Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 387
Filtrar
1.
Front Immunol ; 12: 680442, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956167

RESUMEN

Interleukin-7 (IL-7) is produced by stromal cells, keratinocytes, and epithelial cells in host tissues or tumors and exerts a wide range of immune effects mediated by the IL-7 receptor (IL-7R). IL-7 is primarily involved in regulating the development of B cells, T cells, natural killer cells, and dendritic cells via the JAK-STAT, PI3K-Akt, and MAPK pathways. This cytokine participates in the early generation of lymphocyte subsets and maintain the survival of all lymphocyte subsets; in particular, IL-7 is essential for orchestrating the rearrangement of immunoglobulin genes and T-cell receptor genes in precursor B and T cells, respectively. In addition, IL-7 can aid the activation of immune cells in anti-virus and anti-tumor immunity and plays important roles in the restoration of immune function. These biological functions of IL-7 make it an important molecular adjuvant to improve vaccine efficacy as it can promote and extend systemic immune responses against pathogens by prolonging lymphocyte survival, enhancing effector cell activity, and increasing antigen-specific memory cell production. This review focuses on the biological function and mechanism of IL-7 and summarizes its contribution towards improved vaccine efficacy. We hope to provide a thorough overview of this cytokine and provide strategies for the development of the future vaccines.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Inmunogenicidad Vacunal/fisiología , Inmunomodulación/fisiología , Interleucina-7/fisiología , Desarrollo de Vacunas , Animales , Citocinas/fisiología , Células Dendríticas/citología , Células Dendríticas/inmunología , Humanos , Inmunidad Mucosa , Memoria Inmunológica , Interleucina-7/administración & dosificación , Interleucina-7/deficiencia , Interleucina-7/farmacología , Interleucina-7/uso terapéutico , Linfocitos Intraepiteliales/inmunología , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/inmunología , Ratones , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Transducción de Señal
2.
Genes Dev ; 34(21-22): 1503-1519, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33004416

RESUMEN

EBF1 and PAX5 mutations are associated with the development of B progenitor acute lymphoblastic leukemia (B-ALL) in humans. To understand the molecular networks driving leukemia in the Ebf1+/-Pax5+/- (dHet) mouse model for B-ALL, we interrogated the transcriptional profiles and chromatin status of leukemic cells, preleukemic dHet pro-B, and wild-type pro-B cells with the corresponding EBF1 and Pax5 cistromes. In dHet B-ALL cells, many EBF1 and Pax5 target genes encoding pre-BCR signaling components and transcription factors were down-regulated, whereas Myc and genes downstream from IL-7 signaling or associated with the folate pathway were up-regulated. We show that blockade of IL-7 signaling in vivo and methotrexate treatment of leukemic cells in vitro attenuate the expansion of leukemic cells. Single-cell RNA-sequencing revealed heterogeneity of leukemic cells and identified a subset of wild-type pro-B cells with reduced Ebf1 and enhanced Myc expression that show hallmarks of dHet B-ALL cells. Thus, EBF1 and Pax5 may safeguard early stage B cells from transformation to B-ALL by limiting IL-7 signaling, folate metabolism and Myc expression.


Asunto(s)
Ácido Fólico/metabolismo , Interleucina-7/fisiología , Factor de Transcripción PAX5/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/fisiopatología , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal/genética , Transactivadores/metabolismo , Animales , Carbono/metabolismo , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Ratones , Factor de Transcripción PAX5/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Células Precursoras de Linfocitos B/patología , Unión Proteica , Análisis de la Célula Individual , Transactivadores/genética
3.
Crit Care ; 23(1): 131, 2019 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-30995946

RESUMEN

BACKGROUND: Sepsis is the leading cause of mortality for critically ill patients worldwide. Patients develop T lymphocyte dysfunctions leading to T-cell exhaustion associated with increased risk of death. As interleukin-7 (IL-7) is currently tested in clinical trials to reverse these dysfunctions, it is important to evaluate the expression of its specific CD127 receptor on the T-cell surface of patients with septic shock. Moreover, the CD127lowPD-1high phenotype has been proposed as a T-cell exhaustion marker in chronic viral infections but has never been evaluated in sepsis. The objective of this study was first to evaluate CD127 and CD127lowPD-1high phenotype in septic shock in parallel with functional T-cell alterations. Second, we aimed to reproduce septic shock-induced T-cell alterations in an ex vivo model. METHODS: CD127 expression was followed at the protein and mRNA levels in patients with septic shock and healthy volunteers. CD127lowPD-1high phenotype was also evaluated in parallel with T-cell functional alterations after ex vivo activation. To reproduce T-cell alterations observed in patients, purified T cells from healthy volunteers were activated ex vivo and their phenotype and function were evaluated. RESULTS: In patients, neither CD127 expression nor its corresponding mRNA transcript level was modified compared with normal values. However, the percentage of CD127lowPD-1high T cells was increased while T cells also presented functional alterations. CD127lowPD-1high T cells co-expressed HLA-DR, an activation marker, suggesting a role for T-cell activation in the development of this phenotype. Indeed, T-cell receptor (TCR) activation of normal T lymphocytes ex vivo reproduced the increase of CD127lowPD-1high T cells and functional alterations following a second stimulation, as observed in patients. Finally, in this model, as observed in patients, IL-7 could improve T-cell proliferation. CONCLUSIONS: The proportion of CD127lowPD-1high T cells in patients was increased compared with healthy volunteers, although no global CD127 regulation was observed. Our results suggest that TCR activation participates in the occurrence of this T-cell population and in the development of T-cell alterations in septic shock. Furthermore, we provide an ex vivo model for the investigation of the pathophysiology of sepsis-induced T-cell immunosuppression and the testing of innovative immunostimulant treatments.


Asunto(s)
Choque Séptico/sangre , Linfocitos T/fisiología , Anciano , Femenino , Francia , Humanos , Interleucina-7/análisis , Interleucina-7/sangre , Interleucina-7/fisiología , Subunidad alfa del Receptor de Interleucina-7/análisis , Subunidad alfa del Receptor de Interleucina-7/sangre , Recuento de Linfocitos/métodos , Masculino , Persona de Mediana Edad , Fenotipo , Receptor de Muerte Celular Programada 1/análisis , Receptor de Muerte Celular Programada 1/sangre , Choque Séptico/fisiopatología
4.
Artículo en Inglés | MEDLINE | ID: mdl-28620098

RESUMEN

Cytokine signaling is indispensable for regulatory T-cell (Treg) development in the thymus, and also influences the homeostasis, phenotypic diversity, and function of Tregs in the periphery. Because Tregs are required for establishment and maintenance of immunological self-tolerance, investigating the role of cytokines in Treg biology carries therapeutic potential in the context of autoimmune disease. This review discusses the potent and diverse influences of interleukin (IL)-2 signaling on the Treg compartment, an area of knowledge that has led to the use of low-dose IL-2 as a therapy to reregulate autoaggressive immune responses. Evidence suggesting Treg-specific impacts of the cytokines transforming growth factor ß (TGF-ß), IL-7, thymic stromal lymphopoietin (TSLP), IL-15, and IL-33 is also presented. Finally, we consider the technical challenges and knowledge limitations that must be overcome to bring other cytokine-based, Treg-targeted therapies into clinical use.


Asunto(s)
Citocinas/fisiología , Homeostasis , Transducción de Señal/fisiología , Linfocitos T Reguladores/inmunología , Animales , Humanos , Interleucina-15/fisiología , Interleucina-2/fisiología , Interleucina-33/fisiología , Interleucina-7/fisiología , Factor de Crecimiento Transformador beta/fisiología
5.
Artículo en Inglés | MEDLINE | ID: mdl-28716889

RESUMEN

Multipotent blood progenitor cells migrate into the thymus and initiate the T-cell differentiation program. T-cell progenitor cells gradually acquire T-cell characteristics while shedding their multipotentiality for alternative fates. This process is supported by extracellular signaling molecules, including Notch ligands and cytokines, provided by the thymic microenvironment. T-cell development is associated with dynamic change of gene regulatory networks of transcription factors, which interact with these environmental signals. Together with Notch or pre-T-cell-receptor (TCR) signaling, cytokines always control proliferation, survival, and differentiation of early T cells, but little is known regarding their cross talk with transcription factors. However, recent results suggest ways that cytokines expressed in distinct intrathymic niches can specifically modulate key transcription factors. This review discusses how stage-specific roles of cytokines and transcription factors can jointly guide development of early T cells.


Asunto(s)
Diferenciación Celular , Linfocitos T/citología , Proliferación Celular , Microambiente Celular , Citocinas/genética , Citocinas/metabolismo , Citocinas/fisiología , Interleucina-7/genética , Interleucina-7/metabolismo , Interleucina-7/fisiología , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/fisiología , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/metabolismo , Receptores de Interleucina-7/fisiología , Transducción de Señal , Linfocitos T/metabolismo , Timo/citología , Timo/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
6.
J Exp Med ; 214(10): 3123-3144, 2017 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-28904110

RESUMEN

Aging is linked to deficiencies in immune responses and increased systemic inflammation. To unravel the regulatory programs behind these changes, we applied systems immunology approaches and profiled chromatin accessibility and the transcriptome in PBMCs and purified monocytes, B cells, and T cells. Analysis of samples from 77 young and elderly donors revealed a novel and robust aging signature in PBMCs, with simultaneous systematic chromatin closing at promoters and enhancers associated with T cell signaling and a potentially stochastic chromatin opening mostly found at quiescent and repressed sites. Combined analyses of chromatin accessibility and the transcriptome uncovered immune molecules activated/inactivated with aging and identified the silencing of the IL7R gene and the IL-7 signaling pathway genes as potential biomarkers. This signature is borne by memory CD8+ T cells, which exhibited an aging-related loss in binding of NF-κB and STAT factors. Thus, our study provides a unique and comprehensive approach to identifying candidate biomarkers and provides mechanistic insights into aging-associated immunodeficiency.


Asunto(s)
Envejecimiento/fisiología , Linfocitos T CD8-positivos/fisiología , Cromatina/fisiología , Adulto , Anciano , Envejecimiento/inmunología , Biomarcadores , Linfocitos T CD8-positivos/inmunología , Epigénesis Genética , Femenino , Humanos , Interleucina-7/fisiología , Subunidad alfa del Receptor de Interleucina-7/fisiología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/fisiología , Masculino , Transducción de Señal/fisiología , Adulto Joven
7.
Anticancer Res ; 37(3): 963-967, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28314253

RESUMEN

Interleukin-7 (IL-7) is a cytokine that has been known since long in immunology, mainly regarding its effects on T-cells and B-cells. IL-7 has been demonstrated to be necessary for both B-cell and T-cell proliferation and lack of IL-7 causes immature immune cell arrest. Interestingly, in recent years, certain studies have strongly suggested that the role of IL-7 is far beyond the field of immunology, it might have direct or indirect effect on cancer. This review aims to summarize the role of IL-7 in immunity and its role in the pathogenesis of neoplasia.


Asunto(s)
Sistema Inmunológico/metabolismo , Interleucina-7/fisiología , Neoplasias/inmunología , Neoplasias/metabolismo , Fosfatasa Ácida/metabolismo , Animales , Antineoplásicos/uso terapéutico , Linfocitos B/citología , Diferenciación Celular , Proliferación Celular , Reordenamiento Génico , Humanos , Activación de Linfocitos , Regiones Promotoras Genéticas , Linfocitos T/citología
8.
Oncotarget ; 7(34): 54370-54379, 2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-27494846

RESUMEN

Adenosine deaminase acting on RNA (ADAR) 1 is the master editor of the transcriptome, catalyzing the conversion of adenosine to inosine (A-to-I). RNA transcripts fold into a variety of secondary structures including long intramolecular RNA duplexes that are the major substrate of ADAR1. Most A-to-I editing sites occur within RNA duplexes formed by complementary pairing of inverted retrotransposable elements interspersed within noncoding regions of transcripts. This catalytic activity of ADAR1 most likely prevents the abnormal activation of cytosolic nucleic acid sensors by self-dsRNAs. Homozygous disruption of mouse Adar is embryonic lethal due to a toxic type-I interferons response and correspondingly biallelic missense mutations in human ADAR1 cause a severe congenital interferonopathy. Here, we report that Cd19-Cre-mediated Adar gene ablation in the mouse causes a significant defect in the final stages of B cell development with an almost complete absence of newly formed immature and CD23+ mature recirculating B cells in the BM. Adar ablation in pre-B cells induced upregulation of typical interferon-stimulated genes (ISGs) and apoptosis upon further maturation. ADAR1 deficiency also inhibited the in vitro, IL-7-mediated, differentiation of BM-derived B cell precursors. In summary, ADAR1 is required, non-redundantly, for normal B lymphopoiesis in the BM and peripheral maintenance.


Asunto(s)
Adenosina Desaminasa/fisiología , Linfocitos B/fisiología , Médula Ósea/fisiología , Linaje de la Célula/fisiología , Linfopoyesis , Animales , Antígenos CD19/fisiología , Apoptosis , Interleucina-7/fisiología , Ratones , Ratones Endogámicos C57BL
9.
Haematologica ; 101(11): 1368-1379, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27470599

RESUMEN

Interleukin-7 and interleukin-7 receptor are essential for normal T-cell development and homeostasis, whereas excessive interleukin-7/interleukin-7 receptor-mediated signaling promotes leukemogenesis. The protein kinase, casein kinase 2, is overexpressed and hyperactivated in cancer, including T-cell acute lymphoblastic leukemia. Herein, we show that while interleukin-7 had a minor but significant positive effect on casein kinase 2 activity in leukemia T-cells, casein kinase 2 activity was mandatory for optimal interleukin-7/interleukin-7 receptor-mediated signaling. Casein kinase 2 pharmacological inhibition impaired signal transducer and activator of transcription 5 and phosphoinositide 3-kinase/v-Akt murine thymoma viral oncogene homolog 1 pathway activation triggered by interleukin-7 or by mutational activation of interleukin-7 receptor. By contrast, forced expression of casein kinase 2 augmented interleukin-7 signaling in human embryonic kidney 293T cells reconstituted with the interleukin-7 receptor machinery. Casein kinase 2 inactivation prevented interleukin-7-induced B-cell lymphoma 2 upregulation, maintenance of mitochondrial homeostasis and viability of T-cell acute lymphoblastic leukemia cell lines and primary leukemia cells collected from patients at diagnosis. Casein kinase 2 inhibition further abrogated interleukin-7-mediated cell growth and upregulation of the transferrin receptor, and blocked cyclin A and E upregulation and cell cycle progression. Notably, casein kinase 2 was also required for the viability of mutant interleukin-7 receptor expressing leukemia T-cells. Overall, our study identifies casein kinase 2 as a major player in the effects of interleukin-7 and interleukin-7 receptor in T-cell acute lymphoblastic leukemia. This further highlights the potential relevance of targeting casein kinase 2 in this malignancy.


Asunto(s)
Quinasa de la Caseína II/fisiología , Subunidad alfa del Receptor de Interleucina-7/fisiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Transducción de Señal , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/metabolismo , Ciclo Celular , Supervivencia Celular , Células Cultivadas , Células HEK293 , Humanos , Interleucina-7/fisiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/mortalidad
10.
Int J Hematol ; 104(3): 358-67, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27272942

RESUMEN

Chronic myeloid leukemia (CML) is a three-stage myeloproliferative disease caused by translocation between chromosomes 9 and 22. Although tyrosine kinase inhibitors (TKI) are highly effective in the treatment of CML, numerous clinical trials have shown that many patients become refractory or drug resistance, especially those in the blastic crisis of CML. The molecular mechanisms underlying CML, however, remain poorly understood. In the present study, we used a coculture model to address possible mechanisms underlying the involvement of bone marrow microenvironment in the drug resistance of CML. Our data show that interleukin-7(IL-7) levels in the bone marrow of CML patients in blastic crisis are significantly higher than those of both healthy persons and CML patients in chronic and accelerated phases. The increased IL-7 was secreted by mesenchymal stem cells (MSC) in the bone marrow, which may protect leukemic cells from apoptosis induced by imatinib through JAK1/STAT5 signaling pathway. Our findings suggest that therapeutic strategies IL-7 signaling pathway may represent a promising approach for improving CML therapy, especially for patients in blastic crisis.


Asunto(s)
Médula Ósea/metabolismo , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Mesilato de Imatinib/farmacología , Interleucina-7/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Apoptosis/efectos de los fármacos , Crisis Blástica/tratamiento farmacológico , Células Cultivadas , Técnicas de Cocultivo , Humanos , Interleucina-7/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Cell Rep ; 14(9): 2100-2107, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26923586

RESUMEN

HIV-1 post-integration latency in CD4+ lymphocytes is responsible for viral persistence despite treatment, but mechanisms involved in the establishment of latent viral reservoirs are not fully understood. We determined that both interleukin 2 (IL-2) and IL-7 induced SAMHD1 phosphorylation in T592, abrogating its antiviral activity. However, IL-7 caused a much more profound stimulatory effect on HIV-1 reverse transcription and integration than IL-2 that required chemokine co-stimulation. Both cytokines barely induced transcription due to low NF-κB induction, favoring the establishment of latent reservoirs. Effect of IL-7 on SAMHD1 phosphorylation was confirmed in IL-7-treated patients (ACTG 5214 study). Dasatinib--a tyrosine-kinase inhibitor--blocked SAMHD1 phosphorylation induced by IL-2 and IL-7 and restored HIV-1 restriction. We propose that γc-cytokines play a major role in the reservoir establishment not only by driving homeostatic proliferation but also by increasing susceptibility of CD4+ lymphocytes to HIV-1 infection through SAMHD1 inactivation.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , VIH-1/fisiología , Interleucina-7/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Integración Viral , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Humanos , Interleucina-2/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Transcripción Reversa , Proteína 1 que Contiene Dominios SAM y HD
12.
Cancer Immunol Immunother ; 65(3): 341-54, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26880265

RESUMEN

Induction of lymphopenia before adoptive transfer of T cells was followed by lymphopenia-induced proliferation (LIP) and generated a potent anti-tumor immune response in rodents and in a clinical setting. Previously, we reported that CD28 signaling is essential for the differentiation of functional effector cytotoxic T lymphocytes (CTLs) under lymphopenic conditions and sequential LIP of T cells. In this study, to clarify the correlation between LIP and the anti-tumor effect, LIP was inhibited with interleukin 7 (IL7) receptor blockade at various stages, and the anti-tumor effect then assessed. We confirmed that IL7 signaling at the start of LIP is crucial for the anti-tumor immune response. In contrast, continuous IL7 signaling was not required for tumor regression, although LIP of naïve CD8+ T cells is usually regulated by IL7. The expansion and migration of CTLs in lymphopenic hosts depend on IL7 signaling during the induction phase. Here, we propose that IL7 signaling and subsequent LIP of T cells have distinct roles in the induction of T cell immunity during lymphopenia.


Asunto(s)
Interleucina-7/fisiología , Neoplasias Pulmonares/inmunología , Linfopenia/inmunología , Transducción de Señal/fisiología , Animales , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Lectinas Tipo C , Ratones , Ratones Endogámicos C57BL , Receptores Inmunológicos/análisis , Receptores de Interleucina-7/fisiología , Linfocitos T Citotóxicos/inmunología
13.
Toxicol Sci ; 149(2): 289-99, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26518055

RESUMEN

Our previously published data show that As(+3) in vivo and in vitro, at very low concentrations, inhibits lymphoid, but not myeloid stem cell development in mouse bone marrow. We also showed that the As(+3) metabolite, monomethylarsonous acid (MMA(+3)), was responsible for the observed pre-B cell toxicity caused by As(+3). Interleukin-7 (IL-7) is the primary growth factor responsible for pre-lymphoid development in mouse and human bone marrow, and Signal Transducer and Activator of Transcription 5 (STAT5) is a transcriptional factor in the IL-7 signaling pathway. We found that MMA(+3) inhibited STAT5 phosphorylation at a concentration as low as 50 nM in mouse bone marrow pre-B cells. Inhibition of STAT5 phosphorylation by As(+3) occurred only at a concentration of 500 nM. In the IL-7 dependent mouse pre-B 2E8 cell line, we also found selective inhibition of STAT5 phosphorylation by MMA(+3), and this inhibition was dependent on effects on JAK3 phosphorylation. IL-7 receptor expression on 2E8 cell surface was also suppressed by 50 nM MMA(+3) at 18 h. As further evidence for the inhibition of STAT5, we found that the induction of several genes required in B cell development, cyclin D1, E2A, EBF1, and PAX5, were selectively inhibited by MMA(+3). Since 2E8 cells lack the enzymes responsible for the conversion of As(+3) to MMA(+3) in vitro, the results of these studies suggest that As(+3) induced inhibition of pre-B cell formation in vivo is likely dependent on the formation of MMA(+3) which in turn inhibits IL-7 signaling at several steps in mouse pre-B cells.


Asunto(s)
Interleucina-7/antagonistas & inhibidores , Compuestos Organometálicos/toxicidad , Células Precursoras de Linfocitos B/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Arsénico/toxicidad , Interleucina-7/fisiología , Janus Quinasa 1/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/fisiología , Factor de Transcripción STAT5/antagonistas & inhibidores , Factor de Transcripción STAT5/metabolismo
14.
Immunol Cell Biol ; 94(2): 196-207, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26272555

RESUMEN

Interleukin-7 (IL-7), a key immunoregulatory cytokine, plays an essential role in peripheral T-cell homeostasis and function. Signaling via the IL-7 receptor is tightly regulated and we and others have shown IL-7 provides negative feedback on its own signaling by downregulating expression of the IL-7 receptor alpha-chain (CD127) through both suppression of CD127 gene transcription and by internalization of existing CD127 proteins from the cell membrane. We show here for the first time in primary human CD8 T cells that upon stimulation with IL-7, CD127 is internalized through clathrin-coated pits, a process dependent on both lipid-raft formation and the activity of dynamin. As visualized by confocal microscopy, CD127 shows increased co-localization with clathrin within 5 min of IL-7 stimulation and within 15-30 min is seen in multiple intracellular punctae co-localizing with the early endosomal marker EEA1. By 2 h after addition of IL-7, CD127 staining associates with the late endosomal marker RAB7 and with the proteasomal 20S subunit. By inducing receptor internalization and translocation from early endosomes to the proteasome, IL-7 directly influences its receptor density on the cell surface and thus regulates the intensity of its own signaling cascades. Given the important role IL-7 plays in T-cell development, homeostasis and function, deciphering how expression of its receptor is controlled on the cell surface is essential in understanding how T-cell activity can be regulated in different microenvironments and in response to different pathogens.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vesículas Cubiertas por Clatrina/metabolismo , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Interleucina-7/fisiología , Microdominios de Membrana/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Células Cultivadas , Clatrina/metabolismo , Endocitosis , Retroalimentación Fisiológica , Homeostasis , Humanos , Microscopía Confocal , Proteolisis , Transducción de Señal
15.
PLoS One ; 10(7): e0132249, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26133981

RESUMEN

The formation and maintenance of granulomas is central to the host response to Mycobacterium tuberculosis (Mtb) infection. It is widely accepted that the lungs of patients with tuberculosis (TB) usually contain multiple infection foci, and that the granulomas evolve and differentiate independently, resulting in considerable heterogeneity. Although gene expression profiles of human blood cells have been proposed as biomarkers of Mtb infection and/or active disease, the immune profiles of discrete lesion types has not been studied extensively. Using histology, immunopathology and genome-wide transcriptome analysis, we explored the immunological profile of human lung TB granulomas. We show that although the different granulomas share core similarities in their immunological/inflammatory characteristics, they also exhibit significant divergence. Despite similar numbers of CD68+ macrophages in the different lesions, the extent of immune reactivity, as determined by the density of CD3+ T cells in the macrophage rich areas, and the extent of fibrosis, shows considerable variation. Both quantitative and qualitative differences among significantly differentially expressed genes (SDEG) were noted in each of the lesion types studied. Further, network/pathway analysis of SDEG revealed differential regulation of inflammatory response, immune cell trafficking, and cell mediated immune response in the different lesions. Our data highlight the formidable challenges facing ongoing efforts to identify peripheral blood biomarkers due to the diversity of lesion types and complexity of local immune responses in the lung.


Asunto(s)
Granuloma del Sistema Respiratorio/patología , Pulmón/patología , Tuberculosis Pulmonar/patología , Microambiente Celular , Fibrosis , Perfilación de la Expresión Génica , Granuloma del Sistema Respiratorio/genética , Granuloma del Sistema Respiratorio/inmunología , Humanos , Inflamación , Interleucina-7/fisiología , Activación de Linfocitos , Macrófagos/inmunología , Necrosis , Proyectos Piloto , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación , Receptores de Calcitriol/fisiología , Factor de Transcripción STAT1/fisiología , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Transcriptoma , Tuberculosis Resistente a Múltiples Medicamentos/genética , Tuberculosis Resistente a Múltiples Medicamentos/inmunología , Tuberculosis Resistente a Múltiples Medicamentos/patología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología
16.
Cell Signal ; 27(10): 2110-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26192098

RESUMEN

Type 1 diabetes mellitus (T1D) is associated with increased type 1 interferon (IFN) levels and subsequent severe defects in lymphocyte function, which increase susceptibility to infections. The blockade of type 1 IFN receptor 1 (IFNAR1) in non-obese diabetic mice has been shown to delay T1D onset and decrease T1D incidence by enhancing spleen CD4+ T cells and restoring B cell function. However, the effect of type 1 IFN blockade during T1D on splenic CD8+ T cells has not previously been studied. Therefore, we investigated, for the first time, the effect of IFNAR1 blockade on the survival and architecture of spleen-homing CD8+ T cells in a streptozotocin-induced T1D mouse model. Three groups of mice were examined: a non-diabetic control group; a diabetic group; and a diabetic group treated with an anti-IFNAR1 blocking antibody. We observed that T1D induction was accompanied by a marked destruction of ß cells followed by a marked reduction in insulin levels and increased IFN-α and IFN-ß levels in the diabetic group. The diabetic mice also exhibited many abnormal changes including an elevation in blood and spleen free radical (reactive oxygen species and nitric oxide) and pro-inflammatory cytokine (IL-6 and TNF-α) levels, a significant decrease in IL-7 levels, and subsequently, a significant decrease in the numbers of spleen-homing CD8+ T cells. This decrease in spleen-homing CD8+ T cells resulted from a marked reduction in the CCL21-mediated entry of CD8+ T cells into the spleen and from increased apoptosis due to a marked reduction in IL-7-mediated STAT5 and AKT phosphorylation. Interestingly, type 1 IFN signaling blockade in diabetic mice significantly restored the numbers of splenic CD8+ T cells by restoring free radical, pro-inflammatory cytokine and IL-7 levels. These effects subsequently rescued splenic CD8+ T cells from apoptosis through a mechanism that was dependent upon CCL21- and IL-7-mediated signaling. Our data suggest that type 1 IFN is an essential mediator of pathogenesis in T1D and that this role results from the negative effect of IFN signaling on the survival of splenic CD8+ T cells.


Asunto(s)
Apoptosis , Linfocitos T CD8-positivos/fisiología , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Tipo 1/sangre , Bazo/inmunología , Animales , Quimiocina CCL21/fisiología , Interferón-alfa , Interferón beta/sangre , Interleucina-7/fisiología , Subunidad alfa del Receptor de Interleucina-7/fisiología , Potencial de la Membrana Mitocondrial , Ratones Endogámicos BALB C , Estrés Oxidativo , Especies Reactivas de Oxígeno , Receptor de Interferón alfa y beta/metabolismo , Receptores CCR7/fisiología , Transducción de Señal , Bazo/patología , Estreptozocina
17.
Biol Blood Marrow Transplant ; 21(10): 1721-31, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26151303

RESUMEN

Graft-versus-host disease (GVHD) impairs immune reconstitution after allogeneic stem cell transplantation (allo-SCT) and effective therapies aimed at restoring T cell counts in GVHD patients have yet to be developed. During GVHD, CD4(+) T cell reconstitution is particularly affected and current models hold that GVHD insult to the peripheral lymphoid niche is responsible for this effect. Here, we show that naïve CD4(+) T cell homeostatic proliferation (HP) is lost during GVHD because of low systemic IL-7 and impaired dendritic cell (DC) regeneration. We assessed factors involved in DC differentiation and found that although fms-like tyrosine kinase 3 ligand (Flt3-L) levels were normal, stromal-derived factor-1α (SDF-1α) was diminished in the blood of GVHD mice. Unlike Flt3-L treatment, the administration of SDF-1α specifically increased CD8α(+) DC numbers and did not worsen GVHD. Importantly, CD4(+) T cell HP was enhanced only when IL-7 and SDF-1α or Flt3L were coadministered, confirming the crucial role of DCs and IL-7 in restoring CD4(+) T cell regeneration during GVHD. Altogether, our results indicate that CD8α(+) DCs are part of the peripheral niche that controls CD4(+) T cell HP and that their depletion, combined with low systemic IL-7, explains how GVHD constrains naïve CD4(+) T cell reconstitution after allo-SCT.


Asunto(s)
Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/inmunología , Quimiocina CXCL12/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Interleucina-7/uso terapéutico , Proteínas de la Membrana/uso terapéutico , Traslado Adoptivo , Animales , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/trasplante , Quimiocina CXCL12/sangre , Quimiocina CXCL12/deficiencia , Células Dendríticas/inmunología , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Enfermedad Injerto contra Huésped/sangre , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Interleucina-7/deficiencia , Interleucina-7/fisiología , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-7/deficiencia , Proteínas Recombinantes/uso terapéutico , Células del Estroma/metabolismo , Linfocitos T Citotóxicos/inmunología , Trasplante Homólogo
18.
Exp Hematol ; 43(4): 319-30.e10, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550197

RESUMEN

HSPA9 is located on chromosome 5q31.2 in humans, a region that is commonly deleted in patients with myeloid malignancies [del(5q)], including myelodysplastic syndrome (MDS). HSPA9 expression is reduced by 50% in patients with del(5q)-associated MDS, consistent with haploinsufficient levels. Zebrafish mutants and knockdown studies in human and mouse cells have implicated a role for HSPA9 in hematopoiesis. To comprehensively evaluate the effects of Hspa9 haploinsufficiency on hematopoiesis, we generated an Hspa9 knockout mouse model. Although homozygous knockout of Hspa9 is embryonically lethal, mice with heterozygous deletion of Hspa9 (Hspa9(+/-)) are viable and have a 50% reduction in Hspa9 expression. Hspa9(+/-) mice have normal basal hematopoiesis and do not develop MDS. However, Hspa9(+/-) mice have a cell-intrinsic reduction in bone marrow colony-forming unit-PreB colony formation without alterations in the number of B-cell progenitors in vivo, consistent with a functional defect in Hspa9(+/-) B-cell progenitors. We further reduced Hspa9 expression (<50%) using RNA interference and observed reduced B-cell progenitors in vivo, indicating that appropriate levels (≥50%) of Hspa9 are required for normal B lymphopoiesis in vivo. Knockdown of Hspa9 in an interleukin 7 (IL-7)-dependent mouse B-cell line reduced signal transducer and activator of transcription 5 (Stat5) phosphorylation following IL-7 receptor stimulation, supporting a role for Hspa9 in Stat5 signaling in B cells. Collectively, these data imply a role for Hspa9 in B lymphopoiesis and Stat5 activation downstream of IL-7 signaling.


Asunto(s)
Linfocitos B/metabolismo , Proteínas Portadoras/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Linfocitos B/citología , Proteínas Portadoras/genética , Proliferación Celular , Proteínas HSP70 de Choque Térmico/genética , Interleucina-7/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética
19.
Gut ; 64(7): 1072-81, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25298539

RESUMEN

OBJECTIVE: Under both physiological and pathological conditions, bone volume is determined by the rate of bone formation by osteoblasts and bone resorption by osteoclasts. Excessive bone loss is a common complication of human IBD whose mechanisms are not yet completely understood. Despite the role of activated CD4(+) T cells in inflammatory bone loss, the nature of the T cell subsets involved in this process in vivo remains unknown. The aim of the present study was to identify the CD4(+) T cell subsets involved in the process of osteoclastogenesis in vivo, as well as their mechanism of action. DESIGN: CD4(+) T cells were studied in IL10-/- mice and Rag1-/- mice adoptively transferred with naive CD4(+)CD45RB(high) T cells, representing two well-characterised animal models of IBD and in patients with Crohn's disease. They were phenotypically and functionally characterised by flow cytometric and gene expression analysis, as well as in in vitro cocultures with osteoclast precursors. RESULTS: In mice, we identified bone marrow (BM) CD4(+) T cells producing interleukin (IL)-17 and tumour necrosis factor (TNF)-α as an osteoclastogenic T cell subset referred to as Th17 TNF-α(+) cells. During chronic inflammation, these cells migrate to the BM where they survive in an IL-7-dependent manner and where they promote the recruitment of inflammatory monocytes, the main osteoclast progenitors. A population equivalent to the Th17 TNF-α(+) cells was also detected in patients with Crohn's disease. CONCLUSIONS: Our results highlight the osteoclastogenic function of the Th17 TNF-α(+) cells that contribute to bone loss in vivo in IBD.


Asunto(s)
Enfermedades Óseas/fisiopatología , Células de la Médula Ósea/fisiología , Enfermedades Inflamatorias del Intestino/fisiopatología , Osteoclastos/fisiología , Subgrupos de Linfocitos T/fisiología , Células Th17/fisiología , Inmunidad Adaptativa/fisiología , Animales , Enfermedades Óseas/inmunología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/fisiopatología , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Enfermedades Inflamatorias del Intestino/inmunología , Interleucina-7/fisiología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Osteoclastos/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/fisiología
20.
J Acquir Immune Defic Syndr ; 67(4): 357-64, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25202922

RESUMEN

BACKGROUND: HIV-1 shedding from the female genital tract is associated with increased sexual and perinatal transmission and has been broadly evaluated in cross-sectional studies. However, few longitudinal studies have evaluated how the immune microenvironment effects shedding. METHODS: Thirty-nine HIV-1-infected women had blood, cervicovaginal lavage, and biopsies of the uterine cervix taken quarterly for up to 5 years. Cytokines/chemokines were quantified by Luminex assay in cervicovaginal lavage, and cellular phenotypes were characterized using immunohistochemistry in cervical biopsies. Comparisons of cytokine/chemokine concentrations and the percent of tissue staining positive for T cells were compared using generalized estimating equations between non-shedding and shedding visits across all women and within a subgroup of women who intermittently shed HIV-1. RESULTS: Genital HIV-1 shedding was more common when plasma HIV-1 was detected. Cytokines associated with cell growth (interleukin-7), Th1 cells/inflammation (interleukin-12p70), and fractalkine were significantly increased at shedding visits compared with non-shedding visits within intermittent shedders and across all subjects. Within intermittent shedders and across all subjects, FOXP3 T cells were significantly decreased at shedding visits. However, there were significant increases in CD8 cells and proportions of CD8FOXP3 T cells associated with HIV-1 shedding. CONCLUSIONS: Within intermittent HIV-1 shedders, decreases in FOXP3 T cells at the shedding visit suggests that local HIV-1 replication leads to CD4 T-cell depletion, with increases in the proportion of CD8FOXP3 cells. HIV-1-infected cell loss may promote a cytokine milieu that maintains cellular homeostasis and increases immune suppressor cells in response to HIV-1 replication in the cervical tissues.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Quimiocinas/fisiología , Citocinas/fisiología , Factores de Transcripción Forkhead/fisiología , Genitales Femeninos/virología , Infecciones por VIH/virología , VIH-1/fisiología , Células TH1/fisiología , Esparcimiento de Virus/fisiología , Linfocitos T CD8-positivos/virología , Quimiocina CX3CL1/fisiología , Femenino , Homeostasis/fisiología , Humanos , Interleucina-7/fisiología , Subgrupos de Linfocitos T/fisiología , Células TH1/virología , Carga Viral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...