Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
J Biol Chem ; 299(12): 105418, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37923138

ABSTRACT

Most uveal melanoma cases harbor activating mutations in either GNAQ or GNA11. Despite activation of the mitogen-activated protein kinase (MAPK) signaling pathway downstream of Gαq/11, there are no effective targeted kinase therapies for metastatic uveal melanoma. The human genome encodes numerous understudied kinases, also called the "dark kinome". Identifying additional kinases regulated by Gαq/11 may uncover novel therapeutic targets for uveal melanoma. In this study, we treated GNAQ-mutant uveal melanoma cell lines with a Gαq/11 inhibitor, YM-254890, and conducted a kinase signaling proteomic screen using multiplexed-kinase inhibitors followed by mass spectrometry. We observed downregulated expression and/or activity of 22 kinases. A custom siRNA screen targeting these kinases demonstrated that knockdown of microtubule affinity regulating kinase 3 (MARK3) and serine/threonine kinase 10 (STK10) significantly reduced uveal melanoma cell growth and decreased expression of cell cycle proteins. Additionally, knockdown of MARK3 but not STK10 decreased ERK1/2 phosphorylation. Analysis of RNA-sequencing and proteomic data showed that Gαq signaling regulates STK10 expression and MARK3 activity. Our findings suggest an involvement of STK10 and MARK3 in the Gαq/11 oncogenic pathway and prompt further investigation into the specific roles and targeting potential of these kinases in uveal melanoma.


Subject(s)
Melanoma , Protein Serine-Threonine Kinases , Uveal Neoplasms , Humans , Cell Line, Tumor , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteomics , Uveal Neoplasms/drug therapy , Uveal Neoplasms/enzymology , Uveal Neoplasms/genetics
2.
Pigment Cell Melanoma Res ; 35(1): 78-87, 2022 01.
Article in English | MEDLINE | ID: mdl-34347929

ABSTRACT

Metastatic uveal melanoma (UM) responds poorly to targeted therapies and immune checkpoint inhibitors. Loss of BRCA1-associated protein 1 (BAP1) via inactivating mutations in the BAP1 gene is associated with UM progression. Thus, molecular alterations caused by BAP1 dysfunction may be novel therapeutic targets for metastatic UM. Here, we found that phosphorylation of AMP-dependent kinase (AMPK) was elevated in BAP1-altered (or mutant) compared to BAP1-unaltered (or wild-type [WT]) UM tumors. As a readout of AMPK pathway activation, phosphorylation of an AMPK downstream effector, acetyl-CoA-carboxylase (ACC), was also elevated. BAP1 re-expression in BAP1-null UM cell lines decreased phospho-AMPK (pAMPK) and phospho-ACC (pACC) levels. AMPK phosphorylation is mediated by calcium/calmodulin dependent protein kinase kinase 2 (CaMKK2) and potentially liver kinase B1 (LKB1) in BAP1 mutant UM cells. Knockdown of AMPKα1/2 reduced the viability of BAP1 mutant UM cells, indicating a survival function of AMPK in BAP1 mutant UM. Our data suggest that the AMPK pathway is an important mechanism mediating the survival of BAP1 mutant UM. Targeting the AMPK pathway may be a novel therapeutic strategy for metastatic UM.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Melanoma/enzymology , Mutation , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Uveal Neoplasms/enzymology , AMP-Activated Protein Kinases/genetics , Acetyl-CoA Carboxylase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Line, Tumor , Cell Survival , Enzyme Activation , Humans , Melanoma/genetics , Melanoma/pathology , Phosphorylation , Signal Transduction , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology
3.
BMC Cancer ; 21(1): 1068, 2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34587931

ABSTRACT

BACKGROUND: Uveal melanoma (UVM) is the leading cause of eye-related mortality worldwide. This study aimed to explore the expression and prognostic value of matrix metalloproteinases (MMPs) in UVM. METHODS: Gene expression levels were obtained from the Gene Expression Omnibus (GEO) and Oncomine databases. Functional and pathway enrichment analyses were performed using the Metascape database. GeneMANIA was then applied to construct a protein-protein interaction network and identify the hub genes. Moreover, overall survival (OS) and disease-free survival (DFS) analysis for the hub genes was performed using the UALCAN and Gene Expression Profiling Interactive Analysis (GEPIA) online tool. Furthermore, TRRUST was used to predict the targets of the MMPs. RESULTS: Our results revealed that the transcriptional levels of MMP1, MMP9, MMP10, MMP11, MMP13, MMP14, and MMP17 were upregulated in UVM tissues compared to normal tissues. A protein-protein interaction (PPI) network was constructed and the top 50 hub genes were identified. The functions of MMPs and their neighboring proteins are mainly associated with ECM-receptor interaction, proteoglycans in cancer, the IL-17 signaling pathway, and microRNAs in cancer. Among the MMPs, MMP1/2/9/11/14/15/16/17/24 played significant roles in the progression of UVM from stage 3 to stage 4. We also found that the expression of MMP1, MMP2, MMP9, and MMP16 positively correlated with OS and DFS in patients with UVM. Additionally, 18 transcription factors associated with nine MMPs were identified. CONCLUSIONS: The results of this study may provide potential biomarkers and targets for UVM. However, further studies are required to confirm these results.


Subject(s)
Biomarkers, Tumor/metabolism , Collagenases/metabolism , Melanoma/enzymology , Protein Interaction Maps/genetics , Uveal Neoplasms/enzymology , Biomarkers, Tumor/genetics , Collagenases/genetics , Databases, Genetic , Disease Progression , Disease-Free Survival , Gene Expression Profiling/methods , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Melanoma/genetics , Melanoma/mortality , Melanoma/pathology , Prognosis , Transcription Factors/metabolism , Up-Regulation , Uvea/enzymology , Uveal Neoplasms/genetics , Uveal Neoplasms/mortality , Uveal Neoplasms/pathology
4.
Nanomedicine ; 35: 102391, 2021 07.
Article in English | MEDLINE | ID: mdl-33794371

ABSTRACT

Uveal melanoma (UM) is an intraocular tumor which is almost lethal at the metastatic stage due to the lack of effective treatments. In this regard, we have developed an albumin-based nanostructure (ABN) containing AZD8055 (ABN-AZD), which is a potent mTOR kinase inhibitor, for its efficient delivery to the tumors. The drug has been conjugated to ABN using tailored linkers that have a disulfide moiety, allowing its release selectively and effectively in the presence of an elevated concentration of glutathione, such as inside the tumoral cells. Our therapeutic approach induced significant cellular toxicity in uveal melanoma cells, but not in non-tumoral keratinocytes, highlighting the excellent selectivity of the system. In addition, these nanostructures showed excellent activity in vivo, decreasing the tumor surface compared to the free AZD8055 in mice models. Remarkably, the results obtained were achieved employing a dose 23 times lower than those used in previous reports.


Subject(s)
Melanoma/drug therapy , Morpholines , Nanostructures , Serum Albumin, Human , Uveal Neoplasms/drug therapy , Animals , Feeder Cells , Humans , Melanoma/enzymology , Mice , Mice, Nude , Morpholines/chemistry , Morpholines/pharmacology , Nanostructures/chemistry , Nanostructures/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Serum Albumin, Human/chemistry , Serum Albumin, Human/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Uveal Neoplasms/enzymology , Xenograft Model Antitumor Assays
5.
Biomed Pharmacother ; 133: 111006, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33202284

ABSTRACT

The crosstalk between Notch and MAPK pathway plays a role in MEK inhibitor resistance in BRAFV600E metastatic melanoma (MM) and promotes migration in GNAQQ209L uveal melanoma (UM) cells. We determined the cytotoxicity of combinatorial inhibition of MEK and Notch by cobimetinib and γ-secretase inhibitor (GSI) nirogacestat, in BRAFV600E and BRAF wt MM and GNAQQ209L UM cells displaying different Erk1/2 and Notch activation status, with the aim to elucidate the impact of Notch signaling in the response to MEK inhibitor. Overall the combination was synergic in BRAFV600E MM and GNAQQ209L UM cells and antagonistic in BRAF wt one. Focusing on UM cells, we found that cobimetinib resulted in G0/G1 phase arrest and apoptosis induction, whereas the combination with GSI increased treatment efficacy by inducing a senescent-like state of cells and by blocking migration towards liver cancer cells. Mechanistically, this was reflected in a strong reduction of cyclin D1, in the inactivation of retinoblastoma protein and in the increase of p27KIP1 expression levels. Of note, each drug alone prevented Notch signaling activation resulting in inhibition of c-jun(Ser63) and Hes-1 expression. The combination achieved the strongest inhibition on Notch signaling and on both c-jun(Ser63) and Erk1/2 activation level. In conclusion we unveiled a coordinate action of MAPK and Notch signaling in promoting proliferation of BRAFV600E MM and GNAQQ209L UM cells. Remarkably, the simultaneous inhibition of MEK and Notch signaling highlighted a role for the second pathway in protecting cells against senescence in GNAQQ209L UM cells treated with the MEK inhibitor.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azetidines/pharmacology , Melanoma/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Quinolines/pharmacology , Receptors, Notch/antagonists & inhibitors , Skin Neoplasms/drug therapy , Uveal Neoplasms/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Movement/drug effects , Cellular Senescence/drug effects , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Hep G2 Cells , Humans , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Invasiveness , Phosphorylation , Proto-Oncogene Proteins B-raf/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Uveal Neoplasms/enzymology , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology
6.
Int J Mol Sci ; 21(15)2020 Jul 23.
Article in English | MEDLINE | ID: mdl-32718045

ABSTRACT

Melanoma of the ocular region (ocular melanoma) comprises about 5% of all patients with melanoma and covers posterior uveal melanoma, iris melanoma, and conjunctival melanoma. The risk of metastasis is much higher in patients with ocular melanoma compared to a primary melanoma of the skin. The subtypes of ocular melanoma have distinct genetic features, which should be taken into consideration when making clinical decisions. Most relevant for current practice is the absence of BRAF mutations in posterior uveal melanoma, although present in some iris melanomas and conjunctival melanomas. In this review, we discuss the genetic biomarkers of the subtypes of ocular melanoma and their impacts on the clinical care of these patients.


Subject(s)
Melanoma , Mutation , Proto-Oncogene Proteins B-raf , Uveal Neoplasms , Humans , Melanoma/enzymology , Melanoma/genetics , Melanoma/therapy , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Uveal Neoplasms/enzymology , Uveal Neoplasms/genetics , Uveal Neoplasms/therapy
7.
Pigment Cell Melanoma Res ; 33(3): 507-514, 2020 05.
Article in English | MEDLINE | ID: mdl-31758842

ABSTRACT

MEK inhibitors (MEKi) demonstrate anti-proliferative activity in patients with metastatic uveal melanoma, but responses are short-lived. In the present study, we evaluated the MEKi trametinib alone and in combination with drugs targeting epigenetic regulators, including DOT1L, EZH2, LSD1, DNA methyltransferases, and histone acetyltransferases. The DNA methyltransferase inhibitor (DNMTi) decitabine effectively enhanced the anti-proliferative activity of trametinib in cell viability, colony formation, and 3D organoid assays. RNA-Seq analysis showed the MEKi-DNMTi combination primarily affected the expression of genes involved in G1 and G2/2M checkpoints, cell survival, chromosome segregation and mitotic spindle. The DNMTi-MEKi combination did not appear to induce a DNA damage response (as measured by γH2AX foci) or senescence (as measured by ß-galactosidase staining) compared to either MEKi or DNMTi alone. Instead, the combination increased expression of the CDK inhibitor p21 and the pro-apoptotic protein BIM. In vivo, the DNMTi-MEKi combination was more effective at suppressing growth of MP41 uveal melanoma xenografts than either drug alone. Our studies indicate that DNMTi may enhance the activity of MEKi in uveal melanoma.


Subject(s)
Decitabine/therapeutic use , Melanoma/drug therapy , Melanoma/enzymology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Uveal Neoplasms/drug therapy , Uveal Neoplasms/enzymology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Decitabine/pharmacology , Female , Humans , Melanoma/pathology , Mice , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Uveal Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Biosci Biotechnol Biochem ; 84(3): 471-480, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31694485

ABSTRACT

This study aimed to explore the influence of Tryptophanyl-tRNA synthetase (WARS) expression on the proliferation and migration of uveal melanoma (UM) cells, and the potential mechanisms. Bioinformatics analysis based on Gene Expression Omnibus (GEO) database showed that WARS expression in metastatic cancer was significantly higher than that in no-metastatic group. Kaplan-Meier analysis based on The Cancer Genome Atlas (TCGA) database showed that high WARS expression was associated with lower survival. Biological function experiments showed that overexpression of WARS in OCM-1A cells can promote cell proliferation, migration, and invasion, whereas knockdown of WARS in C918 cells showed the opposite effect. Finally, we observed that the up-regulation of WARS induced the activation of phosphatidylinositol 3-kinase/AKT (PI3K/AKT) signaling, whilst depletion of WARS resulted in opponent outcomes. Taken together, our results illustrated that WARS was overexpressed in UM cells and contributed to the viability and motility of UM cells via modulating PI3K/AKT signaling pathway.


Subject(s)
Melanoma/metabolism , Tryptophan-tRNA Ligase/metabolism , Uveal Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Melanoma/enzymology , Melanoma/pathology , Up-Regulation , Uveal Neoplasms/enzymology , Uveal Neoplasms/pathology
9.
Invest Ophthalmol Vis Sci ; 60(7): 2474-2480, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31173078

ABSTRACT

Purpose: The activation of the mitogen-activated protein kinase (MAPK) pathway has been suggested as the major downstream target when GNAQ and GNA11 (GNAQ/11) are mutated in uveal melanoma (UM). However, clinical trials with single agent MEK inhibitor showed no clinical significance in altering the overall outcome of the disease in UM; therefore, we investigated the correlation between naturally occurring mutations in GNAQ/11 and activation of MAPK pathway in vivo in primary UM. Methods: Screening for activating mutations in codons 183 and 209 of GNAQ/11 was carried out by sequencing and restriction fragment length polymorphism (RFLP) in a cohort of 42 primary UM. Activation of the MAPK pathway and other potential downstream signals was assessed by immunohistochemistry and/or Western blot analysis. Potential downstream signaling of mutant and wild type GNAQ/11 was studied by transient transfection assay in nonmutant cell lines. Results: Somatic mutations in GNAQ/11 were observed in 35/42 (83.3%) of primary UM. Tumors with GNAQ/11 mutations showed variations in the activation of ERK1/2 with significant tumor heterogeneity. Weak and undetectable ERK1/2 activation was observed in 4/35 (11.4%) and 8/35 (22.9%) of the GNAQ/11 mutant UM, respectively. Tumor heterogeneity of GNAQ/11 mutations was also observed in a subset of tumors. Conclusions: Our results indicate that there is marked variation in MAPK activation in UM with GNAQ/11 mutations. Thus, GNAQ/11 mutational status is not a sufficient biomarker to adequately predict UM patient responses to single-agent selective MEK inhibitor therapy.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits/genetics , Melanoma/enzymology , Melanoma/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , Uveal Neoplasms/enzymology , Uveal Neoplasms/genetics , Blotting, Western , Cell Line, Tumor , Cohort Studies , DNA Mutational Analysis , Humans , Immunohistochemistry , Plasmids , Polymorphism, Restriction Fragment Length , Sequence Analysis, DNA , Signal Transduction , Transfection
10.
Sci Signal ; 12(573)2019 03 19.
Article in English | MEDLINE | ID: mdl-30890659

ABSTRACT

Somatic gain-of-function mutations of GNAQ and GNA11, which encode α subunits of heterotrimeric Gαq/11 proteins, occur in about 85% of cases of uveal melanoma (UM), the most common cancer of the adult eye. Molecular therapies to directly target these oncoproteins are lacking, and current treatment options rely on radiation, surgery, or inhibition of effector molecules downstream of these G proteins. A hallmark feature of oncogenic Gαq/11 proteins is their reduced intrinsic rate of hydrolysis of guanosine triphosphate (GTP), which results in their accumulation in the GTP-bound, active state. Here, we report that the cyclic depsipeptide FR900359 (FR) directly interacted with GTPase-deficient Gαq/11 proteins and preferentially inhibited mitogenic ERK signaling rather than canonical phospholipase Cß (PLCß) signaling driven by these oncogenes. Thereby, FR suppressed the proliferation of melanoma cells in culture and inhibited the growth of Gαq-driven UM mouse xenografts in vivo. In contrast, FR did not affect tumor growth when xenografts carried mutated B-RafV600E as the oncogenic driver. Because FR enabled suppression of malignant traits in cancer cells that are driven by activating mutations at codon 209 in Gαq/11 proteins, we envision that similar approaches could be taken to blunt the signaling of non-Gαq/11 G proteins.


Subject(s)
Depsipeptides/pharmacology , Drug Delivery Systems , GTP-Binding Protein alpha Subunits, Gq-G11 , GTP-Binding Protein alpha Subunits , Gain of Function Mutation , Melanoma , Neoplasm Proteins , Uveal Neoplasms , Animals , Cell Line, Tumor , Depsipeptides/chemistry , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Uveal Neoplasms/drug therapy , Uveal Neoplasms/enzymology , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Xenograft Model Antitumor Assays
11.
Cancer Res ; 79(5): 899-904, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30622115

ABSTRACT

Chimeric antigen receptors (CAR) can transmit signals akin to those from activated T-cell receptors when bound to a cell surface target. CAR-expressing T cells against CD19 can cause curative effects in leukemia and lymphoma and is approved for clinical use. However, no CAR-T therapy is currently approved for use in solid tumors. We hypothesize that the resistance of solid tumors to CAR-T can be overcome by similar means as those used to reactivate tumor-infiltrating T lymphocytes (TIL), for example, by cytokines or immune checkpoint blockade. Here we demonstrate that CAR-T cells directed against HER2 can kill uveal and cutaneous melanoma cells in vitro and in vivo. Curative effects in vivo were only observed in xenografts grown in a NOD/SCID IL2 receptor gamma (NOG) knockout mouse strain transgenic for human IL2. The effect was target-specific, as CRISPR/Cas9-mediated disruption of HER2 in the melanoma cells abrogated the killing effect of the CAR-T cells. The CAR-T cells were also able to kill melanoma cells from patients resistant to adoptive T-cell transfer (ACT) of autologous TILs. Thus, CAR-T therapy represents an option for patients that do not respond to immunotherapy with ACT of TIL or immune checkpoint blockade. In addition, our data highlight the use of IL2 transgenic NOG mice as models to prove efficacy of CAR-T-cell products, possibly even in a personalized manner. SIGNIFICANCE: These findings demonstrate that a novel humanized mouse model can help clinical translation of CAR-T cells against uveal and cutaneous melanoma that do not respond to TIL therapy or immune checkpoint blockade.


Subject(s)
Immunotherapy, Adoptive/methods , Melanoma/therapy , Receptor, ErbB-2/immunology , T-Lymphocytes/transplantation , Uveal Neoplasms/therapy , Animals , Cell Line, Tumor , Humans , Interleukin Receptor Common gamma Subunit/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , Melanoma/enzymology , Melanoma/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptor, ErbB-2/metabolism , Skin Neoplasms/enzymology , Skin Neoplasms/immunology , Skin Neoplasms/therapy , T-Lymphocytes/immunology , Uveal Neoplasms/enzymology , Uveal Neoplasms/immunology , Xenograft Model Antitumor Assays , Melanoma, Cutaneous Malignant
12.
ACS Chem Biol ; 14(1): 132-136, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30525429

ABSTRACT

In uveal melanoma (UM) cells, the protein kinase C (pathway) is almost generally constitutively activated as a result of an activating mutation in either the GNAQ or the GNA11 G-protein. A pan-PKC inhibitor, sotrastaurin (also named AEB071), is in clinical trials for treatment of UM patients with limited success and eliciting adverse effects. Interestingly, genetic interference with expression of just one PKC isoform, e.g., PKCδ, is sufficient to reduce UM cell proliferation. Therefore, we tested the effect of a recently described specific PKCδ inhibitor, B106, on growth and survival of UM cell lines. Surprisingly, we found that B106 efficiently induced apoptosis in several cell lines, but apparently independent of activated PKCδ.


Subject(s)
Carbazoles/pharmacology , Chromans/pharmacology , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Uveal Neoplasms/pathology , Enzyme Activation , Humans , Protein Kinase C-delta/metabolism , Uveal Neoplasms/enzymology
13.
Eur Rev Med Pharmacol Sci ; 22(19): 6230-6238, 2018 10.
Article in English | MEDLINE | ID: mdl-30338785

ABSTRACT

OBJECTIVE: Thrombospondin 2 (THBS2) expression and its prognostic value have been documented in several types of cancer. Nevertheless, the potential role and clinical significance of THBS2 in uveal melanoma (UM) have never been reported. Thus, in our study, we aimed to explore the clinical significance and prognostic impact of THBS2 in UM. MATERIALS AND METHODS: Survival and prognosis analyses were implemented using the Kaplan-Meier method and COX's proportional hazards model based on the clinical data retrieved from The Cancer Genome Atlas (TCGA) database. Colony formation, cell proliferation, invasion and migration assays in M23 cell line were performed to evaluate the effects of THBS2 on UM in vitro. To further reveal whether the dysregulated THBS2 expression regulates the UM metastasis, protein biomarkers including serine-threonine kinase (AKT), p-AKT, phosphoinositide 3-kinase (PI3K), p-PI3K, and p70S6K were measured using Western blotting analysis. RESULTS: THBS2 was up-regulated in metastatic UMs. Relationship of THBS2 expression level with the clinicopathological factors demonstrated that the expression level of THBS2 was significantly correlated to histological type, recurrence, and dead. Univariate as well as multivariate COX analyses demonstrated that THBS2 could serve as an independent prognostic factor for overall survival of UM. The knockdown of THBS2 significantly inhibited the proliferation rate of M23 cells, suppressed the colony numbers of M23 cells, lowered the invasive and migratory cell proportion. Importantly, Western blotting results implicated that THBS2 knockdown significantly decreased the expression level of p-AKT, p-PI3K, and p70S6K in M23 cell line. CONCLUSIONS: This is the first study to report that THBS2 may play important roles in UM progression and might be a novel prognosis biomarker for UM.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement , Melanoma/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Thrombospondins/metabolism , Uveal Neoplasms/enzymology , Biomarkers, Tumor/genetics , Cell Line, Tumor , Databases, Genetic , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Melanoma/genetics , Melanoma/mortality , Melanoma/secondary , Middle Aged , Phosphorylation , Prognosis , Signal Transduction , Thrombospondins/genetics , Uveal Neoplasms/genetics , Uveal Neoplasms/mortality , Uveal Neoplasms/pathology
14.
Anticancer Res ; 38(7): 3817-3824, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29970501

ABSTRACT

Uveal melanoma is the most common intraocular malignancy in adults, representing approximately 3% of all melanoma cases. Despite progress in chemotherapy, radiation and surgical treatment options, the prognosis and survival rates remain poor. Acetylation of histone proteins causes transcription of genes involved in cell growth, DNA replication and progression of cell cycle. Overexpression of histone deacetylases occurs in a wide spectrum of malignancies. Histone deacetylase inhibitors block the action of histone deacetylases, leading to inhibition of tumor cell proliferation. This article reviewed the potential therapeutic effects of histone deacetylase inhibitors on uveal melanoma. MEDLINE database was used under the key words/phrases: histone deacetylase, inhibitors, uveal melanoma and targeted therapies for uveal melanoma. A total of 47, English articles, not only referring to uveal melanoma, published up to February 2018 were used. Valproic acid, trichostatin A, tenovin-6, depsipeptide, panobinostat (LBH-589), vorinostat (suberanilohydroxamic acid) entinostat (MS-275), quisinostat, NaB, JSL-1, MC1568 and MC1575 are histone deacetylase inhibitors that have demonstrated promising antitumor effects against uveal melanoma. Histone deacetylase inhibitors represent a promising therapeutic approach for the treatment of uveal melanoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Melanoma/drug therapy , Uveal Neoplasms/drug therapy , Histone Deacetylases/metabolism , Humans , Melanoma/enzymology , Melanoma/pathology , Neoplasm Metastasis , Uveal Neoplasms/enzymology , Uveal Neoplasms/pathology
15.
Biochem Biophys Res Commun ; 501(4): 885-892, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29763606

ABSTRACT

Hexokinase-2 (HK2), the initial as well as the rate-limiting step in glycolysis, is overexpressed in many human cancers, and correlates with poor clinical outcomes. Aerobic glycolysis is a hallmark of cancer, and drugs targeting its enzymes, including HK2, are being developed. However, the mechanisms of HK2 inhibition and the physiological significance of the HK2 inhibitors in cancer cells are rarely reported. Here, we show that microRNA-216a-5p (miR-216a-5p) inhibits HK2 expression by directly targeting its 3'-UTR in uveal melanoma cells. Through inhibition of HK2, miR-216a-5p dampens glycolysis by reducing HK activity, glucose uptake, lactate production, ATP generation, extracellular acidification rate (ECAR), and increasing oxygen consumption rate (OCR) in uveal melanoma cells. Importantly, glycolysis regulated by miR-216a-5p is critical for its regulating uveal melanoma tumor growth both in vitro and in vivo. miR-216a-5p expression is negatively correlated with HK2 expression and predicts better outcome in uveal melanoma patients. Our findings provide clues regarding the role of miR-216a-5p as a tumor suppressor in uveal melanoma through the inhibition of HK2. Targeting HK2 through miR-216a-5p could be a promising therapeutic strategy in uveal melanoma.


Subject(s)
Glycolysis , Hexokinase/metabolism , Melanoma/genetics , Melanoma/pathology , MicroRNAs/metabolism , Signal Transduction , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Aerobiosis , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Hexokinase/genetics , Humans , Melanoma/enzymology , Mice , MicroRNAs/genetics , Prognosis , Uveal Neoplasms/enzymology
16.
Am J Vet Res ; 79(1): 90-97, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29287164

ABSTRACT

OBJECTIVE To characterize the distribution and intensity of cyclooxygenase (COX)-2 expression in the eyes of cats with and without uveitis and to determine whether COX-2 expression is correlated with severity of inflammation. SAMPLES Archived ocular tissue specimens from 51 cats with and 10 cats without ocular disease. PROCEDURES Specimens from only 1 eye were evaluated for each cat. Specimens were stained with H&E stain or immunohistochemical stain for detection of COX-2 and reviewed. For each eye, the type, severity, and distribution of inflammation and the distribution and intensity of COX-2 expression were determined for the uvea and other ocular tissues. Correlation between COX-2 expression and inflammation severity was also assessed. RESULTS COX-2 was not expressed in any nondiseased eye. Of the 51 diseased eyes, 20 had histologic evidence of lymphocytic-plasmacytic uveitis, 13 had neutrophilic uveitis, 11 had diffuse iris melanoma with uveitis, and 7 had diffuse iris melanoma without uveitis. Of the 44 eyes with uveitis, COX-2 was detected in the uvea of 16, including 11 eyes with lymphocytic-plasmacytic uveitis, 4 with neutrophilic uveitis, and 1 with diffuse iris melanoma-induced uveitis. Inflammation was severe, moderate, or mild in 10, 5, and 1 of those eyes, respectively. Cyclooxygenase-2 was detected in the cornea of 21 eyes with uveitis and 1 eye with diffuse iris melanoma without uveitis. Uveitis severity was positively correlated with COX-2 expression in both the uvea and cornea. CONCLUSIONS AND CLINICAL RELEVANCE Results suggested that COX-2 is an inflammatory mediator in feline uveitis but not diffuse iris melanoma.


Subject(s)
Cyclooxygenase 2/biosynthesis , Eye/enzymology , Uveitis/veterinary , Animals , Cats , Cornea/enzymology , Cyclooxygenase 2/metabolism , Female , Male , Melanoma/enzymology , Melanoma/veterinary , Uvea/enzymology , Uveal Neoplasms/enzymology , Uveal Neoplasms/veterinary , Uveitis/enzymology
17.
Cancer Cell ; 31(5): 685-696.e6, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28486107

ABSTRACT

Constitutive activation of Gαq signaling by mutations in GNAQ or GNA11 occurs in over 80% of uveal melanomas (UMs) and activates MAPK. Protein kinase C (PKC) has been implicated as a link, but the mechanistic details remained unclear. We identified PKC δ and ɛ as required and sufficient to activate MAPK in GNAQ mutant melanomas. MAPK activation depends on Ras and is caused by RasGRP3, which is significantly and selectively overexpressed in response to GNAQ/11 mutation in UM. RasGRP3 activation occurs via PKC δ- and ɛ-dependent phosphorylation and PKC-independent, DAG-mediated membrane recruitment, possibly explaining the limited effect of PKC inhibitors to durably suppress MAPK in UM. The findings nominate RasGRP3 as a therapeutic target for cancers driven by oncogenic GNAQ/11.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Guanine Nucleotide Exchange Factors/metabolism , MAP Kinase Signaling System , Melanoma/enzymology , Mutation , Uveal Neoplasms/enzymology , Animals , Cell Line, Tumor , Cell Proliferation , Diglycerides/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Guanine Nucleotide Exchange Factors/genetics , Humans , MAP Kinase Signaling System/drug effects , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Mice, Nude , Phosphorylation , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference , Time Factors , Transfection , Tumor Burden , Uveal Neoplasms/drug therapy , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , ras Guanine Nucleotide Exchange Factors
18.
Cancer Lett ; 400: 47-60, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28455241

ABSTRACT

Uveal melanoma (UM) is the most common intraocular malignant neoplasm in adults. Despite the availability of enucleation, radiation and chemotherapy, the prognosis of patients with metastasis remains poor. Therefore, novel effective therapies for patients with metastatic UM are urgently needed. In the present study, we demonstrated that JSL-1, a novel HDAC inhibitor, effectively inhibited the proliferation. JSL-1 induced apoptosis with increased expression of proapoptotic BH3-only protein BIM in UM cells. JSL-1 suppressed migration and invasion of UM cells with MMP-2 decreased. Furthermore, JSL-1 blocked the canonical Wnt/ß-catenin pathway, impaired self-renewal capacity and decreased percentage of ALDH+ cells, thereby reflecting elimination of UM cancer stem-like cells (CSCs) which are believed seeds of metastasis. Importantly, JSL-1 potently inhibited the growth of uveal melanoma xenograft in NOD-SCID mice. These results suggested that JSL-1 may be a promising therapeutic agent for UM.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Depsipeptides/pharmacology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Melanoma/drug therapy , Neoplastic Stem Cells/drug effects , Uveal Neoplasms/drug therapy , Aldehyde Dehydrogenase/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Cell Line, Tumor , Cell Self Renewal/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Humans , Male , Matrix Metalloproteinase 2/metabolism , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Neoplasm Invasiveness , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , RNA Interference , Time Factors , Transfection , Tumor Burden/drug effects , Uveal Neoplasms/enzymology , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Vinblastine/pharmacology , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays
19.
Hum Pathol ; 60: 137-146, 2017 02.
Article in English | MEDLINE | ID: mdl-27825810

ABSTRACT

Previous studies have found a link between high expression levels of the Deleted in Split hand/Split foot 1 (DSS1) gene and cancer progression. The aim of this study was to examine whether overexpression of DSS1 is a feature of melanoma and squamous cell carcinoma (SCC) and if any epigenetic modifications are involved. Evaluation of DSS1 expression profile indicated that the gene is overexpressed in 112 of 130 cutaneous melanomas (86.1%), 41 of 64 uveal melanomas (64.1%), 67 of 82 mucosal melanomas (81.7%), and 61 of 75 SCC samples (81.3%), relative to normal skin. An inverse correlation between DSS1 expression and methylation status of the promoter was found. In vitro studies showed that treatment of DSS1-methylated melanoma and SCC cells with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine significantly increased DSS1 expression at mRNA and protein levels. Interestingly, a significant association between high DSS1 expression levels and some clinicopathological variables, such as metastasis, ulceration, and reduced overall/disease-free survival was observed. In summary, these data suggest that the extent of promoter methylation plays a role in modulating DSS1 gene expression and highlight that promoter hypomethylation is a frequent event in melanoma and SCC closely linked to poor prognosis.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , DNA Methylation , Epigenesis, Genetic , Melanoma/genetics , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/genetics , Skin Neoplasms/genetics , Uveal Neoplasms/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , Cell Line, Tumor , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Decitabine , Disease-Free Survival , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Melanoma/enzymology , Melanoma/pathology , Melanoma/surgery , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Skin Neoplasms/surgery , Time Factors , Treatment Outcome , Up-Regulation , Uveal Neoplasms/enzymology , Uveal Neoplasms/pathology , Uveal Neoplasms/surgery
20.
Biomed Pharmacother ; 84: 1538-1550, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27881235

ABSTRACT

Uveal melanoma (UM) is the most common primary intraocular malignant tumor of adults. It has high mortality rate due to liver metastasis. However, the epidemiology and pathogenesis of liver metastasis in UM are not elucidated and there is no effective therapy available for preventing the development of this disease. IGF-1 is a growth factor involved in cell proliferation, malignant transformation and inhibition of apoptosis. In previous report, IGF-1 receptor was found to be highly expressed in UM and this was related to tumor prognosis. FoxO3a is a Forkhead box O (FOXO) transcription factor and a downstream target of the IGF-1R/PI3K/Akt pathway involved in a number of physiological and pathological processes including cancer. However, the role of FoxO3a in UM is unknown. In the present study, we investigated fundamental mechanisms in the growth, migration and invasion of UM and the involvement of FoxO3a. IGF-1 increased the cell viability, invasion, migration and S-G2/M cell cycle phase accumulation of UM cells. Western blot analysis showed that IGF-1 led to activation of Akt and concomitant phosphorylation of FoxO3a. FoxO3a phosphorylation was associated with its translocation into the cytoplasm from the nucleus and its functional inhibition led to the inhibition of expression of Bim and p27, but an increase in the expression of Cyclin D1. The effects of IGF-1 on UM cells were reversed by LY294002 (a PI3K inhibitor) or Akt siRNA, and the overexpression of FoxO3a also attenuated basal invasion and migration of UM. Taken all together, these results suggest that inhibition of FoxO3a by IGF-1 via the PI3K/Akt pathway has an important role in IGF-1 induced proliferation and invasion of UM cells. These findings also support FoxO3a and IGF signaling may represent a valid target for investigating the development of new strategies for the treatment and prevention of the pathology of UM.


Subject(s)
Cell Movement/drug effects , Forkhead Box Protein O3/metabolism , Insulin-Like Growth Factor I/pharmacology , Melanoma/pathology , Uveal Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Chromones/pharmacology , Flavonoids/pharmacology , Forkhead Box Protein O3/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , Melanoma/enzymology , Melanoma/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Morpholines/pharmacology , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Uveal Neoplasms/enzymology , Uveal Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL