Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.176
Filtrar
1.
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
2.
J Neuroimmunol ; 357: 577620, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34062352

RESUMEN

Neuropsychiatric systemic lupus erythematosus (NPSLE) is the most serious and complicated clinical manifestation of lupus erythematosus. Cognitive dysfunction is the most common symptom of NPSLE. A variety of potential mechanisms or mediators related to the pathogenesis of NPSLE cognitive dysfunction have been proposed. However, the involvement of microglia CD40 has not been reported yet. This study aimed to investigate whether hippocampal microglia CD40 of MRL/MpJ-Faslpr (MRL/lpr) mice was involved in NPSLE cognitive dysfunction. This study found, using quantitative polymerase chain reaction, western blotting and immunohistochemistry, that hippocampal CD40 was aberrantly overexpressed in the MRL/lpr lupus mice. It also determined using flow cytometry and immunofluorescence that the aberrantly overexpressed CD40 was mainly derived from hippocampal microglia. The adeno-associated virus was used to inhibit microglia CD40 expression, and the brain damage and cognitive dysfunction of MRL/lpr mice improved. Also, imiquimod (IMQ)-induced lupus mice had the same NPSLE cognitive dysfunction, brain damage, and overexpressed hippocampal microglia CD40 as MRL/lpr mice. Therefore, IMQ-induced lupus mouse was proposed as one of the mouse models for studying NPSLE cognitive dysfunction for the first time in this study. The findings indicated that hippocampal microglia CD40 was involved in the development of NPSLE cognitive dysfunction, thus providing a novel research direction for the study of the pathogenesis of NPSLE.


Asunto(s)
Antígenos CD40/metabolismo , Disfunción Cognitiva/metabolismo , Hipocampo/metabolismo , Vasculitis por Lupus del Sistema Nervioso Central/metabolismo , Microglía/metabolismo , Animales , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Femenino , Imiquimod/farmacología , Inductores de Interferón/farmacología , Vasculitis por Lupus del Sistema Nervioso Central/complicaciones , Ratones , Ratones Endogámicos MRL lpr
3.
Mol Immunol ; 136: 110-117, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34098343

RESUMEN

Reticuloendothelial virus (REV) is widely found in many domestic poultry areas and results in severe immunosuppression of infected chickens. This increases the susceptibility to other pathogens, which causes economic losses to the poultry industry. The aim of our study was to determine whether polyinosinic-polycytidylic acid [Poly (I: C)] treatment could inhibit REV replication in chicken macrophage-like cell line, HD11. We found that Poly (I: C) treatment could markedly inhibit REV replication in HD11 from 24 to 48 h post infection (hpi). Additionally, Poly (I: C) treatment could switch HD11 from an inactive type into M1-like polarization from 24 to 48 hpi. Furthermore, Poly (I: C) treatment promoted interferon-ß secretion from HD11 post REV infection. Moreover, Toll-like receptor-3 (TLR-3) mRNA and protein levels in HD11 treated with Poly (I: C) were markedly increased compared to those of HD11 not treated with Poly (I: C). The above results suggested that Poly (I: C) treatment switches HD11 into M1-like polarization to secret more interferon-ß and activate TLR-3 signaling, which contributes to block REV replication. Our findings provide a theoretical reference for further studying the underlying pathogenic mechanism of REV and Poly (I: C) as a potential therapeutic intervention against REV infection.


Asunto(s)
Antivirales/farmacología , Inductores de Interferón/farmacología , Poli I-C/farmacología , Virus de la Reticuloendoteliosis Aviar/crecimiento & desarrollo , Receptor Toll-Like 3/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Pollos , Interferón beta/biosíntesis , Interferón beta/metabolismo , Macrófagos/inmunología , Macrófagos/virología , Virus de la Reticuloendoteliosis Aviar/efectos de los fármacos , Infecciones por Retroviridae/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Infecciones Tumorales por Virus/tratamiento farmacológico
4.
J Leukoc Biol ; 110(6): 1197-1208, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33988261

RESUMEN

Macrophage phagocytosis plays essential roles in antitumor immunity. CD47/SIRPα phagocytosis checkpoint blockade has demonstrated therapeutic potential in several hematopoietic cancers, but recent clinical studies reported very limited efficacy against solid malignancies. Here, we show that polyinosinic-polycytidylic acid (Poly(I:C)), a synthetic analog of double-stranded RNA, enhances the antitumor activity of CD47 blockade in colorectal cancer in vitro and in vivo. Poly(I:C) activation leads to a potent immune response characterized by the production of proinflammatory cytokines, especially IL-6. Stimulation with IL-6 promotes the PI3K signaling and cytoskeletal reorganization required for macrophage phagocytosis mediated by CD47 blockade. Our findings demonstrate the potential of Poly(I:C) to synergize the efficacy of CD47 blockade therapy and a novel role for IL-6 in macrophage phagocytosis, which provide new strategy for combinational cancer immunotherapy.


Asunto(s)
Neoplasias Colorrectales/inmunología , Inmunoterapia/métodos , Interleucina-6/biosíntesis , Macrófagos/inmunología , Poli I-C/farmacología , Animales , Antígeno CD47/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/inmunología , Inductores de Interferón/farmacología , Ratones Endogámicos C57BL , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología
5.
JCI Insight ; 6(8)2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33884963

RESUMEN

It remains unresolved how retinal pigment epithelial cell metabolism is regulated following immune activation to maintain retinal homeostasis and retinal function. We exposed retinal pigment epithelium (RPE) to several stress signals, particularly Toll-like receptor stimulation, and uncovered an ability of RPE to adapt their metabolic preference on aerobic glycolysis or oxidative glucose metabolism in response to different immune stimuli. We have identified interleukin-33 (IL-33) as a key metabolic checkpoint that antagonizes the Warburg effect to ensure the functional stability of the RPE. The identification of IL-33 as a key regulator of mitochondrial metabolism suggests roles for the cytokine that go beyond its extracellular "alarmin" activities. IL-33 exerts control over mitochondrial respiration in RPE by facilitating oxidative pyruvate catabolism. We have also revealed that in the absence of IL-33, mitochondrial function declined and resultant bioenergetic switching was aligned with altered mitochondrial morphology. Our data not only shed new light on the molecular pathway of activation of mitochondrial respiration in RPE in response to immune stressors but also uncover a potentially novel role of nuclear intrinsic IL-33 as a metabolic checkpoint regulator.


Asunto(s)
Respiración de la Célula/fisiología , Glucólisis/fisiología , Interleucina-33/metabolismo , Mitocondrias/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular , Metabolismo Energético , Técnicas de Silenciamiento del Gen , Glucólisis/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Inductores de Interferón/farmacología , Interleucina-33/efectos de los fármacos , Interleucina-33/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Oxidantes/farmacología , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo , Poli I-C/farmacología , Cultivo Primario de Células , Ácido Pirúvico/metabolismo
6.
Cancer Res ; 81(12): 3374-3386, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33837043

RESUMEN

Screening for sensitizers of cancer cells to TRAIL-mediated apoptosis identified a natural product of the 17ß-hydroxywithanolide (17-BHW) class, physachenolide C (PCC), as a promising hit. In this study, we show that PCC was also able to sensitize melanoma and renal carcinoma cells to apoptosis in response not only to TRAIL, but also to the synthetic polynucleotide poly I:C, a viral mimetic and immune activator, by reducing levels of antiapoptotic proteins cFLIP and Livin. Both death receptor and TLR3 signaling elicited subsequent increased assembly of a proapoptotic ripoptosome signaling complex. Administration of a combination of PCC and poly I:C in human M14 melanoma xenograft and a syngeneic B16 melanoma model provided significant therapeutic benefit as compared with individual agents. In addition, PCC enhanced melanoma cell death in response to activated human T cells in vitro and in vivo in a death ligand-dependent manner. Biochemical mechanism-of-action studies established bromo and extraterminal domain (BET) proteins as major cellular targets of PCC. Thus, by targeting of BET proteins to reduce antiapoptotic proteins and enhance caspase-8-dependent apoptosis of cancer cells, PCC represents a unique agent that can potentially be used in combination with various immunotherapeutic approaches to promote tumor regression and improve outcome. SIGNIFICANCE: These findings demonstrate that PCC selectively sensitizes cancer cells to immune-mediated cell death, potentially improving the efficacy of cancer immunotherapies. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/12/3374/F1.large.jpg.


Asunto(s)
Productos Biológicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Inmunoterapia/métodos , Melanoma Experimental/tratamiento farmacológico , Poli I-C/farmacología , Factores de Transcripción/antagonistas & inhibidores , Witanólidos/farmacología , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Proliferación Celular , Quimioterapia Combinada , Femenino , Humanos , Inductores de Interferón/farmacología , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Methods Mol Biol ; 2239: 153-162, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33226618

RESUMEN

The discovery of induced pluripotent stem cells (iPSCs) allows for establishment of human embryonic stem-like cells from various adult human somatic cells (e.g., fibroblasts), without the need for destruction of human embryos. This provides an unprecedented opportunity where patient-specific iPSCs can be subsequently differentiated to many cell types, e.g., cardiac cells and neurons, so that we can use these iPSC-derived cells to study patient-specific disease mechanisms and conduct drug testing and screening. Critically, these cells have unlimited therapeutic potentials, and there are many ongoing clinical trials to investigate the regenerative potentials of these iPSC-derivatives in humans. However, the traditional iPSC reprogramming methods have problem of insertional mutagenesis because of use of the integrating viral vectors. While a number of advances have been made to mitigate this issue, including the use of chemicals, excisable and non-integrating vectors, and use of the modified mRNA, safety remains a concern. Both integrating and non-integrating methods also suffer from many other limitations including low efficiency, variability, and tumorigenicity. The non-integrating mRNA reprogramming is of high efficiency, but it is sensitive to reagents and need approaches to reduce the immunogenic reaction. An alternative non-integrating and safer way of generating iPSCs is via direct delivery of recombinant cell-penetrating reprogramming proteins into the cells to be reprogrammed, but reprogramming efficiency of the protein-based approach is extremely low compared to the conventional virus-based nuclear reprogramming. Herein, we describe detailed steps for efficient generation of human iPSCs by protein-based reprogramming in combination with stimulation of the Toll-like receptor 3 (TLR3) innate immune pathway.


Asunto(s)
Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Poli I-C/farmacología , Proteínas Recombinantes/metabolismo , Receptor Toll-Like 3/metabolismo , Técnicas de Cultivo de Célula/métodos , Permeabilidad de la Membrana Celular , Células Cultivadas , Criopreservación , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Inductores de Interferón/farmacología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Péptidos/química , Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Recombinantes/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal/genética , Receptor Toll-Like 3/genética
8.
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
9.
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
10.
J Infect ; 81(4): e1-e10, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32707230

RESUMEN

OBJECTIVES: Respiratory and intestinal tract are two primary target organs of SARS-CoV-2 infection. However, detailed characterization of the host-virus interplay in infected human lung and intestinal epithelial cells is lacking. METHODS: We utilized immunofluorescence assays, flow cytometry, and RT-qPCR to delineate the virological features and the innate immune response of the host cells against SARS-CoV-2 infection in two prototype human cell lines representing the human lung (Calu3) and intestinal (Caco2) epithelium when compared with SARS-CoV. RESULTS: Lung epithelial cells were significantly more susceptible to SARS-CoV-2 compared to SARS-CoV. However, SARS-CoV-2 infection induced an attenuated pro-inflammatory cytokines/chemokines induction and type I and type II IFN responses. A single dose of 10 U/mL interferon-ß (IFNß) pretreatment potently protected both Calu3 and Caco2 against SARS-CoV-2 infection. Interestingly, SARS-CoV-2 was more sensitive to the pretreatment with IFNß and IFN inducer than SARS-CoV in Calu3. CONCLUSIONS: Despite robust infection in both human lung and intestinal epithelial cells, SARS-CoV-2 could attenuate the virus-induced pro-inflammatory response and IFN response. Pre-activation of the type I IFN signaling pathway primed a highly efficient antiviral response in the host against SARS-CoV-2 infection, which could serve as a potential therapeutic and prophylactic maneuver to COVID-19 patients.


Asunto(s)
Infecciones por Coronavirus/inmunología , Inductores de Interferón/farmacología , Interferón beta/farmacología , Mucosa Intestinal/inmunología , Neumonía Viral/inmunología , Mucosa Respiratoria/inmunología , Síndrome Respiratorio Agudo Grave/inmunología , Antivirales/farmacología , Betacoronavirus/inmunología , COVID-19 , Células CACO-2 , Línea Celular Tumoral , Infecciones por Coronavirus/tratamiento farmacológico , Células Epiteliales/virología , Humanos , Inmunidad Innata , Pulmón/inmunología , Pandemias , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19
11.
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
12.
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
13.
Colloids Surf B Biointerfaces ; 182: 110352, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31306831

RESUMEN

Psoriasis is a severe disfiguring skin disease affecting approximately 3% of people worldwide and negatively affecting their daily lives. The pathogenesis of psoriasis is complicated, and typical therapeutic strategies for psoriasis mainly focus on anti-inflammation. Considering the side effects, withdrawal rebound, high cost, and many other disadvantages of existing treatments, we developed a new topical therapeutic formulation consisting of niosomes loaded with celastrol, a triterpenoid extracted from Tripterygium. Celastrol niosomes were prepared by the thin film hydration method and probe sonication. The niosomes were composed of Span 20, Span 60, and cholesterol at a weight ratio of 3:1:1. The particle size of the niosomes was approximately 147 nm, with yield of up to 90%. Celastrol niosomes showed improved in vitro permeation ability compared to the raw drug. In our in vivo study, celastrol niosomes effectively alleviated erythema and scaling on the dorsal skin of psoriasis mouse models. Spleen weight and the levels of cytokines, including IL-22, IL-23, and IL-17, decreased after the treatment, indicating the high therapeutic potential of this formulation for psoriasis. In conclusion, encapsulation of celastrol by niosomes increased the water-solubility and permeation of celastrol into the skin, significantly improving its anti-psoriasis activity in mice.


Asunto(s)
Antiinflamatorios/farmacología , Liposomas/química , Psoriasis/tratamiento farmacológico , Piel/efectos de los fármacos , Tripterygium/química , Triterpenos/farmacología , Administración Cutánea , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/metabolismo , Biomarcadores/metabolismo , Colesterol/química , Modelos Animales de Enfermedad , Composición de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/métodos , Femenino , Hexosas/química , Imiquimod/farmacología , Inductores de Interferón/farmacología , Interleucinas/antagonistas & inhibidores , Interleucinas/inmunología , Interleucinas/metabolismo , Liposomas/administración & dosificación , Liposomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Triterpenos Pentacíclicos , Extractos Vegetales/química , Psoriasis/inmunología , Psoriasis/metabolismo , Psoriasis/patología , Piel/inmunología , Piel/patología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología , Triterpenos/aislamiento & purificación , Triterpenos/metabolismo
14.
Dev Comp Immunol ; 100: 103423, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31254564

RESUMEN

Rainbow trout (Oncorhynchus mykiss) face low environmental temperatures over winter months and during extreme low temperature events. Suboptimal temperatures are known to negatively impact the teleost immune system, although there is mixed evidence in rainbow trout as to the effect on the endogenous antigen processing and presentation pathway (EAPP). The EAPP is an important pathway for antiviral defense that involves the presentation of endogenous peptides on the cell surface for recognition by cytotoxic T cells. Using a rainbow trout hypodermal fibroblast (RTHDF) cell line as an in vitro model, we determined that constitutive EAPP transcript levels are not impaired at low temperature, but induction of up-regulation of these transcripts is delayed at the suboptimal temperature following exposure to poly(I:C) or viral haemorrhagic septicaemia virus IVb, which was still able to enter and replicate in the cell line at 4 °C, albeit with reduced efficiency. The delay in the induction of EAPP mRNA level up-regulation following poly(I:C) stimulation coincided with a delay in ifn1 transcript levels and secretion, which is important since interferon-stimulated response elements were identified in the promoter regions of the EAPP-specific members of the pathway, implying that IFN1 is involved in the regulation of these genes. Our results suggest that the ability of rainbow trout to mount an effective immune response to viral pathogens may be lessened at suboptimal temperatures.


Asunto(s)
Frío/efectos adversos , Fibroblastos/inmunología , Enfermedades de los Peces/inmunología , Proteínas de Peces/metabolismo , Oncorhynchus mykiss/inmunología , Aclimatación/inmunología , Animales , Presentación de Antígeno , Línea Celular , Fibroblastos/metabolismo , Enfermedades de los Peces/virología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Inductores de Interferón/farmacología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Novirhabdovirus/inmunología , Oncorhynchus mykiss/virología , Poli I-C/farmacología , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
15.
J Immunol Res ; 2019: 1749803, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31093508

RESUMEN

Plasmacytoid dendritic cells (pDCs) express high levels of the toll-like receptors (TLRs) TLR7 and TLR9. In response to TLR7 and TLR9 ligands, pDCs are primary producers of type I interferons. Our previous study demonstrated that pDCs activated by the TLR7 ligand imiquimod (IMQ) and the TLR9 ligand CpG A can kill breast cancer cells in vitro and inhibit tumor growth in vivo. Moreover, we observed a distinctive morphological, phenotypic change in pDCs after activation by IMQ and CpG A. However, the effect of other TLR7 and TLR9 ligands on pDCs remains less understood. In this study, we treat pDCs with the TLR7 ligand IMQ, TLR7/8 ligands (CL097 and CL075), and three TLR9 ligands (different types of CpGs). The size of pDCs increased significantly after activation by TLR7, or TLR7/8 ligands. TLR7, TLR7/8, and TLR9 ligands similarly modulated cytokine release, as well as protein expression of pDC markers, costimulatory molecules, and cytotoxic molecules. Interestingly, TLR7/8 ligands, especially CL097, induced stronger responses. These results are relevant to the further study of the role and mechanism of pDC-induced antitumor effects and may aid in the development of a new strategy for future tumor immunotherapy.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Citocinas/inmunología , Células Dendríticas/efectos de los fármacos , Imiquimod/farmacología , Glicoproteínas de Membrana/inmunología , Oligodesoxirribonucleótidos/farmacología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/inmunología , Animales , Femenino , Imidazoles/farmacología , Inductores de Interferón/farmacología , Interferón Tipo I/inmunología , Ligandos , Ratones , Ratones Endogámicos BALB C , Quinolinas/farmacología , Tiazoles/farmacología
16.
Int J Mol Sci ; 20(9)2019 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-31067687

RESUMEN

Rhinovirus (RV) is the predominant virus causing respiratory tract infections. Bronchobini® is a low dose multi component, multi target preparation used to treat inflammatory respiratory diseases such as the common cold, described to ease severity of symptoms such as cough and viscous mucus production. The aim of the study was to assess the efficacy of Bronchobini® in RV infection and to elucidate its mode of action. Therefore, Bronchobini®'s ingredients (BRO) were assessed in an ex vivo model of RV infection using mouse precision-cut lung slices, an organotypic tissue capable to reflect the host immune response to RV infection. Cytokine profiles were assessed using enzyme-linked immunosorbent assay (ELISA) and mesoscale discovery (MSD). Gene expression analysis was performed using Affymetrix microarrays and ingenuity pathway analysis. BRO treatment resulted in the significant suppression of RV-induced antiviral and pro-inflammatory cytokine release. Transcriptome analysis revealed a multifactorial mode of action of BRO, with a strong inhibition of the RV-induced pro-inflammatory and antiviral host response mediated by nuclear factor kappa B (NFkB) and interferon signaling pathways. Interestingly, this was due to priming of these pathways in the absence of virus. Overall, BRO exerted its beneficial anti-inflammatory effect by priming the antiviral host response resulting in a reduced inflammatory response to RV infection, thereby balancing an otherwise excessive inflammatory response.


Asunto(s)
Antivirales/farmacología , Inductores de Interferón/farmacología , Interferones/metabolismo , Pulmón/efectos de los fármacos , Infecciones por Picornaviridae/tratamiento farmacológico , Extractos Vegetales/farmacología , Transcriptoma , Animales , Antivirales/uso terapéutico , Femenino , Inductores de Interferón/uso terapéutico , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/virología , Extractos Vegetales/uso terapéutico , Rhinovirus/efectos de los fármacos , Rhinovirus/patogenicidad , Transducción de Señal
17.
eNeuro ; 6(2)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31016229

RESUMEN

Sex differences are documented in psychiatric and neurological disorders, yet most preclinical animal research has been conducted in males only. There is a need to better understand of the nature of sex differences in brain disease in order to meet the needs of psychiatric patients. We present the behavior profile of adult female offspring produced using a maternal immune activation (MIA) model where pregnant rats receive an immune stimulant and the offspring typically show various abnormalities consistent with psychiatric illnesses such as schizophrenia and autism. The results in female offspring were compared to a previously published cohort of their male siblings (Lins et al., 2018). We examined prepulse inhibition (PPI), sociability, MK-801-induced locomotor activity, crossmodal object recognition (CMOR), and oddity discrimination; behaviors relevant to the positive, negative, and cognitive symptoms of schizophrenia. No between-treatment differences in PPI or locomotor activity were noted. Tactile memory was observed in the control and treated female offspring, visual recognition memory was deficient in the polyinosinic:polycytidylic acid (polyI:C) offspring only, and both groups lacked crossmodal recognition. PolyI:C offspring were impaired in oddity preference and had reduced preference for a stranger conspecific in a sociability assay. Systemic maternal CXCL1, IL-6, and TNF-a levels 3 h after polyI:C treatment were determined, but no relationship was found between these cytokines and the behavior seen in the adult female offspring. Overall, female offspring of polyI:C-treated dams display an array of behavior abnormalities relevant to psychiatric illnesses such as schizophrenia similar to those previously reported in male rats.


Asunto(s)
Conducta Animal/fisiología , Complicaciones del Embarazo/inmunología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Inhibición Prepulso/fisiología , Reconocimiento en Psicología/fisiología , Conducta Social , Animales , Modelos Animales de Enfermedad , Femenino , Inductores de Interferón/farmacología , Poli I-C/farmacología , Embarazo , Ratas , Ratas Sprague-Dawley , Esquizofrenia/fisiopatología , Caracteres Sexuales
18.
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
19.
Artículo en Inglés | MEDLINE | ID: mdl-30292753

RESUMEN

Bcl6B, also known as BAZF, plays important roles in the immune response, repression of cancers, and maintenance of spermatogonial stem cells in mammals. In this study, the homologous gene bcl6b and its 5 alternative splicing variants, namely bcl6bX1 to bcl6bX5, were isolated from medaka fish, Oryzias latipes. Medaka bcl6b possesses conserved domains such as BTB domain, RD2 domain and four zinc fingers. Medaka bcl6bX1 to bcl6bX3 possess all three previously mentioned domains with minor differences in sequences. Medaka bcl6bX4 possesses only the BTB domain due to premature stopping, and bcl6bX5 possesses both the BTB domain and zinc fingers without the RD2 domain. Medaka bcl6b was expressed in the tissues including the brain, heart, gill, muscle, spleen, kidney, intestine, ovary and testes of adult fish. Medaka bcl6b was expressed in the embryos from very early stage, and could be detected clearly in the developing eyes by RT-PCR and in situ hybridization. Medaka bcl6b could respond to the stimuli of polyI:C and LPS in the kidney and spleen. Medaka bcl6bX1 to bcl6bX3 were the majority of the variants expressed in the adult tissues and the embryos, and were the major response to the stimulation of polyI:C and LPS in the spleen. These results suggested that bcl6b, including its isoforms, could function in various tissues and embryogenesis. Moreover, bcl6b might be a factor for immune response in medaka.


Asunto(s)
Empalme Alternativo , Desarrollo Embrionario , Proteínas de Peces/metabolismo , Regulación del Desarrollo de la Expresión Génica , Oryzias/fisiología , Proteínas Represoras/metabolismo , Empalme Alternativo/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Embrión no Mamífero/inmunología , Embrión no Mamífero/fisiología , Ojo/embriología , Ojo/metabolismo , Proteínas de Peces/química , Proteínas de Peces/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Inductores de Interferón/farmacología , Riñón/efectos de los fármacos , Riñón/crecimiento & desarrollo , Riñón/inmunología , Riñón/metabolismo , Lipopolisacáridos/farmacología , Especificidad de Órganos , Oryzias/embriología , Oryzias/crecimiento & desarrollo , Oryzias/inmunología , Poli I-C/farmacología , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Distribución Aleatoria , Proteínas Represoras/química , Proteínas Represoras/genética , Alineación de Secuencia , Bazo/efectos de los fármacos , Bazo/crecimiento & desarrollo , Bazo/inmunología , Bazo/metabolismo
20.
PLoS One ; 13(11): e0206827, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30383838

RESUMEN

Hyperuricaemia is associated with various metabolic dysfunctions including obesity, type 2 diabetes mellitus, hypertension and in general metabolic syndrome, which are all associated with increased risk of cancer. However, the direct association between elevated uricemia and cancer mortality still remains unclear. In this study, we used a mouse model of hyperuricemia, the Urahplt2/plt2 (PLT2) mouse, to investigate the effect of high uric acid levels on anti-tumor immune responses and tumor growth. In normo-uricaemic C57BL/6 mice injected with B16 melanomas, immunotherapy by treatment with Poly I:C at the tumor site delayed tumor growth compared to PBS treatment. In contrast, Poly I:C-treated hyper-uricaemic PLT2 mice were unable to delay tumor growth. Conventional and monocyte-derived dendritic cells in the tumor-draining lymph nodes (dLN) of C57BL/6 and PLT2 mice were similarly increased after Poly I:C immunotherapy, and expressed high levels of CD40 and CD86. CD8+ T cells in the tumor-dLN and tumor of both WT and PLT2 mice were also increased after Poly I:C immunotherapy, and were able to secrete increased IFNγ upon in vitro restimulation. Surprisingly, tumor-specific CD8+ T cells in dLN were less abundant in PLT2 mice compared to C57BL/6, but showed a greater ability to proliferate even in the absence of cognate antigen. These data suggest that hyperuricaemia may affect the functionality of CD8+ T cells in vivo, leading to dysregulated T cell proliferation and impaired anti-tumor activity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Hiperuricemia/inmunología , Inductores de Interferón/farmacología , Melanoma Experimental/inmunología , Poli I-C/farmacología , Neoplasias Cutáneas/inmunología , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Carcinogénesis/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Humanos , Hiperuricemia/sangre , Hiperuricemia/genética , Hiperuricemia/metabolismo , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/inmunología , Inmunoterapia/métodos , Inductores de Interferón/uso terapéutico , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Masculino , Melanoma Experimental/sangre , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Poli I-C/uso terapéutico , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/metabolismo , Resultado del Tratamiento , Ácido Úrico/sangre , Ácido Úrico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...