Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Annu Rev Immunol ; 40: 1-14, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-34871102

RESUMEN

I've had serious misgivings about writing this article, because from living the experience day by day, it's hard to believe my accomplishments merit the attention. To skirt this roadblock, I forced myself to pretend I was in a conversation with my trainees, trying to distill the central driving forces of my career in science. The below chronicles my evolution from would-be astronaut/ballerina to budding developmental biologist to devoted T cell immunologist. It traces my work from a focus on intrathymic events that mold developing T cells into self-major histocompatibility complex (MHC)-restricted lymphocytes to extrathymic events that fine-tune the T cell receptor (TCR) repertoire and impose the finishing touches on T cell maturation. It is a story of a few personal attributes multiplied by generous mentors, good luck, hard work, perseverance, and knowing when to step down.


Asunto(s)
Complejo Mayor de Histocompatibilidad , Linfocitos T , Animales , Diferenciación Celular , Humanos , Timo
2.
Annu Rev Immunol ; 31: 31-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23121398

RESUMEN

The generation of the TCRαß lineage of T cells occurs in the thymus through a series of orchestrated developmental events that result in a carefully selected population of CD4 or CD8 lineage-committed TCR(+) thymocytes capable of recognizing foreign antigen in the context of self MHC. T cells first exit the thymus in a phenotypically and functionally immature state and require an approximately 3-week period of post-thymic maturation before transitioning into the mature T cell compartment. A greater understanding of recent thymic emigrant biology has come with the development of methods to exclusively identify and isolate this population for further characterization. I now review current knowledge about the phenotype and function of this key but understudied population of peripheral T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Linaje de la Célula/inmunología , Movimiento Celular/inmunología , Senescencia Celular/inmunología , Timo/citología , Timo/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Movimiento Celular/genética , Senescencia Celular/genética , Humanos , Receptores de Antígenos de Linfocitos T alfa-beta/química , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Timo/metabolismo
3.
J Immunol ; 201(6): 1627-1632, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30068595

RESUMEN

Recent thymic emigrants (RTEs) are peripheral T cells that have most recently completed selection and thymic egress and constitute a population that is phenotypically and functionally distinct from its more mature counterpart. Ag-activated RTEs are less potent effectors than are activated mature T cells, due in part to reduced aerobic glycolysis (correctable by exogenous IL-2), which in turn impacts IFN-γ production. Mitochondria serve as nodal regulators of cell function, but their contribution to the unique biology of RTEs is unknown. In this study, we show that activated mouse RTEs have impaired oxidative phosphorylation, even in the presence of exogenous IL-2. This altered respiratory phenotype is the result of decreased CD28 signaling, reduced glutaminase induction, and diminished mitochondrial mass in RTEs relative to mature T cells. These results suggest an uncoupling whereby IL-2 tunes the rate of RTE glycolytic metabolism, whereas the unique profile of RTE mitochondrial metabolism is "hard wired."


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Glucólisis/inmunología , Activación de Linfocitos , Mitocondrias/inmunología , Timo/inmunología , Animales , Antígenos CD28/genética , Antígenos CD28/inmunología , Linfocitos T CD8-positivos/citología , Movimiento Celular/genética , Glucólisis/genética , Interleucina-2/genética , Interleucina-2/inmunología , Ratones , Ratones Noqueados , Mitocondrias/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Timo/citología
4.
J Immunol ; 198(12): 4575-4580, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28507025

RESUMEN

Recent thymic emigrants (RTEs) are the youngest peripheral T cells that have completed thymic selection and egress to the lymphoid periphery. RTEs are functionally distinct from their more mature but still naive T cell counterparts, because they exhibit dampened proliferation and reduced cytokine production upon activation. In this article, we show that, compared with more mature but still naive T cells, RTEs are impaired in their ability to perform aerobic glycolysis following activation. Impaired metabolism underlies the reduced IFN-γ production observed in activated RTEs. This failure to undergo Ag-induced aerobic glycolysis is caused by reduced mTORC1 activity and diminished Myc induction in RTEs. Critically, exogenous IL-2 restores Myc expression in RTEs, driving aerobic glycolysis and IFN-γ production to the level of mature T cells. These results reveal a previously unknown metabolic component to postthymic T cell maturation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Movimiento Celular , Glucólisis , Activación de Linfocitos , Timo/citología , Animales , Diferenciación Celular , Genes myc , Glucólisis/efectos de los fármacos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-2/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Timo/inmunología
5.
J Immunol ; 196(6): 2450-5, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26873989

RESUMEN

The youngest peripheral T cells (recent thymic emigrants [RTEs]) are functionally distinct from naive T cells that have completed postthymic maturation. We assessed the RTE memory response and found that RTEs produced less granzyme B than their mature counterparts during infection but proliferated more and, therefore, generated equivalent target killing in vivo. Postinfection, RTE numbers contracted less dramatically than those of mature T cells, but RTEs were delayed in their transition to central memory, displaying impaired expression of CD62L, IL-2, Eomesodermin, and CXCR4, which resulted in impaired bone marrow localization. RTE-derived and mature memory cells expanded equivalently during rechallenge, indicating that the robust proliferative capacity of RTEs was maintained independently of central memory phenotype. Thus, the diminished effector function and delayed central memory differentiation of RTE-derived memory cells are counterbalanced by their increased proliferative capacity, driving the efficacy of the RTE response to that of mature T cells.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Activación de Linfocitos/inmunología , Traslado Adoptivo , Animales , Diferenciación Celular/inmunología , Separación Celular , Citometría de Flujo , Ratones , Ratones Transgénicos , Timo/citología , Timo/inmunología
6.
J Immunol ; 194(4): 1677-85, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25582857

RESUMEN

All aerobic cells and organisms must synthesize heme from the amino acid glycine and the tricarboxylic acid cycle intermediate succinyl CoA for incorporation into hemoproteins, such as the cytochromes needed for oxidative phosphorylation. Most studies on heme regulation have been done in erythroid cells or hepatocytes; however, much less is known about heme metabolism in other cell types. The feline leukemia virus subgroup C receptor (FLVCR) is a 12-transmembrane domain surface protein that exports heme from cells, and it was shown to be required for erythroid development. In this article, we show that deletion of Flvcr in murine hematopoietic precursors caused a complete block in αß T cell development at the CD4(+)CD8(+) double-positive stage, although other lymphoid lineages were not affected. Moreover, FLVCR was required for the proliferation and survival of peripheral CD4(+) and CD8(+) T cells. These studies identify a novel and unexpected role for FLVCR, a major facilitator superfamily metabolite transporter, in T cell development and suggest that heme metabolism is particularly important in the T lineage.


Asunto(s)
Diferenciación Celular/inmunología , Hemo/inmunología , Proteínas de Transporte de Membrana/inmunología , Receptores Virales/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Separación Celular , Supervivencia Celular/inmunología , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Proc Natl Acad Sci U S A ; 111(15): 5652-7, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24706795

RESUMEN

Peripheral CD4 T cells in Vß5 transgenic (Tg) C57BL/6J mice undergo tolerance to an endogenous superantigen encoded by mouse mammary tumor virus 8 (Mtv-8) by either deletion or T-cell receptor (TCR) revision. Revision is a process by which surface expression of the Vß5(+) TCR is down-regulated in response to Mtv-8 and recombination activating genes are expressed to drive rearrangement of the endogenous TCRß locus, effecting cell rescue through the expression of a newly generated, non-self-reactive TCR. In an effort to identify the microenvironment in which revision takes place, we show here that the proportion of T follicular helper cells (Tfh) and production of high-affinity antibody during a primary response are increased in Vß5 Tg mice in an Mtv-8-dependent manner. Revising T cells have a Tfh-like surface phenotype and transcription factor profile, with elevated expression of B-cell leukemia/lymphoma 6 (Bcl-6), CXC chemokine receptor 5, programmed death-1, and other Tfh-associated markers. Efficient revision requires Bcl-6 and is inhibited by B lymphocyte-induced maturation protein-1. Revision completes less efficiently in the absence of signaling lymphocytic activation molecule-associated protein although initiation proceeds normally. These data indicate that Tfh formation is required for the initiation of revision and germinal-center interactions for its completion. The germinal center is known to provide a confined space in which B-cell antigen receptors undergo selection. Our data extend the impact of this selective microenvironment into the arena of T cells, suggesting that this fluid structure also provides a regulatory environment in which TCR revision can safely take place.


Asunto(s)
Reordenamiento Génico de Linfocito T/inmunología , Centro Germinal/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Cartilla de ADN/genética , Citometría de Flujo , Ratones , Reacción en Cadena de la Polimerasa , Receptores de Antígenos de Linfocitos T/metabolismo , Recombinación Genética/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Colaboradores-Inductores/metabolismo
8.
J Immunol ; 193(7): 3262-6, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25172492

RESUMEN

To explore the TCR sensitivity of recent thymic emigrants (RTEs), we triggered T cells with altered peptide ligands (APLs). Upon peptide stimulation in vitro, RTEs exhibited increased TCR signal transduction, and following infection in vivo with APL-expressing bacteria, CD8 RTEs expanded to a greater extent in response to low-affinity Ags than did their mature T cell counterparts. RTEs skewed to short-lived effector cells in response to all APLs but also were characterized by diminished cytokine production. RTEs responding to infection expressed increased levels of VLA-4, with consequent improved entry into inflamed tissue and pathogen clearance. These positive outcomes were offset by the capacity of RTEs to elicit autoimmunity. Overall, salient features of CD8 RTE biology should inform strategies to improve neonatal vaccination and therapies for cancer and HIV, because RTEs make up a large proportion of the T cells in lymphodepleted environments.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Timo/inmunología , Animales , Antígenos/genética , Antígenos/inmunología , Linfocitos T CD8-positivos/patología , Movimiento Celular/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Integrina alfa4beta1/genética , Integrina alfa4beta1/inmunología , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética , Timo/patología
9.
PLoS Genet ; 9(8): e1003708, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23990801

RESUMEN

Most yeast ribosomal protein genes are duplicated and their characterization has led to hypotheses regarding the existence of specialized ribosomes with different subunit composition or specifically-tailored functions. In yeast, ribosomal protein genes are generally duplicated and evidence has emerged that paralogs might have specific roles. Unlike yeast, most mammalian ribosomal proteins are thought to be encoded by a single gene copy, raising the possibility that heterogenous populations of ribosomes are unique to yeast. Here, we examine the roles of the mammalian Rpl22, finding that Rpl22(-/-) mice have only subtle phenotypes with no significant translation defects. We find that in the Rpl22(-/-) mouse there is a compensatory increase in Rpl22-like1 (Rpl22l1) expression and incorporation into ribosomes. Consistent with the hypothesis that either ribosomal protein can support translation, knockdown of Rpl22l1 impairs growth of cells lacking Rpl22. Mechanistically, Rpl22 regulates Rpl22l1 directly by binding to an internal hairpin structure and repressing its expression. We propose that ribosome specificity may exist in mammals, providing evidence that one ribosomal protein can influence composition of the ribosome by regulating its own paralog.


Asunto(s)
Proteínas de Unión al ARN/genética , ARN/genética , Proteínas Ribosómicas/genética , Ribosomas/genética , Homología de Secuencia de Aminoácido , Secuencia de Aminoácidos , Animales , Regulación de la Expresión Génica , Ratones , Datos de Secuencia Molecular , Biosíntesis de Proteínas , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo
11.
J Immunol ; 190(12): 6180-6, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23686491

RESUMEN

Recent thymic emigrants (RTEs) are the youngest T cells in the lymphoid periphery and exhibit phenotypic and functional characteristics distinct from those of their more mature counterparts in the naive peripheral T cell pool. We show in this study that the Il2 and Il4 promoter regions of naive CD4(+) RTEs are characterized by site-specific hypermethylation compared with those of both mature naive (MN) T cells and the thymocyte precursors of RTEs. Thus, RTEs do not merely occupy a midpoint between the thymus and the mature T cell pool, but represent a distinct transitional T cell population. Furthermore, RTEs and MN T cells exhibit distinct CpG DNA methylation patterns both before and after activation. Compared with MN T cells, RTEs express higher levels of several enzymes that modify DNA methylation, and inhibiting methylation during culture allows RTEs to reach MN T cell levels of cytokine production. Collectively, these data suggest that the functional differences that distinguish RTEs from MN T cells are influenced by epigenetic mechanisms and provide clues to a mechanistic basis for postthymic maturation.


Asunto(s)
Diferenciación Celular/genética , Citocinas/genética , Metilación de ADN/inmunología , Subgrupos de Linfocitos T/citología , Linfocitos T/citología , Timocitos/citología , Animales , Diferenciación Celular/inmunología , Separación Celular , Citocinas/inmunología , Metilación de ADN/genética , Citometría de Flujo , Interleucina-2/genética , Interleucina-4/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Timocitos/inmunología
12.
J Immunol ; 198(2): 553, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28069748
13.
Proc Natl Acad Sci U S A ; 108(13): 5366-71, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21402911

RESUMEN

Recent thymic emigrants (RTEs) are the youngest subset of peripheral T cells, and they differ functionally and phenotypically from the rest of the naïve T-cell pool. RTEs are present in the peripheral T-cell pool throughout life but are the most common subset of T cells in neonates and adults recovering from lymphoablation. Using a murine model to study the homeostasis of RTEs, we show that under lymphoreplete conditions, RTEs are at a competitive disadvantage to already established mature naïve (MN) T cells. This disadvantage may be caused by a defect in survival, because RTEs may transduce homeostatic signals inefficiently, and their ability to survive is enhanced with increased expression of IL-7 receptor or B-cell lymphoma 2 (Bcl-2). Conversely, under lymphopenic conditions, enhanced proliferation by RTEs allows them to out-compete their MN T-cell counterparts. These results suggest that in times of need, such as in neonates or lymphopenic adults, RTEs perform well to fill the gaps in the peripheral T-cell pool, but when the periphery already is full, many RTEs are not incorporated into the pool of recirculating lymphocytes.


Asunto(s)
Linfocitos T/citología , Linfocitos T/inmunología , Timo/citología , Timo/inmunología , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Interleucina-7/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología
14.
Blood ; 117(4): 1239-49, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21048154

RESUMEN

After intrathymic development, T cells exit the thymus and join the peripheral T-cell pool. Such recent thymic emigrants (RTEs) undergo both phenotypic and functional maturation during the first 3 weeks they reside in the periphery. Using a well-controlled in vitro polarization scheme, we now show that CD4(+) RTEs are defective in T-helper (Th) type 0 (Th0), Th1, Th17, and regulatory T-cell lineage commitment, with dampened cytokine production and transcription factor expression. In contrast, CD4(+) RTES are biased toward the Th2 lineage both in vitro and in vivo, with more robust interleukin-4, interleukin-5, and interleukin-13 production than their mature naive counterparts. Coculture experiments demonstrate that mature naive T cells influence neighboring RTEs in their Th responses. In adoptive hosts, CD4(+) RTEs drive production of the Th2-associated antibody isotype immunoglobulin G1 and mediate airway inflammatory disease. This bias in RTEs likely results from dampened negative regulation of the Th2 lineage by diminished levels of T-bet, a key Th1 transcription factor. CD4(+) RTEs thus represent a transitional population with a distinct interpretation of, and response to, immunologic cues. These characteristics may be beneficial during the postthymic maturation period by leading to the avoidance of inappropriate immune responses, particularly in lymphopenic neonates and adults.


Asunto(s)
Linaje de la Célula/inmunología , Movimiento Celular , Células TH1/fisiología , Células Th2/fisiología , Timo/citología , Envejecimiento/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/fisiología , Diferenciación Celular/inmunología , Linaje de la Célula/fisiología , Movimiento Celular/inmunología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T Colaboradores-Inductores/clasificación , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/fisiología , Células TH1/inmunología , Células Th2/inmunología , Timo/inmunología , Factores de Tiempo
17.
J Immunol ; 186(2): 799-806, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21148799

RESUMEN

Peripheral CD4(+)Vß5(+) T cells are tolerized to an endogenous mouse mammary tumor virus superantigen either by deletion or TCR revision. Through TCR revision, RAG reexpression mediates extrathymic TCRß rearrangement and results in a population of postrevision CD4(+)Vß5(-) T cells expressing revised TCRß chains. We have hypothesized that cell death pathways regulate the selection of cells undergoing TCR revision to ensure the safety and utility of the postrevision population. In this study, we investigate the role of Bcl-2-interacting mediator of cell death (Bim)-mediated cell death in autoantigen-driven deletion and TCR revision. Bim deficiency and Bcl-2 overexpression in Vß5 transgenic (Tg) mice both impair peripheral deletion. Vß5 Tg Bim-deficient and Bcl-2 Tg mice exhibit an elevated frequency of CD4(+) T cells expressing both the transgene-encoded Vß5 chain and a revised TCRß chain. We now show that these dual-TCR-expressing cells are TCR revision intermediates and that the population of RAG-expressing, revising CD4(+) T cells is increased in Bim-deficient Vß5 Tg mice. These findings support a role for Bim and Bcl-2 in regulating the balance of survival versus apoptosis in peripheral T cells undergoing RAG-dependent TCR rearrangements during TCR revision, thereby ensuring the utility of the postrevision repertoire.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Autoantígenos/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Depleción Linfocítica , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Proteínas Proto-Oncogénicas/fisiología , Receptores de Antígenos de Linfocitos T/biosíntesis , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteína 11 Similar a Bcl2 , Linfocitos T CD4-Positivos/virología , Muerte Celular/genética , Muerte Celular/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Reordenamiento Génico de Linfocito T/inmunología , Genes RAG-1/inmunología , Humanos , Virus del Tumor Mamario del Ratón/inmunología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/deficiencia , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética , Transducción de Señal/inmunología
18.
J Immunol ; 186(1): 156-63, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21098226

RESUMEN

We have previously shown that regulatory T cells (Treg) accumulate dramatically in aged animals and negatively impact the ability to control persistent infection. However, the mechanisms underlying the age-dependent accrual of Treg remain unclear. In this study, we show that Treg accumulation with age is progressive and likely not the result of increased thymic output, increased peripheral proliferation, or from enhanced peripheral conversion. Instead, we found that Treg from aged mice are more resistant to apoptosis than Treg from young mice. Although Treg from aged mice had increased expression of functional IL-7Rα, we found that IL-7R signaling was not required for maintenance of Treg in vivo. Notably, aged Treg exhibit decreased expression of the proapoptotic molecule Bim compared with Treg from young mice. Furthermore, in the absence of Bim, Treg accumulate rapidly, accounting for >25% of the CD4(+) T cell compartment by 6 mo of age. Additionally, accumulation of Treg in Bim-deficient mice occurred after the cells left the transitional recent thymic emigrant compartment. Mechanistically, we show that IL-2 drives preferential proliferation and accumulation of Bim(lo) Treg. Collectively, our data suggest that chronic stimulation by IL-2 leads to preferential expansion of Treg having low expression of Bim, which favors their survival and accumulation in aged hosts.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Homeostasis/inmunología , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas/fisiología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Envejecimiento/genética , Envejecimiento/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Técnicas de Sustitución del Gen , Homeostasis/genética , Humanos , Recuento de Linfocitos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Timo/citología , Timo/inmunología
19.
Clin Dev Immunol ; 2013: 781320, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23762096

RESUMEN

One of the challenges faced by the infant immune system is learning to distinguish the myriad of foreign but nonthreatening antigens encountered from those expressed by true pathogens. This balance is reflected in the diminished production of proinflammatory cytokines by both innate and adaptive immune cells in the infant. A downside of this bias is that several factors critical for controlling Mycobacterium tuberculosis infection are significantly restricted in infants, including TNF, IL-1, and IL-12. Furthermore, infant T cells are inherently less capable of differentiating into IFN- γ -producing T cells. As a result, infected infants are 5-10 times more likely than adults to develop active tuberculosis (TB) and have higher rates of severe disseminated disease, including miliary TB and meningitis. Infant TB is a fundamentally different disease than TB in immune competent adults. Immunotherapeutics, therefore, should be specifically evaluated in infants before they are routinely employed to treat TB in this age group. Modalities aimed at reducing inflammation, which may be beneficial for adjunctive therapy of some forms of TB in older children and adults, may be of no benefit or even harmful in infants who manifest much less inflammatory disease.


Asunto(s)
Antituberculosos/uso terapéutico , Inmunidad Innata , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Tuberculosis/fisiopatología , Adulto , Factores de Edad , Antituberculosos/farmacología , Niño , Susceptibilidad a Enfermedades , Humanos , Factores Inmunológicos/farmacología , Factores Inmunológicos/uso terapéutico , Lactante , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-1/biosíntesis , Interleucina-1/inmunología , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/tratamiento farmacológico
20.
Cell Immunol ; 274(1-2): 39-45, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22398309

RESUMEN

Recent thymic emigrants, the youngest T cells in the lymphoid periphery, undergo a 3 week-long period of functional and phenotypic maturation before being incorporated into the pool of mature, naïve T cells. Previous studies indicate that this maturation requires T cell exit from the thymus and access to secondary lymphoid organs, but is MHC-independent. We now show that post-thymic T cell maturation is independent of homeostatic and costimulatory pathways, requiring neither signals delivered by IL-7 nor CD80/86. Furthermore, while CCR7/CCL19,21-regulated homing of recent thymic emigrants to the T cell zones within the secondary lymphoid organs is not required for post-thymic T cell maturation, an intact dendritic cell compartment modulates this process. It is thus clear that, unlike T cell development and homeostasis, post-thymic maturation is focused not on interrogating the T cell receptor or the cell's responsiveness to homeostatic or costimulatory signals, but on some as yet unrecognized property.


Asunto(s)
Células Dendríticas/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Células Dendríticas/inmunología , Homeostasis , Interleucina-7/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores CCR7/metabolismo , Timo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA