Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(5): 1214-1231.e16, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33636133

RESUMEN

Although enteric helminth infections modulate immunity to mucosal pathogens, their effects on systemic microbes remain less established. Here, we observe increased mortality in mice coinfected with the enteric helminth Heligmosomoides polygyrus bakeri (Hpb) and West Nile virus (WNV). This enhanced susceptibility is associated with altered gut morphology and transit, translocation of commensal bacteria, impaired WNV-specific T cell responses, and increased virus infection in the gastrointestinal tract and central nervous system. These outcomes were due to type 2 immune skewing, because coinfection in Stat6-/- mice rescues mortality, treatment of helminth-free WNV-infected mice with interleukin (IL)-4 mirrors coinfection, and IL-4 receptor signaling in intestinal epithelial cells mediates the susceptibility phenotypes. Moreover, tuft cell-deficient mice show improved outcomes with coinfection, whereas treatment of helminth-free mice with tuft cell-derived cytokine IL-25 or ligand succinate worsens WNV disease. Thus, helminth activation of tuft cell-IL-4-receptor circuits in the gut exacerbates infection and disease of a neurotropic flavivirus.


Asunto(s)
Coinfección , Nematospiroides dubius/fisiología , Transducción de Señal , Infecciones por Strongylida/patología , Virus del Nilo Occidental/fisiología , Animales , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Mucosa Intestinal/parasitología , Mucosa Intestinal/virología , Ratones , Ratones Endogámicos C57BL , Neuronas/parasitología , Neuronas/virología , Receptores de Interleucina-4/metabolismo , Factor de Transcripción STAT6/genética , Índice de Severidad de la Enfermedad , Infecciones por Strongylida/parasitología
2.
Cell ; 178(5): 1088-1101.e15, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442402

RESUMEN

Mammals evolved in the face of fluctuating food availability. How the immune system adapts to transient nutritional stress remains poorly understood. Here, we show that memory T cells collapsed in secondary lymphoid organs in the context of dietary restriction (DR) but dramatically accumulated within the bone marrow (BM), where they adopted a state associated with energy conservation. This response was coordinated by glucocorticoids and associated with a profound remodeling of the BM compartment, which included an increase in T cell homing factors, erythropoiesis, and adipogenesis. Adipocytes, as well as CXCR4-CXCL12 and S1P-S1P1R interactions, contributed to enhanced T cell accumulation in BM during DR. Memory T cell homing to BM during DR was associated with enhanced protection against infections and tumors. Together, this work uncovers a fundamental host strategy to sustain and optimize immunological memory during nutritional challenges that involved a temporal and spatial reorganization of the memory pool within "safe haven" compartments.


Asunto(s)
Médula Ósea/metabolismo , Memoria Inmunológica , Animales , Médula Ósea/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Restricción Calórica/veterinaria , Línea Celular Tumoral , Quimiocina CXCL12/metabolismo , Dieta Reductora/veterinaria , Metabolismo Energético , Regulación de la Expresión Génica , Glucocorticoides , Melanoma Experimental/mortalidad , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores CXCR4/metabolismo , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
3.
Nat Immunol ; 22(1): 41-52, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33139915

RESUMEN

Personalized cancer vaccines are a promising approach for inducing T cell immunity to tumor neoantigens. Using a self-assembling nanoparticle vaccine that links neoantigen peptides to a Toll-like receptor 7/8 agonist (SNP-7/8a), we show how the route and dose alter the magnitude and quality of neoantigen-specific CD8+ T cells. Intravenous vaccination (SNP-IV) induced a higher proportion of TCF1+PD-1+CD8+ T cells as compared to subcutaneous immunization (SNP-SC). Single-cell RNA sequencing showed that SNP-IV induced stem-like genes (Tcf7, Slamf6, Xcl1) whereas SNP-SC enriched for effector genes (Gzmb, Klrg1, Cx3cr1). Stem-like cells generated by SNP-IV proliferated and differentiated into effector cells upon checkpoint blockade, leading to superior antitumor response as compared to SNP-SC in a therapeutic model. The duration of antigen presentation by dendritic cells controlled the magnitude and quality of CD8+ T cells. These data demonstrate how to optimize antitumor immunity by modulating vaccine parameters for specific generation of effector or stem-like CD8+ T cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Factor Nuclear 1-alfa del Hepatocito/análisis , Nanopartículas , Animales , Presentación de Antígeno , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Femenino , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Vacunación
4.
Cell ; 171(5): 1015-1028.e13, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29056339

RESUMEN

Laboratory mice, while paramount for understanding basic biological phenomena, are limited in modeling complex diseases of humans and other free-living mammals. Because the microbiome is a major factor in mammalian physiology, we aimed to identify a naturally evolved reference microbiome to better recapitulate physiological phenomena relevant in the natural world outside the laboratory. Among 21 distinct mouse populations worldwide, we identified a closely related wild relative to standard laboratory mouse strains. Its bacterial gut microbiome differed significantly from its laboratory mouse counterpart and was transferred to and maintained in laboratory mice over several generations. Laboratory mice reconstituted with natural microbiota exhibited reduced inflammation and increased survival following influenza virus infection and improved resistance against mutagen/inflammation-induced colorectal tumorigenesis. By demonstrating the host fitness-promoting traits of natural microbiota, our findings should enable the discovery of protective mechanisms relevant in the natural world and improve the modeling of complex diseases of free-living mammals. VIDEO ABSTRACT.


Asunto(s)
Microbioma Gastrointestinal , Ratones/clasificación , Ratones/microbiología , Animales , Animales de Laboratorio , Animales Salvajes , Carcinogénesis/inmunología , Resistencia a la Enfermedad , Femenino , Masculino , Maryland , Ratones/inmunología , Ratones Endogámicos C57BL , Peromyscus , Virosis/inmunología
5.
Nat Immunol ; 20(5): 602-612, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30886418

RESUMEN

Despite intense interest in antiviral T cell priming, the routes by which virions move in lymph nodes (LNs) are imperfectly understood. Current models fail to explain how virus-infected cells rapidly appear within the LN interior after viral infection. To better understand virion trafficking in the LN, we determined the locations of virions and infected cells after administration to mice of vaccinia virus or Zika virus. Notably, many rapidly infected cells in the LN interior were adjacent to LN conduits. Through the use of confocal and electron microscopy, we clearly visualized virions within conduits. Functionally, CD8+ T cells rapidly and preferentially associated with vaccinia virus-infected cells in the LN paracortex, which led to T cell activation in the LN interior. These results reveal that it is possible for even large virions to flow through LN conduits and infect dendritic cells within the T cell zone to prime CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Virión/inmunología , Animales , Linfocitos T CD8-positivos/virología , Femenino , Ganglios Linfáticos/ultraestructura , Ganglios Linfáticos/virología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Virus Vaccinia/inmunología , Virus Vaccinia/fisiología , Virión/fisiología , Virión/ultraestructura , Virosis/inmunología , Virosis/virología , Virus Zika/inmunología , Virus Zika/fisiología
6.
Cell ; 166(5): 1117-1131.e14, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27565342

RESUMEN

Cancer cells must evade immune responses at distant sites to establish metastases. The lung is a frequent site for metastasis. We hypothesized that lung-specific immunoregulatory mechanisms create an immunologically permissive environment for tumor colonization. We found that T-cell-intrinsic expression of the oxygen-sensing prolyl-hydroxylase (PHD) proteins is required to maintain local tolerance against innocuous antigens in the lung but powerfully licenses colonization by circulating tumor cells. PHD proteins limit pulmonary type helper (Th)-1 responses, promote CD4(+)-regulatory T (Treg) cell induction, and restrain CD8(+) T cell effector function. Tumor colonization is accompanied by PHD-protein-dependent induction of pulmonary Treg cells and suppression of IFN-γ-dependent tumor clearance. T-cell-intrinsic deletion or pharmacological inhibition of PHD proteins limits tumor colonization of the lung and improves the efficacy of adoptive cell transfer immunotherapy. Collectively, PHD proteins function in T cells to coordinate distinct immunoregulatory programs within the lung that are permissive to cancer metastasis. PAPERCLIP.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Pulmón/inmunología , Oxígeno/metabolismo , Prolil Hidroxilasas/metabolismo , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/enzimología , Glucólisis/inmunología , Interferón gamma/inmunología , Pulmón/patología , Neoplasias Pulmonares/terapia , Activación de Linfocitos , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Neuropilina-1/metabolismo , Prolil Hidroxilasas/genética , Linfocitos T Reguladores/enzimología , Células TH1/enzimología , Células TH1/inmunología
7.
Nat Immunol ; 19(2): 173-182, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29311694

RESUMEN

CD8+ T cell immunosurveillance dynamics influence the outcome of intracellular infections and cancer. Here we used two-photon intravital microscopy to visualize the responses of CD8+ resident memory T cells (TRM cells) within the reproductive tracts of live female mice. We found that mucosal TRM cells were highly motile, but paused and underwent in situ division after local antigen challenge. TRM cell reactivation triggered the recruitment of recirculating memory T cells that underwent antigen-independent TRM cell differentiation in situ. However, the proliferation of pre-existing TRM cells dominated the local mucosal recall response and contributed most substantially to the boosted secondary TRM cell population. We observed similar results in skin. Thus, TRM cells can autonomously regulate the expansion of local immunosurveillance independently of central memory or proliferation in lymphoid tissue.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Mucosa/inmunología , Memoria Inmunológica/inmunología , Vigilancia Inmunológica/inmunología , Membrana Mucosa/inmunología , Animales , Femenino , Microscopía Intravital , Ratones , Membrana Mucosa/citología , Piel/inmunología
8.
Immunity ; 54(2): 276-290.e5, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33434494

RESUMEN

The oropharyngeal mucosa serves as a perpetual pathogen entry point and a critical site for viral replication and spread. Here, we demonstrate that type 1 innate lymphoid cells (ILC1s) were the major immune force providing early protection during acute oral mucosal viral infection. Using intravital microscopy, we show that ILC1s populated and patrolled the uninfected labial mucosa. ILC1s produced interferon-γ (IFN-γ) in the absence of infection, leading to the upregulation of key antiviral genes, which were downregulated in uninfected animals upon genetic ablation of ILC1s or antibody-based neutralization of IFN-γ. Thus, tonic IFN-γ production generates increased oral mucosal viral resistance even before infection. Our results demonstrate barrier-tissue protection through tissue surveillance in the absence of rearranged-antigen receptors and the induction of an antiviral state during homeostasis. This aspect of ILC1 biology raises the possibility that these cells do not share true functional redundancy with other tissue-resident lymphocytes.


Asunto(s)
Interferón gamma/metabolismo , Linfocitos/inmunología , Orofaringe/inmunología , Mucosa Respiratoria/inmunología , Virus Vaccinia/fisiología , Vaccinia/inmunología , Animales , Células Cultivadas , Resistencia a la Enfermedad , Humanos , Inmunidad Innata , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética , Células TH1/inmunología
9.
Nat Immunol ; 18(4): 456-463, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28192417

RESUMEN

Immunodominance (ID) defines the hierarchical immune response to competing antigens in complex immunogens. Little is known regarding B cell and antibody ID despite its importance in immunity to viruses and other pathogens. We show that B cells and serum antibodies from inbred mice demonstrate a reproducible ID hierarchy to the five major antigenic sites in the influenza A virus hemagglutinin globular domain. The hierarchy changed as the immune response progressed, and it was dependent on antigen formulation and delivery. Passive antibody transfer and sequential infection experiments demonstrated 'original antigenic suppression', a phenomenon in which antibodies suppress memory responses to the priming antigenic site. Our study provides a template for attaining deeper understanding of antibody ID to viruses and other complex immunogens.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Interacciones Huésped-Patógeno/inmunología , Epítopos Inmunodominantes/inmunología , Virosis/inmunología , Virus/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Antígenos Virales/química , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antecedentes Genéticos , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Interacciones Huésped-Patógeno/genética , Inmunización , Epítopos Inmunodominantes/química , Memoria Inmunológica , Virus de la Influenza A/inmunología , Ganglios Linfáticos/inmunología , Ratones , Modelos Moleculares , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Conformación Proteica , Virosis/genética , Virosis/virología
10.
PLoS Pathog ; 20(7): e1012339, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38950078

RESUMEN

The regulation of inflammatory responses and pulmonary disease during SARS-CoV-2 infection is incompletely understood. Here we examine the roles of the prototypic pro- and anti-inflammatory cytokines IFNγ and IL-10 using the rhesus macaque model of mild COVID-19. We find that IFNγ drives the development of 18fluorodeoxyglucose (FDG)-avid lesions in the lungs as measured by PET/CT imaging but is not required for suppression of viral replication. In contrast, IL-10 limits the duration of acute pulmonary lesions, serum markers of inflammation and the magnitude of virus-specific T cell expansion but does not impair viral clearance. We also show that IL-10 induces the subsequent differentiation of virus-specific effector T cells into CD69+CD103+ tissue resident memory cells (Trm) in the airways and maintains Trm cells in nasal mucosal surfaces, highlighting an unexpected role for IL-10 in promoting airway memory T cells during SARS-CoV-2 infection of macaques.


Asunto(s)
COVID-19 , Memoria Inmunológica , Interleucina-10 , Macaca mulatta , Células T de Memoria , SARS-CoV-2 , Animales , Interleucina-10/inmunología , Interleucina-10/metabolismo , COVID-19/inmunología , SARS-CoV-2/inmunología , Células T de Memoria/inmunología , Células T de Memoria/metabolismo , Memoria Inmunológica/inmunología , Pulmón/inmunología , Pulmón/virología , Pulmón/patología , Modelos Animales de Enfermedad , Interferón gamma/metabolismo , Interferón gamma/inmunología , Linfocitos T/inmunología
11.
Immunity ; 46(1): 13-14, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28099861

RESUMEN

Although CD8+ T cells provide protection against many viral infections, their role in Zika virus (ZIKV) immunity has not been extensively examined. In a recent issue of Cell Host & Microbe, Elong Ngono et al. (2017) define antigenic epitopes determining CD8+ T cell immunity in murine models of ZIKV infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Virus Zika , Animales , Ratones , Infección por el Virus Zika
12.
Immunol Rev ; 306(1): 200-217, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34796538

RESUMEN

The past decade has seen near continual global public health crises caused by emerging viral infections. Extraordinary increases in our knowledge of the mechanisms underlying successful antiviral immune responses in animal models and during human infection have accompanied these viral outbreaks. Keeping pace with the rapidly advancing field of viral immunology, innovations in microscopy have afforded a previously unseen view of viral infection occurring in real-time in living animals. Here, we review the contribution of intravital imaging to our understanding of cell-mediated immune responses to viral infections, with a particular focus on studies that visualize the antiviral effector cells responding to infection as well as virus-infected cells. We discuss methods to visualize viral infection in vivo using intravital microscopy (IVM) and significant findings arising through the application of IVM to viral infection. Collectively, these works underscore the importance of developing a comprehensive spatial understanding of the relationships between immune effectors and virus-infected cells and how this has enabled unique discoveries about virus/host interactions and antiviral effector cell biology.


Asunto(s)
Virosis , Virus , Animales , Antivirales , Humanos , Inmunidad Celular , Microscopía Intravital/métodos
13.
EMBO J ; 40(22): e108966, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34618370

RESUMEN

Viremia in the vertebrate host is a major determinant of arboviral reservoir competency, transmission efficiency, and disease severity. However, immune mechanisms that control arboviral viremia are poorly defined. Here, we identify critical roles for the scavenger receptor MARCO in controlling viremia during arthritogenic alphavirus infections in mice. Following subcutaneous inoculation, arthritogenic alphavirus particles drain via the lymph and are rapidly captured by MARCO+ lymphatic endothelial cells (LECs) in the draining lymph node (dLN), limiting viral spread to the bloodstream. Upon reaching the bloodstream, alphavirus particles are cleared from the circulation by MARCO-expressing Kupffer cells in the liver, limiting viremia and further viral dissemination. MARCO-mediated accumulation of alphavirus particles in the draining lymph node and liver is an important host defense mechanism as viremia and viral tissue burdens are elevated in MARCO-/- mice and disease is more severe. In contrast to prior studies implicating a key role for lymph node macrophages in limiting viral dissemination, these findings exemplify a previously unrecognized arbovirus-scavenging role for lymphatic endothelial cells and improve our mechanistic understanding of viremia control during arthritogenic alphavirus infection.


Asunto(s)
Infecciones por Alphavirus/virología , Ganglios Linfáticos/citología , Receptores Inmunológicos/metabolismo , Viremia/patología , Alphavirus/patogenicidad , Animales , Fiebre Chikungunya/genética , Fiebre Chikungunya/virología , Células Endoteliales/virología , Interacciones Huésped-Patógeno , Macrófagos del Hígado/virología , Ganglios Linfáticos/virología , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , ARN Viral/metabolismo , Receptores Inmunológicos/genética , Análisis de la Célula Individual , Viremia/virología
14.
J Virol ; 97(12): e0127223, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38009914

RESUMEN

IMPORTANCE: Human poxvirus infections have caused significant public health burdens both historically and recently during the unprecedented global Mpox virus outbreak. Although vaccinia virus (VACV) infection of mice is a commonly used model to explore the anti-poxvirus immune response, little is known about the metabolic changes that occur in vivo during infection. We hypothesized that the metabolome of VACV-infected skin would reflect the increased energetic requirements of both virus-infected cells and immune cells recruited to sites of infection. Therefore, we profiled whole VACV-infected skin using untargeted mass spectrometry to define the metabolome during infection, complementing these experiments with flow cytometry and transcriptomics. We identified specific metabolites, including nucleotides, itaconic acid, and glutamine, that were differentially expressed during VACV infection. Together, this study offers insight into both virus-specific and immune-mediated metabolic pathways that could contribute to the clearance of cutaneous poxvirus infection.


Asunto(s)
Reprogramación Metabólica , Metaboloma , Piel , Virus Vaccinia , Vaccinia , Animales , Ratones , Citometría de Flujo , Perfilación de la Expresión Génica , Glutamina/metabolismo , Espectrometría de Masas , Nucleótidos/metabolismo , Piel/inmunología , Piel/metabolismo , Piel/virología , Vaccinia/inmunología , Vaccinia/metabolismo , Vaccinia/virología , Virus Vaccinia/metabolismo , Carga Viral
15.
Immunity ; 42(3): 524-37, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25769612

RESUMEN

CD8(+) T cells play a critical role in limiting peripheral virus replication, yet how they locate virus-infected cells within tissues is unknown. Here, we have examined the environmental signals that CD8(+) T cells use to localize and eliminate virus-infected skin cells. Epicutaneous vaccinia virus (VV) infection, mimicking human smallpox vaccination, greatly increased expression of the CXCR3 chemokine receptor ligands CXCL9 and CXCL10 in VV-infected skin. Despite normal T cell numbers in the skin, Cxcr3(-/-) mice exhibited dramatically impaired CD8(+)-T-cell-dependent virus clearance. Intravital microscopy revealed that Cxcr3(-/-) T cells were markedly deficient in locating, engaging, and killing virus-infected cells. Further, transfer of wild-type CD8(+) T cells restored viral clearance in Cxcr3(-/-) animals. These findings demonstrate a function for CXCR3 in enhancing the ability of tissue-localized CD8(+) T cells to locate virus-infected cells and thereby exert anti-viral effector functions.


Asunto(s)
Queratinocitos/inmunología , Infecciones por Poxviridae/inmunología , Receptores CXCR3/inmunología , Piel/inmunología , Linfocitos T Citotóxicos/inmunología , Virus Vaccinia/inmunología , Traslado Adoptivo , Animales , Movimiento Celular , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL9/genética , Quimiocina CXCL9/inmunología , Femenino , Regulación de la Expresión Génica , Humanos , Queratinocitos/patología , Queratinocitos/virología , Ratones Transgénicos , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/patología , Infecciones por Poxviridae/virología , Receptores CXCR3/deficiencia , Receptores CXCR3/genética , Transducción de Señal , Piel/patología , Piel/virología , Linfocitos T Citotóxicos/patología , Linfocitos T Citotóxicos/trasplante , Carga Viral
16.
PLoS Pathog ; 16(1): e1008292, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31999809

RESUMEN

Humoral immune responses initiate in the lymph node draining the site of viral infection (dLN). Some viruses subvert LN B cell activation; however, our knowledge of viral hindrance of B cell responses of important human pathogens is lacking. Here, we define mechanisms whereby chikungunya virus (CHIKV), a mosquito-transmitted RNA virus that causes outbreaks of acute and chronic arthritis in humans, hinders dLN antiviral B cell responses. Infection of WT mice with pathogenic, but not acutely cleared CHIKV, induced MyD88-dependent recruitment of monocytes and neutrophils to the dLN. Blocking this influx improved lymphocyte accumulation, dLN organization, and CHIKV-specific B cell responses. Both inducible nitric oxide synthase (iNOS) and the phagocyte NADPH oxidase (Nox2) contributed to impaired dLN organization and function. Infiltrating monocytes expressed iNOS through a local IRF5- and IFNAR1-dependent pathway that was partially TLR7-dependent. Together, our data suggest that pathogenic CHIKV triggers the influx and activation of monocytes and neutrophils in the dLN that impairs virus-specific B cell responses.


Asunto(s)
Linfocitos B/inmunología , Fiebre Chikungunya/inmunología , Factores Reguladores del Interferón/inmunología , Monocitos/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , NADPH Oxidasa 2/inmunología , Neutrófilos/inmunología , Óxido Nítrico Sintasa de Tipo II/inmunología , Animales , Fiebre Chikungunya/virología , Virus Chikungunya/fisiología , Humanos , Factores Reguladores del Interferón/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , NADPH Oxidasa 2/genética , Óxido Nítrico Sintasa de Tipo II/genética
17.
Proc Natl Acad Sci U S A ; 116(27): 13474-13479, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31213541

RESUMEN

A major obstacle to vaccination against antigenically variable viruses is skewing of antibody responses to variable immunodominant epitopes. For influenza virus hemagglutinin (HA), the immunodominance of the variable head impairs responses to the highly conserved stem. Here, we show that head immunodominance depends on the physical attachment of head to stem. Stem immunogenicity is enhanced by immunizing with stem-only constructs or by increasing local HA concentration in the draining lymph node. Surprisingly, coimmunization of full-length HA and stem alters stem-antibody class switching. Our findings delineate strategies for overcoming immunodominance, with important implications for human vaccination.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Epítopos/inmunología , Hemaglutininas/inmunología , Epítopos Inmunodominantes/inmunología , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Animales , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Femenino , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Células Madre/inmunología
18.
J Immunol ; 203(4): 775-781, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31383748

RESUMEN

Scientists have long valued the power of in vivo observation to answer fundamental biological questions. Over the last 20 years, the application and evolution of intravital microscopy (IVM) has vastly increased our ability to directly visualize immune responses as they are occurring in vivo after infection or immunization. Many IVM strategies employ a strong multiphoton laser that penetrates deeply into the tissues of living, anesthetized mice, allowing the precise tracking of the movement of cells as they navigate complex tissue environments. In the realm of viral infections, IVM has been applied to better understand many critical phases of effector T cell responses, from activation in the draining lymph node, to the execution of effector functions, and finally to the development of tissue-resident memory. In this review, we discuss seminal studies incorporating IVM that have advanced our understanding of the biology of antiviral CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Microscopía Intravital/métodos , Virosis/inmunología , Animales , Humanos , Ratones
19.
Nat Immunol ; 9(2): 155-65, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18193049

RESUMEN

It is uncertain how antiviral lymphocytes are activated in draining lymph nodes, the site where adaptive immune responses are initiated. Here, using intravital microscopy we show that after infection of mice with vaccinia virus (a large DNA virus) or vesicular stomatitis virus (a small RNA virus), virions drained to the lymph node and infected cells residing just beneath the subcapsular sinus. Naive CD8+ T cells rapidly migrated to infected cells in the peripheral interfollicular region and then formed tight interactions with dendritic cells, leading to complete T cell activation. Thus, antigen presentation at the lymph node periphery, not at lymphocyte exit sites in deeper lymph node venules, as dogma dictates, has a dominant function in antiviral CD8+ T cell activation.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Animales , Células Presentadoras de Antígenos/inmunología , Movimiento Celular , Células Dendríticas/inmunología , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Transgénicos , Virus Vaccinia/inmunología , Vesiculovirus/inmunología
20.
J Immunol ; 201(4): 1222-1228, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30012850

RESUMEN

Probing the limits of CD8+ T cell immunosurveillance, we inserted the SIINFEKL peptide into influenza A virus (IAV)-negative strand gene segments. Although IAV genomic RNA is considered noncoding, there is a conserved, relatively long open reading frame present in segment 8, encoding a potential protein termed NEG8. The biosynthesis of NEG8 from IAV has yet to be demonstrated. Although we failed to detect NEG8 protein expression in IAV-infected mouse cells, cell surface Kb-SIINFEKL complexes are generated when SIINFEKL is genetically appended to the predicted C terminus of NEG8, as shown by activation of OT-I T cells in vitro and in vivo. Moreover, recombinant IAV encoding of SIINFEKL embedded in the negative strand of the neuraminidase-stalk coding sequence also activates OT-I T cells in mice. Together, our findings demonstrate both the translation of sequences on the negative strand of a single-stranded RNA virus and its relevance in antiviral immunosurveillance.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vigilancia Inmunológica/inmunología , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , ARN Viral/inmunología , Animales , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/inmunología , Biosíntesis de Proteínas/fisiología , ARN Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA