Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Intervalo de año de publicación
1.
Chem Res Toxicol ; 32(2): 294-303, 2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30638013

RESUMEN

Retigabine (RTG) is an antiepileptic drug approved as an adjunctive treatment for refractory partial-onset seizures in adults. In April 2013, the Food and Drug Administration issued a warning that RTG could cause changes in retinal pigmentation and discoloration of skin, resulting in a blue appearance. As part of a larger preclinical effort to gain a mechanistic understanding as to the origins of retinal pigment changes associated with RTG, we conducted a long-term repeat dosing study in rats. Matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) was used to determine the distribution of RTG and its metabolites in the rat eye following 13 and 39 weeks of dosing. IMS revealed the presence of RTG, a previously characterized N-acetyl metabolite of RTG (NAMR), and several species structurally related through the dimerization of RTG and NAMR. These species were highly localized to the melanin-containing layers of the uveal tract of the rat eye including the choroid, ciliary body, and iris, suggesting that the formation of these dimers occurs from melanin bound RTG and NAMR. Furthermore, several of the RTG-related dimers have UV absorbance which give them a purple color in solution. We propose that the melanin binding of RTG and NAMR effectively concentrates the two compounds to enable mixed condensation reactions to occur when the binding provides the proper geometry in the redox environment of the uveal tissues. High lateral resolution images illustrate that the blood-retinal barrier effectively restricts retinal access to RTG-related compounds. The spatial information provided by MALDI IMS was critical in contextualizing the homogenate concentrations of key RTG-related compounds and helped provide a basis for the mechanism of dimer formation.


Asunto(s)
Carbamatos/metabolismo , Fenilendiaminas/metabolismo , Pigmentos Retinianos/análisis , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Úvea/efectos de los fármacos , Animales , Carbamatos/farmacología , Dimerización , Masculino , Melaninas/química , Melaninas/metabolismo , Fenilendiaminas/farmacología , Ratas , Ratas Long-Evans , Úvea/metabolismo , Úvea/patología
2.
Cutan Ocul Toxicol ; 38(4): 360-369, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31213109

RESUMEN

Purpose: To compare the efficacy of systemic and intravitreal infliximab treatments in an experimental endotoxin-induced uveitis (EIU) model. Methods: Twenty-eight white New Zealand rabbits were equally divided into 4 groups. Group 1 received an intravitreal injection of 0.1 cc saline, group 2 received an intravitreal injection of 2 µg/0.1 cc lipopolysaccharide (LPS), group 3 received an intravitreal injection of 2 µg/0.1 cc LPS and 2 mg/0.1 cc infliximab, and group 4 received intravitreal injection of 2 µg/0.1 cc LPS and intravenous injection of 5 mg/kg infliximab. Clinical, biochemical (aqueous and vitreous humour protein levels and TNF-α concentrations), and histopathological evaluations were performed. Results: The clinical examination score was lower in group 4 than in group 2 (p = 0.006); but there was no significant difference between groups 2 and 3 (Bonferroni correction, p = 0.016). No statistically significant difference was found among groups 2, 3, and 4 for aqueous humour protein levels (p > 0.05). Significantly higher aqueous humour concentrations of TNF-α was measured in group 3 comparing to both group 1 and 4 (p = 0.003 and p = 0.002, respectively). No significant difference was found in vitreous protein levels or TNF-α concentrations among all study groups (Bonferroni correction, p = 0.026 and p = 0.101, respectively). Histopathological evaluation of the uveal tissue and anterior chamber reaction revealed the highest inflammation in group 3 (p < 0.001). In group 4, histopathological evaluation of uveal tissue was lower than in groups 2 and 3 (p < 0.001 and p = 0.001, respectively); whereas there was no difference in anterior chamber inflammation between groups 2 and 4 (p = 1.00). Conclusion: Intravitreal 2 mg/0.1 cc infliximab injection exacerbated inflammation in an EIU model; whereas systemic infliximab treatment at a dose of 5 mg/kg suppressed inflammation effectively and rapidly.


Asunto(s)
Antiinflamatorios/administración & dosificación , Infliximab/administración & dosificación , Uveítis/tratamiento farmacológico , Animales , Cámara Anterior/efectos de los fármacos , Cámara Anterior/inmunología , Antiinflamatorios/efectos adversos , Proteínas del Ojo/metabolismo , Infliximab/efectos adversos , Inyecciones Intravenosas , Inyecciones Intravítreas , Lipopolisacáridos , Conejos , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Úvea/efectos de los fármacos , Úvea/patología , Uveítis/inducido químicamente , Uveítis/inmunología , Uveítis/patología , Cuerpo Vítreo/inmunología
3.
Artículo en Inglés | MEDLINE | ID: mdl-30232034

RESUMEN

Uveitis is inflammation of the uvea which consists of the iris, ciliary body and the choroid of the eye. Uveitis can lead to impaired vision and is responsible for 10% of all cases of blindness globally. Using an endotoxin-induced uveitis (EIU) rodent model, our previous data implicated the endogenous cannabinoid system (ECS) in the amelioration of many of the components of the inflammatory response. Here, we test the hypothesis that the reduction in inflammatory mediators in the EIU model by the CB2 agonist, HU308, is associated with changes in ECS endogenous ligands as well as related lipids, prostaglandins (PGs), 2-acyl glycerols, and lipoamines. Analysis of leukocytes and neutrophils, CB2 mRNA, and 26 lipids in the eye of WT mice after EIU induction and HU308 treatment were compared to the same analyses in the CB2 knock-out (CB2 KO) mouse. Endothelial leukocyte adhesion and neutrophil migration were significantly increased in both WT and CB2 KO after EIU. HU308 significantly reduced the leukocyte adhesion and neutrophil recruitment in the WT animals. HU308 also significantly reduced leukocyte adhesion in the CB2 KO mouse, yet, had no effect on neutrophil infiltration suggesting an important off-target effect of HU308. Lipidomics profiles revealed significant increases in 6 non-ECS lipids after EIU in the WT and that HU308 effectively reduced these back to control levels; in addition, HU308 increased levels of 2-acyl glycerols and decreased all N-acyl glycines. CB2 KOs with saline-injection compared to WT had significantly elevated levels of 2-acyl glycerols, whereas levels of N-oleoyl ethanolamine (OEA), N-stearoyl ethanolamine (SEA), and PGE2 were reduced. CB2 KOs with EIU had 13 lipids that were significantly lower than WT with EIU including 4 N-acyl glycines. HU308 had no effect on lipid concentrations in the CB2 KOs with EIU, however, it did cause further reductions on 3 additional lipids compared to saline controls. HU308 appears to be acting at a non-CB2 target for the reduction of leukocyte infiltration in the EIU model; however, our data suggest that HU308 is working through CB2 to reduce neutrophil migration and for the regulation of multiple lipid signaling pathways including PGs, lipoamines, and 2-acyl glycerols. These data implicate ocular CB2 as a key component of lipid signaling in the eye and part of the regulatory processes of inflammation.


Asunto(s)
Cannabinoides/administración & dosificación , Ojo/efectos de los fármacos , Inflamación/tratamiento farmacológico , Receptor Cannabinoide CB2/genética , Uveítis/tratamiento farmacológico , Animales , Endocannabinoides/genética , Endocannabinoides/metabolismo , Endotoxinas/toxicidad , Ojo/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Leucocitos/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Prostaglandinas/genética , ARN Mensajero/genética , Receptor Cannabinoide CB2/agonistas , Transducción de Señal/efectos de los fármacos , Úvea/efectos de los fármacos , Úvea/patología , Uveítis/inducido químicamente , Uveítis/metabolismo , Uveítis/patología
4.
Neurochem Res ; 41(5): 1020-8, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26700431

RESUMEN

In the present study, we investigated the effect of three different sources of hydrogen sulfide (H2S) on sympathetic neurotransmission from isolated superfused bovine iris-ciliary bodies. The three agents under consideration were: ACS67, a hybrid of latanoprost and a H2S-donating moiety; L-cysteine, a substrate for endogenous production of H2S and GYY 4137, a slow donor of H2S. We also examined the contribution of prostaglandins to the pharmacological actions of the H2S donors on release of [(3)H]-norepinephrine ([(3)H]NE) triggered by electrical field stimulation. ACS67, L-cysteine and GYY 4137 caused a concentration-dependent inhibition of electrically-evoked [(3)H]NE release from isolated bovine iris-ciliary bodies without affecting basal [(3)H]NE efflux. The cyclooxygenase inhibitor, flurbiprofen enhanced the inhibitory action of ACS67 and L-cysteine on stimulated [(3)H]NE release. Both aminooxyacetic acid, an inhibitor of cystathionine-ß-synthase and glibenclamide, a KATP channel blocker reversed the inhibition of evoked NE release induced by the H2S donors. We conclude that H2S donors can inhibit sympathetic neurotransmission from isolated bovine iris-ciliary bodies, an effect partially dependent on the in situ production of H2S and prostanoids, and is mediated by an action on KATP channels.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos , Úvea/efectos de los fármacos , Animales , Bovinos , Cuerpo Ciliar/efectos de los fármacos , Cuerpo Ciliar/metabolismo , Cisteína/farmacología , Estimulación Eléctrica , Técnicas In Vitro , Morfolinas/farmacología , Norepinefrina/metabolismo , Compuestos Organotiofosforados/farmacología , Prostaglandinas/metabolismo , Prostaglandinas F Sintéticas/farmacología , Sistema Nervioso Simpático/fisiología , Transmisión Sináptica , Ácido Tióctico/análogos & derivados , Ácido Tióctico/farmacología , Úvea/fisiología
5.
Clin Immunol ; 157(2): 198-204, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25701800

RESUMEN

Uveitis is a sight-threatening inflammatory disease of the eye which represents the third leading cause of blindness in the developed countries. The conventional pharmacological treatment includes corticosteroids and immunosuppressive agents, which are limited by their side effects. New therapeutic strategies are thus strongly needed. Exogenously-administered carbon monoxide (CO) may represent an effective treatment for conditions characterized by a dysregulated inflammatory response. Carbon monoxide-releasing molecules (CORMs) are a novel group of compounds capable of carrying and liberating controlled quantities of CO. Among CORMs, CORM-A1 represents the first example of water soluble CO releaser. We show here that CORM-A1 under a late prophylactic regime is able to significantly ameliorate the natural course of experimental autoimmune uveoretinitis, a rodent model of immunoinflammatory posterior uveitis. The present study strongly supports the development of CORM-A1 as a potential new drug for treatment of patients with non-infectious posterior uveitis.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Boranos/farmacología , Carbonatos/farmacología , ARN Mensajero/efectos de los fármacos , Retina/efectos de los fármacos , Retinitis/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Úvea/efectos de los fármacos , Uveítis/inmunología , Animales , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/patología , Citocinas/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas Lew , Retina/inmunología , Retina/patología , Retinitis/inducido químicamente , Retinitis/patología , Proteínas de Unión al Retinol/toxicidad , Bazo/efectos de los fármacos , Bazo/metabolismo , Linfocitos T Reguladores/inmunología , Úvea/inmunología , Úvea/patología , Uveítis/inducido químicamente , Uveítis/patología
6.
Mol Vis ; 21: 403-11, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25883526

RESUMEN

PURPOSE: Our previous study demonstrated that an intraperitoneal injection of Diminazene Aceturate (DIZE) attenuated uveitis by activating ocular angiotensin-converting enzyme 2 (ACE2). Here, we investigated the anti-inflammatory effects on the ocular anterior segment of a topical administration of a DIZE solution and explored the downstream target molecules involved in the anti-inflammatory mechanism after ACE2 activation. METHODS: Endotoxin-induced uveitis (EIU) in rats was induced by a subcutaneous injection of lipopolysaccharides (LPS, 200 µg) in 0.1 ml of sterile saline. DIZE (0.025, 0.05, or 0.1%) and dexamethasone (0.1%) solutions were applied topically (10 µl eyedrops) to both eyes 6X every two hours before and after LPS injection. The inflammation of the ocular anterior segment was observed and the clinical scores were evaluated 24 h after LPS injection. The total protein concentration and levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in the aqueous humor were determined. CD11b-positive cells adjacent to the iris ciliary body (ICB) were stained by immunohistochemistry. The mRNA levels of inflammatory cytokines and mediators, including IL-1ß, TNF-α, COX-2, and iNOS or NF-κB subunit p65 in the ICB, were analyzed by real time RT-PCR. The protein expression of NF-κB p65 and the phosphorylated protein of p38 MAPK were detected by western blotting. RESULTS: A topical administration of DIZE decreased clinical scores and the total protein concentration, as well as TNF-α and IL-6 levels in the aqueous humor. Meanwhile, the mRNA levels of inflammatory cytokines and mediators, including IL-1ß, TNF-α, COX-2, and iNOS in the ICB, were downregulated. DIZE reduced the recruitment of CD11b-positive cells adjacent to the ICB. Furthermore, DIZE downregulated the expressions of NF-κB subunit p65 at protein and mRNA levels and inhibited the phosphorylation of p38 MAPK protein in the ICB. CONCLUSIONS: A topical administration of DIZE suppressed ocular inflammation in EIU and decreased the levels of inflammatory cytokines. DIZE attenuated the activation of NF-κB and p38 MAPK in EIU, which may be associated with ACE2-mediated anti-inflammatory effects. Our data provided further evidence that DIZE may represent a novel class of drug for the management of ocular inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Dexametasona/farmacología , Diminazeno/análogos & derivados , Úvea/efectos de los fármacos , Uveítis/tratamiento farmacológico , Administración Oftálmica , Enzima Convertidora de Angiotensina 2 , Animales , Humor Acuoso/química , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Diminazeno/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Inyecciones Intraperitoneales , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolisacáridos , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Soluciones Oftálmicas , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Úvea/metabolismo , Úvea/patología , Uveítis/inducido químicamente , Uveítis/genética , Uveítis/patología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Front Immunol ; 12: 623725, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995347

RESUMEN

The profound impact that vision loss has on human activities and quality of life necessitates understanding the etiology of potentially blinding diseases and their clinical management. The unique anatomic features of the eye and its sequestration from peripheral immune system also provides a framework for studying other diseases in immune privileged sites and validating basic immunological principles. Thus, early studies of intraocular inflammatory diseases (uveitis) were at the forefront of research on organ transplantation. These studies laid the groundwork for foundational discoveries on how immune system distinguishes self from non-self and established current concepts of acquired immune tolerance and autoimmunity. Our charge in this review is to examine how advances in molecular cell biology and immunology over the past 3 decades have contributed to the understanding of mechanisms that underlie immunopathogenesis of uveitis. Particular emphasis is on how advances in biotechnology have been leveraged in developing biologics and cell-based immunotherapies for uveitis and other neuroinflammatory diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Autoinmunidad/efectos de los fármacos , Productos Biológicos/uso terapéutico , Tolerancia Inmunológica/efectos de los fármacos , Inmunoterapia , Úvea/efectos de los fármacos , Uveítis/terapia , Animales , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Terapia Molecular Dirigida , Transducción de Señal , Úvea/inmunología , Úvea/metabolismo , Uveítis/inmunología , Uveítis/metabolismo
9.
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
10.
Front Immunol ; 11: 608134, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33469457

RESUMEN

There has been steady progress in understanding the pathogenesis, clinical features, and effective treatment of acute anterior uveitis (AU) over the past 5 years. Large gene wide association studies have confirmed that AU is a polygenic disease, with overlaps with the seronegative arthropathies and inflammatory bowel diseases, associations that have been repeatedly confirmed in clinical studies. The role of the microbiome in AU has received increased research attention, with recent evidence indicating that human leukocyte antigen B27 (HLA B27) may influence the composition of the gut microbiome in experimental animals. Extensive clinical investigations have confirmed the typical features of acute AU (AAU) and its response to topical, regional and systemic immunosuppressive treatment. Increased understanding of the role of cytokines has resulted in studies confirming the value of anti-cytokine therapy [anti-tumor necrosis factor (anti-TNF) and interleukin 6 (IL-6) therapy] in severe and recurrent cases of AAU, particularly in subjects with an associated spondyloarthopathy (SpA) and in juvenile idiopathic arthritis (JIA)-associated AAU.


Asunto(s)
Antígeno HLA-B27/inmunología , Úvea/inmunología , Uveítis Anterior/inmunología , Enfermedad Aguda , Animales , Bacterias/inmunología , Bacterias/metabolismo , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Microbioma Gastrointestinal , Predisposición Genética a la Enfermedad , Antígeno HLA-B27/genética , Haplotipos , Humanos , Inmunosupresores/uso terapéutico , Intestinos/microbiología , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Úvea/efectos de los fármacos , Úvea/metabolismo , Uveítis Anterior/tratamiento farmacológico , Uveítis Anterior/genética , Uveítis Anterior/microbiología
11.
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
12.
Exp Eye Res ; 88(4): 760-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19150349

RESUMEN

The uveoscleral outflow route was described more than 40 years ago. Part of aqueous leaves the eye through the iris root. The ciliary muscle, and there are large species differences in the fraction of aqueous outflow that leaves the eye through this route. In non-human primates 40-50% of aqueous leaves the eye by the uveoscleral route. In human eyes most data has been collected by indirect calculations, with results suggesting a similar fraction, at least in eyes from younger individuals. An age-dependent reduction in uveoscleral flow in human eyes may explain the initial difference seen between non-human primate and human eyes. Unlike trabecular outflow, intraocular pressures within the normal range have little effect on uveoscleral outflow. This may be explained by the fact that changes in intraocular pressure have little effect on the pressure gradient for flow through the ciliary muscle, which is likely to be the rate-limiting step in uveoscleral outflow. The state of the ciliary muscle is important and contraction reduces while relaxation increases uveoscleral flow. Similar effects are achieved with cholinergic agonists and antagonists. Epinephrine increases uveoscleral flow, most likely through stimulating beta(2)-adrenergic receptors. Prostaglandin F(2alpha) and prostaglandin F(2alpha)-analogues effectively reduce intraocular pressure by increasing uveoscleral flow. This is mediated by structural changes in the extracellular matrix of the ciliary muscle, and is likely to contribute to a valuable excess route for aqueous and proteins during intraocular inflammation. Whether uveoscleral flow plays a significant role in any other eye disease is not clear. Thus, 40 years later we are able to successfully increase aqueous flow through the uveoscleral route, a valuable contribution to glaucoma treatment, but we still have only a limited understanding on its physiological role.


Asunto(s)
Humor Acuoso/fisiología , Esclerótica/fisiología , Úvea/fisiología , Adrenérgicos/farmacología , Adulto , Anciano de 80 o más Años , Envejecimiento/fisiología , Animales , Colinérgicos/farmacología , Humanos , Macaca fascicularis , Prostaglandinas/farmacología , Esclerótica/efectos de los fármacos , Especificidad de la Especie , Úvea/efectos de los fármacos
13.
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
14.
Anticancer Agents Med Chem ; 17(2): 190-205, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27109020

RESUMEN

Epigenetic modifications can affect numerous mechanisms used by neoplastic cells to evade immune control. In melanoma epigenetic defects, caused by dysregulations in the expression of genome writers, erasers, or readers, play a significant role in the reduced expression of molecules required for efficient immune recognition as well as antigen presentation and processing. Alterations in gene expression were identified in tumor-associated antigens (TAAs), human leukocyte antigen (HLA) complex, co-stimulatory/accessory molecules, antigen processing machinery (APM), and NKG2D ligands that have shown to be silenced or down-regulated in melanoma. In agreement with the inherent reversibility of epigenetic silencing, epigenetic drugs such as inhibitors of DNA methyltransferases (DNMTs), histone deacetylases (HDACs), histone methyltransferase enhancer of Zeste homolog 2 (EZH2), and modifiers of microRNA (miRNA) dysregulation or antagomirs can restore the expression of these molecules, favouring the recognition of cancer cells by immune responses, reducing the resistance to Natural Killer (NK) and cytotoxic T cells (CTL), and enhancing the functions of antigen presenting cells. Moreover, inhibitors of reader proteins seem to preferentially affect the NF-kB-induced activation of pro-inflammatory cytokine genes. At present an increasing interest is shown toward new combined therapeutic approaches employing epidrugs or new molecular inhibitors and in vivo immunotherapies, such as vaccines and adoptive T-cell transfer (ACT). This review summarizes the current understanding of the role of epidrugs in the modulation of molecules involved in the melanoma immune response and focuses on their future clinical use in new therapeutic combinations for melanoma treatment.


Asunto(s)
Antineoplásicos/farmacología , Epigénesis Genética/efectos de los fármacos , Inmunoterapia/métodos , Melanoma/genética , Melanoma/terapia , Úvea/efectos de los fármacos , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/terapia , Animales , Antineoplásicos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunidad/efectos de los fármacos , Melanoma/inmunología , Melanoma/patología , Úvea/inmunología , Úvea/metabolismo , Úvea/patología , Neoplasias de la Úvea/inmunología , Neoplasias de la Úvea/patología
15.
Sci Rep ; 7: 40462, 2017 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-28084469

RESUMEN

Biocompatibility of intraocular lens (IOL) is critical to vision reconstruction after cataract surgery. Foldable hydrophobic acrylic IOL is vulnerable to the adhesion of extracellular matrix proteins and cells, leading to increased incidence of postoperative inflammation and capsule opacification. To increase IOL biocompatibility, we synthesized a hydrophilic copolymer P(MPC-MAA) and grafted the copolymer onto the surface of IOL through air plasma treatment. X-ray photoelectron spectroscopy, atomic force microscopy and static water contact angle were used to characterize chemical changes, topography and hydrophilicity of the IOL surface, respectively. Quartz crystal microbalance with dissipation (QCM-D) showed that P(MPC-MAA) modified IOLs were resistant to protein adsorption. Moreover, P(MPC-MAA) modification inhibited adhesion and proliferation of lens epithelial cells (LECs) in vitro. To analyze uveal and capsular biocompatibility in vivo, we implanted the P(MPC-MAA) modified IOLs into rabbits after phacoemulsification. P(MPC-MAA) modification significantly reduced postoperative inflammation and anterior capsule opacification (ACO), and did not affect posterior capsule opacification (PCO). Collectively, our study suggests that surface modification by P(MPC-MAA) can significantly improve uveal and capsular biocompatibility of hydrophobic acrylic IOL, which could potentially benefit patients with blood-aqueous barrier damage.


Asunto(s)
Cápsula Anterior del Cristalino/fisiología , Materiales Biocompatibles/farmacología , Interacciones Hidrofóbicas e Hidrofílicas , Lentes Intraoculares , Ensayo de Materiales , Metacrilatos/farmacología , Fosforilcolina/análogos & derivados , Úvea/fisiología , Adsorción , Animales , Cápsula Anterior del Cristalino/efectos de los fármacos , Catarata/patología , Extracción de Catarata/efectos adversos , Adhesión Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Humanos , Inflamación/etiología , Microscopía de Fuerza Atómica , Fosforilcolina/farmacología , Cápsula Posterior del Cristalino/efectos de los fármacos , Complicaciones Posoperatorias/etiología , Conejos , Propiedades de Superficie , Úvea/efectos de los fármacos
16.
Cytometry B Clin Cytom ; 92(4): 286-290, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-26860883

RESUMEN

BACKGROUND: The uveitis masquerade syndromes (UMS) are a group of ocular diseases that may mimic chronic intraocular inflammation. Many malignant entities such as non-Hodgkin's lymphomas may masquerade as uveitis. We report a case of an HIV-positive patient with masquerade syndrome presenting unilateral uveitis. CASE REPORT: 45-year-old Caucasian man with a diagnosis of diffuse large B-cell lymphoma (DLBCL). The patient was diagnosed by a biopsy of an abdominal mass which showed fragments of gastric mucosa with diffuse growth of neoplastic cells. At diagnosis, the patient suffered from unilateral blurring of vision and a sudden decrease of left-eye visual acuity. A slit-lamp examination of the left eye revealed a diagnosis of anterior uveitis. The patient exhibited no signs of posterior uveitis. An anterior-chamber paracentesis was performed and analyzed by multiparameter flow cytometry (MFC), showing cells CD45, CD19, CD20, CD22, and CD38 positives, and moderate expression of CD10 with kappa light chain restriction, showing a monoclonal B-cell population. The patient received CHOP-R with intrathecal methotrexate followed by consolidation high dose methotrexate obtaining a complete response which is ongoing. CONCLUSION: Differential diagnosis between chronic uveitis and ocular lymphoma may be challenging. We advocate anterior-chamber paracentesis in cases of refractory uveitis in patients with hematologic malignancies. © 2016 International Clinical Cytometry Society.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Doxorrubicina/análogos & derivados , Linfoma de Células B Grandes Difuso/diagnóstico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Metotrexato/uso terapéutico , Uveítis/diagnóstico , Antígenos CD/genética , Antígenos CD/inmunología , Humor Acuoso/efectos de los fármacos , Humor Acuoso/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/patología , Biomarcadores/metabolismo , Ciclofosfamida/uso terapéutico , Diagnóstico Diferencial , Doxorrubicina/uso terapéutico , Citometría de Flujo/métodos , Expresión Génica , Humanos , Inflamación , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/patología , Masculino , Persona de Mediana Edad , Paracentesis , Prednisona/uso terapéutico , Resultado del Tratamiento , Úvea/efectos de los fármacos , Úvea/inmunología , Úvea/patología , Uveítis/tratamiento farmacológico , Uveítis/inmunología , Uveítis/patología , Vincristina/uso terapéutico
17.
Int J Mol Med ; 40(4): 1277-1284, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28849034

RESUMEN

The mechanisms underlying cutaneous melanogenesis have been widely studied; however, very little is known about uveal melanogenesis. Melanin is normally produced by uveal melanocytes and gives the color to the iris. A derangement from this normal production may occur, for instance, by iatrogenic events, such as glaucoma therapy with prostaglandins that may enhance cutaneous and iris pigmentation. In this study, we investigated the mechanisms that regulate uveal melanogenesis in human uveal melanoma cells (92.1) and murine cutaneous melanoma cells (B16-F1). In the first part of the study, we compared the effects of known cutaneous pigmenting agents on the B16-F1 and 92.1 cells, showing an opposite response of the two cell lines. Subsequently, using argan oil, a known depigmenting agent for murine cutaneous melanoma cells, on 92.1 cells, we found that in these cells, it also functioned as an inhibitor of melanogenesis and tyrosinase expression. From a molecular perspective, treatment of the 92.1 cells with argan oil decreased melanogenesis-associated transcription factor (MITF) gene expression by inducing MITF phosphorylation at Ser73, thus leading to MITF ubiquitination and disposal. It also led to the downregulation of the extracellular signal-regulated kinase (ERK)1/2 and Akt pathways, also known to be involved in cutaneous melanogenesis, although with an opposing function. Taken together, our data indicate that: ⅰ) some differences exist in the regulation of melanogenesis between cutaneous and uveal melanoma cells; and ⅱ) argan oil exerts a depigmenting effect on 92.1 cells through its action on the ERK1/2 and Akt pathways.


Asunto(s)
Melaninas/antagonistas & inhibidores , Melanocitos/efectos de los fármacos , Monofenol Monooxigenasa/antagonistas & inhibidores , Aceites de Plantas/farmacología , Úvea/efectos de los fármacos , Animales , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Melaninas/biosíntesis , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Ratones , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Monofenol Monooxigenasa/genética , Monofenol Monooxigenasa/metabolismo , Especificidad de Órganos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Ubiquitinación/efectos de los fármacos , Úvea/metabolismo , Úvea/patología , Neoplasias de la Úvea/tratamiento farmacológico , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/metabolismo , Neoplasias de la Úvea/patología
18.
Invest Ophthalmol Vis Sci ; 47(9): 4042-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16936121

RESUMEN

PURPOSE: To investigate the ocular hypotensive effect of the prostanoid EP2 receptor agonist butaprost and to establish its mechanism of action. METHODS: All experiments were performed in cynomolgus monkeys after topical application of butaprost (0.1%). The effects of butaprost on aqueous humor flow were determined by fluorophotometry. Total outflow facility was measured by the two-level, constant-pressure perfusion method, and uveoscleral outflow was determined by perfusion of FITC-labeled dextran through the anterior chamber. Effects on ocular morphology were studied after tissue fixation with transcardial perfusion by paraformaldehyde and immersion fixation of the globe, in animals subjected to long-term treatment with butaprost. Conscious ocular normotensive monkeys and monkeys with unilateral ocular hypertension were used for intraocular pressure (IOP) studies. RESULTS: Butaprost had no significant effect on aqueous humor flow or total outflow facility in ocular normotensive monkeys. Uveoscleral outflow was significantly higher in the butaprost treated eyes than in vehicle treated eyes, 1.03 +/- 0.20 vs. 0.53 +/- 0.18 microL.min(-1). After a 1-year treatment with butaprost, the morphology of the ciliary muscle was changed, showing increased spaces between ciliary muscle bundles and the apparent formation of new outflow channels. In many instances, changes were observed in the trabecular meshwork as well. Butaprost, in a single 0.1% dose, decreased IOP significantly in ocular normotensive monkeys and reduced IOP in laser-induced glaucomatous monkey eyes to the same level as that in the ocular normotensive contralateral eyes. CONCLUSIONS: The prostanoid EP2 receptor agonist butaprost appears to lower IOP by increasing uveoscleral outflow, according to both physiological and morphologic findings. Although the prostanoid EP2 receptor is structurally and functionally distinct from the FP receptor, the effects of EP2 and FP receptor stimulation on aqueous humor outflow are similar.


Asunto(s)
Alprostadil/análogos & derivados , Humor Acuoso/metabolismo , Presión Intraocular/efectos de los fármacos , Hipertensión Ocular/tratamiento farmacológico , Prostaglandinas E Sintéticas/farmacología , Receptores de Prostaglandina E/agonistas , Esclerótica/efectos de los fármacos , Úvea/efectos de los fármacos , Administración Tópica , Alprostadil/farmacología , Animales , Cuerpo Ciliar/efectos de los fármacos , Cuerpo Ciliar/patología , Dextranos/metabolismo , Modelos Animales de Enfermedad , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Fluorofotometría , Macaca fascicularis , Músculo Liso/efectos de los fármacos , Músculo Liso/patología , Hipertensión Ocular/patología , Subtipo EP2 de Receptores de Prostaglandina E , Esclerótica/metabolismo , Malla Trabecular , Úvea/metabolismo
19.
Ocul Immunol Inflamm ; 14(5): 277-83, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17056461

RESUMEN

PURPOSE: Demonstration of experimental autoimmune uveitis (EAU) with extremely small, fragmented peptides (12-30 amino acid residues) of interphotoreceptor retinoid-binding protein (IRPB). METHOD: Very small fragmented peptides (no. 854, 888, 907, and 1057) were conjugated to heat-killed Group A Streptococcus cells and administered as a single intravenous injection to Lewis rats. A non-uveitogenic peptide 950 was also conjugated to heat-killed Streptococcus and administered. Administration of a mixture of small peptides and Streptococcus was a control for the peptides conjugated with Streptococcus. RESULTS: The uveitogenic peptide/Streptococcus conjugates produced uveitis inflammatory responses in the uvea, retina and pineal gland. Administration of mixtures of small peptides and Streptococcus cells, and a non-uveitogenic peptide 950 conjugated with Streptococcus did not produce autoimmune uveitis. CONCLUSIONS: Since mixtures of small uveitogenic peptides and Streptococcal cells did not develop autoimmune uveitis, conjugated Streptococcal cells provided a vehicle for macrophage phagocytosos of very small uveitogenic IRBP peptides. Subsequent antigen presentation from macrophages to lymphocytes developed autoimmune uveitis. Peptide 888, one of four IRBP peptides that encompass the major uveitogenic domain, proved to be the most effective in development of uveitis.


Asunto(s)
Enfermedades Autoinmunes/inducido químicamente , Proteínas del Ojo/inmunología , Inmunoconjugados/toxicidad , Fragmentos de Péptidos/inmunología , Proteínas de Unión al Retinol/inmunología , Streptococcus pyogenes/inmunología , Uveítis/inducido químicamente , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Activación de Macrófagos , Modelos Animales , Glándula Pineal/efectos de los fármacos , Glándula Pineal/inmunología , Glándula Pineal/patología , Ratas , Ratas Endogámicas Lew , Retina/efectos de los fármacos , Retina/inmunología , Retina/patología , Úvea/efectos de los fármacos , Úvea/inmunología , Úvea/patología , Uveítis/inmunología , Uveítis/patología
20.
J Ocul Pharmacol Ther ; 22(2): 86-92, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16722794

RESUMEN

This study examines, in 11 cynomolgus monkeys with unilateral laser-induced glaucoma, the ocular hypotensive mechanism of action of AL-6598, partial agonist at the DP and EP prostanoid receptors. In a crossover fashion, both eyes of each monkey were dosed twice daily with 25 microL of either AL-6598 0.01% or vehicle for 2 days and on the morning of the 3rd day. Measurements were made on day 3 of each treatment. Alternative treatments were separated by at least 2 weeks. Intraocular pressures (IOPs) were measured by pneumatonometry and aqueous flow and outflow facility by fluorophotometry. Uveoscleral outflow was calculated mathematically. In the normotensive eyes, compared to vehicle treatment, AL-6598 decreased IOP from 22.5 +/- 0.7 to 18.7 +/- 0.9 mmHg (P = 0.006), increased uveoscleral outflow from 0.47 +/- 0.17 to 1.22 +/- 0.17 microL/min (P = 0.03), and increased aqueous flow from 1.49 +/- 0.10 to 1.93 +/- 0.13 microL/min (P = 0.01). No measurement in AL-6598-treated hypertensive eyes was significantly different from vehicle treatment. It is concluded that AL-6598 reduces IOP by increasing uveoscleral outflow in normotensive eyes of ketamine-sedated monkeys, despite an increase in aqueous flow. This effect is different from that of PGD(2), which decreases aqueous flow, and of the selective DP receptor agonist, BW245C, which increases both outflow facility and uveoscleral outflow in addition to decreasing aqueous flow.


Asunto(s)
Humor Acuoso/fisiología , Dinoprost/análogos & derivados , Glaucoma/tratamiento farmacológico , Presión Intraocular/efectos de los fármacos , Receptores Inmunológicos/agonistas , Receptores de Prostaglandina/agonistas , Animales , Dinoprost/administración & dosificación , Dinoprost/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Fluorofotometría , Glaucoma/metabolismo , Glaucoma/fisiopatología , Macaca fascicularis , Esclerótica/efectos de los fármacos , Esclerótica/fisiología , Resultado del Tratamiento , Úvea/efectos de los fármacos , Úvea/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA