidad Inmunológica , Inmunoterapia Adoptiva , Proteína Jagged-2 , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Neoplasias/inmunología , Neoplasias/patología , Unión Proteica , Transducción de Señal
11.
Sci Rep ; 7(1): 10696, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28878284

RESUMEN

MicroRNAs have broad roles in tumorigenesis and cell differentiation through regulation of target genes. Notch signaling also controls cell differentiation and tumorigenesis. However, the mechanisms through which Notch mediates microRNA expression are still unclear. In this study, we aimed to identify microRNAs regulated by Notch signaling. Our analysis found that microRNA-449a (miR-449a) was indirectly regulated by Notch signaling. Although miR-449a-deficient mice did not show any Notch-dependent defects in immune cell development, treatment of miR-449a-deficient mice with azoxymethane (AOM) or dextran sodium sulfate (DSS) increased the numbers and sizes of colon tumors. These effects were associated with an increase in intestinal epithelial cell proliferation following AOM/DSS treatment. In patients with colon cancer, miR-449a expression was inversely correlated with disease-free survival and histological scores and was positively correlated with the expression of MLH1 for which loss-of function mutations have been shown to be involved in colon cancer. Colon tissues of miR-449a-deficient mice showed reduced Mlh1 expression compared with those of wild-type mice. Thus, these data suggested that miR-449a acted as a key regulator of colon tumorigenesis by controlling the proliferation of intestinal epithelial cells. Additionally, activation of miR-449a may represent an effective therapeutic strategy and prognostic marker in colon cancer.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , MicroARNs/genética , Animales , Biomarcadores , Biomarcadores de Tumor , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias del Colon/mortalidad , Neoplasias del Colon/patología , Perfilación de la Expresión Génica , Inmunofenotipificación , Ratones , Ratones Noqueados , Ratones Transgénicos , Homólogo 1 de la Proteína MutL/genética , Homólogo 1 de la Proteína MutL/metabolismo , Pronóstico , Receptores Notch/metabolismo , Transducción de Señal , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
12.
Sci Rep ; 6: 33692, 2016 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-27659682

RESUMEN

Notch expressed on CD4+ T cells transduces signals that mediate their effector functions and survival. Although Notch signaling is known to be cis-inhibited by Notch ligands expressed on the same cells, the role of Notch ligands on T cells remains unclear. In this report we demonstrate that the CD4+ T cell Notch ligand Dll1 transduces signals required for their survival. Co-transfer of CD4+ T cells from Dll1-/- and control mice into recipient mice followed by immunization revealed a rapid decline of CD4+ T cells from Dll1-/- mice compared with control cells. Dll1-/- mice exhibited lower clinical scores of experimental autoimmune encephalitis than control mice. The expression of Notch target genes in CD4+ T cells from Dll1-/- mice was not affected, suggesting that Dll1 deficiency in T cells does not affect cis Notch signaling. Overexpression of the intracellular domain of Dll1 in Dll1-deficient CD4+ T cells partially rescued impaired survival. Our data demonstrate that Dll1 is an independent regulator of Notch-signaling important for the survival of activated CD4+ T cells, and provide new insight into the physiological roles of Notch ligands as well as a regulatory mechanism important for maintaining adaptive immune responses.

13.
Nat Commun ; 7: 12597, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27576369

RESUMEN

A population of monocytes, known as Ly6C(lo) monocytes, patrol blood vessels by crawling along the vascular endothelium. Here we show that endothelial cells control their origin through Notch signalling. Using combinations of conditional genetic deletion strategies and cell-fate tracking experiments we show that Notch2 regulates conversion of Ly6C(hi) monocytes into Ly6C(lo) monocytes in vivo and in vitro, thereby regulating monocyte cell fate under steady-state conditions. This process is controlled by Notch ligand delta-like 1 (Dll1) expressed by a population of endothelial cells that constitute distinct vascular niches in the bone marrow and spleen in vivo, while culture on recombinant DLL1 induces monocyte conversion in vitro. Thus, blood vessels regulate monocyte conversion, a form of committed myeloid cell fate regulation.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Monocitos/fisiología , Receptor Notch2/metabolismo , Transducción de Señal/fisiología , Traslado Adoptivo , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/metabolismo , Proteínas de Unión al Calcio , Diferenciación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Proteínas Ligadas a GPI/metabolismo , Voluntarios Sanos , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , Ratones Noqueados , Receptores de IgG/metabolismo , Proteínas Recombinantes/metabolismo , Bazo/citología
14.
Nat Med ; 21(1): 55-61, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25501905

RESUMEN

CD4+ T cells differentiate into memory T cells that protect the host from subsequent infection. In contrast, autoreactive memory CD4+ T cells harm the body by persisting in the tissues. The underlying pathways controlling the maintenance of memory CD4+ T cells remain undefined. We show here that memory CD4+ T cell survival is impaired in the absence of the Notch signaling protein known as recombination signal binding protein for immunoglobulin κ J region (Rbpj). Treatment of mice with a Notch inhibitor reduced memory CD4+ T cell numbers and prevented the recurrent induction of experimental autoimmune encephalomyelitis. Rbpj-deficient CD4+ memory T cells exhibit reduced glucose uptake due to impaired AKT phosphorylation, resulting in low Glut1 expression. Treating mice with pyruvic acid, which bypasses glucose uptake and supplies the metabolite downstream of glucose uptake, inhibited the decrease of autoimmune memory CD4+ T cells in the absence of Notch signaling, suggesting memory CD4+ T cell survival relies on glucose metabolism. Together, these data define a central role for Notch signaling in maintaining memory CD4+ T cells through the regulation of glucose uptake.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Glucosa/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Linfocitos T CD4-Positivos/metabolismo , Glucosa/inmunología , Transportador de Glucosa de Tipo 1/biosíntesis , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Ratones , Proteína Oncogénica v-akt/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Notch/antagonistas & inhibidores
15.
PLoS One ; 10(10): e0139692, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26444422

RESUMEN

Upon their recognition of antigens presented by the MHC, T cell proliferation is vital for clonal expansion and the acquisition of effector functions, which are essential for mounting adaptive immune responses. The CD98 heavy chain (CD98hc, Slc3a2) plays a crucial role in the proliferation of both CD4+ and CD8+ T cells, although it is unclear if CD98hc directly regulates the T cell effector functions that are not linked with T cell proliferation in vivo. Here, we demonstrate that CD98hc is required for both CD4+ T cell proliferation and Th1 functional differentiation. T cell-specific deletion of CD98hc did not affect T cell development in the thymus. CD98hc-deficient CD4+ T cells proliferated in vivo more slowly as compared with control T cells. C57BL/6 mice lacking CD98hc in their CD4+ T cells could not control Leishmania major infections due to lowered IFN-γ production, even with massive CD4+ T cell proliferation. CD98hc-deficient CD4+ T cells exhibited lower IFN-γ production compared with wild-type T cells, even when comparing IFN-γ expression in cells that underwent the same number of cell divisions. Therefore, these data indicate that CD98hc is required for CD4+ T cell expansion and functional Th1 differentiation in vivo, and suggest that CD98hc might be a good target for treating Th1-mediated immune disorders.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular/fisiología , Cadena Pesada de la Proteína-1 Reguladora de Fusión/inmunología , Interferón gamma/inmunología , Animales , Diferenciación Celular/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Células TH1/inmunología
16.
J Leukoc Biol ; 98(5): 781-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26188077

RESUMEN

Invariant natural killer T cells are a distinct subset of T cells that exert Janus-like functions. Moreover, Notch signaling is known to have critical roles in the development and functions of T cells. However, it is not known whether Notch signaling contributes to the development or functions of invariant natural killer T cells. Here, we found that CD4-specific gene ablation of Notch 1 and Notch 2 (N1N2(-/-)) increased the number of invariant natural killer T cells in the thymus but decreased them in the liver. N1N2(-/-) mice showed impaired thymic maturation of invariant natural killer T cells from the NK1.1(-)CD44(+) to the NK1.1(+)CD44(+) stage, resulting in accumulation of NK1.1(-)CD44(+) invariant natural killer T cells in the thymus. Upon activation, hepatic invariant natural killer T cells from N1N2(-/-) mice produced lower cytokine levels and increased apoptosis versus wild-type invariant natural killer T cells. Furthermore, Notch 1/Notch 2-deficient, but not wild type, invariant natural killer T cells failed to promote antibody-induced arthritis in CD1d(-/-) mice. Unlike N1N2(-/-) mice, RBP-j(lox) (/) (lox) CD4-Cre mice showed similar percentages and numbers of thymic invariant natural killer T cells to wild-type mice but had defects in their homeostasis, maturation, and cytokine production in the liver. Taken together, our data indicate distinct effects of Notch signaling on invariant natural killer T cells in the thymus and liver, which are at least partly independent of RBP-j in the thymus.


Asunto(s)
Diferenciación Celular/inmunología , Células T Asesinas Naturales/inmunología , Receptor Notch1/inmunología , Receptor Notch2/inmunología , Transducción de Señal/inmunología , Animales , Antígenos CD1d/genética , Antígenos CD1d/inmunología , Antígenos Ly/genética , Antígenos Ly/inmunología , Diferenciación Celular/genética , Ratones , Ratones Noqueados , Subfamilia B de Receptores Similares a Lectina de Células NK/genética , Subfamilia B de Receptores Similares a Lectina de Células NK/inmunología , Células T Asesinas Naturales/citología , Receptor Notch1/genética , Receptor Notch2/genética , Transducción de Señal/genética , Timo/citología , Timo/inmunología
17.
PLoS One ; 9(2): e89266, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586644

RESUMEN

Experimental autoimmune uveoretinitis (EAU) is an organ-specific T cell-mediated disease induced by immunizing mice with interphotoreceptor retinoid binding protein (IRBP). Autoaggressive CD4(+) T cells are the major pathogenic population for EAU. We investigated the contribution of Notch signaling in T cells to EAU pathogenesis because Notch signaling regulates various aspects of CD4(+) T cell functions. Rbpj is required for Notch signaling, and Rbpj deficiency in T cells inhibited EAU disease severity. The amelioration of EAU in T cell-specific Rbpj-deficient mice correlated with low levels of IL-22 production from CD4(+) T cells, although IRBP-specific CD4(+) T cell proliferation and Th17 differentiation were unaffected. Administration of recombinant IL-22 during the late phase, but not the early phase, of EAU increased EAU clinical scores in T cell-specific Rbpj-deficient mice. Notch inhibition in mice immunized with IRBP with a γ-secretase inhibitor (GSI) suppressed EAU progression, even when GSI was administered as late as 13 days after IRBP immunization. Our data demonstrate that Rbpj/Notch-mediated IL-22 production in T cells has a key pathological role in the late phase of EAU, and suggest that Notch blockade might be a useful therapeutic approach for treating EAU.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Proteínas del Ojo/metabolismo , Interleucinas/metabolismo , Retinitis/metabolismo , Proteínas de Unión al Retinol/metabolismo , Uveítis/metabolismo , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/terapia , Linfocitos T CD4-Positivos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas del Ojo/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Retinitis/genética , Retinitis/terapia , Proteínas de Unión al Retinol/deficiencia , Proteínas de Unión al Retinol/genética , Células Th17/metabolismo , Uveítis/genética , Uveítis/terapia , Interleucina-22
18.
Nat Commun ; 4: 2112, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23836150

RESUMEN

Intestinal intraepithelial T cells contribute to the regulation of inflammatory responses in the intestine; however, the molecular basis for their development and maintenance is unknown. The aryl hydrocarbon receptor complexes with the aryl hydrocarbon receptor nuclear translocator (ARNT) and senses environmental factors, including gut microbiota. Here, we identify ARNT as a critical regulator of the differentiation of TCRαß(+)CD8αα(+) intestinal intraepithelial T cells. Mice deficient in either ARNT or aryl hydrocarbon receptor show a greater than- eight-fold reduction in the number of TCRαß(+)CD8αα(+) intestinal intraepithelial T cells. The number of TCRαß(+)CD8αα(+) intestinal intraepithelial T cells is increased by treatment with an aryl hydrocarbon receptor agonist in germ-free mice and is decreased by antibiotic treatment. The Arnt-deficient precursors of TCRαß(+)CD8αα(+) intestinal intraepithelial T cells express low amounts of STAT3 and fail to differentiate towards the TCRαß(+)CD8αα(+) cell fate after IL-15 stimulation, a deficiency that is overcome by overexpression of Stat3. These data demonstrate that the ARNT-STAT3 axis is a critical regulator of TCRαß(+)CD8αα(+) intestinal intraepithelial T-cell development and differentiation.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Linfocitos T CD8-positivos/citología , Mucosa Intestinal/citología , Intestinos/citología , Células Precursoras de Linfocitos T/citología , Factor de Transcripción STAT3/genética , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/deficiencia , Translocador Nuclear del Receptor de Aril Hidrocarburo/inmunología , Antígenos CD8/genética , Antígenos CD8/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Interleucina-15/farmacología , Mucosa Intestinal/inmunología , Intestinos/efectos de los fármacos , Intestinos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Precursoras de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/inmunología , Transducción de Señal , beta-naftoflavona/farmacología
20.
DNA Res ; 18(4): 291-303, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21791478

RESUMEN

Numerous microbes inhabit the mammalian intestinal track and strongly impact host physiology; however, our understanding of this ecosystem remains limited owing to the high complexity of the microbial community and the presence of numerous non-culturable microbes. Segmented filamentous bacteria (SFBs), which are clostridia-related Gram-positive bacteria, are among such non-culturable populations and are well known for their unique morphology and tight attachment to intestinal epithelial cells. Recent studies have revealed that SFBs play crucial roles in the post-natal maturation of gut immune function, especially the induction of Th17 lymphocytes. Here, we report the complete genome sequence of mouse SFBs. The genome, which comprises a single circular chromosome of 1 620 005 bp, lacks genes for the biosynthesis of almost all amino acids, vitamins/cofactors and nucleotides, but contains a full set of genes for sporulation/germination and, unexpectedly, for chemotaxis/flagella-based motility. These findings suggest a triphasic lifestyle of the SFB, which comprises two types of vegetative (swimming and epicellular parasitic) phases and a dormant (spore) phase. Furthermore, SFBs encode four types of flagellin, three of which are recognized by Toll-like receptor 5 and could elicit the innate immune response. Our results reveal the non-culturability, lifestyle and immunostimulation mechanisms of SFBs and provide a genetic basis for the future development of the SFB cultivation and gene-manipulation techniques.


Asunto(s)
Bacterias/genética , Genoma Bacteriano/genética , Intestinos/microbiología , Animales , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/ultraestructura , Vías Biosintéticas/genética , Quimiotaxis , Cromosomas Bacterianos/genética , Flagelos/metabolismo , Flagelina/inmunología , Flagelina/metabolismo , Orden Génico , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Filogenia , Análisis de Secuencia de ADN , Esporas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA