Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.034
Filter
1.
Cell Death Dis ; 15(10): 713, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39353898

ABSTRACT

Compelling evidence has revealed a novel function of the STAT pathway in the pathophysiology of uveal melanoma (UM); however, its regulatory mechanisms remain unclear. Here, we analyzed the clinical prognostic value of STAT family genes in UM patients using bioinformatics approaches and found that high STAT6 expression is associated with poor prognosis. Furthermore, cellular experiments and a nude mouse model demonstrated that STAT6 promotes UM progression through the autophagy pathway both in vivo and in vitro. Next, RIP-PCR revealed that STAT6 protein binds to LINC01637 mRNA, which in turn regulates STAT6 expression to promote UM growth. Finally, molecular docking indicated that STAT6 is a target of Zoledronic Acid, which can delay UM tumorigenicity by inhibiting STAT6 expression. Taken together, our results indicate that the STAT6/LINC01637 axis promotes UM progression via autophagy and may serve as a potential therapeutic target for UM.


Subject(s)
Autophagy , Cell Proliferation , Melanoma , Mice, Nude , STAT6 Transcription Factor , Uveal Neoplasms , Autophagy/drug effects , Humans , Uveal Neoplasms/pathology , Uveal Neoplasms/metabolism , Uveal Neoplasms/genetics , Uveal Neoplasms/drug therapy , Melanoma/pathology , Melanoma/metabolism , Melanoma/genetics , Melanoma/drug therapy , Animals , Cell Line, Tumor , Mice , STAT6 Transcription Factor/metabolism , STAT6 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Zoledronic Acid/pharmacology , Male , Female , Mice, Inbred BALB C , Signal Transduction
2.
Cell Death Dis ; 15(10): 720, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39353904

ABSTRACT

Nucleotide-binding oligomerization domain 2 (NOD2) is an immune sensor crucial for eliciting the innate immune responses. Nevertheless, discrepancies exist regarding the effect of NOD2 on different types of cancer. This study aimed to investigate these function of NOD2 in melanoma and its underlying mechanisms. We have validated the tumor suppressor effect of NOD2 in melanoma. NOD2 inhibited the proliferation of melanoma cells, hindering their migration and invasion while promoting the onset of apoptosis. Our study showed that NOD2 expression is closely related to pyrimidine and folate metabolism. NOD2 inhibits thymidylate synthase (TYMS) expression by promoting K48-type ubiquitination modification of TYMS, thereby decreasing the resistance of melanoma cells to 5-fluorouracil (5-FU) and capecitabine (CAP). TYMS was identified to form a complex with Polo-like Kinase 1 (PLK1) and activate the PLK1 signaling pathway. Furthermore, we revealed that the combination of the PLK1 inhibitor volasertib (BI6727) with 5-FU or CAP had a synergistic effect repressing the proliferation, migration, and autophagy of melanoma cells. Overall, our research highlights the protective role of NOD2 in melanoma and suggests that targeting NOD2 and the TYMS/PLK1 signaling axis is a high-profile therapy that could be a prospect for melanoma treatment.


Subject(s)
Cell Cycle Proteins , Cell Proliferation , Drug Resistance, Neoplasm , Melanoma , Nod2 Signaling Adaptor Protein , Polo-Like Kinase 1 , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Signal Transduction , Thymidylate Synthase , Humans , Protein Serine-Threonine Kinases/metabolism , Melanoma/drug therapy , Melanoma/pathology , Melanoma/metabolism , Melanoma/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Signal Transduction/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Thymidylate Synthase/metabolism , Thymidylate Synthase/genetics , Nod2 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Cell Proliferation/drug effects , Cell Movement/drug effects , Fluorouracil/pharmacology , Apoptosis/drug effects , Pteridines/pharmacology , Animals , Mice , Ubiquitination/drug effects , Autophagy/drug effects
3.
J Exp Clin Cancer Res ; 43(1): 276, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39354629

ABSTRACT

BACKGROUND: In order for cancers to progress, they must evade elimination by CD8 T cells or other immune mechanisms. CD8 T cells recognize and kill tumor cells that display immunogenic tumor peptides bound to MHC I molecules. One of the ways that cancers can escape such killing is by reducing expression of MHC I molecules, and loss of MHC I is frequently observed in tumors. There are multiple different mechanisms that can underly the loss of MHC I complexes on tumor and it is currently unclear whether there are particular mechanisms that occur frequently and, if so, in what types of cancers. Also of importance to know is whether the loss of MHC I is reversible and how such loss and/or its restoration would impact responses to immunotherapy. Here, we investigate these issues for loss of IRF1 and IRF2, which are transcription factors that drive expression of MHC I pathway genes and some killing mechanisms. METHODS: Bioinformatics analyses of IRF2 and IRF2-dependent gene transcripts were performed for all human cancers in the TCGA RNAseq database. IRF2 protein-DNA-binding was analyzed in ChIPseq databases. CRISRPcas9 was used to knock out IRF1 and IRF2 genes in human and mouse melanoma cells and the resulting phenotypes were analyzed in vitro and in vivo. RESULTS: Transcriptomic analysis revealed that IRF2 expression was reduced in a substantial subset of cases in almost all types of human cancers. When this occurred there was a corresponding reduction in the expression of IRF2-regulated genes that were needed for CD8 T cell recognition. To test cause and effect for these IRF2 correlations and the consequences of IRF2 loss, we gene-edited IRF2 in a patient-derived melanoma and a mouse melanoma. The IRF2 gene-edited melanomas had reduced expression of transcripts for genes in the MHC I pathway and decreased levels of MHC I complexes on the cell surface. Levels of Caspase 7, an IRF2 target gene involved in CD8 T cell killing of tumors, were also reduced. This loss of IRF2 caused both human and mouse melanomas to become resistant to immunotherapy with a checkpoint inhibitor. Importantly, these effects were reversible. Stimulation of the IRF2-deficient melanomas with interferon induced the expression of a functionally homologous transcription factor, IRF1, which then restored the MHC I pathway and responsiveness to CPI. CONCLUSIONS: Our study shows that a subset of cases within most types of cancers downregulates IRF2 and that this can allow cancers to escape immune control. This can cause resistance to checkpoint blockade immunotherapy and is reversible with currently available biologics.


Subject(s)
Immunotherapy , Interferon Regulatory Factor-2 , Melanoma , Animals , Humans , Mice , Interferon Regulatory Factor-2/genetics , Interferon Regulatory Factor-2/metabolism , Melanoma/genetics , Melanoma/immunology , Melanoma/drug therapy , Melanoma/therapy , Immunotherapy/methods , Melanoma, Experimental/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Cell Line, Tumor
4.
Front Immunol ; 15: 1451103, 2024.
Article in English | MEDLINE | ID: mdl-39355255

ABSTRACT

Background: Immunotherapy has revolutionized skin cutaneous melanoma treatment, but response variability due to tumor heterogeneity necessitates robust biomarkers for predicting immunotherapy response. Methods: We used weighted gene co-expression network analysis (WGCNA), consensus clustering, and 10 machine learning algorithms to develop the immunotherapy-related gene model (ITRGM) signature. Multi-omics analyses included bulk and single-cell RNA sequencing of melanoma patients, mouse bulk RNA sequencing, and pathology sections of melanoma patients. Results: We identified 66 consensus immunotherapy prognostic genes (CITPGs) using WGCNA and differentially expressed genes (DEGs) from two melanoma cohorts. The CITPG-high group showed better prognosis and enriched immune activities. DEGs between CITPG-high and CITPG-low groups in the TCGA-SKCM cohort were analyzed in three additional melanoma cohorts using univariate Cox regression, resulting in 44 consensus genes. Using 101 machine learning algorithm combinations, we constructed the ITRGM signature based on seven model genes. The ITRGM outperformed 37 published signatures in predicting immunotherapy prognosis across the training cohort, three testing cohorts, and a meta-cohort. It effectively stratified patients into high-risk or low-risk groups for immunotherapy response. The low-risk group, with high levels of model genes, correlated with increased immune characteristics such as tumor mutation burden and immune cell infiltration, indicating immune-hot tumors with a better prognosis. The ITRGM's relationship with the tumor immune microenvironment was further validated in our experiments using pathology sections with GBP5, an important model gene, and CD8 IHC analysis. The ITRGM also predicted better immunotherapy response in eight cohorts, including urothelial carcinoma and stomach adenocarcinoma, indicating broad applicability. Conclusions: The ITRGM signature is a stable and robust predictor for stratifying melanoma patients into 'immune-hot' and 'immune-cold' tumors, enhancing prognosis and response to immunotherapy.


Subject(s)
Biomarkers, Tumor , Immunotherapy , Machine Learning , Melanoma , Humans , Melanoma/therapy , Melanoma/immunology , Melanoma/genetics , Immunotherapy/methods , Biomarkers, Tumor/genetics , Prognosis , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Skin Neoplasms/genetics , Animals , Gene Expression Profiling , Transcriptome , Gene Expression Regulation, Neoplastic , Mice , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Treatment Outcome , Gene Regulatory Networks
5.
Mol Biol (Mosk) ; 58(2): 189-203, 2024.
Article in Russian | MEDLINE | ID: mdl-39355878

ABSTRACT

Uveal melanoma (UM) is a neuroectodermal tumor that results from malignant transformation of melanocytes in the eye uvea, including the iris, the ciliary body, and the choroid. UM accounts for 5% of all melanoma cases and is extremely aggressive with half of the UM patients developing metastases within the first 1-2 years after tumor development. Molecular mechanisms of UM carcinogenesis are poorly understood, but are known to differ from those of skin melanoma. Activating mutations of the GNAQ and GNA11 genes, which code for the large G protein subunits Gq and G11, respectively, are found in 90% of UM patients. The Gaq/PKC/MAPK signaling pathway is a main signaling cascade that leads to the transformation of melanocytes of the uveal tract, and major regulators of the cascade provide targets for the development of drugs. Metastatic UM (MUM) is most often associated with mutations of BAP1, EIF1AX, GNA11, GNAQ, and SF3B1. A combination of a commercial expression test panel of 15 genes and a mutation panel of 7 genes, supplemented with data on the size of the primary tumor, is highly efficient in predicting the risk of metastasis. The risk of metastasis determines the choice of therapy and the patient follow-up regimen. However, no systemic therapy for MUM has been developed to date. New drugs undergoing clinical trials are mostly targeted drugs designed to inhibit the protein products of mutant genes or immunotherapeutic agents designed to stimulate the immune response against specific antigens. In addition to these approaches, potential therapeutic targets of epigenetic regulation of UM development are considered in the review.


Subject(s)
Melanoma , Mutation , Uveal Neoplasms , Humans , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Uveal Neoplasms/metabolism , Uveal Neoplasms/drug therapy , Uveal Neoplasms/therapy , Melanoma/genetics , Melanoma/pathology , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/therapy , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Signal Transduction/drug effects , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects
6.
Mol Biol (Mosk) ; 58(2): 295-304, 2024.
Article in Russian | MEDLINE | ID: mdl-39355886

ABSTRACT

Multiple exogenous or endogenous factors alter gene expression patterns by different mechanisms that are poorly understood. We used RNA-Seq analysis in order to study changes in gene expression in melanoma cells that are capable of vasculogenic mimicry that is inhibited upon the action of an inhibitor of vasculogenic mimicry. Here, we show that the drug induces a strong upregulation of 50 genes that control the cell cycle and microtubule cytoskeleton coupled with a strong downregulation of 50 genes that control different cellular metabolic processes. We found that both groups of genes are simultaneously regulated by multiple sets of transcription factors. We conclude that one way for coordinated regulation of large groups of genes is regulation simultaneously by multiple transcription factors.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma , Humans , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Melanoma/drug therapy , Cell Line, Tumor , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/biosynthesis , Cell Cycle/drug effects
7.
Cancer Res ; 84(17): 2792-2805, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39228255

ABSTRACT

Neoantigen-based immunotherapy is an attractive potential treatment for previously intractable tumors. To effectively broaden the application of this approach, stringent biomarkers are crucial to identify responsive patients. ARID1A, a frequently mutated subunit of SWI/SNF chromatin remodeling complex, has been reported to determine tumor immunogenicity in some cohorts; however, mutations and deletions of ARID1A are not always linked to clinical responses to immunotherapy. In this study, we investigated immunotherapeutic responses based on ARID1A status in targeted therapy-resistant cancers. Mouse and human BRAFV600E melanomas with or without ARID1A expression were transformed into resistant to vemurafenib, an FDA-approved specific BRAFV600E inhibitor. Anti-PD-1 antibody treatment enhanced antitumor immune responses in vemurafenib-resistant ARID1A-deficient tumors but not in ARID1A-intact tumors or vemurafenib-sensitive ARID1A-deficient tumors. Neoantigens derived from accumulated somatic mutations during vemurafenib resistance were highly expressed in ARID1A-deficient tumors and promoted tumor immunogenicity. Furthermore, the newly generated neoantigens could be utilized as immunotherapeutic targets by vaccines. Finally, targeted therapy resistance-specific neoantigen in experimental human melanoma cells lacking ARID1A were validated to elicit T-cell receptor responses. Collectively, the classification of ARID1A-mutated tumors based on vemurafenib resistance as an additional indicator of immunotherapy response will enable a more accurate prediction to guide cancer treatment. Furthermore, the neoantigens that emerge with therapy resistance can be promising therapeutic targets for refractory tumors. Significance: Chemotherapy resistance promotes the acquisition of immunogenic neoantigens in ARID1A-deficient tumors that confer sensitivity to immune checkpoint blockade and can be utilized for developing antitumor vaccines, providing strategies to improve immunotherapy efficacy.


Subject(s)
Antigens, Neoplasm , DNA-Binding Proteins , Drug Resistance, Neoplasm , Melanoma , Transcription Factors , Vemurafenib , Animals , Humans , Transcription Factors/genetics , Transcription Factors/immunology , Mice , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Drug Resistance, Neoplasm/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/genetics , Vemurafenib/pharmacology , Vemurafenib/therapeutic use , Melanoma/immunology , Melanoma/drug therapy , Melanoma/genetics , Melanoma/therapy , Immunotherapy/methods , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/immunology , Cell Line, Tumor , Female , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mutation , Molecular Targeted Therapy/methods , Mice, Inbred C57BL
8.
Biol Res ; 57(1): 59, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39223638

ABSTRACT

BACKGROUND: Tumour dormancy, a resistance mechanism employed by cancer cells, is a significant challenge in cancer treatment, contributing to minimal residual disease (MRD) and potential relapse. Despite its clinical importance, the mechanisms underlying tumour dormancy and MRD remain unclear. In this study, we employed two syngeneic murine models of myeloid leukemia and melanoma to investigate the genetic, epigenetic, transcriptomic and protein signatures associated with tumour dormancy. We used a multiomics approach to elucidate the molecular mechanisms driving MRD and identify potential therapeutic targets. RESULTS: We conducted an in-depth omics analysis encompassing whole-exome sequencing (WES), copy number variation (CNV) analysis, chromatin immunoprecipitation followed by sequencing (ChIP-seq), transcriptome and proteome investigations. WES analysis revealed a modest overlap of gene mutations between melanoma and leukemia dormancy models, with a significant number of mutated genes found exclusively in dormant cells. These exclusive genetic signatures suggest selective pressure during MRD, potentially conferring resistance to the microenvironment or therapies. CNV, histone marks and transcriptomic gene expression signatures combined with Gene Ontology (GO) enrichment analysis highlighted the potential functional roles of the mutated genes, providing insights into the pathways associated with MRD. In addition, we compared "murine MRD genes" profiles to the corresponding human disease through public datasets and highlighted common features according to disease progression. Proteomic analysis combined with multi-omics genetic investigations, revealed a dysregulated proteins signature in dormant cells with minimal genetic mechanism involvement. Pathway enrichment analysis revealed the metabolic, differentiation and cytoskeletal remodeling processes involved in MRD. Finally, we identified 11 common proteins differentially expressed in dormant cells from both pathologies. CONCLUSIONS: Our study underscores the complexity of tumour dormancy, implicating both genetic and nongenetic factors. By comparing genomic, transcriptomic, proteomic, and epigenomic datasets, our study provides a comprehensive understanding of the molecular landscape of minimal residual disease. These results provide a robust foundation for forthcoming investigations and offer potential avenues for the advancement of targeted MRD therapies in leukemia and melanoma patients, emphasizing the importance of considering both genetic and nongenetic factors in treatment strategies.


Subject(s)
Disease Models, Animal , Melanoma , Neoplasm, Residual , Animals , Melanoma/genetics , Melanoma/pathology , Mice , Leukemia/genetics , Leukemia/pathology , DNA Copy Number Variations , Exome Sequencing , Mice, Inbred C57BL , Proteomics , Transcriptome , Gene Expression Profiling , Multiomics
9.
J Drugs Dermatol ; 23(9): 717-723, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39231084

ABSTRACT

BACKGROUND: Numerous melanoma-specific dermoscopic features have been described in invasive melanomas, while fewer features are found in melanoma in situ (MIS) and atypical nevi (ATN). Consensus regarding which features are critical for the differentiation of MIS from ATN has not been reached. PURPOSE: Determine 1) whether there are dermoscopic features that differentiate early MIS from ATN, and 2) whether non-invasive assessment of genomic biomarkers (LINC00518 and PRAME) can aid in patient management. METHODS: From 2018 to 2023, 56 melanomas were evaluated for 5 clinical and 13 dermoscopic features and melanoma-associated genomic biomarkers. Two groups of ATN with positive and negative genomic biomarkers were randomly selected for comparison. RESULTS: All melanomas in this study expressed one or both melanoma-associated genomic markers. MIS had an average of 3.90 (range, 2-7) of the 13 dermoscopic features, while invasive melanomas had an average of 4.44 (range, 3-6). Sixteen of 40 (40%) MIS and 3 of 16 (18.8%) invasive melanomas had 3 or fewer dermoscopic features. These findings were comparable to those observed in both ATN groups. The most common dermoscopic features were absent or diminished pigment network, regression structures, and granularity. This combination of features was most helpful in identifying lesions for genomic testing. CONCLUSIONS: Clinical and dermoscopic features alone could not differentiate MIS from ATN. Non-invasive genomic testing helped differentiate lower from higher-risk lesions and aid in clinical management decisions. Genomic testing was particularly helpful in patients with large numbers of lesions with several being considered for biopsy based on clinical and dermoscopic examination. J Drugs Dermatol. 2024;23(9):717-723. doi:10.36849/JDD.8454.


Subject(s)
Dermoscopy , Melanoma , Skin Neoplasms , Humans , Skin Neoplasms/pathology , Skin Neoplasms/genetics , Skin Neoplasms/diagnosis , Melanoma/genetics , Melanoma/pathology , Melanoma/diagnosis , Female , Male , Middle Aged , Diagnosis, Differential , Aged , Adult , Genomics , Biomarkers, Tumor/genetics , Nevus, Pigmented/genetics , Nevus, Pigmented/diagnosis , Nevus, Pigmented/pathology , Aged, 80 and over
10.
Medicine (Baltimore) ; 103(36): e39385, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39252325

ABSTRACT

Uveal melanoma (UM) is a common health challenge worldwide as a prevalent intraocular malignancy because of its high mortality rate. However, clinical workers do not have an accurate prognostic tool now. Immune function is closely related to tumor development. Interestingly, researchers have identified that long noncoding RNAs (lncRNAs) are tightly associated with biological processes at the cellular level, particularly their involvements in immune response and its regulation of the growth of tumor cells. Hence, lncRNAs may be involved in the progression of uveal melanoma. UM patients' RNA expression matrices were extracted from TCGA database. The targeted immune genes were filtered by weighted correlation network analysis and the immune-related lncRNAs with a high prognostic relevance were obtained by Cox regression analysis and least absolute shrinkage and selection operator regression analysis. Each sample was scored according to those lncRNA expression and divided into high-risk and low-risk group. We confirmed the sensitivity and independence of our risk model compared to the tumor mutation burden score. Finally, we demonstrated the clinical relevance of our model by examining its sensitivity to different drugs. The risk score based on our risk model was significantly independent of other clinical parameters in either univariate (hazard ratio = 109.852 [15.738-766.749], P value < .001) or multivariate (hazard ratio = 114.075 [15.207-855.735], P value < .001) analyses. The ROC curves of this model imply high predictive accuracy for 1-year, 3-year, and 5-year survival (1-year area under the curve [AUC] = 0.849, 3-years AUC = 0.848, and 5-years AUC = 0.761). Our study revealed that immune-related lncRNAs are significant in the clinical diagnosis, treatment and prognosis of UM patients. We successfully constructed a lncRNA-based prognostic risk model which may serve as a future reference for the diagnosis and prognosis of UM. Based on this model we also validated the sensitivity of some cancer drugs, which has implications for the future immunotherapy and drug development.


Subject(s)
Melanoma , RNA, Long Noncoding , Uveal Neoplasms , Uveal Neoplasms/genetics , Uveal Neoplasms/mortality , Uveal Neoplasms/immunology , Humans , Melanoma/genetics , Melanoma/mortality , Melanoma/immunology , RNA, Long Noncoding/genetics , Prognosis , Male , Female , Risk Assessment/methods , Biomarkers, Tumor/genetics , Middle Aged , Proportional Hazards Models
11.
Cell Mol Biol Lett ; 29(1): 117, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237877

ABSTRACT

BACKGROUND: PD-L1 expression on cancer cells is an important mechanism of tumor immune escape, and immunotherapy targeting the PD-L1/PD1 interaction is a common treatment option for patients with melanoma. However, many patients do not respond to treatment and novel predictors of response are emerging. One suggested modifier of PD-L1 is the p53 pathway, although the relationship of p53 pathway function and activation is poorly understood. METHODS: The study was performed on human melanoma cell lines with various p53 status. We investigated PD-L1 and proteins involved in IFNγ signaling by immunoblotting and mRNA expression, as well as membrane expression of PD-L1 by flow cytometry. We evaluated differences in the ability of NK cells to recognize and kill target tumor cells on the basis of p53 status. We also investigated the influence of proteasomal degradation and protein half-life, IFNγ signaling and p53 activation on biological outcomes, and performed bioinformatic analysis using available data for melanoma cell lines and melanoma patients. RESULTS: We demonstrate that p53 status changes the level of membrane and total PD-L1 protein through IRF1 regulation and show that p53 loss influences the recently discovered SOX10/IRF1 regulatory axis. Bioinformatic analysis identified a dependency of SOX10 on p53 status in melanoma, and a co-regulation of immune signaling by both transcription factors. However, IRF1/PD-L1 regulation by p53 activation revealed complicated regulatory mechanisms that alter IRF1 mRNA but not protein levels. IFNγ activation revealed no dramatic differences based on TP53 status, although dual p53 activation and IFNγ treatment confirmed a complex regulatory loop between p53 and the IRF1/PD-L1 axis. CONCLUSIONS: We show that p53 loss influences the level of PD-L1 through IRF1 and SOX10 in an isogenic melanoma cell model, and that p53 loss affects NK-cell cytotoxicity toward tumor cells. Moreover, activation of p53 by MDM2 inhibition has a complex effect on IRF1/PD-L1 activation. These findings indicate that evaluation of p53 status in patients with melanoma will be important for predicting the response to PD-L1 monotherapy and/or dual treatments where p53 pathways participate in the overall response.


Subject(s)
B7-H1 Antigen , Interferon Regulatory Factor-1 , Melanoma , SOXE Transcription Factors , Signal Transduction , Tumor Suppressor Protein p53 , Humans , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Melanoma/genetics , Melanoma/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , SOXE Transcription Factors/metabolism , SOXE Transcription Factors/genetics , Interferon-gamma/metabolism , Interferon-gamma/genetics , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Gene Expression Regulation, Neoplastic
12.
Cancer Med ; 13(17): e70199, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39240165

ABSTRACT

BACKGROUND: Most primary cutaneous melanomas have pathogenesis driven by ultraviolet exposure and genetic mutations, whereas acral lentiginous melanoma (ALM) and metastatic melanoma are much less, if at all, linked with the former. Thus, we evaluated both ultraviolet related and non-ultraviolet related melanomas. Mutations in the MUC16 and TTN genes commonly occur concurrently in these melanoma patients, but their combined prognostic significance stratified by gender and cancer subtype remains unclear. METHODS: The cBioPortal database was queried for melanoma studies and returned 16 independent studies. Data from 2447 melanoma patients were utilized including those with ALM, cutaneous melanoma (CM), and melanoma of unknown primary (MUP). Patients were grouped based on the presence or absence of MUC16 and TTN mutations. Univariate Cox regression and Student's t-tests were used to analyze hazard ratios and total mutation count comparisons, respectively. RESULTS: TTN mutations, either alone or concurrently with MUC16 mutations, significantly correlated with worse prognosis overall, in both genders, and in CM patients. ALM patients with both mutations had better prognoses than CM patients, while ALM patients with neither mutation had worse prognosis than CM patients. For MUP patients, only MUC16 mutations correlated with worse prognosis. ALM patients with neither MUC16 nor TTN mutations had significantly more total mutations than MUP patients, followed by CM patients. CONCLUSION: TTN mutations are a potential marker of poor prognosis in melanoma, which is amplified in the presence of concurrent MUC16 mutations. ALM patients with neither gene mutations had worse prognosis, suggesting a protective effect of having both MUC16 and TTN mutations. Only MUC16 mutations conferred a worse prognosis for MUP patients. Comprehensive genetic profiling in melanoma patients may facilitate personalized treatment strategies to optimize patient outcomes.


Subject(s)
Connectin , Melanoma , Mutation , Skin Neoplasms , Humans , Melanoma/genetics , Melanoma/mortality , Melanoma/pathology , Female , Male , Prognosis , Retrospective Studies , Middle Aged , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Connectin/genetics , CA-125 Antigen , Sex Factors , Membrane Proteins/genetics , Aged , Biomarkers, Tumor/genetics , Adult
13.
Sci Adv ; 10(36): eadn9361, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39231216

ABSTRACT

Kv1.3 is a multifunctional potassium channel implicated in multiple pathologies, including cancer. However, how it is involved in disease progression is not fully clear. We interrogated the interactome of Kv1.3 in intact cells using BioID proximity labeling, revealing that Kv1.3 interacts with STAT3- and p53-linked pathways. To prove the relevance of Kv1.3 and of its interactome in the context of tumorigenesis, we generated stable melanoma clones, in which ablation of Kv1.3 remodeled gene expression, reduced proliferation and colony formation, yielded fourfold smaller tumors, and decreased metastasis in vivo in comparison to WT cells. Kv1.3 deletion or pharmacological inhibition of mitochondrial Kv1.3 increased mitochondrial Reactive Oxygen Species release, decreased STAT3 phosphorylation, stabilized the p53 tumor suppressor, promoted metabolic switch, and altered the expression of several BioID-identified Kv1.3-networking proteins in tumor tissues. Collectively, our work revealed the tumor-promoting Kv1.3-interactome landscape, thus opening the way to target Kv1.3 not only as an ion-conducting entity but also as a signaling hub.


Subject(s)
Kv1.3 Potassium Channel , STAT3 Transcription Factor , Signal Transduction , Tumor Suppressor Protein p53 , Kv1.3 Potassium Channel/metabolism , Kv1.3 Potassium Channel/genetics , Tumor Suppressor Protein p53/metabolism , STAT3 Transcription Factor/metabolism , Humans , Animals , Mice , Cell Line, Tumor , Melanoma/metabolism , Melanoma/pathology , Melanoma/genetics , Mitochondria/metabolism , Cell Proliferation , Reactive Oxygen Species/metabolism
14.
BMC Bioinformatics ; 25(1): 300, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271985

ABSTRACT

BACKGROUND: Overall Survival (OS) and Progression-Free Interval (PFI) as survival times have been collected in The Cancer Genome Atlas (TCGA). It is of biomedical interest to consider their dependence in pathway detection and survival prediction. We intend to develop novel methods for integrating PFI as condition based on parametric survival models for identifying pathways associated with OS and predicting OS. RESULTS: Based on the framework of conditional probability, we developed a family of frailty-based parametric-models for this purpose, with exponential or Weibull distribution as baseline. We also considered two classes of existing methods with PFI as a covariate. We evaluated the performance of three approaches by analyzing RNA-seq expression data from TCGA for lung squamous cell carcinoma and lung adenocarcinoma (LUNG), brain lower grade glioma and glioblastoma multiforme (GBMLGG), as well as skin cutaneous melanoma (SKCM). Our focus was on fourteen general cancer-related pathways. The 10-fold cross-validation was employed for the evaluation of predictive accuracy. For LUNG, p53 signaling and cell cycle pathways were detected by all approaches. Furthermore, three approaches with the consideration of PFI demonstrated significantly better predictive performance compared to the approaches without the consideration of PFI. For GBMLGG, ten pathways (e.g., Wnt signaling, JAK-STAT signaling, ECM-receptor interaction, etc.) were detected by all approaches. Furthermore, three approaches with the consideration of PFI demonstrated better predictive performance compared to the approaches without the consideration of PFI. For SKCM, p53 signaling pathway was detected only by our Weibull-baseline-based model. And three approaches with the consideration of PFI demonstrated significantly better predictive performance compared to the approaches without the consideration of PFI. CONCLUSIONS: Based on our study, it is necessary to incorporate PFI into the survival analysis of OS. Furthermore, PFI is a survival-type time, and improved results can be achieved by our conditional-probability-based approach.


Subject(s)
RNA-Seq , Humans , RNA-Seq/methods , Survival Analysis , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/metabolism , Melanoma/genetics , Melanoma/mortality , Melanoma/metabolism
15.
Cells ; 13(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273022

ABSTRACT

Melanoma is the most severe type of skin cancer and among the most malignant neoplasms in humans. With the growing incidence of melanoma, increased numbers of therapeutic options, and the potential to target specific proteins, understanding the basic mechanisms underlying the disease's progression and resistance to treatment has never been more important. LOXL3, SNAI1, and NES are key factors in melanoma genesis, regulating tumor growth, metastasis, and cellular differentiation. In our study, we explored the potential role of LOXL3, SNAI1, and NES in melanoma progression and metastasis among patients with dysplastic nevi, melanoma in situ, and BRAF+ and BRAF- metastatic melanoma, using immunofluorescence and qPCR analysis. Our results reveal a significant increase in LOXL3 expression and the highest NES expression in BRAF+ melanoma compared to BRAF-, dysplastic nevi, and melanoma in situ. As for SNAI1, the highest expression was observed in the metastatic melanoma group, without significant differences among groups. We found co-expression of LOXL3 and SNAI1 in the perinuclear area of all investigated subgroups and NES and SNAI1 co-expression in melanoma cells. These findings suggest a codependence or collaboration between these markers in melanoma EMT, suggesting new potential therapeutic interventions to block the EMT cascade that could significantly affect survival in many melanoma patients.


Subject(s)
Disease Progression , Melanoma , Snail Family Transcription Factors , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Humans , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Cell Line, Tumor , Male , Female , Neoplasm Metastasis , Middle Aged
16.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273363

ABSTRACT

MDM4 is upregulated in the majority of melanoma cases and has been described as a "key therapeutic target in cutaneous melanoma". Numerous isoforms of MDM4 exist, with few studies examining their specific expression in human tissues. The changes in splicing of MDM4 during human melanomagenesis are critical to p53 activity and represent potential therapeutic targets. Compounding this, studies relying on short reads lose "connectivity" data, so full transcripts are frequently only inferred from the presence of splice junction reads. To address this problem, long-read nanopore sequencing was utilized to read the entire length of transcripts. Here, MDM4 transcripts, both alternative and canonical, are characterized in a pilot cohort of human melanoma specimens. RT-PCR was first used to identify the presence of novel splice junctions in these specimens. RT-qPCR then quantified the expression of major MDM4 isoforms observed during sequencing. The current study both identifies and quantifies MDM4 isoforms present in melanoma tumor samples. In the current study, we observed high expression levels of MDM4-S, MDM4-FL, MDM4-A, and the previously undescribed Ensembl transcript MDM4-209. A novel transcript lacking both exons 6 and 9 is observed and named MDM4-A/S for its resemblance to both MDM4-A and MDM4-S isoforms.


Subject(s)
Melanoma , Protein Isoforms , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Alternative Splicing , Gene Expression Regulation, Neoplastic , Nanopore Sequencing/methods
17.
Int J Mol Sci ; 25(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273574

ABSTRACT

Primary intracranial melanoma is a very rare brain tumor, especially when accompanied by benign intramedullary melanocytoma. Distinguishing between a primary central nervous system (CNS) lesion and metastatic melanoma is extremely difficult, especially when the primary cutaneous lesion is not visible. Here we report a 13-year-old girl admitted to the Neurosurgery Department of the Institute of Polish Mother's Health Centre in Lodz due to upper limb paresis. An intramedullary tumor of the cervical C3-C4 and an accompanying syringomyelic cavity C1-C7 were revealed. The child underwent partial removal of the tumor due to the risk of damage to spinal cord motor centers. The removed part of the tumor was diagnosed as melanocytoma. Eight months later, a neurological examination revealed paresis of the right sixth cranial nerve, accompanied by bilateral optic disc edema. Diagnostic imaging revealed a brain tumor. The girl underwent resection of both detected the tumors and an additional satellite lesion revealed during the surgery. The removed tumors were diagnosed as malignant melanomas in pathomorphological examination. Molecular analysis revealed NRASQ61K mutation in both the intracranial and the intramedullary tumor. It should be noted that in cases where available evidence is inconclusive, an integrative diagnostic process is essential to reach a definitive diagnosis.


Subject(s)
Melanoma , Humans , Female , Adolescent , Melanoma/genetics , Melanoma/pathology , Melanoma/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/diagnosis , Membrane Proteins/genetics , Spinal Cord Neoplasms/genetics , Spinal Cord Neoplasms/pathology , Spinal Cord Neoplasms/diagnosis , Spinal Cord Neoplasms/surgery , Mutation , GTP Phosphohydrolases
18.
Hum Genomics ; 18(1): 96, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39244622

ABSTRACT

BACKGROUND: Precancerous and malignant tumours arise within the oral cavity from a predisposed "field" of epithelial cells upon exposure to carcinogenic stimulus. This phenomenon is known as "Field Cancerization". The molecular genomic and transcriptomic alterations that lead to field cancerization and tumour progression is unknown in Indian Oral squamous cell carcinoma (OSCC) patients. METHODS: We have performed whole exome sequencing, copy-number variation array and whole transcriptome sequencing from five tumours and dysplastic lesions (sampled from distinct anatomical subsites - one each from buccal anterior and posterior alveolus, dorsum of tongue-mucosal melanoma, lip and left buccal mucosa) and blood from a rare OSCC patient with field cancerization. RESULTS: A missense CASP8 gene mutation (p.S375F) was observed to be the initiating event in oral tumour field development. APOBEC mutation signatures, arm-level copy number alterations, depletion of CD8 + T cells and activated NK cells and enrichment of pro-inflammatory mast cells were features of early-originating tumours. Pharmacological inhibition of CASP8 protein in a CASP8-wild type OSCC cell line showed enhanced levels of cellular migration and viability. CONCLUSION: CASP8 alterations are the earliest driving events in oral field carcinogenesis, whereas additional somatic mutational, copy number and transcriptomic alterations ultimately lead to OSCC tumour formation and progression.


Subject(s)
Caspase 8 , DNA Copy Number Variations , Melanoma , Mouth Neoplasms , Transcriptome , Humans , Caspase 8/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Melanoma/genetics , Melanoma/pathology , Transcriptome/genetics , DNA Copy Number Variations/genetics , Exome Sequencing , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Male , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Mutation, Missense/genetics , Female , Middle Aged , CD8-Positive T-Lymphocytes
19.
Cancer Immunol Immunother ; 73(11): 218, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39235510

ABSTRACT

Epigenetic modifications to DNA and chromatin control oncogenic and tumor-suppressive mechanisms in melanoma. Ezh2, the catalytic component of the Polycomb Repressive Complex 2 (PRC2), which mediates methylation of lysine 27 on histone 3 (H3K27me3), can regulate both melanoma initiation and progression. We previously found that mutant Ezh2Y641F interacts with the immune regulator Stat3 and together they affect anti-tumor immunity. However, given the numerous downstream targets and pathways affected by Ezh2, many mechanisms that determine its oncogenic activity remain largely unexplored. Using genetically engineered mouse models, we further investigated the role of pathways downstream of Ezh2 in melanoma carcinogenesis and identified significant enrichment in several autophagy signatures, along with increased expression of autophagy regulators, such as Atg7. In this study, we investigated the effect of Atg7 on melanoma growth and tumor immunity within the context of a wild-type or Ezh2Y641F epigenetic state. We found that the Atg7 locus is controlled by multiple Ezh2 and Stat3 binding sites, Atg7 expression is dependent on Stat3 expression, and that deletion of Atg7 slows down melanoma cell growth in vivo, but not in vitro. Atg7 deletion also results in increased CD8 + T cells in Ezh2Y641F melanomas and reduced myelosuppressive cell infiltration in the tumor microenvironment, particularly in Ezh2WT melanomas, suggesting a strong immune system contribution in the role of Atg7 in melanoma progression. These findings highlight the complex interplay between genetic mutations, epigenetic regulators, and autophagy in shaping tumor immunity in melanoma.


Subject(s)
Autophagy-Related Protein 7 , Enhancer of Zeste Homolog 2 Protein , STAT3 Transcription Factor , Animals , STAT3 Transcription Factor/metabolism , Mice , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Tumor Microenvironment/immunology , Mice, Inbred C57BL , Gene Expression Regulation, Neoplastic , Melanoma/immunology , Melanoma/metabolism , Melanoma/genetics , Melanoma/pathology , Epigenesis, Genetic , Cell Line, Tumor , Humans , Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism
20.
Sci Signal ; 17(854): eadp3967, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39288219

ABSTRACT

In contrast with sun exposure-induced melanoma, rarer melanocytic tumors and neoplasms with low mutational burden present opportunities to study isolated signaling mechanisms. These include uveal melanoma and blue nevi, which are often driven by mutations within the G protein-coupled signaling cascade downstream of cysteinyl leukotriene receptor 2. Here, we review how the same mutations within this pathway drive the growth of melanocytes in one tissue but can inhibit the growth of those in another, exemplifying the role of the tissue environment in the delicate balance between uncontrolled cell growth and senescence.


Subject(s)
Cell Proliferation , Cellular Senescence , Melanocytes , Receptors, Leukotriene , Signal Transduction , Receptors, Leukotriene/metabolism , Receptors, Leukotriene/genetics , Humans , Melanocytes/metabolism , Melanocytes/cytology , Melanocytes/pathology , Melanoma/metabolism , Melanoma/genetics , Melanoma/pathology , Animals , Mutation , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Uveal Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL