Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.438
Filter
1.
Biochemistry ; 63(9): 1107-1117, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38671548

ABSTRACT

DNA polymerase θ (Pol θ or POLQ) is primarily involved in repairing double-stranded breaks in DNA through an alternative pathway known as microhomology-mediated end joining (MMEJ) or theta-mediated end joining (TMEJ). Unlike other DNA repair polymerases, Pol θ is thought to be highly error-prone yet critical for cell survival. We have identified several POLQ gene variants from human melanoma tumors that experience altered DNA polymerase activity, including a propensity for incorrect nucleotide selection and reduced polymerization rates compared to WT Pol θ. Variants are 30-fold less efficient at incorporating a nucleotide during repair and up to 70-fold less accurate at selecting the correct nucleotide opposite a templating base. This suggests that aberrant Pol θ has reduced DNA repair capabilities and may also contribute to increased mutagenesis. Moreover, the variants were identified in established tumors, suggesting that cancer cells may use mutated polymerases to promote metastasis and drug resistance.


Subject(s)
DNA Polymerase theta , DNA-Directed DNA Polymerase , Melanoma , Humans , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/chemistry , Melanoma/genetics , Melanoma/enzymology , DNA Repair , Mutation
2.
Biosensors (Basel) ; 14(4)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667195

ABSTRACT

Tyrosinase (TYR) emerges as a key enzyme that exerts a regulatory influence on the synthesis of melanin, thereby assuming the role of a critical biomarker for the detection of melanoma. Detecting the authentic concentration of TYR in the skin remains a primary challenge. Distinguished from ex vivo detection methods, this study introduces a novel sensor platform that integrates a microneedle (MN) biosensor with surface-enhanced Raman spectroscopy (SERS) technology for the in situ detection of TYR in human skin. The platform utilized dopamine (DA)-functionalized gold nanoparticles (Au NPs) as the capturing substrate and 4-mercaptophenylboronic acid (4-MPBA)-modified silver nanoparticles (Ag NPs) acting as the SERS probe. Here, the Au NPs were functionalized with mercaptosuccinic acid (MSA) for DA capture. In the presence of TYR, DA immobilized on the MN is preferentially oxidized to dopamine quinone (DQ), a process that results in a decreased density of SERS probes on the platform. TYR concentration was detected through variations in the signal intensity emitted by the phenylboronic acid. The detection system was able to evaluate TYR concentrations within a linear range of 0.05 U/mL to 200 U/mL and showed robust anti-interference capabilities. The proposed platform, integrating MN-based in situ sensing, SERS technology, and TYR responsiveness, holds significant importance for diagnosing cutaneous melanoma.


Subject(s)
Biosensing Techniques , Early Detection of Cancer , Melanoma , Monophenol Monooxygenase , Spectrum Analysis, Raman , Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Monophenol Monooxygenase/analysis , Monophenol Monooxygenase/metabolism , Skin/enzymology , Animals , Mice , Melanoma/diagnosis , Melanoma/enzymology , Metal Nanoparticles/chemistry , Gold/chemistry , Needles/standards , Enzyme-Linked Immunosorbent Assay , Silver/chemistry , Sensitivity and Specificity , Early Detection of Cancer/instrumentation , Early Detection of Cancer/methods
3.
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
4.
J Nat Prod ; 85(11): 2557-2569, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36351173

ABSTRACT

A library of more than 2500 plant extracts was screened for activity on oncogenic signaling in melanoma cells. The ethyl acetate extract from the aerial parts of Artemisia argyi displayed pronounced inhibition of the PI3K/AKT pathway. Active compounds were tracked with the aid of HPLC-based activity profiling, and altogether 21 active compounds were isolated, including one novel dimerosequiterpenoid (1), one new disesquiterpenoid (2), three new guaianolides (3-5), 12 known sesquiterpenoids (6-17), and four known flavonoids (19-22). A new eudesmanolide derivative (13b) was isolated as an artifact formed by methanolysis. Compound 1 is the first adduct comprising a sesquiterpene lactone and a methyl jasmonate moiety. The absolute configurations of compounds 1 and 3-18 were established by comparison of their experimental and calculated ECD spectra. The absolute configuration for 2 was determined by X-ray diffraction analysis. Guaianolide 8 was the most potent sesquiterpene lactone, inhibiting the PI3K/AKT pathway with an IC50 value of 8.9 ± 0.9 µM.


Subject(s)
Antineoplastic Agents , Artemisia , Lactones , Melanoma , Phosphatidylinositol 3-Kinases , Phytochemicals , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-akt , Sesquiterpenes , Artemisia/chemistry , Lactones/chemistry , Lactones/isolation & purification , Lactones/pharmacology , Melanoma/enzymology , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Phytochemicals/chemistry , Phytochemicals/isolation & purification , Phytochemicals/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Sesquiterpenes/chemistry , Sesquiterpenes/isolation & purification , Sesquiterpenes/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/isolation & purification , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology
5.
Int J Mol Sci ; 23(13)2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35805902

ABSTRACT

Melanoma is a relatively rare disease worldwide; nevertheless, it has a great relevance in some countries, such as in Europe. In order to shed some light upon the transcriptional profile of skin melanoma, we compared the gene expression of six independent tumours (all progressed towards metastatic disease and with wild type BRAF) to the expression profile of non-dysplastic melanocytes (considered as a healthy control) in a pilot study. Paraffin-embedded samples were manually micro-dissected to obtain enriched samples, and then, RNA was extracted and analysed through a microarray-based approach. An exhaustive bioinformatics analysis was performed to identify differentially expressed transcripts between the two groups, as well as enriched functional terms. Overall, 50 up- and 19 downregulated transcripts were found to be significantly changed in the tumour compared to the control tissue. Among the upregulated transcripts, the majority belonged to the immune response group and to the proteasome, while most of the downregulated genes were related to cytosolic ribosomes. A Gene Set Enrichment Analysis (GSEA), along with the RNA-Seq data retrieved from the TCGA/GTEx databases, confirmed the general trend of downregulation affecting cytoribosome proteins. In contrast, transcripts coding for mitoribosome proteins showed the opposite trend.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Skin Neoplasms , Humans , Melanocytes/metabolism , Melanoma/enzymology , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Neoplasm Metastasis , Pilot Projects , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
6.
Phytochemistry ; 198: 113154, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35245525

ABSTRACT

Three undescribed diterpenes including two ent-abietanes, euphomauritanol A, and euphomauritanol B, and one jatrophane, euphomauritanophane A, in addition to eight previously described metabolites were isolated from the MeOH-CH2Cl2 (1:1) extract of the Euphorbia mauritanica. The chemical structures of isolates were established based on the spectroscopic means including FT-IR, HRMS, 1D and 2D NMR. The absolute stereochemistry of the undescribed diterpenes was deduced by experimental and calculated TDDFT-electronic circular dichroism (ECD). The anti-proliferative effects of the isolated diterpenes were evaluated against B16-BL6, Hep G2, and Caco-2. The euphomauritanol A, euphomauritanol B, and euphomauritanophane A significantly inhibited the growth of murine melanoma B16-BL6 cell lines with IC50 10.28, 20.22, and 38.81 µM, respectively with no responses against the other cells. These activities were rationalized by molecular docking of the active compounds in BRAFV600E and MEK1 active sites. Moreover, the in-silico pharmacokinetics predictions by Swiss ADME revealed that the active compounds possessed favorable oral bioavailability and drug-likeness properties.


Subject(s)
Diterpenes , Euphorbia , MAP Kinase Kinase 1 , Melanoma , Proto-Oncogene Proteins B-raf , Animals , Caco-2 Cells , Diterpenes/chemistry , Diterpenes/pharmacology , Egypt , Euphorbia/chemistry , Hep G2 Cells , Humans , MAP Kinase Kinase 1/metabolism , Melanoma/drug therapy , Melanoma/enzymology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/enzymology , Mice , Molecular Docking Simulation , Molecular Structure , Proto-Oncogene Proteins B-raf/metabolism , Spectroscopy, Fourier Transform Infrared
7.
Molecules ; 27(4)2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35208960

ABSTRACT

Melanoma is the most dangerous skin malignancy due to its strong metastatic potential with high mortality. Activation of crucial signaling pathways enforcing melanoma progression depends on phosphorylation of distinct tyrosine kinases and oxidative stress. We here investigated the effect of a bis-coumarin derivative [3, 3'- ((3″, 5'-Dichlorophenyl) methylene) bis (4-hydroxy-2H-chromen-2-one)] [3, 3'- (3, 5-DCPBC)] on human melanoma cell survival, growth, proliferation, migration, intracellular redox state, and deciphered associated signaling pathways. This derivative is toxic for melanoma cells and non-toxic for melanocytes, their benign counterpart, and fibroblasts. 3, 3'- (3, 5-DCPBC) inhibits cell survival, migration, and proliferation of different metastatic and non-metastatic melanoma cell lines through profound suppression of the phosphorylation of Epidermal Growth Factor receptor (EGFR) and proto-oncogene cellular sarcoma (c-SRC) related downstream pathways. Thus, 3, 3'- (3, 5-DCPBC) endowed with the unique property to simultaneously suppress phosphorylation of multiple downstream kinases, such as EGFR/JAK/STAT and EGFR/SRC and their corresponding transcription factors.


Subject(s)
Coumarins , Down-Regulation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Melanoma , Neoplasm Proteins/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Signal Transduction/drug effects , Cell Line, Tumor , Coumarins/chemistry , Coumarins/pharmacology , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Humans , Melanoma/drug therapy , Melanoma/enzymology , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics
8.
Int J Mol Sci ; 23(4)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35216470

ABSTRACT

The role of proline dehydrogenase/proline oxidase (PRODH/POX) in the mechanism of antineoplastic activity of metformin (MET) was studied in C32 melanoma cells. PRODH/POX is a mitochondrial enzyme-degrading proline that is implicated in the regulation of cancer cell survival/apoptosis. The enzyme is activated by AMP kinase (AMPK). It has been found that MET induced a significant decrease in cell viability and DNA biosynthesis accompanied by an increase in the expressions of AMPK and PRODH/POX in C32 cells. The mechanism for MET-dependent cytotoxicity on C32 cells was found at the level of PRODH/POX-induced ROS generation and activation of Caspase-3 and Caspase-9 expressions in these cells. The effects were not observed in MET-treated PRODH/POX knock-out C32 cells. Of interest is an MET-dependent increase in the concentration of proline, which is a substrate for PRODH/POX. This phenomenon is due to the MET-dependent inhibition of collagen biosynthesis, which is the main proline-utilizing process. It has been found that the underlying mechanism of anticancer activity of MET involves the activation of AMPK, PRODH/POX, increase in the cytoplasmic concentration of proline, inhibition of collagen biosynthesis, and stimulation of PRODH/POX-dependent ROS generation, which initiate the apoptosis of melanoma cells.


Subject(s)
Apoptosis , Melanoma/drug therapy , Metformin/pharmacology , Proline Oxidase/metabolism , AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Melanoma/enzymology , Melanoma/physiopathology , Metformin/therapeutic use , Mitochondria/enzymology
9.
Pigment Cell Melanoma Res ; 35(1): 66-77, 2022 01.
Article in English | MEDLINE | ID: mdl-34482636

ABSTRACT

Melanoma cells expressing mutant B-RAF V600E are susceptible to treatment with the combination of a B-RAF inhibitor and a MEK1/2 inhibitor. We investigated the impact of the ERBB family and MAP4K inhibitor neratinib on the biology of PDX isolates of cutaneous melanoma expressing B-RAF V600E. Neratinib synergized with HDAC inhibitors to kill melanoma cells at their physiologic concentrations. Neratinib activated ATM, AMPK, ULK1, and PERK and inactivated mTORC1/2, ERK1/2, eIF2 alpha, and STAT3. Neratinib increased expression of Beclin1, ATG5, CD95, and FAS-L and decreased levels of multiple toxic BH3 domain proteins, MCL1, BCL-XL, FLIP-s, and ERBB1/2/4. ATG13 S318 phosphorylation and autophagosome formation was dependent upon ATM, and activation of ATM was dependent on reactive oxygen species. Reduced expression of ERBB1/2/4 required autophagosome formation and reduced MCL1/BCL-XL levels required eIF2 alpha phosphorylation. Maximal levels of eIF2 alpha phosphorylation required signaling by ATM-AMPK and autophagosome formation. Knock down of eIF2 alpha, CD95, FAS-L, Beclin1, and ATG5 or over-expression of FLIP-s significantly reduced killing. Combined knock down of Beclin1 and CD95 abolished cell death. Our data demonstrate that PDX melanoma cells expressing B-RAF V600E are susceptible to being killed by neratinib and more so when combined with HDACi.


Subject(s)
Autophagosomes/drug effects , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Death Domain/metabolism , Skin Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagosomes/genetics , Autophagosomes/metabolism , Autophagosomes/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Histone Deacetylase Inhibitors/pharmacology , Humans , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mutation , Proto-Oncogene Proteins B-raf/genetics , Receptors, Death Domain/genetics , Signal Transduction , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology
10.
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
11.
Br J Cancer ; 125(11): 1552-1560, 2021 11.
Article in English | MEDLINE | ID: mdl-34621046

ABSTRACT

BACKGROUND: Simultaneous inhibition of multiple components of the BRAF-MEK-ERK cascade (vertical inhibition) has become a standard of care for treating BRAF-mutant melanoma. However, the molecular mechanism of how vertical inhibition synergistically suppresses intracellular ERK activity, and consequently cell proliferation, are yet to be fully elucidated. METHODS: We develop a mechanistic mathematical model that describes how the mutant BRAF inhibitor, dabrafenib, and the MEK inhibitor, trametinib, affect BRAFV600E-MEK-ERK signalling. The model is based on a system of chemical reactions that describes cascade signalling dynamics. Using mass action kinetics, the chemical reactions are re-expressed as ordinary differential equations that are parameterised by in vitro data and solved numerically to obtain the temporal evolution of cascade component concentrations. RESULTS: The model provides a quantitative method to compute how dabrafenib and trametinib can be used in combination to synergistically inhibit ERK activity in BRAFV600E-mutant melanoma cells. The model elucidates molecular mechanisms of vertical inhibition of the BRAFV600E-MEK-ERK cascade and delineates how elevated BRAF concentrations generate drug resistance to dabrafenib and trametinib. The computational simulations further suggest that elevated ATP levels could be a factor in drug resistance to dabrafenib. CONCLUSIONS: The model can be used to systematically motivate which dabrafenib-trametinib dose combinations, for treating BRAFV600E-mutated melanoma, warrant experimental investigation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Models, Biological , Models, Chemical , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/genetics , Mitogen-Activated Protein Kinase Kinases/chemistry , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Oximes/chemistry , Oximes/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Pyridones/chemistry , Pyridones/pharmacology , Pyrimidinones/chemistry , Pyrimidinones/pharmacology
12.
Molecules ; 26(19)2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34641584

ABSTRACT

Despite its classification as a non-life-threatening disease, increased skin pigmentation adversely affects quality of life and leads to loss of self-confidence. Until now, there are no recommended remedies with high efficacy and human safety for hyperpigmentation. This study aimed to investigate anti-melanogenic activity and underlying mechanism of cajanin, an isoflavonoid extracted from Dalbergia parviflora Roxb. (Leguminosae) in human melanin-producing cells. Culture with 50 µM cajanin for 48-72 h significantly suppressed proliferation in human melanoma MNT1 cells assessed via MTT viability assay. Interestingly, cajanin also efficiently diminished melanin content in MNT1 cells with the half maximum inhibitory concentration (IC50) at 77.47 ± 9.28 µM. Instead of direct inactivating enzymatic function of human tyrosinase, down-regulated mRNA and protein expression levels of MITF and downstream melanogenic enzymes, including tyrosinase, TRP-1 and Dct (TRP-2) were observed in MNT1 cells treated with 50 µM cajanin for 24-72 h. Correspondingly, treatment with cajanin modulated the signaling pathway of CREB and ERK which both regulate MITF expression level. Targeted suppression on MITF-related proteins in human melanin-producing cells strengthens the potential development of cajanin as an effective treatment for human hyperpigmented disorders.


Subject(s)
Isoflavones/pharmacology , Melanoma/drug therapy , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/drug effects , Microphthalmia-Associated Transcription Factor/metabolism , Signal Transduction/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Dalbergia/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Hyperpigmentation/drug therapy , Interferon Type I/metabolism , Intramolecular Oxidoreductases/metabolism , Isoflavones/chemistry , Melanins/biosynthesis , Melanocytes/drug effects , Melanocytes/enzymology , Melanocytes/metabolism , Melanoma/enzymology , Monophenol Monooxygenase/metabolism , Plant Extracts/pharmacology , Pregnancy Proteins/metabolism , Quality of Life
13.
Int J Mol Sci ; 22(17)2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34502464

ABSTRACT

Melanoma is the most aggressive type of skin cancer due to its high capability of developing metastasis and acquiring chemoresistance. Altered redox homeostasis induced by increased reactive oxygen species is associated with melanomagenesis through modulation of redox signaling pathways. Dysfunctional endothelial nitric oxide synthase (eNOS) produces superoxide anion (O2-•) and contributes to the establishment of a pro-oxidant environment in melanoma. Although decreased tetrahydrobiopterin (BH4) bioavailability is associated with eNOS uncoupling in endothelial and human melanoma cells, in the present work we show that eNOS uncoupling in metastatic melanoma cells expressing the genes from de novo biopterin synthesis pathway Gch1, Pts, and Spr, and high BH4 concentration and BH4:BH2 ratio. Western blot analysis showed increased expression of Nos3, altering the stoichiometry balance between eNOS and BH4, contributing to NOS uncoupling. Both treatment with L-sepiapterin and eNOS downregulation induced increased nitric oxide (NO) and decreased O2• levels, triggering NOS coupling and reducing cell growth and resistance to anoikis and dacarbazine chemotherapy. Moreover, restoration of eNOS activity impaired tumor growth in vivo. Finally, NOS3 expression was found to be increased in human metastatic melanoma samples compared with the primary site. eNOS dysfunction may be an important mechanism supporting metastatic melanoma growth and hence a potential target for therapy.


Subject(s)
Biopterins/biosynthesis , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Melanoma/enzymology , Neoplasm Proteins/biosynthesis , Nitric Oxide Synthase Type III/biosynthesis , Animals , Biopterins/genetics , Female , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Neoplasm Metastasis , Neoplasm Proteins/genetics , Nitric Oxide Synthase Type III/genetics
14.
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
15.
Front Immunol ; 12: 736498, 2021.
Article in English | MEDLINE | ID: mdl-34557196

ABSTRACT

Immune escape is an early phenomenon in cancer development/progression. Indoleamine 2,3-dioxygenase 1 (IDO1) is a normal endogenous mechanism of acquired peripheral immune tolerance and may therefore be tumor-promoting. This study investigated the clinical relevance of IDO1 expression by immune cells in the lymph nodes and blood and of the serum kynurenine/tryptophan (Kyn/Trp) ratio in 65 systemic treatment naïve stage I-III melanoma patients. Blood samples were collected within the first year of diagnosis. Patients had a median follow-up of 61 months. High basal IDO1 expression in peripheral monocytes and low IFNγ-induced IDO1 upregulation correlated with worse outcome independent from disease stage. Interestingly studied factors were not interrelated. During follow-up, the risk of relapse was 9% (2/22) in the subgroup with high IFNγ-induced IDO1 upregulation in monocytes. In contrast, if IDO1 upregulation was low, relapse occurred in 30% (3/10) of patients with low basal IDO1 expression in monocytes and in 61.5% (8/13) in the subgroup with high basal IDO1 expression in monocytes (Log-Rank test, p=0.008). This study reveals some immune features in the blood of early stage melanoma that may be of relevance for disease outcome. These may offer a target for sub-stratification and early intervention.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Interferon-gamma/pharmacology , Kynurenine/blood , Melanoma/blood , Monocytes/drug effects , Skin Neoplasms/blood , Tryptophan/blood , Adult , Cells, Cultured , Enzyme Induction , Female , Humans , Male , Melanoma/enzymology , Melanoma/immunology , Melanoma/therapy , Middle Aged , Monocytes/enzymology , Monocytes/immunology , Neoplasm Recurrence, Local , Neoplasm Staging , Retrospective Studies , Skin Neoplasms/enzymology , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Time Factors , Treatment Outcome , Tumor Escape
16.
Eur J Cancer ; 156: 149-163, 2021 10.
Article in English | MEDLINE | ID: mdl-34454317

ABSTRACT

BACKGROUND: Melanoma brain metastases (MBM) have a poor prognosis. Systemic treatments that have improved outcomes in advanced melanoma have been shown to have an intracranial (IC) effect. We studied the efficacy and outcomes of combined immune checkpoint inhibitor ipilimumab/nivolumab (Combi-ICI) or targeted therapy (Combi-TT) as first-line treatment in MBM. METHODS: MBM patients treated with Combi-ICI or Combi-TT within 3 months after MBM diagnosis. Endpoints were progression-free survival (PFS) and overall survival (OS). RESULTS: 53 patients received Combi-ICI, 32% had symptomatic MBM and 33.9% elevated LDH. 71.7% required local treatment. The disease control rate was 60.3%. IC response rate (RR) was 43.8% at 3-months with durable responses at 6- (46.5%) and 12-months (53.1%). Extracranial (EC) RR was 44.7% at 3-months and 50% at 12-months. Median PFS was 9.6 months (95% CI 3.6-NR) and median overall survival (mOS) 44.8 months (95% CI; 26.2-NR). 63 patients received Combi-TT, 55.6% of patients had symptomatic MBM, 57.2% of patients had elevated LDH and 68.3% of patients required local treatment. The disease control rate was 60.4%. ICRR was 50% at 3-months, but dropped at 6-months (20.9%). ECRR was 69.2% at 3-months and 17.6% at 12-months. Median PFS was 5.8 months (95% CI 4.2-7.6) and mOS 14.2 months (95% CI 8.99-26.8). In BRAFV600 patients, 26.7% of patients received Combi-ICI and 73.3% Combi-TT with OS (p = 0.0053) and mPFS (p = 0.03) in favour to Combi-ICI. CONCLUSION: Combi-ICI showed prolonged mOS with sustainable IC and EC responses. Despite the initially increased efficacy, Combi-TT responses at 12 months were low. Combi-ICI appeared superior to Combi-TT for OS and PFS in BRAFV600 patients. Other clinical factors are determinants for first-line treatment choice.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Neoplasms/enzymology , Brain Neoplasms/immunology , Brain Neoplasms/secondary , CTLA-4 Antigen/antagonists & inhibitors , Europe , Female , Humans , Immune Checkpoint Inhibitors/adverse effects , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Melanoma/enzymology , Melanoma/immunology , Melanoma/secondary , Middle Aged , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Progression-Free Survival , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Retrospective Studies , Skin Neoplasms/enzymology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Time Factors , Victoria , Young Adult
17.
J Biochem Mol Toxicol ; 35(11): e22895, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34423490

ABSTRACT

Spindle and kinetochore-associated complex subunit 3 (SKA3) is reportedly a key contributor to the progression of various cancers. The present work aimed to evaluate the possible role of SKA3 in cutaneous melanoma (CM). A high SKA3 level was found in CM tissues and predicted a poor prognosis. SKA3 silencing markedly repressed the proliferation, invasion, and epithelial-mesenchymal transition and induced the apoptosis of CM cells. SKA3 silencing decreased the phosphorylation of PI3K and Akt. Akt inhibition markedly reversed SKA3 overexpression-induced oncogenic effects on CM cells. SKA3 silencing significantly prohibited the formation and growth of CM-derived xenograft tumors in nude mice in vivo. Our findings demonstrated SKA3 inhibition repressed the progression of CM by downregulating the PI3K/Akt pathway. This study indicates that SKA3 has potential as an anticancer candidate for CM.


Subject(s)
Kinetochores/metabolism , Melanoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/pathology , Spindle Apparatus/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Proliferation , Gene Silencing , Humans , Melanoma/enzymology , Mice , Mice, Nude , Microtubule-Associated Proteins/genetics , Neoplasm Invasiveness , Skin Neoplasms/enzymology , Up-Regulation
18.
Aging (Albany NY) ; 13(14): 18006-18017, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34257160

ABSTRACT

This investigation addressed the impact of integrin-initiated signaling pathways on senescence of tumor cells. In a model of human SK-Mel-147 melanoma cells, the silencing of integrin α2ß1 strongly reduced cell proliferation and enhanced the percentage of SA-ß-Gal-positive cells, a phenotypic feature of cellular senescence. These changes were accompanied by a significant increase in the activity of Akt and mTOR protein kinases and also in the expression of p53 and p21 oncosuppressors. Pharmacological inhibition of Akt and mTORC1 and genetic inhibition of p53 and p21 reduced the senescence of α2ß1-depleted SK-Mel-147 cells to the level of control cells. Based on our earlier data on the non-canonical functions of Akt isomers in the invasion and anoikis of SK-Mel-147 cells, we investigated the role of Akt isomers in senescence induced by α2ß1 suppression. The inhibition of Akt1 strongly reduced the percentage of SA-ß-Gal-positive cells in the α2ß1-depleted cell population, while the inhibition of Akt2 did not have a noticeable effect. Our data demonstrated for the first time that α2ß1 is involved in the protection of tumor cells against senescence and that senescence, which is induced by the downregulation of α2ß, is based on a signaling mechanism in which Akt1 performs a non-canonical function.


Subject(s)
Cellular Senescence/drug effects , Integrin alpha2beta1/metabolism , Melanoma/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/enzymology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Humans , Integrin alpha2beta1/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism
19.
Int J Mol Sci ; 22(11)2021 May 25.
Article in English | MEDLINE | ID: mdl-34070680

ABSTRACT

We previously reported (E)-ß-phenyl-α,ß-unsaturated carbonyl scaffold ((E)-PUSC) played an important role in showing high tyrosinase inhibitory activity and that derivatives with a 4-substituted resorcinol moiety as the ß-phenyl group of the scaffold resulted in the greatest tyrosinase inhibitory activity. To examine whether the 4-substituted resorcinol moiety could impart tyrosinase inhibitory activity in the absence of the α,ß-unsaturated carbonyl moiety of the (E)-PUSC scaffold, 10 urolithin derivatives were synthesized. To obtain more candidate samples, the lactone ring in synthesized urolithins was reduced to produce nine reduced urolithins. Compounds 1c (IC50 = 18.09 ± 0.25 µM), 1h (IC50 = 4.14 ± 0.10 µM), and 2a (IC50 = 15.69 ± 0.40 µM) had greater mushroom tyrosinase-inhibitory activities than kojic acid (KA) (IC50 = 48.62 ± 3.38 µM). The SAR results suggest that the 4-substituted resorcinol motif makes an important contribution to tyrosinase inhibition. To investigate whether these compounds bind to human tyrosinase, a human tyrosinase homology model was developed. Docking simulations with mushroom and human tyrosinases showed that 1c, 1h, and 2a bind to the active site of both tyrosinases with higher binding affinities than KA. Pharmacophore analyses showed that two hydroxyl groups of the 4-substituted resorcinol entity act as hydrogen bond donors in both mushroom and human tyrosinases. Kinetic analyses indicated that these compounds were all competitive inhibitors. Compound 2a inhibited cellular tyrosinase activity and melanogenesis in α-MSH plus IBMX-stimulated B16F10 melanoma cells more strongly than KA. These results suggest that 2a is a promising candidate for the treatment of skin pigment disorders, and show the 4-substituted resorcinol entity importantly contributes to tyrosinase inhibition.


Subject(s)
Agaricales/enzymology , Coumarins , Enzyme Inhibitors , Fungal Proteins , Melanoma/enzymology , Monophenol Monooxygenase , Neoplasm Proteins/antagonists & inhibitors , Resorcinols , Animals , Cell Line, Tumor , Coumarins/chemistry , Coumarins/pharmacology , Enzyme Inhibitors/pharmacology , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/metabolism , Humans , Melanins/biosynthesis , Mice , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/metabolism , Neoplasm Proteins/metabolism , Resorcinols/chemistry , Resorcinols/pharmacology
20.
Cancer Treat Rev ; 99: 102253, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34186441

ABSTRACT

In the last decade, immunotherapy and target therapy have revolutionized the prognosis of patients with BRAF-V600 mutation-positive metastatic melanoma. To date, three different combinations of BRAF/MEK inhibitors have been approved for this population, showing comparable efficacy and unique toxicity profiles. Several immune-checkpoint inhibitors, including pembrolizumab, nivolumab and the combination of nivolumab plus ipilimumab, are also available options for untreated metastatic melanoma patients. A novel approach has emerged by combining immune-checkpoint inhibitors and targeted agents, based on preclinical hints of synergy, prompting clinical results from large randomized trials. Specifically, the triplet of atezolizumab, vemurafenib and cobimetinib has been recently approved by FDA for patients with untreated BRAF-mutant metastatic melanoma. With a wide variety of available treatment options in this setting, it is paramount to establish criteria to select the most effective and safe frontline tailored approaches, for each patient. Results from ongoing studies are awaited, to maximise the benefits in survival outcomes and quality of life for patients, balancing adverse events and clinical benefit. The purpose of this review is to summarize the current landscape of standard and experimental treatment strategies for the first line treatment of patients with BRAF-mutated advanced melanoma and discuss the best patient-centered tailored strategies in the first-line setting.


Subject(s)
Melanoma/drug therapy , Melanoma/enzymology , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials, Phase III as Topic , Humans , Immune Checkpoint Inhibitors/administration & dosage , MAP Kinase Signaling System/drug effects , Melanoma/genetics , Melanoma/immunology , Molecular Targeted Therapy , Protein Kinase Inhibitors/administration & dosage , Randomized Controlled Trials as Topic , Skin Neoplasms/drug therapy , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...