RESUMO
Genome instability relies on preserving the chromatin structure, with any histone imbalances threating DNA integrity. Histone synthesis occurs in the cytoplasm, followed by a maturation process before their nuclear translocation. This maturation involves protein folding and the establishment of post-translational modifications. Disruptions in this pathway hinder chromatin assembly and contribute to genome instability. JMJD1B, a histone demethylase, not only regulates gene expression but also ensures a proper supply of histones H3 and H4 for the chromatin assembly. Reduced JMJD1B levels lead to the cytoplasmic accumulation of histones, causing defects in the chromatin assembly and resulting in DNA damage. To investigate the role of JMJD1B in regulating genome stability and the malignancy of melanoma tumors, we used a JMJD1B/KDM3B knockout in B16F10 mouse melanoma cells to perform tumorigenic and genome instability assays. Additionally, we analyzed the transcriptomic data of human cutaneous melanoma tumors. Our results show the enhanced tumorigenic properties of JMJD1B knockout melanoma cells both in vitro and in vivo. The γH2AX staining, Micrococcal Nuclease sensitivity, and comet assays demonstrated increased DNA damage and genome instability. The JMJD1B expression in human melanoma tumors correlates with a lower mutational burden and fewer oncogenic driver mutations. Our findings highlight JMJD1B's role in maintaining genome integrity by ensuring a proper histone supply to the nucleus, expanding its function beyond gene expression regulation. JMJD1B emerges as a crucial player in preserving genome stability and the development of melanoma, with a potential role as a safeguard against oncogenic mutations.
Assuntos
Dano ao DNA , Instabilidade Genômica , Histonas , Histona Desmetilases com o Domínio Jumonji , Melanoma , Neoplasias Cutâneas , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Dano ao DNA/genética , Regulação Neoplásica da Expressão Gênica , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Melanoma/genética , Melanoma/patologia , Melanoma/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/patologia , Melanoma Experimental/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/metabolismoRESUMO
Cutaneous melanoma is an aggressive type of skin cancer that is recognized for its high metastatic potential and the challenges it presents in its treatment. There has been increasing interest in plant extracts and their potential applications in melanoma. The present study aimed to investigate the content of individual phenolic compounds in araçá-boi extract, evaluate their antioxidant activity, and explore their effects on cell viability, migration properties, oxidative stress levels, and protein expression in the human metastatic melanoma cell line SK-MEL-28. HPLC-DAD analysis identified 11 phenolic compounds in the araçá-boi extract. Trans-cinnamic acid was the main phenolic compound identified; therefore, it was used alone to verify its contribution to antitumor activities. SK-MEL-28 melanoma cells were treated for 24 h with different concentrations of araçá-boi extract and trans-cinnamic acid (200, 400, 600, 800, and 1600 µg/mL). Both the araçá-boi extract and trans-cinnamic acid reduced cell viability, cell migration, and oxidative stress in melanoma cells. Additionally, they modulate proteins involved in apoptosis and inflammation. These findings suggest the therapeutic potential of araçá-boi extract and its phenolic compounds in the context of melanoma, especially in strategies focused on preventing metastasis. Additional studies, such as the analysis of specific signaling pathways, would be valuable in confirming and expanding these observations.
Assuntos
Movimento Celular , Sobrevivência Celular , Cinamatos , Melanoma , Fenóis , Extratos Vegetais , Humanos , Melanoma/tratamento farmacológico , Melanoma/patologia , Melanoma/metabolismo , Movimento Celular/efeitos dos fármacos , Extratos Vegetais/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Cinamatos/farmacologia , Linhagem Celular Tumoral , Fenóis/farmacologia , Antioxidantes/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Estresse Oxidativo/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Antineoplásicos Fitogênicos/farmacologiaRESUMO
BACKGROUND: Angiogenesis is a process that many tumors depend on for growth, development, and metastasis. Vascular endothelial growth factor (VEGF) is one of the major players in tumor angiogenesis in several tumor types, including melanoma. VEGF inhibition is achieved by bevacizumab, a humanized monoclonal antibody that binds with high affinity to VEGF and prevents its function. In order to successfully enable in vivo VEGF expression imaging in a murine melanoma model, we previously labeled bevacizumab with [99mTc]Tc. We observed that this was feasible, but it had prolonged blood circulation and delayed tumor uptake. OBJECTIVE: The aim of this study was to develop a radiolabeled Fab bevacizumab fragment, [99mTc]Tc-HYNICFab( bevacizumab), for non-invasive in vivo VEGF expression molecular imaging. METHODS: Flow cytometry was used to examine VEGF presence in the murine melanoma cell line (B16-F10). Bevacizumab was digested with papain for six hours at 37°C to produce Fab(bevacizumab), which was then conjugated to NHS-HYNIC-Tfa for radiolabeling with [99mTc]Tc. Stability and binding affinity assays were also evaluated. Biodistribution and single photon emission computed tomography/computed tomography (SPECT/CT) were performed at 1, 3, and 6 h (n = 4) after injection of [99mTc]Tc-HYNIC-Fab(Bevacizumab) in normal and B16-F10 tumor-bearing C57Bl/6J mice. RESULTS: Using flow cytometry, it was shown that the B16-F10 murine melanoma cell line has intracellular VEGF expression. Papain incubation resulted in the complete digestion of bevacizumab with good purity and homogeneity. The radiolabeling yield of [99mTc]Tc-HYNIC-Fab(bevacizumab) was 85.00 ± 6.06%, with a specific activity of 291.87 ± 18.84 MBq/mg (n=3), showing in vitro stability. Binding assays demonstrated significant intracellular in vitro VEGF expression. Fast blood clearance and high kidney and tumor uptake were observed in biodistribution and SPECT/CT studies. CONCLUSIONS: We present the development and evaluation of [99mTc]Tc-HYNIC-Fab(bevacizumab), a novel molecular VEGF expression imaging agent that may be used for precision medicine in melanoma and potentially in other VEGF-expressing tumors.
Assuntos
Bevacizumab , Compostos de Organotecnécio , Fator A de Crescimento do Endotélio Vascular , Animais , Bevacizumab/química , Bevacizumab/farmacologia , Camundongos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Compostos de Organotecnécio/química , Imagem Molecular , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/síntese química , Camundongos Endogâmicos C57BL , Melanoma/diagnóstico por imagem , Melanoma/metabolismo , Melanoma/tratamento farmacológico , Melanoma/patologia , Distribuição Tecidual , Tecnécio/química , Estrutura Molecular , Humanos , Linhagem Celular Tumoral , Tomografia Computadorizada de Emissão de Fóton Único , Fragmentos Fab das Imunoglobulinas/químicaRESUMO
BACKGROUND: Metastatic melanoma stands out as the most lethal form of skin cancer because of its high propensity to spread and its remarkable resistance to treatment methods. METHODS: In this review article, we address the incidence of melanoma worldwide and its staging phases. We thoroughly investigate the different melanomas and their associated risk factors. In addition, we underscore the principal therapeutic goals and pharmacological methods that are currently used in the treatment of melanoma. RESULTS: The implementation of targeted therapies has contributed to improving the approach to patients. However, because of the emergence of resistance early in treatment, overall survival and progression-free periods continue to be limited. CONCLUSIONS: We provide new insights into plant serine protease inhibitor therapeutics, supporting high-throughput drug screening soon, and seeking a complementary approach to explain crucial mechanisms associated with melanoma.
Assuntos
Melanoma Maligno Cutâneo , Melanoma , Inibidores de Serina Proteinase , Neoplasias Cutâneas , Melanoma/tratamento farmacológico , Melanoma/patologia , Melanoma/metabolismo , Humanos , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Inibidores de Serina Proteinase/uso terapêutico , Inibidores de Serina Proteinase/farmacologia , Inibidores de Serina Proteinase/química , Biotecnologia/métodosRESUMO
Melanoma, a malignant tumor of melanocytes, poses a significant clinical challenge due to its aggressive nature and high potential for metastasis. The advent of targeted therapy has revolutionized the treatment landscape of melanoma, particularly for tumors harboring specific genetic alterations such as BRAF V600E mutations. Despite the initial success of targeted agents, resistance inevitably arises, underscoring the need for novel therapeutic strategies. This review explores the latest advances in targeted therapy for melanoma, focusing on new molecular targets, combination therapies, and strategies to overcome resistance.
Assuntos
Melanoma , Terapia de Alvo Molecular , Proteínas Proto-Oncogênicas B-raf , Humanos , Melanoma/tratamento farmacológico , Melanoma/terapia , Melanoma/genética , Melanoma/patologia , Melanoma/metabolismo , Terapia de Alvo Molecular/métodos , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos , Mutação , Antineoplásicos/uso terapêuticoRESUMO
Melanoma is a type of tumor skin with high metastatic potential. Reconstructed human skin, development for pre-clinic assay, are make using primary human cells, but with same limitations. The aim this study was to characterize a cell culture model, with structure similar to human skin containing melanoma cells entirely from cell lines. Reconstructed skin with melanoma were development using human fibroblasts (MRC5), human epidermal keratinocytes (HaCat), and human melanoma (SK-MEL-28) embedded in collagen type I. The structure was characterized by hematoxylin-eosin stained, as well as points of melanoma cell invasion, which was associated with activity of MMPs (MMP-2 and MMP-9) by zymographic method. Then, the gene expression of the target molecular mechanisms involved in melanoma progression were evaluated. Here, the model development showed a region epidermis organized and separated from the dermis, with fibroblast cells confined and melanoma cells form delimited area invasion. MMP-2 and MMP-9 were identified during of cell culture and gene expression of BRAF, NRAS, and Vimentin was confirmed. The proposed model provides one more opportunity to study in vitro tumor biology of melanoma and also to allows the study of new drugs with more reliable results then whats we would find in vivo.
Assuntos
Fibroblastos , Metaloproteinase 2 da Matriz , Metaloproteinase 9 da Matriz , Melanoma , Neoplasias Cutâneas , Humanos , Melanoma/patologia , Melanoma/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Neoplasias Cutâneas/patologia , Fibroblastos/metabolismo , Fibroblastos/efeitos dos fármacos , Linhagem Celular Tumoral , Pele/metabolismo , Pele/patologia , Invasividade Neoplásica , Queratinócitos/efeitos dos fármacos , Linhagem Celular , Vimentina/metabolismo , Vimentina/genéticaRESUMO
Xeroderma Pigmentosum (XP) is a genetic disorder characterized by photosensitivity, dyschromia, and high risk of skin cancer. From a clinical and histologic view, it can be difficult to diagnose cutaneous melanoma (CM) in XP patients and to define its resection margins. We aimed to study the role of PRAME (PReferentially Expressed Antigen in MElanoma) in differentiating intraepidermal CM from superficial atypical melanocytic proliferation of uncertain significance (SAMPUS) and evaluating the histological margins of CMs. We included XP patients. melanocitic and nonmelanocytic lesions with adjacent skin, and, as control groups, sun-damaged skin from non-XP individuals. Melanocytic lesions with a consensus diagnosis were grouped into CM, SAMPUS, or benign. The selected samples were PRAME-immunoshistochemically stained, and the ratio between immuno-positive cells/mm was recorded, according to Olds and colleagues for intraepidermal lesions. Lezcano and colleagues' method was used for intradermal lesions. Clinical data from XP patients were reviewed. All 9 patients were alive and well at the study closure, even those who developed melanoma metastases. Positive/diffuse PRAME expression was found in 29% (7/24) of intraepidermal CMs and 20% (1/5) SAMPUS samples. All 103 XP control samples and 24 adjacent lesions skin of non-XP patients were PRAME negative. This was a single-center and retrospective study, using a relatively small sample, limiting our conclusions. In XP patients' lesions, PRAME expression could help in the setting of challenging melanocytic tumors and surgical margins evaluation. It is also possible that the method can avoid overdiagnosis and, consequently, more aggressive treatment recommendation in unequivocal CM cases.
Assuntos
Antígenos de Neoplasias , Melanoma Maligno Cutâneo , Melanoma , Neoplasias Cutâneas , Xeroderma Pigmentoso , Humanos , Melanoma/metabolismo , Melanoma/diagnóstico , Melanoma/patologia , Antígenos de Neoplasias/metabolismo , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/diagnóstico , Masculino , Feminino , Xeroderma Pigmentoso/patologia , Xeroderma Pigmentoso/metabolismo , Xeroderma Pigmentoso/diagnóstico , Adulto , Adolescente , Pessoa de Meia-Idade , Criança , Imuno-HistoquímicaRESUMO
Cancer development and progression are intimately related with post-translational protein modifications, e.g., highly reactive thiol moiety of cysteines enables structural rearrangements resulting in redox biological switches. In this context, redox proteomics techniques, such as 2D redox DIGE, biotin switch assay and OxIcat are fundamental tools to identify and quantify redox-sensitive proteins and to understand redox mechanisms behind thiol modifications. Given the great variability in redox proteomics protocols, problems including decreased resolution of peptides and low protein amounts even after enrichment steps may occur. Considering the biological importance of thiol's oxidation in melanoma, we adapted the biotin-switch assay technique for melanoma cells in order to overcome the limitations and improve coverage of detected proteins.
Assuntos
Biotina , Melanoma , Oxirredução , Proteômica , Proteômica/métodos , Melanoma/metabolismo , Melanoma/patologia , Humanos , Linhagem Celular Tumoral , Biotina/química , Biotina/metabolismo , Compostos de Sulfidrila/química , Compostos de Sulfidrila/metabolismoRESUMO
Melanoma, an infrequent yet significant variant of skin cancer, emerges as a primary cause of brain metastasis among various malignancies. Despite recognizing the involvement of inflammatory molecules, particularly chemokines, in shaping the metastatic microenvironment, the intricate cellular signaling mechanisms underlying cerebral metastasis remain elusive. In our pursuit to unravel the role of cytokines in melanoma metastasis, we devised a protocol utilizing mixed cerebral cortical cells and SK-MEL-28 melanoma cell lines. Contrary to expectations, we observed no discernible morphological change in melanoma cells exposed to a cerebral conditioned medium (CM). However, a substantial increase in both migration and proliferation was quantitatively noted. Profiling the chemokine secretion by melanoma in response to the cerebral CM unveiled the pivotal role of interferon gamma-induced protein 10 (CXCL10), inhibiting the secretion of interleukin 8 (CXCL8). Furthermore, through a transwell assay, we demonstrated that knockdown CXCL10 led to a significant decrease in the migration of the SK-MEL-28 cell line. In conclusion, our findings suggest that a cerebral CM induces melanoma cell migration, while modulating the secretion of CXCL10 and CXCL8 in the context of brain metastases. These insights advance our understanding of the underlying mechanisms in melanoma cerebral metastasis, paving the way for further exploration and targeted therapeutic interventions.
Assuntos
Movimento Celular , Quimiocina CXCL10 , Melanoma , Transdução de Sinais , Quimiocina CXCL10/metabolismo , Quimiocina CXCL10/genética , Humanos , Meios de Cultivo Condicionados/farmacologia , Melanoma/patologia , Melanoma/metabolismo , Linhagem Celular Tumoral , Interleucina-8/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Invasividade Neoplásica , Proliferação de Células , Córtex Cerebral/metabolismo , Córtex Cerebral/patologiaRESUMO
Secreted quiescin/sulfhydryl oxidase (QSOX) is overexpressed in many tumor cell lines, including melanoma, and is usually associated with a pro-invasive phenotype. Our previous work described that B16-F10 cells enter in a quiescent state as a protective mechanism against damage generated by reactive oxygen species (ROS) during melanogenesis stimulation. Our present results show that QSOX activity was two-fold higher in cells with stimulated melanogenesis when compared to control cells. Considering that glutathione (GSH) is one of the main factor responsible for controlling redox homeostasis in cells, this work also aimed to investigate the relationship between QSOX activity, GSH levels and melanogenesis stimulation in B16-F10 murine melanoma cell line. The redox homeostasis was impaired by treating cells with GSH in excess or depleting its intracellular levels through BSO treatment. Interestingly, GSH-depleted cells without stimulation of melanogenesis kept high levels of viability, suggesting a possible adaptive mechanism of survival even under low GSH levels. They also showed lower extracellular activity of QSOX, and higher QSOX intracellular immunostaining, suggesting that this enzyme was less excreted from cells and corroborating with a diminished extracellular QSOX activity. On the other hand, cells under melanogenesis stimulation showed a lower GSH/GSSG ratio (8:1) in comparison with control (non-stimulated) cells (20:1), indicating a pro-oxidative state after stimulation. This was accompanied by decreased cell viability after GSH-depletion, no alterations in QSOX extracellular activity, but higher QSOX nucleic immunostaining. We suggest that melanogenesis stimulation and redox impairment caused by GSH-depletion enhanced the oxidative stress in these cells, contributing to additional alterations of its metabolic adaptive response.
Assuntos
Melanoma , Oxirredutases atuantes sobre Doadores de Grupo Enxofre , Animais , Camundongos , Glutationa/metabolismo , Melanoma/metabolismo , Oxirredução , Oxirredutases/metabolismo , Espécies Reativas de Oxigênio , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismoRESUMO
Seriniquinone (SQ) was initially described by our group as an antimelanoma drug candidate and now also as an antifungal drug candidate. Despite its promising in vitro effects, SQ translation has been hindered by poor water-solubility. In this paper, we described the challenging nanoformulation process of SQ, which culminated in the selection of a phosphatidylcholine-based lamellar phase (PLP1). Liposomes and nanostructured lipid carriers were also evaluated but failed to encapsulate the compound. SQ-loaded PLP1 (PLP1-SQ) was characterized for the presence of sedimented or non-dissolved SQ, rheological and thermal behavior, and irritation potential with hen's egg test on the chorioallantoic membrane (HET-CAM). PLP1 influence on transepidermal water loss (TEWL) and skin penetration of SQ was assessed in a porcine ear skin model, while biological activity was evaluated against melanoma cell lines (SK-MEL-28 and SK-MEL-147) and C. albicans SC5314. Despite the presence of few particles of non-dissolved SQ (observed under the microscope 2 days after formulation obtainment), PLP1 tripled SQ retention in viable skin layers compared to SQ solution at 12 h. This effect did not seem to relate to formulation-induced changes on the barrier function, as no increases in TEWL were observed. No sign of vascular toxicity in the HET-CAM model was observed after cutaneous treatment with PLP1. SQ activity was maintained on melanoma cells after 48 h-treatment (IC50 values of 0.59-0.98 µM) whereas the minimum inhibitory concentration (MIC) against C. albicans after 24 h-treatment was 32-fold higher. These results suggest that a safe formulation for SQ topical administration was developed, enabling further in vivo studies.
Assuntos
Melanoma , Micoses , Neoplasias Cutâneas , Animais , Feminino , Suínos , Galinhas , Melanoma/metabolismo , Pele/metabolismo , Neoplasias Cutâneas/metabolismo , Candida albicans , Água/farmacologiaRESUMO
The regulation of metastasis-related cellular aspects of two structurally similar AGIs from prunes tea infusion, with different molar masses, was studied in vitro against Triple Wild-Type metastatic melanoma (TWM) from murine and human origin. The higher molar mass AGI (AGI-78KDa) induced TWMs cells death and, in murine cell line, it decreased some metastasis-related cellular processes: invasiveness capacity, cell-extracellular matrix interaction, and colonies sizes. The lower molar mass AGI (AGI-12KDa) did not induce cell death but decreased TWMs proliferation rate and, in murine cell line, it decreased cell adhesion and colonies sizes. Both AGIs alter the clonogenic capacity of human cell line. In spite to understand why we saw so many differences between AGIs effects on murine and human cell lines we performed in silico analysis that demonstrated differential gene expression profiles between them. Complementary network topological predictions suggested that AGIs can modulate multiple pathways in a specie-dependent manner, which explain differential results obtained in vitro between cell lines. Our results pointed to therapeutic potential of AGIs from prunes tea against TWMs and showed that molecular weight of AGIs may influence their antitumor effects.
Assuntos
Galactanos , Melanoma , Humanos , Camundongos , Animais , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Peso Molecular , Chá , Linhagem Celular TumoralRESUMO
Cutaneous melanoma (CM) is a malignant neoplasm with a high metastatic rate that shows poor response to systemic treatments in patients with advanced stages. Recently, studies have highlighted the antineoplastic potential of natural compounds, such as polyphenols, in the adjuvant therapy context to treat CM. The objective of the present study was to evaluate the effect of different concentrations of curcumin (0.1-100 µM) on the metastatic CM cell line SK-MEL-28. The cells were treated for 6 and 24 h with different concentrations of curcumin. Cell viability was assessed by 3-(4,5-dimethyl-2thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay and fluorescence microscopy. The apoptotic-inducing potential was detected by annexin V flow cytometry. The wound healing assay was used to verify cell migration after the curcumin exposition. The redox profile was evaluated by levels of the pro-oxidant markers reactive oxygen species (ROS) and Nitric oxide (NOx) and antioxidants of total thiols (PSH) and nonprotein thiols. The gene expression and enzymatic activity of caspase 3 were evaluated by reverse transcription-quantitative polymerase chain reaction and a sensitive fluorescence assay, respectively. Curcumin significantly decreased the cell viability of SK-MEL-28 cells at both exposure times. It also induced apoptosis at the highest concentration tested (p < .0001). SK-MEL-28 cell migration was inhibited by curcumin after treatment with 10 µM (p < .0001) and 100 µM (p < .0001) for 6 and 24 h (p = .0006 and p < .0001, respectively). Furthermore, curcumin significantly increased levels of ROS and NOx. Finally, curcumin was capable of increasing the gene expression at 10 µM (p = .0344) and 100 µM (p = .0067) and enzymatic activity at 10 µM (p = .0086) and 100 µM (p < .0001) of caspase 3 after 24 h. For the first time, we elucidated in our study that curcumin increases ROS levels, promoting oxidative stress that activates the caspase pathway and culminates in SK-MEL-28 metastatic CM cell death.
Assuntos
Curcumina , Melanoma , Neoplasias Cutâneas , Humanos , Melanoma/metabolismo , Curcumina/farmacologia , Caspase 3/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , Apoptose , Compostos de Sulfidrila/farmacologia , Linhagem Celular Tumoral , Sobrevivência CelularRESUMO
Over the last years, the incidence of melanoma, the deadliest form of skin cancer, has risen significantly. Nearly half of the melanoma patients exhibit the BRAFV600E mutation. Although the use of BRAF and MEK inhibitors (BRAFi and MEKi) showed an impressive success rate in melanoma patients, durability of response remains an issue because tumor quickly becomes resistant. Here, we generated and characterized Lu1205 and A375 melanoma cells resistant to vemurafenib (BRAFi). Resistant cells (Lu1205R and A375R) exhibit higher IC50 (5-6 fold increase) and phospho-ERK levels and 2-3 times reduced apoptosis than their sensitive parents (Lu1205S and A375S). Moreover, resistant cells are 2-3 times bigger, display a more elongated morphology and have a modulation of migration capacity. Interestingly, pharmacological inhibition of sphingosine kinases, that prevents sphingosine-1-phosphate production, reduces migration of Lu1205R cells by 50 %. In addition, although Lu1205R cells showed increased basal levels of the autophagy markers LC3II and p62, they have decreased autophagosome degradation and autophagy flux. Remarkably, expression of Rab27A and Rab27B, which are involved in the release of extracellular vesicles are dramatically augmented in resistant cells (i.e. 5-7 fold increase). Indeed, conditioned media obtained from Lu1205R cells increased the resistance to vemurafenib of sensitive cells. Hence, these results support that resistance to vemurafenib modulates migration and the autophagic flux and may be transferred to nearby sensitive melanoma cells by factors that are released to the extracellular milieu by resistant cells.
Assuntos
Melanoma , Proteínas Proto-Oncogênicas B-raf , Humanos , Vemurafenib/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Sulfonamidas/farmacologia , Indóis/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Inibidores de Proteínas Quinases/farmacologia , AutofagiaRESUMO
Annexin A1 (AnxA1) is highly secreted by neutrophils and binds to formyl peptide receptors (FPRs) to trigger anti-inflammatory effects and efferocytosis. AnxA1 is also expressed in the tumor microenvironment, being mainly attributed to cancer cells. As recruited neutrophils are player cells at the tumor sites, the role of neutrophil-derived AnxA1 in lung melanoma metastasis was investigated here. Melanoma cells and neutrophils expressing AnxA1 were detected in biopsies from primary melanoma patients, which also presented higher levels of serum AnxA1 and augmented neutrophil-lymphocyte ratio (NLR) in the blood. Lung melanoma metastatic mice (C57BL/6; i.v. injected B16F10 cells) showed neutrophilia, elevated AnxA1 serum levels, and higher labeling for AnxA1 in neutrophils than in tumor cells at the lungs with metastasis. Peritoneal neutrophils collected from naïve mice were co-cultured with B16F10 cells or employed to obtain neutrophil-conditioned medium (NCM; 18 h incubation). B16F10 cells co-cultured with neutrophils or with NCM presented higher invasion, which was abolished if B16F10 cells were previously incubated with FPR antagonists or co-cultured with AnxA1 knockout (AnxA1-/-) neutrophils. The depletion of peripheral neutrophils during lung melanoma metastasis development (anti-Gr1; i.p. every 48 h for 21 days) reduced the number of metastases and AnxA1 serum levels in mice. Our findings show that AnxA1 secreted by neutrophils favors melanoma metastasis evolution via FPR pathways, addressing AnxA1 as a potential biomarker for the detection or progression of melanoma.
Assuntos
Anexina A1 , Melanoma , Animais , Camundongos , Anexina A1/metabolismo , Melanoma/metabolismo , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Fagocitose , Microambiente TumoralRESUMO
Melanoma is the most aggressive and lethal type of skin cancer, characterized by therapeutic resistance. In this context, the present study aimed to investigate the cytotoxic potential of manool, a diterpene from Salvia officinalis L., in human (A375) and murine (B16F10) melanoma cell lines. The analysis of cytotoxicity using the XTT assay showed the lowest IC50 after 48 h of treatment with the manool, being 17.6 and 18.2 µg/ml for A375 and B16F10, respectively. A selective antiproliferative effect of manool was observed on the A375 cells based on the colony formation assay, showing an IC50 equivalent to 5.6 µg/ml. The manool treatments led to 43.5% inhibition of the A375 cell migration at a concentration of 5.0 µg/ml. However, it did not affect cell migration in the B16F10 cells. Cell cycle analysis revealed that the manool interfered in the cell cycle of the A375 cells, blocking the G2/M phase. No changes in the cell cycle were observed in the B16F10 cells. Interestingly, manool did not induce apoptosis in the A375 cells, but apoptosis was observed after treatment of the B16F10 cells. Additionally, manool showed an antimelanoma effect in a reconstructed human skin model. Furthermore, in silico studies, showed that manool is stabilized in the active sites of the tubulin dimer with comparable energy concerning taxol, indicating that both structures can inhibit the proliferation of cancer cells. Altogether, it is concluded that manool, through the modulation of the cell cycle, presents a selective antiproliferative activity and a potential antimelanoma effect.
Assuntos
Diterpenos , Melanoma , Neoplasias Cutâneas , Humanos , Animais , Camundongos , Linhagem Celular Tumoral , Melanoma/metabolismo , Diterpenos/farmacologia , Apoptose , Técnicas de Cultura de Células , Proliferação de CélulasRESUMO
BACKGROUND: Melanoma is an aggressive form of skin cancer worldwide. Phosphoinositide-3-kinase regulatory subunit 2 (PIK3R2) exerts carcinogenic roles in various tumors. So far, the function and mechanism of PIK3R2 in melanoma are not been fully clarified. OBJECTIVE: We aimed to clarify the role of PIK3R2 in melanoma. METHODS: PIK3R2 expressions in melanoma clinical tissues and melanoma cells were measured using quantitative real-time PCR and Western blot. In addition, PIK3R2 expressions in different tumor stages of melanoma were determined by immunohistochemistry assay. Meanwhile, PIK3R2 function was evaluated using loss or gain-of-function assays, Cell Counting Kit-8 assay, flow cytometry, and Transwell analysis. Furthermore, PIK3R2 mechanism in melanoma was assessed by a series of rescue experiments. RESULTS: PIK3R2 was highly expressed in melanoma tissues and cells, and PIK3R2 expressions were the highest in Stage IV. Functionally, PIK3R2 knockdown repressed melanoma cell proliferation, invasion, epithelial-mesenchymal transition, and facilitated cell apoptosis. Also, PIK3R2 overexpression produced an opposite trend. Mechanistically, PIK3R2 facilitated melanoma progression by activating PI3K/AKT/NF-κB pathway. Furthermore, PIK3R2 knockdown restrained the melanoma tumor growth in vivo. CONCLUSIONS: PIK3R2 aggravated melanoma by activating PI3K/AKT/NF-κB pathway, prompting that PIK3R2 might be a therapeutic target for melanoma.
Assuntos
Melanoma , Fosfatidilinositol 3-Quinases , Neoplasias Cutâneas , Humanos , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismoRESUMO
Embora o melanoma seja representado por apenas cerca de 5% dos casos de tumores malignos de pele, é responsável pela ampla maioria de óbitos relacionados a tumores cutâneos. Seu desenvolvimento está principalmente associado à presença de mutações em oncogenes específicos, como BRAF, c-KIT e PDGFRA, os quais estão relacionados ao metabolismo celular. OBJETIVOS: Avaliar retrospectivamente a presença de mutações nos genes BRAF (éxons 15), C-KIT (éxons 9,11,13 e 17) e PDGFRA (éxons 12 ,14 e 18) e correlacionar os resultados com as características clínicas e epidemiológicas e desfecho dos pacientes. METODOLOGIA: Foi realizado um estudo retrospectivo de 94 pacientes com melanoma metastático atendidos no Hospital Amaral Carvalho entre os anos de 2015 a 2022, de acordo com as seguintes variáveis: idade ao diagnóstico, gênero, cor, local do tumor primário, local de metástase, subtipo histológico de melanoma, dosagem de LDH, positividade para marcadores de imuno-histoquímica, espessura, presença de ulceração, metástase no linfonodo ao diagnóstico do tumor primário, espessura Breslow, classificação Clark, índice mitótico e desfecho (óbito, sobrevida). RESULTADOS: A maioria dos pacientes eram do sexo masculino (52,1%), de cor branca (97,9%) e com idade acima de 60 anos (42,5%). O subtipo histológico mais encontrado foi o extensivo superficial (42,5%), sendo os membros inferiores o local mais acometido (35,1%). O nível IV de Clark (39,4%) e índice de Breslow acima de 4mm (33,0%) foram os mais prevalentes. A maior parte dos tumores não apresentou ulceração (53,2%) e mais da metade dos pacientes apresentou mais de uma mitose por mm² (47,9%). A dosagem de desidrogenase láctica (LDH) se manteve dentro da normalidade na maioria dos pacientes no momento do diagnóstico (43,6%) e no surgimento de metástases (30,9%). Todos os pacientes apresentaram ao menos uma metástase, sendo os linfonodos regionais o local de maior incidência. A maioria dos pacientes faleceu em decorrência da doença (66,0%). A análise de BRAF apresentou significância estatística para as variáveis idade ao diagnóstico (p= 0,0085), tipo histológico (p= 0,048) e nível de Clark (p= 0,0290). A mediana de sobrevivência para pacientes com mutações em BRAF foi de 62 meses e 50 meses para aqueles que não apresentaram mutações. Não foram encontradas significâncias estatísticas entre c-KIT e as variáveis analisadas. A mediana de sobrevivência para pacientes com mutações em c-KIT foi de 38 meses e 70 meses para aqueles que não apresentaram mutações. CONCLUSÕES: A presença de mutação em BRAF esteve associada à menor idade e menores índices de Clark, e relacionada à ligeira maior sobrevida. O contrário foi observado para mutações em c-KIT, apesar da ausência de significância estatística: uma ligeira menor sobrevida foi observada em pacientes mutados. O LDH não esteve bem correlacionado à presença do melanoma metastático ao se apresentar normal na maioria dos casos. Grande parte dos pacientes tiveram o tumor primário localizado nos membros inferiores, diferentemente do relatado por outros estudos, e apesar de não significante, os tumores de tronco foram em sua maioria mutados para BRAF, e o oposto observado em membros inferiores e superiores. Assim, o papel prognóstico da mutação em BRAF na progressão da doença é ainda controverso. Tais dados podem fornecer mais informações para a adequação de protocolos de prevenção e tratamento de pacientes acometidos por melanoma.
INTRODUCTION: Although melanoma is represented by only about 5% of cases of malignant skin tumors, it is responsible for the vast majority of deaths related to skin tumors. Its development is mainly associated with the presence of mutations in specific oncogenes, such as BRAF, c-KIT and PDGFRA, which are related to cell metabolism. OBJECTIVES: Retrospectively evaluate the presence of mutations in BRAF (exons 15), C-KIT (exons 9,11,13 and 17) and PDGFRA (exons 12, 14 and 18) genes and correlate the results with the clinical and epidemiological characteristics and patient outcomes. METHODS: A retrospective study of 94 patients with metastatic melanoma treated at Hospital Amaral Carvalho between the years 2015 to 2022 was carried out, according to the following variables: age at diagnosis, gender, color, site of primary tumor, site of metastasis, histological subtype of melanoma, LDH dosage, positivity for immunohistochemical markers, thickness, presence of ulceration, lymph node metastasis at diagnosis of the primary tumor, Breslow thickness, Clark classification, mitotic index and outcome (death, survival). RESULTS: Most patients were male (52.1%), white (97.9%) and aged over 60 years (42.5%). The most common histological subtype was superficial extensive (42.5%), with the lower limbs being the most affected site (35.1%). Clark's level IV (39.4%) and Breslow thickness above 4mm (33.0%) were the most prevalent. Most tumors did not show ulceration (53.2%) and more than half of the patients had more than one mitosis per mm² (47.9%). The lactate dehydrogenase (LDH) dosage remained within the normal range in most patients at the time of diagnosis (43.6%) and at the onset of metastases (30.9%). All patients had at least one metastasis, with regional lymph nodes being the site with the highest incidence. Most patients died as a result of the disease (66.0%). BRAF analysis showed statistical significance for the variables age at diagnosis (p= 0.0085), histological type (p= 0.048) and Clark's level (p= 0.0290). Median survival for patients with BRAF mutations was 62 months and 50 months for those without mutations. No statistical significance was found between c-KIT and the analyzed variables. The median survival for patients with c-KIT mutations was 38 months and 70 months for those without mutations. CONCLUSIONS: The presence of a BRAF mutation was associated with younger age and lower Clark scores, and related to slightly longer survival. The opposite was observed for mutations in c-KIT, despite the absence of statistical significance: a slightly lower survival was observed in mutated patients. LDH was not well correlated with the presence of metastatic melanoma as it was normal in most cases. Most of the patients had the primary tumor located in the lower limbs, unlike what has been reported by other studies, and although not significant, the trunk tumors were mostly mutated to BRAF, and the opposite was observed in the lower and upper limbs. Thus, the prognostic role of the BRAF mutation in disease progression is still controversial. Such data may provide more information for the adequacy of prevention and treatment protocols for patients affected by melanoma.
Assuntos
Humanos , Masculino , Feminino , Proteínas Proto-Oncogênicas c-kit , Proteínas Proto-Oncogênicas B-raf , Melanoma/metabolismo , Mutação , Neoplasias Cutâneas , Institutos de Câncer/estatística & dados numéricos , Estudos Retrospectivos , Epidemiologia Clínica , Melanoma/genéticaRESUMO
CD8+ and CD4+ T-cells play a key role in cellular immune responses against cancer by cytotoxic responses and effector lineages differentiation, respectively. These subsets have been found in different types of cancer; however, it is unclear whether tumor-infiltrating T-cell subsets exhibit similar transcriptome profiling across different types of cancer in comparison with healthy tissue-resident T-cells. Thus, we analyzed the single cell transcriptome of five tumor-infiltrating CD4-T, CD8-T and Treg cells obtained from different types of cancer to identify specific pathways for each subset in malignant environments. An in silico analysis was performed from single-cell RNA-sequencing data available in public repositories (Gene Expression Omnibus) including breast cancer, melanoma, colorectal cancer, lung cancer and head and neck cancer. After dimensionality reduction, clustering and selection of the different subpopulations from malignant and nonmalignant datasets, common genes across different types of cancer were identified and compared to nonmalignant genes for each T-cell subset to identify specific pathways. Exclusive pathways in CD4+ cells, CD8+ cells and Tregs, and common pathways for the tumor-infiltrating T-cell subsets were identified. Finally, the identified pathways were compared with RNAseq and proteomic data obtained from T-cell subsets cultured under malignant environments and we observed that cytokine signaling, especially Th2-type cytokine, was the top overrepresented pathway in Tregs from malignant samples.