Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40.657
Filter
1.
Invest Ophthalmol Vis Sci ; 65(8): 3, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38953846

ABSTRACT

Purpose: To investigate the correlation between apparent diffusion coefficient (ADC) histograms and high-risk clinicopathologic features related to uveal melanoma (UM) prognosis. Methods: This retrospective study included 53 patients with UM who underwent diffusion-weighted imaging (DWI) between August 2015 and March 2024. Axial DWI was performed with a single-shot spin-echo echo-planar imaging sequence. ADC histogram parameters of ADCmean, ADC50%, interquartile range (IQR), skewness, kurtosis, and entropy were obtained from DWI. The relationships between histogram parameters and high-risk clinicopathological characteristics including tumor size, preoperative retinal detachment, histological subtypes, Ki-67 index, and chromosome status, were analyzed by Spearman correlation analysis, Mann-Whitney U test, or Kruskal-Wallis test. Results: A total of 53 patients (mean ± SD age, 55 ± 15 years; 22 men) were evaluated. The largest basal diameter (LBD) was correlated with kurtosis (r = 0.311, P = 0.024). Tumor prominence (TP) was correlated with entropy (r = 0.581, P < 0.001) and kurtosis (r = 0.273, P = 0.048). Additionally, significant correlations were identified between the Ki-67 index and ADCmean (r = -0.444, P = 0.005), ADC50% (r = -0.487, P = 0.002), and skewness (r = 0.394, P = 0.014). Finally, entropy was correlated with monosomy 3 (r = 0.541, P = 0.017). Conclusions: The ADC histograms provided valuable insights into high-risk clinicopathologic features of UM and hold promise in the early prediction of UM prognosis.


Subject(s)
Diffusion Magnetic Resonance Imaging , Melanoma , Uveal Neoplasms , Humans , Uveal Neoplasms/pathology , Uveal Neoplasms/genetics , Male , Female , Middle Aged , Melanoma/pathology , Retrospective Studies , Prognosis , Diffusion Magnetic Resonance Imaging/methods , Adult , Aged , Echo-Planar Imaging/methods
2.
Invest Ophthalmol Vis Sci ; 65(8): 11, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38967943

ABSTRACT

Purpose: Ocular melanoma is a common primary malignant ocular tumor in adults with limited effective treatments. Epigenetic regulation plays an important role in tumor development. The switching/sucrose nonfermentation (SWI/SNF) chromatin remodeling complex and bromodomain and extraterminal domain family proteins are epigenetic regulators involved in several cancers. We aimed to screen a candidate small molecule inhibitor targeting these regulators and investigate its effect and mechanism in ocular melanoma. Methods: We observed phenotypes caused by knockdown of the corresponding gene and synergistic effects with BRD inhibitor treatment and SWI/SNF complex knockdown. The effect of JQ-1 on ocular melanoma cell cycle and apoptosis was analyzed with flow cytometry. Via RNA sequencing, we also explored the mechanism of BRD4. Results: The best tumor inhibitory effect was observed for the BRD4 inhibitor (JQ-1), although there were no statistically obvious changes in the shBRD4 and shBRD9 groups. Interestingly, the inhibitory effect of JQ-1 was decrease in the shBRD4 group. JQ-1 inhibits the growth of melanoma in various cell lines and in tumor-bearing mice. We found 17 of these 28 common differentially expressed genes were downregulated after MEL270 and MEL290 cells treated with JQ-1. Four of these 17 genes, TP53I11, SH2D5, SEMA5A, and MDGA1, were positively correlated with BRD4. In TCGA database, low expression of TP53I11, SH2D5, SEMA5A, and MDGA1 improved the overall survival rate of patients. Furthermore, the disease-free survival rate was increased in the groups with low expression of TP53I11, SH2D5, and SEMA5A. Conclusions: JQ-1 may act downstream of BRD4 and suppress ocular melanoma growth by inducing G1 cell cycle arrest.


Subject(s)
Apoptosis , Azepines , Cell Cycle Checkpoints , Cell Cycle Proteins , Melanoma , Transcription Factors , Triazoles , Animals , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Mice , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Azepines/pharmacology , Triazoles/pharmacology , Triazoles/therapeutic use , Cell Cycle Checkpoints/drug effects , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Uveal Neoplasms/drug therapy , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Uveal Neoplasms/metabolism , Flow Cytometry , Xenograft Model Antitumor Assays , Mice, Nude , Bromodomain Containing Proteins
3.
J Transl Med ; 22(1): 605, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951874

ABSTRACT

BACKGROUND: Uveal melanoma (UM), the most common adult intraocular tumor, is characterized by high malignancy and poor prognosis in advanced stages. Angiogenesis is critical for UM development, however, not only the role of vascular endothelial dysfunction in UM remains unknown, but also their analysis at the single-cell level has been lacking. A comprehensive analysis is essential to clarify the role of the endothelium in the development of UM. METHODS: By using single-cell RNA transcriptomics data of 11 cases of primary and liver metastasis UM, we analyzed the endothelial cell status. In addition, we analyzed and validated ECs in the in vitro model and collected clinical specimens. Subsequently, we explored the impact of endothelial dysfunction on UM cell migration and explored the mechanisms responsible for the endothelial cell abnormalities and the reasons for their peripheral effects. RESULTS: UM metastasis has a significantly higher percentage of vascular endothelial cells compared to in situ tumors, and endothelial cells in metastasis show significant senescence. Senescent endothelial cells in metastatic tumors showed significant Krüppel-like factor 4 (KLF4) upregulation, overexpression of KLF4 in normal endothelial cells induced senescence, and knockdown of KLF4 in senescent endothelium inhibited senescence, suggesting that KLF4 is a driver gene for endothelial senescence. KLF4-induced endothelial senescence drove tumor cell migration through a senescence-associated secretory phenotype (SASP), of which the most important component of the effector was CXCL12 (C-X-C motif chemokine ligand 12), and participated in the composition of the immunosuppressive microenvironment. CONCLUSION: This study provides an undesirable insight of senescent endothelial cells in promoting UM metastasis.


Subject(s)
Cell Movement , Cellular Senescence , Endothelial Cells , Kruppel-Like Factor 4 , Liver Neoplasms , Melanoma , Single-Cell Analysis , Uveal Neoplasms , Humans , Uveal Neoplasms/pathology , Uveal Neoplasms/genetics , Melanoma/pathology , Melanoma/genetics , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Liver Neoplasms/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Cell Line, Tumor , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Gene Expression Regulation, Neoplastic , Female , Male
4.
Oncoimmunology ; 13(1): 2374954, 2024.
Article in English | MEDLINE | ID: mdl-38957477

ABSTRACT

Gut microbiota impacts responses to immune checkpoint inhibitors (ICI). A high level of Faecalibacterium prausnitzii have been associated with a positive response to ICI in multiple cancer types. Here, based on fecal shotgun metagenomics data, we show in two independent cohorts of patients with non-small cell lung cancer and advanced melanoma that a high level of F. prausnitzii at baseline is positively associated with a better clinical response to ICI. In MCA205 tumor-bearing mice, administration of F. prausnitzii strain EXL01, already in clinical development for Inflammatory Bowel Disease, restores the anti-tumor response to ICI in the context of antibiotic-induced microbiota perturbation at clinical and tumor transcriptomics level. In vitro, EXL01 strain enhances T cell activation in the presence of ICI. Interestingly, oral administration of EXL01 strain did not induce any change in fecal microbiota diversity or composition, suggesting a direct effect on immune cells in the small intestine. F. prausnitzii strain EXL01 will be evaluated as an adjuvant to ICI in multiple cancers in the near future.


Subject(s)
Faecalibacterium prausnitzii , Gastrointestinal Microbiome , Immune Checkpoint Inhibitors , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Animals , Humans , Mice , Gastrointestinal Microbiome/drug effects , Faecalibacterium prausnitzii/drug effects , Female , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Melanoma/drug therapy , Melanoma/immunology , Melanoma/pathology , Feces/microbiology , Male , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Cell Line, Tumor , Mice, Inbred C57BL
5.
Dermatol Online J ; 30(2)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38959928

ABSTRACT

Large defects on the face after Mohs surgery have posed significant reconstructive challenges. A 90-year-old man presented with melanoma in situ of the central forehead, which resulted in a 4.5cmx4.3cm defect after multiple stages of Mohs surgery. Although different approaches for forehead repair with nasal root involvement are possible, we demonstrate that the V-Y advancement flap and subsequent Burrow graft for nasal root repair represents a viable closure technique for large circular defects of the central forehead.


Subject(s)
Forehead , Melanoma , Mohs Surgery , Skin Neoplasms , Surgical Flaps , Humans , Male , Forehead/surgery , Aged, 80 and over , Melanoma/surgery , Melanoma/pathology , Skin Neoplasms/surgery , Skin Neoplasms/pathology , Facial Neoplasms/surgery , Facial Neoplasms/pathology
6.
Clin Nucl Med ; 49(8): 748-749, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38967506

ABSTRACT

ABSTRACT: A 51-year-old woman with a 2-mm-Breslow-thickness melanoma on her arm had 99mTc-nanocolloid lymphoscintigraphy to localize the associated sentinel lymph node. A single axillary node was identified, and histology confirmed a micrometastasis of breast tissue origin. Imaging of the patient's breasts and subsequent biopsy confirmed ipsilateral stage III breast cancer, which was treated with lumpectomy and axillary node clearance. This is the first reported case of an incidental solid cancer diagnosis from a sentinel lymph node biopsy undertaken for a different tumor origin. This illustrates the importance of recognizing overlapping lymphatic distribution of sentinel lymph nodes, which can drain multiple organs.


Subject(s)
Arm , Breast Neoplasms , Incidental Findings , Lymphoscintigraphy , Melanoma , Skin Neoplasms , Technetium Tc 99m Aggregated Albumin , Humans , Female , Middle Aged , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Melanoma/diagnostic imaging , Melanoma/pathology , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/pathology , Arm/diagnostic imaging , Melanoma, Cutaneous Malignant , Sentinel Lymph Node Biopsy
7.
Oncoimmunology ; 13(1): 2370928, 2024.
Article in English | MEDLINE | ID: mdl-38948930

ABSTRACT

Deregulation or loss of the human leukocyte antigen class I (HLA-I) molecules on tumor cells leading to inhibition of CD8+ T cell recognition is an important tumor immune escape strategy, which could be caused by a posttranscriptional control of molecules in the HLA-I pathway mediated by RNA-binding proteins (RBPs). So far, there exists only limited information about the interaction of RBPs with HLA-I-associated molecules, but own work demonstrated a binding of the heterogeneous ribonucleoprotein C (hnRNP C) to the 3' untranslated region (UTR) of the TAP-associated glycoprotein tapasin (tpn). In this study, in silico analysis of pan-cancer TCGA datasets revealed that hnRNP C is higher expressed in tumor specimens compared to corresponding normal tissues, which is negatively correlated to tpn expression, T cell infiltration and the overall survival of tumor patients. Functional analysis demonstrated an upregulation of tpn expression upon siRNA-mediated downregulation of hnRNP C, which is accompanied by an increased HLA-I surface expression. Thus, hnRNP C has been identified to target tpn and its inhibition could improve the HLA-I surface expression on melanoma cells suggesting its use as a possible biomarker for T-cell-based tumor immunotherapies.


Subject(s)
3' Untranslated Regions , Heterogeneous-Nuclear Ribonucleoprotein Group C , Melanoma , Membrane Transport Proteins , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Melanoma/immunology , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , 3' Untranslated Regions/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
9.
Arch Dermatol Res ; 316(7): 447, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958761

ABSTRACT

Malignant melanoma presents a formidable challenge due to its aggressive metastatic behavior and limited response to current treatments. To address this, our study delves into the impact of anlotinib on angiogenesis and vasculogenic mimicry using malignant melanoma cells and human umbilical vein endothelial cells. Evaluating tubular structure formation, cell proliferation, migration, invasion, and key signaling molecules in angiogenesis, we demonstrated that anlotinib exerts a dose-dependent inhibition on tubular structures and effectively suppresses cell growth and invasion in both cell types. Furthermore, in a mouse xenograft model, anlotinib treatment resulted in reduced tumor growth and vascular density. Notably, the downregulation of VEGFR-2, FGFR-1, PDGFR-ß, and PI3K underscored the multitargeted antitumor activity of anlotinib. Our findings emphasize the therapeutic potential of anlotinib in targeting angiogenesis and vasculogenic mimicry, contributing to the development of novel strategies for combating malignant melanoma.


Subject(s)
Cell Movement , Cell Proliferation , Human Umbilical Vein Endothelial Cells , Indoles , Melanoma , Neovascularization, Pathologic , Quinolines , Vascular Endothelial Growth Factor Receptor-2 , Xenograft Model Antitumor Assays , Quinolines/pharmacology , Quinolines/therapeutic use , Quinolines/administration & dosage , Humans , Melanoma/drug therapy , Melanoma/pathology , Animals , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Indoles/pharmacology , Indoles/therapeutic use , Mice , Cell Proliferation/drug effects , Cell Line, Tumor , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Cell Movement/drug effects , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Mice, Nude , Angiogenesis
10.
Mol Cancer ; 23(1): 136, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965534

ABSTRACT

BACKGROUND: BRAF inhibitors are widely employed in the treatment of melanoma with the BRAF V600E mutation. However, the development of resistance compromises their therapeutic efficacy. Diverse genomic and transcriptomic alterations are found in BRAF inhibitor resistant melanoma, posing a pressing need for convergent, druggable target that reverse therapy resistant tumor with different resistance mechanisms. METHODS: CRISPR-Cas9 screens were performed to identify novel target gene whose inhibition selectively targets A375VR, a BRAF V600E mutant cell line with acquired resistance to vemurafenib. Various in vitro and in vivo assays, including cell competition assay, water soluble tetrazolium (WST) assay, live-dead assay and xenograft assay were performed to confirm synergistic cell death. Liquid Chromatography-Mass Spectrometry analyses quantified polyamine biosynthesis and changes in proteome in vemurafenib resistant melanoma. EIF5A hypusination dependent protein translation and subsequent changes in mitochondrial biogenesis and activity were assayed by O-propargyl-puromycin labeling assay, mitotracker, mitoSOX labeling and seahorse assay. Bioinformatics analyses were used to identify the association of polyamine biosynthesis with BRAF inhibitor resistance and poor prognosis in melanoma patient cohorts. RESULTS: We elucidate the role of polyamine biosynthesis and its regulatory mechanisms in promoting BRAF inhibitor resistance. Leveraging CRISPR-Cas9 screens, we identify AMD1 (S-adenosylmethionine decarboxylase 1), a critical enzyme for polyamine biosynthesis, as a druggable target whose inhibition reduces vemurafenib resistance. Metabolomic and proteomic analyses reveal that polyamine biosynthesis is upregulated in vemurafenib-resistant cancer, resulting in enhanced EIF5A hypusination, translation of mitochondrial proteins and oxidative phosphorylation. We also identify that sustained c-Myc levels in vemurafenib-resistant cancer are responsible for elevated polyamine biosynthesis. Inhibition of polyamine biosynthesis or c-Myc reversed vemurafenib resistance both in vitro cell line models and in vivo in a xenograft model. Polyamine biosynthesis signature is associated with poor prognosis and shorter progression free survival after BRAF/MAPK inhibitor treatment in melanoma cohorts, highlighting the clinical relevance of our findings. CONCLUSIONS: Our findings delineate the molecular mechanisms involving polyamine-EIF5A hypusination-mitochondrial respiration pathway conferring BRAF inhibitor resistance in melanoma. These targets will serve as effective therapeutic targets that can maximize the therapeutic efficacy of existing BRAF inhibitors.


Subject(s)
Drug Resistance, Neoplasm , Eukaryotic Translation Initiation Factor 5A , Melanoma , Mutation , Peptide Initiation Factors , Polyamines , Proto-Oncogene Proteins B-raf , RNA-Binding Proteins , Vemurafenib , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Animals , Polyamines/metabolism , Mice , Peptide Initiation Factors/metabolism , Peptide Initiation Factors/genetics , Cell Line, Tumor , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Vemurafenib/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Xenograft Model Antitumor Assays , CRISPR-Cas Systems , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Lysine/analogs & derivatives
11.
Skin Res Technol ; 30(7): e13842, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38965799

ABSTRACT

BACKGROUND: As the most important modifications on the RNA level, N6-methyladenosine (m6A-) and 5-methylcytosine (m5C-) modification could have a direct influence on the RNAs. Long non-coding RNAs (lncRNAs) could also be modified by methylcytosine modification. Compared with mRNAs, the function of lncRNAs could be more potent to some extent in biological processes like tumorigenesis. Until now, rare reports have been done associated with cutaneous melanoma. Herein, we wonder if the m6A- and m5C- modified lncRNAs could influence the immune landscape and prognosis in melanoma, and we also want to find some lncRNAs which could directly affect the malignant behaviors of melanoma. METHODS: Systematically, we explored the expression pattern of m6A- and m5C- modified lncRNAs in melanoma from datasets including UCSC Xena and NCBI GEO, and the prognostic lncRNAs were selected. Then, according to the expression pattern of lncRNAs, melanoma samples from these datasets were divided into several subtypes. Prognostic model, nomogram survival model, drug sensitivity, GO, and KEGG pathway analysis were performed. Furthermore, among several selected lncRNAs, we identified one lncRNA named LINC00893 and investigated its expression pattern and its biological function in melanoma cell lines. RESULTS: We identified 27 m6A- and m5C- related lncRNAs which were significantly associated with survival, and we made a subtype analysis of melanoma samples based on these 27 lncRNAs. Among the two subtypes, we found differences of immune cells infiltration between these two subtypes. Then, LASSO algorithm was used to screen the optimized lncRNAs combination including ZNF252P-AS1, MIAT, FAM13A-AS1, LINC-PINT, LINC00893, AGAP2-AS1, OIP5-AS1, and SEMA6A-AS1. We also found that there was a significant correlation between the different risk groups predicted based on RS model and the actual prognosis. The nomogram survival model based on independent survival prognostic factors was also constructed. Besides, sensitivity to chemotherapeutic agents, GO and KEGG analysis were performed. In different risk groups, a total of 14 drug molecules with different distributions were obtained, which included AZD6482, AZD7762, AZD8055, camptothecin, dasatinib, erlotinib, gefitinib, gemcitabine, GSK269962A, nilotinib, rapamycin, and sorafenib. A total of 55 significantly related biological processes and 17 KEGG signaling pathways were screened. At last, we noticed that LINC00893 had a relatively lower expression in melanoma tissue and cell lines compared with adjacent tissues and epidermal melanocyte, and down-regulation of LINC00893 could promote the malignant behavior of melanoma cells in A875 and MV3. In these two melanoma cell lines, down-regulation of m6A-related molecules like YTHDF3 and METTL3 could promote the expression of LINC00893. CONCLUSION: We made an analysis of m6A- and m5C- related lncRNAs in melanoma samples and a prediction of these lncRNAs' role in prognosis, tumor microenvironment, immune infiltration, and clinicopathological features. We also found that LINC00893, which is potentially regulated by m6A modification, could serve as a tumor-suppressor in melanoma and play an inhibitory role in melanoma metastasis.


Subject(s)
Adenosine , Melanoma , RNA, Long Noncoding , Skin Neoplasms , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Melanoma/genetics , Melanoma/pathology , Melanoma/mortality , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/mortality , Adenosine/analogs & derivatives , Adenosine/metabolism , Prognosis , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Melanoma, Cutaneous Malignant , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Nomograms
12.
Cesk Slov Oftalmol ; 80(Ahead of print): 1-6, 2024.
Article in English | MEDLINE | ID: mdl-38925897

ABSTRACT

AIM: To demonstrate a rare case of ciliary body leiomyoma in our patient Case report: A 72-year-old female reported to our clinic for a preventive examination, upon which we found a dome-shaped grey-brownish mass on the retinal periphery. After completing gonioscopic and ultrasound examinations, we referred the patient to a specialist facility. Due to a finding of suspicious malignant melanoma, we completed the MRI scan and recommended enucleation of the eyeball. A histopathological examination showed a leiomyoma of the ciliary body. CONCLUSION: The aim of this case report is to demonstrate the difficulty of intraocular leiomyoma diagnosis. Only immunohistochemical examination differentiated the tumor from malignant melanoma and determined the diagnosis of ciliary body leiomyoma. Perhaps because of the extreme rarity of this type of tumor, we often neglect to consider a diagnosis of leiomyoma.


Subject(s)
Ciliary Body , Leiomyoma , Uveal Neoplasms , Humans , Leiomyoma/pathology , Leiomyoma/diagnostic imaging , Leiomyoma/diagnosis , Leiomyoma/surgery , Female , Ciliary Body/pathology , Ciliary Body/diagnostic imaging , Aged , Uveal Neoplasms/pathology , Uveal Neoplasms/diagnostic imaging , Uveal Neoplasms/diagnosis , Uveal Neoplasms/surgery , Melanoma/pathology , Melanoma/diagnostic imaging , Melanoma/diagnosis , Melanoma/surgery , Diagnosis, Differential
13.
Discov Med ; 36(185): 1231-1240, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38926109

ABSTRACT

BACKGROUND: Cutaneous melanoma is a malignant tumor with an increasing incidence, prone to recurrence and metastasis. This study aims to explore the effects and mechanisms of the novel shikonin derivative 5,8-dimethyl alkannin oxime derivative (DMAKO-20) on the metastasis and invasion of melanoma cells. METHODS: The inhibitory effects of DMAKO-20 on the melanoma cell line A375 were investigated through Cell Counting Kit-8 (CCK-8), Transwell and angiogenesis experiments. Network pharmacology and Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were employed to explore potential sites and pathways involved in this process. Additionally, quantitative polymerase chain reaction (qPCR) and Western blot experiments were conducted before and after drug treatment to verify the expression trends of related pathways and proteins. RESULTS: DMAKO-20 demonstrated selective inhibition of proliferation, invasion and migration of melanoma cells at low concentrations. The WNT pathway appears to be implicated in this process, as DMAKO-20 effectively attenuates its activation, consequently reducing matrix metalloproteinase 9 (MMP9) and Cellular Communication Network Factor 1 (CCN1)/cysteine-rich angiogenic inducer 61 (CYR61) levels. Such modulation inhibits melanoma dissemination and invasion into other tissues. CONCLUSION: DMAKO-20 exhibits the capability to suppress metastasis and invasion of melanoma cells, suggesting its potential for clinical application as an adjuvant therapy against melanoma.


Subject(s)
Cell Movement , Cell Proliferation , Melanoma , Naphthoquinones , Neoplasm Invasiveness , Humans , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , Melanoma/drug therapy , Melanoma/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Neoplasm Metastasis , Wnt Signaling Pathway/drug effects , Skin Neoplasms/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Melanoma, Cutaneous Malignant
14.
Oncoimmunology ; 13(1): 2372118, 2024.
Article in English | MEDLINE | ID: mdl-38939518

ABSTRACT

The need for reliable biomarkers to predict clinical benefit from anti-PD1 treatment in metastatic melanoma (MM) patients remains unmet. Several parameters have been considered in the tumor environment or the blood, but none has yet achieved sufficient accuracy for routine clinical practice. Whole blood samples from MM patients receiving second-line anti-PD1 treatment (NCT02626065), collected longitudinally, were analyzed by flow cytometry to assess the immune cell subsets absolute numbers, the expression of immune checkpoints or ligands on T cells and the functionality of innate immune cells and T cells. Clinical response was assessed according to Progression-Free Survival (PFS) status at one-year following initiation of anti-PD1 (responders: PFS > 1 year; non-responders: PFS ≤ 1 year). At baseline, several phenotypic and functional alterations in blood immune cells were observed in MM patients compared to healthy donors, but only the proportion of polyfunctional memory CD4+ T cells was associated with response to anti-PD1. Under treatment, a decreased frequency of HVEM on CD4+ and CD8+ T cells after 3 months of treatment identified responding patients, whereas its receptor BTLA was not modulated. Both reduced proportion of CD69-expressing CD4+ and CD8+ T cells and increased number of polyfunctional blood memory T cells after 3 months of treatment were associated with response to anti-PD1. Of upmost importance, the combination of changes of all these markers accurately discriminated between responding and non-responding patients. These results suggest that drugs targeting HVEM/BTLA pathway may be of interest to improve anti-PD1 efficacy.


Subject(s)
Melanoma , Programmed Cell Death 1 Receptor , Receptors, Immunologic , Receptors, Tumor Necrosis Factor, Member 14 , Humans , Melanoma/drug therapy , Melanoma/immunology , Melanoma/pathology , Male , Receptors, Immunologic/metabolism , Female , Middle Aged , Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Adult , Treatment Outcome , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism
15.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928399

ABSTRACT

The ability of tumor-derived extracellular vesicles (EVs) to modulate the function of myeloid cells is widely recognized. Hence, a comprehensive understanding of the distinct components associated with EVs and the signals that they deliver to myeloid cells could provide potential approaches to impede the immunosuppression by myeloid-derived suppressor cells (MDSCs). We investigated melanoma EV-associated microRNAs (miRs) using the RET transgenic melanoma mouse model and simulated their transfer to normal myeloid cells by transfecting immature mouse myeloid cells and human monocytes. We observed elevated levels of miR-125a-5p, -125b-5p, and let-7e-5p in mouse melanoma-infiltrating MDSCs. In addition, miR-125a-5p levels in the tumor microenvironment correlated with mouse melanoma progression. The delivery of miR-125a-5p, alone or in combination with let-7e-5p and miR-99b-5p from the same genomic cluster, to normal myeloid cells resulted in their conversion to MDSC-like cells. Our findings indicate that miR-125a-5p could modulate myeloid cell activation in the melanoma microenvironment via a NF-κB-dependent mechanism.


Subject(s)
Melanoma , MicroRNAs , Myeloid-Derived Suppressor Cells , Tumor Microenvironment , MicroRNAs/genetics , MicroRNAs/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Animals , Humans , Mice , Tumor Microenvironment/genetics , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice, Transgenic , NF-kappa B/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Mice, Inbred C57BL , Monocytes/metabolism
16.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928466

ABSTRACT

Cutaneous melanoma is the most dangerous and deadly form of human skin malignancy. Despite its rarity, it accounts for a staggering 80% of deaths attributed to cutaneous cancers overall. Moreover, its final stages often exhibit resistance to drug treatments, resulting in unfavorable outcomes. Hence, ensuring access to novel and improved chemotherapeutic agents is imperative for patients grappling with this severe ailment. Pyrazole and its fused systems derived thereof are heteroaromatic moieties widely employed in medicinal chemistry to develop effective drugs for various therapeutic areas, including inflammation, pain, oxidation, pathogens, depression, and fever. In a previous study, we described the biochemical properties of a newly synthesized group of imidazo-pyrazole compounds. In this paper, to improve our knowledge of the pharmacological properties of these molecules, we conduct a differential proteomic analysis on a human melanoma cell line treated with one of these imidazo-pyrazole derivatives. Our results detail the changes to the SKMEL-28 cell line proteome induced by 24, 48, and 72 h of 3e imidazo-pyrazole treatment. Notably, we highlight the down-regulation of the Ras-responsive element binding protein 1 (RREB1), a member of the zinc finger transcription factors family involved in the tumorigenesis of melanoma. RREB1 is a downstream element of the MAPK pathway, and its activation is mediated by ERK1/2 through phosphorylation.


Subject(s)
Melanoma , Proteomics , Pyrazoles , Humans , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Pyrazoles/pharmacology , Pyrazoles/chemistry , Proteomics/methods , Cell Line, Tumor , Transcription Factors/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , DNA-Binding Proteins/metabolism , Imidazoles/pharmacology , Imidazoles/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Proteome/metabolism
17.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928474

ABSTRACT

Chitosan is a natural polymer with numerous biomedical applications. The cellular activity of chitosan has been studied in various types of cancer, including melanoma, and indicates that these molecules can open new perspectives on antiproliferative action and anticancer therapy. This study analyzes how different chitosan conformations, such as α-chitosan (CH) or ß-oligochitosan (CO), with various degrees of deacetylation (DDA) and molar mass (MM), both in different concentrations and in CH-CO mixtures, influence the cellular processes of SK-MEL-28 melanocytes, to estimate the reactivity of these cells to the applied treatments. The in vitro evaluation was carried out, aiming at the cellular metabolism (MTT assay), cellular morphology, and chitinase-like glycoprotein YKL-40 expression. The in vitro effect of the CH-CO mixture application on melanocytes is obvious at low concentrations of α-chitosan/ß-oligochitosan (1:2 ratio), with the cell's response supporting the hypothesis that ß-oligo-chitosan amplifies the effect. This oligochitosan mixture, favored by the ß conformation and its small size, penetrates faster into the cells, being more reactive when interacting with some cellular components. Morphological effects expressed by the loss of cell adhesion and the depletion of YKL-40 synthesis are significant responses of melanocytes. ß-oligochitosan (1.5 kDa) induces an extension of cytophysiological effects and limits the cell viability compared to α-chitosan (400-900 kDa). Statistical analysis using multivariate techniques showed differences between the CH samples and CH-CO mixtures.


Subject(s)
Chitin , Chitinase-3-Like Protein 1 , Chitosan , Melanocytes , Oligosaccharides , Chitosan/chemistry , Chitosan/pharmacology , Melanocytes/drug effects , Melanocytes/metabolism , Humans , Chitin/analogs & derivatives , Chitin/pharmacology , Chitin/chemistry , Oligosaccharides/pharmacology , Chitinase-3-Like Protein 1/metabolism , Cell Survival/drug effects , Cell Proliferation/drug effects , Cell Line, Tumor , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology
18.
Exp Dermatol ; 33(6): e15092, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38888196

ABSTRACT

Secreted protein acidic and cysteine rich/osteonectin, cwcv and kazal-like domain proteoglycan 2 (SPOCK2) is a protein that regulates cell differentiation and growth. Recent studies have reported that SPOCK2 plays important roles in the progression of various human cancers; however, the role of SPOCK2 in melanoma remains unknown. Therefore, this study investigated the roles of SPOCK2 and the related mechanisms in melanoma progression. To evaluate the clinical significance of SPOCK2 expression in patients with melanoma, we analysed the association between SPOCK2 expression and its prognostic value for patients with melanoma using systematic multiomic analysis. Subsequently, to investigate the roles of Spock2 in melanoma progression in vitro and in vivo, we knocked down Spock2 in the B16F10 melanoma cell line. High SPOCK2 levels were positively associated with good prognosis and long survival rate of patients with melanoma. Spock2 knockdown promoted melanoma cell proliferation by inducing the cell cycle and inhibiting apoptosis. Moreover, Spock2 downregulation significantly increased cell migration and invasion by upregulating MMP2 and MT1-MMP. The increased cell proliferation and migration were inhibited by MAPK inhibitor, and ERK phosphorylation was considerably enhanced in Spock2 knockdown cells. Therefore, Spock2 could function as a tumour suppressor gene to regulate melanoma progression by regulating the MAPK/ERK signalling pathway. Additionally, Spock2 knockdown cell injection induced considerable tumour growth and lung metastasis in C57BL6 mice compared to that in the control group. Our findings suggest that SPOCK2 plays crucial roles in malignant progression of melanoma and functions as a novel therapeutic target of melanoma.


Subject(s)
Apoptosis , Cell Movement , Cell Proliferation , Disease Progression , Melanoma , Skin Neoplasms , Animals , Female , Humans , Male , Mice , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Cell Cycle , Cell Line, Tumor , Gene Knockdown Techniques , MAP Kinase Signaling System , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Neoplasm Invasiveness , Prognosis , Proteoglycans/metabolism , Proteoglycans/genetics , Skin Neoplasms/pathology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
19.
Arch Dermatol Res ; 316(7): 419, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38904763

ABSTRACT

High-frequency ultrasound has been used to visualize depth and vascularization of cutaneous neoplasms, but little has been synthesized as a review for a robust level of evidence about the diagnostic accuracy of high-frequency ultrasound in dermatology. A narrative review of the PubMed database was performed to establish the correlation between ultrasound findings and histopathologic/dermoscopic findings for cutaneous neoplasms. Articles were divided into the following four categories: melanocytic, keratinocytic/epidermal, appendageal, and soft tissue/neural neoplasms. Review of the literature revealed that ultrasound findings and histopathology findings were strongly correlated regarding the depth of a cutaneous neoplasm. Morphological characteristics were correlated primarily in soft tissue/neural neoplasms. Overall, there is a paucity of literature on the correlation between high-frequency ultrasound and histopathology of cutaneous neoplasms. Further studies are needed to investigate this correlation in various dermatologic conditions.


Subject(s)
Skin Neoplasms , Ultrasonography , Humans , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/diagnosis , Skin Neoplasms/pathology , Ultrasonography/methods , Skin/diagnostic imaging , Skin/pathology , Dermoscopy/methods , Melanoma/diagnostic imaging , Melanoma/diagnosis , Melanoma/pathology
20.
Cells ; 13(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38920653

ABSTRACT

Uveal melanoma (UM), a distinct subtype of melanoma, presents unique challenges in its clinical management due to its complex molecular landscape and tendency for liver metastasis. This review highlights recent advancements in understanding the molecular pathogenesis, genetic alterations, and immune microenvironment of UM, with a focus on pivotal genes, such as GNAQ/11, BAP1, and CYSLTR2, and delves into the distinctive genetic and chromosomal classifications of UM, emphasizing the role of mutations and chromosomal rearrangements in disease progression and metastatic risk. Novel diagnostic biomarkers, including circulating tumor cells, DNA and extracellular vesicles, are discussed, offering potential non-invasive approaches for early detection and monitoring. It also explores emerging prognostic markers and their implications for patient stratification and personalized treatment strategies. Therapeutic approaches, including histone deacetylase inhibitors, MAPK pathway inhibitors, and emerging trends and concepts like CAR T-cell therapy, are evaluated for their efficacy in UM treatment. This review identifies challenges in UM research, such as the limited treatment options for metastatic UM and the need for improved prognostic tools, and suggests future directions, including the discovery of novel therapeutic targets, immunotherapeutic strategies, and advanced drug delivery systems. The review concludes by emphasizing the importance of continued research and innovation in addressing the unique challenges of UM to improve patient outcomes and develop more effective treatment strategies.


Subject(s)
Melanoma , Uveal Neoplasms , Humans , Uveal Neoplasms/genetics , Uveal Neoplasms/therapy , Uveal Neoplasms/pathology , Uveal Neoplasms/diagnosis , Melanoma/genetics , Melanoma/therapy , Melanoma/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Tumor Microenvironment/genetics , Mutation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...