Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Nat Immunol ; 18(4): 422-432, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28218746

RESUMEN

During microbial infection, responding CD8+ T lymphocytes differentiate into heterogeneous subsets that together provide immediate and durable protection. To elucidate the dynamic transcriptional changes that underlie this process, we applied a single-cell RNA-sequencing approach and analyzed individual CD8+ T lymphocytes sequentially throughout the course of a viral infection in vivo. Our analyses revealed a striking transcriptional divergence among cells that had undergone their first division and identified previously unknown molecular determinants that controlled the fate specification of CD8+ T lymphocytes. Our findings suggest a model for the differentiation of terminal effector cells initiated by an early burst of transcriptional activity and subsequently refined by epigenetic silencing of transcripts associated with memory lymphocytes, which highlights the power and necessity of single-cell approaches.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Epigénesis Genética , Transcripción Genética , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Análisis por Conglomerados , Biología Computacional/métodos , Perfilación de la Expresión Génica , Silenciador del Gen , Heterogeneidad Genética , Histonas/metabolismo , Memoria Inmunológica/genética , Memoria Inmunológica/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Transcriptoma
3.
Immunity ; 49(2): 208-210, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30134200

RESUMEN

In a recent issue of Nature, Borges da Silva et al. (2018) reveal that P2RX7, a receptor for extracellular ATP, promotes CD8 T cell memory by enhancing metabolic fitness. This work links an ancient "danger" signal with long-term immunity.


Asunto(s)
Receptores Purinérgicos , Linfocitos T , Adenosina Trifosfato , Linfocitos T CD8-positivos , Inmunidad
4.
Immunity ; 48(4): 730-744.e5, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29669251

RESUMEN

Although characterization of T cell exhaustion has unlocked powerful immunotherapies, the mechanisms sustaining adaptations of short-lived innate cells to chronic inflammatory settings remain unknown. During murine chronic viral infection, we found that concerted events in bone marrow and spleen mediated by type I interferon (IFN-I) and Toll-like receptor 7 (TLR7) maintained a pool of functionally exhausted plasmacytoid dendritic cells (pDCs). In the bone marrow, IFN-I compromised the number and the developmental capacity of pDC progenitors, which generated dysfunctional pDCs. Concurrently, exhausted pDCs in the periphery were maintained by self-renewal via IFN-I- and TLR7-induced proliferation of CD4- subsets. On the other hand, pDC functional loss was mediated by TLR7, leading to compromised IFN-I production and resistance to secondary infection. These findings unveil the mechanisms sustaining a self-perpetuating pool of functionally exhausted pDCs and provide a framework for deciphering long-term exhaustion of other short-lived innate cells during chronic inflammation.


Asunto(s)
Autorrenovación de las Células/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Glicoproteínas de Membrana/inmunología , Receptor Toll-Like 7/inmunología , Células 3T3 , Animales , Proteínas Portadoras/biosíntesis , Línea Celular , Proliferación Celular , Proteínas de Unión al ADN/biosíntesis , Células Dendríticas/citología , Humanos , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/biosíntesis , Proteínas Represoras , Transducción de Señal/inmunología , Factor de Transcripción 4/biosíntesis , Factores de Transcripción/biosíntesis
5.
PLoS Biol ; 21(1): e3001983, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36716323

RESUMEN

During a microbial infection, responding CD8+ T cells give rise to effector cells that provide acute host defense and memory cells that provide sustained protection. An alternative outcome is exhaustion, a state of T cell dysfunction that occurs in the context of chronic infections and cancer. Although it is evident that exhausted CD8+ T (TEX) cells are phenotypically and molecularly distinct from effector and memory CD8+ T cells, the factors regulating the earliest events in the differentiation process of TEX cells remain incompletely understood. Here, we performed single-cell RNA-sequencing and single-cell ATAC-sequencing of CD8+ T cells responding to LCMV-Armstrong (LCMV-Arm) or LCMV-Clone 13 (LCMV-Cl13), which result in acute or chronic infections, respectively. Compared to CD8+ T cells that had undergone their first division in response to LCMV-Arm (Div1ARM) cells, CD8+ T cells that had undergone their first division in response to LCMV-Cl13 (Div1CL13) expressed higher levels of genes encoding transcription factors previously associated with exhaustion, along with higher levels of Ezh2, the catalytic component of the Polycomb Repressive Complex 2 (PRC2) complex, which mediates epigenetic silencing. Modulation of Ezh2 resulted in altered expression of exhaustion-associated molecules by CD8+ T cells responding to LCMV-Cl13, though the specific cellular and infectious contexts, rather than simply the level of Ezh2 expression, likely determine the eventual outcome. Taken together, these findings suggest that the differentiation paths of CD8+ T cells responding to acute versus chronic infections may diverge earlier than previously appreciated.


Asunto(s)
Coriomeningitis Linfocítica , Humanos , Animales , Ratones , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/metabolismo , Infección Persistente , Linfocitos T CD8-positivos/metabolismo , Virus de la Coriomeningitis Linfocítica , Epigénesis Genética , Ratones Endogámicos C57BL
6.
Immunol Rev ; 309(1): 12-24, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35775361

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has caused millions of deaths in the past two years. Although initially little was understood about this virus, recent research has significantly advanced and landed interferons (IFNs) in the spotlight. While Type I and III IFN have long been known as central to antiviral immunity, in the case of COVID-19 their role was initially controversial. However, the protective function of IFN is now well supported by the identification of human deficiencies in IFN responses as a predictor of disease severity. Here, we will review the cell types and pathways that lead to IFN production as well as the importance of IFN timing and location for disease outcome. We will further discuss the mechanisms that SARS-CoV-2 uses to evade IFN responses, and the current efforts to implement IFNs as therapeutics in the treatment of COVID-19. It is essential to understand the relationships between SARS-CoV-2 and IFN to better inform treatments that exploit IFN functions to alleviate COVID-19.


Asunto(s)
COVID-19 , Interferón Tipo I , Antivirales/uso terapéutico , Humanos , Interferón Tipo I/farmacología , Interferones , SARS-CoV-2
7.
Trends Immunol ; 43(7): 500-502, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672237

RESUMEN

Emerging immunotherapies offer a new hope for cancer patients but are not always effective even when a tumor is recognized by the immune system. Baldominos and colleagues address this challenge by characterizing a resilient niche of metabolically unique quiescent cancer cells (QCCs) that resist T cell-mediated control.


Asunto(s)
Evasión Inmune , Neoplasias , Humanos , Inmunoterapia , Linfocitos T , Microambiente Tumoral
8.
Immunity ; 44(2): 274-86, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26885856

RESUMEN

Despite the importance of the co-receptor PD-1 in T cell immunity, the upstream signaling pathway that regulates PD-1 expression has not been defined. Glycogen synthase kinase 3 (GSK-3, isoforms α and ß) is a serine-threonine kinase implicated in cellular processes. Here, we identified GSK-3 as a key upstream kinase that regulated PD-1 expression in CD8(+) T cells. GSK-3 siRNA downregulation, or inhibition by small molecules, blocked PD-1 expression, resulting in increased CD8(+) cytotoxic T lymphocyte (CTL) function. Mechanistically, GSK-3 inactivation increased Tbx21 transcription, promoting enhanced T-bet expression and subsequent suppression of Pdcd1 (encodes PD-1) transcription in CD8(+) CTLs. Injection of GSK-3 inhibitors in mice increased in vivo CD8(+) OT-I CTL function and the clearance of murine gamma-herpesvirus 68 and lymphocytic choriomeningitis clone 13 and reversed T cell exhaustion. Our findings identify GSK-3 as a regulator of PD-1 expression and demonstrate the applicability of GSK-3 inhibitors in the modulation of PD-1 in immunotherapy.


Asunto(s)
Aminofenoles/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Glucógeno Sintasa Quinasa 3/metabolismo , Infecciones por Herpesviridae/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Maleimidas/administración & dosificación , Receptor de Muerte Celular Programada 1/metabolismo , Rhadinovirus/fisiología , Proteínas de Dominio T Box/metabolismo , Linfocitos T Citotóxicos/inmunología , Aminofenoles/efectos adversos , Animales , Linfocitos T CD8-positivos/virología , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Glucógeno Sintasa Quinasa 3/genética , Maleimidas/efectos adversos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/genética , ARN Interferente Pequeño/genética , Proteínas de Dominio T Box/genética , Linfocitos T Citotóxicos/virología , Carga Viral/efectos de los fármacos , Carga Viral/genética
9.
Nat Immunol ; 13(12): 1162-70, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23086447

RESUMEN

The NF-κB protein RelB controls dendritic cell (DC) maturation and may be targeted therapeutically to manipulate T cell responses in disease. Here we report that RelB promoted DC activation not as the expected RelB-p52 effector of the noncanonical NF-κB pathway, but as a RelB-p50 dimer regulated by canonical IκBs, IκBα and IκBɛ. IκB control of RelB minimized spontaneous maturation but enabled rapid pathogen-responsive maturation. Computational modeling of the NF-κB signaling module identified control points of this unexpected cell type-specific regulation. Fibroblasts that we engineered accordingly showed DC-like RelB control. Canonical pathway control of RelB regulated pathogen-responsive gene expression programs. This work illustrates the potential utility of systems analyses in guiding the development of combination therapeutics for modulating DC-dependent T cell responses.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Activación de Linfocitos , FN-kappa B/metabolismo , Factor de Transcripción ReIB/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Quinasa I-kappa B/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , Multimerización de Proteína , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/metabolismo , Factor de Transcripción ReIB/genética
10.
Proc Natl Acad Sci U S A ; 117(51): 32574-32583, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33288689

RESUMEN

It is known that a subpopulation of T cells expresses two T cell receptor (TCR) clonotypes, though the extent and functional significance of this is not established. To definitively evaluate dual TCRα cells, we generated mice with green fluorescent protein and red fluorescent protein reporters linked to TCRα, revealing that ∼16% of T cells express dual TCRs, notably higher than prior estimates. Importantly, dual TCR expression has functional consequences, as dual TCR cells predominated response to lymphocytic choriomeningitis virus infection, comprising up to 60% of virus-specific CD4+ and CD8+ T cells during acute responses. Dual receptor expression selectively influenced immune memory, as postinfection memory CD4+ populations contained significantly increased frequencies of dual TCR cells. These data reveal a previously unappreciated contribution of dual TCR cells to the immune repertoire and highlight their potential effects on immune responses.


Asunto(s)
Genes Codificadores de la Cadena alfa de los Receptores de Linfocito T/fisiología , Coriomeningitis Linfocítica/inmunología , Linfocitos T/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD4-Positivos/virología , Antígenos CD5/inmunología , Antígenos CD5/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Chlorocebus aethiops , Femenino , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Memoria Inmunológica/genética , Virus de la Coriomeningitis Linfocítica/patogenicidad , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Timocitos/inmunología , Timocitos/fisiología , Células Vero
11.
Proc Natl Acad Sci U S A ; 117(40): 24998-25007, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32958643

RESUMEN

Infections elicit immune adaptations to enable pathogen resistance and/or tolerance and are associated with compositional shifts of the intestinal microbiome. However, a comprehensive understanding of how infections with pathogens that exhibit distinct capability to spread and/or persist differentially change the microbiome, the underlying mechanisms, and the relative contribution of individual commensal species to immune cell adaptations is still lacking. Here, we discovered that mouse infection with a fast-spreading and persistent (but not a slow-spreading acute) isolate of lymphocytic choriomeningitis virus induced large-scale microbiome shifts characterized by increased Verrucomicrobia and reduced Firmicute/Bacteroidetes ratio. Remarkably, the most profound microbiome changes occurred transiently after infection with the fast-spreading persistent isolate, were uncoupled from sustained viral loads, and were instead largely caused by CD8 T cell responses and/or CD8 T cell-induced anorexia. Among the taxa enriched by infection with the fast-spreading virus, Akkermansia muciniphila, broadly regarded as a beneficial commensal, bloomed upon starvation and in a CD8 T cell-dependent manner. Strikingly, oral administration of A. muciniphila suppressed selected effector features of CD8 T cells in the context of both infections. Our findings define unique microbiome differences after chronic versus acute viral infections and identify CD8 T cell responses and downstream anorexia as driver mechanisms of microbial dysbiosis after infection with a fast-spreading virus. Our data also highlight potential context-dependent effects of probiotics and suggest a model in which changes in host behavior and downstream microbiome dysbiosis may constitute a previously unrecognized negative feedback loop that contributes to CD8 T cell adaptations after infections with fast-spreading and/or persistent pathogens.


Asunto(s)
Anorexia/inmunología , Antígenos CD8/inmunología , Memoria Inmunológica/inmunología , Coriomeningitis Linfocítica/inmunología , Virosis/inmunología , Akkermansia , Animales , Anorexia/microbiología , Anorexia/virología , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Disbiosis/inmunología , Disbiosis/microbiología , Disbiosis/virología , Firmicutes/inmunología , Firmicutes/metabolismo , Microbioma Gastrointestinal/inmunología , Humanos , Coriomeningitis Linfocítica/microbiología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Linfocitos T/inmunología , Linfocitos T/microbiología , Verrucomicrobia/inmunología , Verrucomicrobia/patogenicidad , Virosis/microbiología , Virosis/patología
12.
Immunity ; 39(3): 548-59, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-23993651

RESUMEN

The outcome of chronic viral infections, which affect millions of people worldwide, is greatly dependent on CD4⁺ T cells. Here we showed that T cell-specific ablation of the common interleukin-6 (IL-6) family receptor, gp130, profoundly compromised virus-specific CD4⁺ T cell survival, T follicular helper responses, and IL-21 production at late stages of chronic lymphocytic choriomeningitis virus (LCMV) infection. These effects were cell intrinsic for CD4⁺ T cells and were accompanied by a reduction of CD8⁺ T cells, antibodies, and a severe failure in viral control. We identified IL-27 as a gp130 cytokine that promoted antiviral CD4⁺ T cell accumulation in vivo and that rapidly induced IL-21 ex vivo. Furthermore, IL-27R was critical for control of persistent LCMV in vivo. These results reveal that gp130 cytokines (particularly IL-27) are key regulators of CD4⁺ T cell responses during an established chronic viral infection, empowering both humoral and cytotoxic immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Receptor gp130 de Citocinas/metabolismo , Interleucina-27/metabolismo , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Receptores de Citocinas/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular , Interleucinas/biosíntesis , Coriomeningitis Linfocítica/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Citocinas/genética , Receptores de Interleucina , Receptores de Interleucina-6
13.
J Immunol ; 203(6): 1509-1520, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31413107

RESUMEN

The pleiotropic cytokine IL-6 plays an integral role not only in innate inflammatory responses but also in the activation and differentiation of lymphocyte subsets. In this study, by using a conditional knockout (cKO) model with selective IL-6 receptor deletion in T cells (IL-6R-cKO), we demonstrated that T cell-specific IL-6R signaling is essential for viral control during persistent lymphocytic choriomeningitis virus clone 13 infection. Strikingly, we observed that in contrast to previous studies with ubiquitous IL-6 deletion or blockade, specific IL-6R deletion in T cells did not affect T follicular helper (Tfh) cell accumulation unless IL-6R-deficient T cells were competing with wild-type cells in mixed bone marrow chimeras. In contrast, Tfh cells from IL-6R-cKO-infected mice exhibited reduced ICOS expression in both chimeric and nonchimeric settings, and this sole identifiable Tfh defect was associated with reduced germinal centers, compromised Ig switch and low avidity of lymphocytic choriomeningitis virus-specific Abs despite intact IL-6R expression in B cells. We posit that IL-6R cis-signaling is absolutely required for appropriate ICOS expression in Tfh cells and provides a competitive advantage for Tfh accumulation, enabling generation of optimal B cell and Ab responses, and ultimately viral control during in vivo chronic infection.


Asunto(s)
Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Receptores de Interleucina-6/metabolismo , Transducción de Señal/fisiología , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Linfocitos B/metabolismo , Linfocitos B/virología , Diferenciación Celular/fisiología , Centro Germinal/metabolismo , Centro Germinal/virología , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Colaboradores-Inductores/virología
14.
J Virol ; 92(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29593047

RESUMEN

Chronic viral infections represent a major challenge to the host immune response, and a unique network of immunological elements, including cytokines, are required for their containment. By using a model persistent infection with the natural murine pathogen lymphocytic choriomeningitis virus clone 13 (LCMV Cl13) we investigated the role of one such cytokine, interleukin-27 (IL-27), in the control of chronic infection. We found that IL-27 receptor (IL-27R) signaling promoted control of LCMV Cl13 as early as days 1 and 5 after infection and that il27p28 transcripts were rapidly elevated in multiple subsets of dendritic cells (DCs) and myeloid cells. In particular, plasmacytoid DCs (pDCs), the most potent type 1 interferon (IFN-I)-producing cells, significantly increased il27p28 in a Toll-like receptor 7 (TLR7)-dependent fashion. Notably, mice deficient in an IL-27-specific receptor, WSX-1, exhibited a pleiotropy of innate and adaptive immune alterations after chronic lymphocytic choriomeningitis virus (LCMV) infection, including compromised NK cell cytotoxicity and antibody responses. While, the majority of these immune alterations appeared to be cell extrinsic, cell-intrinsic IL-27R was necessary to maintain early pDC numbers, which, alongside lower IFN-I transcription in CD11b+ DCs and myeloid cells, may explain the compromised IFN-I elevation that we observed early after LCMV Cl13 infection in IL-27R-deficient mice. Together, these data highlight the critical role of IL-27 in enabling optimal antiviral immunity early and late after infection with a systemic persistent virus and suggest that a previously unrecognized positive-feedback loop mediated by IL-27 in pDCs might be involved in this process.IMPORTANCE Persistently replicating pathogens, such as human immunodeficiency virus, hepatitis B virus, and hepatitis C virus, represent major health problems worldwide. These infections impose a long-term challenge on the host immune system, which must be heavily and continuously regulated to keep pathogen replication in check without causing fatal immunopathology. Using a persistently replicating rodent pathogen, LCMV, in its natural host, we identified the cellular sources and effects of one important regulatory pathway, interleukin-27 receptor WSX-1 signaling, that is required for both very early and late restriction of chronic (but not acute) infection. We found that WSX-1 was necessary to promote innate immunity and the development of aberrant adaptive immune responses. This not only highlights the role of IL-27 receptor signaling in regulating distinct host responses that are known to be necessary to control chronic infections, but also positions IL-27 as a potential therapeutic target for their modulation.


Asunto(s)
Inmunidad Adaptativa/inmunología , Células Dendríticas/inmunología , Inmunidad Innata/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Receptores de Citocinas/inmunología , Animales , Enfermedad Crónica , Interleucina-27/inmunología , Células Asesinas Naturales/inmunología , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Citocinas/genética , Receptores de Interleucina , Transducción de Señal
15.
PLoS Pathog ; 13(8): e1006588, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28837667

RESUMEN

The multifunctional NS1 protein of influenza A viruses suppresses host cellular defense mechanisms and subverts other cellular functions. We report here on a new role for NS1 in modifying cell-cell signaling via the Hedgehog (Hh) pathway. Genetic epistasis experiments and FRET-FLIM assays in Drosophila suggest that NS1 interacts directly with the transcriptional mediator, Ci/Gli1. We further confirmed that Hh target genes are activated cell-autonomously in transfected human lung epithelial cells expressing NS1, and in infected mouse lungs. We identified a point mutation in NS1, A122V, that modulates this activity in a context-dependent fashion. When the A122V mutation was incorporated into a mouse-adapted influenza A virus, it cell-autonomously enhanced expression of some Hh targets in the mouse lung, including IL6, and hastened lethality. These results indicate that, in addition to its multiple intracellular functions, NS1 also modifies a highly conserved signaling pathway, at least in part via cell autonomous activities. We discuss how this new Hh modulating function of NS1 may influence host lethality, possibly through controlling cytokine production, and how these new insights provide potential strategies for combating infection.


Asunto(s)
Proteínas Hedgehog/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Transducción de Señal/fisiología , Proteínas no Estructurales Virales/metabolismo , Animales , Drosophila , Humanos , Inmunohistoquímica , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Ratones , Ratones Endogámicos C57BL
16.
Immunity ; 31(1): 145-57, 2009 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-19604493

RESUMEN

Although deficient CD8(+) T cell responses have long been associated with chronic viral infections, the underlying mechanisms are still unclear. Here we report that sustained transforming growth factor-beta (TGF-beta) expression and phosphorylation of its signaling mediator, Smad-2, were distinctive features of virus-specific CD8(+) T cells during chronic versus acute viral infections in vivo. The result was TGF-beta-dependent apoptosis of virus-specific CD8(+) T cells that related to upregulation of the proapoptotic protein Bim during chronic infection. Moreover, selective attenuation of TGF-beta signaling in T cells increased the numbers and multiple functions of antiviral CD8(+) T cells and enabled rapid eradication of the persistence-prone virus and memory generation. Finally, we found that cell-intrinsic TGF-beta signaling was responsible for virus-specific-CD8(+) T cell apoptosis and decreased numbers but was not necessary for their functional exhaustion. Our findings reveal persisting TGF-beta-Smad signaling as a hallmark and key regulator of CD8(+) T cell responses during chronic viral infections in vivo.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de la Membrana/inmunología , Proteínas Proto-Oncogénicas/inmunología , Proteína Smad2/inmunología , Factor de Crecimiento Transformador beta/inmunología , Traslado Adoptivo , Animales , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/metabolismo , Infecciones por Arenaviridae/virología , Proteína 11 Similar a Bcl2 , Linfocitos T CD8-positivos/metabolismo , Depleción Linfocítica , Virus de la Coriomeningitis Linfocítica/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosforilación/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/inmunología , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
17.
J Immunol ; 196(4): 1900-9, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26773151

RESUMEN

Type I IFNs (IFN-I) are key innate mediators that create a profound antiviral state and orchestrate the activation of almost all immune cells. Plasmacytoid dendritic cells (pDCs) are the most powerful IFN-I-producing cells and play important roles during viral infections, cancer, and autoimmune diseases. By comparing gene expression profiles of murine pDCs and conventional DCs, we found that CD28, a prototypic T cell stimulatory receptor, was highly expressed in pDCs. Strikingly, CD28 acted as a negative regulator of pDC IFN-I production upon TLR stimulation but did not affect pDC survival or maturation. Importantly, cell-intrinsic CD28 expression restrained pDC (and systemic) IFN-I production during in vivo RNA and DNA viral infections, limiting antiviral responses and enhancing viral growth early after exposure. Finally, CD28 also downregulated IFN-I response upon skin injury. Our study identified a new pDC regulatory mechanism by which the same CD28 molecule that promotes stimulation in most cells that express it is co-opted to negatively regulate pDC IFN-I production and limit innate responses.


Asunto(s)
Antígenos CD28/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Animales , Células Dendríticas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma
18.
J Immunol ; 195(3): 1071-81, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26085685

RESUMEN

The IL-6 cytokine family utilizes the common signal transduction molecule gp130, which can mediate a diverse range of outcomes. To clarify the role of gp130 signaling in vivo during acute viral infection, we infected Cd4-cre Il6st(fl/fl) mice, in which gp130 is conditionally ablated in T cells, with acute lymphocytic choriomeningitis virus. We found that by day 12, but not at day 8, after infection the number of virus-specific CD4(+) T cells was reduced in the absence of gp130, and this was sustained for up to 2 mo postinfection. Additionally, gp130-deficient T follicular helper cells had lower expression of Maf, IL-21, and ICOS, and this was accompanied by a reduction in the proportion of germinal center B cells and plasmablasts. Remarkably, at 2 mo postinfection the proportion of IgG2a/c(+) memory B cells and the systemic levels of lymphocytic choriomeningitis virus-specific IgG2 Abs were dramatically decreased, whereas there was a corresponding increase in IgG1(+) memory B cells and virus-specific IgG1 Abs. In the same animals gp130-deficient virus-specific CD8(+) T cells showed a reduced proportion of memory cells, which expressed lower levels of Tcf7, and displayed diminished recall responses on secondary infection. Mixed bone marrow chimeras revealed that the aforementioned gp130 effects on CD4(+) T cells were cell intrinsic. Overall, our data show that gp130 signaling in T cells influences the quantity and quality of long-lasting CD4(+) T cell responses as well as CD8(+) T cell- and Ab-mediated immunity after acute viral infection.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Receptor gp130 de Citocinas/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/citología , Receptor gp130 de Citocinas/genética , Centro Germinal/citología , Centro Germinal/inmunología , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Inmunoglobulina G/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/biosíntesis , Interleucina-6/inmunología , Interleucinas/biosíntesis , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-maf/biosíntesis , Transducción de Señal/inmunología
19.
J Virol ; 89(6): 3343-55, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25589641

RESUMEN

UNLABELLED: Rapid innate responses to viral encounters are crucial to shaping the outcome of infection, from viral clearance to persistence. Transforming growth factor ß (TGF-ß) is a potent immune suppressor that is upregulated early upon viral infection and maintained during chronic infections in both mice and humans. However, the role of TGF-ß signaling in regulating individual cell types in vivo is still unclear. Using infections with two different persistent viruses, murine cytomegalovirus (MCMV) and lymphocytic choriomeningitis virus (LCMV; Cl13), in their natural rodent host, we observed that TGF-ß signaling on dendritic cells (DCs) did not dampen DC maturation or cytokine production in the early stages of chronic infection with either virus in vivo. In contrast, TGF-ß signaling prior to (but not during) chronic viral infection directly restricted the natural killer (NK) cell number and effector function. This restriction likely compromised both the early control of and host survival upon MCMV infection but not the long-term control of LCMV infection. These data highlight the context and timing of TGF-ß signaling on different innate cells that contribute to the early host response, which ultimately influences the outcome of chronic viral infection in vivo. IMPORTANCE: In vivo host responses to pathogens are complex processes involving the cooperation of many different immune cells migrating to specific tissues over time, but these events cannot be replicated in vitro. Viruses causing chronic infections are able to subvert this immune response and represent a human health burden. Here we used two well-characterized viruses that are able to persist in their natural mouse host to dissect the role of the suppressive molecule TGF-ß in dampening host responses to infection in vivo. This report presents information that allows an increased understanding of long-studied TGF-ß signaling by examining its direct effect on different immune cells that are activated very early after in vivo viral infection and may aid with the development of new antiviral therapeutic strategies.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Herpesviridae/veterinaria , Células Asesinas Naturales/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Muromegalovirus/fisiología , Enfermedades de los Roedores/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Citocinas/inmunología , Femenino , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Humanos , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades de los Roedores/genética , Enfermedades de los Roedores/virología , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
20.
J Immunol ; 192(5): 2133-42, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24493818

RESUMEN

Dendritic cells (DCs) are potent APCs essential for initiating adaptive immunity. Following pathogen exposure, trafficking of DCs to lymph nodes (LNs) through afferent lymphatic vessels constitutes a crucial step in the execution of their functions. The mechanisms regulating this process are poorly understood, although the involvement of certain chemokines in this process has recently been reported. In this study, we demonstrate that genetically altering the fine structure (N-sulfation) of heparan sulfate (HS) specifically in mouse lymphatic endothelium significantly reduces DC trafficking to regional LNs in vivo. Moreover, this alteration had the unique functional consequence of reducing CD8(+) T cell proliferative responses in draining LNs in an ovalbumin immunization model. Mechanistic studies suggested that lymphatic endothelial HS regulates multiple steps during DC trafficking, including optimal presentation of chemokines on the surface of DCs, thus acting as a co-receptor that may function "in trans" to mediate chemokine receptor binding. This study not only identifies novel glycan-mediated mechanisms that regulate lymphatic DC trafficking, but it also validates the fine structure of lymphatic vascular-specific HS as a novel molecular target for strategies aiming to modulate DC behavior and/or alter pathologic T cell responses in lymph nodes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Células Dendríticas/inmunología , Endotelio Linfático/inmunología , Heparitina Sulfato/inmunología , Ganglios Linfáticos/inmunología , Animales , Linfocitos T CD8-positivos/citología , Movimiento Celular/genética , Quimiocinas/genética , Quimiocinas/inmunología , Células Dendríticas/citología , Endotelio Linfático/citología , Heparitina Sulfato/genética , Humanos , Ganglios Linfáticos/citología , Ratones , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA