Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Immunol ; 14: 1261081, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37868985

RESUMEN

Thymic epithelial cells (TECs) are essential for T cell development in the thymus, yet the mechanisms governing their differentiation are not well understood. Lin28, known for its roles in embryonic development, stem cell pluripotency, and regulating cell proliferation and differentiation, is expressed in endodermal epithelial cells during embryogenesis and persists in adult epithelia, implying postnatal functions. However, the detailed expression and function of Lin28 in TECs remain unknown. In this study, we examined the expression patterns of Lin28 and its target Let-7g in fetal and postnatal TECs and discovered opposing expression patterns during postnatal thymic growth, which correlated with FOXN1 and MHCII expression. Specifically, Lin28b showed high expression in MHCIIhi TECs, whereas Let-7g was expressed in MHCIIlo TECs. Deletion of Lin28a and Lin28b specifically in TECs resulted in reduced MHCII expression and overall TEC numbers. Conversely, overexpression of Lin28a increased total TEC and thymocyte numbers by promoting the proliferation of MHCIIlo TECs. Additionally, our data strongly suggest that Lin28 and Let-7g expression is reliant on FOXN1 to some extent. These findings suggest a critical role for Lin28 in regulating the development and differentiation of TECs by modulating MHCII expression and TEC proliferation throughout thymic ontogeny and involution. Our study provides insights into the mechanisms underlying TEC differentiation and highlights the significance of Lin28 in orchestrating these processes.


Asunto(s)
Células Epiteliales , Timo , Embarazo , Femenino , Humanos , Timo/metabolismo , Células Epiteliales/metabolismo , Timocitos , Epitelio , Diferenciación Celular/genética
2.
Development ; 150(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36975725

RESUMEN

The transcription factor FOXN1 is essential for fetal thymic epithelial cell (TEC) differentiation and proliferation. Postnatally, Foxn1 levels vary widely between TEC subsets, from low/undetectable in putative TEC progenitors to highest in differentiated TEC subsets. Correct Foxn1 expression is required to maintain the postnatal microenvironment; premature downregulation of Foxn1 causes a rapid involution-like phenotype, and transgenic overexpression can cause thymic hyperplasia and/or delayed involution. We investigated a K5.Foxn1 transgene that drives overexpression in mouse TECs, but causes neither hyperplasia nor delay or prevention of aging-related involution. Similarly, this transgene cannot rescue thymus size in Foxn1lacZ/lacZ mice, which undergo premature involution as a result of reduced Foxn1 levels. However, TEC differentiation and cortico-medullary organization are maintained with aging in both K5.Foxn1 and Foxn1lacZ/lacZ mice. Analysis of candidate TEC markers showed co-expression of progenitor and differentiation markers as well as increased proliferation in Plet1+ TECs associated with Foxn1 expression. These results demonstrate that the functions of FOXN1 in promoting TEC proliferation and differentiation are separable and context dependent, and suggest that modulating Foxn1 levels can regulate the balance of proliferation and differentiation in TEC progenitors.


Asunto(s)
Regulación de la Expresión Génica , Timo , Animales , Ratones , Diferenciación Celular/genética , Proliferación Celular/genética , Regulación hacia Abajo , Células Epiteliales/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Ratones Endogámicos C57BL
3.
J Exp Med ; 219(10)2022 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-35997680

RESUMEN

The generation of a functional, self-tolerant T cell receptor (TCR) repertoire depends on interactions between developing thymocytes and antigen-presenting thymic epithelial cells (TECs). Cortical TECs (cTECs) rely on unique antigen-processing machinery to generate self-peptides specialized for T cell positive selection. In our current study, we focus on the lipid kinase Vps34, which has been implicated in autophagy and endocytic vesicle trafficking. We show that loss of Vps34 in TECs causes profound defects in the positive selection of the CD4 T cell lineage but not the CD8 T cell lineage. Utilizing TCR sequencing, we show that T cell selection in conditional mutants causes altered repertoire properties including reduced clonal sharing. cTECs from mutant mice display an increased abundance of invariant chain intermediates bound to surface MHC class II molecules, indicating altered antigen processing. Collectively, these studies identify lipid kinase Vps34 as an important contributor to the repertoire of selecting ligands processed and presented by TECs to developing CD4 T cells.


Asunto(s)
Linfocitos T CD8-positivos , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Activación de Linfocitos , Animales , Antígenos CD8 , Células Epiteliales , Lípidos , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T , Timo
4.
Development ; 147(12)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32467240

RESUMEN

The cortical and medullary thymic epithelial cell (cTEC and mTEC) lineages are essential for inducing T cell lineage commitment, T cell positive selection and the establishment of self-tolerance, but the mechanisms controlling their fetal specification and differentiation are poorly understood. Here, we show that notch signaling is required to specify and expand the mTEC lineage. Notch1 is expressed by and active in TEC progenitors. Deletion of Notch1 in TECs resulted in depletion of mTEC progenitors and dramatic reductions in mTECs during fetal stages, consistent with defects in mTEC specification and progenitor expansion. Conversely, forced notch signaling in all TECs resulted in widespread expression of mTEC progenitor markers and profound defects in TEC differentiation. In addition, lineage-tracing analysis indicated that all mTECs have a history of receiving a notch signal, consistent with notch signaling occurring in mTEC progenitors. These data provide strong evidence for a requirement for notch signaling in specification of the mTEC lineage.


Asunto(s)
Desarrollo Fetal/genética , Receptor Notch1/metabolismo , Timo/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis , Receptor Notch1/deficiencia , Receptor Notch1/genética , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Timo/citología , Timo/crecimiento & desarrollo
5.
PLoS One ; 13(2): e0193188, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29462197

RESUMEN

The postnatal thymus is an efficient microenvironment for T cell specification and differentiation. B cells are also present in the thymus and have been recently shown to impact T cell selection, however, the mechanisms controlling B cell development in the thymus are largely unknown. In Foxn1lacZ mutant mice, down-regulation of Foxn1 expression in thymic epithelial cells beginning 1 week after birth caused a dramatic reduction of T progenitors and an increase of B cell progenitors. This time point is coincident with the switch from fetal to adult-type hematopoietic stem cells (HSCs), which is regulated by the Lin28-Let7 system. We hypothesize that the thymic environment might regulate this process to suppress fetal-type B cell development in the thymus. In this study we show that in the Foxn1lacZ thymus, although the down-regulation of Lin28 in thymocytes was normal, up-regulation of Let-7 was impaired. The failure to up-regulate Let-7 caused a transient increase of Arid3a in B precursors, which is known to promote fetal-type B cell fate. Over-expression of Lin28a in HSCs also reduced Let-7 and promoted Arid3a expression in BM and thymic B progenitors, increasing B cell production in the thymus. The level of Let-7 in thymic B progenitors was up regulated by in vitro co-culture with IL15, Vitamin-D3, and retinoic acid, thus down-regulating Arid3a to promote B cell differentiation. All of these signals were produced in thymic epithelial cells (TECs) related to Let-7 expression in thymic B progenitors, and down-regulated in Foxn1lacZ mutants. Our data show that signals provided by TEC control thymic B cell development by up-regulating Let-7, suppressing Arid3a expression in intrathymic progenitor B cells to limit their proliferation during the neonatal to adult transition.


Asunto(s)
Linfocitos B/metabolismo , Proliferación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , MicroARNs/metabolismo , Células Madre/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Factores de Transcripción/metabolismo , Animales , Linfocitos B/citología , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Ratones , Ratones Transgénicos , MicroARNs/genética , Células Madre/citología , Timocitos/citología , Timo/citología , Factores de Transcripción/genética
6.
PLoS One ; 13(2): e0193189, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29462202

RESUMEN

BACKGROUND: Hematopoietic stem cells (HSCs) derived from birth through adult possess differing differentiation potential for T or B cell fate in the thymus; neonatal bone marrow (BM) cells also have a higher potential for B cell production in BM compared to adult HSCs. We hypothesized that this hematopoietic-intrinsic B potential might also regulate B cell development in the thymus during ontogeny. METHODS: Foxn1lacZ mutant mice are a model in which down regulation of a thymic epithelial cell (TEC) specific transcription factor beginning one week postnatal causes a dramatic reduction of thymocytes production. In this study, we found that while T cells were decreased, the frequency of thymic B cells was greatly increased in these mutants in the perinatal period. We used this model to characterize the mechanisms in the thymus controlling B cell development. RESULTS: Foxn1lacZ mutants, T cell committed intrathymic progenitors (DN1a,b) were progressively reduced beginning one week after birth, while thymic B cells peaked at 3-4 weeks with pre-B-II progenitor phenotype, and originated in the thymus. Heterochronic chimeras showed that the capacity for thymic B cell production was due to a combination of higher B potential of neonatal HSCs, combined with a thymic microenvironment deficiency including reduction of DL4 and increase of IL-7 that promoted B cell fate. CONCLUSION: Our findings indicate that the capacity and time course for thymic B-cell production are primarily controlled by the hematopoietic-intrinsic potential for B cells themselves during ontogeny, but that signals from TECs microenvironment also influence the frequency and differentiation potential of B cell development in the thymus.


Asunto(s)
Linfocitos B/citología , Células Madre Hematopoyéticas/citología , Activación de Linfocitos/fisiología , Timocitos/citología , Timo/citología , Animales , Linfocitos B/metabolismo , Diferenciación Celular/fisiología , Células Epiteliales/citología , Células Madre Hematopoyéticas/metabolismo , Ratones , Timocitos/metabolismo , Timo/metabolismo
7.
Sci Immunol ; 3(19)2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29330161

RESUMEN

The thymus is not only extremely sensitive to damage but also has a remarkable ability to repair itself. However, the mechanisms underlying this endogenous regeneration remain poorly understood, and this capacity diminishes considerably with age. We show that thymic endothelial cells (ECs) comprise a critical pathway of regeneration via their production of bone morphogenetic protein 4 (BMP4) ECs increased their production of BMP4 after thymic damage, and abrogating BMP4 signaling or production by either pharmacologic or genetic inhibition impaired thymic repair. EC-derived BMP4 acted on thymic epithelial cells (TECs) to increase their expression of Foxn1, a key transcription factor involved in TEC development, maintenance, and regeneration, and its downstream targets such as Dll4, a key mediator of thymocyte development and regeneration. These studies demonstrate the importance of the BMP4 pathway in endogenous tissue regeneration and offer a potential clinical approach to enhance T cell immunity.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Células Endoteliales/metabolismo , Regeneración/fisiología , Timo/metabolismo , Timo/fisiología , Animales , Proliferación Celular/fisiología , Células Endoteliales/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Femenino , Factores de Transcripción Forkhead/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Células Madre/metabolismo , Células Madre/fisiología , Linfocitos T/metabolismo , Linfocitos T/fisiología
8.
J Immunol ; 199(8): 2701-2712, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28931604

RESUMEN

Total body irradiation (TBI) damages hematopoietic cells in the bone marrow and thymus; however, the long-term effects of irradiation with aging remain unclear. In this study, we found that the impact of radiation on thymopoiesis in mice varied by sex and dose but, overall, thymopoiesis remained suppressed for ≥12 mo after a single exposure. Male and female mice showed a long-term dose-dependent reduction in thymic cKit+ lymphoid progenitors that was maintained throughout life. Damage to hematopoietic stem cells (HSCs) in the bone marrow was dose dependent, with as little as 0.5 Gy causing a significant long-term reduction. In addition, the potential for T lineage commitment was radiation sensitive with aging. Overall, the impact of irradiation on the hematopoietic lineage was more severe in females. In contrast, the rate of decline in thymic epithelial cell numbers with age was radiation-sensitive only in males, and other characteristics including Ccl25 transcription were unaffected. Taken together, these data suggest that long-term suppression of thymopoiesis after sublethal irradiation was primarily due to fewer progenitors in the BM combined with reduced potential for T lineage commitment. A single irradiation dose also caused synchronization of thymopoiesis, with a periodic thymocyte differentiation profile persisting for at least 12 mo postirradiation. This study suggests that the number and capability of HSCs for T cell production can be dramatically and permanently damaged after a single relatively low TBI dose, accelerating aging-associated thymic involution. Our findings may impact evaluation and therapeutic intervention of human TBI events.


Asunto(s)
Células de la Médula Ósea/fisiología , Hematopoyesis/efectos de la radiación , Síndromes de Inmunodeficiencia/inmunología , Células Progenitoras Linfoides/fisiología , Linfocitos T/fisiología , Timo/efectos de la radiación , Envejecimiento , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Femenino , Síndromes de Inmunodeficiencia/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-kit/metabolismo , Timo/inmunología , Irradiación Corporal Total/efectos adversos
9.
Nat Commun ; 4: 2959, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24343363

RESUMEN

The thoracic thymus is the primary vertebrate organ for T-cell generation. Accessory cervical thymi have also been identified in humans and mice, and shown in mice to be independent functional organs that support T-cell development. However, their origin and functional significance remain unclear. Here we show that cervical thymi in mice have following two origins: delayed differentiation of endodermal precursors and transdifferentiation of parathyroid-fated cells. Compared with thoracic thymus, parathyroid-origin cervical thymi (pCT) express low levels of the thymic epithelial cell-specific transcription factor FOXN1. Consequently, pCT form a distinct microenvironment that supports an atypical thymocyte development pathway, generating T cells with unconventional phenotypic characteristics. Our data demonstrate a transdifferentiation origin for a subset of cervical thymi, with specific functional consequences for T-cell development.


Asunto(s)
Transdiferenciación Celular , Glándulas Paratiroides/patología , Linfocitos T/citología , Timo/patología , Animales , Diferenciación Celular , Linaje de la Célula , Células Epiteliales/citología , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo
10.
Blood ; 113(3): 567-74, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18978204

RESUMEN

The postnatal thymus is the primary source of T cells in vertebrates, and many if not all stages of thymocyte development require interactions with thymic epithelial cells (TECs). The Foxn1 gene is a key regulator of TEC differentiation, and is required for multiple aspects of fetal TEC differentiation. Foxn1 is also expressed in the postnatal thymus, but its function after birth is unknown. We generated a Foxn1 allele with normal fetal expression and thymus development, but decreased expression in the postnatal thymus. This down-regulation causes rapid thymic compartment degeneration and reduced T-cell production. TEC subsets that express higher Foxn1 levels are most sensitive to its down-regulation, in particular MHCII(hi)UEA-1(hi) medullary TECs. The requirement for Foxn1 is extremely dosage sensitive, with small changes in Foxn1 levels having large effects on thymus phenotypes. Our results provide the first evidence that Foxn1 is required to maintain the postnatal thymus. Furthermore, the similarities of this phenotype to accelerated aging-related thymic involution support the possibility that changes in Foxn1 expression in TECs during aging contribute to the mechanism of involution.


Asunto(s)
Células Epiteliales/citología , Factores de Transcripción Forkhead/metabolismo , Timo/citología , Timo/crecimiento & desarrollo , Animales , Células Epiteliales/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Ratones , Ratones Mutantes , Fenotipo , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Timo/inmunología
11.
J Immunol ; 180(2): 914-21, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18178831

RESUMEN

Foxn1Delta is a hypomorphic allele of the nude gene that causes arrested thymic epithelial cell differentiation and abnormal thymic architecture lacking cortical and medullary domains. T cells develop in the Foxn1Delta/Delta adult thymus to the double- and single-positive stages, but in the apparent absence of double-negative 3 (DN3) cells; however, DN3 cells are present in the fetal thymus. To investigate the origin of this seemingly contradictory phenotype, we performed an analysis of fetal and adult DN cells in these mutants. Neither adult bone marrow-derived cells nor fetal liver cells from wild-type or Rag1-/- mice were able to differentiate to the DN2 or DN3 stage in the Foxn1Delta/Delta thymus. Our data suggest that thymopoiesis in the Foxn1Delta/Delta adult thymus proceeds from CD117- atypical progenitors, while CD117+ DN1a cells are absent or blocked in their ability to differentiate to the T lineage. Wild-type cells generated by this pathway in the postnatal thymus were exported to the periphery, demonstrating that these atypical cells contributed to the peripheral T cell pool. The Foxn1Delta/Delta adult (but not fetal) thymus also preferentially supports B cell development, specifically of the B-1 type, and this phenotype correlated with reduced Notch ligand expression in the adult stroma.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Proteínas Proto-Oncogénicas c-kit/análisis , Células Madre/inmunología , Linfocitos T/inmunología , Timo/crecimiento & desarrollo , Animales , Linfocitos B/inmunología , Diferenciación Celular/genética , Factores de Transcripción Forkhead/genética , Eliminación de Gen , Péptidos y Proteínas de Señalización Intracelular , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Mutación , Linfocitos T/citología , Timo/citología , Timo/inmunología
12.
BMC Dev Biol ; 7: 69, 2007 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-17577402

RESUMEN

BACKGROUND: Thymic epithelial cells (TECs) promote thymocyte maturation and are required for the early stages of thymocyte development and for positive selection. However, investigation of the mechanisms by which TECs perform these functions has been inhibited by the lack of genetic tools. Since the Foxn1 gene is expressed in all presumptive TECs from the early stages of thymus organogenesis and broadly in the adult thymus, it is an ideal locus for driving gene expression in differentiating and mature TECs. RESULTS: We generated two knock-in alleles of Foxn1 by inserting IRES-Cre or IRES-lacZ cassettes into the 3' UTR of the Foxn1 locus. We simultaneously electroporated the two targeting vectors to generate the two independent alleles in the same experiment, demonstrating the feasibility of multiplex gene targeting at this locus. Our analysis shows that the knockin alleles drive expression of Cre or lacZ in all TECs in the fetal thymus. Furthermore, the knockin alleles express Cre or lacZ in a Foxn1-like pattern without disrupting Foxn1 function as determined by phenotype analysis of Foxn1 knockin/Foxn1 null compound heterozygotes. CONCLUSION: These data show that multiplex gene targeting into the 3' UTR of the Foxn1 locus is an efficient method to express any gene of interest in TECs from the earliest stage of thymus organogenesis. The resulting alleles will make possible new molecular and genetic studies of TEC differentiation and function. We also discuss evidence indicating that gene targeting into the 3' UTR is a technique that may be broadly applicable for the generation of genetically neutral driver strains.


Asunto(s)
Células Epiteliales/fisiología , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Integrasas/genética , Timo/citología , Timo/embriología , Regiones no Traducidas 3'/genética , Animales , Animales Recién Nacidos , Línea Celular , Operón Lac , Ratones , Ratones Endogámicos C57BL , Timo/crecimiento & desarrollo
13.
J Immunol ; 172(4): 2424-30, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-14764713

RESUMEN

Memory peripheral Th2 immune responses to infectious pathogens are not well studied due to the lack of suitable models and the difficulty of assessing Th2 cytokine expression at sites of inflammation. We have examined the localized immune response to a nematode parasite that encysts in the small intestine. An unexpected architecture was observed on day 4 of the memory response, with granulocytes and macrophages infiltrating the cyst and CD4(+), TCR-alphabeta(+) T cells surrounding the cyst. Laser capture microdissection analysis showed a pronounced CD4-dependent Th2 cytokine pattern at the cyst region only during the memory response, demonstrating that the Th2 memory response is readily distinguished from the primary response by the rapid accumulation of Th2 effector cells at the host:parasite interface.


Asunto(s)
Linfocitos T CD4-Positivos/patología , Movimiento Celular/inmunología , Memoria Inmunológica , Mucosa Intestinal/patología , Yeyuno/patología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Células Th2/patología , Administración Oral , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD4/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/parasitología , Femenino , Regulación de la Expresión Génica/inmunología , Inmunización Secundaria , Inflamación/inmunología , Inflamación/parasitología , Inflamación/patología , Interleucina-13/antagonistas & inhibidores , Interleucina-13/biosíntesis , Interleucina-13/genética , Interleucina-4/antagonistas & inhibidores , Interleucina-4/biosíntesis , Interleucina-4/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Yeyuno/inmunología , Yeyuno/parasitología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/parasitología , Ganglios Linfáticos/patología , Mesenterio/inmunología , Mesenterio/parasitología , Mesenterio/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología , Células Th2/inmunología , Células Th2/parasitología
14.
J Immunol ; 170(1): 384-93, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12496423

RESUMEN

In these studies, we examined the effects of OX40 ligand (OX40L) deficiency on the development of Th2 cells during the Th2 immune response to the intestinal nematode parasite Heligmosomoides polygyrus. Elevations in IL-4 production and total and Ag-specific serum IgE levels were partially inhibited during both the primary and memory immune responses to H. polygyrus in OX40L(-/-) mice. The host-protective memory response was compromised in OX40L(-/-) mice, as decreased worm expulsion and increased egg production were observed compared with H. polygyrus-inoculated OX40L(+/+) mice. To further examine the nature of the IL-4 defect during priming, adoptively transferred DO11.10 T cells were analyzed in the context of the H. polygyrus response. Although Ag-specific T cell IL-4 production was reduced in the OX40L(-/-) mice following immunization with OVA peptide plus H. polygyrus, Ag-specific T cell expansion, cell cycle progression, CXCR5 expression, and migration were comparable between OX40L(+/+) and OX40L(-/-) mice inoculated with OVA and H. polygyrus. These studies suggest an important role for OX40/OX40L interactions in specifically promoting IL-4 production, as well as associated IgE elevations, in Th2 responses to H. polygyrus. However, OX40L interactions were not required for serum IgG1 elevations, increases in germinal center formation, and Ag-specific Th2 cell expansion and migration to the B cell zone.


Asunto(s)
Parasitosis Intestinales/inmunología , Glicoproteínas de Membrana/fisiología , Nematospiroides dubius/inmunología , Receptores del Factor de Necrosis Tumoral , Infecciones por Strongylida/inmunología , Células Th2/inmunología , Células Th2/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos Helmínticos/inmunología , Subgrupos de Linfocitos B/inmunología , Ciclo Celular/genética , Ciclo Celular/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Quimiocinas CXC/metabolismo , Epítopos de Linfocito T/inmunología , Centro Germinal/inmunología , Centro Germinal/patología , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología , Inmunidad Innata/genética , Inmunización Secundaria , Inmunoglobulina E/biosíntesis , Inmunoglobulina E/sangre , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Inyecciones Subcutáneas , Interleucina-4/antagonistas & inhibidores , Interleucina-4/biosíntesis , Parasitosis Intestinales/genética , Ligandos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Activación de Linfocitos/genética , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Cuello , Ligando OX40 , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Receptores CXCR5 , Receptores de Quimiocina , Receptores de Citocinas/biosíntesis , Receptores OX40 , Infecciones por Strongylida/genética , Células Th2/patología , Factores de Necrosis Tumoral , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA