Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Pigment Cell Melanoma Res ; 36(5): 330-347, 2023 09.
Article in English | MEDLINE | ID: mdl-37132530

ABSTRACT

Melanoma, a lethal malignancy that arises from melanocytes, exhibits a multiplicity of clinico-pathologically distinct subtypes in sun-exposed and non-sun-exposed areas. Melanocytes are derived from multipotent neural crest cells and are present in diverse anatomical locations, including skin, eyes, and various mucosal membranes. Tissue-resident melanocyte stem cells and melanocyte precursors contribute to melanocyte renewal. Elegant studies using mouse genetic models have shown that melanoma can arise from either melanocyte stem cells or differentiated pigment-producing melanocytes depending on a combination of tissue and anatomical site of origin and activation of oncogenic mutations (or overexpression) and/or the repression in expression or inactivating mutations in tumor suppressors. This variation raises the possibility that different subtypes of human melanomas (even subsets within each subtype) may also be a manifestation of malignancies of distinct cells of origin. Melanoma is known to exhibit phenotypic plasticity and trans-differentiation (defined as a tendency to differentiate into cell lineages other than the original lineage from which the tumor arose) along vascular and neural lineages. Additionally, stem cell-like properties such as pseudo-epithelial-to-mesenchymal (EMT-like) transition and expression of stem cell-related genes have also been associated with the development of melanoma drug resistance. Recent studies that employed reprogramming melanoma cells to induced pluripotent stem cells have uncovered potential relationships between melanoma plasticity, trans-differentiation, and drug resistance and implications for cell or origin of human cutaneous melanoma. This review provides a comprehensive summary of the current state of knowledge on melanoma cell of origin and the relationship between tumor cell plasticity and drug resistance.


Subject(s)
Induced Pluripotent Stem Cells , Melanoma , Skin Neoplasms , Animals , Mice , Humans , Melanoma/pathology , Skin Neoplasms/pathology , Cell Plasticity , Melanocytes/metabolism , Cell Differentiation , Drug Resistance , Induced Pluripotent Stem Cells/metabolism , Neural Crest/metabolism
2.
J Genet ; 1012022.
Article in English | MEDLINE | ID: mdl-35975816

ABSTRACT

With more than 33,000 sting cases and 47 deaths recorded between 2000 and 2016, Panama is the country with the highest incidence of envenomations by scorpions in Central America. Species in the genus Centruroides are responsible for most scorpion sting reports, however, identification at the species level is complicated because the genus has considerable intraspecific morphological variation. To date no molecular data have been reported from Panama that would help to estimate their genetic diversity and validate morphometric identification methods. We provide here the first genetic diversity data of the two endemic species (C. granosus and C. panamensis) and other two species reported in Panama (C. bicolor and C. limbatus). A total of 41 specimens were sequenced for COI and 16S rDNA mitochondrial genes. The phylogenetic concatenated analysis separates the Panamanian samples into four well-supported clades represented by C. bicolor, C. granosus and (C. panamensis + C. limbatus). The two endemic species are not the closest relatives in the tree. Low diversity in combination with its very narrow distribution suggest that C. panamensis is susceptible to environmental degradation. A single specimen of Coiba island is intermediate in the tree structure between C. bicolor and C. panamensis and may represent an early stage of speciation. The haplotype network is also consistent with the phylogenetic trees.


Subject(s)
Genetic Variation , Scorpions , Animals , Haplotypes , Panama/epidemiology , Phylogeny , Scorpions/genetics
3.
Cancers (Basel) ; 14(5)2022 Feb 27.
Article in English | MEDLINE | ID: mdl-35267541

ABSTRACT

BRAFV600E is the most common mutation driver in melanoma. This mutation is known to cause a brief burst of proliferation followed by growth arrest and senescence, which prevent an uncontrolled cell proliferation. This phenomenon is known as oncogene-induced senescence (OIS) and OIS escape is thought to lead to melanomagenesis. Much attention has been focused on the melanocyte-intrinsic mechanisms that contribute to senescence escape. Additional genetic events such as the loss of tumor suppressor PTEN and/or epigenetic changes that contribute to senescence escape have been described. However, the role of the skin microenvironment-specifically, the role of epidermal keratinocytes-on melanomagenesis is not fully understood. In this study, we employ a microfluidic platform to study the interaction between melanocytes expressing the BRAFV600E mutation as well as keratinocytes and dermal fibroblasts. We demonstrate that keratinocytes suppress senescence-related genes and promote the proliferation of transformed melanocytes. We also show that a keratinocyte-conditioned medium can alter the secretion of both pro- and anti-tumorigenic factors by transformed melanocytes. In addition, we show that melanocytes and keratinocytes from donors of white European and black African ancestry display different crosstalks; i.e., white keratinocytes appear to promote a more pro-tumorigenic phenotype compared with black keratinocytes. These data suggest that keratinocytes exert their influence on melanomagenesis both by suppressing senescence-related genes in melanocytes and by affecting the balance of the melanocyte-secreted factors that favor tumorigenesis.

4.
Biosens Bioelectron ; 142: 111522, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31336226

ABSTRACT

We report the fabrication of polyaniline nanofiber (PANI)-modified screen-printed electrode (PANI/SPE) incorporated in a poly-dimethylsiloxane (PDMS) microfluidic channel for the detection of circulating tumor cells. We employed this device to detect melanoma skin cancer cells through specific immunogenic binding of cell surface biomarker melanocortin 1 receptor (MC1R) to anti-MC1R antibody. The antibody-functionalized PANI/SPE was used in batch-continuous flow-through fashion. An aqueous cell suspension of ferri/ferrocyanide at a flow rate of 1.5 mL/min was passed over the immunosensor, which allowed for continuous electrochemical measurements. The sensor performed exceptionally well affording an ultralow limit of quantification of 1 melanoma cell/mL, both in buffer and when mixed with peripheral blood mononuclear cells, and the response was log-linear over the range of 10-9000 melanoma cells/10 mL.


Subject(s)
Biosensing Techniques/instrumentation , Cell Count/instrumentation , Melanoma/blood , Microfluidic Analytical Techniques/instrumentation , Neoplastic Cells, Circulating/pathology , Aniline Compounds/chemistry , Antibodies, Immobilized/chemistry , Cell Line, Tumor , Electrodes , Equipment Design , Humans , Immunoassay/instrumentation , Limit of Detection , Melanoma/pathology , Nanofibers/chemistry , Nanofibers/ultrastructure , Receptor, Melanocortin, Type 1/analysis
5.
Stem Cell Reports ; 13(1): 177-192, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31231022

ABSTRACT

Melanomas are known to exhibit phenotypic plasticity. However, the role cellular plasticity plays in melanoma tumor progression and drug resistance is not fully understood. Here, we used reprogramming of melanocytes and melanoma cells to induced pluripotent stem cell (iPSCs) to investigate the relationship between cellular plasticity and melanoma progression and mitogen-activated protein kinase (MAPK) inhibitor resistance. We found that melanocyte reprogramming is prevented by the expression of oncogenic BRAF, and in melanoma cells harboring oncogenic BRAF and sensitive to MAPK inhibitors, reprogramming can be restored by inhibition of the activated oncogenic pathway. Our data also suggest that melanoma tumor progression acts as a barrier to reprogramming. Under conditions that promote melanocytic differentiation of fibroblast- and melanocyte-derived iPSCs, melanoma-derived iPSCs exhibited neural cell-like dysplasia and increased MAPK inhibitor resistance. These data suggest that iPSC-like reprogramming and drug resistance of differentiated cells can serve as a model to understand melanoma cell plasticity-dependent mechanisms in recurrence of aggressive drug-resistant melanoma.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Melanoma/metabolism , Melanoma/pathology , Neoplastic Stem Cells/metabolism , Phenotype , Biomarkers, Tumor , Cell Differentiation , Cell Line, Tumor , Cell Plasticity , Cellular Reprogramming/drug effects , Cellular Reprogramming/genetics , Disease Progression , Drug Resistance, Neoplasm/genetics , Gene Expression , Humans , Induced Pluripotent Stem Cells/pathology , Melanocytes/metabolism , Melanoma/genetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutation , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/pharmacology
6.
Cancer Lett ; 414: 268-277, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29179997

ABSTRACT

Melanocyte development and differentiation are regulated by cAMP, which is produced by the adenylate cyclase (AC) enzyme upon activation of the melanocortin-1-receptor (MC1R). Individuals carrying single amino acid substitution variants of MC1R have impaired cAMP signaling and higher risk of melanoma. However, the contribution of AC to this risk is not clear. Downstream of AC, the phosphorylated transcription factor, cyclic AMP Responsive Element Binding Protein (pCREB), which is activated by protein kinase A, regulates the expression of several genes including the melanocyte master regulator MITF. The roles of AC and CREB in melanoma development and growth are not well understood. Here, we investigated the effect of topical application of AC inhibitor on BrafCA/Pten-/- mouse melanoma development. We show that AC inhibitor delays melanoma growth independent of MAPK pathway activity and melanin content. Next, employing a primary melanoma tissue microarray and quantitative immunohistochemistry, we show that pCREB levels are positively correlated with the proliferative status of melanoma, but low pCREB expression is associated with tumor aggressiveness and metastatic recurrence. These data suggest that low cAMP signaling inhibits tumor growth but is a predictor of melanoma aggressiveness.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/metabolism , Melanoma/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adenylyl Cyclase Inhibitors/pharmacology , Adult , Aged , Animals , Cell Line, Tumor , Disease Progression , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/genetics , Melanoma/prevention & control , Mice, Knockout , Mice, Transgenic , Middle Aged , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins B-raf/genetics , Tumor Burden/drug effects
7.
Mol Cancer Res ; 15(12): 1792-1802, 2017 12.
Article in English | MEDLINE | ID: mdl-28851815

ABSTRACT

Cyclic AMP (cAMP) is an important second messenger that regulates a wide range of physiologic processes. In mammalian cutaneous melanocytes, cAMP-mediated signaling pathways activated by G-protein-coupled receptors (GPCR), like melanocortin 1 receptor (MC1R), play critical roles in melanocyte homeostasis including cell survival, proliferation, and pigment synthesis. Impaired cAMP signaling is associated with increased risk of cutaneous melanoma. Although mutations in MAPK pathway components are the most frequent oncogenic drivers of melanoma, the role of cAMP in melanoma is not well understood. Here, using the Braf(V600E)/Pten-null mouse model of melanoma, topical application of an adenylate cyclase agonist, forskolin (a cAMP inducer), accelerated melanoma tumor development in vivo and stimulated the proliferation of mouse and human primary melanoma cells, but not human metastatic melanoma cells in vitro The differential response of primary and metastatic melanoma cells was also evident upon pharmacologic inhibition of the cAMP effector protein kinase A. Pharmacologic inhibition and siRNA-mediated knockdown of other cAMP signaling pathway components showed that EPAC-RAP1 axis, an alternative cAMP signaling pathway, mediates the switch in response of primary and metastatic melanoma cells to cAMP. Evaluation of pERK levels revealed that this phenotypic switch was not correlated with changes in MAPK pathway activity. Although cAMP elevation did not alter the sensitivity of metastatic melanoma cells to BRAF(V600E) and MEK inhibitors, the EPAC-RAP1 axis appears to contribute to resistance to MAPK pathway inhibition. These data reveal a MAPK pathway-independent switch in response to cAMP signaling during melanoma progression.Implications: The prosurvival mechanism involving the cAMP-EPAC-RAP1 signaling pathway suggest the potential for new targeted therapies in melanoma. Mol Cancer Res; 15(12); 1792-802. ©2017 AACR.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Melanoma/drug therapy , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins B-raf/genetics , Telomere-Binding Proteins/genetics , Adenylyl Cyclases/drug effects , Adenylyl Cyclases/genetics , Animals , Cell Line, Tumor , Colforsin/administration & dosage , Cyclic AMP/genetics , Cyclic AMP/metabolism , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/genetics , Neoplasm Metastasis , Receptor, Melanocortin, Type 1/genetics , Shelterin Complex , Signal Transduction/drug effects
8.
Neural Plast ; 2016: 1752176, 2016.
Article in English | MEDLINE | ID: mdl-26843989

ABSTRACT

An increasing body of evidence suggests that mechanisms related to the introduction and repair of DNA double strand breaks (DSBs) may be associated with long-term memory (LTM) processes. Previous studies from our group suggested that factors known to function in DNA recombination/repair machineries, such as DNA ligases, polymerases, and DNA endonucleases, play a role in LTM. Here we report data using C57BL/6 mice showing that the V(D)J recombination-activating gene 1 (RAG1), which encodes a factor that introduces DSBs in immunoglobulin and T-cell receptor genes, is induced in the amygdala, but not in the hippocampus, after context fear conditioning. Amygdalar induction of RAG1 mRNA, measured by real-time PCR, was not observed in context-only or shock-only controls, suggesting that the context fear conditioning response is related to associative learning processes. Furthermore, double immunofluorescence studies demonstrated the neuronal localization of RAG1 protein in amygdalar sections prepared after perfusion and fixation. In functional studies, intra-amygdalar injections of RAG1 gapmer antisense oligonucleotides, given 1 h prior to conditioning, resulted in amygdalar knockdown of RAG1 mRNA and a significant impairment in LTM, tested 24 h after training. Overall, these findings suggest that the V(D)J recombination-activating gene 1, RAG1, may play a role in LTM consolidation.


Subject(s)
Association Learning/physiology , Conditioning, Psychological/physiology , Fear/physiology , Genes, RAG-1 , Homeodomain Proteins/genetics , Memory, Long-Term/physiology , Amygdala/metabolism , Animals , Electroshock , Hippocampus/metabolism , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...