Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Exp Med ; 216(8): 1762-1776, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31201208

RESUMEN

Recently, human ILCs that express CD117 and CD127 but lack CRTH2 and NKp44 have been shown to contain precursors of ILC1, ILC2, and ILC3. However, these ILCs have not been extensively characterized. We performed an unbiased hierarchical stochastic neighbor embedding (HSNE) analysis of the phenotype of peripheral blood CD117+ ILCs, which revealed the presence of three major subsets: the first expressed NKp46, the second expressed both NKp46 and CD56, and the third expressed KLRG1, but not NKp46 or CD56. Analysis of their cytokine production profiles and transcriptome revealed that NKp46+ ILCs predominantly develop into ILC3s; some of them can differentiate into ILC1/NK-like cells, but they are unable to develop into ILC2s. In contrast, KLRG1+ ILCs predominantly differentiate into ILC2s. Single-cell cultures demonstrate that KLRG1+ ILCs can also differentiate into other ILC subsets depending on the signals they receive. Epigenetic profiling of KLRG1+ ILCs is consistent with the broad differentiation potential of these cells.


Asunto(s)
Diferenciación Celular/inmunología , Células Asesinas Naturales/metabolismo , Lectinas Tipo C/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Animales , Donantes de Sangre , Línea Celular , Citocinas/metabolismo , Epigénesis Genética , Humanos , Inmunidad Innata , Células Asesinas Naturales/inmunología , Ratones , Receptor 2 Gatillante de la Citotoxidad Natural/metabolismo , Tonsila Palatina/patología , Fenotipo , Transcriptoma
2.
Nat Commun ; 10(1): 2162, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31089134

RESUMEN

Innate lymphoid cells (ILCs) are crucial for the immune surveillance at mucosal sites. ILCs coordinate early eradication of pathogens and contribute to tissue healing and remodeling, features that are dysfunctional in patients with cystic fibrosis (CF). The mechanisms by which ILCs contribute to CF-immunopathology are ill-defined. Here, we show that group 2 ILCs (ILC2s) transdifferentiated into IL-17-secreting cells in the presence of the epithelial-derived cytokines IL-1ß, IL-23 and TGF-ß. This conversion is abrogated by IL-4 or vitamin D3. IL-17 producing ILC2s induce IL-8 secretion by epithelial cells and their presence in nasal polyps of CF patients is associated with neutrophilia. Our data suggest that ILC2s undergo transdifferentiation in CF nasal polyps in response to local cytokines, which are induced by infectious agents.


Asunto(s)
Plasticidad de la Célula/inmunología , Fibrosis Quística/inmunología , Inflamación/inmunología , Pólipos Nasales/inmunología , Células Th17/inmunología , Adulto , Animales , Línea Celular , Fibrosis Quística/sangre , Fibrosis Quística/patología , Femenino , Humanos , Inmunidad Innata , Inflamación/sangre , Inflamación/patología , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Interleucina-23/inmunología , Interleucina-23/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Mucosa Nasal/citología , Mucosa Nasal/inmunología , Mucosa Nasal/patología , Pólipos Nasales/sangre , Pólipos Nasales/patología , Neutrófilos/inmunología , Adulto Joven
3.
Curr Protoc Immunol ; 122(1): e55, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29957859

RESUMEN

Innate lymphoid cells (ILCs) are innate immune cells of lymphoid origin that have important effector and regulatory functions in the first line of defense against pathogens, but also regulate tissue homeostasis, remodeling, and repair. Their function mirrors T helper cells and cytotoxic CD8+ T lymphocytes, but they lack expression of rearranged antigen-specific receptors. Distinct ILC subsets are classified in group 1 ILCs (ILC1s), group 2 ILCs (ILC2s), and group 3 ILCs (ILC3s and lymphoid tissue-inducer cells), based on the expression of transcription factors and the cytokines they produce. As the frequency of ILCs is low, their isolation requires extensive depletion of other cell types. The lack of unique cell surface antigens further complicates the identification of these cells. Here, methods for ILC isolation and characterization from human peripheral blood and different tissues are described. © 2018 by John Wiley & Sons, Inc.

4.
Artículo en Inglés | MEDLINE | ID: mdl-29229782

RESUMEN

Innate lymphoid cells (ILCs) have emerged as an expanding family of effector cells particularly enriched in the mucosal barriers. ILCs are promptly activated by stress signals and multiple epithelial- and myeloid-cell-derived cytokines. In response, ILCs rapidly secrete effector cytokines, which allow them to survey and maintain the mucosal integrity. Uncontrolled action of ILCs might contribute to tissue damage, chronic inflammation, metabolic diseases, autoimmunity, and cancer. Here we discuss the recent advances in our understanding of the cytokine network that modulate ILC immune responses: stimulating cytokines, signature cytokines secreted by ILC subsets, autocrine cytokines, and cytokines that induce cell plasticity.


Asunto(s)
Citocinas/metabolismo , Homeostasis/fisiología , Inmunidad Innata/fisiología , Linfocitos/fisiología , Humanos
5.
Front Immunol ; 8: 1047, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28912776

RESUMEN

Innate lymphoid cells (ILCs) have emerged as a key cell type involved in surveillance and maintenance of mucosal tissues. Mouse ILCs rely on the transcriptional regulator Inhibitor of DNA-binding protein 2 (Id2) for their development. Here, we show that Id2 also drives development of human ILC because forced expression of Id2 in human thymic progenitors blocked T cell commitment, upregulated CD161 and promyelocytic leukemia zinc finger (PLZF), and maintained CD127 expression, markers that are characteristic for human ILCs. Surprisingly CD5 was also expressed on these in vitro generated ILCs. This was not an in vitro artifact because CD5 was also found on ex vivo isolated ILCs from thymus and from umbilical cord blood. CD5 was also expressed on small proportions of ILC2 and ILC3. CD5+ ILCs were functionally immature, but could further differentiate into mature CD5- cytokine-secreting ILCs. Our data show that Id2 governs human ILC development from thymic progenitor cells toward immature CD5+ ILCs.

6.
J Immunol ; 198(6): 2310-2319, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28159903

RESUMEN

Although CD31 expression on human thymocytes has been reported, a detailed analysis of CD31 expression at various stages of T cell development in the human thymus is missing. In this study, we provide a global picture of the evolution of CD31 expression from the CD34+ hematopoietic precursor to the CD45RA+ mature CD4+ and CD8+ single-positive (SP) T cells. Using nine-color flow cytometry, we show that CD31 is highly expressed on CD34+ progenitors and stays high until the early double-positive stage (CD3-CD4+CD8α+ß-). After ß-selection, CD31 expression levels become low to undetectable. CD31 expression then increases and peaks on CD3highCD4+CD8+ double-positive thymocytes. However, following positive selection, CD31 expression differs dramatically between CD4+ and CD8+ lineages: homogeneously high on CD8 SP but lower or negative on CD4 SP cells, including a subset of CD45RA+CD31- mature CD4+ thymocytes. CD31 expression on TCRγδ thymocytes is very similar to that of CD4 SP cells. Remarkably, there is a substantial subset of semimature (CD45RA-) CD4 SP thymocytes that lack CD31 expression. Moreover, FOXP3+ and ICOS+ cells are overrepresented in this CD31- subpopulation. Despite this CD31-CD45RA- subpopulation, most egress-capable mature CD45RA+ CD4 SP thymocytes express CD31. The variations in CD31 expression appear to coincide with three major selection processes occurring during thymopoiesis: ß-selection, positive selection, and negative selection. Considering the ability of CD31 to modulate the TCR's activation threshold via the recruitment of tyrosine phosphatases, our results suggest a significant role for CD31 during T cell development.


Asunto(s)
Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Timo/inmunología , Antígenos CD34/metabolismo , Diferenciación Celular , Células Cultivadas , Supresión Clonal , Selección Clonal Mediada por Antígenos , Factores de Transcripción Forkhead/metabolismo , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo
7.
Nat Immunol ; 17(6): 636-45, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27111145

RESUMEN

Group 2 innate lymphoid cells (ILC2s) secrete type 2 cytokines, which protect against parasites but can also contribute to a variety of inflammatory airway diseases. We report here that interleukin 1ß (IL-1ß) directly activated human ILC2s and that IL-12 induced the conversion of these activated ILC2s into interferon-γ (IFN-γ)-producing ILC1s, which was reversed by IL-4. The plasticity of ILCs was manifested in diseased tissues of patients with severe chronic obstructive pulmonary disease (COPD) or chronic rhinosinusitis with nasal polyps (CRSwNP), which displayed IL-12 or IL-4 signatures and the accumulation of ILC1s or ILC2s, respectively. Eosinophils were a major cellular source of IL-4, which revealed cross-talk between IL-5-producing ILC2s and IL-4-producing eosinophils. We propose that IL-12 and IL-4 govern ILC2 functional identity and that their imbalance results in the perpetuation of type 1 or type 2 inflammation.


Asunto(s)
Plasticidad de la Célula , Eosinófilos/inmunología , Inmunidad Innata , Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Interleucina-4/metabolismo , Linfocitos/inmunología , Pólipos Nasales/inmunología , Neumonía/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Rinitis/inmunología , Sinusitis/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Femenino , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones SCID , Células TH1/inmunología , Balance Th1 - Th2 , Células Th2/inmunología
8.
Immunol Cell Biol ; 90(8): 802-11, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22491065

RESUMEN

Interleukin (IL)-21-producing CD4(+)T cells are central to humoral immunity. Deciphering the signals that induce IL-21 production in CD4(+) T cells and those triggered by IL-21 in B cells are, therefore, of importance for understanding the generation of antibody (Ab) responses. Here, we show that IL-6 increased IL-21 production by human CD4(+) T cells, particularly in those that express the transcriptional regulator B cell lymphoma (BCL)6, which is required in mice for the development of C-X-C chemokine receptor type 5 (CXCR5(+)) IL-21-producing T follicular helper (T(FH)) cells. However, retroviral overexpression of BCL6 in total human CD4(+) T cells only transiently increased CXCR5, the canonical T(FH)-defining surface marker. We show here that IL-21 was required for the induction of Ab production by IL-6. In IL-21-treated B cells, signal transducer and activator of transcription (STAT)3 was required for optimal immunoglobulin production and upregulation of PR domain containing 1 (PRDM1(+)), the master plasma cell factor. These results, therefore, demonstrate the critical importance of STAT3 activation in B cells during IL-21-driven humoral immunity and suggest that BCL6 expression, although not sufficient, may serve as a platform for the acquisition of a T(FH)-like phenotype by human CD4(+) T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Interleucina-6/farmacología , Interleucinas/biosíntesis , Células Plasmáticas/citología , Factor de Transcripción STAT3/metabolismo , Animales , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Humanos , Inmunoglobulinas/biosíntesis , Ratones , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/inmunología , Proteínas Proto-Oncogénicas c-bcl-6
9.
Blood ; 119(22): 5191-200, 2012 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-22510878

RESUMEN

Plasmacytoid dendritic cells (pDCs) selectively express Toll-like receptor (TLR)-7 and TLR-9, which allow them to rapidly secrete massive amounts of type I interferons after sensing nucleic acids derived from viruses or bacteria. It is not completely understood how development and function of pDCs are controlled at the transcriptional level. One of the main factors driving pDC development is the ETS factor Spi-B, but little is known about its target genes. Here we demonstrate that Spi-B is crucial for the differentiation of hematopoietic progenitor cells into pDCs by controlling survival of pDCs and its progenitors. In search for Spi-B target genes, we identified the antiapoptotic gene Bcl2-A1 as a specific and direct target gene, thereby consolidating the critical role of Spi-B in cell survival.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Plasmáticas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factores de Transcripción/metabolismo , Supervivencia Celular/fisiología , Células Cultivadas , Preescolar , Proteínas de Unión al ADN/genética , Células Dendríticas/citología , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Lactante , Masculino , Antígenos de Histocompatibilidad Menor , Células Plasmáticas/citología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Factores de Transcripción/genética
10.
Proc Natl Acad Sci U S A ; 108(32): 13224-9, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21788504

RESUMEN

The homeostatic control mechanisms regulating human leukocyte numbers are poorly understood. Here, we assessed the role of phagocytes in this process using human immune system (HIS) BALB/c Rag2(-/-)IL-2Rγc(-/-) mice in which human leukocytes are generated from transplanted hematopoietic progenitor cells. Interactions between signal regulatory protein alpha (SIRPα; expressed on phagocytes) and CD47 (expressed on hematopoietic cells) negatively regulate phagocyte activity of macrophages and other phagocytic cells. We previously showed that B cells develop and survive robustly in HIS mice, whereas T and natural killer (NK) cells survive poorly. Because human CD47 does not interact with BALB/c mouse SIRPα, we introduced functional CD47/SIRPα interactions in HIS mice by transducing mouse CD47 into human progenitor cells. Here, we show that this procedure resulted in a dramatic and selective improvement of progenitor cell engraftment and human T- and NK-cell homeostasis in HIS mouse peripheral lymphoid organs. The amount of engrafted human B cells also increased but much less than that of T and NK cells, and total plasma IgM and IgG concentrations increased 68- and 35-fold, respectively. Whereas T cells exhibit an activated/memory phenotype in the absence of functional CD47/SIRPα interactions, human T cells accumulated as CD4(+) or CD8(+) single-positive, naive, resting T cells in the presence of functional CD47/SIRPα interactions. Thus, in addition to signals mediated by T cell receptor (TCR)/MHC and/or IL/IL receptor interactions, sensing of cell surface CD47 expression by phagocyte SIRPα is a critical determinant of T- and NK-cell homeostasis under steady-state conditions in vivo.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Antígeno CD47/metabolismo , Homeostasis , Células Asesinas Naturales/metabolismo , Receptores Inmunológicos/metabolismo , Linfocitos T/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Células Asesinas Naturales/citología , Cinética , Linfopoyesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Unión Proteica , Receptores de Interleucina-2/deficiencia , Receptores de Interleucina-2/metabolismo , Bazo/citología , Bazo/inmunología , Análisis de Supervivencia , Linfocitos T/citología , Timo/metabolismo , Trasplante Heterólogo
11.
J Immunol ; 184(12): 6670-9, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20483740

RESUMEN

The cytokine IL-15 and the inhibitor of DNA binding (Id)2, which negatively regulates the activity of basic helix-loop-helix transcription factors, have been shown to play key roles in NK cell development. Consistent with this, exogenous IL-15 added to human thymic progenitor cells stimulated their development into NK cells at the expense of T cells both in fetal thymic organ culture and in coculture with stromal cells expressing the Notch ligand Delta-like 1. Overexpression of Id2 in thymic progenitor cells stimulated NK cell development and blocked T cell development. This, in part, is attributed to inhibition of the transcriptional activity of the E protein HEB, which we show in this study is the only E protein that enhanced T cell development. Notably, Id2 increased a pool of lineage CD1a-CD5+ progenitor cells that in synergy with IL-15 furthered expansion and differentiation into NK cells. Taken together, our findings point to a dualistic function of Id2 in controlling T/NK cell lineage decisions; T cell development is impaired by Id2, most likely by sequestering HEB, whereas NK cell development is promoted by increasing a pool of CD1a-CD5+ NK cell progenitors, which together with IL-15 differentiate into mature NK cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Diferenciación Celular/inmunología , Células Madre Hematopoyéticas/inmunología , Proteína 2 Inhibidora de la Diferenciación/inmunología , Interleucina-15/inmunología , Células Asesinas Naturales/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linaje de la Célula/inmunología , Separación Celular , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Humanos , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Interleucina-15/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Eur J Immunol ; 40(4): 955-65, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20127673

RESUMEN

Thymic stromal lymphopoietin (TSLP) is a cytokine that binds the IL-7-receptor-alpha chain and a unique TSLP receptor (TSLPR) chain. The role of TSLP in human B-cell development has not been elucidated. We show that TSLPR transcripts are expressed most prominently in CD34(+) cells from fetal liver and BM. In general, cell surface expression of TSLPR was low, except on a subset of multilineage-commited progenitor cells. TSLP induced the tyrosine-phosphorylation of STAT5 and the proliferation of multilineage-commited progenitor cells, pro-B cells and pre-B cells. Compared with IL-7, the levels of proliferation after stimulation of the B-cell progenitors with TSLP were lower. Expression of the BCR on the cell surface of fetal cells was inversely correlated to TSLP or IL-7 responsiveness. Pre-B cells from fetal BM, but not fetal liver, were refractory to TSLP or IL-7 stimulation. When employing an in vitro B-cell differentiation culture system starting from CD34(+)CD38(-) multipotent HSC, IL-7 induced a short wave of precursor cell expansion but did not result in long-term survival of mature B cells. TSLP was capable of increasing the proportion and the absolute numbers of more mature human B cells. Overall, we provide evidence that TSLP supports human B-cell differentiation from fetal hematopoietic progenitors.


Asunto(s)
Linfocitos B/citología , Citocinas/fisiología , Células Madre Multipotentes/citología , Células Precursoras de Linfocitos B/citología , Médula Ósea/embriología , División Celular/efectos de los fármacos , Linaje de la Célula , Células Cultivadas/citología , Células Cultivadas/efectos de los fármacos , Técnicas de Cocultivo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-7/farmacología , Hígado/citología , Hígado/embriología , Linfopoyesis/efectos de los fármacos , Células Madre Multipotentes/efectos de los fármacos , Fosforilación , Células Precursoras de Linfocitos B/efectos de los fármacos , Procesamiento Proteico-Postraduccional , Proteínas Tirosina Quinasas/metabolismo , Receptores de Citocinas/biosíntesis , Receptores de Citocinas/genética , Receptores de Citocinas/fisiología , Receptores de Interleucina-7/biosíntesis , Receptores de Interleucina-7/genética , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT5/metabolismo , Linfopoyetina del Estroma Tímico
13.
Methods Mol Biol ; 595: 67-85, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19941106

RESUMEN

Our understanding of human lymphocyte development has increased significantly over the past 20 years. In particular, our insight into human T- and B-cell development has improved (1, 2). Nonetheless, there are many gaps in our understanding, particularly regarding the early stages of development of hematopoietic progenitor cells (HPCs) into downstream lineage-biased and lineage-restricted precursors and the molecular mechanisms underlying these activities. The same holds true for our knowledge of human dendritic cell (DC) development. While the amount of data on the different subsets of conventional DCs (cDCs) and plasmacytoid DCs (pDCs) rapidly increases in mice (3, 4), the developmental stages of different DC subsets in humans remain poorly defined (2). The relatively easy access to patient material and therefore human precursor cells that can be isolated from these tissues combined with the availability of in vitro and in vivo differentiation assays allows studies in the field of human hematopoietic development, including that of DCs. In addition, the opportunities to manipulate gene expression, by stable overexpression of a gene of interest or RNA interference-mediated knockdown, generate valuable information about the mechanisms underlying lineage commitment and differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Células Dendríticas/citología , Animales , Antígenos CD34/metabolismo , Células Dendríticas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-3/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Ratones
14.
Methods Mol Biol ; 595: 87-115, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19941107

RESUMEN

Over the last two decades, several humanized mouse models have been used to experimentally analyze the function and development of the human immune system. Recent advances have lead to the establishment of new murine-human chimeric models with improved characteristics, both in terms of human engraftment efficiency and in situ multilineage human hematopoietic development. We describe here the use of newborn BALB/c Rag2(-/-)gamma(c) (-/-) mice as recipients of human hematopoietic progenitor cells to produce "human immune system" (HIS) (BALB-Rag/gamma) mice, using human fetal liver progenitors. The two major subsets of the human dendritic cell lineage, namely, BDCA2(+)CD11c(-) plasmacytoid dendritic cells and BDCA2(-)CD11c(+) conventional dendritic cells, can be found in HIS (BALB-Rag/gamma) mice. In order to manipulate the expression of genes of interest, the human hematopoietic progenitor cells can be genetically engineered ex vivo by lentiviral transduction before performing xenograft transplantation. Using this mouse model, the human immune system can be assessed for both fundamental and pre-clinical purposes.


Asunto(s)
Proteínas de Unión al ADN/genética , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Transducción Genética/métodos , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Animales Recién Nacidos , Antígenos CD34/metabolismo , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/inmunología , Citometría de Flujo , Humanos , Lentivirus/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
15.
J Immunol ; 183(12): 7645-55, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19923447

RESUMEN

IL-7 is a central cytokine in the development of hematopoietic cells, although interspecies discrepancies have been reported. By coculturing human postnatal thymus hematopoietic progenitors and OP9-huDL1 stromal cells, we found that murine IL-7 is approximately 100-fold less potent than human IL-7 for supporting human T cell development in vitro. We investigated the role of human IL-7 in newborn BALB/c Rag2(-/-)gamma(c)(-/-) mice transplanted with human hematopoietic stem cells (HSC) as an in vivo model of human hematopoiesis using three approaches to improve IL-7 signaling: administration of human IL-7, ectopic expression of human IL-7 by the transplanted human HSC, or enforced expression of a murine/human chimeric IL-7 receptor binding murine IL-7. We show that premature IL-7 signaling at the HSC stage, before entrance in the thymus, impeded T cell development, whereas increased intrathymic IL-7 signaling significantly enhanced the maintenance of immature thymocytes. Increased thymopoiesis was also observed when we transplanted BCL-2- or BCL-x(L)-transduced human HSC. Homeostasis of peripheral mature T cells in this humanized mouse model was not improved by any of these strategies. Overall, our results provide evidence for an important role of IL-7 in human T cell development in vivo and highlight the notion that IL-7 availability is but one of many signals that condition peripheral T cell homeostasis.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proteínas de Unión al ADN/genética , Homeostasis/inmunología , Subunidad gamma Común de Receptores de Interleucina/genética , Interleucina-7/fisiología , Proteínas Nucleares/genética , Subgrupos de Linfocitos T/inmunología , Timo/inmunología , Animales , Células Cultivadas , Técnicas de Cocultivo , Proteínas de Unión al ADN/deficiencia , Homeostasis/genética , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Mutantes Quiméricas/deficiencia , Proteínas Mutantes Quiméricas/genética , Proteínas Nucleares/deficiencia , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Timo/citología , Timo/metabolismo
16.
J Exp Med ; 205(9): 2033-42, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18695003

RESUMEN

IgM(+)IgD(+)CD27(+) B cells from peripheral blood have been described as circulating marginal zone B cells. It is still unknown when and where these cells develop. These IgM(+)IgD(+)CD27(+) B cells exhibit somatic hypermutations (SHMs) in their B cell receptors, but the exact nature of the signals leading to induction of these SHMs remains elusive. Here, we show that IgM(+)IgD(+)CD27(+) B cells carrying SHMs are observed during human fetal development. To examine the role of T cells in human IgM(+)IgD(+)CD27(+) B cell development we used an in vivo model in which Rag2(-/-)gamma(C)(-/-) mice were repopulated with human hematopoietic stem cells. Using Rag2(-/-)gamma(C)(-/-) mice on a Nude background, we demonstrated that development and induction of SHMs of human IgM(+)IgD(+)CD27(+) B cells can occur in a T cell-independent manner.


Asunto(s)
Linfocitos B/metabolismo , Inmunoglobulina D/metabolismo , Inmunoglobulina M/metabolismo , Mutación , Linfocitos T/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Animales , Sangre Fetal/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Transducción de Señal
17.
Blood ; 112(5): 1804-12, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18552212

RESUMEN

The terminal differentiation of B cells into antibody-secreting plasma cells is tightly regulated by a complex network of transcription factors. Here we evaluated the role of the Ets factor Spi-B during terminal differentiation of human B cells. All mature tonsil and peripheral blood B-cell subsets expressed Spi-B, with the exception of plasma cells. Overexpression of Spi-B in CD19(+) B cells inhibited, similar to the known inhibitor BCL-6, the expression of plasma cell-associated surface markers and transcription factors as well as immunoglobulin production, ie, in vitro plasma cell differentiation. The arrest in B-cell differentiation enforced by Spi-B was independent of the transactivation domain, but dependent on the Ets-domain. By chromatin immunoprecipitation and assays using an inducible Spi-B construct BLIMP1 and XBP-1 were identified as direct target genes of Spi-B mediated repression. We propose a novel role for Spi-B in maintenance of germinal center and memory B cells by direct repression of major plasma cell factors and thereby plasma cell differentiation.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Células Plasmáticas/citología , Células Plasmáticas/fisiología , Proteínas Proto-Oncogénicas c-ets/fisiología , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/fisiología , Sitios de Unión/genética , Diferenciación Celular/fisiología , Proteínas de Unión al ADN/genética , Expresión Génica , Humanos , Inmunoglobulinas/biosíntesis , Técnicas In Vitro , Células Plasmáticas/inmunología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-ets/química , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Transcripción del Factor Regulador X , Proteínas Represoras/genética , Factores de Transcripción/genética , Transducción Genética , Proteína 1 de Unión a la X-Box
18.
J Immunol ; 180(7): 4805-15, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18354204

RESUMEN

STAT family members have been implicated in regulating the balance between B cell lymphoma (BCL)6 and B lymphocyte induced maturation protein (BLIMP)1 to control plasma cell differentiation. We previously showed that STAT5 induces BCL6 to block plasma cell differentiation and extend the life span of human B cells. The heterogeneity in STAT activation by cytokines and their effects on B cell differentiation prompted us to investigate the effect of STAT3 activation in plasma cell differentiation. First stimulation with IL-21, which promotes plasma cell differentiation, induced robust and prolonged STAT3 activation in primary human B cells. We then investigated effects of direct STAT3 activation on regulation of plasma cell genes, cellular phenotype, and Ig production. Activation of a tamoxifen-regulated STAT3-estrogen receptor fusion protein triggered BLIMP1 mRNA and protein up-regulation, plasma cell phenotypic features, and Ig secretion. When STAT3 was activated by IL-21 in B cells ectopically expressing BCL6, BLIMP1 was up-regulated, but only partial plasma cell differentiation was achieved. Lastly, through coexpression of BCL6 and STAT3-ER, we verified that STAT3 activation functionally mimicked IL-21 treatment and that STAT3-mediated BLIMP1 up-regulation occurred despite high BCL6 expression levels indicating that BCL6 is not the dominant repressor of BLIMP1. Thus, up-regulation of BLIMP1 alone is not sufficient for differentiation of primary human B cells into plasma cells; concomitant down-regulation of BCL6 is absolutely required for completion of the plasma cell differentiation program.


Asunto(s)
Diferenciación Celular , Regulación hacia Abajo , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Proteínas Represoras/metabolismo , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Humanos , Inmunoglobulinas/inmunología , Interleucinas/farmacología , Imitación Molecular , Fenotipo , Fosforilación/efectos de los fármacos , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/inmunología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Proto-Oncogénicas c-bcl-6 , Factor de Transcripción STAT3/genética
19.
J Immunol ; 179(8): 4959-68, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17911580

RESUMEN

In vitro production of human T cells with known Ag specificity is of major clinical interest for immunotherapy against tumors and infections. We have performed TCRalphabeta gene transfer into human hemopoietic progenitors from postnatal thymus or umbilical cord blood, and subsequently cultured these precursors on OP9 stromal cells expressing the Notch human ligand Delta-like1. We report here that fully mature, functional T cells with controlled Ag specificity are obtained from such cultures. Using vectors encoding TCRalphabeta-chains directed against melanoma (MART-1), viral (CMV), and minor histocompatibility (HA-2) Ags, we show that the obtained Ag-specific T cells exert cytolytic activity against their cognate Ag and expand in vitro upon specific TCR stimulation. Therapeutic applications may arise from these results because they provide a way to produce large numbers of autologous mature Ag-specific T cells in vitro from undifferentiated hemopoietic progenitors.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/inmunología , Epítopos de Linfocito T/fisiología , Células Madre Hematopoyéticas/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Retroviridae/genética , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Línea Celular , Preescolar , Técnicas de Cocultivo , Técnicas de Transferencia de Gen , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Subgrupos de Linfocitos T/metabolismo , Transducción Genética
20.
Nat Immunol ; 6(3): 303-13, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15711548

RESUMEN

It is unknown how B cells that mature during a germinal center reaction 'decide' between plasma or memory cell fate. Here we describe a previously unknown subpopulation of B cells in the human germinal center that is characterized by tyrosine phosphorylated transcriptional activator STAT5. These cells had an activated centrocyte phenotype and had abundant expression of BCL6 but low expression of PRDM1, both encoding transcriptional repression proteins. Using RNA interference and ectopic expression of constitutively activated forms of STAT5, we demonstrate here a function for STAT5 in the self-renewal of B cells in vitro. STAT5b isoform seemed to directly upregulate Bcl-6, and ectopic expression of Bcl-6 in B cells resulted in self-renewal and inhibition of plasma cell differentiation. These data indicate that activation of STAT5 is involved in regulation of memory B cell differentiation.


Asunto(s)
Linfocitos B/inmunología , Proteínas de Unión al ADN/metabolismo , Memoria Inmunológica , Proteínas de la Leche/metabolismo , Transactivadores/metabolismo , Linfocitos B/citología , Diferenciación Celular , Proteínas de Unión al ADN/efectos de los fármacos , Humanos , Proteínas de la Leche/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-6 , Factor de Transcripción STAT5 , Tamoxifeno/farmacología , Transactivadores/efectos de los fármacos , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA