Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
1.
Invest Ophthalmol Vis Sci ; 62(10): 10, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34379096

RESUMO

Purpose: In spite of clear differences in tissue function and significance to ocular disease, little is known about how immune responses differ between the retina and uveal tract. To this end we compared the effects of acute systemic inflammation on myeloid cells within the mouse retina, iris-ciliary body, and choroid. Methods: Systemic inflammation was induced in Cx3cr1gfp/gfp and CD11c-eYFP Crb1wt/wtmice by intraperitoneal lipopolysaccharide (LPS). In vivo fundus imaging was performed at two, 24, and 48 hours after LPS, and ocular tissue wholemounts were immunostained and studied by confocal microscopy. Flow cytometry was used to investigate the expression of activation markers (MHC class II, CD80, CD86) on myeloid cell populations at 24 hours. For functional studies, retinal microglia were isolated from LPS-exposed mice and cocultured with naïve OT-II CD4+ T-cells and ovalbumin peptide. T-cell proliferation was measured by flow cytometry and cytokine assays. Results: Systemic LPS altered the density and morphology of retinal microglia; however, retinal microglia did not upregulate antigen presentation markers and failed to stimulate naïve CD4+ T-cell proliferation in vitro. In contrast, uveal tract myeloid cells displayed a phenotype consistent with late-activated antigen-presenting cells at 24 hours. Systemic LPS induced remodeling of myeloid populations within the uveal tract, particularly in the choroid, where dendritic cells were partially displaced by macrophages at 24 hours. Conclusions: The disparate myeloid cell responses in the retina and uveal tract after systemic LPS highlight differential regulation of innate immunity within these tissue environments, observations that underpin and advance our understanding of ocular immune privilege.


Assuntos
Células Dendríticas/patologia , Inflamação/patologia , Macrófagos/patologia , Células Mieloides/patologia , Retina/patologia , Úvea/patologia , Animais , Células Dendríticas/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/imunologia , Camundongos Endogâmicos BALB C , Microscopia Confocal , Células Mieloides/imunologia , Retina/imunologia , Úvea/imunologia
2.
Front Immunol ; 12: 623725, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995347

RESUMO

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.


Assuntos
Anti-Inflamatórios/uso terapêutico , Autoimunidade/efeitos dos fármacos , Produtos Biológicos/uso terapêutico , Tolerância Imunológica/efeitos dos fármacos , Imunoterapia , Úvea/efeitos dos fármacos , Uveíte/terapia , Animais , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Terapia de Alvo Molecular , Transdução de Sinais , Úvea/imunologia , Úvea/metabolismo , Uveíte/imunologia , Uveíte/metabolismo
3.
Biomed Pharmacother ; 136: 111291, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33493870

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Doenças Autoimunes/prevenção & controle , Diferenciação Celular/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Receptor Notch1/metabolismo , Células Th17/efeitos dos fármacos , Úvea/efeitos dos fármacos , Uveíte/prevenção & controle , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mapas de Interação de Proteínas , Ratos Endogâmicos Lew , Receptor Notch1/genética , Transdução de Sinais , Células Th17/imunologia , Células Th17/metabolismo , Úvea/imunologia , Úvea/metabolismo , Uveíte/genética , Uveíte/imunologia , Uveíte/metabolismo
4.
Front Immunol ; 11: 601272, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33343573

RESUMO

Previous studies have shown that CD73 is pivotal in the conversion of pro-inflammatory adenosine triphosphate into anti-inflammatory adenosine and that immune cells of the same type that express different levels of CD73 are functionally distinct. In this study we show that adenosine enhances the Th17 promoting effect of dendritic cells (DCs), and DCs expressing CD73 critically augment Th17 responses. Bone marrow dendritic cells (BMDCs) do not constantly express CD73; however, a significant portion of the BMDCs expressed CD73 after exposure to Toll-like receptor ligand, leading to stronger Th17 responses by converting adenosine monophosphate to adenosine. We show that the CD73+ BMDCs play a critical role in cascading Th17 responses, and CD73+ BMDCs are functionally augmented after treatment with Toll-like receptor ligand. Splenic antigen presenting cells (DCs) of CD73-/- mouse have a poor Th17-stimulating effect, even after exposure to lipopolysaccharide (LPS) or γδ T cells, indicating that induction of CD73+ DCs is critically involved in augmented Th17 responses. We conclude that CD73+ DCs critically trigger cascading Th17 responses, and the activated Th17 cells that express CD73 further augment Th17 responses, leading to cascading exacerbation. Hence, disabling the CD73 function of DCs should block this cascading response and mitigate Th17 responses.


Assuntos
5'-Nucleotidase/metabolismo , Doenças Autoimunes/enzimologia , Células Dendríticas/enzimologia , Células Th17/metabolismo , Úvea/enzimologia , Uveíte/enzimologia , 5'-Nucleotidase/genética , Adenosina/metabolismo , Monofosfato de Adenosina/farmacologia , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Genes Codificadores da Cadeia delta de Receptores de Linfócitos T , Lipopolissacarídeos/farmacologia , Ativação Linfocitária , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Células Th17/imunologia , Úvea/imunologia , Úvea/patologia , Uveíte/genética , Uveíte/imunologia , Uveíte/patologia
5.
Front Immunol ; 11: 575669, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33117376

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Doenças Autoimunes/prevenção & controle , Linfócitos T CD4-Positivos/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Lamiaceae/química , Extratos Vegetais/farmacologia , Uveíte/prevenção & controle , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Autoimunidade/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Proteína Quinase CDC2/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Fosforilação , Transdução de Sinais , Úvea/efeitos dos fármacos , Úvea/imunologia , Úvea/metabolismo , Úvea/patologia , Uveíte/imunologia , Uveíte/metabolismo , Uveíte/patologia
6.
Sci Rep ; 10(1): 11377, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32647297

RESUMO

This study reports the use of cell-type-specific in vivo bioluminescence to measure intraocular immune cell population dynamics during the course of inflammation in a mouse model of uveitis. Transgenic lines expressing luciferase in inflammatory cell subsets (myeloid cells, T cells, and B cells) were generated and ocular bioluminescence was measured serially for 35 days following uveitis induction. Ocular leukocyte populations were identified using flow cytometry and compared to the ocular bioluminescence profile. Acute inflammation is neutrophilic (75% of ocular CD45 + cells) which is reflected by a significant increase in ocular bioluminescence in one myeloid reporter line on day 2. By day 7, the ocular T cell population increases to 50% of CD45 + cells, leading to a significant increase in ocular bioluminescence in the T cell reporter line. While initially negligible (< 1% of CD45 + cells), the ocular B cell population increases to > 4% by day 35. This change is reflected by a significant increase in the ocular bioluminescence of the B cell reporter line starting on day 28. Our data demonstrates that cell-type-specific in vivo bioluminescence accurately detects changes in multiple intraocular immune cell populations over time in experimental uveitis. This assay could also be useful in other inflammatory disease models.


Assuntos
Modelos Animais de Doenças , Medições Luminescentes/métodos , Tomografia de Coerência Óptica , Uveíte/diagnóstico , Animais , Animais Geneticamente Modificados , Linfócitos B/química , Linfócitos B/imunologia , Estudos de Viabilidade , Feminino , Genes Reporter/genética , Humanos , Luciferases/química , Luciferases/genética , Masculino , Camundongos , Células Mieloides/química , Células Mieloides/imunologia , Linfócitos T/química , Linfócitos T/imunologia , Úvea/citologia , Úvea/imunologia , Uveíte/imunologia
7.
Front Immunol ; 11: 975, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32508841

RESUMO

Experimental autoimmune uveoretinitis (EAU) is a mouse model of human autoimmune uveitis. EAU spontaneously resolves and is marked by ocular autoantigen-specific regulatory immunity in the spleen. Kallikrein binding protein (KBP) or kallistatin is a serine proteinase inhibitor that inhibits angiogenesis and inflammation, but its role in autoimmune uveitis has not been explored. We report that T cells activation is inhibited and EAU is attenuated in human KBP (HKBP) mice with no significant difference in the Treg population that we previously identified both before and after recovery from EAU. Moreover, following EAU immunization HKBP mice have potent ocular autoantigen specific regulatory immunity that is functionally suppressive.


Assuntos
Doenças Autoimunes/prevenção & controle , Autoimunidade , Ativação Linfocitária , Serpinas/metabolismo , Baço/metabolismo , Linfócitos T/metabolismo , Úvea/metabolismo , Uveíte/prevenção & controle , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Serpinas/genética , Baço/imunologia , Linfócitos T/imunologia , Úvea/imunologia , Úvea/patologia , Uveíte/genética , Uveíte/imunologia , Uveíte/metabolismo
8.
Front Immunol ; 11: 603939, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33613524

RESUMO

A20 is a negative regulator of inflammation and immunity and plays a role in several autoimmune and inflammatory diseases. Here, we demonstrate that A20 overexpression significantly ameliorates severity of EAU by inhibiting the infiltration of Th1 and Th17 cells, and by protecting integrity of the blood retinal barrier. In vitro studies showed that A20 silencing could promote CD4+T cells toward a Th1 and Th17 phenotype. A decreased expression of A20 in CD4+T cells was noticed in active BD patients but not in VKH patients. Furthermore, silencing of A20 in hRPE cells induced the production of IL-6, IL-8, and MCP-1 and downregulated ZO-1 and occludin expression which is mediated by inhibition of MAPK and NF-κB pathways. This study reveals a mechanism by which A20 prevents autoimmune uveitis.


Assuntos
Doenças Autoimunes/metabolismo , Barreira Hematorretiniana , Linfócitos T CD4-Positivos/metabolismo , Quimiotaxia de Leucócito , Células Epiteliais/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Úvea/metabolismo , Uveíte/metabolismo , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Doenças Autoimunes/prevenção & controle , Linfócitos T CD4-Positivos/imunologia , Estudos de Casos e Controles , Citocinas/metabolismo , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/patologia , Humanos , Camundongos , Fenótipo , Epitélio Pigmentado da Retina/imunologia , Epitélio Pigmentado da Retina/patologia , Transdução de Sinais , Proteínas de Junções Íntimas/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Úvea/imunologia , Úvea/patologia , Uveíte/imunologia , Uveíte/patologia , Uveíte/prevenção & controle
9.
Front Immunol ; 11: 608377, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33569055

RESUMO

Immune privilege (IP), a term introduced to explain the unpredicted acceptance of allogeneic grafts by the eye and the brain, is considered a unique property of these tissues. However, immune responses are modified by the tissue in which they occur, most of which possess IP to some degree. The eye therefore displays a spectrum of IP because it comprises several tissues. IP as originally conceived can only apply to the retina as it contains few tissue-resident bone-marrow derived myeloid cells and is immunologically shielded by a sophisticated barrier - an inner vascular and an outer epithelial barrier at the retinal pigment epithelium. The vascular barrier comprises the vascular endothelium and the glia limitans. Immune cells do not cross the blood-retinal barrier (BRB) despite two-way transport of interstitial fluid, governed by tissue oncotic pressure. The BRB, and the blood-brain barrier (BBB) mature in the neonatal period under signals from the expanding microbiome and by 18 months are fully established. However, the adult eye is susceptible to intraocular inflammation (uveitis; frequency ~200/100,000 population). Uveitis involving the retinal parenchyma (posterior uveitis, PU) breaches IP, while IP is essentially irrelevant in inflammation involving the ocular chambers, uveal tract and ocular coats (anterior/intermediate uveitis/sclerouveitis, AU). Infections cause ~50% cases of AU and PU but infection may also underlie the pathogenesis of immune-mediated "non-infectious" uveitis. Dysbiosis accompanies the commonest form, HLA-B27-associated AU, while latent infections underlie BRB breakdown in PU. This review considers the pathogenesis of uveitis in the context of IP, infection, environment, and the microbiome.


Assuntos
Bactérias/imunologia , Microbioma Gastrointestinal , Privilégio Imunológico , Intestinos/microbiologia , Úvea/imunologia , Uveíte/imunologia , Animais , Bactérias/metabolismo , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Disbiose , Predisposição Genética para Doença , Antígeno HLA-B27/genética , Antígeno HLA-B27/imunologia , Humanos , Fatores de Risco , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/microbiologia , Úvea/metabolismo , Uveíte/genética , Uveíte/metabolismo , Uveíte/microbiologia
10.
Front Immunol ; 11: 608134, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33469457

RESUMO

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.


Assuntos
Antígeno HLA-B27/imunologia , Úvea/imunologia , Uveíte Anterior/imunologia , Doença Aguda , Animais , Bactérias/imunologia , Bactérias/metabolismo , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Microbioma Gastrointestinal , Predisposição Genética para Doença , Antígeno HLA-B27/genética , Haplótipos , Humanos , Imunossupressores/uso terapêutico , Intestinos/microbiologia , Inibidores do Fator de Necrose Tumoral/uso terapêutico , Úvea/efeitos dos fármacos , Úvea/metabolismo , Uveíte Anterior/tratamento farmacológico , Uveíte Anterior/genética , Uveíte Anterior/microbiologia
11.
Biomed Pharmacother ; 120: 109381, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31542616

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Bases de Dados de Proteínas , Medicamentos de Ervas Chinesas/farmacologia , Mapas de Interação de Proteínas , Biologia de Sistemas , Úvea/efeitos dos fármacos , Uveíte/tratamento farmacológico , Animais , Modelos Animais de Doenças , Feminino , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células T Matadoras Naturais/efeitos dos fármacos , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Ratos Endogâmicos Lew , Transdução de Sinais , Úvea/imunologia , Úvea/metabolismo , Úvea/patologia , Uveíte/imunologia , Uveíte/metabolismo , Uveíte/patologia
12.
Cytometry B Clin Cytom ; 92(4): 286-290, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-26860883

RESUMO

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.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Doxorrubicina/análogos & derivados , Linfoma Difuso de Grandes Células B/diagnóstico , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Metotrexato/uso terapêutico , Uveíte/diagnóstico , Antígenos CD/genética , Antígenos CD/imunologia , Humor Aquoso/efeitos dos fármacos , Humor Aquoso/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/patologia , Biomarcadores/metabolismo , Ciclofosfamida/uso terapêutico , Diagnóstico Diferencial , Doxorrubicina/uso terapêutico , Citometria de Fluxo/métodos , Expressão Gênica , Humanos , Inflamação , Linfoma Difuso de Grandes Células B/imunologia , Linfoma Difuso de Grandes Células B/patologia , Masculino , Pessoa de Meia-Idade , Paracentese , Prednisona/uso terapêutico , Resultado do Tratamento , Úvea/efeitos dos fármacos , Úvea/imunologia , Úvea/patologia , Uveíte/tratamento farmacológico , Uveíte/imunologia , Uveíte/patologia , Vincristina/uso terapêutico
14.
Anticancer Agents Med Chem ; 17(2): 190-205, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27109020

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Epigênese Genética/efeitos dos fármacos , Imunoterapia/métodos , Melanoma/genética , Melanoma/terapia , Úvea/efeitos dos fármacos , Neoplasias Uveais/genética , Neoplasias Uveais/terapia , Animais , Antineoplásicos/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imunidade/efeitos dos fármacos , Melanoma/imunologia , Melanoma/patologia , Úvea/imunologia , Úvea/metabolismo , Úvea/patologia , Neoplasias Uveais/imunologia , Neoplasias Uveais/patologia
15.
Immunology ; 146(2): 301-11, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26152845

RESUMO

Experimental autoimmune uveoretinitis (EAU) in the C57BL/6J mouse is a model of non-infectious posterior segment intraocular inflammation that parallels clinical features of the human disease. The purpose of this study was to analyse the immune response to the four murine subunits of retinol binding protein-3 (RBP-3) to identify pathogenic epitopes to investigate the presence of intramolecular epitope spreading during the persistent inflammation phase observed in this model of EAU. Recombinant murine subunits of the RBP-3 protein were purified and used to immunize C57BL/6J mice to induce EAU. An overlapping peptide library was used to screen RBP-3 subunit 3 for immunogenicity and pathogenicity. Disease phenotype and characterization of pathogenic subunits and peptides was undertaken by topical endoscopic fundal imaging, immunohistochemistry, proliferation assays and flow cytometry. RBP-3 subunits 1, 2 and 3 induced EAU in the C57BL/6J mice, with subunit 3 eliciting the most destructive clinical disease. Within subunit 3 we identified a novel uveitogenic epitope, 629-643. The disease induced by this peptide was comparable to that produced by the uveitogenic 1-20 peptide. Following immunization, peptide-specific responses by CD4(+) and CD8(+) T-cell subsets were detected, and cells from both populations were present in the retinal inflammatory infiltrate. Intramolecular epitope spreading between 629-643 and 1-20 was detected in mice with clinical signs of disease. The 629-643 RBP-3 peptide is a major uveitogenic peptide for the induction of EAU in C57BL/6J mice and the persistent clinical disease induced with one peptide leads to epitope spreading.


Assuntos
Doenças Autoimunes/imunologia , Epitopos/imunologia , Proteínas do Olho/imunologia , Fragmentos de Peptídeos/imunologia , Retina/imunologia , Retinite/imunologia , Proteínas de Ligação ao Retinol/imunologia , Úvea/imunologia , Uveíte/imunologia , Animais , Doenças Autoimunes/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Mapeamento de Epitopos , Epitopos/genética , Proteínas do Olho/genética , Feminino , Humanos , Ativação Linfocitária , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/genética , Fenótipo , Retina/patologia , Retinite/patologia , Proteínas de Ligação ao Retinol/genética , Índice de Gravidade de Doença , Úvea/patologia , Uveíte/patologia
16.
Clin Immunol ; 157(2): 198-204, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25701800

RESUMO

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.


Assuntos
Doenças Autoimunes/imunologia , Boranos/farmacologia , Carbonatos/farmacologia , RNA Mensageiro/efeitos dos fármacos , Retina/efeitos dos fármacos , Retinite/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Úvea/efeitos dos fármacos , Uveíte/imunologia , Animais , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/patologia , Citocinas/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Retina/imunologia , Retina/patologia , Retinite/induzido quimicamente , Retinite/patologia , Proteínas de Ligação ao Retinol/toxicidade , Baço/efeitos dos fármacos , Baço/metabolismo , Linfócitos T Reguladores/imunologia , Úvea/imunologia , Úvea/patologia , Uveíte/induzido quimicamente , Uveíte/patologia
17.
J Proteome Res ; 12(12): 5812-9, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24144192

RESUMO

Autoimmune uveitis is characterized by crossing of blood-retinal barrier (BRB) by autoaggressive immune cells. Equine recurrent uveitis (ERU) is a valuable spontaneous model for autoimmune uveitis and analyses of differentially expressed proteins in ERU unraveled changed protein clusters in target tissues and immune system. Healthy eyes are devoid of leukocytes. In ERU, however, leukocytes enter the inner eye and subsequently destroy it. Molecular mechanisms enabling cell migration through BRB still remain elusive. Previously, we detected decreased talin 1 expression in blood-derived granulocytes of ERU cases, linking the innate immune system to ERU. Because changes in leukocyte protein expression pattern may play a role in pathological abnormalities leading to migration ability, we aimed at identifying interactors of talin 1 in leukocytes with immunoprecipitation, followed by LC-MS/MS for candidate identification. This enabled us to identify CD90 (Thy1) as novel interactor of talin 1 besides several other interactors. In blood-derived granulocytes from healthy individuals, CD90 was highly abundant and significantly reduced in ERU, especially in effector cells. Connection between talin 1 and CD90 and their expression differences in inflammation is an interesting novel finding allowing deeper insight into immune response of innate immune system and granulocyte migration ability in this organ-specific autoimmune disease.


Assuntos
Granulócitos/metabolismo , Doenças dos Cavalos/genética , Talina/genética , Antígenos Thy-1/genética , Úvea/metabolismo , Uveíte/veterinária , Animais , Autoanticorpos/biossíntese , Doenças Autoimunes , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Barreira Hematorretiniana , Estudos de Casos e Controles , Movimento Celular , Cromatografia Líquida , Regulação da Expressão Gênica , Granulócitos/imunologia , Granulócitos/patologia , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/metabolismo , Doenças dos Cavalos/patologia , Cavalos , Imunoprecipitação , Espectrometria de Massas , Anotação de Sequência Molecular , Ligação Proteica , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Talina/imunologia , Talina/metabolismo , Antígenos Thy-1/imunologia , Antígenos Thy-1/metabolismo , Úvea/imunologia , Úvea/patologia , Uveíte/imunologia , Uveíte/metabolismo , Uveíte/patologia
18.
Invest Ophthalmol Vis Sci ; 53(7): 4158-69, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-22562518

RESUMO

PURPOSE: To investigate the anti-inflammatory effect of an adenosine monophosphate (AMP) analog, aminoimidazole carboxamide ribonucleotide (AICAR), in experimental autoimmune uveoretinitis (EAU). METHODS: C57BL/6 mice were injected daily with AICAR (200 mg/kg, intraperitoneally [IP]) from day 0, the day of interphotoreceptor retinoid-binding protein (IRBP) immunization, until day 21. The severity of uveitis was assessed clinically and histopathologically. T-cell proliferation and cytokine production of IFN-γ, IL-17, and IL-10 in response to IRBP stimulation were determined. In addition, regulatory T-cell (Treg) populations were measured. Co-stimulatory molecule expression (CD40, 80, 86, and I-Ab) on dendritic cells (DCs) in EAU and on bone marrow-derived dendritic cells (BMDCs) treated with AICAR was measured. RESULTS: AICAR treatment significantly reduced clinical and histologic severity of EAU as well as ocular cytokine production. An anti-inflammatory effect associated with the inhibition of T-cell proliferation and Th1 and Th17 cytokine production was observed. Increases in the Th2 response and Treg population were not observed with AICAR treatment. AICAR did significantly inhibit BMDC maturation by reducing co-stimulatory molecule expression. CONCLUSIONS: AICAR attenuates EAU by preventing generation of Ag-specific Th1 and Th17 cells. Impaired DC maturation may be an underlying mechanism for this anti-inflammatory effect observed with AICAR.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Doenças Autoimunes/tratamento farmacológico , Imunidade Celular , Ribonucleotídeos/administração & dosagem , Linfócitos T/imunologia , Uveíte/tratamento farmacológico , Aminoimidazol Carboxamida/administração & dosagem , Aminoimidazol Carboxamida/uso terapêutico , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Western Blotting , Proliferação de Células , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/uso terapêutico , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Ribonucleotídeos/uso terapêutico , Resultado do Tratamento , Úvea/imunologia , Úvea/metabolismo , Úvea/patologia , Uveíte/imunologia , Uveíte/metabolismo
19.
Invest Ophthalmol Vis Sci ; 51(9): 4661-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20375337

RESUMO

PURPOSE: To characterize the regulatory effect of gammadelta T cells in the activation of IL-17+ uveitogenic T cells. METHODS: The authors administered the gammadelta TCR-specific antibody GL3 to B6 mice before or after antigen immunization and examined Th1- or Th17-polarized T-cell responses. The intensity of Th17 responses was also examined in responder T cells containing varying numbers of gammadelta T cells. RESULTS: GL3 treatment resulted in varying degrees of depletion of circulating gammadelta T cells, depending on when the antibody was administered. The intensity of the alphabetaTCR+IL-17+, but not the alphabetaTCR+IFN-gamma+, IRBP-specific T-cell responses was correlated to the percentage of gammadelta T cells in the responder T cells. Kinetic studies showed that early IL-17+ T cells were primarily gammadelta T cells, with a later gradual shift to alphabeta T cells. A close association was seen between the intensity of the IL-17+ autoreactive T-cell response and the percentage of gammadelta T cells in the responder T cells. Although a modest increase in gammadelta T cells among the responder T cells promoted the expansion of IL-17+ alphabetaTCR+ T cells, a higher proportion of gammadelta T cells inhibited it. CONCLUSIONS: gammadelta T cells are actively involved in the generation of alphabetaTCR+IL-17+ T cells. The number of gammadelta T cells and the alphabeta/gammadelta T-cell ratio in the responder T cells regulate the intensity of the Th17-type autoreactive T-cell response.


Assuntos
Interleucina-17/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/metabolismo , Úvea/imunologia , Uveíte/imunologia , Animais , Autoanticorpos/farmacologia , Autoantígenos/farmacologia , Autoimunidade/imunologia , Feminino , Imunização , Interleucina-17/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linfócitos T/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Úvea/citologia
20.
Gene Ther ; 15(22): 1478-88, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18580969

RESUMO

Uveitis is a sight threatening inflammatory disorder that remains a significant cause of visual loss. We investigated lentiviral gene delivery of interleukin 1 receptor antagonist (IL-1ra) or interleukin (IL)-10 to ameliorate murine endotoxin-induced uveitis (EIU). An human immunodeficiency virus-1-based vector containing the mIL-1ra or mIL-10 cDNA demonstrated high expression of biologically active cytokine. Following administration of Lenti.GFP into the anterior chamber, transgene expression was observed in corneal endothelial cells, trabecular meshwork and iris cells. To treat EIU, mice were injected with Lenti.IL-1ra, Lenti.IL-10 or a combination of these. EIU was induced 14 days after vector administration and mice were culled 12 h following disease induction. Lenti.IL-1ra or Lenti.IL-10-treated eyes showed significantly lower mean inflammatory cell counts in the anterior and posterior chambers compared with controls. The aqueous total protein content was also significantly lower in treated eyes, demonstrating better preservation of the blood-ocular barrier. Furthermore, the treated eyes showed less in vivo fluorescein leakage from inner retinal vessels compared with controls. The combination of both IL-1ra and IL-10 had no additive effect. Thus, lentiviral gene delivery of IL-1ra or IL-10 significantly reduces the severity of experimental uveitis, suggesting that lentiviral-mediated expression of immunomodulatory genes in the anterior chamber offers an opportunity to treat uveitis.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , HIV-1/genética , Proteína Antagonista do Receptor de Interleucina 1/genética , Interleucina-10/genética , Uveíte/terapia , Animais , Feminino , Expressão Gênica , Vetores Genéticos/genética , Humanos , Injeções , Proteína Antagonista do Receptor de Interleucina 1/imunologia , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Transdução Genética/métodos , Transgenes , Úvea/imunologia , Uveíte/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...