Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(17): e2318420121, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38621136

RESUMEN

In response to an immune challenge, naive T cells undergo a transition from a quiescent to an activated state acquiring the effector function. Concurrently, these T cells reprogram cellular metabolism, which is regulated by iron. We and others have shown that iron homeostasis controls proliferation and mitochondrial function, but the underlying mechanisms are poorly understood. Given that iron derived from heme makes up a large portion of the cellular iron pool, we investigated iron homeostasis in T cells using mice with a T cell-specific deletion of the heme exporter, FLVCR1 [referred to as knockout (KO)]. Our finding revealed that maintaining heme and iron homeostasis is essential to keep naive T cells in a quiescent state. KO naive CD4 T cells exhibited an iron-overloaded phenotype, with increased spontaneous proliferation and hyperactive mitochondria. This was evidenced by reduced IL-7R and IL-15R levels but increased CD5 and Nur77 expression. Upon activation, however, KO CD4 T cells have defects in proliferation, IL-2 production, and mitochondrial functions. Iron-overloaded CD4 T cells failed to induce mitochondrial iron and exhibited more fragmented mitochondria after activation, making them susceptible to ferroptosis. Iron overload also led to inefficient glycolysis and glutaminolysis but heightened activity in the hexosamine biosynthetic pathway. Overall, these findings highlight the essential role of iron in controlling mitochondrial function and cellular metabolism in naive CD4 T cells, critical for maintaining their quiescent state.


Asunto(s)
Linfocitos T CD4-Positivos , Hierro , Ratones , Animales , Hierro/metabolismo , Mitocondrias/metabolismo , Transducción de Señal , Hemo/metabolismo
2.
J Immunol ; 204(7): 1708-1713, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32122995

RESUMEN

Iron has long been established as a critical mediator of T cell development and proliferation. However, the mechanisms by which iron controls CD4 T cell activation and expansion remain poorly understood. In this study, we show that stimulation of CD4 T cells from C57BL/6 mice not only decreases total and labile iron levels but also leads to changes in the expression of iron homeostatic machinery. Additionally, restraining iron availability in vitro severely inhibited CD4 T cell proliferation and cell cycle progression. Although modulating cellular iron levels increased IL-2 production by activated T lymphocytes, CD25 expression and pSTAT5 levels were decreased, indicating that iron is necessary for IL-2R-mediated signaling. We also found that iron deprivation during T cell stimulation negatively impacts mitochondrial function, which can be reversed by iron supplementation. In all, we show that iron contributes to activation-induced T cell expansion by positively regulating IL-2R signaling and mitochondrial function.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular/fisiología , Hierro/inmunología , Mitocondrias/inmunología , Receptores de Interleucina-2/inmunología , Animales , Femenino , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología
3.
Proc Natl Acad Sci U S A ; 116(15): 7439-7448, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30910955

RESUMEN

Cellular metabolism and signaling pathways are key regulators to determine conventional T cell fate and function, but little is understood about the role of cell metabolism for natural killer T (NKT) cell survival, proliferation, and function. We found that NKT cells operate distinct metabolic programming from CD4 T cells. NKT cells are less efficient in glucose uptake than CD4 T cells with or without activation. Gene-expression data revealed that, in NKT cells, glucose is preferentially metabolized by the pentose phosphate pathway and mitochondria, as opposed to being converted into lactate. In fact, glucose is essential for the effector functions of NKT cells and a high lactate environment is detrimental for NKT cell survival and proliferation. Increased glucose uptake and IFN-γ expression in NKT cells is inversely correlated with bacterial loads in response to bacterial infection, further supporting the significance of glucose metabolism for NKT cell function. We also found that promyelocytic leukemia zinc finger seemed to play a role in regulating NKT cells' glucose metabolism. Overall, our study reveals that NKT cells use distinct arms of glucose metabolism for their survival and function.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Mitocondrias/metabolismo , Células T Asesinas Naturales/inmunología , Fosforilación Oxidativa , Vía de Pentosa Fosfato/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Glucosa/genética , Glucosa/inmunología , Ratones , Ratones Noqueados , Mitocondrias/genética , Células T Asesinas Naturales/citología , Vía de Pentosa Fosfato/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/inmunología
4.
PLoS Pathog ; 15(10): e1007903, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31584995

RESUMEN

HIV-1 gene expression is regulated by host and viral factors that interact with viral motifs and is influenced by proviral integration sites. Here, expression variation among integrants was followed for hundreds of individual proviral clones within polyclonal populations throughout successive rounds of virus and cultured cell replication, with limited findings using CD4+ cells from donor blood consistent with observations in immortalized cells. Tracking clonal behavior by proviral "zip codes" indicated that mutational inactivation during reverse transcription was rare, while clonal expansion and proviral expression states varied widely. By sorting for provirus expression using a GFP reporter in the nef open reading frame, distinct clone-specific variation in on/off proportions were observed that spanned three orders of magnitude. Tracking GFP phenotypes over time revealed that as cells divided, their progeny alternated between HIV transcriptional activity and non-activity. Despite these phenotypic oscillations, the overall GFP+ population within each clone was remarkably stable, with clones maintaining clone-specific equilibrium mixtures of GFP+ and GFP- cells. Integration sites were analyzed for correlations between genomic features and the epigenetic phenomena described here. Integrants inserted in the sense orientation of genes were more frequently found to be GFP negative than those in the antisense orientation, and clones with high GFP+ proportions were more distal to repressive H3K9me3 peaks than low GFP+ clones. Clones with low frequencies of GFP positivity appeared to expand more rapidly than clones for which most cells were GFP+, even though the tested proviruses were Vpr-. Thus, much of the increase in the GFP- population in these polyclonal pools over time reflected differential clonal expansion. Together, these results underscore the temporal and quantitative variability in HIV-1 gene expression among proviral clones that are conferred in the absence of metabolic or cell-type dependent variability, and shed light on cell-intrinsic layers of regulation that affect HIV-1 population dynamics.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Infecciones por VIH/virología , VIH-1/fisiología , Provirus/genética , Integración Viral/genética , Replicación Viral , Linfocitos T CD4-Positivos/metabolismo , Infecciones por VIH/genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Células Jurkat , Transducción Genética
6.
Eur J Immunol ; 48(7): 1255-1257, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29572809

RESUMEN

We show the presence of lymphoid tissue-resident PLZF+ CD45RA+ RO+ CD4 T cells in humans. They express HLA-DR, granzyme B, and perforin and are low on CCR7 like terminally differentiated effector memory (Temra) cells and are likely generated from effector T cells (Te) or from central (Tcm) or effector (Tem) memory T (Tcm) cells during immune responses. Tn, Naïve T cells.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Genotipo , Tejido Linfoide/inmunología , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Subgrupos de Linfocitos T/fisiología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Células Cultivadas , Granzimas/metabolismo , Antígenos HLA-DR/metabolismo , Humanos , Inmunidad Celular , Memoria Inmunológica , Perforina/metabolismo
7.
J Immunol ; 199(10): 3478-3487, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29021374

RESUMEN

Reactive oxygen species (ROS) are byproducts of aerobic metabolism and contribute to both physiological and pathological conditions as second messengers. ROS are essential for activation of T cells, but how ROS influence NKT cells is unknown. In the present study, we investigated the role of ROS in NKT cell function. We found that NKT cells, but not CD4 or CD8 T cells, have dramatically high ROS in the spleen and liver of mice but not in the thymus or adipose tissues. Accordingly, ROS-high NKT cells exhibited increased susceptibility and apoptotic cell death with oxidative stress. High ROS in the peripheral NKT cells were primarily produced by NADPH oxidases and not mitochondria. We observed that sorted ROS-high NKT cells were enriched in NKT1 and NKT17 cells, whereas NKT2 cells were dominant in ROS-low cells. Furthermore, treatment of NKT cells with antioxidants led to reduced frequencies of IFN-γ- and IL-17-expressing cells, indicating that ROS play a role in regulating the inflammatory function of NKT cells. The transcription factor promyelocytic leukemia zinc finger (PLZF) seemed to control the ROS levels. NKT cells from adipose tissues that do not express PLZF and those from PLZF haplodeficient mice have low ROS. Conversely, ROS were highly elevated in CD4 T cells from mice ectopically expressing PLZF. Thus, our findings demonstrate that PLZF controls ROS levels, which in turn governs the inflammatory function of NKT cells.


Asunto(s)
Hígado/inmunología , Células T Asesinas Naturales/inmunología , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Bazo/inmunología , Animales , Apoptosis , Células Cultivadas , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética
8.
J Hepatol ; 67(1): 100-109, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28267623

RESUMEN

BACKGROUND & AIMS: The liver is an immunologically-privileged organ. Breakdown of liver immune privilege has been reported in chronic liver disease; however, the role of adaptive immunity in liver injury is poorly defined. Nuclear factor-κB-inducing kinase (NIK) is known to regulate immune tissue development, but its role in maintaining liver homeostasis remains unknown. This study aimed to assess the role of NIK, particularly thymic NIK, in regulating liver adaptive immunity. METHODS: NIK was deleted systemically or conditionally using the Cre/loxp system. Cluster of differentiation [CD]4+ or CD8+ T cells were depleted using anti-CD4 or anti-CD8 antibody. Donor bone marrows or thymi were transferred into recipient mice. Immune cells were assessed by immunohistochemistry and flow cytometry. RESULTS: Global, but not liver-specific or hematopoietic lineage cell-specific, deletion of NIK induced fatal liver injury, inflammation, and fibrosis. Likewise, adoptive transfer of NIK-null, but not wild-type, thymi into immune-deficient mice induced liver inflammation, injury, and fibrosis in recipients. Liver inflammation was characterized by a massive expansion of T cells, particularly the CD4+ T cell subpopulation. Depletion of CD4+, but not CD8+, T cells fully protected against liver injury, inflammation, and fibrosis in NIK-null mice. NIK deficiency also resulted in inflammation in the lung, kidney, and pancreas, but to a lesser degree relative to the liver. CONCLUSIONS: Thymic NIK suppresses development of autoreactive T cells against liver antigens, and NIK deficiency in the thymus results in CD4+ T cell-orchestrated autoimmune hepatitis and liver fibrosis. Thus, thymic NIK is essential for the maintenance of liver immune privilege and liver homeostasis. LAY SUMMARY: We found that global or thymus-specific ablation of the NIK gene results in fatal autoimmune liver disease in mice. NIK-deficient mice develop liver inflammation, injury, and fibrosis. Our findings indicate that thymic NIK is essential for the maintenance of liver integrity and homeostasis.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Hepatitis Autoinmune/etiología , Cirrosis Hepática Experimental/etiología , Hígado/inmunología , Proteínas Serina-Treonina Quinasas/fisiología , Timo/fisiología , Inmunidad Adaptativa , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quinasa de Factor Nuclear kappa B
9.
J Immunol ; 194(1): 223-30, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25404366

RESUMEN

The mammalian target of rapamycin (mTOR) senses and incorporates different environmental cues via the two signaling complexes mTOR complex 1 (mTORC1) and mTORC2. As a result, mTOR controls cell growth and survival, and also shapes different effector functions of the cells including immune cells such as T cells. We demonstrate in this article that invariant NKT (iNKT) cell development is controlled by mTORC2 in a cell-intrinsic manner. In mice deficient in mTORC2 signaling because of the conditional deletion of the Rictor gene, iNKT cell numbers were reduced in the thymus and periphery. This is caused by decreased proliferation of stage 1 iNKT cells and poor development through subsequent stages. Functionally, iNKT cells devoid of mTORC2 signaling showed reduced number of IL-4-expressing cells, which correlated with a decrease in the transcription factor GATA-3-expressing cells. However, promyelocytic leukemia zinc-finger (PLZF), a critical transcription factor for iNKT cell development, is expressed at a similar level in mTORC2-deficient iNKT cells compared with that in the wild type iNKT cells. Furthermore, cellular localization of PLZF was not altered in the absence of mTOR2 signaling. Thus, our study reveals the PLZF-independent mechanisms of the development and function of iNKT cells regulated by mTORC2.


Asunto(s)
Proteínas Portadoras/inmunología , Factor de Transcripción GATA3/biosíntesis , Factores de Transcripción de Tipo Kruppel/biosíntesis , Complejos Multiproteicos/inmunología , Células T Asesinas Naturales/citología , Serina-Treonina Quinasas TOR/inmunología , Animales , Proteínas Portadoras/genética , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Interleucina-4/biosíntesis , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/genética , Células T Asesinas Naturales/inmunología , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Proteína Asociada al mTOR Insensible a la Rapamicina , Transducción de Señal/genética , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/genética
10.
Proc Natl Acad Sci U S A ; 110(6): 2270-5, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23341605

RESUMEN

Myeloid differentiation primary response protein 88 (MyD88) is classically known as an adaptor, linking TLR and IL-1R to downstream signaling pathways in the innate immune system. In addition to its role in innate immune cells, MyD88 has been shown to play an important role in T cells. How MyD88 regulates helper T-cell differentiation remains largely unknown, however. Here we demonstrate that MyD88 is an important regulator of IL-17-producing CD4(+) T helper cells (Th17) cell proliferation. MyD88-deficient CD4(+) T cells showed a defect in Th17 cell differentiation, but not in Th1 cell or Th2 cell differentiation. The impaired IL-17 production from MyD88-deficient CD4(+) T cells is not a result of defective RAR-related orphan receptor γt (RORγt) expression. Instead, MyD88 is essential for sustaining the mammalian target of rapamycin (mTOR) activation necessary to promote Th17 cell proliferation by linking IL-1 and IL-23 signaling. MyD88-deficient CD4(+) T cells showed impaired mTOR activation and, consequently, reduced Th17 cell proliferation. Importantly, the absence of MyD88 in T cells ameliorated disease in the experimental autoimmune encephalomyelitis model. Taken together, our results demonstrate that MyD88 has a dual function in Th17 cells by delivering IL-1 signaling during the early differentiation stage and integrating IL-23 signaling to the mTOR complex to expand committed Th17 cells.


Asunto(s)
Interleucina-1/metabolismo , Interleucina-23/metabolismo , Factor 88 de Diferenciación Mieloide/inmunología , Serina-Treonina Quinasas TOR/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular , Proliferación Celular , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Inmunidad Innata , Interleucina-17/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Células Th17/citología , Células Th17/metabolismo
11.
BMC Immunol ; 16: 62, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26482437

RESUMEN

BACKGROUND: Invariant Natural Killer T (iNKT) cells have been implicated in lung inflammation in humans and also shown to be a key cell type in inducing allergic lung inflammation in mouse models. iNKT cells differentiate and acquire functional characteristics during development in the thymus. However, the correlation between development of iNKT cells in the thymus and role in lung inflammation remains unknown. In addition, transcriptional control of differentiation of iNKT cells into iNKT cell effector subsets in the thymus during development is also unclear. In this report we show that ß-catenin dependent mechanisms direct differentiation of iNKT2 and iNKT17 subsets but not iNKT1 cells. METHODS: To study the role for ß-catenin in lung inflammation we utilize mice with conditional deletion and enforced expression of ß-catenin in a well-established mouse model for IL-25-dependen lung inflammation. RESULTS: Specifically, we demonstrate that conditional deletion of ß-catenin permitted development of mature iNKT1 cells while impeding maturation of iNKT2 and 17 cells. A role for ß-catenin expression in promoting iNKT2 and iNKT17 subsets was confirmed when we noted that enforced transgenic expression of ß-catenin in iNKT cell precursors enhanced the frequency and number of iNKT2 and iNKT17 cells at the cost of iNKT1 cells. This effect of expression of ß-catenin in iNKT cell precursors was cell autonomous. Furthermore, iNKT2 cells acquired greater capability to produce type-2 cytokines when ß-catenin expression was enhanced. DISCUSSION: This report shows that ß-catenin deficiency resulted in a profound decrease in iNKT2 and iNKT17 subsets of iNKT cells whereas iNKT1 cells developed normally. By contrast, enforced expression of ß-catenin promoted the development of iNKT2 and iNKT17 cells. It was important to note that the majority of iNKT cells in the thymus of C57BL/6 mice were iNKT1 cells and enforced expression of ß-catenin altered the pattern to iNKT2 and iNKT17 cells suggesting that ß-catenin may be a major factor in the distinct pathways that critically direct differentiation of iNKT effector subsets. CONCLUSIONS: Thus, we demonstrate that ß-catenin expression in iNKT cell precursors promotes differentiation toward iNKT2 and iNKT17 effector subsets and supports enhanced capacity to produce type 2 and 17 cytokines which in turn augment lung inflammation in mice.


Asunto(s)
Diferenciación Celular , Interleucina-17/metabolismo , Células T Asesinas Naturales/inmunología , Neumonía/inmunología , Neumonía/patología , beta Catenina/metabolismo , Animales , Hiperreactividad Bronquial/complicaciones , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Neumonía/complicaciones
12.
J Immunol ; 191(2): 737-44, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23776174

RESUMEN

MHC class II-expressing thymocytes can efficiently mediate positive selection of CD4 T cells in mice. Thymocyte-selected CD4 (T-CD4) T cells have an innate-like phenotype similar to invariant NKT cells. To investigate the development and function of T-CD4 T cells in-depth, we cloned TCR genes from T-CD4 T cells and generated transgenic mice. Remarkably, positive selection of T-CD4 TCR transgenic (T3) thymocytes occurred more efficiently when MHC class II was expressed by thymocytes than by thymic epithelial cells. Similar to polyclonal T-CD4 T cells and also invariant NKT cells, T3 CD4 T cell development is controlled by signaling lymphocyte activation molecule/signaling lymphocyte activation molecule-associated protein signaling, and the cells expressed both IL-4 and promyelocytic leukemia zinc finger (PLZF). Surprisingly, the selected T3 CD4 T cells were heterogeneous in that only half expressed IL-4 and only half expressed PLZF. IL-4- and PLZF-expressing cells were first found at the double-positive cell stage. Thus, the expression of IL-4 and PLZF seems to be determined by an unidentified event that occurs postselection and is not solely dependent on TCR specificity or the selection process, per se. Taken together, our data show for the first time, to our knowledge, that the TCR specificity regulates but does not determine the development of innate CD4 T cells by thymocytes.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interleucina-4/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/genética , Animales , Antígenos CD/metabolismo , Células de la Médula Ósea , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Quimera/genética , Antígenos de Histocompatibilidad Clase II , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Nucleares/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal/inmunología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Timocitos/metabolismo , Transactivadores/genética
13.
Proc Natl Acad Sci U S A ; 109(40): 16264-9, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-22988097

RESUMEN

MHC class II-expressing thymocytes and thymic epithelial cells can mediate CD4 T-cell selection resulting in functionally distinct thymocyte-selected CD4 (T-CD4) and epithelial-selected CD4 (E-CD4) T cells, respectively. However, little is known about how T-cell receptor (TCR) signaling influences the development of these two CD4 T-cell subsets. To study TCR signaling for T-CD4 T-cell development, we used a GFP reporter system of Nur77 in which GFP intensity directly correlates with TCR signaling strength. T-CD4 T cells expressed higher levels of GFP than E-CD4 T cells, suggesting that T-CD4 T cells received stronger TCR signaling than E-CD4 T cells during selection. Elimination of Ras GTPase-activating protein enhanced E-CD4 but decreased T-CD4 T-cell selection efficiency, suggesting a shift to negative selection. Conversely, the absence of IL-2-inducible T-cell kinase that causes poor E-CD4 T-cell selection due to insufficient TCR signaling improved T-CD4 T-cell generation, consistent with rescue from negative selection. Strong TCR signaling during T-CD4 T-cell development correlates with the expression of the transcription factor promyelocytic leukemia zinc finger protein. However, although modulation of the signaling strength affected the efficiency of T-CD4 T-cell development during positive and negative selection, the signaling strength is not as important for the effector function of T-CD4 T cells. These findings indicate that innate T-CD4 T cells, together with invariant natural killer T cells and γδ T cells, receive strong TCR signals during their development and that signaling requirements for the development and the effector functions are distinct.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/inmunología , Factores de Transcripción de Tipo Kruppel/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Trasplante de Médula Ósea , Epitelio/inmunología , Citometría de Flujo , Proteínas Fluorescentes Verdes , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Ratones Noqueados , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Proteínas Tirosina Quinasas/genética , Especificidad del Receptor de Antígeno de Linfocitos T , Timocitos/citología , Timocitos/inmunología
14.
J Exp Med ; 204(9): 2145-57, 2007 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-17724129

RESUMEN

Recently, a new developmental pathway for CD4 T cells that is mediated by major histocompatibility complex class II-positive thymocytes was identified (Choi, E.Y., K.C. Jung, H.J. Park, D.H. Chung, J.S. Song, S.D. Yang, E. Simpson, and S.H. Park. 2005. Immunity. 23:387-396; Li, W., M.G. Kim, T.S. Gourley, B.P. McCarthy, D.B. Sant'angelo, and C.H. Chang. 2005. Immunity. 23:375-386). We demonstrate that thymocyte-selected CD4 (T-CD4) T cells can rapidly produce interferon gamma and interleukin (IL) 4 upon in vivo and in vitro T cell receptor stimulation. These T-CD4 T cells appear to be effector cells producing both T helper type 1 (Th1) and Th2 cytokines, and they maintain a potential to produce Th2 cytokines under Th1-skewing conditions in a signal transducer and activator of transcription 6-independent manner. The IL-4 mRNA level is high in CD4 single-positive thymocytes if they are selected on thymocytes, which is at least partly caused by enhanced histone acetylation of the IL-4 locus. However, mice that can generate T-CD4 T cells showed attenuated immune responses in an allergen-induced airway inflammation model, suggesting a protective role for T-CD4 T cells during an airway challenge. Our results imply that this thymic selection pathway plays an important role in determining the effector function of the resulting CD4 cells and in regulating immune response.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Selección Genética , Timo/inmunología , Alérgenos , Animales , Presentación de Antígeno/inmunología , Células Epiteliales/inmunología , Regulación de la Expresión Génica , Antígenos de Histocompatibilidad Clase II , Humanos , Inflamación , Interferón gamma/biosíntesis , Interleucina-4/biosíntesis , Interleucina-4/genética , Células Asesinas Naturales/inmunología , Ratones , Ratones Transgénicos , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sistema Respiratorio/patología , Factor de Transcripción STAT6/metabolismo , Células TH1/metabolismo , Células Th2/metabolismo
15.
J Virol ; 86(8): 4194-203, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22345470

RESUMEN

There is an incomplete understanding of the differences between neonatal immune responses that contribute to the increased susceptibility of neonates to some viral infections. We tested the hypothesis that neonates are more susceptible than adults to mouse adenovirus type 1 (MAV-1) respiratory infection and are impaired in the ability to generate a protective immune response against a second infection. Following intranasal infection, lung viral loads were greater in neonates than in adults during the acute phase but the virus was cleared from the lungs of neonates as efficiently as it was from adult lungs. Lung gamma interferon (IFN-γ) responses were blunted and delayed in neonates, and lung viral loads were higher in adult IFN-γ(-/-) mice than in IFN-γ(+/+) controls. However, administration of recombinant IFN-γ to neonates had no effect on lung viral loads. Recruitment of inflammatory cells to the airways was impaired in neonates. CD4 and CD8 T cell responses were similar in the lungs of neonates and adults, although a transient increase in regulatory T cells occurred only in the lungs of infected neonates. Infection of neonates led to protection against reinfection later in life that was associated with increased effector memory CD8 T cells in the lungs. We conclude that neonates are more susceptible than adults to acute MAV-1 respiratory infection but are capable of generating protective immune responses.


Asunto(s)
Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/prevención & control , Adenoviridae/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/prevención & control , Infecciones por Adenoviridae/genética , Animales , Línea Celular , Citocinas/biosíntesis , Interferón gamma/deficiencia , Interferón gamma/genética , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Infecciones del Sistema Respiratorio/genética , Linfocitos T/inmunología , Carga Viral
16.
J Immunol ; 187(1): 151-63, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21646295

RESUMEN

Activation of the Ras small GTP-binding protein is necessary for normal T cell development and function. However, it is unknown which Ras GTPase-activating proteins (RasGAPs) inactivate Ras in T cells. We used a T cell-specific RASA1-deficient mouse model to investigate the role of the p120 RasGAP (RASA1) in T cells. Death of CD4(+)CD8(+) double-positive thymocytes was increased in RASA1-deficient mice. Despite this finding, on an MHC class II-restricted TCR transgenic background, evidence was obtained for increased positive selection of thymocytes associated with augmented activation of the Ras-MAPK pathway. In the periphery, RASA1 was found to be dispensable as a regulator of Ras-MAPK activation and T cell functional responses induced by full agonist peptides. However, numbers of naive T cells were substantially reduced in RASA1-deficient mice. Loss of naive T cells in the absence of RASA1 could be attributed in part to impaired responsiveness to the IL-7 prosurvival cytokine. These findings reveal an important role for RASA1 as a regulator of double-positive survival and positive selection in the thymus as well as naive T cell survival in the periphery.


Asunto(s)
Fase de Descanso del Ciclo Celular/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Timo/citología , Timo/inmunología , Proteína Activadora de GTPasa p120/fisiología , Proteínas Activadoras de ras GTPasa/fisiología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fase de Descanso del Ciclo Celular/genética , Subgrupos de Linfocitos T/metabolismo , Timo/metabolismo , Proteína Activadora de GTPasa p120/deficiencia , Proteína Activadora de GTPasa p120/genética , Proteínas Activadoras de ras GTPasa/deficiencia , Proteínas Activadoras de ras GTPasa/genética
17.
J Immunol ; 186(5): 2792-9, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21282512

RESUMEN

IL-4 expression is known to be activated in CD4 T cells when they are differentiated to Th2 but not Th1 cells. However, CD4 T cells selected by MH class II-expressing thymocytes, named thymocyte-selected CD4 T cells (T-CD4 T cells), express IL-4 under both Th1 and Th2 conditions. In this study, we investigated molecular mechanisms by which IL-4 gene expression is regulated in T-CD4 T cells. We found that T-CD4 T cells express IL-4 soon after selection in the thymus. Deficiency of DNase I hypersensitive (HS) sites HS5a and HS5 at the 3'-enhancer region in the IL-4 gene decreased IL-4 production, but T-CD4 T cells were able to make IL-4 under the Th1-inducing condition. Consistent with this, IL-4 was expressed in Th1 differentiated T-CD4 T cells in the absence of recombination signal binding protein-J that interacts with HS5. When HS5 was examined separately from other endogenous regulatory elements using a reporter system, CD4 T cells that are selected by thymic epithelial cells cannot transcribe the IL-4 reporter gene with HS5 alone. However, HS5 was able to induce the expression of the IL-4 reporter gene in T-CD4 T cells. Interestingly, the Th1 differentiating signal led to deacetylation at HS5 of the IL-4 endogenous gene, whereas the Th2-inducing environment had no effect. Therefore, in T-CD4 T cells, HS5 plays an essential role during the induction phase of IL-4 expression, but the maintenance of IL-4 expression in Th1 cells requires additional regulatory elements.


Asunto(s)
Regiones no Traducidas 3'/inmunología , Elementos de Facilitación Genéticos/inmunología , Regulación de la Expresión Génica/inmunología , Interleucina-4/biosíntesis , Interleucina-4/genética , Células TH1/inmunología , Células Th2/inmunología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Desoxirribonucleasa I/deficiencia , Desoxirribonucleasa I/genética , Genes Reporteros/inmunología , Humanos , Interleucina-4/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células TH1/citología , Células TH1/metabolismo , Células Th2/citología , Células Th2/metabolismo , Timo/citología , Timo/inmunología , Timo/metabolismo
18.
J Immunol ; 186(10): 5749-57, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21478404

RESUMEN

We have recently shown that MHC class II-dependent thymocyte-thymocyte (T-T) interaction successfully generates CD4(+) T cells (T-T CD4(+) T cells), and that T-T CD4(+) T cells expressing promyelocytic leukemia zinc finger protein (PLZF) show an innate property both in mice and humans. In this article, we report that the thymic T-T interaction is essential for the conversion of CD8(+) T cells into innate phenotype in the physiological condition. CD8(+) T cells developed in the presence of PLZF(+) CD4(+) T cells showed marked upregulation of eomesodermin (Eomes), activation/memory phenotype, and rapid production of IFN-γ on ex vivo stimulation. Their development was highly dependent on the PLZF expression in T-T CD4(+) T cells and the IL-4 secreted by PLZF(+) T-T CD4(+) T cells. The same events may take place in humans, as a substantial number of Eomes expressing innate CD8(+) T cells were found in human fetal thymi and spleens. It suggests that PLZF(+) T-T CD4(+) T cells in combination with Eomes(+) CD8(+) T cells might actively participate in the innate immune response against various pathogens, particularly in human perinatal period.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Comunicación Celular , Antígenos de Histocompatibilidad Clase II/inmunología , Inmunidad Innata , Factores de Transcripción de Tipo Kruppel/genética , Timo/citología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Citometría de Flujo , Humanos , Interferón gamma/biosíntesis , Interleucina-4/inmunología , Activación de Linfocitos , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Fenotipo , Reacción en Cadena de la Polimerasa , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Proteínas de Dominio T Box/metabolismo , Timo/embriología , Timo/inmunología , Timo/metabolismo
19.
Proc Natl Acad Sci U S A ; 107(18): 8340-5, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20404153

RESUMEN

IL-12 and IL-23 are produced by activated antigen-presenting cells but the two induce distinct immune responses by promoting Th1 and Th17 cell differentiation, respectively. IL-23 is a heterodimeric cytokine consisting of two subunits: p40 that is shared with IL-12 and p19 unique to IL-23. In this study, we showed that the production of IL-23 but not IL-12 was negatively regulated by protein phosphatase 2A (PP2A) in dendritic cells (DC). PP2A inhibits IL-23 production by suppressing the expression of the IL-23p19 gene. Treating DC with okadaic acid that inhibits the PP2A activity or knocking down the catalytic subunit of PP2A with siRNA enhanced IL-23 but not IL-12 production. Unlike PP2A, MAP kinase phosphatase-1 or CYLD did not show an effect on IL-23 production supporting the specificity of PP2A. PP2A-mediated inhibition requires a newly made protein that is likely responsible for bringing PP2A and IKKbeta together upon LPS stimulation, which then results in the termination of IKK phosphorylation. Thus, our results uncovered an important role of the protein phosphatase in the regulation of IL-23 production and identified PP2A as a previously uncharacterized inhibitor of IL-23p19 expression in DC.


Asunto(s)
Células Dendríticas/inmunología , Regulación hacia Abajo , Subunidad p19 de la Interleucina-23/inmunología , Proteína Fosfatasa 2/metabolismo , Animales , Células Cultivadas , Células Dendríticas/metabolismo , Fosfatasa 1 de Especificidad Dual/deficiencia , Fosfatasa 1 de Especificidad Dual/metabolismo , Quinasa I-kappa B/metabolismo , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Subunidad p19 de la Interleucina-23/biosíntesis , Subunidad p19 de la Interleucina-23/genética , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fosforilación , Unión Proteica , Proteína Fosfatasa 2/genética , ARN Interferente Pequeño/genética
20.
Immunohorizons ; 7(3): 235-242, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36951874

RESUMEN

The E3 ubiquitin ligase cullin 3 (Cul3) is critical for invariant NKT (iNKT) cell development, as iNKT cells lacking Cul3 accumulate in the immature developmental stages. However, the mechanisms by which Cul3 mediates iNKT cell development remain unknown. In this study, we investigated the role of Cul3 in both immature and mature thymic iNKT cells using a mouse model with a T cell-specific deletion of Cul3. We found that mature iNKT cells lacking Cul3 proliferated and died more than wild-type cells did. These cells also displayed increased glucose metabolism and autophagy. Interestingly, we found that tight regulation of iron homeostasis is critical for iNKT cell development. Without Cul3, mature iNKT cells harbored higher levels of cytosolic iron, a phenotype associated with increased cell death. Taken together, our data suggest that Cul3 promotes iNKT cell development partially through intracellular iron homeostasis.


Asunto(s)
Células T Asesinas Naturales , Animales , Ratones , Diferenciación Celular/genética , Células T Asesinas Naturales/metabolismo , Ratones Noqueados , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Homeostasis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA