Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Immunity ; 42(2): 239-251, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25692700

RESUMEN

T follicular helper (Tfh) cells are essential in the induction of high-affinity, class-switched antibodies. The differentiation of Tfh cells is a multi-step process that depends upon the co-receptor ICOS and the activation of phosphoinositide-3 kinase leading to the expression of key Tfh cell genes. We report that ICOS signaling inactivates the transcription factor FOXO1, and a Foxo1 genetic deletion allowed for generation of Tfh cells with reduced dependence on ICOS ligand. Conversely, enforced nuclear localization of FOXO1 inhibited Tfh cell development even though ICOS was overexpressed. FOXO1 regulated Tfh cell differentiation through a broad program of gene expression exemplified by its negative regulation of Bcl6. Final differentiation to germinal center Tfh cells (GC-Tfh) was instead FOXO1 dependent as the Foxo1(-/-) GC-Tfh cell population was substantially reduced. We propose that ICOS signaling transiently inactivates FOXO1 to initiate a Tfh cell contingency that is completed in a FOXO1-dependent manner.


Asunto(s)
Diferenciación Celular/inmunología , Proteínas de Unión al ADN/biosíntesis , Factores de Transcripción Forkhead/genética , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Linfocitos T Colaboradores-Inductores/citología , Animales , Activación Enzimática , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6 , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología
2.
J Neuroinflammation ; 20(1): 8, 2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36631780

RESUMEN

BACKGROUND: The innate lymphoid cell (ILC) family consists of NK cells, ILC type 1, 2, 3 and lymphoid tissue inducer cells. They have been shown to play important roles in homeostasis and immune responses and are generally considered tissue resident. Not much is known about the presence of ILC members within the central nervous system and whether they are tissue resident in this organ too. Therefore, we studied the presence of all ILC members within the central nervous system and after ischemic brain insult. METHODS: We used the photothrombotic ischemic lesion method to induce ischemic lesions within the mouse brain. Using whole-mount immunofluorescence imaging, we established that the ILCs were present at the rim of the lesion. We quantified the increase of all ILC members at different time-points after the ischemic lesion induction by flow cytometry. Their migration route via chemokine CXCL12 was studied by using different genetic mouse models, in which we induced deletion of Cxcl12 within the blood-brain barrier endothelium, or its receptor, Cxcr4, in the ILCs. The functional role of the ILCs was subsequently established using the beam-walk sensorimotor test. RESULTS: Here, we report that ILCs are not resident within the mouse brain parenchyma during steady-state conditions, but are attracted towards the ischemic stroke. Specifically, we identify NK cells, ILC1s, ILC2s and ILC3s within the lesion, the highest influx being observed for NK cells and ILC1s. We further show that CXCL12 expressed at the blood-brain barrier is essential for NK cells and NKp46+ ILC3s to migrate toward the lesion. Complementary, Cxcr4-deficiency in NK cells prevents NK cells from entering the infarct area. Lack of NK cell migration results in a higher neurological deficit in the beam-walk sensorimotor test. CONCLUSIONS: This study establishes the lack of ILCs in the mouse central nervous system at steady-state and their migration towards an ischemic brain lesion. Our data show a role for blood-brain barrier-derived CXCL12 in attracting protective NK cells to ischemic brain lesions and identifies a new CXCL12/CXCR4-mediated component of the innate immune response to stroke.


Asunto(s)
Quimiocina CXCL12 , Accidente Cerebrovascular Isquémico , Células Asesinas Naturales , Animales , Ratones , Encéfalo/metabolismo , Encéfalo/patología , Quimiocina CXCL12/metabolismo , Células Endoteliales , Inmunidad Innata , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Células Asesinas Naturales/metabolismo , Linfocitos
3.
Nat Immunol ; 10(2): 176-84, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19136962

RESUMEN

Foxo transcription factors have a conserved role in the adaptation of cells and organisms to nutrient and growth factor availability. Here we show that Foxo1 has a crucial, nonredundant role in T cells. In naive T cells, Foxo1 controlled the expression of the adhesion molecule L-selectin, the chemokine receptor CCR7 and the transcription factor Klf2, and its deletion was sufficient to alter lymphocyte trafficking. Furthermore, Foxo1 deficiency resulted in a severe defect in interleukin 7 receptor alpha-chain (IL-7Ralpha) expression associated with its ability to bind an Il7r enhancer. Finally, growth factor withdrawal induced a Foxo1-dependent increase in Sell, Klf2 and Il7r expression. These data suggest that Foxo1 regulates the homeostasis and life span of naive T cells by sensing growth factor availability and regulating homing and survival signals.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Factores de Transcripción Forkhead/metabolismo , Selectina L/biosíntesis , Receptores CCR7/biosíntesis , Receptores de Interleucina-7/biosíntesis , Linfocitos T/metabolismo , Animales , Western Blotting , Diferenciación Celular/inmunología , Supervivencia Celular , Citometría de Flujo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Expresión Génica , Regulación de la Expresión Génica/inmunología , Homeostasis/inmunología , Inmunoprecipitación , Selectina L/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , ARN Mensajero/análisis , Receptores CCR7/inmunología , Receptores de Interleucina-7/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología , Linfocitos T/inmunología
4.
Nat Immunol ; 10(5): 504-13, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19363483

RESUMEN

Foxo transcription factors regulate cell cycle progression, cell survival and DNA-repair pathways. Here we demonstrate that deficiency in Foxo3 resulted in greater expansion of T cell populations after viral infection. This exaggerated expansion was not T cell intrinsic. Instead, it was caused by the enhanced capacity of Foxo3-deficient dendritic cells to sustain T cell viability by producing more interleukin 6. Stimulation of dendritic cells mediated by the coinhibitory molecule CTLA-4 induced nuclear localization of Foxo3, which in turn inhibited the production of interleukin 6 and tumor necrosis factor. Thus, Foxo3 acts to constrain the production of key inflammatory cytokines by dendritic cells and to control T cell survival.


Asunto(s)
Células Dendríticas/inmunología , Factores de Transcripción Forkhead/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos CD/inmunología , Antígenos CD/metabolismo , Infecciones por Arenaviridae/inmunología , Western Blotting , Antígeno CTLA-4 , Células Dendríticas/metabolismo , Citometría de Flujo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Congénicos , Ratones Transgénicos , Transporte de Proteínas/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
6.
Semin Immunol ; 31: 55-63, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28943093

RESUMEN

After many years of research, recent advances have shed new light on the role of the immune system in advanced-stage cancer. Various types of immune cells may be useful for therapeutic purposes, along with chemical molecules and engineered monoclonal antibodies. The immune effectors suitable for manipulation for adoptive transfer or drug targeting in vivo include natural killer (NK) cells. These cells are of particular interest because they are tightly regulated by an array of inhibitory and activating receptors, enabling them to kill tumor cells while sparing normal cells. New therapeutic antibodies blocking the interactions of inhibitory receptors (immune checkpoint inhibitors, ICI) with their ligands have been developed and can potentiate NK cell functions in vivo.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Traslado Adoptivo , Animales , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Citotoxicidad Inmunológica , Ingeniería Genética , Humanos , Células Asesinas Naturales/trasplante , Neoplasias/inmunología
7.
Immunity ; 33(6): 890-904, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21167754

RESUMEN

Foxo transcription factors integrate extrinsic signals to regulate cell division, differentiation and survival, and specific functions of lymphoid and myeloid cells. Here, we showed the absence of Foxo1 severely curtailed the development of Foxp3(+) regulatory T (Treg) cells and those that developed were nonfunctional in vivo. The loss of function included diminished CTLA-4 receptor expression as the Ctla4 gene was a direct target of Foxo1. T cell-specific loss of Foxo1 resulted in exocrine pancreatitis, hind limb paralysis, multiorgan lymphocyte infiltration, anti-nuclear antibodies and expanded germinal centers. Foxo-mediated control over Treg cell specification was further revealed by the inability of TGF-ß cytokine to suppress T-bet transcription factor in the absence of Foxo1, resulting in IFN-γ secretion. In addition, the absence of Foxo3 exacerbated the effects of the loss of Foxo1. Thus, Foxo transcription factors guide the contingencies of T cell differentiation and the specific functions of effector cell populations.


Asunto(s)
Antígenos CD/biosíntesis , Factores de Transcripción Forkhead/metabolismo , Proteínas de Dominio T Box/metabolismo , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Antígenos CD/genética , Autoinmunidad/genética , Antígeno CTLA-4 , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/inmunología , Tolerancia Inmunológica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Balance Th1 - Th2 , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
8.
Proc Natl Acad Sci U S A ; 108(45): 18324-9, 2011 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-22021440

RESUMEN

NKp46 is a cell surface receptor expressed on natural killer (NK) cells, on a minute subset of T cells, and on a population of innate lymphoid cells that produce IL-22 and express the transcription factor retinoid-related orphan receptor (ROR)-γt, referred to as NK cell receptor (NKR)(+)ROR-γt(+) cells. Here we describe Nkp46(iCre) knock-in mice in which the gene encoding the improved Cre (iCre) recombinase was inserted into the Nkp46 locus. This mouse was used to noninvasively trace cells expressing NKp46 in vivo. Fate mapping experiments demonstrated the stable expression of NKp46 on NK cells and allowed a reappraisal of the sequential steps of NK cell maturation. NKp46 genetic tracing also showed that gut NKR(+)ROR-γt(+) and NK cells represent two distinct lineages. In addition, the genetic heterogeneity of liver NK cells was evidenced. Finally, Nkp46(iCre) mice also represent a unique mouse model of conditional mutagenesis specifically in NKp46(+) cells, paving the way for further developments in the biology of NKp46(+) NK, T, and NKR(+)ROR-γt(+) cells.


Asunto(s)
Antígenos Ly/metabolismo , Tejido Linfoide/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Linfocitos T/metabolismo , Animales , Antígenos Ly/genética , Diferenciación Celular , Linaje de la Célula , Intestinos/citología , Hígado/citología , Tejido Linfoide/citología , Ratones , Ratones Transgénicos , Receptor 1 Gatillante de la Citotoxidad Natural/genética
9.
Int Immunol ; 23(7): 427-31, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21665959

RESUMEN

NK cells are considered as prototypical innate immune cells. However, recent discoveries have tended to refine the dogmatic concepts of innate and adaptive immunity. In many ways, NK cells are highly related to T cells and represent the closest innate immune cell lineage to adaptive immune cell populations. Here, we review the relationships between NK cells and T cells and discuss the recently described cell-intrinsic-adaptive features of NK cells.


Asunto(s)
Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Proliferación Celular , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Inmunidad Innata
10.
Cell Rep Med ; 3(11): 100812, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36384102

RESUMEN

Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1s) are populations of non-T, non-B lymphocytes in peripheral tissues. Although NK and ILC1 subsets have been described, their identification and characteristics remain unclear. We performed single-cell RNA sequencing and CITE-seq to explore NK and ILC1 heterogeneity between tissues. We observed that although NK1 and NK2 subsets are conserved in spleen and liver, ILC1s are heterogeneous across tissues. We identified sets of genes expressed by related subsets or characterizing unique ILC1 populations in each organ. The syndecan-4 appeared as a marker discriminating murine ILC1 from NK cells across organs. Finally, we revealed that the expressions of EOMES, GZMA, IRF8, JAK1, NKG7, PLEK, PRF1, and ZEB2 define NK cells and that IL7R, LTB, and RGS1 differentiate ILC1s from NK cells in mice and humans. Our data constitute an important resource to improve our understanding of NK-ILC1 origin, phenotype, and biology.


Asunto(s)
Inmunidad Innata , Células Asesinas Naturales , Animales , Humanos , Ratones , Inmunidad Innata/genética , Células Asesinas Naturales/metabolismo
11.
J Exp Med ; 219(2)2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35024767

RESUMEN

Gut innate lymphoid cells (ILCs) show remarkable phenotypic diversity, yet microenvironmental factors that drive this plasticity are incompletely understood. The balance between NKp46+, IL-22-producing, group 3 ILCs (ILC3s) and interferon (IFN)-γ-producing group 1 ILCs (ILC1s) contributes to gut homeostasis. The gut mucosa is characterized by physiological hypoxia, and adaptation to low oxygen is mediated by hypoxia-inducible transcription factors (HIFs). However, the impact of HIFs on ILC phenotype and gut homeostasis is not well understood. Mice lacking the HIF-1α isoform in NKp46+ ILCs show a decrease in IFN-γ-expressing, T-bet+, NKp46+ ILC1s and a concomitant increase in IL-22-expressing, RORγt+, NKp46+ ILC3s in the gut mucosa. Single-cell RNA sequencing revealed HIF-1α as a driver of ILC phenotypes, where HIF-1α promotes the ILC1 phenotype by direct up-regulation of T-bet. Loss of HIF-1α in NKp46+ cells prevents ILC3-to-ILC1 conversion, increases the expression of IL-22-inducible genes, and confers protection against intestinal damage. Taken together, our results suggest that HIF-1α shapes the ILC phenotype in the gut.


Asunto(s)
Antígenos Ly/metabolismo , Plasticidad de la Célula/inmunología , Tracto Gastrointestinal/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunidad Innata , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Animales , Biomarcadores , Susceptibilidad a Enfermedades , Expresión Génica , Perfilación de la Expresión Génica , Homeostasis , Inmunidad Mucosa , Inmunofenotipificación , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Subgrupos Linfocitarios , Ratones , Ratones Noqueados , Microbiota , Análisis de la Célula Individual
12.
Front Immunol ; 12: 768989, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868026

RESUMEN

Natural killer (NK) cells are known to be able to kill established tumor cell lines, but important caveats remain regarding their roles in the detection and elimination of developing primary tumors. Using a genetic model of selective ILC1 and NK cell deficiency, we showed that these cells were dispensable for tumor immunosurveillance and immunoediting in the MCA-induced carcinogenesis model. However, we were able to generate primary cell lines derived from MCA-induced tumors with graded sensitivity to NK1.1+ cells (including NK cells and ILC1). This differential sensitivity was associated neither with a modulation of intratumoral NK cell frequency, nor the capacity of tumor cells to activate NK cells. Instead, ILC1 infiltration into the tumor was found to be a critical determinant of NK1.1+ cell-dependent tumor growth. Finally, bulk tumor RNAseq analysis identified a gene expression signature associated with tumor sensitivity to NK1.1+ cells. ILC1 therefore appear to play an active role in inhibiting the antitumoral immune response, prompting to evaluate the differential tumor infiltration of ILC1 and NK cells in patients to optimize the harnessing of immunity in cancer therapies.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Linfocitos/inmunología , Sarcoma Experimental/inmunología , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Inmunidad Innata , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL
13.
Front Immunol ; 12: 730970, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975835

RESUMEN

Natural Killer (NK) cells are potent anti-leukemic immune effectors. However, they display multiple defects in acute myeloid leukemia (AML) patients leading to reduced anti-tumor potential. Our limited understanding of the mechanisms underlying these defects hampers the development of strategies to restore NK cell potential. Here, we have used a mouse model of AML to gain insight into these mechanisms. We found that leukemia progression resulted in NK cell maturation defects and functional alterations. Next, we assessed NK cell cytokine signaling governing their behavior. We showed that NK cells from leukemic mice exhibit constitutive IL-15/mTOR signaling and type I IFN signaling. However, these cells failed to respond to IL-15 stimulation in vitro as illustrated by reduced activation of the mTOR pathway. Moreover, our data suggest that mTOR-mediated metabolic responses were reduced in NK cells from AML-bearing mice. Noteworthy, the reduction of mTOR-mediated activation of NK cells during AML development partially rescued NK cell metabolic and functional defects. Altogether, our data strongly suggest that NK cells from leukemic mice are metabolically and functionally exhausted as a result of a chronic cytokine activation, at least partially IL-15/mTOR signaling. NK cells from AML patients also displayed reduced IL-2/15Rß expression and showed cues of reduced metabolic response to IL-15 stimulation in vitro, suggesting that a similar mechanism might occur in AML patients. Our study pinpoints the dysregulation of cytokine stimulation pathways as a new mechanism leading to NK cell defects in AML.


Asunto(s)
Interleucina-15/farmacología , Células Asesinas Naturales/inmunología , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/inmunología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Animales , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-15/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/genética
14.
Cell Rep ; 32(6): 108004, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32783932

RESUMEN

During embryogenesis, lymphoid tissue inducer (LTi) cells are essential for lymph node organogenesis. These cells are part of the innate lymphoid cell (ILC) family. Although their earliest embryonic hematopoietic origin is unclear, other innate immune cells have been shown to be derived from early hemogenic endothelium in the yolk sac as well as the aorta-gonad-mesonephros. A proper model to discriminate between these locations was unavailable. In this study, using a Cxcr4-CreERT2 lineage tracing model, we identify a major contribution from embryonic hemogenic endothelium, but not the yolk sac, toward LTi progenitors. Conversely, embryonic LTi cells are replaced by hematopoietic stem cell-derived cells in adults. We further show that, in the fetal liver, common lymphoid progenitors differentiate into highly dynamic alpha-lymphoid precursor cells that, at this embryonic stage, preferentially mature into LTi precursors and establish their functional LTi cell identity only after reaching the periphery.


Asunto(s)
Hemangioblastos/metabolismo , Hematopoyesis/fisiología , Tejido Linfoide/embriología , Receptores CXCR4/metabolismo , Animales , Desarrollo Embrionario/fisiología , Hemangioblastos/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunidad Innata , Hígado/embriología , Linfocitos/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Saco Vitelino/embriología
15.
Sci Immunol ; 2(10)2017 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-28480349

RESUMEN

Innate lymphoid cells (ILCs) are involved in immune responses to microbes and various stressed cells, such as tumor cells. They include group 1 [such as natural killer (NK) cells and ILC1], group 2, and group 3 ILCs. Besides their capacity to respond to cytokines, ILCs detect their targets through a series of cell surface-activating receptors recognizing microbial and nonmicrobial ligands. The nature of some of these ligands remains unclear, limiting our understanding of ILC biology. We focused on NKp46, which is highly conserved in mammals and expressed by all mature NK cells and subsets of ILC1 and ILC3. We show here that NKp46 binds to a soluble plasma glycoprotein, the complement factor P (CFP; properdin), the only known positive regulator of the alternative complement pathway. Consistent with the selective predisposition of patients lacking CFP to lethal Neisseria meningitidis (Nm) infections, NKp46 and group 1 ILCs bearing this receptor were found to be required for mice to survive Nm infection. Moreover, the beneficial effects of CFP treatment for Nm infection were dependent on NKp46 and group 1 NKp46+ ILCs. Thus, group 1 NKp46+ ILCs interact with the complement pathway, via NKp46, revealing a cross-talk between two partners of innate immunity in the response to an invasive bacterial infection.

16.
Viral Immunol ; 19(2): 324-34, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16817775

RESUMEN

Morbillivirus infections have been known for a long time to be associated with an acute immunosuppression in their natural hosts. Here, we show that recombinant Morbillivirus nucleoproteins from canine distemper virus, peste-des-petits-ruminants virus, and Rinderpest virus bind B-lymphocytes from dogs, goats, and cattle, respectively, similarly to measles virus nucleoprotein in humans. The use of surface plasmon resonance imaging allowed the real time detection of differential interactions between Morbillivirus nucleoproteins and FcgammaRIIb (CD32). Moreover, those nucleoproteins which bind murine Fcgamma receptor inhibited the inflammatory immune responses in mice in a Fc receptor- dependent manner. In contrast, nucleoprotein from closely related Henipavirus genus, belonging to the Paramyxoviridae family as Morbillivirus, was devoid of capacity either to bind FcgammaRIIb or to inhibit inflammatory response. Altogether, these results suggest that nucleoprotein-FcR interaction is a common mechanism used by different Morbilliviruses to modulate the immune response.


Asunto(s)
Terapia de Inmunosupresión , Infecciones por Morbillivirus/inmunología , Morbillivirus/patogenicidad , Nucleoproteínas/metabolismo , Animales , Baculoviridae/genética , Baculoviridae/metabolismo , Bovinos , Línea Celular , Perros , Humanos , Ratones , Ratones Endogámicos C57BL , Morbillivirus/clasificación , Morbillivirus/genética , Morbillivirus/inmunología , Infecciones por Morbillivirus/virología , Nucleoproteínas/genética , Receptores de IgG/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie
17.
Antioxid Redox Signal ; 14(4): 663-74, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20673126

RESUMEN

Recent studies have highlighted a fundamental role for Forkhead box O (Foxo) transcription factors in immune system homeostasis. Initial reports designed to dissect function of individual Foxo isoforms in the immune system were based on in vitro overexpression systems, and these experiments suggested that Foxo1 and Foxo3 are important for growth factor withdrawal-induced cell death. Moreover, Foxo factors importantly regulate basic cell cycle progression, and so the implication was that these factors may control lymphocyte homeostasis, including a critical function in the termination and resolution of an immune response. Most recently, cell-type-specific loss mutants for the different Foxo isoforms have revealed unexpected and highly specialized functions in the control of multiple cell types in the immune system, but they have yet to reveal a role in cell death or proliferation. This review will focus on the recent advances made in the understanding of the many ways that Foxo factors regulate the immune system, including a discussion of how the specialized versus redundant functions of Foxo transcription factors impact immune system homeostasis.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Sistema Inmunológico/metabolismo , Animales , Factores de Transcripción Forkhead/genética , Humanos , Modelos Biológicos
18.
Sci Signal ; 4(167): ra21, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21467299

RESUMEN

Natural killer (NK) cell tolerance to self is partly ensured by major histocompatibility complex (MHC) class I-specific inhibitory receptors on NK cells, which dampen their reactivity when engaged. However, NK cells that do not detect self MHC class I are not autoreactive. We used dynamic fluorescence correlation spectroscopy to show that MHC class I-independent NK cell tolerance in mice was associated with the presence of hyporesponsive NK cells in which both activating and inhibitory receptors were confined in an actin meshwork at the plasma membrane. In contrast, the recognition of self MHC class I by inhibitory receptors "educated" NK cells to become fully reactive, and activating NK cell receptors became dynamically compartmentalized in membrane nanodomains. We propose that the confinement of activating receptors at the plasma membrane is pivotal to ensuring the self-tolerance of NK cells.


Asunto(s)
Membrana Celular/metabolismo , Células Asesinas Naturales/inmunología , Autotolerancia/inmunología , Transducción de Señal/inmunología , Actinas/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Cartilla de ADN/genética , Citometría de Flujo , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Ratones , Espectrometría de Fluorescencia
19.
J Clin Invest ; 119(11): 3373-83, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19809162

RESUMEN

A key adaptation to environmental hypoxia is an increase in erythropoiesis, driven by the hormone erythropoietin (EPO) through what is traditionally thought to be primarily a renal response. However, both neurons and astrocytes (the largest subpopulation of glial cells in the CNS) also express EPO following ischemic injury, and this response is known to ameliorate damage to the brain. To investigate the role of glial cells as a component of the systemic response to hypoxia, we created astrocyte-specific deletions of the murine genes encoding the hypoxia-inducible transcription factors HIF-1alpha and HIF-2alpha and their negative regulator von Hippel-Lindau (VHL) as well as astrocyte-specific deletion of the HIF target gene Vegf. We found that loss of the hypoxic response in astrocytes does not cause anemia in mice but is necessary for approximately 50% of the acute erythropoietic response to hypoxic stress. In accord with this, erythroid progenitor cells and reticulocytes were substantially reduced in number in mice lacking HIF function in astrocytes following hypoxic stress. Thus, we have demonstrated that the glial component of the CNS is an essential component of hypoxia-induced erythropoiesis.


Asunto(s)
Eritropoyesis/fisiología , Hipoxia/fisiopatología , Neuroglía/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Eritropoyesis/genética , Eliminación de Gen , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Neuroglía/citología , Fenotipo , ARN Mensajero/metabolismo , Recombinación Genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Rev Med Virol ; 16(1): 49-63, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16237742

RESUMEN

Measles virus (MV) causes transient but profound immunosuppression resulting in increased susceptibility to secondary bacterial and viral infections. Due to the development of these opportunistic infections, measles remains the leading vaccine-preventable cause of child death worldwide. Different immune abnormalities have been associated with measles, including disappearance of delayed-type hypersensitivity reactions, impaired lymphocyte and antigen-presenting cell functions, down-regulation of pro-inflammatory interleukin 12 production and altered interferon alpha/beta signalling pathways. Several MV proteins have been suggested to hinder immune functions: hemagglutinin, fusion protein, nucleoprotein and the non-structural V and C proteins. This review will focus on the novel functions attributed to MV proteins in the immunosuppression associated with measles. Here, we highlight new advances in the field, emphasising the interaction between MV proteins and their cellular targets, in particular the cell membrane receptors, CD46, CD150, TLR2 and FcgammaRII in the induction of immunological abnormalities associated with measles.


Asunto(s)
Tolerancia Inmunológica , Virus del Sarampión/inmunología , Sarampión/complicaciones , Sarampión/inmunología , Proteínas Virales/fisiología , Humanos , Terapia de Inmunosupresión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA