Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Adv Sci (Weinh) ; 11(19): e2308031, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38493498

RESUMEN

Activated microglia in the retina are essential for the development of autoimmune uveitis. Yin-Yang 1 (YY1) is an important transcription factor that participates in multiple inflammatory and immune-mediated diseases. Here, an increased YY1 lactylation in retinal microglia within in the experimental autoimmune uveitis (EAU) group is observed. YY1 lactylation contributed to boosting microglial activation and promoting their proliferation and migration abilities. Inhibition of lactylation suppressed microglial activation and attenuated inflammation in EAU. Mechanistically, cleavage under targets & tagmentation ï¼ˆCUT&Tag) analysis revealed that YY1 lactylation promoted microglial activation by regulating the transcription of a set of inflammatory genes, including STAT3, CCL5, IRF1, IDO1, and SEMA4D. In addition, p300 is identified as the writer of YY1 lactylation. Inhibition of p300 decreased YY1 lactylation and suppressed microglial inflammation in vivo and in vitro. Collectively, the results showed that YY1 lactylation promoted microglial dysfunction in autoimmune uveitis by upregulating inflammatory cytokine secretion and boosting cell migration and proliferation. Therapeutic effects can be achieved by targeting the lactate/p300/YY1 lactylation/inflammatory genes axis.


Asunto(s)
Enfermedades Autoinmunes , Modelos Animales de Enfermedad , Microglía , Uveítis , Factor de Transcripción YY1 , Animales , Microglía/metabolismo , Microglía/inmunología , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo , Ratones , Uveítis/genética , Uveítis/inmunología , Uveítis/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Inflamación/genética , Inflamación/metabolismo , Ratones Endogámicos C57BL , Proliferación Celular/genética
2.
Biomed Pharmacother ; 136: 111291, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33493870

RESUMEN

This study aimed to investigate the dynamic effects of the traditional Chinese medicine compound Longdan Xiegan Decoction (LXD) on the inhibition of Notch signaling pathway activation and T helper (Th) cell differentiation in rats with experimental autoimmune uveitis (EAU). Based on a network pharmacology strategy, we conducted protein interaction network analysis to construct an active ingredient-disease treatment network. Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were further used to screen out the possible signaling pathways regulated by LXD in the treatment of uveitis. In the subsequent functional studies, we established an EAU rat model and investigated the regulatory role of LXD in the Notch signaling pathway and Th cell differentiation in rats with EAU. Female Lewis rats were randomly divided into a normal control (NC) group, an EAU group, and an LXD group. After the induction of EAU, the ocular inflammation and pathological changes in the rats in each group were observed; for documentation, a scanning laser ophthalmoscope (SLO) was used to observe fundus inflammation on day 12 after immunization. Additionally, quantitative polymerase chain reaction (Q-PCR) and enzyme-linked immunosorbent assay (ELISA) were used to detect the expression of Notch1, DLL4, IL-10 and IL-17A in the spleen, lymph nodes and ocular tissues of each group at 0, 6, 9, 12, 15 and 18 days after immunization. In addition, the dynamic frequencies of the CD4+, CD8+, Th17 and Treg cell subsets in the spleen, lymph nodes and ocular tissues were measured by flow cytometry. We found that the Notch signaling pathway was activated and the Th17 frequency was elevated in rats with EAU, leading to disrupted CD4+/CD8+ and Th17/Treg balance. The expression of Notch1, DLL4 and IL-17 mRNA and proteins in the EAU and LXD groups reached a peak on day 12, and then gradually decreased (all P < 0.05), and the ratios of the CD4+/CD8+ and Th17/Treg also peaked on day 12. However, after treatment with LXD, the expression of Notch1, DLL4 and IL-17 mRNA and proteins was significantly decreased (all P < 0.05), and the CD4+/CD8+ and Th17/Treg ratios significantly gradually returns to balance. LXD can efficiently inhibit Th17 cell differentiation, decrease inflammatory cytokine expression, and restore the CD4+/CD8+ and Th17/Treg balance by inhibiting the activation of the Notch signaling pathway in rats with EAU, thus effectively alleviating eye inflammation, protecting eye tissue structures, and positively regulating the immune state of the whole body and the intraocular microenvironment.


Asunto(s)
Antiinflamatorios/farmacología , Enfermedades Autoinmunes/prevención & control , Diferenciación Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Receptor Notch1/metabolismo , Células Th17/efectos de los fármacos , Úvea/efectos de los fármacos , Uveítis/prevención & control , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mapas de Interacción de Proteínas , Ratas Endogámicas Lew , Receptor Notch1/genética , Transducción de Señal , Células Th17/inmunología , Células Th17/metabolismo , Úvea/inmunología , Úvea/metabolismo , Uveítis/genética , Uveítis/inmunología , Uveítis/metabolismo
3.
Front Immunol ; 11: 2071, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33013877

RESUMEN

We specify the clinical features of a spontaneous experimental autoimmune uveitis (EAU) model, in which foreign hen-egg lysozyme (HEL) is expressed in the retina, controlled by the promoter for interphotoreceptor retinol binding protein (IRBP). We previously reported 100% P21 (post-partum day) IRBP:HEL single transgenic (sTg) mice, when crossed to transgenic T cell receptor mice (3A9) generating the double transgenic (dTg) genotype, develop EAU despite profound lymphopenia (thymic HEL-specific T cell deletion). In this work, we characterized the immune component of this model and found conventional dTg CD4+ T cells were less anergic than those from 3A9 controls. Furthermore, prior in vitro HEL-activation of 3A9 anergic T cells (Tan) rendered them uveitogenic upon adoptive transfer (Tx) to sTg mice, while antigen-experienced (AgX, dTg), but not naïve (3A9) T cells halted disease in P21 dTg mice. Flow cytometric analysis of the AgX cells elucidated the underlying pathology: FoxP3+CD25hiCD4+ T regulatory cells (Treg) comprised ∼18%, while FR4+CD73+FoxP3-CD25lo/-CD4+ Tan comprised ∼1.2% of total cells. Further Treg-enrichment (∼80%) of the AgX population indicated FoxP3+CD25hiCD4+ Treg played a key role in EAU-suppression while FoxP3-CD25lo/-CD4+ T cells did not. Here we present the novel concept of dual immunological tolerance where spontaneous EAU is due to escape from anergy with consequent failure of Treg induction and subsequent imbalance in the [Treg:Teffector] cell ratio. The reduced numbers of Tan, normally sustaining Treg to prevent autoimmunity, are the trigger for disease, while immune homeostasis can be restored by supplementation with AgX, but not naïve, antigen-specific Treg.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Inmunoterapia Adoptiva/métodos , Retina/patología , Linfocitos T Reguladores/inmunología , Uveítis/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas del Ojo/inmunología , Factores de Transcripción Forkhead/metabolismo , Humanos , Tolerancia Inmunológica , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Unión al Retinol/inmunología , Linfocitos T Reguladores/trasplante
4.
Front Immunol ; 11: 575669, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117376

RESUMEN

Experimental autoimmune uveitis (EAU) is a CD4+ T cell-mediated organ-specific autoimmune disease and has been considered as a model of human autoimmune uveitis. Dracocephalum heterophyllum (DH) is a Chinese herbal medicine used in treating hepatitis. DH suppressed the production of inflammatory cytokines through the recruitment of myeloid-derived suppressor cells (MDSCs) to the liver. However, it remains elusive whether DH can directly regulate CD4+ T cell biology and hence ameliorates the development of CD4+ T cell-mediated autoimmune disease. In the current study, we found that DH extract significantly suppressed the production of pro-inflammatory cytokines by CD4+ T cells. Further study showed that DH didn't affect the activation, differentiation, and apoptosis of CD4+ T cells. Instead, it significantly suppressed the proliferation of conventional CD4+ T cells both in vitro and in vivo. Mechanistic study showed that DH-treated CD4+ T cells were partially arrested at the G2/M phase of the cell cycle because of the enhanced inhibitory phosphorylation of Cdc2 (Tyr15). In addition, we demonstrated that treatment with DH significantly ameliorated EAU in mice through suppressing the proliferation of autoreactive antigen specific CD4+ T cells. Taken together, the current study indicates that DH-mediated suppression of CD4+ T cell proliferation may provide a promising therapeutic strategy for treating CD4+ T cell-mediated diseases.


Asunto(s)
Antiinflamatorios/farmacología , Enfermedades Autoinmunes/prevención & control , Linfocitos T CD4-Positivos/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Lamiaceae/química , Extractos Vegetales/farmacología , Uveítis/prevención & control , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Autoinmunidad/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proteína Quinasa CDC2/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Fosforilación , Transducción de Señal , Úvea/efectos de los fármacos , Úvea/inmunología , Úvea/metabolismo , Úvea/patología , Uveítis/inmunología , Uveítis/metabolismo , Uveítis/patología
5.
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
6.
Prog Brain Res ; 256(1): 99-124, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32958217

RESUMEN

The chapter is a review enclosed in the volume "Glaucoma: A pancitopatia of the retina and beyond." No cure exists for glaucoma. Knowledge on the molecular and cellular alterations underlying glaucoma neurodegeneration (GL-ND) includes innovative and path-breaking research on neuroinflammation and neuroprotection. A series of events involving immune response (IR), oxidative stress and gene expression are occurring during the glaucoma course. Uveitic glaucoma (UG) is a prevalent acute/chronic complication, in the setting of chronic anterior chamber inflammation. Managing the disease requires a team approach to guarantee better results for eyes and vision. Advances in biomedicine/biotechnology are driving a tremendous revolution in ophthalmology and ophthalmic research. New diagnostic and imaging modalities, constantly refined, enable outstanding criteria for delimiting glaucomatous neurodegeneration. Moreover, biotherapies that may modulate or inhibit the IR must be considered among the first-line for glaucoma neuroprotection. This review offers the readers useful and practical information on the latest updates in this regard.


Asunto(s)
Inteligencia Artificial , Terapia Biológica , Glaucoma , Inflamación , Degeneración Nerviosa , Uveítis , Glaucoma/diagnóstico por imagen , Glaucoma/inmunología , Glaucoma/metabolismo , Glaucoma/terapia , Humanos , Inflamación/diagnóstico por imagen , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/terapia , Degeneración Nerviosa/diagnóstico por imagen , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/terapia , Uveítis/diagnóstico por imagen , Uveítis/inmunología , Uveítis/metabolismo , Uveítis/terapia
7.
Front Immunol ; 11: 1053, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32612602

RESUMEN

Purpose: Previous studies have shown that parental abnormal physiological conditions such as inflammation, stress, and obesity can be transferred to offspring. The purpose of this study was to investigate the impact of parental uveitis on the development and susceptibility to experimental autoimmune uveitis (EAU) in offspring. Methods: Parental male and female B10RIII mice were immunized with interphotoreceptor retinoid binding protein (IRBP) 161-180 in complete Freund's adjuvant and were immediately allowed to mate. Gross examination of the offspring gestated with EAU was performed to determine the influence of parental uveitis on offspring development after birth. Gene expression profiles were analyzed in the affected eyes of offspring under EAU to identify differentially expressed genes (DEGs). Adult offspring were given 5, 25, and 50 µg IRBP161-180 to compare their susceptibility to EAU. Immunized mice were clinically and pathologically evaluated for the development of EAU. Ag-specific T-cell proliferation and IL-17 production from spleens and lymph nodes were evaluated on day 14 or 35 after immunization. Results: Hair loss, delay of eye opening, and swollen spleens in the offspring from parents with uveitis were observed from day 14 to 39 after birth. DEGs were involved in the immune system process, muscle system process, and cell development. The altered antigen processing and presentation, cell adhesion molecules, and phagosome in the eyes of the offspring from uveitis-affected parents were enriched. Offspring gestated with EAU showed a susceptibility to EAU and an earlier onset and higher severity of EAU compared to the control group mice. IRBP-specific lymphocyte proliferation and IL-17 production were observed in the EAU offspring with exposure to parental uveitis. Conclusions: The results suggest that mouse parents with uveitis can increase their offspring's susceptibility to EAU, probably through altering cell adhesion molecules and antigen processing and presentation related to the T-cell proliferation and Th17 response.


Asunto(s)
Enfermedades Autoinmunes/etiología , Uveítis/etiología , Animales , Autoantígenos/inmunología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Proliferación Celular , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Proteínas del Ojo/inmunología , Femenino , Perfilación de la Expresión Génica , Inmunización , Masculino , Herencia Materna/genética , Herencia Materna/inmunología , Intercambio Materno-Fetal/genética , Intercambio Materno-Fetal/inmunología , Ratones , Herencia Paterna/genética , Herencia Paterna/inmunología , Fragmentos de Péptidos/inmunología , Embarazo , Proteínas de Unión al Retinol/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Células Th17/inmunología , Uveítis/genética , Uveítis/inmunología
8.
J Neuroimmunol ; 345: 577286, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32559555

RESUMEN

We investigated the effects of matairesinol (MAT) in the experimental autoimmune uveitis (EAU), a classical animal model of uveitis. We found that treatment with MAT could alleviate intraocular inflammation of EAU. Notably, Th17 cells in eyes of EAU mice could be predominantly restrained by MAT. Furthermore, MAT could inhibit Th17 differentiation in vitro. In addition, MAT inhibited the signaling of MAPK and ROR-γt, a pivotal transcription factor for Th17 cell differentiation in vitro and in vivo. Taken together, these results suggested that MAT had immune-suppressive effects on autoimmune inflammation through Th17 cells.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Proteínas del Ojo/antagonistas & inhibidores , Furanos/uso terapéutico , Lignanos/uso terapéutico , Proteínas de Unión al Retinol/antagonistas & inhibidores , Células Th17/efectos de los fármacos , Uveítis/tratamiento farmacológico , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Proteínas del Ojo/inmunología , Proteínas del Ojo/metabolismo , Femenino , Adyuvante de Freund/toxicidad , Furanos/farmacología , Lignanos/farmacología , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión al Retinol/inmunología , Proteínas de Unión al Retinol/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Uveítis/inmunología , Uveítis/metabolismo
9.
Biomed Pharmacother ; 120: 109381, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31542616

RESUMEN

BACKGROUND: Qinghuo Rougan Formula (QHRGF) is a traditional Chinese medicine (TCM) that has been widely apllied to treat uveitis for several decades. However, the inhibitory mechanism of QHRGF in uveitis has remained to be an enigma. METHODS: The Chinese herbal medicine pharmacology data and analysis platform wereused to search and screen for the effective components of the QHRGF compound injection and to analyse possible therapeutic targets based on network topology. In addition, various known disease target databases were enraolled, the therapeutic target proteins in uveitis were screened, and a protein-protein interaction (PPI) network was constructed. Enrichment analysis was performed on key nodes. Finally, the inhibitory effect of QHRGF on uveitis was verified by experiments. RESULTS: We identified 259 major candidate targets of QHRGF and successfully constructed a 'QHRGF-compound-target-uveitis' network. Above-mentioned targets revealed by Gene enrichment analysis have played an significant role in the cell cycle, autoimmune disease, apoptosis and related signal pathways. We demonstrated that QHRGF attenuates local inflammation in experimental autoimmune uveoretinitis (EAU) rats by regulating natural killer T (NKT) cells and inhibiting MAPK signal pathways. CONCLUSION: QHRGF may regulate the local immune response and inflammatory factors mainly through the MAPK signal pathway. For autoimmune uveitis, QHRGF may be a promising, long-lasting treatment strategy.


Asunto(s)
Antiinflamatorios/farmacología , Bases de Datos de Proteínas , Medicamentos Herbarios Chinos/farmacología , Mapas de Interacción de Proteínas , Biología de Sistemas , Úvea/efectos de los fármacos , Uveítis/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células T Asesinas Naturales/efectos de los fármacos , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Ratas Endogámicas Lew , Transducción de Señal , Úvea/inmunología , Úvea/metabolismo , Úvea/patología , Uveítis/inmunología , Uveítis/metabolismo , Uveítis/patología
10.
Invest Ophthalmol Vis Sci ; 60(1): 420-429, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30695094

RESUMEN

Purpose: We determine the changes in intestinal microbiota and/or disruptions in intestinal homeostasis during uveitis. Methods: Experimental autoimmune uveitis (EAU) was induced in B10.RIII mice with coadministration of interphotoreceptor retinoid-binding protein peptide (IRBP) and killed mycobacterial antigen (MTB) as an adjuvant. Using 16S rRNA gene sequencing, we looked at intestinal microbial differences during the course of uveitis, as well as intestinal morphologic changes, changes in intestinal permeability by FITC-dextran leakage, antimicrobial peptide expression in the gastrointstinal tract, and T lymphocyte prevalence before and at peak intraocular inflammation. Results: We demonstrate that increased intestinal permeability and antimicrobial peptide expression in the intestinal tract coincide in timing with increased effector T cells in the mesenteric lymph nodes, during the early stages of uveitis, before peak inflammation. Morphologic changes in the intestine were most prominent during this phase, but also occurred with adjuvant MTB alone, whereas increased intestinal permeability was found only in IRBP-immunized mice that develop uveitis. We also demonstrate that the intestinal microbiota were altered during the course of uveitis, and that some of these changes are specific to uveitic animals, whereas others are influenced by adjuvant MTB alone. Intestinal permeability peaked at 2 weeks, coincident with an increase in intestinal bacterial strain differences, peak lipocalin production, and peak uveitis. Conclusions: An intestinal dysbiosis accompanies a disruption in intestinal homeostasis in autoimmune uveitis, although adjuvant MTB alone promotes intestinal disruption as well. This may indicate a novel axis for future therapeutic targeting experimentally or clinically.


Asunto(s)
Enfermedades Autoinmunes/microbiología , Disbiosis/microbiología , Microbioma Gastrointestinal/inmunología , Homeostasis/fisiología , Intestinos/fisiología , Uveítis/microbiología , Animales , Antígenos Bacterianos/inmunología , Enfermedades Autoinmunes/inmunología , Ensayo de Inmunoadsorción Enzimática , Proteínas del Ojo , Citometría de Flujo , Lipocalinas/metabolismo , Ratones , Ratones Mutantes , Modelos Animales , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/inmunología , ARN Ribosómico 16S/genética , Proteínas de Unión al Retinol , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/inmunología , Uveítis/inmunología , Proteína de la Zonula Occludens-1/metabolismo
11.
Eur J Pharm Biopharm ; 134: 88-95, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30465821

RESUMEN

Corticosteroids and macrolide immunomodulators such as tacrolimus are effective drugs for the topical treatment of inflammatory eye diseases like allergic conjunctivitis or dry eye. However, tacrolimus is practically insoluble in aqueous solutions and is therefore currently formulated as dispersion. This leads to low bioavailability. Here, we present a novel pharmacologically acceptable, aqueous formulation of tacrolimus based on the "Marinosolv formulation platform". Marinosolv allows the solubilization and thereby improvement of the bioavailability of many otherwise practically insoluble drugs, since dissolved drugs permeate faster into tissues, including ocular tissues. To visualize the benefits of Marinosolv in ophthalmic formulations, we investigated the permeation of a fluorescently labeled estradiol dissolved in Marinosolv compared to a formulation containing the compound as dispersion. Permeation was studied ex-vivo and in-vivo in porcine eyes. Further, we evaluated the improved permeation of topically applied tacrolimus dissolved in Marinosolv compared to a commercially available topically applied tacrolimus dispersion. The Marinosolv formulation was also compared to oral tacrolimus treatment, the standard application route for this drug in case of severe posterior uveitis. Finally, the ocular tissue levels of tacrolimus in all groups were determined using HPLC/MS. We demonstrated that tacrolimus dissolved in Marinosolv reached significantly higher levels in ocular tissues compared to the marketed topical product or after oral application and thus may be a suitable novel option for the treatment of several eye diseases, such as allergic conjunctivitis or uveitis. Thus, Marinosolv may be considered as a new vehicle for tacrolimus eye drops.


Asunto(s)
Composición de Medicamentos/métodos , Inmunosupresores/farmacocinética , Soluciones Oftálmicas/farmacocinética , Tacrolimus/farmacocinética , Uveítis/tratamiento farmacológico , Administración Oftálmica , Administración Oral , Animales , Disponibilidad Biológica , Evaluación Preclínica de Medicamentos , Excipientes/química , Inmunosupresores/administración & dosificación , Inmunosupresores/química , Modelos Animales , Soluciones Oftálmicas/administración & dosificación , Solubilidad , Sus scrofa , Tacrolimus/administración & dosificación , Tacrolimus/química , Uveítis/inmunología , Agua/química
12.
Intern Med J ; 49(11): 1352-1363, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30582273

RESUMEN

The uveitides are a heterogenous group of ocular inflammatory disorders that account for the third highest cause of blindness worldwide, responsible for 5-10% of visual impairment globally. Up to 35% of patients with uveitis can suffer significant vision loss. To prevent irreversible structural damage and blindness, it is important that the diagnosis and commencement of appropriate therapy occurs promptly. Management includes topical and systemic corticosteroid therapy and conventional immunomodulatory agents, including methotrexate, azathioprine, mycophenolate mofetil and cyclosporin. Significant progress has been made in the past decade in our understanding of the immunopathological pathways that drive intraocular inflammation, allowing the development of targeted therapy with biologic agents. These include TNF-α inhibitors, such as infliximab, adalimumab and etanercept; interleukin blockers, such as tocilizumab and daclizumab; and other targeted therapies, such as rituximab and abatacept. The efficacy of these agents has been studied in cases of severe uveitis that are refractory to conventional immunomodulatory agents and provide exciting results that have revolutionised uveitis management. Though the biologic era has provided a large armamentarium to treat uveitis, ongoing challenges and cases of recalcitrant uveitis remain, posing a challenge to internal medicine physicians. This comprehensive review aims to construct an updated summary on the existing evidence pertaining to the use of biologic agents in the treatment of uveitis. Methods include a systematic search for studies between 2000 and 2018 using PubMed, EMBASE, Ovid MEDLINE and Cochrane libraries.


Asunto(s)
Factores Biológicos/uso terapéutico , Terapia Biológica/tendencias , Uveítis/terapia , Corticoesteroides/uso terapéutico , Citocinas/antagonistas & inhibidores , Manejo de la Enfermedad , Humanos , Inmunosupresores/uso terapéutico , Terapia Molecular Dirigida , Uveítis/inmunología
13.
Ophthalmology ; 125(12): 1977-1983, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30458922

RESUMEN

The interleukin (IL)-23/IL-17 axis plays a central role in the pathogenesis of immune-mediated diseases such as psoriasis, psoriatic arthritis, Crohn's disease, and uveitis. Therefore, targeting the IL-23/IL-17 axis has become the focus of multiple clinical trials for drug development in patients with autoimmune diseases. We briefly describe the biology of the IL-23/IL-17 axis and its relevance to the pathogenesis of experimental and clinical uveitis, and review the monoclonal antibody therapies targeting this pathway. Finally, 2 ongoing phase 2 trials of the anti-IL-23 biologic therapy ustekinumab (STELARA, Janssen Biotech Inc, Horsham, PA) in patients with noninfectious uveitis are introduced.


Asunto(s)
Enfermedades Autoinmunes/terapia , Terapia Biológica , Interleucina-23/inmunología , Uveítis/terapia , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Ensayos Clínicos como Asunto , Fármacos Dermatológicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Interleucina-17/inmunología , Células Th17/inmunología , Investigación Biomédica Traslacional , Ustekinumab/uso terapéutico , Uveítis/inmunología
14.
Int J Med Sci ; 15(8): 758-764, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30008584

RESUMEN

PURPOSE: Recent studies have reported the anti-inflammatory effect of Sinomenium acutum. We investigated the anti-inflammatory effect of sinomenine on endotoxin-induced uveitis in a rat model. METHODS: Endotoxin-induced uveitis was induced in rat by lipopolysaccharide (LPS) immunization. Sinomenine (50mg/kg and 100mg/kg) was administered at 30 minutes before, 6 hours and 12 hours after LPS immunization. Clinical and histological severity was evaluated. Protein concentration and levels of tumor necrosis factor (TNF)-α and prostaglandin (PG)-E2 in aqueous humor were measured. Expression of activated Nuclear factor (NF)-κB p65 in ciliary body was also observed. RESULTS: Clinical and histological severities were significantly milder in sinomenine-treated rat than in controls (P < 0.001). Sinomenine suppressed protein leakage and down-regulated the production of TNF-α and PG-E2 in a dose-dependent manner. Sinomenine treatment suppressed the translocation of the NF-κB p65 subunit into the nuclei. CONCLUSION: Systemic administration of sinomenine suppressed the inflammation of ocular tissues. These findings suggest that sinomenine could be a novel therapeutic agent for the control of endogenous ocular inflammatory disease.


Asunto(s)
Antiinflamatorios/farmacología , Extractos Vegetales/farmacología , Sinomenium , Uveítis/tratamiento farmacológico , Animales , Humor Acuoso , Modelos Animales de Enfermedad , Endotoxinas , Lipopolisacáridos , Masculino , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Ratas , Ratas Endogámicas Lew , Factor de Necrosis Tumoral alfa , Uveítis/inmunología
15.
Inflammopharmacology ; 26(5): 1293-1303, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29752572

RESUMEN

In this study, we evaluated the preventive and curative effects of ethanolic extract of Propolis (EEP) during α-Tropomyosin-induced uveitis in an experimental model using Wistar rats, through the regulation of inducible nitric oxide synthase (NOS2) and arginase-1. In this context, rats received daily injection of EEP (100 mg/kg) for 5 days prior to immunization or for 9 days commencing 5 days post immunization with α-Tropomyosin extract, then were sacrificed at day 14. Histological examination, NOS2, arginase-1, and nuclear factor-κB (NF-κB) expression were evaluated in the retinas. Plasmatic production of nitric oxide (NO), urea, IL-4, and TNF-α was assessed. We have found that treatment with EEP substantially reduced the retinal histological damages induced by α-Tropomyosin. In the same context, a significant decrease of NO and TNF-α levels was noticed. Interestingly, EEP down-modulated NOS2 and NF-κB expression in retina. Also, an increase in urea and IL-4 levels was concomitant to an up-modulation of arginase-1 expression. Hence, it appears that EEP attenuated retinal damages through the induction of Th2 response and the inhibition of NF-κB/NOS2 pathway.


Asunto(s)
Apiterapia/métodos , Enfermedades Autoinmunes/terapia , Própolis/farmacología , Uveítis/terapia , Animales , Arginasa/metabolismo , Enfermedades Autoinmunes/inmunología , Modelos Animales de Enfermedad , Etanol/química , Masculino , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Própolis/administración & dosificación , Ratas , Ratas Wistar , Células Th2/inmunología , Tropomiosina/toxicidad , Factor de Necrosis Tumoral alfa/metabolismo , Uveítis/inmunología
16.
Biochem Pharmacol ; 154: 18-27, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29656117

RESUMEN

Autoimmune uveitis is a sight-threatening ocular inflammatory disorder. Immunological inflammation is regarded as the key to pathogenesis in autoimmune uveitis. Baicalin, the major bioactive component of Scutellaria baicalensis, possesses immunomodulatory properties. However, the role of baicalin in uveitis and its underlying mechanisms remain unclear. In the current study, we found that baicalin treatment obviously inhibited the intraocular inflammatory process in mice with experimental autoimmune uveitis, along with clear declines in infiltrated inflammatory cells and inflammatory cytokine transcription in the retina and draining lymph nodes. Furthermore, baicalin treatment increased the frequency and number of regulatory T cells and decreased the frequency and number of effector T cells (Th1 and Th17 cells) in the draining lymph nodes of mice with experimental autoimmune uveitis. In vitro, baicalin treatment suppressed interphotoreceptor retinoid binding protein-specific CD4+ T cell proliferation and converted CD4+ T cell differentiation. Furthermore, the expression of aryl hydrocarbon receptor was activated by baicalin treatment. Baicalin-mediated modulation of CD4+ T cell differentiation was partially abrogated by the suppression of aryl hydrocarbon receptor. These findings suggest that baicalin modulates the Treg/Teff balance and CD4+ T cell proliferation to ameliorate experimental autoimmune uveitis by activating the aryl hydrocarbon receptor.


Asunto(s)
Flavonoides/uso terapéutico , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T Reguladores/metabolismo , Células TH1/metabolismo , Células Th17/metabolismo , Uveítis/tratamiento farmacológico , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Flavonoides/farmacología , Ratones , Ratones Endogámicos C57BL , Receptores de Hidrocarburo de Aril/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Uveítis/inmunología , Uveítis/patología
17.
Int J Immunopathol Pharmacol ; 32: 394632017751547, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29363368

RESUMEN

Total glucosides of paeony (TGP) are active components extracted from the roots of Paeonia lactiflora Pall. In this study, we investigated the role and mechanisms of TGP in experimental autoimmune uveitis (EAU) model of mice. The C57BL/6 mice were randomly divided into three groups: sham group, EAU-control group, and EAU-TGP group. Clinical score of images of the eye fundus were taken on 7, 14, 21, and 28 days after induction of EAU. The concentrations of proinflammatory cytokines in intraocular fluid were measured at 14 days after EAU induction with the use of a multiplex assay system. Flow cytometry was used to analyze the frequency of CD4+, CD8+, interferon-gamma (IFN-γ), and CD4+/CD8+ ratio in spleen and lymph nodes. Western blotting was used to measure expressions of mitogen-activated protein kinase (MAPK) pathway-related proteins in retina. Clinical scores for uveitis were lower in TGP-treated EAU mice than those without TGP treatment. Importantly, the concentrations of cytokines induced by T-helper 1 (Th1) and T-helper 2 (Th2) cells in intraocular fluid were reduced in EAU mice treated with TGP. Furthermore, the frequency of CD4+, IFN-γ, and CD4+/CD8+ ratio was decreased and the frequency of CD8+ was increased in spleen and lymph nodes of mice treated with TGP. The anti-inflammatory effects of TGP were mediated by inhibiting the MAPK signaling pathways. Our results showed that TGP suppressed uveitis in mice via the inhibition of Th1 and Th2 cell function. Thus, TGP may be a promising therapeutic strategy for uveitis, as well as other ocular inflammatory diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Glucósidos/uso terapéutico , Paeonia , Células TH1/inmunología , Células Th2/inmunología , Uveítis/inmunología , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Femenino , Glucósidos/aislamiento & purificación , Glucósidos/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Células TH1/efectos de los fármacos , Células Th2/efectos de los fármacos , Uveítis/tratamiento farmacológico
18.
Neuroimmunomodulation ; 24(2): 87-99, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28848192

RESUMEN

OBJECTIVE: We examined the role of behavioral conditioning of immune responses with cyclosporine A (CsA) on the development of Th1/Th17-driven experimental autoimmune uveoretinitis (EAU). METHODS: Mice received a 0.2% w/v saccharin solution as conditioned stimulus combined with CsA (20 mg/kg) in 6 association trials at 72-h intervals. For evocation periods, conditioned mice were reexposed to saccharin, whereas the conditioned but not reexposed group received water only. Animals were immunized with human interphotoreceptor-retinoid-binding protein peptide 161-180 (hIRBPp161-180) peptide in complete Freund adjuvant (CFA) and a concomitant injection of pertussis toxin. RESULTS: In naïve mice subjected to the behavioral conditioning regimen, mitogen-induced interleukin (IL)-2 production was decreased in conditioned mice compared to conditioned but not reexposed animals. Incidence and severity of EAU were not significantly lower in behaviorally conditioned and immunized mice. ELISA analysis of splenocytes revealed a reduced interferon (IFN)-γ/IL-17 ratio in CsA-treated, conditioned but not reexposed, and conditioned animals. The adoptive transfer of antigen-specific splenocytes from animals behaviorally conditioned with CsA to naïve mice decreased the severity of EAU in recipient mice compared to the control group. In vitro activation of splenocytes isolated from immunized mice with agonists targeting TLR2 and NOD2 together with ß2-adrenergic activation (induced by epinephrine, norepinephrine, or salbutamol) resulted in decreased IFN-γ but increased IL-17 immune responses. The ß2-adrenergic antagonist propranolol could restore IFN-γ production, whereas only the norepinephrine-induced increase in IL-17 production was abrogated. CONCLUSIONS: We conclude that CsA conditioning in the EAU model mitigates Th1 but enhances Th17 immune responses, and does not ameliorate disease. The results imply that in EAU the mechanism of immune conditioning interacts with CFA components during active immunization, most likely via the TLR2/NOD2 pathway, and induces differentiation of Th17 cells that drive autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Ciclosporina/farmacología , Inmunosupresores/farmacología , Neuroinmunomodulación/fisiología , Uveítis/inmunología , Animales , Condicionamiento Clásico/fisiología , Modelos Animales de Enfermedad , Masculino , Ratones , Células Th17/inmunología
19.
Clin Exp Immunol ; 190(3): 293-303, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28763100

RESUMEN

Uveitis (intraocular inflammation) is a leading cause of loss of vision. Although its aetiology is largely speculative, it is thought to arise from complex genetic-environmental interactions that break immune tolerance to generate eye-specific autoreactive T cells. Experimental autoimmune uveitis (EAU), induced by immunization with the ocular antigen, interphotoreceptor retinoid binding protein (IRBP), in combination with mycobacteria-containing complete Freund's adjuvant (CFA), has many clinical and histopathological features of human posterior uveitis. Studies in EAU have focused on defining pathogenic CD4+ T cell effector responses, such as those of T helper type 17 (Th17) cells, but the innate receptor pathways precipitating development of autoreactive, eye-specific T cells remain poorly defined. In this study, we found that fungal-derived antigens possess autoimmune uveitis-promoting function akin to CFA in conventional EAU. The capacity of commensal fungi such as Candida albicans or Saccharomyces cerevisae to promote IRBP-triggered EAU was mediated by Card9. Because Card9 is an essential signalling molecule of a subgroup of C-type lectin receptors (CLRs) important in host defence, we evaluated further the proximal Card9-activating CLRs. Using single receptor-deficient mice we identified Dectin-2, but not Mincle or Dectin-1, as a predominant mediator of fungal-promoted uveitis. Conversely, Dectin-2 activation by α-mannan reproduced the uveitic phenotype of EAU sufficiently, in a process mediated by the Card9-coupled signalling axis and interleukin (IL)-17 production. Taken together, this report relates the potential of the Dectin-2/Card9-coupled pathway in ocular autoimmunity. Not only does it contribute to understanding of how innate immune receptors orchestrate T cell-mediated autoimmunity, it also reveals a previously unappreciated ability of fungal-derived signals to promote autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Proteínas Adaptadoras de Señalización CARD/inmunología , Candida albicans/inmunología , Candidiasis/inmunología , Lectinas Tipo C/inmunología , Saccharomyces cerevisiae/inmunología , Uveítis/inmunología , Animales , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/patología , Proteínas Adaptadoras de Señalización CARD/genética , Candidiasis/inducido químicamente , Candidiasis/patología , Proteínas del Ojo/toxicidad , Lectinas Tipo C/genética , Ratones , Ratones Mutantes , Proteínas de Unión al Retinol/toxicidad , Células Th17/inmunología , Células Th17/patología , Uveítis/inducido químicamente , Uveítis/genética , Uveítis/patología
20.
Ocul Immunol Inflamm ; 25(2): 169-178, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27749140

RESUMEN

The treatment of articular and extra-articular manifestations associated with HLA-B27 has undergone dramatic changes over the past two decades, mainly as a consequence of the introduction of biologic agents and in particular anti-tumor necrosis factor α (anti-TNFα) agents. Uveitis is known to be the most frequent extra-articular feature in HLA-B27-associated spondyloarthritides. Topical corticosteroids and cycloplegic agents remain the cornerstones of treatment. However, biologic therapy may be effective in the management of refractory or recurrent forms of uveitis. This review gives an update on the management of HLA-B27-associated ocular disorders with biologics, including anti-TNFα agents and non-anti-TNFα biologic modifier drugs. There is an emerging role for newer biologics targeting interleukin-12/23 and interleukin-17 for the treatment of spondyloarthritides but data on their efficacy on anterior uveitis are sparse.


Asunto(s)
Terapia Biológica , Antígeno HLA-B27/inmunología , Espondiloartropatías/terapia , Uveítis/terapia , Glucocorticoides/uso terapéutico , Humanos , Midriáticos/uso terapéutico , Espondiloartropatías/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Uveítis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA