Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 37(2): 109803, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34644581

RESUMEN

Human respiratory syncytial virus (RSV) is a common cause of lower respiratory tract infections in the pediatric, elderly, and immunocompromised individuals. RSV non-structural protein NS1 is a known cytosolic immune antagonist, but how NS1 modulates host responses remains poorly defined. Here, we observe NS1 partitioning into the nucleus of RSV-infected cells, including the human airway epithelium. Nuclear NS1 coimmunoprecipitates with Mediator complex and is chromatin associated. Chromatin-immunoprecipitation demonstrates enrichment of NS1 that overlaps Mediator and transcription factor binding within the promoters and enhancers of differentially expressed genes during RSV infection. Mutation of the NS1 C-terminal helix reduces NS1 impact on host gene expression. These data suggest that nuclear NS1 alters host responses to RSV infection by binding at regulatory elements of immune response genes and modulating host gene transcription. Our study identifies another layer of regulation by virally encoded proteins that shapes host response and impacts immunity to RSV.


Asunto(s)
Núcleo Celular/metabolismo , Cromatina/metabolismo , Células Dendríticas/metabolismo , Células Epiteliales/metabolismo , Pulmón/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitial Respiratorio Humano/metabolismo , Transcripción Genética , Proteínas no Estructurales Virales/metabolismo , Células A549 , Animales , Sitios de Unión , Núcleo Celular/virología , Cromatina/genética , Cromatina/virología , Células Dendríticas/virología , Células Epiteliales/virología , Femenino , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Pulmón/virología , Complejo Mediador/genética , Complejo Mediador/metabolismo , Ratones Endogámicos BALB C , Regiones Promotoras Genéticas , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/patogenicidad , Proteínas no Estructurales Virales/genética
2.
Cell ; 183(3): 818-834.e13, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33038342

RESUMEN

Many approaches to identify therapeutically relevant neoantigens couple tumor sequencing with bioinformatic algorithms and inferred rules of tumor epitope immunogenicity. However, there are no reference data to compare these approaches, and the parameters governing tumor epitope immunogenicity remain unclear. Here, we assembled a global consortium wherein each participant predicted immunogenic epitopes from shared tumor sequencing data. 608 epitopes were subsequently assessed for T cell binding in patient-matched samples. By integrating peptide features associated with presentation and recognition, we developed a model of tumor epitope immunogenicity that filtered out 98% of non-immunogenic peptides with a precision above 0.70. Pipelines prioritizing model features had superior performance, and pipeline alterations leveraging them improved prediction performance. These findings were validated in an independent cohort of 310 epitopes prioritized from tumor sequencing data and assessed for T cell binding. This data resource enables identification of parameters underlying effective anti-tumor immunity and is available to the research community.


Asunto(s)
Antígenos de Neoplasias/inmunología , Epítopos/inmunología , Neoplasias/inmunología , Alelos , Presentación de Antígeno/inmunología , Estudios de Cohortes , Humanos , Péptidos/inmunología , Receptor de Muerte Celular Programada 1 , Reproducibilidad de los Resultados
3.
Nature ; 574(7780): 696-701, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31645760

RESUMEN

The ability of the immune system to eliminate and shape the immunogenicity of tumours defines the process of cancer immunoediting1. Immunotherapies such as those that target immune checkpoint molecules can be used to augment immune-mediated elimination of tumours and have resulted in durable responses in patients with cancer that did not respond to previous treatments. However, only a subset of patients benefit from immunotherapy and more knowledge about what is required for successful treatment is needed2-4. Although the role of tumour neoantigen-specific CD8+ T cells in tumour rejection is well established5-9, the roles of other subsets of T cells have received less attention. Here we show that spontaneous and immunotherapy-induced anti-tumour responses require the activity of both tumour-antigen-specific CD8+ and CD4+ T cells, even in tumours that do not express major histocompatibility complex (MHC) class II molecules. In addition, the expression of MHC class II-restricted antigens by tumour cells is required at the site of successful rejection, indicating that activation of CD4+ T cells must also occur in the tumour microenvironment. These findings suggest that MHC class II-restricted neoantigens have a key function in the anti-tumour response that is nonoverlapping with that of MHC class I-restricted neoantigens and therefore needs to be considered when identifying patients who will most benefit from immunotherapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Neoplasias Experimentales/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunoterapia , Ratones , Neoplasias Experimentales/terapia
4.
J Virol ; 94(1)2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31619554

RESUMEN

Type I interferons (IFNs) are key mediators of the innate immune response. Although members of this family of cytokines signal through a single shared receptor, biochemical and functional variation exists in response to different IFN subtypes. While previous work has demonstrated that type I IFNs are essential to control infection by chikungunya virus (CHIKV), a globally emerging alphavirus, the contributions of individual IFN subtypes remain undefined. To address this question, we evaluated CHIKV pathogenesis in mice lacking IFN-ß (IFN-ß knockout [IFN-ß-KO] mice or mice treated with an IFN-ß-blocking antibody) or IFN-α (IFN regulatory factor 7 knockout [IRF7-KO] mice or mice treated with a pan-IFN-α-blocking antibody). Mice lacking either IFN-α or IFN-ß developed severe clinical disease following infection with CHIKV, with a marked increase in foot swelling compared to wild-type mice. Virological analysis revealed that mice lacking IFN-α sustained elevated infection in the infected ankle and in distant tissues. In contrast, IFN-ß-KO mice displayed minimal differences in viral burdens within the ankle or at distal sites and instead had an altered cellular immune response. Mice lacking IFN-ß had increased neutrophil infiltration into musculoskeletal tissues, and depletion of neutrophils in IFN-ß-KO but not IRF7-KO mice mitigated musculoskeletal disease caused by CHIKV. Our findings suggest disparate roles for the IFN subtypes during CHIKV infection, with IFN-α limiting early viral replication and dissemination and IFN-ß modulating neutrophil-mediated inflammation.IMPORTANCE Type I interferons (IFNs) possess a range of biological activity and protect against a number of viruses, including alphaviruses. Despite signaling through a shared receptor, there are established biochemical and functional differences among the IFN subtypes. The significance of our research is in demonstrating that IFN-α and IFN-ß both have protective roles during acute chikungunya virus (CHIKV) infection but do so by distinct mechanisms. IFN-α limits CHIKV replication and dissemination, whereas IFN-ß protects from CHIKV pathogenesis by limiting inflammation mediated by neutrophils. Our findings support the premise that the IFN subtypes have distinct biological activities in the antiviral response.


Asunto(s)
Fiebre Chikungunya/genética , Virus Chikungunya/patogenicidad , Factor 7 Regulador del Interferón/genética , Interferón-alfa/genética , Interferón beta/genética , Neutrófilos/inmunología , Animales , Anticuerpos Neutralizantes/farmacología , Huesos/inmunología , Huesos/patología , Huesos/virología , Fiebre Chikungunya/inmunología , Fiebre Chikungunya/patología , Fiebre Chikungunya/virología , Virus Chikungunya/inmunología , Femenino , Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Inflamación , Factor 7 Regulador del Interferón/deficiencia , Factor 7 Regulador del Interferón/inmunología , Interferón-alfa/antagonistas & inhibidores , Interferón-alfa/deficiencia , Interferón-alfa/inmunología , Interferón beta/antagonistas & inhibidores , Interferón beta/deficiencia , Interferón beta/inmunología , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Músculo Esquelético/virología , Infiltración Neutrófila , Neutrófilos/patología , Neutrófilos/virología , Tarso Animal/inmunología , Tarso Animal/patología , Tarso Animal/virología , Replicación Viral
6.
Cell ; 175(4): 1014-1030.e19, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30343900

RESUMEN

Although current immune-checkpoint therapy (ICT) mainly targets lymphoid cells, it is associated with a broader remodeling of the tumor micro-environment. Here, using complementary forms of high-dimensional profiling, we define differences across all hematopoietic cells from syngeneic mouse tumors during unrestrained tumor growth or effective ICT. Unbiased assessment of gene expression of tumor-infiltrating cells by single-cell RNA sequencing (scRNAseq) and longitudinal assessment of cellular protein expression by mass cytometry (CyTOF) revealed significant remodeling of both the lymphoid and myeloid intratumoral compartments. Surprisingly, we observed multiple subpopulations of monocytes/macrophages, distinguishable by the markers CD206, CX3CR1, CD1d, and iNOS, that change over time during ICT in a manner partially dependent on IFNγ. Our data support the hypothesis that this macrophage polarization/activation results from effects on circulatory monocytes and early macrophages entering tumors, rather than on pre-polarized mature intratumoral macrophages.


Asunto(s)
Linfocitos/inmunología , Células Mieloides/inmunología , Neoplasias/inmunología , Análisis de la Célula Individual , Transcriptoma , Animales , Línea Celular Tumoral , Citometría de Flujo , Inmunoterapia/métodos , Interferón gamma/inmunología , Activación de Macrófagos , Masculino , Espectrometría de Masas , Ratones , Células Precursoras de Monocitos y Macrófagos/inmunología , Neoplasias/terapia
7.
PLoS One ; 10(5): e0128636, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26010249

RESUMEN

Although type I interferons (IFNs) were first described almost 60 years ago, the ability to monitor and modulate the functional activities of the individual IFN subtypes that comprise this family has been hindered by a lack of reagents. The major type I IFNs, IFN-ß and the multiple subtypes of IFN-α, are expressed widely and induce their effects on cells by interacting with a shared heterodimeric receptor (IFNAR). In the mouse, the physiologic actions of IFN-α and IFN-ß have been defined using polyclonal anti-type I IFN sera, by targeting IFNAR using monoclonal antibodies or knockout mice, or using Ifnb-/- mice. However, the corresponding analysis of IFN-α has been difficult because of its polygenic nature. Herein, we describe two monoclonal antibodies (mAbs) that differentially neutralize murine IFN-ß or multiple subtypes of murine IFN-α. Using these mAbs, we distinguish specific contributions of IFN-ß versus IFN-α in restricting viral pathogenesis and identify IFN-α as the key mediator of the antiviral response in mice infected with West Nile virus. This study thus suggests the utility of these new reagents in dissecting the antiviral and immunomodulatory roles of IFN-ß versus IFN-α in murine models of infection, immunity, and autoimmunity.


Asunto(s)
Anticuerpos Bloqueadores/aislamiento & purificación , Anticuerpos Monoclonales/aislamiento & purificación , Interferón Tipo I/inmunología , Fiebre del Nilo Occidental/inmunología , Animales , Anticuerpos Bloqueadores/inmunología , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Modelos Animales de Enfermedad , Humanos , Interferón-alfa/inmunología , Interferón beta/inmunología , Ratones , Receptor de Interferón alfa y beta/metabolismo , Fiebre del Nilo Occidental/sangre , Fiebre del Nilo Occidental/virología
8.
Cell Host Microbe ; 17(5): 653-61, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25974304

RESUMEN

Although type I interferon (IFN-I) is thought to be beneficial against microbial infections, persistent viral infections are characterized by high interferon signatures suggesting that IFN-I signaling may promote disease pathogenesis. During persistent lymphocytic choriomeningitis virus (LCMV) infection, IFNα and IFNß are highly induced early after infection, and blocking IFN-I receptor (IFNAR) signaling promotes virus clearance. We assessed the specific roles of IFNß versus IFNα in controlling LCMV infection. While blockade of IFNß alone does not alter early viral dissemination, it is important in determining lymphoid structure, lymphocyte migration, and anti-viral T cell responses that lead to accelerated virus clearance, approximating what occurs during attenuation of IFNAR signaling. Comparatively, blockade of IFNα was not associated with improved viral control, but with early dissemination of virus. Thus, despite their use of the same receptor, IFNß and IFNα have unique and distinguishable biologic functions, with IFNß being mainly responsible for promoting viral persistence.


Asunto(s)
Interferón-alfa/metabolismo , Interferón beta/metabolismo , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Noqueados
9.
PLoS One ; 8(10): e76145, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24155889

RESUMEN

Dap12 and FcRγ, the two transmembrane ITAM-containing signaling adaptors expressed in dendritic cells (DC), are implicated in the regulation of DC function. Several activating and adhesion receptors including integrins require these chains for their function in triggering downstream signaling and effector pathways, however the exact role(s) for Dap12 and FcRγ remains elusive as their loss can lead to both attenuating and enhancing effects. Here, we report that mice congenitally lacking both Dap12 and FcRγ chains (DF) show a massively enhanced effector CD8(+) T cell response to protein antigen immunization or West Nile Virus (WNV) infection. Thus, immunization of DF mice with MHCI-restricted OVA peptide leads to accumulation of IL-12-producing monocyte-derived dendritic cells (Mo-DC) in draining lymph nodes, followed by vastly enhanced generation of antigen-specific IFNγ-producing CD8(+) T cells. Moreover, DF mice show increased viral clearance in the WNV infection model. Depletion of CCR2+ monocytes/macrophages in vivo by administration anti-CCR2 antibodies or clodronate liposomes completely prevents the exaggerated CD8+ T cell response in DF mice. Mechanistically, we show that the loss of Dap12 and FcRγ-mediated signals in Mo-DC leads to a disruption of GM-CSF receptor-induced STAT5 activation resulting in upregulation of expression of IRF8, a transcription factor. Consequently, Dap12- and FcRγ-deficiency exacerbates GM-CSF-driven monocyte differentiation and production of inflammatory Mo-DC. Our data suggest a novel cross-talk between DC-ITAM and GM-CSF signaling pathways, which controls Mo-DC differentiation, IL-12 production, and CD8(+) T cell responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/metabolismo , Interleucina-12/biosíntesis , Monocitos/citología , Receptores CCR2/metabolismo , Receptores de IgG/deficiencia , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos Virales/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Proliferación Celular/efectos de los fármacos , Reactividad Cruzada/efectos de los fármacos , Reactividad Cruzada/inmunología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Ratones , Ratones Endogámicos C57BL , Receptores de IgG/metabolismo , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/metabolismo , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/inmunología
10.
J Immunol ; 191(8): 4223-34, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24048899

RESUMEN

Although IFN-γ is required for resolution of Listeria monocytogenes infection, the identities of the IFN-γ-responsive cells that initiate the process remain unclear. We addressed this question using novel mice with conditional loss of IFN-γR (IFNGR1). Itgax-cre(+)Ifngr1(f/f) mice with selective IFN-γ unresponsiveness in CD8α(+) dendritic cells displayed increased susceptibility to infection. This phenotype was due to the inability of IFN-γ-unresponsive CD8α(+) dendritic cells to produce the initial burst of IL-12 induced by IFN-γ from TNF-α-activated NK/NKT cells. The defect in early IL-12 production resulted in increased IL-4 production that established a myeloid cell environment favoring Listeria growth. Neutralization of IL-4 restored Listeria resistance in Itgax-cre(+)Ifngr1(f/f) mice. We also found that Itgax-cre(+)Ifngr1(f/f) mice survived infection with low-dose Listeria as the result of a second wave of IL-12 produced by Ly6C(hi) monocytes. Thus, an IFN-γ-driven cascade involving CD8α(+) dendritic cells and NK/NKT cells induces the rapid production of IL-12 that initiates the anti-Listeria response.


Asunto(s)
Interferón gamma/inmunología , Interleucina-12/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Receptores de Interferón/inmunología , Animales , Antígenos Ly/metabolismo , Antígenos CD8/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interleucina-12/biosíntesis , Interleucina-4/biosíntesis , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Factor de Necrosis Tumoral alfa/metabolismo , Receptor de Interferón gamma
11.
Science ; 340(6129): 207-11, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23580529

RESUMEN

During persistent viral infections, chronic immune activation, negative immune regulator expression, an elevated interferon signature, and lymphoid tissue destruction correlate with disease progression. We demonstrated that blockade of type I interferon (IFN-I) signaling using an IFN-I receptor neutralizing antibody reduced immune system activation, decreased expression of negative immune regulatory molecules, and restored lymphoid architecture in mice persistently infected with lymphocytic choriomeningitis virus. IFN-I blockade before and after establishment of persistent virus infection resulted in enhanced virus clearance and was CD4 T cell-dependent. Hence, we demonstrate a direct causal link between IFN-I signaling, immune activation, negative immune regulator expression, lymphoid tissue disorganization, and virus persistence. Our results suggest that therapies targeting IFN-I may help control persistent virus infections.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/virología , Interferón Tipo I/metabolismo , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Transducción de Señal , Animales , Anticuerpos Antivirales/sangre , Infecciones por Arenaviridae/patología , Antígeno B7-H1/metabolismo , Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/virología , Femenino , Tolerancia Inmunológica , Interferón Tipo I/inmunología , Interleucina-10/metabolismo , Linfocitos/inmunología , Linfocitos/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor de Interferón alfa y beta/inmunología , Receptor de Interferón alfa y beta/metabolismo , Bazo/inmunología , Bazo/patología , Viremia
12.
PLoS Pathog ; 7(12): e1002407, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22144897

RESUMEN

A genetic absence of the common IFN-α/ß signaling receptor (IFNAR) in mice is associated with enhanced viral replication and altered adaptive immune responses. However, analysis of IFNAR(-/-) mice is limited for studying the functions of type I IFN at discrete stages of viral infection. To define the temporal functions of type I IFN signaling in the context of infection by West Nile virus (WNV), we treated mice with MAR1-5A3, a neutralizing, non cell-depleting anti-IFNAR antibody. Inhibition of type I IFN signaling at or before day 2 after infection was associated with markedly enhanced viral burden, whereas treatment at day 4 had substantially less effect on WNV dissemination. While antibody treatment prior to infection resulted in massive expansion of virus-specific CD8(+) T cells, blockade of type I IFN signaling starting at day 4 induced dysfunctional CD8(+) T cells with depressed cytokine responses and expression of phenotypic markers suggesting exhaustion. Thus, only the later maturation phase of anti-WNV CD8(+) T cell development requires type I IFN signaling. WNV infection experiments in BATF3(-/-) mice, which lack CD8-α dendritic cells and have impaired priming due to inefficient antigen cross-presentation, revealed a similar effect of blocking IFN signaling on CD8(+) T cell maturation. Collectively, our results suggest that cell non-autonomous type I IFN signaling shapes maturation of antiviral CD8(+) T cell response at a stage distinct from the initial priming event.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interferón Tipo I/inmunología , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/inmunología , Enfermedad Aguda , Animales , Anticuerpos Neutralizantes/farmacología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Antígenos CD8/genética , Antígenos CD8/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/genética , Ratones , Ratones Noqueados , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Factores de Tiempo , Fiebre del Nilo Occidental/genética
13.
J Immunol ; 181(12): 8492-503, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19050267

RESUMEN

Gene expression analysis previously revealed a robust IFN-responsive gene induction profile that was selectively up-regulated in Borrelia burgdorferi-infected C3H mice at 1 wk postinfection. This profile was correlated with arthritis development, as it was absent from infected, mildly arthritic C57BL/6 mice. In this report we now demonstrate that profile induction in infected C3H scid mice occurs independently of B or T lymphocyte infiltration in the joint tissue. Additionally, type I IFN receptor-blocking Abs, but not anti-IFN-gamma Abs, dramatically reduced arthritis, revealing a critical but previously unappreciated role for type I IFN in Lyme arthritis development. Certain examined IFN-inducible transcripts were also significantly diminished within joint tissue of mice treated with anti-IFNAR1, whereas expression of other IFN-responsive genes was more markedly altered by anti-IFN-gamma treatment. These data indicate that induction of the entire IFN profile is not necessary for arthritis development. These findings further tie early type I IFN induction to Lyme arthritis development, a connection not previously made. Bone marrow-derived macrophages readily induced IFN-responsive genes following B. burgdorferi stimulation, and this expression required a functional type I IFN receptor. Strikingly, induction of these genes was independent of TLRs 2,4, and 9 and of the adapter molecule MyD88. These data demonstrate that the extracellular pathogen B. burgdorferi uses a previously unidentified receptor and a pathway traditionally associated with viruses and intracellular bacteria to initiate transcription of type I IFN and IFN-responsive genes and to initiate arthritis development.


Asunto(s)
Borrelia burgdorferi/inmunología , Interferón Tipo I/fisiología , Enfermedad de Lyme/inmunología , Enfermedad de Lyme/microbiología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/patología , Femenino , Perfilación de la Expresión Génica , Inmunidad Innata/genética , Interferón Tipo I/biosíntesis , Interferón Tipo I/deficiencia , Enfermedad de Lyme/metabolismo , Enfermedad de Lyme/terapia , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología
14.
Transplantation ; 86(1): 137-47, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18622291

RESUMEN

BACKGROUND: Acute allograft rejection requires a multifaceted immune response involving trafficking of immune cells into the transplant and expression of effector cell functions leading to graft destruction. The chemokine receptor CXCR3 and its ligands, CXCL9, CXCL10 and CXCL11, constitute an important pathway for effector cell recruitment posttransplant. However, analysis of CXCR3 expression and function has been hampered by a general lack of availability of a neutralizing anti-CXCR3 monoclonal antibody (mAb) for use in experimental models. METHODS: We report the generation, characterization, and use of CXCR3-173, a new hamster mAb specific for mouse CXCR3 that recognizes CXCR3 on cells from wild-type but not CXCR3-/- mice. RESULTS: Using CXCR3-173 mAb, we demonstrate CXCR3 expression on primary memory phenotype CD4+ and CD8+ T cells, naturally occurring CD4+CD25+ Foxp3+ regulatory T cells, natural killer T cells, and approximately 25% of NK cells. CXCR3-173 blocked chemotaxis in vitro in response to CXCL10 or CXCL11 but not CXCL9. When injected into mice, this mAb significantly prolonged both cardiac and islet allograft survival. When combined with a subtherapeutic regimen of rapamycin, CXCR3-173 mAb induced long-term (>100 day) survival of cardiac and islet allografts. The in vivo effects of CXCR3-173 mAb were not associated with effector lymphocyte depletion. CONCLUSION: These data highlight the utility of CXCR3-173 mAb in developing immunotherapeutic approaches to inhibit transplant rejection and potentially other immune-mediated diseases in murine models.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Corazón , Inmunosupresores/farmacología , Trasplante de Islotes Pancreáticos , Subgrupos Linfocitarios/efectos de los fármacos , Receptores CXCR3/inmunología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Línea Celular , Quimiotaxis de Leucocito/efectos de los fármacos , Cricetinae , Citocinas/genética , Citocinas/metabolismo , Quimioterapia Combinada , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Células Asesinas Naturales/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales , ARN Mensajero/metabolismo , Receptores CXCR3/genética , Sirolimus/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Factores de Tiempo , Trasplante Homólogo
15.
Immunity ; 27(4): 597-609, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17950005

RESUMEN

Invariant natural killer T (iNKT) cells are a subset of innate lymphocytes that recognize lipid antigens in the context of CD1d and mediate potent immune regulatory functions via the rapid production of interferon-gamma (IFN-gamma) and interleukin-4 (IL-4). We investigated whether diverse Toll-like receptor (TLR) signals in myeloid dendritic cells (DCs) could differentially stimulate iNKT cells. Together with the lipopolysaccharide-detecting receptor TLR4, activation of the nucleic acid sensors TLR7 and TLR9 in DCs were particularly potent in stimulating iNKT cells to produce IFN-gamma, but not IL-4. iNKT cell activation in response to TLR9 stimulation required combined synthesis of type I interferon and de novo production of charged beta-linked glycosphingolipid(s) by DCs. In addition, DCs stimulated via TLR9 activated both iNKT cells and NK cells in vivo and protected mice against B16F10-induced melanoma metastases. These data underline the role of TLR9 in iNKT cell activation and might have relevance to infectious diseases and cancer.


Asunto(s)
Glicoesfingolípidos Acídicos/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Receptor Toll-Like 9/inmunología , Glicoesfingolípidos Acídicos/metabolismo , Animales , Antígenos CD1/inmunología , Antígenos CD1/metabolismo , Antígenos CD1d , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Femenino , Inmunoterapia Adoptiva , Interferón Tipo I/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Linfocitos T/metabolismo , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo
16.
J Immunol ; 178(12): 7540-9, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17548588

RESUMEN

This study demonstrates that type I IFNs are an early and critical regulator of NK cell numbers, activation, and antitumor activity. Using both IFNAR1- and IFNAR2-deficient mice, as well as an IFNAR1-blocking Ab, we demonstrate that endogenous type I IFN is critical for controlling NK cell-mediated antitumor responses in many experimental tumor models, including protection from methylcholanthrene-induced sarcomas, resistance to the NK cell-sensitive RMA-S tumor and cytokine immunotherapy of lung metastases. Protection from RMA-S afforded by endogenous type I IFN is more potent than that of other effector molecules such as IFN-gamma, IL-12, IL-18, and perforin. Furthermore, cytokine immunotherapy using IL-12, IL-18, or IL-21 was effective in the absence of endogenous type I IFN, however the antimetastatic activity of IL-2 was abrogated in IFNAR-deficient mice, primarily due to a defect in IL-2-induced cytotoxic activity. This study demonstrates that endogenous type I IFN is a central mediator of NK cell antitumor responses.


Asunto(s)
Interferón Tipo I/fisiología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Animales , Anticuerpos/farmacología , Citocinas/uso terapéutico , Homeostasis , Inmunoterapia , Interferón Tipo I/uso terapéutico , Células Asesinas Naturales/efectos de los fármacos , Activación de Linfocitos , Ratones , Ratones Mutantes , Neoplasias/prevención & control , Neoplasias/terapia , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Receptor de Interferón alfa y beta/química , Receptor de Interferón alfa y beta/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Interferon Cytokine Res ; 26(11): 804-19, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17115899

RESUMEN

Herein we report the generation of mouse monoclonal antibodies (mAbs) specific for the IFNAR-1 subunit of the mouse interferon-alpha/beta (IFN-alpha/beta) receptor (MAR1 mAbs) that block type I IFN receptor signaling and biologic response induction in vitro and in vivo. These mAbs were generated from Ifnar1 (/) mice immunized by in vivo hydrodynamic transfection with a plasmid encoding the extracellular domain (ECD) of murine IFNAR-1. All MAR1 mAbs bound native receptor expressed on cell surfaces and immunoprecipitated IFNAR-1 from solubilized cells, and two mAbs also detected IFNAR-1 by Western blot analysis. in vitro, the mAbs prevented ligand-induced intracellular signaling and induction of a variety of type I IFN-induced biologic responses but had no effect on IFN-gamma-induced responses. The most effective in vitro blocker, MAR1-5A3, also blocked type I IFN-induced antiviral, antimicrobial, and antitumor responses in vivo. We also explored whether murine IFNAR-1 surface expression required the presence of Tyk2. In contrast to Tyk2-deficient human cell lines, comparable IFNAR-1 expression was found on primary cells derived either from wild-type or Tyk2 (/) mice. These mAbs represent much needed tools to more clearly elucidate the biochemistry, cell biology, and physiologic function of the type I IFNs and their receptor in mediating host-protective immunity and immunopathology.


Asunto(s)
Anticuerpos Bloqueadores/inmunología , Anticuerpos Monoclonales/inmunología , Receptor de Interferón alfa y beta/inmunología , Animales , Anticuerpos Bloqueadores/aislamiento & purificación , Anticuerpos Monoclonales/aislamiento & purificación , Especificidad de Anticuerpos , Expresión Génica , Inmunización/métodos , Ratones , Ratones Noqueados , Plásmidos/genética , Plásmidos/inmunología , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , TYK2 Quinasa/deficiencia , TYK2 Quinasa/inmunología , Transfección/métodos
18.
Immunol Res ; 32(1-3): 231-45, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16106075

RESUMEN

Over the last 12 yr, we have shown that interferon-gamma and lymphocytes collaborate to regulate tumor development in mice. Specifically, we found that the immune system not only prevents the growth of primary (carcinogen-induced and spontaneous) and transplanted tumors but also sculpts the immunogenicity of tumors that form. These observations led us to refine the old and controversial "cancer immunosurveillance" hypothesis of Burnet and Thomas into one that we termed cancer immunoediting that better emphasizes the paradoxical host-protective and tumor-sculpting roles of immunity on developing tumors. Our current work focuses on defining the molecular mechanisms that underlie cancer immunoediting and exploring the implications of this process for cancer immunotherapy.


Asunto(s)
Interferón gamma/inmunología , Neoplasias/inmunología , Animales , Genes p53 , Rechazo de Injerto/inmunología , Humanos , Inmunoterapia , Linfocitos/inmunología , Ratones , Ratones Noqueados , Modelos Inmunológicos , Monitorización Inmunológica , Trasplante de Neoplasias , Neoplasias/prevención & control , Neoplasias/terapia , Transducción de Señal/inmunología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/inmunología
19.
Nat Immunol ; 6(7): 722-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15951814

RESUMEN

'Cancer immunoediting' is a process wherein the immune system protects hosts against tumor development and facilitates outgrowth of tumors with reduced immunogenicity. Although interferon-gamma (IFN-gamma) is known to be involved in this process, the involvement of type I interferons (IFN-alpha/beta) has not been elucidated. We now show that, like IFN-gamma, endogenously produced IFN-alpha/beta was required for the prevention of the growth of primary carcinogen-induced and transplantable tumors. Although tumor cells are important IFN-gamma targets, they are not functionally relevant sites of the actions of the type I interferons. Instead, host hematopoietic cells are critical IFN-alpha/beta targets during development of protective antitumor responses. Therefore, type I interferons are important components of the cancer immunoediting process and function in a way that does not completely overlap the functions of IFN-gamma.


Asunto(s)
Interferón-alfa/inmunología , Proteínas de la Membrana/inmunología , Neoplasias Experimentales/inmunología , Receptores de Interferón/inmunología , Sarcoma/inmunología , Escape del Tumor/inmunología , Animales , Proteínas de Unión al ADN/inmunología , Hematopoyesis/inmunología , Metilcolantreno , Ratones , Ratones Noqueados , Quimera por Radiación , Receptor de Interferón alfa y beta
20.
Cancer Res ; 65(8): 3447-53, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15833880

RESUMEN

We reported previously that 23% of human lung adenocarcinoma cell lines were unresponsive to IFN-gamma. To extend this finding to cancer cells derived from distinct tissues of origin, we assessed IFN-gamma receptor signaling in the LNCaP human prostate adenocarcinoma cell line, which in previous experiments by others failed to induce a range of IFN-dependent biological responses. In this report, we show that LNCaP cells fail to respond to either IFN-gamma or IFN-alpha because of an impairment in the proximal signaling events downstream of both IFN-gamma and IFN-alpha/beta receptors that lead to the activation of STAT1. Furthermore, we show that LNCaP insensitivity to the IFNs is a result of the absence of expression of the JAK1 kinase, an obligate component shared by both IFN-gamma and IFN-alpha/beta receptors. JAK1 was undetectable in LNCaP cells at both protein and message levels. Treatment of LNCaP cells with a combination of inhibitors of DNA methyltransferases and histone deacetylases induced expression of JAK1 message. These results identify the molecular basis for IFN insensitivity in the LNCaP cell line and suggest that epigenetic silencing of key immunologic signaling components may be one mechanism by which tumor cells evade immune detection and elimination.


Asunto(s)
Interferón Tipo I/farmacología , Interferón gamma/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/enzimología , Proteínas Tirosina Quinasas/deficiencia , Proteínas Recombinantes de Fusión/farmacología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Expresión Génica , Inhibidores de Histona Desacetilasas , Humanos , Interferón-alfa , Janus Quinasa 1 , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes , Transducción de Señal , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...