Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nat Immunol ; 25(3): 448-461, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38351322

RESUMEN

Conventional dendritic cells (cDCs) include functionally and phenotypically diverse populations, such as cDC1s and cDC2s. The latter population has been variously subdivided into Notch-dependent cDC2s, KLF4-dependent cDC2s, T-bet+ cDC2As and T-bet- cDC2Bs, but it is unclear how all these subtypes are interrelated and to what degree they represent cell states or cell subsets. All cDCs are derived from bone marrow progenitors called pre-cDCs, which circulate through the blood to colonize peripheral tissues. Here, we identified distinct mouse pre-cDC2 subsets biased to give rise to cDC2As or cDC2Bs. We showed that a Siglec-H+ pre-cDC2A population in the bone marrow preferentially gave rise to Siglec-H- CD8α+ pre-cDC2As in tissues, which differentiated into T-bet+ cDC2As. In contrast, a Siglec-H- fraction of pre-cDCs in the bone marrow and periphery mostly generated T-bet- cDC2Bs, a lineage marked by the expression of LysM. Our results showed that cDC2A versus cDC2B fate specification starts in the bone marrow and suggest that cDC2 subsets are ontogenetically determined lineages, rather than cell states imposed by the peripheral tissue environment.


Asunto(s)
Células Dendríticas , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Animales , Ratones , Diferenciación Celular
2.
EMBO Mol Med ; 15(11): e17694, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37635627

RESUMEN

Therapies reconstituting autologous antiviral immunocompetence may represent an important prophylaxis and treatment for immunosuppressed individuals. Following hematopoietic cell transplantation (HCT), patients are susceptible to Herpesviridae including cytomegalovirus (CMV). We show in a murine model of HCT that macrophage colony-stimulating factor (M-CSF) promoted rapid antiviral activity and protection from viremia caused by murine CMV. M-CSF given at transplantation stimulated sequential myeloid and natural killer (NK) cell differentiation culminating in increased NK cell numbers, production of granzyme B and interferon-γ. This depended upon M-CSF-induced myelopoiesis leading to IL15Rα-mediated presentation of IL-15 on monocytes, augmented by type I interferons from plasmacytoid dendritic cells. Demonstrating relevance to human HCT, M-CSF induced myelomonocytic IL15Rα expression and numbers of functional NK cells in G-CSF-mobilized hematopoietic stem and progenitor cells. Together, M-CSF-induced myelopoiesis triggered an integrated differentiation of myeloid and NK cells to protect HCT recipients from CMV. Thus, our results identify a rationale for the therapeutic use of M-CSF to rapidly reconstitute antiviral activity in immunocompromised individuals, which may provide a general paradigm to boost innate antiviral immunocompetence using host-directed therapies.


Asunto(s)
Infecciones por Citomegalovirus , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Animales , Citomegalovirus , Factor Estimulante de Colonias de Macrófagos , Trasplante de Células Madre Hematopoyéticas/métodos , Infecciones por Citomegalovirus/prevención & control , Hematopoyesis , Antivirales/farmacología , Antivirales/uso terapéutico , Diferenciación Celular
3.
Nat Immunol ; 24(4): 714-728, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36928414

RESUMEN

Plasmacytoid dendritic cells (pDCs) are the main source of type I interferon (IFN-I) during viral infections. Their other functions are debated, due to a lack of tools to identify and target them in vivo without affecting pDC-like cells and transitional DCs (tDCs), which harbor overlapping phenotypes and transcriptomes but a higher efficacy for T cell activation. In the present report, we present a reporter mouse, pDC-Tom, designed through intersectional genetics based on unique Siglech and Pacsin1 coexpression in pDCs. The pDC-Tom mice specifically tagged pDCs and, on breeding with Zbtb46GFP mice, enabled transcriptomic profiling of all splenic DC types, unraveling diverging activation of pDC-like cells versus tDCs during a viral infection. The pDC-Tom mice also revealed initially similar but later divergent microanatomical relocation of splenic IFN+ versus IFN- pDCs during infection. The mouse models and specific gene modules we report here will be useful to delineate the physiological functions of pDCs versus other DC types.


Asunto(s)
Células Dendríticas , Interferón Tipo I , Animales , Ratones , Interferón Tipo I/metabolismo , Perfilación de la Expresión Génica , Fenotipo , Transcriptoma
4.
Clin Transl Immunology ; 10(7): e1305, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34277006

RESUMEN

OBJECTIVES: To better understand how immune responses may be harnessed against breast cancer, we investigated which immune cell types and signalling pathways are required for spontaneous control of a mouse model of mammary adenocarcinoma. METHODS: The NOP23 mammary adenocarcinoma cell line expressing epitopes derived from the ovalbumin model antigen is spontaneously controlled when orthotopically engrafted in syngeneic C57BL/6 mice. We combined this breast cancer model with antibody-mediated depletion of lymphocytes and with mutant mice affected in interferon (IFN) or type 1 conventional dendritic cell (cDC1) responses. We monitored tumor growth and immune infiltration including the activation of cognate ovalbumin-specific T cells. RESULTS: Breast cancer immunosurveillance required cDC1, NK/NK T cells, conventional CD4+ T cells and CD8+ cytotoxic T lymphocytes (CTLs). cDC1 were required constitutively, but especially during T-cell priming. In tumors, cDC1 were interacting simultaneously with CD4+ T cells and tumor-specific CTLs. cDC1 expression of the XCR1 chemokine receptor and of the T-cell-attracting or T-cell-activating cytokines CXCL9, IL-12 and IL-15 was dispensable for tumor rejection, whereas IFN responses were necessary, including cDC1-intrinsic signalling by STAT1 and IFN-γ but not type I IFN (IFN-I). cDC1 and IFNs promoted CD4+ and CD8+ T-cell infiltration, terminal differentiation and effector functions. In breast cancer patients, high intratumor expression of genes specific to cDC1, CTLs, CD4+ T cells or IFN responses is associated with a better prognosis. CONCLUSION: Interferons and cDC1 are critical for breast cancer immunosurveillance. IFN-γ plays a prominent role over IFN-I in licensing cDC1 for efficient T-cell activation.

5.
Nat Immunol ; 21(9): 983-997, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32690951

RESUMEN

Plasmacytoid dendritic cells (pDCs) are a major source of type I interferon (IFN-I). What other functions pDCs exert in vivo during viral infections is controversial, and more studies are needed to understand their orchestration. In the present study, we characterize in depth and link pDC activation states in animals infected by mouse cytomegalovirus by combining Ifnb1 reporter mice with flow cytometry, single-cell RNA sequencing, confocal microscopy and a cognate CD4 T cell activation assay. We show that IFN-I production and T cell activation were performed by the same pDC, but these occurred sequentially in time and in different micro-anatomical locations. In addition, we show that pDC commitment to IFN-I production was marked early on by their downregulation of leukemia inhibitory factor receptor and was promoted by cell-intrinsic tumor necrosis factor signaling. We propose a new model for how individual pDCs are endowed to exert different functions in vivo during a viral infection, in a manner tightly orchestrated in time and space.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Infecciones por Herpesviridae/inmunología , Muromegalovirus/fisiología , Animales , Células Cultivadas , Interferón Tipo I/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual , Factor de Necrosis Tumoral alfa/metabolismo
6.
EMBO J ; 37(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30131424

RESUMEN

Plasmacytoid dendritic cells (pDC) are the major source of type I interferons (IFN-I) during viral infections, in response to triggering of endosomal Toll-like receptors (TLRs) 7 or 9 by viral single-stranded RNA or unmethylated CpG DNA, respectively. Synthetic ligands have been used to disentangle the underlying signaling pathways. The adaptor protein AP3 is necessary to transport molecular complexes of TLRs, synthetic CpG DNA, and MyD88 into endosomal compartments allowing interferon regulatory factor 7 (IRF7) recruitment whose phosphorylation then initiates IFN-I production. High basal expression of IRF7 by pDC and its further enhancement by positive IFN-I feedback signaling appear to be necessary for robust cytokine production. In contrast, we show here that in vivo during mouse cytomegalovirus (MCMV) infection pDC produce high amounts of IFN-I downstream of the TLR9-to-MyD88-to-IRF7 signaling pathway without requiring IFN-I positive feedback, high IRF7 expression, or AP3-driven endosomal routing of TLRs. Hence, the current model of the molecular requirements for professional IFN-I production by pDC, established by using synthetic TLR ligands, does not strictly apply to a physiological viral infection.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Herpesviridae/inmunología , Interferón Tipo I/inmunología , Muromegalovirus/inmunología , Transducción de Señal/inmunología , Complejo 3 de Proteína Adaptadora/genética , Complejo 3 de Proteína Adaptadora/inmunología , Animales , Células Dendríticas/patología , Endosomas/genética , Endosomas/inmunología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/patología , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Interferón Tipo I/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/genética , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología
7.
PLoS Pathog ; 11(5): e1004897, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25954804

RESUMEN

In mice, plasmacytoid dendritic cells (pDC) and natural killer (NK) cells both contribute to resistance to systemic infections with herpes viruses including mouse Cytomegalovirus (MCMV). pDCs are the major source of type I IFN (IFN-I) during MCMV infection. This response requires pDC-intrinsic MyD88-dependent signaling by Toll-Like Receptors 7 and 9. Provided that they express appropriate recognition receptors such as Ly49H, NK cells can directly sense and kill MCMV-infected cells. The loss of any one of these responses increases susceptibility to infection. However, the relative importance of these antiviral immune responses and how they are related remain unclear. In humans, while IFN-I responses are essential, MyD88 is dispensable for antiviral immunity. Hence, a higher redundancy has been proposed in the mechanisms promoting protective immune responses against systemic infections by herpes viruses during natural infections in humans. It has been assumed, but not proven, that mice fail to mount protective MyD88-independent IFN-I responses. In humans, the mechanism that compensates MyD88 deficiency has not been elucidated. To address these issues, we compared resistance to MCMV infection and immune responses between mouse strains deficient for MyD88, the IFN-I receptor and/or Ly49H. We show that selective depletion of pDC or genetic deficiencies for MyD88 or TLR9 drastically decreased production of IFN-I, but not the protective antiviral responses. Moreover, MyD88, but not IFN-I receptor, deficiency could largely be compensated by Ly49H-mediated antiviral NK cell responses. Thus, contrary to the current dogma but consistent with the situation in humans, we conclude that, in mice, in our experimental settings, MyD88 is redundant for IFN-I responses and overall defense against a systemic herpes virus infection. Moreover, we identified direct NK cell sensing of infected cells as one mechanism able to compensate for MyD88 deficiency in mice. Similar mechanisms likely contribute to protect MyD88- or IRAK4-deficient patients from viral infections.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Interacciones Huésped-Patógeno , Interferón Tipo I/metabolismo , Células Asesinas Naturales/inmunología , Muromegalovirus/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor de Interferón alfa y beta/agonistas , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Infecciones por Herpesviridae/sangre , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Inmunidad Innata , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/virología , Interferón Tipo I/sangre , Interleucina-12/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/virología , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Mutantes , Muromegalovirus/fisiología , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Subfamilia A de Receptores Similares a Lectina de Células NK/deficiencia , Subfamilia A de Receptores Similares a Lectina de Células NK/genética , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Enfermedades de Inmunodeficiencia Primaria , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal , Organismos Libres de Patógenos Específicos , Bazo/inmunología , Bazo/metabolismo , Bazo/virología , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
8.
Cell Host Microbe ; 12(4): 571-84, 2012 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-23084923

RESUMEN

Type I interferons (IFNs) are central to antiviral defense, but how they orchestrate immune cell function is incompletely understood. We determined that IFNs produced during murine cytomegalovirus (MCMV) infection differentially affect dendritic cells (DCs) and natural killer (NK) cells. IFNs induce cell-intrinsic responses in DCs, activating antiproliferative, antiviral, and lymphocyte-activating gene networks, consistent with high activity of the transcription factor STAT1 in these cells. By comparison, NK cells exhibit lower STAT1 expression and reduced IFN responsiveness. Rather, IFNs indirectly affect NK cells by inducing IL-15, which activates the transcription factor E2F and stimulates genes promoting cell expansion. IFN cell-intrinsic responses are necessary in DCs, but not NK cells, for MCMV resistance. Thus, sensitivity to IFN-induced cytokines and differences in IFN receptor signaling program immune cells to mount distinct responses that promote viral control.


Asunto(s)
Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Células Asesinas Naturales/inmunología , Muromegalovirus/inmunología , Animales , Células Cultivadas , Citometría de Flujo , Perfilación de la Expresión Génica , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/veterinaria , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Transducción de Señal
9.
J Immunol ; 180(9): 5799-803, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18424698

RESUMEN

As initially demonstrated with murine cytomegalovirus (MCMV), plasmacytoid dendritic cells (pDCs) are the major source of IFN-alpha/beta in response to a variety of viruses in vivo. However, contradictory results have been obtained pertaining to the mechanisms promoting IFN-alpha/beta production by pDCs in response to MCMV. In this study we show that TLR7 and TLR9 exert redundant functions for IFN-alpha/beta, IL-12p40, and TNF-alpha production by pDCs in vivo during MCMV infection. In contrast, we confirm that systemic production of IL-12p70 strictly depends on TLR9. The combined loss of TLR7 and TLR9 recapitulates critical features of the phenotype of MyD88-deficient mice, including a dramatic decrease in systemic IFN-alpha/beta levels, an increase in viral load, and increased susceptibility to MCMV-induced mortality. This is the first demonstration of the implication of TLR7 in the recognition of a DNA virus.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Inmunidad Innata , Glicoproteínas de Membrana/inmunología , Muromegalovirus/inmunología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/inmunología , Animales , Células Dendríticas/inmunología , Infecciones por Herpesviridae/genética , Inmunidad Innata/genética , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferón beta/genética , Interferón beta/inmunología , Subunidad p40 de la Interleucina-12/genética , Subunidad p40 de la Interleucina-12/inmunología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Receptor Toll-Like 7/genética , Receptor Toll-Like 9/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Carga Viral
10.
Genome Biol ; 9(1): R17, 2008 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-18218067

RESUMEN

BACKGROUND: Dendritic cells (DCs) are a complex group of cells that play a critical role in vertebrate immunity. Lymph-node resident DCs (LN-DCs) are subdivided into conventional DC (cDC) subsets (CD11b and CD8alpha in mouse; BDCA1 and BDCA3 in human) and plasmacytoid DCs (pDCs). It is currently unclear if these various DC populations belong to a unique hematopoietic lineage and if the subsets identified in the mouse and human systems are evolutionary homologs. To gain novel insights into these questions, we sought conserved genetic signatures for LN-DCs and in vitro derived granulocyte-macrophage colony stimulating factor (GM-CSF) DCs through the analysis of a compendium of genome-wide expression profiles of mouse or human leukocytes. RESULTS: We show through clustering analysis that all LN-DC subsets form a distinct branch within the leukocyte family tree, and reveal a transcriptomal signature evolutionarily conserved in all LN-DC subsets. Moreover, we identify a large gene expression program shared between mouse and human pDCs, and smaller conserved profiles shared between mouse and human LN-cDC subsets. Importantly, most of these genes have not been previously associated with DC function and many have unknown functions. Finally, we use compendium analysis to re-evaluate the classification of interferon-producing killer DCs, lin-CD16+HLA-DR+ cells and in vitro derived GM-CSF DCs, and show that these cells are more closely linked to natural killer and myeloid cells, respectively. CONCLUSION: Our study provides a unique database resource for future investigation of the evolutionarily conserved molecular pathways governing the ontogeny and functions of leukocyte subsets, especially DCs.


Asunto(s)
Linaje de la Célula/genética , Células Dendríticas/citología , Perfilación de la Expresión Génica , Genoma/genética , Animales , Análisis por Conglomerados , Genoma Humano , Humanos , Leucocitos , Ratones
11.
Int Immunol ; 20(1): 45-56, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18000008

RESUMEN

Plasmacytoid dendritic cells (pDCs) are an important source of IFN-alpha/beta in response to a variety of viruses in vivo, including murine cytomegalovirus (MCMV). However, the respective contributions of various infected organs, and within these of pDCs, conventional dendritic cells and other cells, to the systemic production of IFN-alpha/beta or other innate cytokines during viral infections in vivo is largely unknown. Whether a specialization of pDC subsets in the production of different patterns of innate cytokines exists in vivo in response to a viral infection has not been investigated. Here, by analyzing for the first time directly ex vivo, at the single-cell level, the simultaneous production of up to three cytokines in pDCs isolated from MCMV-infected mice, we show that (i) pDCs are the quasi-exclusive source of IFN-alpha/beta, IL-12 and tumor necrosis factor (TNF)-alpha, early during MCMV infection, in two immunocompetent mouse lines and with two viral strains, (ii) pDC activation for IFN-alpha/beta production is organ specific and (iii) a significant proportion of pDCs simultaneously produce IFN-alpha/beta, TNF-alpha and IL-12, although TNF-alpha and IFN-alpha/beta appear more often co-expressed with one another than each of them with IL-12. Altogether, these results show a broad and non-redundant role of pDCs in early innate detection of, and defense against, viral infection. The data also show differences in the responsiveness of pDCs from different tissues and suggest distinct molecular requirements for pDC production of various cytokines. These observations must be taken into account when designing new antiviral vaccination strategies aimed at harnessing pDC responses.


Asunto(s)
Citocinas/biosíntesis , Células Dendríticas/citología , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Muromegalovirus/patogenicidad , Animales , Citocinas/genética , Células Dendríticas/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Inmunidad Innata , Interferón-alfa/genética , Interferón beta/genética , Interleucina-12/biosíntesis , Interleucina-12/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Muromegalovirus/inmunología , Especificidad de Órganos , Organismos Libres de Patógenos Específicos , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
12.
PLoS Pathog ; 3(8): e123, 2007 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-17722980

RESUMEN

Understanding the mechanisms that help promote protective immune responses to pathogens is a major challenge in biomedical research and an important goal for the design of innovative therapeutic or vaccination strategies. While natural killer (NK) cells can directly contribute to the control of viral replication, whether, and how, they may help orchestrate global antiviral defense is largely unknown. To address this question, we took advantage of the well-defined molecular interactions involved in the recognition of mouse cytomegalovirus (MCMV) by NK cells. By using congenic or mutant mice and wild-type versus genetically engineered viruses, we examined the consequences on antiviral CD8 T cell responses of specific defects in the ability of the NK cells to control MCMV. This system allowed us to demonstrate, to our knowledge for the first time, that NK cells accelerate CD8 T cell responses against a viral infection in vivo. Moreover, we identify the underlying mechanism as the ability of NK cells to limit IFN-alpha/beta production to levels not immunosuppressive to the host. This is achieved through the early control of cytomegalovirus, which dramatically reduces the activation of plasmacytoid dendritic cells (pDCs) for cytokine production, preserves the conventional dendritic cell (cDC) compartment, and accelerates antiviral CD8 T cell responses. Conversely, exogenous IFN-alpha administration in resistant animals ablates cDCs and delays CD8 T cell activation in the face of NK cell control of viral replication. Collectively, our data demonstrate that the ability of NK cells to respond very early to cytomegalovirus infection critically contributes to balance the intensity of other innate immune responses, which dampens early immunopathology and promotes optimal initiation of antiviral CD8 T cell responses. Thus, the extent to which NK cell responses benefit the host goes beyond their direct antiviral effects and extends to the prevention of innate cytokine shock and to the promotion of adaptive immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Herpesviridae/inmunología , Células Asesinas Naturales/inmunología , Muromegalovirus/inmunología , Animales , Linfocitos T CD8-positivos/citología , Citotoxicidad Inmunológica , Células Dendríticas/citología , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Infecciones por Herpesviridae/metabolismo , Inmunidad Innata , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Células Asesinas Naturales/citología , Activación de Linfocitos , Ratones , Ratones Congénicos , Ratones Endogámicos BALB C , Organismos Libres de Patógenos Específicos , Bazo/citología , Bazo/virología , Replicación Viral/inmunología
13.
J Immunol ; 177(5): 2908-16, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16920926

RESUMEN

DAP12 is an ITAM-containing adaptor molecule conveying activating properties to surface receptors on many cell types. We show here that DAP12 paradoxically down-modulates plasmacytoid dendritic cell (pDC) cytokine production in vivo during murine CMV (MCMV) infection. Higher levels of IFN-alphabeta and IL-12 were detected upon MCMV infection or CpG treatment in DAP12-deficient (DAP12(o)) mice as compared with wild-type (WT) mice. This resulted from altered homeostasis and enhanced responsiveness of pDCs in DAP12(o) animals. Increased numbers of pDCs were observed in the periphery of both naive and MCMV-infected DAP12(o) mice. A higher proportion of pDCs was activated in infected DAP12(o) mice, as demonstrated by intracellular staining using an optimized protocol for simultaneous detection of IFN-alpha and IFN-beta. The homeostasis of WT and DAP12(o) pDCs did not differ in mixed bone marrow chimeric mice. In addition, a similar efficiency of pDC differentiation was observed in vitro in Fms-like tyrosine kinase receptor 3 ligand cultures of WT and DAP12(o) bone marrow cells. This suggests that DAP12 signaling effects on pDC homeostasis are indirect. In contrast, in response to CpG, DAP12-mediated effects on both IL-12 and IFN-alphabeta production were intrinsic to the pDCs. However, in response to MCMV, only IL-12 but not IFN-alphabeta production was affected by pDC-intrinsic DAP12 signaling. Thus, DAP12 signaling in pDCs can mediate different regulatory effects on their functions, depending on the mechanisms of pDC activation. The potential implications of the regulation of pDC functions by DAP12 for promoting health over disease are discussed.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Dendríticas/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/metabolismo , Homeostasis/inmunología , Muromegalovirus/inmunología , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Citocinas/biosíntesis , Células Dendríticas/citología , Regulación de la Expresión Génica , Infecciones por Herpesviridae/virología , Interferón-alfa/biosíntesis , Interferón-alfa/sangre , Interferón beta/biosíntesis , Interferón beta/sangre , Interleucina-12/biosíntesis , Ratones , ARN Mensajero/genética
14.
Biochem Biophys Res Commun ; 329(3): 917-24, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15752743

RESUMEN

The main function of Vif is to limit the antiviral activity of APOBEC3G by counteracting its packaging into HIV-1 virions. In this work, we examine the possible functional interactions between Vif, APOBEC3G, and two Src family tyrosine kinases, Fyn and Hck, present in T lymphocytes and in monocyte-macrophages, respectively. By GST pull-down, we show that the SH3 domains of Fyn and Hck, and the corresponding full-length proteins bind Vif of HIV-1. One consequence of this interaction is a reduction in their catalytic activity. Interestingly, we also observed that APOBEC3G can be phosphorylated on tyrosine in the presence of Fyn or Hck, suggesting that both kinases may regulate APOBEC3G function. Accordingly, we demonstrate that in the presence of Fyn or Hck and in the absence of Vif, the overall level of APOBEC3G incorporated into HIV-1 particles is decreased, whereas the level of encapsidation of its phosphorylated form is significantly enhanced.


Asunto(s)
Productos del Gen vif/metabolismo , VIH-1/fisiología , Proteínas Tirosina Quinasas/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Tirosina/metabolismo , Familia-src Quinasas/metabolismo , Desaminasa APOBEC-3G , Sitios de Unión , Citidina Desaminasa , Células HeLa , Humanos , Riñón/metabolismo , Riñón/virología , Monocitos/metabolismo , Monocitos/virología , Nucleósido Desaminasas , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-fyn , Proteínas Proto-Oncogénicas c-hck , Proteínas Represoras , Linfocitos T/metabolismo , Linfocitos T/virología , Virión/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana
15.
J Mol Biol ; 345(3): 547-58, 2005 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-15581898

RESUMEN

APOBEC3G is a cytidine deaminase that limits the replication of many retroviruses. This antiviral host factor is packaged into retrovirus particles, where it targets single-stranded DNA generated during reverse transcription and induces up to 2% of G-to-A mutations, which are lethal for the HIV-1 provirus. Vif protein counteracts this antiviral factor by decreasing its packaging into lentivirus particles. Here, we demonstrate that Nedd4-1, an HECT E3 ubiquitin ligase, interacts with APOBEC3G, through its WW2 and WW3 domains. As a result of this interaction, APOBEC3G undergoes post-translational modification by addition of ubiquitin moieties. Accordingly, we demonstrate that the dominant negative Nedd4-1 C/S form prevents APOBEC3G ubiquitination. Moreover, the packaging of APOBEC3G into Pr55 Gag virus-like particles and into HIV-1 virions is reduced when Nedd4-1 C/S is expressed. During HIV-1 viral production in the presence of APOBEC3G, Nedd4-1 C/S restores partially the infectivity of Deltavif HIV-1. We conclude that the ubiquitination of APOBEC3G by Nedd4-1 favors its targeting to the virus assembly site where APOBEC3G interacts with Gag and is packaged into HIV-1 particles in the absence of Vif.


Asunto(s)
Proteínas Portadoras/metabolismo , VIH-1/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas/metabolismo , Ubiquitina/metabolismo , Ensamble de Virus , Desaminasa APOBEC-3G , Citidina Desaminasa , VIH-1/fisiología , Células HeLa , Humanos , Inmunoprecipitación , Nucleósido Desaminasas , Proteínas Represoras
16.
Biochem Biophys Res Commun ; 321(3): 566-73, 2004 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-15358144

RESUMEN

The cytidine deaminase hAPOBEC3G is an antiviral human factor that counteracts the replication of HIV-1 in absence of the Vif protein. hAPOBEC3G is packaged into virus particles and lethally hypermutates HIV-1. In this work, we examine the mechanisms governing hAPOBEC3G packaging. By GST pull-down and co-immunoprecipitation assays, we show that hAPOBEC3G binds to HIV-1 Pr55 Gag and its NC domain and to the RT and IN domains contained in Pr160 Gag-Pol. We demonstrate that the expression of HIV-1 Gag is sufficient to induce the packaging of hAPOBEC3G into Gag particles. Gag-Pol polypeptides containing RT and IN domains, as well as HIV-1 genomic RNA, seem not to be necessary for hAPOBEC3G packaging. Lastly, we show that hAPOBEC3G and its murine ortholog are packaged into HIV-1 and MLV Gag particles. We conclude that the Gag polypeptides from distant retroviruses have conserved domains allowing the packaging of the host antiviral factor APOBEC3G.


Asunto(s)
Productos del Gen gag/metabolismo , Virus de la Leucemia Murina/metabolismo , Precursores de Proteínas/metabolismo , Proteínas/metabolismo , Virión/metabolismo , Desaminasa APOBEC-3G , Animales , Citidina Desaminasa , Productos del Gen gag/genética , VIH-1/genética , VIH-1/fisiología , Células HeLa , Humanos , Virus de la Leucemia Murina/genética , Ratones , Nucleósido Desaminasas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/genética , Estructura Terciaria de Proteína , Proteínas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras , Virión/química , Replicación Viral , Productos del Gen gag del Virus de la Inmunodeficiencia Humana , Productos del Gen pol del Virus de la Inmunodeficiencia Humana
17.
Biochem Biophys Res Commun ; 315(1): 66-72, 2004 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-15013426

RESUMEN

The viral infectivity factor (Vif), one of the six HIV-1 auxiliary genes, is absolutely necessary for productive infection in primary CD4-positive T lymphocytes and macrophages. Vif overcomes the antiviral function of the host factor APOBEC3G. To better understand this mechanism, it is of interest to characterize cellular proteins that interact with Vif and may regulate its function. Here, we show that Vif binds to hNedd4 and AIP4, two HECT E3 ubiquitin ligases. WW domains present in those HECT enzymes contribute to the binding of Vif. Moreover, the region of Vif, which includes amino acids 20-128 and interacts with the hNedd4 WW domains, does not contain proline-rich stretches. Lastly, we show that Vif undergoes post-translational modifications by addition of ubiquitin both in cells overexpressing Vif and in cells expressing HIV-1 provirus. Vif is mainly mono-ubiquitinated, a modification known to address the Gag precursor to the virus budding site.


Asunto(s)
Productos del Gen vif/metabolismo , VIH-1/química , Procesamiento Proteico-Postraduccional , Ubiquitina/metabolismo , Western Blotting , Línea Celular , Complejos de Clasificación Endosomal Requeridos para el Transporte , Expresión Génica , Productos del Gen vif/genética , Glutatión Transferasa/metabolismo , Infecciones por VIH/virología , Células HeLa , Humanos , Ubiquitina-Proteína Ligasas Nedd4 , Pruebas de Precipitina , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/metabolismo , Transfección , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA