Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 197
Filter
1.
Front Immunol ; 12: 623725, 2021.
Article in English | MEDLINE | ID: mdl-33995347

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Autoimmunity/drug effects , Biological Products/therapeutic use , Immune Tolerance/drug effects , Immunotherapy , Uvea/drug effects , Uveitis/therapy , Animals , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Molecular Targeted Therapy , Signal Transduction , Uvea/immunology , Uvea/metabolism , Uveitis/immunology , Uveitis/metabolism
2.
Biomed Pharmacother ; 136: 111291, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33493870

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Autoimmune Diseases/prevention & control , Cell Differentiation/drug effects , Drugs, Chinese Herbal/pharmacology , Receptor, Notch1/metabolism , Th17 Cells/drug effects , Uvea/drug effects , Uveitis/prevention & control , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Female , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Interaction Maps , Rats, Inbred Lew , Receptor, Notch1/genetics , Signal Transduction , Th17 Cells/immunology , Th17 Cells/metabolism , Uvea/immunology , Uvea/metabolism , Uveitis/genetics , Uveitis/immunology , Uveitis/metabolism
3.
Front Immunol ; 11: 575669, 2020.
Article in English | MEDLINE | ID: mdl-33117376

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Autoimmune Diseases/prevention & control , CD4-Positive T-Lymphocytes/drug effects , Drugs, Chinese Herbal/pharmacology , Lamiaceae/chemistry , Plant Extracts/pharmacology , Uveitis/prevention & control , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Autoimmunity/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CDC2 Protein Kinase/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Phosphorylation , Signal Transduction , Uvea/drug effects , Uvea/immunology , Uvea/metabolism , Uvea/pathology , Uveitis/immunology , Uveitis/metabolism , Uveitis/pathology
4.
Front Immunol ; 11: 975, 2020.
Article in English | MEDLINE | ID: mdl-32508841

ABSTRACT

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.


Subject(s)
Autoimmune Diseases/prevention & control , Autoimmunity , Lymphocyte Activation , Serpins/metabolism , Spleen/metabolism , T-Lymphocytes/metabolism , Uvea/metabolism , Uveitis/prevention & control , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Mice, Transgenic , Serpins/genetics , Spleen/immunology , T-Lymphocytes/immunology , Uvea/immunology , Uvea/pathology , Uveitis/genetics , Uveitis/immunology , Uveitis/metabolism
5.
Front Immunol ; 11: 603939, 2020.
Article in English | MEDLINE | ID: mdl-33613524

ABSTRACT

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.


Subject(s)
Autoimmune Diseases/metabolism , Blood-Retinal Barrier , CD4-Positive T-Lymphocytes/metabolism , Chemotaxis, Leukocyte , Epithelial Cells/metabolism , Retinal Pigment Epithelium/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Uvea/metabolism , Uveitis/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , CD4-Positive T-Lymphocytes/immunology , Case-Control Studies , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/pathology , Humans , Mice , Phenotype , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/pathology , Signal Transduction , Tight Junction Proteins/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Uvea/immunology , Uvea/pathology , Uveitis/immunology , Uveitis/pathology , Uveitis/prevention & control
6.
Front Immunol ; 11: 608377, 2020.
Article in English | MEDLINE | ID: mdl-33569055

ABSTRACT

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.


Subject(s)
Bacteria/immunology , Gastrointestinal Microbiome , Immune Privilege , Intestines/microbiology , Uvea/immunology , Uveitis/immunology , Animals , Bacteria/metabolism , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Dysbiosis , Genetic Predisposition to Disease , HLA-B27 Antigen/genetics , HLA-B27 Antigen/immunology , Humans , Risk Factors , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/microbiology , Uvea/metabolism , Uveitis/genetics , Uveitis/metabolism , Uveitis/microbiology
7.
Front Immunol ; 11: 608134, 2020.
Article in English | MEDLINE | ID: mdl-33469457

ABSTRACT

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.


Subject(s)
HLA-B27 Antigen/immunology , Uvea/immunology , Uveitis, Anterior/immunology , Acute Disease , Animals , Bacteria/immunology , Bacteria/metabolism , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Gastrointestinal Microbiome , Genetic Predisposition to Disease , HLA-B27 Antigen/genetics , Haplotypes , Humans , Immunosuppressive Agents/therapeutic use , Intestines/microbiology , Tumor Necrosis Factor Inhibitors/therapeutic use , Uvea/drug effects , Uvea/metabolism , Uveitis, Anterior/drug therapy , Uveitis, Anterior/genetics , Uveitis, Anterior/microbiology
8.
Biomed Pharmacother ; 120: 109381, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31542616

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Databases, Protein , Drugs, Chinese Herbal/pharmacology , Protein Interaction Maps , Systems Biology , Uvea/drug effects , Uveitis/drug therapy , Animals , Disease Models, Animal , Female , Humans , Mitogen-Activated Protein Kinases/metabolism , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Rats, Inbred Lew , Signal Transduction , Uvea/immunology , Uvea/metabolism , Uvea/pathology , Uveitis/immunology , Uveitis/metabolism , Uveitis/pathology
9.
Sci Rep ; 9(1): 9142, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31235702

ABSTRACT

In recent years, a significant number of studies have investigated the preventive role of vitamin D in a number of different neoplasms. In this study, we analyze various components of the vitamin D signaling pathways in the human uveal tract and uveal melanoma, including analysis of the expression of vitamin D receptors (VDR), the activating and inactivating hydroxylases, respectively, CYP27B1 and CYP24A1, and the retinoic acid-related orphan receptors (ROR) α (RORα) and γ (RORγ) in these tissues. We further analyzed the expression of VDR, CYP27B1, CYP24A1, and ROR in relation to melanin levels, clinical stage and prognosis. Our study indicated that the uveal melanoma melanin level inversely correlated with VDR expression. We further showed that vitamin D is metabolized in uveal melanoma. This is significant because until now there has been no paper published, that would describe presence of VDR, hydroxylases CYP27B1 and CYP24A1, and RORα and RORγ in the human uveal tract and uveal melanomas. The outcomes of our research can contribute to the development of new diagnostic and therapeutic methods in uveal tract disorders, especially in uveal melanoma. The presented associations between vitamin D signaling elements and uveal melanoma in comparison to uveal tract encourage future clinical research with larger patients' population.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Melanins/metabolism , Melanoma/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Calcitriol/metabolism , Uveal Neoplasms/metabolism , Vitamin D3 24-Hydroxylase/metabolism , Adult , Aged , Aged, 80 and over , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Uvea/metabolism
10.
Chem Res Toxicol ; 32(2): 294-303, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30638013

ABSTRACT

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.


Subject(s)
Carbamates/metabolism , Phenylenediamines/metabolism , Retinal Pigments/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Uvea/drug effects , Animals , Carbamates/pharmacology , Dimerization , Male , Melanins/chemistry , Melanins/metabolism , Phenylenediamines/pharmacology , Rats , Rats, Long-Evans , Uvea/metabolism , Uvea/pathology
11.
AAPS J ; 20(6): 97, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30187172

ABSTRACT

Different types of imaging modalities are used in the diagnosis of ocular cancer. Selection of an imaging modality is based on the features of a tumor as well as the inherent characteristics of the imaging technique. It is vital to select an appropriate imaging modality in diagnosis of ocular tumor with confidence. This review focuses on five most commonly used imaging modalities, i.e., positron emission tomography-computed tomography (PET/CT), single photon emission computed tomography (SPECT), optical coherence tomography (OCT), ultrasound (US), and magnetic resonance imaging (MRI). The principal of imaging modalities is briefly explained, along with their role in the diagnosis and management of the most common ocular tumors such as retinoblastoma and uveal melanoma. Further, the diagnostic features of ocular tumors corresponding to each imaging modality and possibilities of utilizing imaging techniques in the process of ocular drug development are included in this review.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Drug Development/methods , Melanoma/diagnostic imaging , Retinal Neoplasms/diagnostic imaging , Retinoblastoma/diagnostic imaging , Uveal Neoplasms/diagnostic imaging , Administration, Ophthalmic , Adult , Antineoplastic Agents/administration & dosage , Child , Humans , Magnetic Resonance Imaging/methods , Melanoma/drug therapy , Positron Emission Tomography Computed Tomography/methods , Retina/diagnostic imaging , Retina/metabolism , Retina/pathology , Retinal Neoplasms/drug therapy , Retinoblastoma/drug therapy , Sensitivity and Specificity , Tissue Distribution , Tomography, Emission-Computed, Single-Photon/methods , Tomography, Optical Coherence/methods , Ultrasonography/methods , Uvea/diagnostic imaging , Uvea/metabolism , Uvea/pathology , Uveal Neoplasms/drug therapy
12.
Acta Ophthalmol ; 96(1): 31-38, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28444874

ABSTRACT

PURPOSE: To evaluate clinico-pathological and molecular prognostic factors in a well-defined series of posterior uveal melanoma (UM) with focus on chromosomal aberrations and mutations in the GNAQ, GNA11 and BRCA1-associated protein 1 (BAP1) genes. METHODS: Formalin-fixed paraffin-embedded (FFPE) tissue samples were obtained from 50 consecutive eyes enucleated for UM between 1993 and 2005. The material was tested for loss of chromosome 3 and gain of chromosome 8q gene signatures by selective molecular gene markers using multiplex ligation-dependent probe amplification (MLPA), and for DNA mutations in the GNAQ, GNA11 and BAP1 genes. RESULTS: After a mean follow-up of 83 months (range, 8-205 months), 21 patients had died of metastatic UM and 16 patients of other causes. Tumour diameter, ciliary body involvement, mixed/epithelioid cell types, mitotic index, Ki-67 proliferation index, loss of chromosome 3 and gain of chromosome 8q showed statistically significant associations with metastatic disease. There were no significant differences in the prevalence of GNAQ and GNA11 mutations between patients with or without metastatic disease. Mutational analysis of the BAP1 gene was performed in 32 primary UM and in five UM liver metastases. Nine different BAP1 missense mutations were identified. BAP1 mutations were not more common in metastasizing than in nonmetastasizing UM. CONCLUSION: The molecular gene markers showing loss of chromosome 3 and gain of 8q gene signatures were associated with an increased risk of metastatic disease. BRCA1-associated protein 1 (BAP1) gene mutation status had no prognostic significance. The frequency and spectrum of BAP1 mutations in UM may be more dependent on ethnicity and demographic variables than hitherto considered.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 3/genetics , DNA, Neoplasm/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits/genetics , Melanoma/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Uveal Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , DNA Mutational Analysis , Female , Follow-Up Studies , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Male , Melanoma/metabolism , Melanoma/secondary , Middle Aged , Multiplex Polymerase Chain Reaction , Mutation , Neoplasm Metastasis , Prognosis , Retrospective Studies , Time Factors , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Uvea/metabolism , Uvea/pathology , Uveal Neoplasms/metabolism , Uveal Neoplasms/secondary , Young Adult
13.
PLoS One ; 12(12): e0189675, 2017.
Article in English | MEDLINE | ID: mdl-29244840

ABSTRACT

The plasmacytoma variant translocation 1 gene (PVT1) plays an oncogenic role in the initiation and progression of multiple cancers. In this study, by using deep-sequencing data and follow-up data in the Cancer Genome Atlas-Uveal melanomas (TCGA-UVM), we assessed the association between the expression of PVT1 and clinicopathological characteristics of patients with uveal melanoma, the mechanism of its dysregulation and its prognostic value. Results showed that high PVT1 expression group had a higher proportion of epithelioid cell dominant disease (a more malignant histological subtype than spindle cell dominant disease) and more cases of extrascleral extension (a risk factor for metastasis) compared with the low PVT1 expression group. 61 out of 80 cases (76.3%) of primary uveal melanoma had PVT1 amplification in TCGA-UVM. In addition, PVT1 expression was strongly and negatively correlated with its methylation status (Pearson's r = -0.712, Spearman's r = -0.806). By performing univariate and multivariate analysis, we found that high PVT1 expression was an independent predictor of poor OS in patients with uveal melanoma (HR: 12.015, 95%CI: 1.854-77.876, p = 0.009). Based on these findings, we infer that PVT1 expression is modulated by both DNA amplification and methylation and its expression might serve as a valuable and specific prognostic biomarker in terms of OS in uveal melanoma.


Subject(s)
Biomarkers, Tumor/genetics , Melanoma/genetics , RNA, Long Noncoding/genetics , Uveal Neoplasms/genetics , Adult , Aged , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/pathology , Middle Aged , Prognosis , Uvea/metabolism , Uvea/pathology , Uveal Neoplasms/pathology
14.
Int J Mol Med ; 40(4): 1277-1284, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28849034

ABSTRACT

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.


Subject(s)
Melanins/antagonists & inhibitors , Melanocytes/drug effects , Monophenol Monooxygenase/antagonists & inhibitors , Plant Oils/pharmacology , Uvea/drug effects , Animals , Cell Line, Tumor , Gene Expression Regulation , Humans , Melanins/biosynthesis , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Organ Specificity , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Skin/drug effects , Skin/metabolism , Skin/pathology , Ubiquitination/drug effects , Uvea/metabolism , Uvea/pathology , Uveal Neoplasms/drug therapy , Uveal Neoplasms/genetics , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology
15.
Am J Pathol ; 187(6): 1327-1342, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28412300

ABSTRACT

Alkali burns to the eye constitute a leading cause of worldwide blindness. In recent case series, corneal transplantation revealed unexpected damage to the retina and optic nerve in chemically burned eyes. We investigated the physical, biochemical, and immunological components of retinal injury after alkali burn and explored a novel neuroprotective regimen suitable for prompt administration in emergency departments. Thus, in vivo pH, oxygen, and oxidation reduction measurements were performed in the anterior and posterior segment of mouse and rabbit eyes using implantable microsensors. Tissue inflammation was assessed by immunohistochemistry and flow cytometry. The experiments confirmed that the retinal damage is not mediated by direct effect of the alkali, which is effectively buffered by the anterior segment. Rather, pH, oxygen, and oxidation reduction changes were restricted to the cornea and the anterior chamber, where they caused profound uveal inflammation and release of proinflammatory cytokines. The latter rapidly diffuse to the posterior segment, triggering retinal damage. Tumor necrosis factor-α was identified as a key proinflammatory mediator of retinal ganglion cell death. Blockade, by either monoclonal antibody or tumor necrosis factor receptor gene knockout, reduced inflammation and retinal ganglion cell loss. Intraocular pressure elevation was not observed in experimental alkali burns. These findings illuminate the mechanism by which alkali burns cause retinal damage and may have importance in designing therapies for retinal protection.


Subject(s)
Burns, Chemical/metabolism , Eye Burns/metabolism , Retina/injuries , Alkalies , Animals , Apoptosis/drug effects , Apoptosis/physiology , Burns, Chemical/drug therapy , Burns, Chemical/etiology , Burns, Chemical/pathology , Cornea/immunology , Corneal Injuries/drug therapy , Corneal Injuries/etiology , Corneal Injuries/metabolism , Corneal Injuries/pathology , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Eye Burns/drug therapy , Eye Burns/etiology , Eye Burns/pathology , Hydrogen-Ion Concentration , Infliximab/pharmacology , Infliximab/therapeutic use , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidation-Reduction , Rabbits , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Retina/immunology , Retina/metabolism , Retina/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Sodium Hydroxide , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Uvea/metabolism , Uveitis, Anterior/chemically induced , Uveitis, Anterior/metabolism , Uveitis, Anterior/pathology , Uveitis, Anterior/prevention & control
16.
Mol Med Rep ; 15(4): 2261-2266, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28260033

ABSTRACT

The aim of the present study was to investigate differential modules (DMs) between uveal melanoma (UM) and normal conditions by examining differential networks. Based on a gene expression profile collected from the ArrayExpress database, the inference of DMs involved three steps: The first step was construction of a differential co­expression network (DCN); second, the module algorithm was adapted to identify the DMs presented in DCN; finally, the statistical significance of DMs were assessed based on the null score distribution of DMs generated using randomized networks. A DCN with 309 nodes and 3,729 edges was obtained, and 30 seed genes from the DCN were examined. Subsequently, one DM, which had 179 nodes and 3,068 edges, was investigated. By utilizing randomized networks, the P­value for DM was 0.034, therefore, the DM was statically significant between UM and baseline conditions. In conclusion, the present study successfully identified one DM in UM based on DCN and module algorithm, and this DM may be beneficial in revealing the pathological mechanism of UM and provide insight for future investigation of UM.


Subject(s)
Gene Regulatory Networks , Melanoma/genetics , Transcriptome , Uveal Neoplasms/genetics , Databases, Genetic , Gene Expression Regulation, Neoplastic , Humans , Melanoma/pathology , Uvea/metabolism , Uvea/pathology , Uveal Neoplasms/pathology
17.
Exp Eye Res ; 158: 94-111, 2017 05.
Article in English | MEDLINE | ID: mdl-26850315

ABSTRACT

Aqueous humor flows out of the eye primarily through the conventional outflow pathway that includes the trabecular meshwork and Schlemm's canal. However, a fraction of aqueous humor passes through an alternative or 'unconventional' route that includes the ciliary muscle, supraciliary and suprachoroidal spaces. From there, unconventional outflow may drain through two pathways: a uveoscleral pathway where aqueous drains across the sclera to be resorbed by orbital vessels, and a uveovortex pathway where aqueous humor enters the choroid to drain through the vortex veins. We review the anatomy, physiology and pharmacology of these pathways. We also discuss methods to determine unconventional outflow rate, including direct techniques that use radioactive or fluorescent tracers recovered from tissues in the unconventional pathway and indirect methods that estimate unconventional outflow based on total outflow over a range of pressures. Indirect methods are subject to a number of assumptions and generally give poor agreement with tracer measurements. We review the variety of animal models that have been used to study conventional and unconventional outflow. The mouse appears to be a promising model because it captures several aspects of conventional and unconventional outflow dynamics common to humans, although questions remain regarding the magnitude of unconventional outflow in mice. Finally, we review future directions. There is a clear need to develop improved methods for measuring unconventional outflow in both animals and humans.


Subject(s)
Aqueous Humor/metabolism , Limbus Corneae/metabolism , Sclera/metabolism , Trabecular Meshwork/metabolism , Uvea/metabolism , Animals , Humans , Intraocular Pressure , Models, Animal , Secretory Pathway
18.
Anticancer Agents Med Chem ; 17(2): 190-205, 2017.
Article in English | MEDLINE | ID: mdl-27109020

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Epigenesis, Genetic/drug effects , Immunotherapy/methods , Melanoma/genetics , Melanoma/therapy , Uvea/drug effects , Uveal Neoplasms/genetics , Uveal Neoplasms/therapy , Animals , Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunity/drug effects , Melanoma/immunology , Melanoma/pathology , Uvea/immunology , Uvea/metabolism , Uvea/pathology , Uveal Neoplasms/immunology , Uveal Neoplasms/pathology
19.
J Glaucoma ; 25(8): 681-91, 2016 08.
Article in English | MEDLINE | ID: mdl-27253969

ABSTRACT

AIM: We acquired age-matched and sex-matched Sprague-Dawley rats from 2 independent breeding establishments. Serendipitously, we observed that constitutive, and bacterial toxin-induced, expression of major histocompatibility complex (MHC) class II RT1B chain in the uveal tract was much lower in one of the cohorts. Activated microglia are known to upregulate MHC II RT1B expression during optic nerve (ON) degeneration induced by raised intraocular pressure (IOP). We investigated whether, in a model of experimental glaucoma, microglial upregulation of MHC II RT1B was less efficacious and ON degeneration correspondingly less severe in the cohort of rats with low MHC II RT1B expression. METHODS: Experimental glaucoma was induced by lasering the trabecular meshwork using a standard protocol. After 2 weeks of elevated IOP, retinal ganglion cells (RGC) survival, ON degeneration, and microglial responses were determined in both cohorts of rats. RESULTS: Raised IOP-induced expression of MHC II RT1B by microglia was muted in the "Low" cohort compared with the "High" cohort. Axonal degeneration, RGC loss, and microgliosis were all significantly lower in the cohort of rats with low basal and induced expression of MHC II RT1B, despite both cohorts displaying IOP responses that were indistinguishable in terms of peak IOP and IOP exposure. CONCLUSIONS: Expression of MHC II RT1B by activated microglia in the ON during experimental glaucoma was associated with more severe RGC degeneration. Further studies are needed to elucidate the role of MHC II during experimental glaucoma.


Subject(s)
Genes, MHC Class II , Glaucoma/metabolism , Microglia/metabolism , Nerve Degeneration/metabolism , Optic Nerve Diseases/metabolism , Animals , Disease Models, Animal , Glaucoma/pathology , Intraocular Pressure , Male , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/pathology , Trabecular Meshwork/metabolism , Uvea/metabolism
20.
Invest Ophthalmol Vis Sci ; 57(4): 1982-90, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27096756

ABSTRACT

PURPOSE: To study PTP4A3 phosphatase and MMP14 metalloprotease synergy in uveal melanoma aggressiveness. METHODS: Cell membrane localization of matrix metalloprotease 14 (MMP14) in uveal melanoma cells expressing protein tyrosine phosphatase A3 (PTP4A3) was assessed by flow cytometry or immunohistochemistry. The vesicular trafficking of MMP14 in the presence of PTP4A3 was evaluated in OCM-1 cells expressing either the wild-type or mutated phosphatase. Finally, MMP14 localization at the cell membrane of OCM-1 cells was impaired using RNA interference, and the PTP4A3-related migration in vitro and invasiveness in vivo of the treated cells were evaluated. RESULTS: We found that the membrane-anchored MMP14 is enriched at the cell surface of OCM-1 cells, patient-derived xenograft cells, and human primary uveal melanoma tumors expressing PTP4A3. Moreover, we show that PTP4A3 and MMP14 colocalize and that the vesicular trafficking of MMP14 is faster in the presence of active PTP4A3. Finally, we demonstrate that inhibition of MMP14 expression in uveal melanoma cells expressing PTP4A3 impairs their migration in vitro and invasiveness in vivo. CONCLUSIONS: Our observations indicate that PTP4A3 increases cell membrane accumulation of MMP14 as a result of increased cellular trafficking of the metalloprotease. We also show that downregulation of MMP14 expression reduced PTP4A3-induced cell migration and invasiveness. Taken together, our findings suggest that PTP4A3-related subcellular localization of MMP14 is an important event in metastasis induction.


Subject(s)
Cell Membrane/metabolism , Matrix Metalloproteinase 14/physiology , Melanoma/physiopathology , Neoplasm Proteins/physiology , Protein Tyrosine Phosphatases/physiology , Uveal Neoplasms/physiopathology , Cell Line, Tumor , Cell Movement/physiology , Flow Cytometry , Fluorescent Antibody Technique , Humans , Melanoma/metabolism , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , RNA Interference , Uvea/metabolism , Uvea/physiopathology , Uveal Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...