Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 17(5): 514-522, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27043414

RESUMEN

Cytosolic DNA-mediated activation of the transcription factor IRF3 is a key event in host antiviral responses. Here we found that infection with DNA viruses induced interaction of the metabolic checkpoint kinase mTOR downstream effector and kinase S6K1 and the signaling adaptor STING in a manner dependent on the DNA sensor cGAS. We further demonstrated that the kinase domain, but not the kinase function, of S6K1 was required for the S6K1-STING interaction and that the TBK1 critically promoted this process. The formation of a tripartite S6K1-STING-TBK1 complex was necessary for the activation of IRF3, and disruption of this signaling axis impaired the early-phase expression of IRF3 target genes and the induction of T cell responses and mucosal antiviral immunity. Thus, our results have uncovered a fundamental regulatory mechanism for the activation of IRF3 in the cytosolic DNA pathway.


Asunto(s)
ADN/inmunología , Factor 3 Regulador del Interferón/inmunología , Proteínas de la Membrana/inmunología , Proteínas Quinasas S6 Ribosómicas 90-kDa/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Citosol/inmunología , Citosol/metabolismo , Citosol/virología , ADN/genética , ADN/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células HEK293 , Herpes Simple/inmunología , Herpes Simple/virología , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/fisiología , Humanos , Inmunización/métodos , Immunoblotting , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/inmunología , Nucleotidiltransferasas/metabolismo , Ovalbúmina/genética , Ovalbúmina/inmunología , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
2.
Immunol Rev ; 323(1): 19-39, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38459782

RESUMEN

Natural Killer (NK) cells are a top contender in the development of adoptive cell therapies for cancer due to their diverse antitumor functions and ability to restrict their activation against nonmalignant cells. Despite their success in hematologic malignancies, NK cell-based therapies have been limited in the context of solid tumors. Tumor cells undergo various metabolic adaptations to sustain the immense energy demands that are needed to support their rapid and uncontrolled proliferation. As a result, the tumor microenvironment (TME) is depleted of nutrients needed to fuel immune cell activity and contains several immunosuppressive metabolites that hinder NK cell antitumor functions. Further, we now know that NK cell metabolic status is a main determining factor of their effector functions. Hence, the ability of NK cells to withstand and adapt to these metabolically hostile conditions is imperative for effective and sustained antitumor activity in the TME. With this in mind, we review the consequences of metabolic hostility in the TME on NK cell metabolism and function. We also discuss tumor-like metabolic programs in NK cell induced by STAT3-mediated expansion that adapt NK cells to thrive in the TME. Finally, we examine how other approaches can be applied to enhance NK cell metabolism in tumors.


Asunto(s)
Células Asesinas Naturales , Neoplasias , Microambiente Tumoral , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Humanos , Microambiente Tumoral/inmunología , Animales , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Inmunoterapia Adoptiva/métodos , Adaptación Fisiológica , Factor de Transcripción STAT3/metabolismo , Metabolismo Energético , Escape del Tumor
3.
PLoS Pathog ; 19(6): e1011485, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37384813

RESUMEN

Mucosa-associated invariant T (MAIT) cells are MR1-restricted, innate-like T lymphocytes with tremendous antibacterial and immunomodulatory functions. Additionally, MAIT cells sense and respond to viral infections in an MR1-independent fashion. However, whether they can be directly targeted in immunization strategies against viral pathogens is unclear. We addressed this question in multiple wild-type and genetically altered but clinically relevant mouse strains using several vaccine platforms against influenza viruses, poxviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We demonstrate that 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil (5-OP-RU), a riboflavin-based MR1 ligand of bacterial origin, can synergize with viral vaccines to expand MAIT cells in multiple tissues, reprogram them towards a pro-inflammatory MAIT1 phenotype, license them to bolster virus-specific CD8+ T cell responses, and potentiate heterosubtypic anti-influenza protection. Repeated 5-OP-RU administration did not render MAIT cells anergic, thus allowing for its inclusion in prime-boost immunization protocols. Mechanistically, tissue MAIT cell accumulation was due to their robust proliferation, as opposed to altered migratory behavior, and required viral vaccine replication competency and Toll-like receptor 3 and type I interferon receptor signaling. The observed phenomenon was reproducible in female and male mice, and in both young and old animals. It could also be recapitulated in a human cell culture system in which peripheral blood mononuclear cells were exposed to replicating virions and 5-OP-RU. In conclusion, although viruses and virus-based vaccines are devoid of the riboflavin biosynthesis machinery that supplies MR1 ligands, targeting MR1 enhances the efficacy of vaccine-elicited antiviral immunity. We propose 5-OP-RU as a non-classic but potent and versatile vaccine adjuvant against respiratory viruses.


Asunto(s)
COVID-19 , Células T Invariantes Asociadas a Mucosa , Vacunas , Femenino , Masculino , Humanos , Ratones , Animales , Eficacia de las Vacunas , Leucocitos Mononucleares , COVID-19/metabolismo , SARS-CoV-2 , Riboflavina/metabolismo , Antígenos de Histocompatibilidad Clase I , Antígenos de Histocompatibilidad Menor
4.
PLoS Pathog ; 18(5): e1010471, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35512020

RESUMEN

The ability to treat severe viral infections is limited by our understanding of the mechanisms behind virus-induced immunopathology. While the role of type I interferons (IFNs) in early control of viral replication is clear, less is known about how IFNs can regulate the development of immunopathology and affect disease outcomes. Here, we report that absence of type I IFN receptor (IFNAR) is associated with extensive immunopathology following mucosal viral infection. This pathology occurred independent of viral load or type II immunity but required the presence of macrophages and IL-6. The depletion of macrophages and inhibition of IL-6 signaling significantly abrogated immunopathology. Tissue destruction was mediated by macrophage-derived matrix metalloproteinases (MMPs), as MMP inhibition by doxycycline and Ro 28-2653 reduced the severity of tissue pathology. Analysis of post-mortem COVID-19 patient lungs also displayed significant upregulation of the expression of MMPs and accumulation of macrophages. Overall, we demonstrate that IFNs inhibit macrophage-mediated MMP production to prevent virus-induced immunopathology and uncover MMPs as a therapeutic target towards viral infections.


Asunto(s)
COVID-19 , Interferón Tipo I , Infecciones por Orthomyxoviridae , Humanos , Interleucina-6/metabolismo , Macrófagos/metabolismo , Proteolisis
5.
J Infect Dis ; 228(7): 834-839, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-36994782

RESUMEN

Regulation of immune responses during viral infection is critical to preventing the development immunopathology that impairs host survival. Natural killer (NK) cells are well known for their antiviral functions that promote viral clearance; however, their roles in limiting immune-mediated pathology are still unclear. Using a mouse model for genital herpes simplex virus type 2 infection, we find that NK cell-derived interferon-γ directly counteracts interleukin-6-mediated matrix metalloproteases (MMPs) activity in macrophages to limit MMP-mediated tissue damage. Our findings uncover a key immunoregulatory function of NK cells during host-pathogen interactions that highlight the potential of NK cell therapy for treatment of severe viral infections.


Asunto(s)
Herpes Genital , Interferón gamma , Humanos , Células Asesinas Naturales , Herpesvirus Humano 2 , Macrófagos
6.
Immunol Cell Biol ; 100(6): 468-473, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35355318

RESUMEN

Zika virus (ZIKV) has emerged as a significant health threat worldwide. Although typically mosquito-borne, recent evidence suggests that ZIKV is also a sexually transmitted virus. While persistent ZIKV infections in male reproductive tissues have been identified, little is understood regarding the outcomes of primary sexual transmission in females. We investigated how the route of infection affects vaginal ZIKV shedding and dissemination. In two mouse models, vaginal infection resulted in prolonged ZIKV shedding in the vaginal mucosa with delayed systemic infection. Furthermore, heightened vaginal inflammation did not influence ZIKV replication or dissemination, in contrast to previous studies of mosquito-borne infection. Thus, vaginal infection significantly alters ZIKV infection kinetics and must be considered when developing novel treatments.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Animales , Femenino , Masculino , Ratones , Membrana Mucosa , Vagina , Esparcimiento de Virus
7.
J Immunol ; 202(10): 3041-3052, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30952815

RESUMEN

Serotonin (5-hydroxytryptamine [5-HT]) is a key enteric signaling molecule that mediates various physiological processes in the gut. Enterochromaffin (EC) cells in the mucosal layer of the gut are the main source of 5-HT in the body and are situated in close proximity to the gut microbiota. In this study, we identify a pivotal role of TLR2 in 5-HT production in the gut. Antibiotic treatment reduces EC cell numbers and 5-HT levels in naive C57BL/6 mice, which is associated with downregulation of TLR2 expression but not TLR1 or TLR4. TLR2-deficient (Tlr2 -/-) and Myd88 -/- mice express lower EC cell numbers and 5-HT levels, whereas treatment with TLR2/1 agonist upregulates 5-HT production in irradiated C57BL/6 mice, which are reconstituted with Tlr2 -/- bone marrow cells, and in germ-free mice. Human EC cell line (BON-1 cells) release higher 5-HT upon TLR2/1 agonist via NF-κB pathway. Tlr2 -/- mice and anti-TLR2 Ab-treated mice infected with enteric parasite, Trichuris muris, exhibited attenuated 5-HT production, compared with infected wild-type mice. Moreover, excretory-secretory products from T. muris induce higher 5-HT production in BON-1 cells via TLR2 in a dose-dependent manner, whereby the effect of excretory-secretory products is abrogated by TLR2 antagonist. These findings not only suggest an important role of TLR2 in mucosal 5-HT production in the gut by resident microbiota as well as by a nematode parasite but also provide, to our knowledge, novel information on the potential benefits of targeting TLR2 in various gut disorders that exhibit aberrant 5-HT signaling.


Asunto(s)
Células Enterocromafines/inmunología , Serotonina/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 2/inmunología , Tricuriasis/inmunología , Trichuris/inmunología , Animales , Línea Celular , Células Enterocromafines/patología , Microbioma Gastrointestinal/inmunología , Humanos , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Serotonina/genética , Transducción de Señal/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Tricuriasis/genética , Tricuriasis/patología
8.
Am J Physiol Endocrinol Metab ; 318(4): E579-E585, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32101030

RESUMEN

Defining the host receptors and metabolic consequences of bacterial components can help explain how the microbiome influences metabolic diseases. Bacterial peptidoglycans that activate nucleotide-binding oligomerization domain-containing (NOD)1 worsen glucose control, whereas NOD2 activation improves glycemia. Receptor-interacting serine/threonine-protein kinase 2 (RIPK2) is required for innate immunity instigated by NOD1 and NOD2. The role of RIPK2 in the divergent effects of NOD1 versus NOD2 on blood glucose was unknown. We found that whole body deletion of RIPK2 negated all effects of NOD1 or NOD2 activation on blood glucose during an acute, low level endotoxin challenge in mice. It was known that NOD1 in hematopoietic cells participates in insulin resistance and metabolic inflammation in obese mice. It was unknown if RIPK2 in hematopoietic cells is required for the glucose-lowering and anti-inflammatory effects of NOD2 activation. We hypothesized that RIPK2 in nonhematopoietic cells dictated the glycemic effects of NOD2 activation. We found that whole body deletion of RIPK2 prevented the glucose-lowering effects of repeated NOD2 activation that were evident during a glucose tolerance test (GTT) in high-fat diet (HFD)-fed wild-type (WT) mice. NOD2 activation lowered glucose during a GTT and lowered adipose tissue inflammation in mice with RIPK2 deleted in hematopoietic cells. We conclude that RIPK2 in nonhematopoietic cells mediates the glucose lowering and anti-inflammatory effects of NOD2-activating postbiotics. We propose a model where lipopolysaccharides and NOD1 ligands synergize in hematopoietic cells to promote insulin resistance but NOD2 activation in nonhematopoietic cells promotes RIPK2-dependent immune tolerance and lowering of inflammation and insulin resistance.


Asunto(s)
Glucemia/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Microbiota , Proteína Adaptadora de Señalización NOD2/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Activación Metabólica , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Dieta Alta en Grasa , Prueba de Tolerancia a la Glucosa , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética
9.
Cytokine ; 124: 154439, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-29908921

RESUMEN

Despite effective new treatments for Hepatitis C virus (HCV) infection, development of drug resistance, safety concerns and cost are remaining challenges. More importantly, there is no vaccine available against hepatitis C infection. Recent data suggest that there is a strong correlation between spontaneous HCV clearance and human NK cell function, particularly IFN-γ production. Further, IL-15 has innate antiviral activity and is also one of the main factors that activates NK cells to produce IFN-γ. To examine whether IL-15 and IFN-γ have direct antiviral activity against HCV, Huh7.5 cells were treated with either IFN-γ or IL-15 prior to HCV infection. Our data demonstrate that IFN-γ and IL-15 block HCV replication in vitro. Additionally, we show that IL-15 and IFN-γ do not induce anti-HCV effects through the type I interferon signaling pathway or nitric oxide (NO) production. Instead, IL-15 and IFN-γ provide protection against HCV via the ERK pathway. Treatment of Huh7.5 cells with a MEK/ERK inhibitor abrogated the anti-HCV effects of IL-15 and IFN-γ and overexpression of ERK1 prevented HCV replication compared to control transfection. Our in vitro data support the hypothesis that early production of IL-15 and activation of NK cells in the liver lead to control of HCV replication.


Asunto(s)
Hepacivirus/fisiología , Interferón gamma/farmacología , Interleucina-15/farmacología , Células Asesinas Naturales/inmunología , Hígado/inmunología , Hígado/virología , Sistema de Señalización de MAP Quinasas/inmunología , Replicación Viral , Antivirales/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Hepacivirus/efectos de los fármacos , Hepacivirus/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Interferón Tipo I/metabolismo , Interferón Tipo I/farmacología , Interferón-alfa/farmacología , Hígado/efectos de los fármacos , Hígado/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Óxido Nítrico/farmacología , Regulación hacia Arriba , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
10.
Proc Natl Acad Sci U S A ; 113(42): 11931-11936, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27698132

RESUMEN

The generation of strain-specific neutralizing antibodies against influenza A virus is known to confer potent protection against homologous infections. The majority of these antibodies bind to the hemagglutinin (HA) head domain and function by blocking the receptor binding site, preventing infection of host cells. Recently, elicitation of broadly neutralizing antibodies which target the conserved HA stalk domain has become a promising "universal" influenza virus vaccine strategy. The ability of these antibodies to elicit Fc-dependent effector functions has emerged as an important mechanism through which protection is achieved in vivo. However, the way in which Fc-dependent effector functions are regulated by polyclonal influenza virus-binding antibody mixtures in vivo has never been defined. Here, we demonstrate that interactions among viral glycoprotein-binding antibodies of varying specificities regulate the magnitude of antibody-dependent cell-mediated cytotoxicity induction. We show that the mechanism responsible for this phenotype relies upon competition for binding to HA on the surface of infected cells and virus particles. Nonneutralizing antibodies were poor inducers and did not inhibit antibody-dependent cell-mediated cytotoxicity. Interestingly, anti-neuraminidase antibodies weakly induced antibody-dependent cell-mediated cytotoxicity and enhanced induction in the presence of HA stalk-binding antibodies in an additive manner. Our data demonstrate that antibody specificity plays an important role in the regulation of ADCC, and that cross-talk among antibodies of varying specificities determines the magnitude of Fc receptor-mediated effector functions.


Asunto(s)
Anticuerpos Antivirales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Epítopos/inmunología , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Anticuerpos Neutralizantes/inmunología , Afinidad de Anticuerpos/inmunología , Antígenos Virales/química , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Unión Competitiva , Biomarcadores , Línea Celular , Epítopos/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/inmunología , Gripe Humana/metabolismo , Gripe Humana/virología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Modelos Biológicos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores Fc/metabolismo
11.
Cancer Immunol Immunother ; 67(4): 575-587, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29299659

RESUMEN

Ovarian cancer (OC) is the leading cause of gynecological cancer-related death in North America. Most ovarian cancer patients (OCPs) experience disease recurrence after first-line surgery and chemotherapy; thus, there is a need for novel second-line treatments to improve the prognosis of OC. Although peripheral blood-derived NK cells are known for their ability to spontaneously lyse tumour cells without prior sensitization, ascites-derived NK cells (ascites-NK cells) isolated from OCPs exhibit inhibitory phenotypes, impaired cytotoxicity and may play a pro-tumourigenic role in cancer progression. Therefore, it is of interest to improve the cytotoxic effector function of impaired OCP ascites-NK cells at the tumour environment. We investigated the efficacy of using an artificial APC-based ex vivo expansion technique to generate cytotoxic, expanded NK cells from previously impaired OCP ascites-NK cells, for use in an autologous model of NK cell immunotherapy. We are the first to obtain a log-scale expansion of OCP ascites-NK cells that upregulate the surface expression of activating receptors NKG2D, NKp30, NKp44, produce robust amounts of anti-tumour cytokines in the presence of OC cells and mediate direct tumour cytotoxicity against ascites-derived, primary OC cells obtained from autologous patients. Our findings demonstrate that it is possible to generate cytotoxic OCP ascites-NK cells from previously impaired OCP ascites-NK cells, which presents a promising immunotherapeutic target for the second-line treatment of OC. Future work should focus on evaluating the in vivo efficacy of autologous NK cell immunotherapy through the intraperitoneal delivery of NK cell expansion factors to a preclinical xenograft mouse model of human OC.


Asunto(s)
Ascitis/inmunología , Citotoxicidad Inmunológica/inmunología , Inmunoterapia , Células Asesinas Naturales/inmunología , Neoplasias Ováricas/inmunología , Ascitis/metabolismo , Proliferación Celular , Citocinas/metabolismo , Femenino , Humanos , Células Asesinas Naturales/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Células Tumorales Cultivadas
12.
Inflamm Res ; 67(10): 813-828, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30066126

RESUMEN

Metabolic flux can dictate cell fate, including immune cell effector and regulatory function. The metabolic regulation of cell function is well characterized with respect to effector, memory, and regulatory T cells. This knowledge may allow for manipulation of T cell metabolic pathways that set the stage for more effective T cell therapy. Natural Killer (NK) and T-lymphocytes have complementary roles in the defense against pathogens. However, studies of NK cell metabolism are only beginning to emerge and there is comparatively little knowledge on the metabolic regulation of NK-cell activation and effector function. Given their common lymphoid lineage, effector functions and cellular memory potential our current knowledge on T cell metabolism could inform investigation of metabolic reprogramming in NK cells. In this review, we compare the current knowledge of metabolic regulation in T cell and NK cell development, activation, effector and memory function. Commonalties in glucose transport, hypoxia-inducible factors and mTOR highlight metabolic control points in both cells types. Contrasting the glycolytic and oxidative nodes of metabolic regulation in T cells versus NK cells may provide insight into the contribution of specific immune responses to disease and promote the development of immunotherapeutic approaches targeting both innate and adaptive immune responses.


Asunto(s)
Células Asesinas Naturales/metabolismo , Linfocitos T/metabolismo , Animales , Humanos , Memoria Inmunológica , Inflamación/inmunología , Inflamación/metabolismo , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Linfocitos T/inmunología
13.
J Infect Dis ; 216(1): 135-145, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28531291

RESUMEN

Background: The translation of preclinically promising novel tuberculosis vaccines to ultimate human applications has been challenged by the lack of animal models with an immune system equivalent to the human immune system in its genetic diversity and level of susceptibility to tuberculosis. Methods: We have developed a humanized mice (Hu-mice) tuberculosis model system to investigate the clinical relevance of a novel virus-vectored (VV) tuberculosis vaccine administered via respiratory mucosal or parenteral route. Results: We find that VV vaccine activates T cells in Hu-mice as it does in human vaccinees. The respiratory mucosal route for delivery of VV vaccine in Hu-mice, but not the parenteral route, significantly reduces the humanlike lung tuberculosis outcomes in a human T-cell-dependent manner. Conclusions: Our results suggest that the Hu-mouse can be used to predict the protective efficacy of novel tuberculosis vaccines/strategies before they proceed to large, expensive human trials. This new vaccine testing system will facilitate the global pace of clinical tuberculosis vaccine development.


Asunto(s)
Vacuna BCG/administración & dosificación , Inmunidad Mucosa , Mucosa Respiratoria/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Antígenos Virales/sangre , Antígenos Virales/inmunología , Vacuna BCG/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Vectores Genéticos/inmunología , Humanos , Inmunización , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/prevención & control
14.
Breast Cancer Res ; 19(1): 76, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28668076

RESUMEN

BACKGROUND: Natural killer (NK) cells play a critical role in cancer immunosurveillance. Recent developments in NK cell ex-vivo expansion makes it possible to generate millions of activated NK cells from a small volume of peripheral blood. We tested the functionality of ex vivo expanded NK cells in vitro against breast cancer cell lines and in vivo using a xenograft mouse model. The study aim was to assess functionality and phenotype of expanded NK cells from breast cancer patients against breast cancer cell lines and autologous primary tumours. METHODS: We used a well-established NK cell co-culture system to expand NK cells ex vivo from healthy donors and breast cancer patients and examined their surface marker expression. Moreover, we tested the ability of expanded NK cells to lyse the triple negative breast cancer and HER2-positive breast cancer cell lines MDA-MB-231 and MDA-MB-453, respectively. We also tested their ability to prevent tumour growth in vivo using a xenograft mouse model. Finally, we tested the cytotoxicity of expanded NK cells against autologous and allogeneic primary breast cancer tumours in vitro. RESULTS: After 3 weeks of culture we observed over 1000-fold expansion of NK cells isolated from either breast cancer patients or healthy donors. We also showed that the phenotype of expanded NK cells is comparable between those from healthy donors and cancer patients. Moreover, our results confirm the ability of ex vivo expanded NK cells to lyse tumour cell lines in vitro. While the cell lines examined had differential sensitivity to NK cell killing we found this was correlated with level of major histocompatibility complex (MHC) class I expression. In our in vivo model, NK cells prevented tumour establishment and growth in immunocompromised mice. Finally, we showed that NK cells expanded from the peripheral blood of breast cancer patients show high cytotoxicity against allogeneic and autologous patient-derived tumour cells in vitro. CONCLUSION: NK cells from breast cancer patients can be expanded similarly to those from healthy donors, have a high cytotoxic ability against breast cancer cell lines and patient-derived tumour cells, and can be compatible with current cancer treatments to restore NK cell function in cancer patients.


Asunto(s)
Neoplasias de la Mama/inmunología , Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Animales , Biomarcadores , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva , Células Asesinas Naturales/metabolismo , Ratones , Ratones Noqueados , Receptor ErbB-2/metabolismo
16.
Cytokine ; 95: 7-11, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28189043

RESUMEN

Endotoxin, or LPS tolerance, is an immunomodulatory mechanism that results in a significantly diminished response to secondary LPS exposure, which may serve to protect the host against endotoxic shock. Type I interferons (IFNs) are cytokines released upon LPS binding to TLR4 and have been shown to have immunomodulatory properties. Due to this regulatory function of type I IFN, we aimed to investigate the role of type I IFN signalling in LPS tolerance. Our data suggests that type I IFN does not play a role in LPS tolerance in vitro, as both wild type and IFNAR1-/- peritoneal macrophages showed reduced cytokine production after secondary LPS exposure. Furthermore, both wild type and IFNAR1-/- mice were protected from a lethal dose of LPS after receiving three small doses to induce tolerance. However, IFNAR-/- mice seemed to recover faster than wild type mice, suggesting type I IFN signalling plays a detrimental role in LPS-induced sepsis. Although type I IFN may have a regulatory function in microbial infections, it does not seem to play a role in endotoxin tolerance. Type I IFN involvement in bacterial infection remains complex and further studies are needed to define the regulatory function of type I IFN signalling.


Asunto(s)
Interferón Tipo I/fisiología , Lipopolisacáridos/toxicidad , Choque Séptico/inmunología , Transducción de Señal , Animales , Células Cultivadas , Tolerancia a Medicamentos , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética
17.
BMC Immunol ; 17(1): 18, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27307066

RESUMEN

BACKGROUND: Humanized mouse models are an increasingly popular preclinical model to study the human immune response in a biological system. There are a variety of protocols to generate these mice, each differing in the strain of the recipient, source of hematopoietic stem cells, and mode of transplantation. Though there is well-documented reconstitution information regarding the spleen, blood, and bone marrow, there is little information regarding reconstitution of the lymph node and liver. In this report, we sought to compare reconstitution levels in a variety of immunological tissues, including the lymph node and liver, between mice engrafted intravenously as adults and intrahepatically in newborns. RESULTS: CD34+ cells were enriched from cord blood and transplanted intravenously into irradiated adult NOD-Rag1(-/-)IL2rγ(-/-) (NRG) mice or intra-hepatically into irradiated newborn NRG mice. At 9-28 weeks post-engraftment, immunological tissues were processed and analyzed for human lymphoid and myeloid subsets. Adult and newborn engrafted humanized mice were comparable in long-term reconstitution of human CD45 cells and subsequent lymphoid and myeloid subsets in the spleen, bone marrow, thymus, lymph node, and liver. Mice engrafted as newborns had a higher level of T-cells and a lower level of B-cells compared to mice engrafted as adults. We observed significant levels of human immune cell engraftment in both the lymph node and the liver, with a predominant adaptive immune population in both compartments. CONCLUSIONS: Human immune cells repopulate liver and mesenteric lymph nodes of NRG mice and can be used to study the human immune system in the gastrointestinal tract.


Asunto(s)
Envejecimiento/inmunología , Linfocitos B/fisiología , Células Madre Hematopoyéticas/inmunología , Hígado/fisiología , Ganglios Linfáticos/fisiología , Linfocitos T/fisiología , Animales , Animales Recién Nacidos , Antígenos CD34/metabolismo , Autorrenovación de las Células , Supervivencia Celular , Células Cultivadas , Trasplante de Células Madre Hematopoyéticas , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones Noqueados , Quimera por Trasplante
19.
J Immunol ; 193(12): 6184-91, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25355926

RESUMEN

IL-15 is a cytokine that can affect many immune cells, including NK cells and CD8 T cells. In several tumor models, IL-15 delays primary tumor formation and can prevent or reduce metastasis. In this study, we have employed a model of breast cancer metastasis to examine the mechanism by which IL-15 affects metastasis. When breast tumor cells were injected i.v. into IL-15(-/-), C57BL/6, IL-15 transgenic (TG) and IL-15/IL-15Rα-treated C57BL/6 mice, there were high levels of metastasis in IL-15(-/-) mice and virtually no metastasis in IL-15 TG or IL-15-treated mice. In fact, IL-15(-/-) mice were 10 times more susceptible to metastasis, whereas IL-15 TG mice were at least 10 times more resistant to metastasis when compared with control C57BL/6 mice. Depletion of NK cells from IL-15 TG mice revealed that these cells were important for protection from metastasis. When NK cells were depleted from control C57BL/6 mice, these mice did not form as many metastatic foci as IL-15(-/-) mice, suggesting that other cell types may be contributing to metastasis in the absence of IL-15. We then examined the role of CD4 T cells and macrophages. In IL-15(-/-) mice, in vivo depletion of CD4 T cells decreased metastasis. The lack of IL-15 in IL-15(-/-) mice, and possibly the Th2-polarized CD4 T cells, was found to promote the formation of M2 macrophages that are thought to contribute to metastasis formation. This study reveals that whereas IL-15 effects on NK cells are important, it also has effects on other immune cells that contribute to metastasis.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Linfocitos T CD4-Positivos/metabolismo , Expresión Génica , Interleucina-15/genética , Células Asesinas Naturales/metabolismo , Macrófagos/metabolismo , Animales , Neoplasias de la Mama/inmunología , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Femenino , Interleucina-15/administración & dosificación , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Macrófagos/inmunología , Neoplasias Mamarias Experimentales , Melanoma Experimental , Ratones , Ratones Noqueados , Ratones Transgénicos , Metástasis de la Neoplasia
20.
Eur J Immunol ; 44(5): 1375-86, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24519467

RESUMEN

The immune mechanisms underlying delayed induction of Th1-type immunity in the lungs following pulmonary mycobacterial infection remain poorly understood. We have herein investigated the underlying immune mechanisms for such delayed responses and whether a selected innate immune-modulating strategy can accelerate Th1-type responses. We have found that, in the early stage of pulmonary infection with attenuated Mycobacterium tuberculosis (M.tb H37Ra), the levels of infection in the lung continue to increase logarithmically until days 14 and 21 postinfection in C57BL/6 mice. The activation of innate immune responses, particularly DCs, in the lung is delayed. This results in a delay in the subsequent downstream immune responses including the migration of antigen-bearing DCs to the draining lymph node (dLN), the Th1-cell priming in dLN, and the recruitment of Th1 cells to the lung. However, single lung mucosal exposure to the TLR agonist FimH postinfection is able to accelerate protective Th1-type immunity via facilitating DC migration to the lung and draining lymph nodes, enhancing DC antigen presentation and Th1-cell priming. These findings hold implications for the development of immunotherapeutic and vaccination strategies and suggest that enhancement of early innate immune activation is a viable option for improving Th1-type immunity against pulmonary mycobacterial diseases.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Innata , Pulmón/inmunología , Mycobacterium tuberculosis/inmunología , Células TH1/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Células Dendríticas/microbiología , Células Dendríticas/patología , Pulmón/microbiología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/patología , Ratones , Ratones Transgénicos , Células TH1/patología , Factores de Tiempo , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA