Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.946
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 17(3): 297-303, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26752376

RESUMEN

Inflammation induced during infection can both promote and suppress immunity. This contradiction suggests that inflammatory cytokines affect the immune system in a context-dependent manner. Here we show that nonspecific bystander inflammation conditions naive CD4(+) T cells for enhanced peripheral Foxp3 induction and reduced effector differentiation. This results in inhibition of immune responses in vivo via a Foxp3-dependent effect on antigen-specific naive CD4(+) T cell precursors. Such conditioning may have evolved to allow immunity to infection while limiting subsequent autoimmunity caused by release of self-antigens in the wake of infection. Furthermore, this phenomenon suggests a mechanistic explanation for the idea that early tuning of the immune system by infection affects the long-term quality of immune regulation.


Asunto(s)
Asma/inmunología , Autoinmunidad/inmunología , Efecto Espectador/inmunología , Linfocitos T CD4-Positivos/inmunología , Citocinas/inmunología , Diabetes Mellitus/inmunología , Factores de Transcripción Forkhead/inmunología , Inflamación , Autotolerancia/inmunología , Animales , Autoantígenos , Efecto Espectador/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de los fármacos , Línea Celular Tumoral , Citocinas/efectos de los fármacos , Citocinas/farmacología , Metilación de ADN , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Inductores de Interferón/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Tolerancia Periférica/inmunología , Poli I-C/farmacología , Regiones Promotoras Genéticas , Linfopoyetina del Estroma Tímico
2.
Nat Immunol ; 14(4): 346-55, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23416614

RESUMEN

The production of type I interferon is essential for viral clearance but is kept under tight control to avoid unnecessary tissue damage from hyperinflammatory responses. Here we found that OASL1 inhibited translation of IRF7, the master transcription factor for type I interferon, and thus negatively regulated the robust production of type I interferon during viral infection. OASL1 inhibited the translation of IRF7 mRNA by binding to the 5' untranslated region (UTR) of IRF7 and possibly by inhibiting scanning of the 43S preinitiation complex along the message. Oasl1-/- mice were resistant to viral infection because of the greater abundance of type I interferon, which suggests that OASL1 could be a potential therapeutic target for boosting the production of type I interferon during viral infection.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/metabolismo , Regulación de la Expresión Génica , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Biosíntesis de Proteínas , 2',5'-Oligoadenilato Sintetasa/genética , Regiones no Traducidas 5' , Animales , Secuencia de Bases , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Línea Celular , Virus de la Encefalomiocarditis/inmunología , Homocigoto , Humanos , Inductores de Interferón/administración & dosificación , Inductores de Interferón/farmacología , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Poli I-C/administración & dosificación , Poli I-C/farmacología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , ARN Mensajero/metabolismo , Virosis/genética , Virosis/inmunología
3.
Trends Immunol ; 42(12): 1054-1056, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34764015

RESUMEN

Type 1 interferon (IFN) is a major contributor to the pathogenesis of systemic lupus erythematosus (SLE). A landmark study by Caielli et al. now shows that erythrocytes from lupus patients that fail to switch from glycolysis to oxidative phosphorylation during differentiation retain their mitochondria. These mitochondria-containing erythrocytes represent a novel source of IFN when phagocytosed by macrophages.


Asunto(s)
Interferón Tipo I , Lupus Eritematoso Sistémico , Eritrocitos , Humanos , Inductores de Interferón/metabolismo , Interferón Tipo I/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Mitocondrias
4.
Proc Natl Acad Sci U S A ; 117(11): 5761-5771, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32132203

RESUMEN

The circadian clock coordinates a variety of immune responses with signals from the external environment to promote survival. We investigated the potential reciprocal relationship between the circadian clock and skin inflammation. We treated mice topically with the Toll-like receptor 7 (TLR7) agonist imiquimod (IMQ) to activate IFN-sensitive gene (ISG) pathways and induce psoriasiform inflammation. IMQ transiently altered core clock gene expression, an effect mirrored in human patient psoriatic lesions. In mouse skin 1 d after IMQ treatment, ISGs, including the key ISG transcription factor IFN regulatory factor 7 (Irf7), were more highly induced after treatment during the day than the night. Nuclear localization of phosphorylated-IRF7 was most prominently time-of-day dependent in epidermal leukocytes, suggesting that these cell types play an important role in the diurnal ISG response to IMQ. Mice lacking Bmal1 systemically had exacerbated and arrhythmic ISG/Irf7 expression after IMQ. Furthermore, daytime-restricted feeding, which affects the phase of the skin circadian clock, reverses the diurnal rhythm of IMQ-induced ISG expression in the skin. These results suggest a role for the circadian clock, driven by BMAL1, as a negative regulator of the ISG response, and highlight the finding that feeding time can modulate the skin immune response. Since the IFN response is essential for the antiviral and antitumor effects of TLR activation, these findings are consistent with the time-of-day-dependent variability in the ability to fight microbial pathogens and tumor initiation and offer support for the use of chronotherapy for their treatment.


Asunto(s)
Ritmo Circadiano , Inmunidad Innata/genética , Interferones/genética , Glicoproteínas de Membrana/genética , Piel/metabolismo , Receptor Toll-Like 7/genética , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Imiquimod/farmacología , Inductores de Interferón/farmacología , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Interferones/metabolismo , Masculino , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Piel/efectos de los fármacos , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(10): 5409-5419, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32094169

RESUMEN

Type III IFN lambdas (IFN-λ) have recently been described as important mediators of immune responses at barrier surfaces. However, their role in autoimmune diseases such as systemic lupus erythematosus (SLE), a condition characterized by aberrant type I IFN signaling, has not been determined. Here, we identify a nonredundant role for IFN-λ in immune dysregulation and tissue inflammation in a model of TLR7-induced lupus. IFN-λ protein is increased in murine lupus and IFN-λ receptor (Ifnlr1) deficiency significantly reduces immune cell activation and associated organ damage in the skin and kidneys without effects on autoantibody production. Single-cell RNA sequencing in mouse spleen and human peripheral blood revealed that only mouse neutrophils and human B cells are directly responsive to this cytokine. Rather, IFN-λ activates keratinocytes and mesangial cells to produce chemokines that induce immune cell recruitment and promote tissue inflammation. These data provide insights into the immunobiology of SLE and identify type III IFNs as important factors for tissue-specific pathology in this disease.


Asunto(s)
Interferones/fisiología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Animales , Linfocitos B/inmunología , Línea Celular , Eliminación de Gen , Humanos , Imiquimod/farmacología , Inflamación/inmunología , Inflamación/patología , Inductores de Interferón/farmacología , Interferón Tipo I/fisiología , Interferones/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Queratinocitos/patología , Células Mesangiales/efectos de los fármacos , Células Mesangiales/inmunología , Células Mesangiales/patología , Ratones Endogámicos C57BL , Ratones Mutantes , Receptores de Interferón/genética , Transducción de Señal , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/fisiología , Interferón lambda
6.
J Neurosci ; 41(48): 9971-9987, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34607967

RESUMEN

Human epidemiological studies implicate exposure to infection during gestation in the etiology of neurodevelopmental disorders. Animal models of maternal immune activation (MIA) have identified the maternal immune response as the critical link between maternal infection and aberrant offspring brain and behavior development. Here we evaluate neurodevelopment of male rhesus monkeys (Macaca mulatta) born to MIA-treated dams (n = 14) injected with a modified form of the viral mimic polyinosinic:polycytidylic acid at the end of the first trimester. Control dams received saline injections at the same gestational time points (n = 10) or were untreated (n = 4). MIA-treated dams exhibited a strong immune response as indexed by transient increases in sickness behavior, temperature, and inflammatory cytokines. Although offspring born to control or MIA-treated dams did not differ on measures of physical growth and early developmental milestones, the MIA-treated animals exhibited subtle changes in cognitive development and deviated from species-typical brain growth trajectories. Longitudinal MRI revealed significant gray matter volume reductions in the prefrontal and frontal cortices of MIA-treated offspring at 6 months that persisted through the final time point at 45 months along with smaller frontal white matter volumes in MIA-treated animals at 36 and 45 months. These findings provide the first evidence of early postnatal changes in brain development in MIA-exposed nonhuman primates and establish a translationally relevant model system to explore the neurodevelopmental trajectory of risk associated with prenatal immune challenge from birth through late adolescence.SIGNIFICANCE STATEMENT Women exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder. Preclinical maternal immune activation (MIA) models have demonstrated that the effects of maternal infection on fetal brain development are mediated by maternal immune response. Since the majority of MIA models are conducted in rodents, the nonhuman primate provides a unique system to evaluate the MIA hypothesis in a species closely related to humans. Here we report the first longitudinal study conducted in a nonhuman primate MIA model. MIA-exposed offspring demonstrate subtle changes in cognitive development paired with marked reductions in frontal gray and white matter, further supporting the association between prenatal immune challenge and alterations in offspring neurodevelopment.


Asunto(s)
Encéfalo/patología , Modelos Animales de Enfermedad , Trastornos del Neurodesarrollo/etiología , Complicaciones Infecciosas del Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Femenino , Inductores de Interferón/toxicidad , Macaca mulatta , Masculino , Trastornos del Neurodesarrollo/patología , Neurogénesis/fisiología , Poli I-C/toxicidad , Embarazo , Complicaciones Infecciosas del Embarazo/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
7.
J Neurosci ; 41(32): 6954-6965, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34253630

RESUMEN

Episodic memory requires information to be stored and recalled in sequential order, and these processes are disrupted in schizophrenia. Hippocampal phase precession and theta sequences are thought to provide a biological mechanism for sequential ordering of experience at timescales suitable for plasticity. These phenomena have not previously been examined in any models of schizophrenia risk. Here, we examine these phenomena in a maternal immune activation (MIA) rodent model. We show that while individual pyramidal cells in the CA1 region continue to precess normally in MIA animals, the starting phase of precession as an animal enters a new place field is considerably more variable in MIA animals than in controls. A critical consequence of this change is a disorganization of the ordered representation of experience via theta sequences. These results provide the first evidence of a biological-level mechanism that, if it occurs in schizophrenia, may explain aspects of disorganized sequential processing that contribute to the cognitive symptoms of the disorder.SIGNIFICANCE STATEMENT Hippocampal phase precession and theta sequences have been proposed as biophysical mechanisms by which the sequential structure of cognition might be ordered. Disturbances of sequential processing have frequently been observed in schizophrenia. Here, we show for the first time that phase precession and theta sequences are disrupted in a maternal immune activation (MIA) model of schizophrenia risk. This is a result of greater variability in the starting phase of precession, indicating that the mechanisms that coordinate precession at the assembly level are disrupted. We propose that this disturbance in phase precession underlies some of the disorganized cognitive symptoms that occur in schizophrenia. These findings could have important preclinical significance for the identification and treatment of schizophrenia risk factors.


Asunto(s)
Hipocampo/fisiopatología , Memoria Episódica , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Esquizofrenia/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Inflamación/inducido químicamente , Inductores de Interferón/toxicidad , Masculino , Exposición Materna/efectos adversos , Poli I-C/toxicidad , Embarazo , Ratas Sprague-Dawley , Esquizofrenia/etiología
8.
Cancer Immunol Immunother ; 70(11): 3081-3091, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33751208

RESUMEN

The nanoparticle complex of cholesteryl pullulan (CHP) and NY-ESO-1 antigen protein (CHP-NY-ESO-1) presents multiple epitope peptides to MHC class I and II pathways, leading to CD8+ and CD4+ T cell responses. Poly-ICLC is a synthetic, double-stranded RNA, an agonist of toll-like receptor (TLR)-3, and a cytoplasmic receptor of melanoma differentiation-associated gene (MDA)-5. It should be a suitable immune adjuvant of cancer vaccine to overcome the inhibitory tumor microenvironment. We conducted a phase 1 clinical trial of CHP-NY-ESO-1 with poly-ICLC in patients with advanced or recurrent esophageal cancer. CHP-NY-ESO-1/poly-ICLC (µg/mg) was administered at a dose of 200/0.5 or 200/1.0 (cohorts 1 and 2, respectively) every 2 weeks for a total of six doses. The primary endpoints were safety and immune response. The secondary endpoint was tumor response. In total, 16 patients were enrolled, and six patients in each cohort completed the trial. The most common adverse event (AE) was injection site skin reaction (86.7%). No grade 3 or higher drug-related AEs were observed. No tumor responses were observed, and three patients (30%) had stable disease. The immune response was comparable between the two cohorts, and all patients (100%) achieved antibody responses with a median of 2.5 vaccinations. Comparing CHP-NY-ESO-1 alone to the poly-ICLC combination, all patients in both groups exhibited antibody responses, but the titers were higher in the combination group. In a mouse model, adding anti-PD-1 antibody to the combination of CHP-NY-ESO-1/poly-ICLC suppressed the growth of NY-ESO-1-expressing tumors. Combining the vaccine with PD-1 blockade holds promise in human trials.


Asunto(s)
Antígenos de Neoplasias/uso terapéutico , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Carboximetilcelulosa de Sodio/análogos & derivados , Neoplasias Esofágicas/tratamiento farmacológico , Glucanos/uso terapéutico , Proteínas de la Membrana/uso terapéutico , Poli I-C/uso terapéutico , Polilisina/análogos & derivados , Adyuvantes Inmunológicos/uso terapéutico , Anciano , Anciano de 80 o más Años , Animales , Antígenos de Neoplasias/inmunología , Carboximetilcelulosa de Sodio/uso terapéutico , Neoplasias Esofágicas/inmunología , Femenino , Glucanos/inmunología , Humanos , Inductores de Interferón/inmunología , Inductores de Interferón/uso terapéutico , Masculino , Proteínas de la Membrana/inmunología , Ratones , Persona de Mediana Edad , Nanopartículas , Poli I-C/inmunología , Polilisina/inmunología , Polilisina/uso terapéutico
9.
Int J Mol Sci ; 22(19)2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34639115

RESUMEN

Psoriasis is a chronic inflammatory skin disease. Recently, lysophosphatidic acid (LPA)/LPAR5 signaling has been reported to be involved in both NLRP3 inflammasome activation in macrophages and keratinocyte activation to produce inflammatory cytokines, contributing to psoriasis pathogenesis. However, the effect and molecular mechanisms of LPA/LPAR signaling in keratinocyte proliferation in psoriasis remain unclear. In this study, we investigated the effects of LPAR1/3 inhibition on imiquimod (IMQ)-induced psoriasis-like mice. Treatment with the LPAR1/3 antagonist, ki16425, alleviated skin symptoms in IMQ-induced psoriasis-like mouse models and decreased keratinocyte proliferation in the lesion. It also decreased LPA-induced cell proliferation and cell cycle progression via increased cyclin A2, cyclin D1, cyclin-dependent kinase (CDK)2, and CDK4 expression and decreased p27Kip1 expression in HaCaT cells. LPAR1 knockdown in HaCaT cells reduced LPA-induced proliferation, suppressed cyclin A2 and CDK2 expression, and restored p27Kip1 expression. LPA increased Rho-associated protein kinase 2 (ROCK2) expression and PI3K/AKT activation; moreover, the pharmacological inhibition of ROCK2 and PI3K/AKT signaling suppressed LPA-induced cell cycle progression. In conclusion, we demonstrated that LPAR1/3 antagonist alleviates IMQ-induced psoriasis-like symptoms in mice, and in particular, LPAR1 signaling is involved in cell cycle progression via ROCK2/PI3K/AKT pathways in keratinocytes.


Asunto(s)
Proliferación Celular , Regulación de la Expresión Génica/efectos de los fármacos , Imiquimod/toxicidad , Queratinocitos/citología , Lisofosfolípidos/farmacología , Psoriasis/tratamiento farmacológico , Animales , Apoptosis , Biomarcadores/metabolismo , Ciclo Celular , Células Cultivadas , Humanos , Inductores de Interferón/toxicidad , Queratinocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Psoriasis/inducido químicamente , Psoriasis/metabolismo , Psoriasis/patología , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/metabolismo , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
10.
Bull Exp Biol Med ; 171(6): 736-740, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34705177

RESUMEN

We studied the effect of tilorone on the dynamics of IFNα, IFNγ, and IL-1ß levels in the lung tissue and blood serum in relation to viral load in the lungs of BALB/c mice with pneumonia caused by influenza virus A/Aichi/2/68 (H3N2). Tilorone was administered per os in doses of 40, 150, and 540 µg per mouse 6, 30, and 78 h postinfection, which simulated the drug regimen used in the clinic for the treatment of influenza and acute respiratory viral infections in Russia and post-Soviet countries. Tilorone reduced viral load with the maximum amplitude (2-3 lg) after 1-2 administrations. The results of studying the dynamics of the cytokine levels in the infected animals in general support the previous hypothesis that, in repeated dosing, tilorone enhances the IFN response (compensates for its deficiency) at the early stages of acute respiratory viral infections and suppresses (damps) excessive production of IFN and proinflammatory cytokines at the later stages.


Asunto(s)
Antivirales/farmacología , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Inductores de Interferón/farmacología , Pulmón/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Tilorona/farmacología , Animales , Esquema de Medicación , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Interferón-alfa/sangre , Interferón-alfa/inmunología , Interferón gamma/sangre , Interferón gamma/inmunología , Interleucina-1beta/sangre , Interleucina-1beta/inmunología , Pulmón/inmunología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/sangre , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Carga Viral/efectos de los fármacos
11.
Immunity ; 34(2): 213-23, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21349431

RESUMEN

Type I interferon (IFN) is a common therapy for autoimmune and inflammatory disorders, yet the mechanisms of action are largely unknown. Here we showed that type I IFN inhibited interleukin-1 (IL-1) production through two distinct mechanisms. Type I IFN signaling, via the STAT1 transcription factor, repressed the activity of the NLRP1 and NLRP3 inflammasomes, thereby suppressing caspase-1-dependent IL-1ß maturation. In addition, type I IFN induced IL-10 in a STAT1-dependent manner; autocrine IL-10 then signaled via STAT3 to reduce the abundance of pro-IL-1α and pro-IL-1ß. In vivo, poly(I:C)-induced type I IFN diminished IL-1ß production in response to alum and Candida albicans, thus increasing susceptibility to this fungal pathogen. Importantly, monocytes from multiple sclerosis patients undergoing IFN-ß treatment produced substantially less IL-1ß than monocytes derived from healthy donors. Our findings may thus explain the effectiveness of type I IFN in the treatment of inflammatory diseases but also the observed "weakening" of the immune system after viral infection.


Asunto(s)
Inflamasomas/metabolismo , Interferón Tipo I/fisiología , Interleucina-1/biosíntesis , Animales , Proteínas Reguladoras de la Apoptosis/fisiología , Candida albicans/fisiología , Candidiasis/etiología , Candidiasis/inmunología , Proteínas Portadoras/fisiología , Caspasa 1/deficiencia , Caspasa 1/genética , Caspasa 1/fisiología , Células Cultivadas/metabolismo , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inductores de Interferón/farmacología , Interferón Tipo I/biosíntesis , Interferón Tipo I/genética , Interferón beta/uso terapéutico , Interleucina-1/genética , Interleucina-10/fisiología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Proteína con Dominio Pirina 3 de la Familia NLR , Peritonitis/etiología , Peritonitis/inmunología , Poli I-C/farmacología , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/fisiología , Factor de Transcripción STAT3/fisiología
12.
Alcohol Clin Exp Res ; 44(9): 1760-1768, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32640038

RESUMEN

BACKGROUND: The inbred mouse strain C57BL/6 is widely used in both models of addiction and immunological disease. However, there are pronounced phenotypic differences in ethanol (EtOH) consumption and innate immune response between C57BL/6 substrains. The focus of this study was to examine the effects of substrain on innate immune response and neuroimmune-induced escalation of voluntary EtOH consumption. The main goal was to identify whether substrain differences in immune response can account for differences in EtOH behavior. METHODS: We compared acute innate immune response with a viral dsRNA mimic, polyinosinic:polycytidylic acid (poly(I:C)), in brain using qRT-PCR in both C57BL/6N and C57BL/6J mice. Next, we used a neuroimmune model of escalation using poly(I:C) to compare drinking behavior between substrains. Finally, we compared brain neuroimmune response with both EtOH and repeated poly(I:C) in both substrains as a way to account for differences in EtOH behavior. RESULTS: We found that C57BL/6 substrains have differing immune response and drinking behaviors. C57BL/6N mice have a shorter but more robust inflammatory response to acute poly(I:C). In contrast, C57BL/6J mice have a smaller but longer-lasting acute immune response to poly(I:C). In our neuroimmune-induced escalation model, C57BL/6J mice but not C57BL/6N mice escalate EtOH intake after poly(I:C). Finally, only C57BL/6J mice show enhanced proinflammatory transcript abundance after poly(I:C) and EtOH, suggesting that longer-lasting immune responses are critical to neuroimmune drinking phenotypes. CONCLUSIONS: Altogether, this work has elucidated additional influences that substrain has on both innate immune response and drinking phenotypes. Our observations highlight the importance of considering and reporting the source and background used for production of transgenic and knockout mice. These data provide further evidence that genetic background must be carefully considered when investigating the role of neuroimmune signaling in EtOH abuse.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Conducta Animal , Inmunidad Innata/genética , Neuroinmunomodulación/genética , Consumo de Bebidas Alcohólicas/inmunología , Consumo de Bebidas Alcohólicas/fisiopatología , Animales , Depresores del Sistema Nervioso Central/administración & dosificación , Etanol/administración & dosificación , Inmunidad Innata/inmunología , Inductores de Interferón/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Modelos Animales , Neuroinmunomodulación/inmunología , Poli I-C/farmacología
13.
Alcohol Clin Exp Res ; 44(9): 1728-1733, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32583876

RESUMEN

BACKGROUND: A relationship between alcohol consumption and psoriasis has been reported, but it is unclear whether alcohol consumption aggravates psoriasis. Here, we studied the effect of chronic ethanol (EtOH) consumption in the murine model of Aldara-induced psoriasiform dermatitis. METHODS: C57BL/6 mice received 5% EtOH in their drinking water for 10 weeks. Dermatitis was induced from weeks 9 to 10, by applying Aldara to the shaved patch of skin on the back. Inflammation was characterized by histological and transcriptomic analyses. RESULTS: EtOH consumption aggravated Aldara-induced dermatitis. The scales were more severe, epidermal thickening was more pronounced, and cutaneous expression of Th17-related cytokines was exacerbated. Control mice simply receiving EtOH displayed minimal cutaneous inflammation, characterized by epidermal infiltrates of T lymphocytes and the overexpression of IL-17A and the Th17-recruiting chemokine CCL20. In vitro studies showed that low concentrations of EtOH induce the expression of CCL20 by murine epidermal keratinocytes. CONCLUSION: Alcohol consumption leads to subliminar skin inflammation, which is revealed by the exacerbation of Aldara-induced experimental psoriasiform dermatitis, likely through Th17-type minimal skin inflammation. These results favor the systematic management of alcohol consumption in psoriatic patients.


Asunto(s)
Consumo de Bebidas Alcohólicas , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Psoriasis/patología , Piel/efectos de los fármacos , Animales , Quimiocina CCL20/efectos de los fármacos , Quimiocina CCL20/metabolismo , Perfilación de la Expresión Génica , Imiquimod/toxicidad , Inductores de Interferón/toxicidad , Interleucina-17/genética , Interleucina-23/genética , Interleucinas/genética , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Ratones , Psoriasis/genética , Piel/metabolismo , Piel/patología , Células Th17/efectos de los fármacos , Células Th17/metabolismo , Interleucina-22
14.
Bioorg Med Chem Lett ; 30(24): 127613, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33075488

RESUMEN

Type I Interferon (IFN) signaling plays an important role in the immune defense system against virus infection and in the innate immune response, thus IFNs are widely used as anti-viral agents and treatment for immune disorder or cancer. However, there is a growing demand for novel small-molecule IFN inducer due to tolerance, toxicity, or short duration of action following direct administration of IFNs. In this study, we assessed arylpiperazine (ARP) as a new core skeleton of IFN inducer. To investigate structure-activity relationship, we designed and synthesized a series of ARP analogues and evaluated the ability to stimulate IFN response in THP-1 human monocyte cells. Compound 5i was identified as a potent type I IFN inducer as it significantly increased cytokine secretion and increased expression of various IFN-stimulating genes which are representative biomarkers of type I IFN pathway. Our results suggested a beneficial therapeutic potential of 5i as an anti-viral agent.


Asunto(s)
Inductores de Interferón/química , Inductores de Interferón/farmacología , Monocitos/efectos de los fármacos , Piperazinas/química , Piperazinas/farmacología , Diseño de Fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Inductores de Interferón/síntesis química , Interferón Tipo I/agonistas , Interferón Tipo I/inmunología , Monocitos/inmunología , Piperazinas/síntesis química , Células THP-1
15.
Fish Shellfish Immunol ; 97: 656-668, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31891812

RESUMEN

AquAdvantage Salmon (growth hormone transgenic female triploid Atlantic salmon) are a faster-growing alternative to conventional farmed diploid Atlantic salmon. To investigate optimal rearing conditions for their commercial production, a laboratory study was conducted in a freshwater recirculating aquaculture system (RAS) to examine the effect of rearing temperature (10.5 °C, 13.5 °C, 16.5 °C) on their antiviral immune and stress responses. When each temperature treatment group reached an average weight of 800 g, a subset of fish were intraperitoneally injected with either polyriboinosinic polyribocytidylic acid (pIC, a viral mimic) or an equal volume of sterile phosphate-buffered saline (PBS). Blood and head kidney samples were collected before injection and 6, 24 and 48 h post-injection (hpi). Transcript abundance of 7 antiviral biomarker genes (tlr3, lgp2, stat1b, isg15a, rsad2, mxb, ifng) was measured by real-time quantitative polymerase chain reaction (qPCR) on head kidney RNA samples. Plasma cortisol levels from blood samples collected pre-injection and from pIC and PBS groups at 24 hpi were quantified by ELISA. While rearing temperature and treatment did not significantly affect circulating cortisol, all genes tested were significantly upregulated by pIC at all three temperatures (except for tlr3, which was only upregulated in the 10.5 °C treatment). Target gene activation was generally observed at 24 hpi, with most transcript levels decreasing by 48 hpi in pIC-injected fish. Although a high amount of biological variability in response to pIC was evident across all treatments, rearing temperature significantly influenced transcript abundance and/or fold-changes comparing time- and temperature-matched pIC- and PBS-injected fish for several genes (tlr3, lgp2, stat1b, isg15a, rsad2 and ifng) at 24 hpi. As an example, significantly higher fold-changes of rsad2, isg15a and ifng were found in fish reared at 10.5 °C when compared to 16.5 °C. Multivariate analysis confirmed that rearing temperature modulated antiviral immune response. The present experiment provides novel insight into the relationship between rearing temperature and innate antiviral immune response in AquAdvantage Salmon.


Asunto(s)
Hormona del Crecimiento/inmunología , Inmunidad Innata , Salmo salar/inmunología , Temperatura , Triploidía , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/inmunología , Acuicultura/métodos , Femenino , Expresión Génica/inmunología , Hormona del Crecimiento/genética , Inductores de Interferón/administración & dosificación , Inductores de Interferón/inmunología , Poli I-C/administración & dosificación , Poli I-C/inmunología , Salmo salar/genética , Estrés Fisiológico/efectos de los fármacos , Virosis/inmunología , Virosis/veterinaria
16.
J Immunol ; 200(8): 2978-2986, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29507107

RESUMEN

In the steady state, tumors harbor several populations of dendritic cells (DCs) and myeloid cells that are key regulators of the intratumoral immune environment. Among these cells, migratory CD103+ cross-presenting DCs are thought to be critical for tumor-specific CTL responses and tumor resistance. However, it is unclear whether this prominent role also extends to immunotherapy. We used a murine orthotopic mammary tumor model, as well as Clec9A-diphtheria toxin receptor mice that can be depleted of the specialized cross-presenting CD8α+ and CD103+ DC1 subsets, to investigate the role of these DCs in immunotherapy. Treatment with monosodium urate crystals and mycobacteria at the tumor site delayed tumor growth and required DC1s for efficacy. In contrast, treatment with poly I:C was equally effective regardless of DC1 depletion. Neither treatment affected myeloid-derived suppressor cell numbers in the spleen or tumor. Similar experiments using subcutaneous B16 melanoma tumors in BATF3-knockout mice confirmed that CD103+ DCs were not necessary for successful poly I:C immunotherapy. Nevertheless, adaptive immune responses were essential for the response to poly I:C, because mice depleted of CD8+ T cells or all DC subsets were unable to delay tumor growth. In vivo experiments showed that DC1 and DC2 subsets were able to take up tumor Ags, with DC2s making up the larger proportion of lymph node DCs carrying tumor material. Both DC subsets were able to cross-present OVA to OT-I T cells in vitro. Thus, immunotherapy with poly I:C enables multiple DC subsets to cross-present tumor Ag for effective antitumor immune responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Inductores de Interferón/inmunología , Neoplasias Mamarias Experimentales/inmunología , Melanoma Experimental/inmunología , Poli I-C/inmunología , Animales , Reactividad Cruzada/inmunología , Femenino , Inmunoterapia/métodos , Lectinas Tipo C/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores Inmunológicos/inmunología
17.
Int J Med Sci ; 17(12): 1803-1810, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32714083

RESUMEN

Since the end of 2019, a new type of coronavirus pneumonia (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has been spreading rapidly throughout the world. Previously, there were two outbreaks of severe coronavirus caused by different coronaviruses worldwide, namely Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and the Middle East Respiratory Syndrome Coronavirus (MERS-CoV). This article introduced the origin, virological characteristics and epidemiological overview of SARS-CoV-2, reviewed the currently known drugs that may prevent and treat coronavirus, explained the characteristics of the new coronavirus and provided novel information for the prevention and treatment of COVID-19.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/prevención & control , Amidas/farmacología , Amidas/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Betacoronavirus/aislamiento & purificación , Betacoronavirus/fisiología , COVID-19 , Cloroquina/análogos & derivados , Cloroquina/uso terapéutico , Clorpromazina/uso terapéutico , Coronavirus/genética , Infecciones por Coronavirus/genética , Ciclofilinas/antagonistas & inhibidores , Desarrollo de Medicamentos , Reposicionamiento de Medicamentos , Medicamentos Herbarios Chinos/uso terapéutico , Endocitosis/efectos de los fármacos , Humanos , Sueros Inmunes , Inductores de Interferón/uso terapéutico , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Neumonía Viral/genética , Pirazinas/farmacología , Pirazinas/uso terapéutico , Resveratrol/farmacología , Resveratrol/uso terapéutico , SARS-CoV-2 , Vacunas Virales/uso terapéutico , Tratamiento Farmacológico de COVID-19
18.
Int J Mol Sci ; 21(10)2020 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-32456211

RESUMEN

Interferon regulatory factors (IRFs) play diverse roles in the regulation of the innate and adaptive immune responses in various diseases. In psoriasis, IRF2 is known to be involved in pathogenesis, while studies on other IRFs are limited. In this study, we investigated the role of IRF5 in psoriasis using imiquimod-induced psoriasis-like dermatitis. Although IRF5 is known to play a critical role in the induction of proinflammatory cytokines by immune cells, such as dendritic cells (DCs), macrophages, and monocytes, IRF5 deficiency unexpectedly exacerbated psoriasiform skin inflammation. The interferon-α and tumor necrosis factor-α mRNA expression levels were decreased, while levels of Th17 cytokines including IL-17, IL-22, and IL-23 were increased in IRF5-deficient mice. Furthermore, IL-23 expression in DCs from IRF5-deficient mice was upregulated both in steady state and after toll-like receptor 7/8 agonist stimulation. Importantly, the expression of IRF4, which is also important for the IL-23 production in DCs, was augmented in DCs from IRF5-deficient mice. Taken together, our results suggest that IRF5 deficiency induces the upregulation of IRF4 in DCs followed by augmented IL-23 production, resulting in the amplification of Th17 responses and the exacerbation of imiquimod-induced psoriasis-like skin inflammation. The regulation of IRF4 or IRF5 expression may be a novel therapeutic approach to psoriasis.


Asunto(s)
Factores Reguladores del Interferón/genética , Interleucinas/metabolismo , Psoriasis/metabolismo , Animales , Células Cultivadas , Células Dendríticas/metabolismo , Femenino , Imiquimod/toxicidad , Inductores de Interferón/toxicidad , Factores Reguladores del Interferón/deficiencia , Factores Reguladores del Interferón/metabolismo , Interferones/genética , Interferones/metabolismo , Interleucinas/genética , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Psoriasis/etiología , Psoriasis/genética , Piel/efectos de los fármacos , Piel/metabolismo , Células Th17/metabolismo
19.
Thorax ; 74(1): 43-50, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30076187

RESUMEN

BACKGROUND: We previously reported that microvesicles (MVs) released by human mesenchymal stem cells (MSC) were as effective as the cells themselves in both Escherichia coli lipopolysaccharide and live bacteria-induced acute lung injury (ALI) mice models. However, it remained unclear whether the biological effect of MSC MV can be applied to human ALI. METHODS: In the current study, we tested the therapeutic effects of MSC MVs in a well-established ex vivo perfused human model of bacterial pneumonia. Using human donor lungs not used for transplantation, we instilled E. coli bacteria intrabronchially and, 1 hour later, administered MSC MVs into the perfusate as therapy. RESULTS: After 6 hours, instillation of E. coli bacteria caused influx of inflammatory cells, which resulted in significant inflammation, lung protein permeability and pulmonary oedema formation. Administration of MSC MV significantly increased alveolar fluid clearance and reduced protein permeability and numerically lowered the bacterial load in the injured alveolus. The beneficial effect on bacterial killing was more pronounced with pretreatment of MSCs with a Toll-like receptor 3 agonist, polyinosinic:polycytidylic acid (Poly (I:C)), prior to the isolation of MVs. Isolated human alveolar macrophages had increased antimicrobial activity with MSC MV treatment in vitro as well. Although oxygenation and lung compliance levels were similar between injury and treatment groups, administration of MSC MVs numerically decreased median pulmonary artery pressure at 6 hours. CONCLUSIONS: In summary, MSC MVs increased alveolar fluid clearance and reduced lung protein permeability, and pretreatment with Poly (I:C) enhanced the antimicrobial activity of MVs in an ex vivo perfused human lung with severe bacteria pneumonia.


Asunto(s)
Lesión Pulmonar Aguda/fisiopatología , Lesión Pulmonar Aguda/terapia , Tratamiento Basado en Trasplante de Células y Tejidos , Micropartículas Derivadas de Células , Infecciones por Escherichia coli/complicaciones , Células Madre Mesenquimatosas , Neumonía Bacteriana/complicaciones , Proteínas/metabolismo , Alveolos Pulmonares/metabolismo , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Adulto , Anciano , Presión Arterial , Carga Bacteriana , Micropartículas Derivadas de Células/efectos de los fármacos , Femenino , Humanos , Inductores de Interferón/farmacología , Recuento de Leucocitos , Rendimiento Pulmonar , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Persona de Mediana Edad , Neutrófilos , Técnicas de Cultivo de Órganos , Oxígeno/metabolismo , Permeabilidad , Poli I-C/farmacología , Alveolos Pulmonares/microbiología , Alveolos Pulmonares/patología , Arteria Pulmonar , Edema Pulmonar/microbiología , Edema Pulmonar/terapia , Receptor Toll-Like 3/agonistas , Factor de Necrosis Tumoral alfa/metabolismo
20.
J Neuroinflammation ; 16(1): 18, 2019 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-30691477

RESUMEN

BACKGROUND: Important insight into the mechanisms through which gene-environmental interactions cause schizophrenia can be achieved through preclinical studies combining prenatal immune stimuli with disease-related genetic risk modifications. Accumulating evidence associates JNK signalling molecules, including MKK7/MAP2K7, with genetic risk. We tested the hypothesis that Map2k7 gene haploinsufficiency in mice would alter the prenatal immune response to the viral mimetic polyriboinosinic-polyribocytidylic acid (polyI:C), specifically investigating the impact of maternal versus foetal genetic variants. METHODS: PolyI:C was administered to dams (E12.5), and cytokine/chemokine levels were measured 6 h later, in maternal plasma, placenta and embryonic brain. RESULTS: PolyI:C dramatically elevated maternal plasma levels of most cytokines/chemokines. Induction of IL-1ß, IL-2, IL-10, IL-12, TNF-α and CXCL3 was enhanced, while CCL5 was suppressed, in Map2k7 hemizygous (Hz) dams relative to controls. Maternal polyI:C administration also increased embryonic brain chemokines, influenced by both maternal and embryonic genotype: CCL5 and CXCL10 levels were higher in embryonic brains from Map2k7 dams versus control dams; for CCL5, this was more pronounced in Map2k7 Hz embryos. Placental CXCL10 and CXCL12 levels were also elevated by polyI:C, the former enhanced and the latter suppressed, in placentae from maternal Map2k7 Hzs relative to control dams receiving polyI:C. CONCLUSIONS: The results demonstrate JNK signalling as a mediator of MIA effects on the foetus. Since both elevated CXCL10 and supressed CXCL12 compromise developing GABAergic interneurons, the results support maternal immune challenge contributing to schizophrenia-associated neurodevelopmental abnormalities. The influence of Map2k7 on cytokine/chemokine induction converges the genetic and environmental aspects of schizophrenia, and the overt influence of maternal genotype offers an intriguing new insight into modulation of embryonic neurodevelopment by genetic risk.


Asunto(s)
Inductores de Interferón/toxicidad , Sistema de Señalización de MAP Quinasas/fisiología , Poli I-C/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Esquizofrenia/etiología , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , MAP Quinasa Quinasa 7/genética , MAP Quinasa Quinasa 7/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Esquizofrenia/sangre , Factor A de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA