Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Microbes Infect ; 26(4): 105321, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38461968

RESUMEN

Rabies virus (RABV) is a lethal neurotropic virus that causes 60,000 human deaths every year globally. RABV infection is characterized by the suppression of the interferon (IFN)-mediated antiviral response. However, molecular mechanisms leading to RABV sensing by RIG-I-like receptors (RLR) that initiates IFN signaling currently remain elusive. Here, we showed that RABV RNAs are primarily recognized by the RIG-I RLR, resulting in an IFN response in the infected cells, but this response varied according to the type of RABV used. Pathogenic RABV strain RNAs, Tha, were poorly detected in the cytosol by RIG-I and therefore caused a weak antiviral response. However, we revealed a strong IFN activity triggered by the attenuated RABV vaccine strain RNAs, SAD, mediated by RIG-I. We characterized two major 5' copy-back defective interfering (5'cb DI) genomes generated during SAD replication. Furthermore, we identified an interaction between 5'cb DI genomes, and RIG-I correlated with a high stimulation of the type I IFN signaling. This study indicates that wild-type RABV RNAs poorly activate the RIG-I pathway, while the presence of 5'cb DIs in the live-attenuated vaccine strain serves as an intrinsic adjuvant that strengthens its efficiency by enhancing RIG-I detection thus strongly stimulates the IFN response.


Asunto(s)
Proteína 58 DEAD Box , Virus de la Rabia , Humanos , Línea Celular , Proteína 58 DEAD Box/metabolismo , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/inmunología , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Rabia/inmunología , Rabia/virología , Vacunas Antirrábicas/inmunología , Virus de la Rabia/inmunología , Virus de la Rabia/genética , Virus de la Rabia/patogenicidad , Receptores Inmunológicos/metabolismo , ARN Viral/genética , Transducción de Señal , Replicación Viral
2.
Proc Natl Acad Sci U S A ; 119(33): e2117904119, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35939684

RESUMEN

Many urinary tract infections (UTIs) are recurrent because uropathogens persist within the bladder epithelial cells (BECs) for extended periods between bouts of infection. Because persistent uropathogens are intracellular, they are often refractive to antibiotic treatment. The recent discovery of endogenous Lactobacillus spp. in the bladders of healthy humans raised the question of whether these endogenous bacteria directly or indirectly impact intracellular bacterial burden in the bladder. Here, we report that in contrast to healthy women, female patients experiencing recurrent UTIs have a bladder population of Lactobacilli that is markedly reduced. Exposing infected human BECs to L. crispatus in vitro markedly reduced the intracellular uropathogenic Escherichia coli (UPEC) load. The adherence of Lactobacilli to BECs was found to result in increased type I interferon (IFN) production, which in turn enhanced the expression of cathepsin D within lysosomes harboring UPECs. This lysosomal cathepsin D-mediated UPEC killing was diminished in germ-free mice and type I IFN receptor-deficient mice. Secreted metabolites of L. crispatus seemed to be responsible for the increased expression of type I IFN in human BECs. Intravesicular administration of Lactobacilli into UPEC-infected murine bladders markedly reduced their intracellular bacterial load suggesting that components of the endogenous microflora can have therapeutic effects against UTIs.


Asunto(s)
Antibiosis , Infecciones por Escherichia coli , Interferón Tipo I , Lactobacillus crispatus , Vejiga Urinaria , Infecciones Urinarias , Escherichia coli Uropatógena , Animales , Terapia Biológica , Catepsina D/metabolismo , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/terapia , Femenino , Humanos , Inmunidad Innata , Interferón Tipo I/inmunología , Lactobacillus crispatus/fisiología , Masculino , Ratones , Vejiga Urinaria/inmunología , Vejiga Urinaria/microbiología , Infecciones Urinarias/inmunología , Infecciones Urinarias/microbiología , Infecciones Urinarias/terapia , Escherichia coli Uropatógena/crecimiento & desarrollo
3.
Signal Transduct Target Ther ; 6(1): 331, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34471099

RESUMEN

The recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is the causative agent of ongoing global pandemic of COVID-19, may trigger immunosuppression in the early stage and overactive immune response in the late stage of infection; However, the underlying mechanisms are not well understood. Here we demonstrated that the SARS-CoV-2 nucleocapsid (N) protein dually regulated innate immune responses, i.e., the low-dose N protein suppressed type I interferon (IFN-I) signaling and inflammatory cytokines, whereas high-dose N protein promoted IFN-I signaling and inflammatory cytokines. Mechanistically, the SARS-CoV-2 N protein dually regulated the phosphorylation and nuclear translocation of IRF3, STAT1, and STAT2. Additionally, low-dose N protein combined with TRIM25 could suppress the ubiquitination and activation of retinoic acid-inducible gene I (RIG-I). Our findings revealed a regulatory mechanism of innate immune responses by the SARS-CoV-2 N protein, which would contribute to understanding the pathogenesis of SARS-CoV-2 and other SARS-like coronaviruses, and development of more effective strategies for controlling COVID-19.


Asunto(s)
COVID-19/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , Inmunidad Innata , SARS-CoV-2/inmunología , Transducción de Señal/inmunología , Células A549 , COVID-19/patología , Células CACO-2 , Células HEK293 , Células Hep G2 , Humanos , Interferón Tipo I/inmunología , Fosfoproteínas/inmunología
4.
Front Immunol ; 11: 580974, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33262759

RESUMEN

Novel adjuvants, such as Toll-like receptors (TLRs) agonists, are needed for the development of new formulations able to circumvent limitations of current vaccines. Among TLRs, TLR7/8 agonists represent promising candidates, as they are well described to enhance antigen-specific antibody responses and skew immunity toward T helper (TH) 1 responses. We find here that the incorporation of the synthetic TLR7/8 ligand 3M-052 in a cationic DOEPC-based liposome formulation shifts immunity toward TH1 responses and elicits strong and long-lasting germinal center and follicular T helper cell responses in adult mice. This reflects the prolonged recruitment of innate cells toward the site of immunization and homing of activated antigen-loaded monocytes and monocyte-derived dendritic cells toward draining lymph nodes. We further show that this adjuvanticity is independent of type I IFN but NF-κB-dependent. Overall, our data identify TLR7/8 agonists incorporated in liposomes as promising and effective adjuvants to enhance TH1 and germinal center responses.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Glicoproteínas de Membrana/agonistas , Monocitos/inmunología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Animales , Linfocitos B/inmunología , Células Dendríticas/inmunología , Composición de Medicamentos , Centro Germinal/inmunología , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Inmunidad Innata , Interferón Tipo I/inmunología , Ligandos , Liposomas/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/deficiencia , FN-kappa B/genética , FN-kappa B/inmunología , Fosfatidilcolinas/administración & dosificación , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/inmunología , Ácidos Esteáricos/administración & dosificación , Células TH1/inmunología
5.
Nat Commun ; 11(1): 3819, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32732875

RESUMEN

Hormone receptor (HR)+ breast cancer (BC) causes most BC-related deaths, calling for improved therapeutic approaches. Despite expectations, immune checkpoint blockers (ICBs) are poorly active in patients with HR+ BC, in part reflecting the lack of preclinical models that recapitulate disease progression in immunocompetent hosts. We demonstrate that mammary tumors driven by medroxyprogesterone acetate (M) and 7,12-dimethylbenz[a]anthracene (D) recapitulate several key features of human luminal B HR+HER2- BC, including limited immune infiltration and poor sensitivity to ICBs. M/D-driven oncogenesis is accelerated by immune defects, demonstrating that M/D-driven tumors are under immunosurveillance. Safe nutritional measures including nicotinamide (NAM) supplementation efficiently delay M/D-driven oncogenesis by reactivating immunosurveillance. NAM also mediates immunotherapeutic effects against established M/D-driven and transplantable BC, largely reflecting increased type I interferon secretion by malignant cells and direct stimulation of immune effector cells. Our findings identify NAM as a potential strategy for the prevention and treatment of HR+ BC.


Asunto(s)
Neoplasias de la Mama/terapia , Inmunoterapia/métodos , Niacinamida/administración & dosificación , Receptor ErbB-2/inmunología , 9,10-Dimetil-1,2-benzantraceno , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/inmunología , Progresión de la Enfermedad , Femenino , Humanos , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/prevención & control , Acetato de Medroxiprogesterona , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor ErbB-2/metabolismo , Análisis de Supervivencia
8.
Eur J Immunol ; 50(2): 178-191, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31608441

RESUMEN

Mucosal associated invariant T (MAIT) cells are abundant unconventional T cells that can be stimulated either via their TCR or by innate cytokines. The MAIT cell TCR recognises a pyrimidine ligand, derived from riboflavin synthesising bacteria, bound to MR1. In infection, bacteria not only provide the pyrimidine ligand but also co-stimulatory signals, such as TLR agonists, that can modulate TCR-mediated activation. Recently, type I interferons (T1-IFNs) have been identified as contributing to cytokine-mediated MAIT cell activation. However, it is unknown whether T1-IFNs also have a role during TCR-mediated MAIT cell activation. In this study, we investigated the co-stimulatory role of T1-IFNs during TCR-mediated activation of MAIT cells by the MR1 ligand 5-amino-6-d-ribitylaminouracil/methylglyoxal. We found that T1-IFNs were able to boost interferon-γ and granzyme B production in 5-amino-6-d-ribitylaminouracil/methylglyoxal-stimulated MAIT cells. Similarly, influenza virus-induced T1-IFNs enhanced TCR-mediated MAIT cell activation. An essential role of T1-IFNs in regulating MAIT cell activation by riboflavin synthesising bacteria was also demonstrated. The co-stimulatory role of T1-IFNs was also evident in liver-derived MAIT cells. T1-IFNs acted directly on MAIT cells to enhance their response to TCR stimulation. Overall, our findings establish an important immunomodulatory role of T1-IFNs during TCR-mediated MAIT cell activation.


Asunto(s)
Interferón Tipo I/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Células Cultivadas , Citocinas/inmunología , Humanos , Inmunidad Innata/inmunología , Interferón gamma/inmunología , Ligandos , Activación de Linfocitos/inmunología
9.
Front Immunol ; 9: 2746, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555460

RESUMEN

Chronic plaque psoriasis is a common debilitating skin disease. The identification of the pathogenic role of the TNF/IL-23/TH17 pathway has enabled the development of targeted therapies used in the clinic today. Particularly, TNF inhibitors have become a benchmark for the treatment of numerous chronic inflammatory diseases such as psoriasis. Although being highly effective in psoriasis treatment, anti-TNFs can themselves induce psoriasis-like skin lesions, a side effect called paradoxical psoriasis. In this review, we provide a comprehensive look at the different cellular and molecular players involved in classical plaque psoriasis and contrast its pathogenesis to paradoxical psoriasis, which is clinically similar but immunologically distinct. Classical psoriasis is a T-cell mediated autoimmune disease driven by TNF, characterised by T-cells memory, and a relapsing disease course. In contrast, paradoxical psoriasis is caused by the absence of TNF and represents an ongoing type-I interferon-driven innate inflammation that fails to elicit T-cell autoimmunity and lacks memory T cell-mediated relapses.


Asunto(s)
Memoria Inmunológica , Interferón Tipo I/inmunología , Psoriasis/inmunología , Transducción de Señal/inmunología , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Humanos , Interleucina-23/inmunología , Psoriasis/patología , Células Th17/patología
10.
J Immunol ; 199(7): 2491-2502, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28827286

RESUMEN

The anti-proliferative agent hexamethylene bisacetamide (HMBA) belongs to a class of hybrid bipolar compounds developed more than 30 y ago for their ability to induce terminal differentiation of transformed cells. Recently, HMBA has also been shown to trigger HIV transcription from latently infected cells, via a CDK9/HMBA inducible protein-1 dependent process. However, the effect of HMBA on the immune response has not been explored. We observed that pretreatment of human peripheral blood mononuclear cells with HMBA led to a markedly increased production of IL-12 and IFN-γ, but not of TNF-α, IL-6, and IL-8 upon subsequent infection with Burkholderia pseudomallei and Salmonella enterica HMBA treatment was also associated with better intracellular bacterial control. HMBA significantly improved IL-12p70 production from CD14+ monocytes during infection partly via the induction of type I IFN in these cells, which primed an increased transcription of the p35 subunit of IL-12p70 during infection. HMBA also increased early type I IFN transcription in human monocytic and epithelial cell lines, but this was surprisingly independent of its previously reported effects on positive transcription elongation factor b and HMBA inducible protein-1. Instead, the effect of HMBA was downstream of a calcium influx, and required the pattern recognition receptor and adaptor STING but not cGAS. Our work therefore links the STING-IRF3 axis to enhanced IL-12 production and intracellular bacterial control in primary monocytes. This raises the possibility that HMBA or related small molecules may be explored as therapeutic adjuvants to improve disease outcomes during intracellular bacterial infections.


Asunto(s)
Acetamidas/farmacología , Adyuvantes Inmunológicos , Interferón Tipo I/biosíntesis , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/microbiología , Proteínas de la Membrana/metabolismo , Acetamidas/uso terapéutico , Burkholderia pseudomallei/efectos de los fármacos , Burkholderia pseudomallei/inmunología , Línea Celular , Células Cultivadas , Citoplasma/inmunología , Citoplasma/microbiología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Interleucina-8/biosíntesis , Interleucina-8/inmunología , Leucocitos Mononucleares/inmunología , Proteínas de la Membrana/inmunología , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Salmonella enterica/efectos de los fármacos , Salmonella enterica/inmunología , Transducción de Señal , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/inmunología
11.
Clin Immunol ; 164: 65-77, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26821304

RESUMEN

Bruton's tyrosine kinase (Btk) is expressed in a variety of immune cells and previous work has demonstrated that blocking Btk is a promising strategy for treating autoimmune diseases. Herein, we utilized a tool Btk inhibitor, M7583, to determine the therapeutic efficacy of Btk inhibition in two mouse lupus models driven by TLR7 activation and type I interferon. In BXSB-Yaa lupus mice, Btk inhibition reduced autoantibodies, nephritis, and mortality. In the pristane-induced DBA/1 lupus model, Btk inhibition suppressed arthritis, but autoantibodies and the IFN gene signature were not significantly affected; suggesting efficacy was mediated through inhibition of Fc receptors. In vitro studies using primary human macrophages revealed that Btk inhibition can block activation by immune complexes and TLR7 which contributes to tissue damage in SLE. Overall, our results provide translational insight into how Btk inhibition may provide benefit to a variety of SLE patients by affecting both BCR and FcR signaling.


Asunto(s)
Lupus Eritematoso Sistémico/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa , Animales , Artritis/tratamiento farmacológico , Artritis/patología , Autoanticuerpos/sangre , Modelos Animales de Enfermedad , Femenino , Articulaciones del Pie/efectos de los fármacos , Articulaciones del Pie/patología , Humanos , Inmunosupresores , Interferón Tipo I/inmunología , Riñón/efectos de los fármacos , Riñón/patología , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Nefritis/tratamiento farmacológico , Nefritis/patología , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Proteinuria/tratamiento farmacológico , Proteinuria/patología , Terpenos , Receptor Toll-Like 7/inmunología
12.
Cell Rep ; 11(6): 957-966, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25937283

RESUMEN

Micro- and nanometer-size particles have become popular candidates for cancer vaccine adjuvants. However, the mechanism by which such particles enhance immune responses remains unclear. Here, we report a porous silicon microparticle (PSM)-based cancer vaccine that greatly enhances cross-presentation and activates type I interferon (IFN-I) response in dendritic cells (DCs). PSM-loaded antigen exhibited prolonged early endosome localization and enhanced cross-presentation through both proteasome- and lysosome-dependent pathways. Phagocytosis of PSM by DCs induced IFN-I responses through a TRIF- and MAVS-dependent pathway. DCs primed with PSM-loaded HER2 antigen produced robust CD8 T cell-dependent anti-tumor immunity in mice bearing HER2+ mammary gland tumors. Importantly, this vaccination activated the tumor immune microenvironment with elevated levels of intra-tumor IFN-I and MHCII expression, abundant CD11c+ DC infiltration, and tumor-specific cytotoxic T cell responses. These findings highlight the potential of PSM as an immune adjuvant to potentiate DC-based cancer immunotherapy.


Asunto(s)
Reactividad Cruzada/inmunología , Inmunidad Innata , Interferón Tipo I/inmunología , Microesferas , Neoplasias/inmunología , Silicio/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Femenino , Neoplasias Mamarias Animales/patología , Ratones Endogámicos C57BL , Neoplasias/patología , Ovalbúmina/inmunología , Fagocitosis , Porosidad , Receptor ErbB-2/metabolismo , Transducción de Señal , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral
13.
J Leukoc Biol ; 95(5): 841-847, 2014 05.
Artículo en Inglés | MEDLINE | ID: mdl-24421266

RESUMEN

Type I IFN signaling is a central pathway that provides critical innate protection from viral and bacterial infection and can have regulatory outcomes in inflammatory settings. We determined previously that OPCs contained in the dietary supplement APP enhanced responses to type I IFN in vitro. Here, we confirm that OPCs from two different sources significantly increased pSTAT1, whereas a monomeric form of procyanidin did not. We hypothesized that similar responses could be induced in vivo following ingestion of APP. Ingestion of APP before injection of polyI:C enhanced in vivo responses to type I IFNs in mice. When human subjects ingested APP, enhanced responses to type I IFN and enhanced pSTAT1 ex vivo were detected, whereas ingestion of RES, a monomeric polyphenol, induced minimal such changes. Polyphenols are best known for induction of anti-inflammatory and antioxidant responses; however, our findings suggest a unique, nonantioxidant aspect of OPCs that is broadly applicable to many disease settings. The capacity of oral OPCs to enhance type I IFN signaling in vivo can augment innate protection and may, in part, contribute to the noted anti-inflammatory outcome of ingestion of OPCs from many sources.


Asunto(s)
Biflavonoides/administración & dosificación , Catequina/administración & dosificación , Ácido Clorogénico/administración & dosificación , Flavonoides/administración & dosificación , Inmunidad Innata/efectos de los fármacos , Interferón Tipo I/inmunología , Proantocianidinas/administración & dosificación , Taninos/administración & dosificación , Administración Oral , Animales , Método Doble Ciego , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C
14.
J Immunol ; 192(3): 1071-8, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24376264

RESUMEN

Developing new adjuvants and vaccination strategies is of paramount importance to successfully fight against many life-threatening infectious diseases and cancer. Very few adjuvants are currently authorized for human use, and these mainly stimulate a humoral response. However, specific Abs are not sufficient to confer protection against persisting infections or cancer. Therefore, development of adjuvants and immunomodulators able to enhance cell-mediated immune responses represents a major medical need. We recently showed that papaya mosaic virus nanoparticles (PapMV), self-assembled from the coat protein of a plant virus and a noncoding ssRNA molecule, are highly immunogenic in mice. PapMV can be used either as a vaccine delivery platform, through fusion of various epitopes to the coat protein or as adjuvant to enhance humoral immune responses against coadministered Ags or vaccines. However, the mechanisms that confer these immunomodulatory properties to PapMV and its ability to enhance T cell vaccines remain unknown. Using immunization studies in mice, we demonstrate in this paper that PapMV represents a novel TLR7 agonist with strong immunostimulatory properties. More importantly, pretreatment with PapMV significantly improves effector and memory CD8(+) T cell responses generated through dendritic cell vaccination increasing protection against a Listeria monocytogenes challenge.


Asunto(s)
Adyuvantes Inmunológicos , Linfocitos T CD8-positivos/inmunología , Listeria monocytogenes/inmunología , Listeriosis/prevención & control , Glicoproteínas de Membrana/agonistas , Subgrupos de Linfocitos T/inmunología , Receptor Toll-Like 7/agonistas , Tymovirus/inmunología , Vacunación , Inmunidad Adaptativa , Animales , Células Dendríticas/inmunología , Evaluación Preclínica de Medicamentos , Femenino , Inmunoglobulina G/biosíntesis , Memoria Inmunológica , Interferón Tipo I/inmunología , Listeriosis/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/inmunología , Nanopartículas , Ovalbúmina/inmunología , ARN Viral/inmunología , Receptor de Interferón alfa y beta/deficiencia , Receptor Toll-Like 7/deficiencia , Receptor Toll-Like 7/inmunología , Tymovirus/genética
15.
Immunity ; 36(2): 166-74, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22365663

RESUMEN

Interferons (IFNs) were discovered as cytokines induced during and protecting from viral infection. They have been documented to play essential roles in numerous physiological processes beyond antiviral and antimicrobial defense, including immunomodulation, cell cycle regulation, cell survival, and cell differentiation. Recent data have also uncovered a potentially darker side to IFN, including roles in inflammatory diseases, such as autoimmunity and diabetes. IFN can have effects in the absence of acute infection, highlighting a physiologic role for constitutive IFN. Type I IFNs are constitutively produced at vanishingly low quantities and yet exert profound effects, mediated in part through modulation of signaling intermediates required for responses to diverse cytokines. We review evidence for a yin-yang of IFN function through its role in modulating crosstalk between multiple cytokines by both feedforward and feedback regulation of common signaling intermediates and postulate a homeostatic role for IFN through tonic signaling in the absence of acute infection.


Asunto(s)
Interferón Tipo I/fisiología , Animales , Antineoplásicos/inmunología , Antineoplásicos/metabolismo , Antivirales/inmunología , Antivirales/metabolismo , Autoinmunidad , Citocinas/fisiología , Retroalimentación Fisiológica , Homeostasis , Humanos , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Regiones Promotoras Genéticas , Transducción de Señal , Factores de Transcripción/metabolismo
16.
Curr Med Chem ; 17(29): 3327-36, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20712571

RESUMEN

Alpha interferons (IFN) are type I IFNs that have pleiotropic effects on cell functions. A wealth of evidence exists that these cytokines exhibit a variety of biological effects different from those on viral replication, including antitumor activity. IFNs-alpha represent the cytokines exhibiting the longest record of use in clinical oncology for the treatment of over a dozen of cancer types, including some hematological malignancies and solid tumors. Although targeted anticancer agents have recently replaced IFN-alpha in the treatment of certain hematological (e.g. chronic myeloid leukemia) and solid (e.g. renal cell carcinoma) malignancies, this cytokine is still used for the treatment of patients with specific tumor types, such as cutaneous melanoma. Despite the intense work in preclinical tumor models and considerable experience in the clinical use of IFN-alpha, the mechanisms of action underlying tumor response is still a matter of open debate. In this review we describe the evidence supporting the main mechanisms underlying IFN-alpha anticancer effects using both preclinical and clinical findings; moreover, we focus on the results of IFN-alpha for the treatment of patients with high-risk cutaneous melanoma, one of the malignancies most resistant to conventional chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Interferón Tipo I/uso terapéutico , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Antineoplásicos/inmunología , Ensayos Clínicos como Asunto , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Evaluación Preclínica de Medicamentos , Humanos , Interferón Tipo I/inmunología , Ratones , Proteínas Recombinantes
17.
J Immunol ; 182(7): 4296-305, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19299729

RESUMEN

The addition of monophosphoryl lipid A, a minimally toxic derivative of LPS, to nonmucosally administered vaccines induced both systemic and mucosal immune responses to coadministered Ags. This was dependent on an up-regulated expression of 1alpha-hydroxylase (CYP27B1, 1alphaOHase), the enzyme that converts 25-hydroxycholecalciferol, a circulating inactive metabolite of vitamin D(3), into 1,25(OH)2D(3) (calcitriol). In response to locally produced calcitriol, myeloid dendritic cells (DCs) migrated from cutaneous vaccination sites into multiple secondary lymphoid organs, including classical inductive sites of mucosal immunity, where they effectively stimulated B and T cell immune responses. The endogenous production of calcitriol by monophosphoryl lipid A-stimulated DCs appeared to be Toll-IL-1R domain-containing adapter-inducing IFN-beta-dependent, mediated through a type 1 IFN-induced expression of 1alphaOHase. Responsiveness to calcitriol was essential to promote the trafficking of mobilized DCs to nondraining lymphoid organs. Collectively, these studies help to expand our understanding of the physiologically important roles played by locally metabolized vitamin D(3) in the initiation and diversification of adaptive immune responses. The influences of locally produced calcitriol on the migration of activated DCs from sites of vaccination/infection into both draining and nondraining lymphoid organs create a condition whereby Ag-responsive B and T cells residing in multiple lymphoid organs are able to simultaneously engage in the induction of adaptive immune responses to peripherally administered Ags as if they were responding to an infection of peripheral or mucosal tissues they were designed to protect.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Colecalciferol/metabolismo , Células Dendríticas/inmunología , Receptores Toll-Like/inmunología , Vacunas/inmunología , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/inmunología , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Animales , Linfocitos B/inmunología , Western Blotting , Linfocitos T CD4-Positivos/inmunología , Calcitriol/inmunología , Calcitriol/metabolismo , Movimiento Celular/inmunología , Colecalciferol/inmunología , Células Dendríticas/metabolismo , Inmunidad Mucosa/inmunología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Lípido A/análogos & derivados , Lípido A/inmunología , Lípido A/farmacología , Activación de Linfocitos/inmunología , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Ratones , Ratones Transgénicos , Receptores Toll-Like/metabolismo
18.
J Immunol ; 178(4): 2204-11, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17277125

RESUMEN

Innate immune receptors that recognize nucleic acids, such as TLRs and RNA helicases, are potent activators of innate immunity that have been implicated in the induction and exacerbation of autoimmunity and inflammatory arthritis. Polyriboinosine-polyribocytidylic acid sodium salt (poly(IC)) is a mimic of dsRNA and viral infection that activates TLR3 and the RNA helicases retinoic acid-induced gene-1 and melanoma differentiation-associated gene-5, and strongly induces type I IFN production. We analyzed the effects of systemic delivery of poly(IC) on the inflammatory effector phase of arthritis using the collagen Ab-induced and KRN TCR-transgenic mouse serum-induced models of immune complex-mediated experimental arthritis. Surprisingly, poly(IC) suppressed arthritis, and suppression was dependent on type I IFNs that inhibited synovial cell proliferation and inflammatory cytokine production. Administration of exogenous type I IFNs was sufficient to suppress arthritis. These results suggest a regulatory role for innate immune receptors for dsRNA in modulating inflammatory arthritis and provide additional support for an anti-inflammatory function of type I IFNs in arthritis that directly contrasts with a pathogenic role in promoting autoimmunity in systemic lupus.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Inductores de Interferón/farmacología , Interferón Tipo I/inmunología , Poli I-C/farmacología , ARN Bicatenario/farmacología , Animales , Antivirales/inmunología , Antivirales/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Autoinmunidad/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Inflamación/inmunología , Inflamación/patología , Inductores de Interferón/inmunología , Interferón Tipo I/farmacología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos NOD , Poli I-C/inmunología , ARN Helicasas/inmunología , ARN Bicatenario/inmunología , Receptor Toll-Like 3/inmunología , Virosis/inmunología , Virosis/patología
19.
Toxicol Sci ; 79(1): 90-7, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14976356

RESUMEN

Immune-mediated idiosyncratic drug reactions are a major problem for susceptible patients, physicians, and the pharmaceutical industry. Validated screening tools to assess the immunosensitizing capacity of orally or intravenously administered pharmaceuticals are currently not available. To date, the popliteal lymph node assay (PLNA) seems the most promising tool for this purpose. The PLNA has recently been extended with the use of reporter antigens (RA) that are coinjected together with the drug of interest. The measurement of isotypes of RA-specific antibody-secreting cells (ASC) enables the distinction of sensitizing chemicals and (nonsensitizing) irritants without radio-isotopic end points. However, the use of footpad injections raises ethical concerns. Therefore, we examined the use of RA after intradermal injection into the ear of BALB/c mice and measured RA-specific ASC in the draining auricular lymph node (ALN). We show that RA-specific IgG isotype ASC numbers are very useful and sensitive parameters to identify drug-induced hypersensitivity in both PLN and ALN. However, the type 1-associated parameters (CD8(+) cells, macrophages, IFN-gamma, TNF-alpha, and IL-1 beta) that are induced in the PLN by streptozotocin were less pronounced in the ALN. Thus, the PLNA may provide more immunologically relevant information on the mechanisms of certain chemical-induced hypersensitivity reactions. The RA-ALN assay may provide an alternative for the RA-PLNA; both assays can be used to distinguish sensitizing compounds from nonsensitizing ones.


Asunto(s)
Antígenos/inmunología , Evaluación Preclínica de Medicamentos/métodos , Inmunosupresores/agonistas , Inmunosupresores/antagonistas & inhibidores , Ensayo del Nódulo Linfático Local , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Oído Externo/anatomía & histología , Femenino , Ficoll/administración & dosificación , Ficoll/inmunología , Inmunización/métodos , Inmunosupresores/inmunología , Inyecciones Intradérmicas , Inyecciones Subcutáneas , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ganglios Linfáticos/anatomía & histología , Ratones , Ratones Endogámicos BALB C , Ofloxacino/administración & dosificación , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Fenitoína/administración & dosificación , Sulfametoxazol/administración & dosificación , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
20.
Cancer Immunol Immunother ; 43(5): 299-306, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9024507

RESUMEN

We have previously shown that levamisole increases the cytotoxic, cytostatic, and proliferative activity of murine nonparenchymal liver cells (NPC) in vitro. We have also shown that the nonadherent subpopulation of NPC, which are composed predominantly of T lymphocytes, is very responsive to this agent when administered to mice. Kupffer cells or immigrant macrophages are also responsive to levamisole but to a lesser extent. These findings prompted us to investigate changes in cytokine production by NPC following-treatment of mice with levamisole (25 mg/kg, i.p.), which may help explain the observed alterations in the immune functions of these cells. We found that levamisole treatment of mice causes a threefold increase in production of interferon (IFN) alpha/beta by adherent NPC (more than 80%-90% Kupffer cells) in vitro. When IFN alpha/beta was added to cultured cells, it decreased the proliferative capacity of liver T cells in a dose-dependent manner. In contrast, the addition of anti-IFN alpha/beta was shown to augment levamisole-induced proliferation of unfractionated NPC and Kupffer cells. NPC production of interleukin 1 (IL-1) and interleukin-6 (IL-6) in vitro was also increased threefold following treatment of mice with levamisole. IL-6 added in vitro to cells significantly augmented levamisole-induced proliferation of liver T cells while anti-IL-6 reduced proliferative activity to control levels. These findings suggested that IFN alpha/beta, IL-6, and IL-1 play important regulatory roles in controlling the proliferative response of murine liver-associated T lymphocytes to levamisole. Finally, the proliferation of bone marrow cells was increased in mice given 5-fluorouracil (5FU). On the other hand, the proliferation of NPC was dramatically suppressed when 5FU was administered. However, the proliferation of these cells was restored when levamisole was given after 5FU.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Macrófagos del Hígado/inmunología , Levamisol/farmacología , Hígado/citología , Linfocitos T/citología , Animales , Médula Ósea/efectos de los fármacos , Células de la Médula Ósea , Fluorouracilo/farmacología , Interferón Tipo I/inmunología , Macrófagos del Hígado/efectos de los fármacos , Hígado/inmunología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA