Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 53(2): 384-397.e5, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32673565

RESUMEN

Dysregulated Th17 cell responses underlie multiple inflammatory and autoimmune diseases, including autoimmune uveitis and its animal model, EAU. However, clinical trials targeting IL-17A in uveitis were not successful. Here, we report that Th17 cells were regulated by their own signature cytokine, IL-17A. Loss of IL-17A in autopathogenic Th17 cells did not reduce their pathogenicity and instead elevated their expression of the Th17 cytokines GM-CSF and IL-17F. Mechanistic in vitro studies revealed a Th17 cell-intrinsic autocrine loop triggered by binding of IL-17A to its receptor, leading to activation of the transcription factor NF-κB and induction of IL-24, which repressed the Th17 cytokine program. In vivo, IL-24 treatment ameliorated Th17-induced EAU, whereas silencing of IL-24 in Th17 cells enhanced disease. This regulatory pathway also operated in human Th17 cells. Thus, IL-17A limits pathogenicity of Th17 cells by inducing IL-24. These findings may explain the disappointing therapeutic effect of targeting IL-17A in uveitis.


Asunto(s)
Citocinas/metabolismo , Interleucina-17/metabolismo , Células Th17/patología , Uveítis/patología , Adulto , Animales , Citocinas/genética , Modelos Animales de Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interleucina-17/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Células Th17/inmunología , Uveítis/inmunología , Adulto Joven
2.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34782464

RESUMEN

Regulatory B cells (Breg cells) that secrete IL-10 or IL-35 (i35-Breg) play key roles in regulating immunity in tumor microenvironment or during autoimmune and infectious diseases. Thus, loss of Breg function is implicated in development of autoimmune diseases while aberrant elevation of Breg prevents sterilizing immunity, exacerbates infectious diseases, and promotes cancer metastasis. Breg cells identified thus far are largely antigen-specific and derive mainly from B2-lymphocyte lineage. Here, we describe an innate-like IL-27-producing natural regulatory B-1a cell (i27-Breg) in peritoneal cavity and human umbilical cord blood. i27-Bregs accumulate in CNS and lymphoid tissues during neuroinflammation and confers protection against CNS autoimmune disease. i27-Breg immunotherapy ameliorated encephalomyelitis and uveitis through up-regulation of inhibitory receptors (Lag3, PD-1), suppression of Th17/Th1 responses, and propagating inhibitory signals that convert conventional B cells to regulatory lymphocytes that secrete IL-10 and/or IL-35 in eye, brain, or spinal cord. Furthermore, i27-Breg proliferates in vivo and sustains IL-27 secretion in CNS and lymphoid tissues, a therapeutic advantage over administering biologics (IL-10, IL-35) that are rapidly cleared in vivo. Mutant mice lacking irf4 in B cells exhibit exaggerated increase of i27-Bregs with few i35-Bregs, while mice with loss of irf8 in B cells have abundance of i35-Bregs but defective in generating i27-Bregs, identifying IRF8/BATF and IRF4/BATF axis in skewing B cell differentiation toward i27-Breg and i35-Breg developmental programs, respectively. Consistent with its developmental origin, disease suppression by innate i27-Bregs is neither antigen-specific nor disease-specific, suggesting that i27-Breg would be effective immunotherapy for a wide spectrum of autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades del Sistema Nervioso Central/inmunología , Interleucina-27/metabolismo , Enfermedades Neuroinflamatorias/inmunología , Animales , Linfocitos B Reguladores/inmunología , Diferenciación Celular , Encefalitis , Factores Reguladores del Interferón , Interleucina-10 , Ratones , Uveítis/inmunología
3.
J Autoimmun ; 114: 102507, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32593472

RESUMEN

IFN-γ and IL-17A can each elicit ocular autoimmunity independently of the other. Since absence of IFN-γ or IL-17A individually failed to abolish pathology of experimental autoimmune uveitis (EAU), we examined EAU development in the absence of both these cytokines. Ifng-/-Il17a-/- mice were fully susceptible to EAU with a characteristic eosinophilic ocular infiltrate, as opposed to a mononuclear infiltrate in WT mice. Retinal pathology in double-deficient mice was ameliorated when eosinophils were genetically absent or their migration was blocked, supporting a pathogenic role for eosinophils in EAU in the concurrent absence of IFN-γ and IL-17A. In EAU-challenged Ifng-/-Il17a-/- mice, ocular infiltrates contained increased GM-CSF-producing CD4+ T cells, and supernatants of retinal antigen-stimulated splenocytes contained enhanced levels of GM-CSF that contributed to activation and migration of eosinophils in vitro. Systemic or local blockade of GM-CSF ameliorated EAU in Ifng-/-Il17a-/- mice, reduced eosinophil peroxidase levels in the eye and in the serum and decreased eosinophil infiltration to the eye. These results support the interpretation that, in the concurrent absence of IFN-γ and IL-17A, GM-CSF takes on a major role as an inflammatory effector cytokine and drives an eosinophil-dominant pathology. Our findings may impact therapeutic strategies aiming to target IFN-γ and IL-17A in autoimmune uveitis.


Asunto(s)
Autoinmunidad , Eosinofilia/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Retinitis/etiología , Retinitis/metabolismo , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Eosinófilos/inmunología , Eosinófilos/metabolismo , Eosinófilos/patología , Interferón gamma/genética , Interleucina-17/genética , Ratones , Ratones Noqueados , Retinitis/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
4.
Mol Vis ; 26: 641-651, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33088168

RESUMEN

Purpose: Tofacitinib is a pan-Janus kinase (JAK) inhibitor that suppresses cytokine signaling and in turn, the cells that participate in inflammatory immunopathogenic processes. We examined the capacity of tofacitinib to inhibit the induction of experimental autoimmune uveitis (EAU) and related immune responses. Methods: EAU was induced in B10.A mice with immunization with bovine interphotoreceptor retinoid-binding protein (IRBP), emulsified in complete Freund's adjuvant (CFA), and a simultaneous injection of pertussis toxin. Tofacitinib, 25 mg/kg, was administered daily, and the vehicle was used for control. EAU development was assessed by histological analysis of the mouse eyes, and related immune responses were assessed by (i) the levels of interferon (IFN)-γ and interleukin (IL)-17, secreted by spleen cells cultured with IRBP; (ii) flow cytometric analysis of intracellular expression by spleen, or eye-infiltrating CD4 or CD8 cells of IFN-γ, IL-17, and their transcription factors, T-bet and RORγt. In addition, the inflammation-related cell markers CD44 and CD62L and Ki67, a proliferation marker, were tested. The proportions of T-regulatory cells expressing FoxP3 were determined by flow cytometric intracellular staining, while levels of antibody to IRBP were measured with enzyme-linked immunosorbent assay (ELISA). Results: Treatment with tofacitinib significantly suppressed the development of EAU and reduced the levels of secreted IFN-γ, but not of IL-17. Further, treatment with tofacitinib reduced in the spleen and eye-infiltrating cells the intracellular expression of IFN-γ and its transcription factor T-bet. In contrast, treatment with tofacitinib had essentially no effect on the intracellular expression of IL-17 and its transcription factor, RORγt. The selective effect of tofacitinib treatment was particularly evident in the CD8 population. Treatment with tofacitinib also increased the population of CD44, but reduced the populations of cells producing CD62L and Ki67. Treatment with tofacitinib had no effect on the proportion of FoxP3 producing regulatory cells and on the antibody production to IRBP. Conclusions: Treatment with tofacitinib inhibited the development of EAU, reduced the production of IFN-γ, but had essentially no effect on the production of IL-17.


Asunto(s)
Ojo/metabolismo , Piperidinas/farmacología , Pirimidinas/farmacología , Células TH1/efectos de los fármacos , Células Th17/efectos de los fármacos , Uveítis/tratamiento farmacológico , Uveítis/inmunología , Animales , Antígenos CD4/sangre , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Antígenos CD8/sangre , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Ojo/efectos de los fármacos , Ojo/patología , Proteínas del Ojo/farmacología , Factores de Transcripción Forkhead/sangre , Receptores de Hialuranos/sangre , Terapia de Inmunosupresión , Interferón gamma/sangre , Interleucina-17/sangre , Antígeno Ki-67/sangre , Selectina L/sangre , Ratones , Piperidinas/administración & dosificación , Pirimidinas/administración & dosificación , Proteínas de Unión al Retinol/farmacología , Células TH1/inmunología , Células Th17/inmunología
5.
Eur J Immunol ; 48(11): 1810-1816, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30218573

RESUMEN

Experimental autoimmune uveitis (EAU), an animal model for severe intraocular inflammatory eye diseases, is mediated by both Th1 and Th17 cells. Here, we examined the capacity of TMP778, a selective inhibitor of RORγt, to inhibit the development of EAU, as well as the related immune responses. EAU was induced in B10.A mice by immunization with interphotoreceptor retinoid-binding protein (IRBP). Treatment with TMP778 significantly inhibited the development of EAU, determined by histological examination. In addition, the treatment suppressed the cellular immune response to IRBP, determined by reduced production of IL-17 and IFN-γ, as well as lower percentages of lymphocytes expressing these cytokines, as compared to vehicle-treated controls. The inhibition of IFN-γ expression by TMP778 is unexpected in view of this compound being a selective inhibitor of RORγt. The observation was further confirmed by the finding of reduced expression of the T-bet (Tbx21) gene, the transcription factor for IFN-γ, by cells of TMP778-treated mice. Thus, these data demonstrate the capacity of TMP778 to inhibit pathogenic autoimmunity in the eye and shed new light on its mode of action in vivo.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Células TH1/efectos de los fármacos , Células Th17/efectos de los fármacos , Uveítis/tratamiento farmacológico , Animales , Enfermedades Autoinmunes/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Femenino , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Ratones , Proteínas de Unión al Retinol/metabolismo , Proteínas de Dominio T Box/metabolismo , Células TH1/metabolismo , Células Th17/metabolismo , Uveítis/metabolismo
6.
J Autoimmun ; 100: 52-61, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30853312

RESUMEN

AS101 is an organotellurium compound with multifaceted immunoregulatory properties that is remarkable for its lack of toxicity. We tested the therapeutic effect of AS101 in experimental autoimmune uveitis (EAU), a model for human autoimmune uveitis. Unexpectedly, treatment with AS101 elicited Treg generation in vivo in otherwise unmanipulated mice. Mice immunized for EAU with the retinal antigen IRBP and treated with AS101 developed attenuated disease, as did AS101-treated recipients of retina-specific T cells activated in vitro. In both settings, eye-infiltrating effector T cells were decreased, whereas regulatory T (Treg) cells in the spleen were increased. Mechanistic studies in vitro revealed that AS101 restricted polarization of retina-specific T cells towards Th1 or Th17 lineage by repressing activation of their respective lineage-specific transcription factors and downstream signals. Retina-specific T cells polarized in vitro towards Th1 or Th17 in the presence of AS101 had impaired ability to induce EAU in naïve recipients. Finally, AS101 promoted differentiation of retina-specific T cells to Tregs in vitro independently of TGF-ß. We conclude that AS101 modulates autoimmune T cells by inhibiting acquisition and expression of effector function and by promoting Treg generation, and suggest that AS101 could be useful as a therapeutic approach for autoimmune uveitis.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Etilenos/farmacología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células Th17/inmunología , Uveítis/tratamiento farmacológico , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Linfocitos T Reguladores/patología , Células TH1/patología , Células Th17/patología , Uveítis/genética , Uveítis/inmunología , Uveítis/patología
7.
Mol Biol Rep ; 46(4): 4443-4452, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31302805

RESUMEN

Containing high concentration of vanadium served by the volcanic bedrock, Jeju ground water has long been known for various implicit health benefits including immune-promotion. Exposure to stress has been reported to be associated with immunosuppression such as reducing lymphocyte population or antibody production due to stress hormones. In this study, we aimed at evaluating the effects of Jeju ground water on chronically stressed mice. C57BL/6 mice were subjected to various stressors such as restraint stress, water swimming stress, heat stress, acoustic stress, and Jeju ground water was supplied for 28 days with two different concentrations, S1 (vanadium 15-20 µg/l, pH 8.3) and S2 (vanadium 20-25 µg/l, pH 8.5). Treatment with Jeju ground water increased CD4+CD8- or CD4-CD8+ single-positive thymocytes. It also increased the proliferation of splenocytes and the populations of CD4+ T cells, CD45R/B220+ B cells, CD11b+ macrophages or Gr-1+ granulocytes in spleen. In addition, the production of IgG was increased in chronically stressed mice by treatment with Jeju ground water. These results suggest vanadium-rich Jeju ground water may be helpful in T cell development in thymus and immune cell proliferation and its function in spleen against chronic stress.


Asunto(s)
Linfocitos T/efectos de los fármacos , Timocitos/efectos de los fármacos , Vanadio/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Femenino , Agua Subterránea/química , Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología , Estrés Fisiológico/fisiología , Timo/inmunología , Vanadio/fisiología
8.
Pharm Biol ; 55(1): 306-319, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27927068

RESUMEN

CONTEXT: Beetroot [Beta vulgaris Linné (Chenopodiaceae)], a vegetable usually consumed as a food or a medicinal plant in Europe, has been reported to have antioxidant and anti-inflammatory properties. Since the lymphohematopoietic system is the most sensitive tissue to ionizing radiation, protecting it from radiation damage is one of the best ways to decrease detrimental effects from radiation exposure. OBJECTIVE: In this study, we evaluated the radio-protective effects of beetroot in hematopoietic stem cells (HSCs) and progenitor cells. MATERIALS AND METHODS: Beetroot extract was administered at a dose of 400 mg/mouse per os (p.o.) three times into C57BL/6 mice and, at day 10 after γ-ray irradiation, diverse molecular presentations were measured and compared against non-irradiated and irradiated mice with PBS treatments. Survival of beetroot-fed and unfed irradiated animal was also compared. RESULTS: Beetroot not only stimulated cell proliferation, but also minimized DNA damage of splenocytes. Beetroot also repopulated S-phase cells and increased Ki-67 or c-Kit positive cells in bone marrow. Moreover, beetroot-treated mice showed notable boosting of differentiation of HSCs into burst-forming units-erythroid along with increased production of IL-3. Also, beetroot-treated mice displayed enhancement in the level of hematocrit and hemoglobin as well as the number of red blood cell in peripheral blood. Beetroot diet improved survival rate of lethally exposed mice with a dose reduction factor (DRF) of 1.1. DISCUSSION AND CONCLUSION: These results suggest that beetroot has the potency to preserve bone marrow integrity and stimulate the differentiation of HSCs against ionizing radiation.


Asunto(s)
Beta vulgaris/química , Médula Ósea/efectos de los fármacos , Rayos gamma/efectos adversos , Hematínicos/farmacología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Factores Inmunológicos/farmacología , Protectores contra Radiación/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Médula Ósea/inmunología , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Células Cultivadas , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Hematínicos/aislamiento & purificación , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/efectos de la radiación , Tolerancia Inmunológica/efectos de la radiación , Factores Inmunológicos/aislamiento & purificación , Interleucina-3/metabolismo , Ratones Endogámicos C57BL , Fitoterapia , Raíces de Plantas , Plantas Medicinales , Protectores contra Radiación/aislamiento & purificación , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/efectos de la radiación , Factores de Tiempo , Irradiación Corporal Total/efectos adversos
9.
Pharm Biol ; 54(12): 2939-2950, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27333995

RESUMEN

CONTEXT: Hallabong [(Citrus unshiu × C. sinensis) X C. reticulata)] (Rutaceae) is a hybrid citrus cultivated in temperate regions of South Korea. Its fruit is well-known for pharmacological properties. OBJECTIVE: This study examined the anti-inflammatory effect of 80% ethanol extract of Hallabong (HE) on concanavalin A (Con A)-stimulated splenocytes and mouse oedema model induced by 12-O-tetradecanoylphorbal acetate (TPA). MATERIALS AND METHODS: Murine splenocytes treated with HE were stimulated with Con A (10 µg/mL, for 24 h) were evaluated for T-cell population and production of inflammatory cytokines IL-2, IL-4 and IFN-γ. Anti-inflammatory effect of topically applied HE (100 µg/20 µL) on TPA (4 µg/20 µL/ear)-induced ear oedema was investigated in mouse model. RESULTS: HE-treated Con A-stimulated murine splenocytes showed a marked decrease in CD44/CD62L+ memory T-cell population, an important marker for anti-inflammatory activity, and a significant inhibition in the production of IL-2 and IFN-γ. HE treatment had reduced the mouse skin oedema (47%) and myeloperoxidase (MPO) activity significantly (40%) in TPA-challenged tissues. More importantly, immunohistochemical localization revealed the suppressed (p < 0.05) expression of inducible nitric oxide (iNOS), cyclooxygenase-2 (COX2). HE decreased the infiltration of CD3+ T cells and F4/80+ macrophages to the site of inflammation and a topical application of HE significantly suppressed the expression of TNF-α (20.2%). DISCUSSION AND CONCLUSION: A topical application of HE can exert a potential anti-inflammatory effect and HE can be explored further as a putative alternative therapeutic agent for inflammatory oedema.


Asunto(s)
Antiinflamatorios/uso terapéutico , Citrus , Modelos Animales de Enfermedad , Edema/tratamiento farmacológico , Extractos Vegetales/uso terapéutico , Bazo/efectos de los fármacos , Acetato de Tetradecanoilforbol/toxicidad , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Citrus sinensis , Relación Dosis-Respuesta a Droga , Oído , Edema/inducido químicamente , Edema/metabolismo , Ratones , Ratones Endogámicos C57BL , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología
10.
J Biol Chem ; 289(24): 16773-89, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24808182

RESUMEN

Lipocalin-2 (LCN2) plays an important role in cellular processes as diverse as cell growth, migration/invasion, differentiation, and death/survival. Furthermore, recent studies indicate that LCN2 expression and secretion by glial cells are induced by inflammatory stimuli in the central nervous system. The present study was undertaken to examine the regulation of LCN2 expression in experimental autoimmune encephalomyelitis (EAE) and to determine the role of LCN2 in the disease process. LCN2 expression was found to be strongly increased in spinal cord and secondary lymphoid tissues after EAE induction. In spinal cords astrocytes and microglia were the major cell types expressing LCN2 and its receptor 24p3R, respectively, whereas in spleens, LCN2 and 24p3R were highly expressed in neutrophils and dendritic cells, respectively. Furthermore, disease severity, inflammatory infiltration, demyelination, glial activation, the expression of inflammatory mediators, and the proliferation of MOG-specific T cells were significantly attenuated in Lcn2-deficient mice as compared with wild-type animals. Myelin oligodendrocyte glycoprotein-specific T cells in culture exhibited an increased expression of Il17a, Ifng, Rorc, and Tbet after treatment with recombinant LCN2 protein. Moreover, LCN2-treated glial cells expressed higher levels of proinflammatory cytokines, chemokines, and MMP-9. Adoptive transfer and recombinant LCN2 protein injection experiments suggested that LCN2 expression in spinal cord and peripheral immune organs contributes to EAE development. Taken together, these results imply LCN2 is a critical mediator of autoimmune inflammation and disease development in EAE and suggest that LCN2 be regarded a potential therapeutic target in multiple sclerosis.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Lipocalinas/metabolismo , Tejido Linfoide/metabolismo , Proteínas Oncogénicas/metabolismo , Médula Espinal/metabolismo , Proteínas de Fase Aguda/genética , Animales , Astrocitos/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , Eliminación de Gen , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Lipocalina 2 , Lipocalinas/genética , Tejido Linfoide/patología , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Neutrófilos/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteínas Oncogénicas/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Médula Espinal/patología , Linfocitos T/metabolismo
11.
Biochim Biophys Acta ; 1830(3): 2820-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23262141

RESUMEN

BACKGROUND: The proven immunomodulatory and immune system activating properties of Ecklonia cava (E. cava) have been attributed to its plentiful polysaccharide content. Therefore, we investigated whether the sulfated polysaccharide (SP) of E. cava specifically activates the protein kinases (MAPKs) and nuclear factor-kappaB (NFkappaB) to incite immune responses. METHODS: To assess immune responsiveness, lymphocytes were isolated from spleens of ICR mice and cultured with SP and its inhibitors. Assays included 3H-thymidine incorporation, flow cytometry, real time polymerase chain reaction (rtPCR), enzyme linked immunosorbent assay (ELISA), intracellular cytokine assay. Western blot, and electrophoretic mobility shift assay (EMSA). RESULTS: SP dose-dependently increased the proliferation of lymphocytes without cytotoxicity. In particular, SP markedly enhanced the proliferation and differentiation of CD3+ mature T cells and CD45R/B220+ pan B cells. Additionally, SP increased the expression and/or production of IL-2, IgG(1a), and IgG(2b) compared to that in untreated cells. The subsequent application of JNK (SP600125), NFkappaB (PDTC), and serine protease (TPCK) inhibitors significantly inhibited the proliferation and IL-2 production of SP-treated lymphocytes as well as the phosphorylation of JNK and IkappaB, the activation of nuclear NFkappaB p65, and binding of NFkappaB p65 DNA. Moreover, co-application of both JNK and NFkappaB inhibitors completely blocked the proliferation of lymphocytes even in the presence of SP. CONCLUSION: These results suggest that SP induced T and B cell responses via both JNK and NFkappaB pathways. GENERAL SIGNIFICANCE: The effect of SP on splenic lymphocyte activation was assayed here for the first time and indicated the underlying functional mechanism.


Asunto(s)
Linfocitos B/efectos de los fármacos , Laminaria/química , MAP Quinasa Quinasa 4/genética , FN-kappa B/genética , Polisacáridos/farmacología , Linfocitos T/efectos de los fármacos , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Mezclas Complejas/química , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Interleucina-2/biosíntesis , Interleucina-2/inmunología , Activación de Linfocitos/efectos de los fármacos , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/inmunología , Ratones , Ratones Endogámicos ICR , FN-kappa B/antagonistas & inhibidores , FN-kappa B/inmunología , Fosforilación/efectos de los fármacos , Polisacáridos/aislamiento & purificación , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Timidina/metabolismo
12.
Nat Biomed Eng ; 8(2): 193-200, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37996615

RESUMEN

Owing to the immunogenicity of adeno-associated viruses (AAVs), gene therapies using AAVs face considerable obstacles. Here, by leveraging ex vivo T-cell assays, the prediction of epitope binding to major histocompatibility complex class-II alleles, sequence-conservation analysis in AAV phylogeny and site-directed mutagenesis, we show that the replacement of amino acid residues in a promiscuous and most immunodominant T-cell epitope in the AAV9 capsid with AAV5 sequences abrogates the immune responses of peripheral blood mononuclear cells to the chimaeric vector while preserving its functions, potency, cellular specificity, transduction efficacy and biodistribution. This rational approach to the immunosilencing of capsid epitopes promiscuously binding to T cells may be applied to other AAV vectors and epitope regions.


Asunto(s)
Cápside , Dependovirus , Cápside/química , Cápside/metabolismo , Dependovirus/genética , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/análisis , Epítopos de Linfocito T/metabolismo , Leucocitos Mononucleares , Distribución Tisular , Proteínas de la Cápside/genética , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo
13.
Front Immunol ; 14: 1211529, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37469509

RESUMEN

During gene therapy trials, immune responses against adeno-associated virus (AAV) vectors are monitored by antibody assays that detect the humoral and T-cell mediated cellular responses to AAV vectors. T cell assays commonly utilize the collection of patients' peripheral blood mononuclear cells (PBMCs) and stimulation with AAV-derived overlapping peptides. We recently described that spontaneous deamidation coincides with T cell epitopes in AAV capsids and that spontaneous deamidation may enhance or decrease immunogenicity in some individuals. This raised the concern for false negative results of antibody detection and PBMC immune monitoring assays because these assays use wild-type (WT) AAV or WT peptides for T cell re-stimulation and these peptides may not re-activate T cells that were stimulated with deamidated AAV capsid. To investigate this concern, we modeled the scenario by expanding T cells with deamidated peptides and evaluated the cross-reactivity of expanded T cells to WT peptides. In the majority of samples, cells that were expanded with deamidated peptides and restimulated with WT peptide had significantly lowered IL-2 and IFN-γ production. Spiking the four deamidated peptides to the WT peptide pool used for re-stimulation, restored the signal and corrected the performance of the assay. We also evaluated the impact of deamidation on anti AAV binding antibodies and did not observe a major impact on seroprevalence detection of AAV9. These data indicate that a high level of deamidation in AAV therapy may result in underestimation or even failure to detect immune responses against WT peptides during cellular immune monitoring.


Asunto(s)
Leucocitos Mononucleares , Linfocitos T , Humanos , Monitorización Inmunológica , Estudios Seroepidemiológicos , Dependovirus , Péptidos/metabolismo
14.
Mol Ther Methods Clin Dev ; 24: 255-267, 2022 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-35211638

RESUMEN

Despite the high safety profile demonstrated in clinical trials, the immunogenicity of adeno-associated virus (AAV)-mediated gene therapy remains a major hurdle. Specifically, T-cell-mediated immune responses to AAV vectors are related to loss of efficacy and potential liver toxicities. As post-translational modifications in T cell epitopes have the potential to affect immune reactions, the cellular immune responses to peptides derived from spontaneously deamidated AAV were investigated. Here, we report that highly deamidated sites in AAV9 contain CD4 T cell epitopes with a Th1 cytokine pattern in multiple human donors with diverse human leukocyte antigen (HLA) backgrounds. Furthermore, some peripheral blood mononuclear cell (PBMC) samples demonstrated differential T cell activation to deamidated or non-deamidated epitopes. Also, in vitro and in silico HLA binding assays showed differential binding to the deamidated or non-deamidated peptides in some HLA alleles. This study provides critical attributes to vector-immune-mediated responses, as AAV deamidation can impact the immunogenicity, safety, and efficacy of AAV-mediated gene therapy in some patients.

15.
Biol Pharm Bull ; 33(7): 1122-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20606300

RESUMEN

Antioxidant property and hematopoietic repair capacity are important characteristics of radioprotective agents. Some studies have demonstrated that 1,2,3,4,6-penta-O-galloyl-beta-D-glucose (PGG), a molecule isolated from the waterlily, has antioxidant, hematopoietic repair, and anti-inflammatory activities. In this study, we try to determine whether PGG extracted from a lily, Nymphaea tetragona var. angusta, has radioprotective effects on splenocytes in vitro against (60)Co gamma-ray irradiation with absorption doses of 2 Gy and 4 Gy. Results show that PGG treatment dramatically enhances the proliferation of splenocytes compared with irradiated but untreated controls. In addition, PGG treatment before irradiation protects the splenocytes from lethal effects of irradiation and decreases DNA damages as identified by the alkaline comet assay. PGG-treated cells also show less radiation-induced apoptosis. These cells have lower concentrations of the pro-apoptotic protein p53 and more of the anti-apoptotic protein Bcl-2. The results presented in this study suggest that PGG has a cytoprotective effect on immune cells exposed to normally damaging amount of radiation. Thus, PGG could be an effective, non-toxic radioprotective agent.


Asunto(s)
Apoptosis/efectos de la radiación , Taninos Hidrolizables/farmacología , Bazo/efectos de los fármacos , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ensayo Cometa , Ratones , Especies Reactivas de Oxígeno/metabolismo , Bazo/citología , Bazo/metabolismo , Bazo/efectos de la radiación
16.
Acta Histochem ; 120(5): 477-489, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29853304

RESUMEN

Oxidative stress plays a crucial role in the progression of alcoholic liver diseases and substances of antioxidant property are of special interest for therapeutic purposes. We investigated the hepatoprotective effect of leaf extracts of Sasa quelpaertensis, an edible bamboo mainly cultivated in Jeju Island, South Korea. We examined the cytotoxicity of different extracts (distilled water, 20-80% EtOH) of S. quelpaertensis on HepG2 cells and their hepatoprotective effect on HepG2 cells stimulated by ethanol (800 mM, 24 h). Furthermore, we measured reactive oxygen species (ROS) production, ethanol toxicity induced cell death, and the activity of antioxidant enzymes. In in vivo experiments, liver damage was induced by oral administration of 5 g/kg ethanol with or without potent ethanol extract of S. quelpaertensis (10 or 100 mg/kg) 12 h interval for a total of 3 doses. Only 80% ethanol extract of S. quelpaertensis (SQEE80) exhibited cytoprotective effect on HepG2 cells against alcohol-induced toxicity. SQEE80 treatment (250, 500 µg/mL) in ethanol exposed HepG2 cells showed significant attenuation of ROS production and ethanol toxicity induced cell death. Furthermore, SQEE80 markedly increased the activity of antioxidant enzyme glutathione peroxidase 1 in ethanol exposed HepG2 cells compared to ethanol stimulated cells. In in vivo experiments, SQEE80 treatment evidently suppressed the alcohol-induced histopathological changes in liver, serum ethanol content, and expression of cytochrome P450 2E1. Furthermore, SQEE80 significantly reversed the reduction of glutathione level in the ethanol challenged liver. Taken together, we suggest the possibility of developing SQEE80 as a natural hepatoprotective substance in attenuating alcohol-induced oxidative stress.


Asunto(s)
Antioxidantes/química , Hepatopatías Alcohólicas/tratamiento farmacológico , Extractos Vegetales/química , Hojas de la Planta/química , Sasa/química , Animales , Antioxidantes/uso terapéutico , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Hep G2/efectos de los fármacos , Humanos , Inmunohistoquímica , Ratones , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/uso terapéutico
17.
Am J Chin Med ; 45(5): 1003-1016, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28659035

RESUMEN

Cells of the hematopoietic system are uniquely radiosensitive due to their rapid proliferation. Consequently, immune suppression readily and undesirably results from irradiation. Our previous studies demonstrated that geraniin isolated from Nymphaea tetragona var. angusta (water lily) had a protective effect on the splenocytes and intestinal tract of irradiated mice. This study was designed to assess the effectiveness of geraniin, an ellagitannin isolated from the water lily, in decreasing gamma ray irradiation-induced destruction of the hematopoietic system in mice. Geraniin treatment improved the survival time of bone marrow cells and maintained bone marrow integrity and also up-regulated the expression of stem cell receptors and the extent of cell mitosis. Geraniin also enhanced the proliferation and differentiation of immune cells that had been suppressed by irradiation. These results suggest geraniin is a promising agent for reconstituting hematopoietic cells after exposure to irradiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Glucósidos/farmacología , Células Madre Hematopoyéticas/citología , Taninos Hidrolizables/farmacología , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Protectores contra Radiación/farmacología , Animales , Células de la Médula Ósea , Células Cultivadas , Rayos gamma/efectos adversos , Glucósidos/administración & dosificación , Glucósidos/aislamiento & purificación , Glucósidos/uso terapéutico , Células Madre Hematopoyéticas/efectos de la radiación , Taninos Hidrolizables/administración & dosificación , Taninos Hidrolizables/aislamiento & purificación , Taninos Hidrolizables/uso terapéutico , Inyecciones Intraperitoneales , Ratones Endogámicos C57BL , Nymphaea/química , Fitoterapia , Traumatismos Experimentales por Radiación/patología , Protectores contra Radiación/uso terapéutico , Bazo/citología
18.
Am J Chin Med ; 44(6): 1099-1110, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27627913

RESUMEN

Bearing pathologic and clinical similarities to human multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE) is used as a murine model to test potential therapeutic agents for MS. Recently, we reported the protective effects of an acidic polysaccharide of Panax ginseng (APG) in C57BL/6 strain-dependent EAE, a model of primary progressive MS. In this study, we extend our previous findings on the therapeutic capacity of APG in relapsing-remitting EAE (rr-EAE), the animal model to closely mimic recurrent inflammatory demyelination lesions of relapsing-remitting MS. Treatments with APG led to a significant reduction of clinical symptoms and the relapse rate of EAE than vehicle treatments. Consistent with this, histological examination revealed that APG markedly modulated the infiltration of CD4[Formula: see text] T cells and CD11b[Formula: see text] macrophages into the spinal cord and the APG-treated CNS was devoid of demyelination and axonal damages. In addition, APG decreased the proliferation of peripheral PLP-reactive T cells and the production of pro-inflammatory factors such as IFN-[Formula: see text], IL-17 and TNF-[Formula: see text]. The fact that APG can induce clinically beneficial effects to distinct types of EAE furthers our understanding on the basis of its immunosuppression in EAE and, possibly, in MS. Our results suggest that APG may serve as a new therapeutic agent for MS as well as other human autoimmune diseases, and warrants continued evaluation for its translation into therapeutic application.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Panax/química , Fitoterapia , Polisacáridos/farmacología , Polisacáridos/uso terapéutico , Animales , Antígeno CD11b , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Enfermedades Desmielinizantes , Modelos Animales de Enfermedad , Femenino , Humanos , Macrófagos/inmunología , Ratones Endogámicos C57BL , Esclerosis Múltiple/tratamiento farmacológico , Polisacáridos/aislamiento & purificación , Recurrencia , Médula Espinal/inmunología , Médula Espinal/patología
19.
Mol Neurodegener ; 11(1): 54, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27450563

RESUMEN

BACKGROUND: The inflammatory myeloid cell activation is one of the hallmarks of experimental autoimmune encephalomyelitis (EAE), yet the in vivo role of the inflammatory myeloid cell activation in EAE has not been clearly resolved. It is well-known that IKK/NF-κB is a key signaling pathway that regulates inflammatory myeloid activation. METHODS: We investigated the in vivo role of inflammatory myeloid cell activation in myelin oligodendrocyte glycoprotein (MOG) peptides-induced EAE using myeloid cell type-specific ikkß gene conditional knockout-mice (LysM-Cre/Ikkß (F/F) ). RESULTS: In our study, LysM-Cre/Ikkß (F/F) mice had alleviated clinical signs of EAE corresponding to the decreased spinal demyelination, microglial activation, and immune cell infiltration in the spinal cord, compared to the wild-type mice (WT, Ikkß (F/F) ). Myeloid ikkß gene deletion significantly reduced the percentage of CD4(+)/IFN-γ(+) (Th1) and CD4(+)/IL-17(+) (Th17) cells but increased the percentages of CD4(+)/CD25(+)/Foxp3(+) (Treg) cells in the spinal cord and lymph nodes, corresponding to the altered mRNA expression of IFN-γ, IL-17, IL-23, and Foxp3 in the spinal cords of LysM-Cre/Ikkß (F/F) EAE mice. Also, the beneficial effect of myeloid IKKß deletion in EAE corresponded to the decreased permeability of the blood brain barrier (BBB). CONCLUSIONS: Our findings strongly suggest that IKK/NF-kB-induced myeloid cell activation exacerbates EAE by activating Th1 and Th17 responses and compromising the BBB. The development of NF-κB inhibitory agents with high efficacy through specific targeting of IKKß in myeloid cells might be of therapeutic potential in MS and other autoimmune disorders.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Quinasa I-kappa B/metabolismo , Inflamación/metabolismo , Células Mieloides/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Citocinas/metabolismo , Activación de Linfocitos , Ratones Transgénicos , Células Mieloides/patología , FN-kappa B/metabolismo , Linfocitos T Reguladores/metabolismo
20.
Food Sci Biotechnol ; 25(6): 1693-1700, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-30263464

RESUMEN

Emodin, one of the major compounds in the herb Reynoutria elliptica, is known to maintain immunosuppressive, anti-allergic, anti-cancer, and anti-inflammatory effects. In this study, we assessed the possibility of using emodin to induce apoptosis in stimulated immune cells in vitro. After treatment with emodin and concanavalin A (Con A), we observed DNA damage-induced apoptosis in splenocytes. Moreover, treatment with emodin and Con A increased the number of apoptotic splenocytes compared with untreated controls. Emodin also diminished the size of CD45R/B220+ cells, CD19+CD69+ cells, and cDC populations. These results indicate that emodin-induced apoptosis was involved in attenuating the immune activity promoted by DNA damage and in decreasing the number of CD45R/B220+ B cells and CD19+CD69+ activating B cells. This demonstration of emodin inducing apoptosis of Con A-stimulated immune cells indicates its potential utility as a therapy for diseases caused by abnormally activated immune cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA