Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 51(4): 409-22, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23973372

RESUMO

The individuals carrying melanocortin-1 receptor (MC1R) variants, especially those associated with red hair color, fair skin, and poor tanning ability (RHC trait), are more prone to melanoma; however, the underlying mechanism is poorly defined. Here, we report that UVB exposure triggers phosphatase and tensin homolog (PTEN) interaction with wild-type (WT), but not RHC-associated MC1R variants, which protects PTEN from WWP2-mediated degradation, leading to AKT inactivation. Strikingly, the biological consequences of the failure of MC1R variants to suppress PI3K/AKT signaling are highly context dependent. In primary melanocytes, hyperactivation of PI3K/AKT signaling leads to premature senescence; in the presence of BRAF(V600E), MC1R deficiency-induced elevated PI3K/AKT signaling drives oncogenic transformation. These studies establish the MC1R-PTEN axis as a central regulator for melanocytes' response to UVB exposure and reveal the molecular basis underlying the association between MC1R variants and melanomagenesis.


Assuntos
Regulação da Expressão Gênica/efeitos da radiação , Melanócitos/metabolismo , Melanoma Experimental/patologia , PTEN Fosfo-Hidrolase/metabolismo , Receptor Tipo 1 de Melanocortina/metabolismo , Pigmentação da Pele/fisiologia , Raios Ultravioleta , Animais , Western Blotting , Células Cultivadas , Humanos , Técnicas Imunoenzimáticas , Melanócitos/efeitos da radiação , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Camundongos , Mutação/genética , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 1 de Melanocortina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Pigmentação da Pele/efeitos da radiação , alfa-MSH/genética , alfa-MSH/metabolismo
2.
Lab Invest ; 97(6): 725-736, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28165469

RESUMO

Melanoma is among the most virulent cancers, owing to its propensity to metastasize and its resistance to current therapies. The treatment failure is largely attributed to tumor heterogeneity, particularly subpopulations possessing stem cell-like properties, ie, melanoma stem-like cells (MSLCs). Evidence indicates that the MSLC phenotype is malleable and may be acquired by non-MSLCs through phenotypic switching upon appropriate stimuli, the so-called 'dynamic stemness'. Since the phenotypic characteristics and functional integrity of MSLCs depend on their vascular niche, using a two-dimensional (2D) melanoma-endothelium co-culture model, where the MSLC niche is recapitulated in vitro, we identified Notch3 signaling pathway as a micro-environmental cue governing MSLC phenotypic plasticity via pathway-specific gene expression arrays. Accordingly, lentiviral shRNA-mediated Notch3 knockdown (KD) in melanoma cell lines exhibiting high levels of endogenous Notch3 led to retarded/abolished tumorigenicity in vivo through both depleting MSLC fractions, evinced by MSLC marker downregulation (eg, CD133 and CD271); and impeding the MSLC niche, corroborated by the attenuated tumor angiogenesis as well as vasculogenic mimicry. In contrast, Notch3 KD affected neither tumor growth nor MSLC subsets in a melanoma cell line with relatively low endogenous Notch3 expression. Thus, Notch3 signaling may facilitate MSLC plasticity and niche morphogenesis in a cell context-dependent manner. Our findings illustrate Notch3 as a molecular switch driving melanoma heterogeneity, and provide the biological rationale for Notch inhibition as a promising therapeutic option.


Assuntos
Melanoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptor Notch3/metabolismo , Nicho de Células-Tronco/fisiologia , Microambiente Tumoral/fisiologia , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Transdução de Sinais
3.
Exp Cell Res ; 342(2): 159-65, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26988422

RESUMO

Chemotaxis is typically studied in vitro using commercially available products such as the Transwell® in which cells migrate through a porous membrane in response to one or more clearly defined chemotactic stimuli. Despite its widespread use, the Transwell assay suffers from being largely an endpoint assay, with built-in errors due to inconsistent pore size and human sampling. In this study, we report a microfluidic chemotactic chip that provides real-time monitoring, consistent paths for cell migration, and easy on-chip staining for quantifying migration. To compare its performance with that of a traditional Transwell chamber, we investigate the chemotactic response of MDA-MB-231 1833 metastatic breast cancer cells to epidermal growth factor (EGF). The results show that while both platforms were able to detect a chemotactic response, we observed a dose-dependent response of breast cancer cells towards EGF with low non-specific migration using the microfluidic platform, whereas we observed a dose-independent response of breast cancer cells towards EGF with high levels of non-specific migration using the commercially available Transwell.The microfluidic platform also allowed EGF-dependent chemotactic responses to be observed 24h, a substantially longer window than seen with the Transwell. Thus the performance of our microfluidic platform revealed phenomena that were not detected in the Transwell under the conditions tested.


Assuntos
Quimiotaxia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/fisiologia , Humanos , Dispositivos Lab-On-A-Chip
4.
Proc Natl Acad Sci U S A ; 108(19): 7814-9, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21518915

RESUMO

Histone modifications are reported to show different behaviors, associations, and functions in different genomic niches and organisms. We show here that rapid, continuous turnover of acetylation specifically targeted to all K4-trimethylated H3 tails (H3K4me3), but not to bulk histone H3 or H3 carrying other methylated lysines, is a common uniform characteristic of chromatin biology in higher eukaryotes, being precisely conserved in human, mouse, and Drosophila. Furthermore, dynamic acetylation targeted to H3K4me3 is mediated by the same lysine acetyltransferase, p300/cAMP response element binding (CREB)-binding protein (CBP), in both mouse and fly cells. RNA interference or chemical inhibition of p300/CBP using a newly discovered small molecule inhibitor, C646, blocks dynamic acetylation of H3K4me3 globally in mouse and fly cells, and locally across the promoter and start-site of inducible genes in the mouse, thereby disrupting RNA polymerase II association and the activation of these genes. Thus, rapid dynamic acetylation of all H3K4me3 mediated by p300/CBP is a general, evolutionarily conserved phenomenon playing an essential role in the induction of immediate-early (IE) genes. These studies indicate a more global function of p300/CBP in mediating rapid turnover of acetylation of all H3K4me3 across the nuclei of higher eukaryotes, rather than a tight promoter-restricted function targeted by complex formation with specific transcription factors.


Assuntos
Histonas/química , Histonas/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação , Animais , Sítios de Ligação , Linhagem Celular , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Evolução Molecular , Técnicas de Silenciamento de Genes , Genes Precoces , Genes fos , Genes jun , Histona Acetiltransferases/antagonistas & inibidores , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Lisina/química , Camundongos , Regiões Promotoras Genéticas , Especificidade da Espécie , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Fatores de Transcrição de p300-CBP/genética
5.
bioRxiv ; 2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-39229179

RESUMO

Malignant peripheral nerve sheath tumor (MPNST) is a highly aggressive sarcoma that may be seen in patients with neurofibromatosis type 1 (NF1) or occur sporadically. While surgery is the primary treatment for localized MPNST with a 61.9% overall survival rate, metastatic disease is often fatal due to resistance to systemic therapies which underscores the urgent need for effective treatments. MPNSTs frequently harbor inactivating driver mutations in the PRC2 epigenetic repressor complex suggesting epigenetic therapies may represent a specific vulnerability in these tumors. Here, we investigate the role of the LSD1-HDAC1-CoREST (LHC) repressor complex in mediating MPNST tumor growth and progression. Our findings demonstrate that the LHC small molecule inhibitor, corin, induces apoptosis and significantly inhibits proliferation in MPNST cells. Transcriptomic analysis of corin-treated MPNST cells demonstrates specific increases in genes associated with axonogenesis and neuronal differentiation as well as altered extracellular matrix; additionally, corin treatment is shown to inhibit MPNST invasion in vitro. These results underscore the critical role of the LHC complex in facilitating MPNST growth and progression and suggest that targeting the LHC complex represents a promising therapeutic approach for this aggressive malignancy.

6.
J Invest Dermatol ; 144(2): 378-386.e2, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37633457

RESUMO

Wound healing is a complex process involving phases of hemostasis, inflammation, proliferation, and remodeling. The regenerative process in the skin requires coordination between many regulators, including signaling molecules, transcription factors, and the epigenetic machinery. In this study, we show that chromatin regulators HDAC1 and LSD1, key components of the CoREST repressor complex, are upregulated in the regenerating epidermis during wound repair. We also show that corin, a synthetic dual inhibitor of the CoREST complex and HDAC1/LSD1 activities, significantly accelerates wound closure through enhanced re-epithelialization in a mouse tail wound model. Acetylated H3K9 (methylation of histone H3 at lysine 9) expression, a histone modification targeted by HDAC1, is increased in keratinocytes after topical treatment with 100 nM and 1 µM of corin. In vitro experiments demonstrate that corin promotes migration and inhibits the proliferation of human keratinocytes. Furthermore, expression levels of genes promoting keratinocyte migration, such as AREG, CD24, EPHB2, ITGAX, PTGS, SCT1, SERPINB2, SERPINE1, SLPI, SNAI2, and TWIST, increased in keratinocytes treated with corin. These data demonstrate that dual inhibition of class I histone deacetylases and LSD1 by corin may serve as a new approach for promoting wound re-epithelialization and provide a platform for further applications of corin for the treatment of chronic wounds.


Assuntos
Reepitelização , Pele , Camundongos , Animais , Humanos , Pele/lesões , Queratinócitos/metabolismo , Cicatrização/fisiologia , Modelos Animais de Doenças , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Movimento Celular
7.
Cancer Res Commun ; 4(8): 1894-1907, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38994683

RESUMO

SOX10 is a lineage-specific transcription factor critical for melanoma tumor growth; on the other hand, SOX10 loss-of-function drives the emergence of therapy-resistant, invasive melanoma phenotypes. A major challenge has been developing therapeutic strategies targeting SOX10's role in melanoma proliferation while preventing a concomitant increase in tumor cell invasion. In this study, we report that the lysine acetyltransferase (KAT) EP300 and SOX10 gene loci on chromosome 22 are frequently co-amplified in melanomas, including UV-associated and acral tumors. We further show that p300 KAT activity mediates SOX10 protein stability and that the p300 inhibitor A-485 downregulates SOX10 protein levels in melanoma cells via proteasome-mediated degradation. Additionally, A-485 potently inhibits proliferation of SOX10+ melanoma cells while decreasing invasion in AXLhigh/MITFlow melanoma cells through downregulation of metastasis-related genes. We conclude that the SOX10/p300 axis is critical to melanoma growth and invasion and that inhibition of p300 KAT activity through A-485 may be a worthwhile therapeutic approach for SOX10-reliant tumors. SIGNIFICANCE: The p300 KAT inhibitor A-485 blocks SOX10-dependent proliferation and SOX10-independent invasion in hard-to-treat melanoma cells.


Assuntos
Proliferação de Células , Proteína p300 Associada a E1A , Melanoma , Fatores de Transcrição SOXE , Humanos , Melanoma/patologia , Melanoma/genética , Melanoma/metabolismo , Fatores de Transcrição SOXE/metabolismo , Fatores de Transcrição SOXE/genética , Proteína p300 Associada a E1A/metabolismo , Proteína p300 Associada a E1A/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Estabilidade Proteica , Animais , Compostos Heterocíclicos de 4 ou mais Anéis
8.
J Invest Dermatol ; 2024 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-39009280

RESUMO

Keloids are pathological fibroproliferative scars resulting from abnormal collagen deposition within and beyond the margins of the initial cutaneous insult. Keloids negatively impact QOL functionally and cosmetically, with current treatment modalities unsatisfactory. Recent studies indicate that epigenetic dysregulation is central to the development and progression of keloids. In this study, we evaluate the functional significance of epigenetic targeting strategies in vitro using patient-derived keloid fibroblasts treated with small-molecule inhibitors of histone deacetylases, LSD1, CoREST, and p300, as potential therapies for keloids. We find that both the dual-acting CoREST inhibitor corin and the histone deacetylase inhibitor entinostat reduce fibroblast proliferation more than the LSD1 inhibitor GSK-LSD1; in addition, corin was the most effective inhibitor of migration and invasion across keloid fibroblasts. RNA-sequencing analysis of keloid fibroblasts treated with corin demonstrates coordinate upregulation of many genes, including key mediators of cell adhesion such as claudins. Corin also downregulates gene sets involved in cell cycle progression, including reduced expression of cyclins A1 and B2 compared with that of DMSO. These results highlight a significant role for epigenetic regulation of pathologic mediators of keloidal scarring and suggest that inhibitors of the epigenetic CoREST repressor complex may prove beneficial in the prevention and/or treatment of keloidal scarring in patients.

9.
bioRxiv ; 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38469149

RESUMO

SOX10 is a lineage-specific transcription factor critical for melanoma tumor growth, while SOX10 loss-of-function drives the emergence of therapy-resistant, invasive melanoma phenotypes. A major challenge has been developing therapeutic strategies targeting SOX10's role in melanoma proliferation, while preventing a concomitant increase in tumor cell invasion. Here, we report that the lysine acetyltransferase (KAT) EP300 and SOX10 gene loci on Chromosome 22 are frequently co-amplified in melanomas, including UV-associated and acral tumors. We further show that p300 KAT activity mediates SOX10 protein stability and that the p300 inhibitor, A-485, downregulates SOX10 protein levels in melanoma cells via proteasome-mediated degradation. Additionally, A-485 potently inhibits proliferation of SOX10+ melanoma cells while decreasing invasion in AXLhigh/MITFlow melanoma cells through downregulation of metastasis-related genes. We conclude that the SOX10/p300 axis is critical to melanoma growth and invasion, and that inhibition of p300 KAT activity through A-485 may be a worthwhile therapeutic approach for SOX10-reliant tumors.

10.
J Clin Invest ; 134(6)2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38300709

RESUMO

Virtually all patients with BRAF-mutant melanoma develop resistance to MAPK inhibitors largely through nonmutational events. Although the epigenetic landscape is shown to be altered in therapy-resistant melanomas and other cancers, a specific targetable epigenetic mechanism has not been validated. Here, we evaluated the corepressor for element 1-silencing transcription factor (CoREST) epigenetic repressor complex and the recently developed bivalent inhibitor corin within the context of melanoma phenotype plasticity and therapeutic resistance. We found that CoREST was a critical mediator of the major distinct melanoma phenotypes and that corin treatment of melanoma cells led to phenotype reprogramming. Global assessment of transcript and chromatin changes conferred by corin revealed specific effects on histone marks connected to epithelial-mesenchymal transition-associated (EMT-associated) transcription factors and the dual-specificity phosphatases (DUSPs). Remarkably, treatment of BRAF inhibitor-resistant (BRAFi-R) melanomas with corin promoted resensitization to BRAFi therapy. DUSP1 was consistently downregulated in BRAFi-R melanomas, which was reversed by corin treatment and associated with inhibition of p38 MAPK activity and resensitization to BRAFi therapies. Moreover, this activity was recapitulated by the p38 MAPK inhibitor BIRB 796. These findings identify the CoREST repressor complex as a central mediator of melanoma phenotype plasticity and resistance to targeted therapy and suggest that CoREST inhibitors may prove beneficial for patients with BRAFi-resistant melanoma.


Assuntos
Melanoma , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Correpressoras/genética , Resistencia a Medicamentos Antineoplásicos/genética , Linhagem Celular Tumoral , Inibidores de Proteínas Quinases/farmacologia , Fenótipo , Proteínas Quinases p38 Ativadas por Mitógeno
11.
Carcinogenesis ; 34(1): 86-92, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23027618

RESUMO

The hypothesis that germ-line polymorphisms in DNA repair genes influence cancer risk has previously been tested primarily on a cancer site-specific basis. The purpose of this study was to test the hypothesis that DNA repair gene allelic variants contribute to globally elevated cancer risk by measuring associations with risk of all cancers that occurred within a population-based cohort. In the CLUE II cohort study established in 1989 in Washington County, MD, this study was comprised of all 3619 cancer cases ascertained through 2007 compared with a sample of 2296 with no cancer. Associations were measured between 759 DNA repair gene single nucleotide polymorphisms (SNPs) and risk of all cancers. A SNP in O(6)-methylguanine-DNA methyltransferase, MGMT, (rs2296675) was significantly associated with overall cancer risk [per minor allele odds ratio (OR) 1.30, 95% confidence interval (CI) 1.19-1.43 and P-value: 4.1 × 10(-8)]. The association between rs2296675 and cancer risk was stronger among those aged ≤54 years old than those who were ≥55 years at baseline (P-for-(interaction) = 0.021). OR were in the direction of increased risk for all 15 categories of malignancies studied (P < 0.0001), ranging from 1.22 (P = 0.42) for ovarian cancer to 2.01 (P = 0.008) for urinary tract cancers; the smallest P-value was for breast cancer (OR 1.45, P = 0.0002). The results indicate that the minor allele of MGMT SNP rs2296675, a common genetic marker with 37% carriers, was significantly associated with increased risk of cancer across multiple tissues. Replication is needed to more definitively determine the scientific and public health significance of this observed association.


Assuntos
Reparo do DNA/genética , Neoplasias/genética , Adulto , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Maryland/epidemiologia , Neoplasias/epidemiologia , Vigilância da População , Fatores de Risco
12.
Epigenomics ; 15(3): 167-187, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-37020393

RESUMO

Epigenetics encompasses heritable, reversible gene expression patterns that do not arise from mutations in genomic DNA but, rather, are regulated by DNA methylation, histone modifications, RNA modifications and ncRNAs; and epigenetic dysregulation is increasingly recognized as a mechanism of neoplastic disease progression as well as resistance to cancer therapy. This review article focuses on epigenetic modifications implicated in the progression and therapeutic resistance of common cutaneous malignancies, including basal cell carcinoma, squamous cell carcinoma, T-cell lymphoma and malignant melanoma, with an emphasis on therapeutic strategies that may be used to target such disease-associated alterations.


Epigenetics involves the study of how genes can be turned on or off by factors that affect how these genes are packaged and regulated. In cancer, there are often epigenetic changes that contribute to the formation of tumors. Many of these epigenetic changes, some of which can be passed down through generations, increase the risk of skin cancers such as basal cell carcinoma, squamous cell carcinoma, T-cell lymphoma and malignant melanoma. Emerging therapies designed to target these epigenetic changes may be effective treatments for these types of skin cancers. Researchers are currently investigating how to best use these therapies to help the ever-increasing number of people with skin cancer.


Assuntos
Carcinoma Basocelular , Melanoma , Neoplasias Cutâneas , Humanos , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/terapia , Neoplasias Cutâneas/patologia , Melanoma/genética , Epigênese Genética , Carcinoma Basocelular/genética , Carcinoma Basocelular/patologia , Metilação de DNA
13.
Melanoma Res ; 33(4): 283-292, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37276030

RESUMO

Melanoma is a highly aggressive form of skin cancer and the most frequent lethal malignancy diagnosed by dermatologists. Although there have been advances for predicting melanoma prognosis, there are few highly sensitive and specific diagnostic tools for clinically evaluating suspicious melanocytic lesions prior to biopsy. We have recently determined that alterations in cellular lipid and pigment content are associated with tumor progression and melanoma metastasis. Here, we seek to determine if lipid droplet and pigment content assessments near the skin's surface are able to distinguish benign from malignant melanocytic lesions. We obtained 14 benign melanocytic lesions, classified as Melanocytic Pathology Assessment Tool and Hierarchy for Diagnosis (MPATH-Dx) class 1, and 22 malignant melanomas, classified as MPATH-Dx class 4 or 5, from Boston Medical Center. The malignant melanomas had an average greatest thickness of 1.8 ±â€…2.1 mm with 7/22 biopsies showing the presence of ulceration. Tissues were stained with the Fontana Masson stain to detect pigment or immunohistochemically stained for adipophilin, the main protein component of lipid droplets, to detect lipid droplets. Pigment and lipid droplets were quantified using ImageJ and CellProfiler, respectively. We found no significant difference in total pigment area between benign melanocytic lesions and malignant melanoma, and a 66% decrease in lipid content and 68% reduction in lipid/pigment content between benign melanocytic lesions and malignant melanoma ( P  < 0.05). Our results suggest that lipid content and lipid/pigment content ratios may distinguish benign and malignant melanocytic lesions, which may be useful as a diagnostic tool for histopathologically challenging pigmented lesions.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Melanoma/diagnóstico , Melanoma/patologia , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/patologia , Melanócitos/patologia , Prognóstico , Lipídeos
14.
Carcinogenesis ; 33(9): 1692-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22581838

RESUMO

For unknown reasons, non-melanoma skin cancer (NMSC) is associated with increased risk of other malignancies. Focusing solely on DNA repair or DNA repair-related genes, this study tested the hypothesis that DNA repair gene variants contribute to the increased cancer risk associated with a personal history of NMSC. From the parent CLUE II cohort study, established in 1989 in Washington County, MD, the study consisted of a cancer-free control group (n 5 2296) compared with three mutually exclusive groups of cancer cases ascertained through 2007: (i) Other (non-NMSC) cancer only (n 5 2349); (ii) NMSC only (n 5 694) and (iii) NMSC plus other cancer (n 5 577). The frequency of minor alleles in 759 DNA repair gene single nucleotide polymorphisms (SNPs) was compared in these four groups. Comparing those with both NMSC and other cancer versus those with no cancer, 10 SNPs had allelic trend P-values <0.01. The two top-ranked SNPs were both within the thymine DNA glycosylase gene (TDG). One was a non-synonymous coding SNP (rs2888805) [per allele odds ratio (OR) 1.40, 95% confidence interval (CI) 1.16-1.70; P-value 5 0.0006] and the other was an intronic SNP in high linkage disequilibrium with rs2888805 (rs4135150). None of the associations had a P-value <6.6310(-5), the threshold for statistical significance after correcting for multiple comparisons. The results pinpoint DNA repair genes most likely to contribute to the NMSC cancer-prone phenotype. A promising lead is genetic variants in TDG, important not only in base excision repair but also in regulating the epigenome and gene expression, which may contribute to the NMSC-associated increase in overall cancer risk.


Assuntos
Reparo do DNA/genética , Polimorfismo de Nucleotídeo Único , Neoplasias Cutâneas/genética , Timina DNA Glicosilase/genética , Adulto , Idoso , Biomarcadores Tumorais , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo
15.
Cancer Cell ; 3(3): 199-200, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12676576

RESUMO

The formation of a blood supply is critical for tumor growth and metastasis; however, understanding the relationship of cellular transformation to tumor angiogenesis has been limited by the multifactorial nature of both processes. In this issue of Cancer Cell, Watnick and colleagues use a genetically defined tumor model system to determine the link between ras, myc, and angiogenesis and identify Thrombospondin-1 as being the critical regulator of angiogenesis in this system (Watnick et al., 2003).


Assuntos
Neovascularização Patológica/fisiopatologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Trombospondina 1/metabolismo , Proteínas ras/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Modelos Biológicos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Trombospondina 1/genética
16.
Cancer Cell ; 3(6): 525-30, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12842081

RESUMO

Since the gene encoding Id1 was cloned in 1990, Id proteins have been implicated in regulating a variety of cellular processes, including cellular growth, senescence, differentiation, apoptosis, angiogenesis, and neoplastic transformation. The development of knockout and transgenic animal models for many members of the Id gene family has been particularly useful in sorting out the biologic relevance of these genes and their expression during normal development, malignant transformation, and tumor progression. Here we review the current understanding of Id gene function, the biologic consequences of Id gene expression, and the implications for Id gene regulation of cell growth and tumorigenesis.


Assuntos
Neoplasias/metabolismo , Proteínas Repressoras , Fatores de Transcrição/metabolismo , Animais , Apoptose , Ciclo Celular , Diferenciação Celular , Divisão Celular/fisiologia , Transformação Celular Neoplásica , Senescência Celular , Regulação da Expressão Gênica , Sequências Hélice-Alça-Hélice , Humanos , Proteína 1 Inibidora de Diferenciação , Camundongos
17.
Cancer Cell ; 4(4): 291-9, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14585356

RESUMO

Id genes regulate tumor angiogenesis and loss of Id1 inhibits tumor xenograft growth in mice. Here we evaluate the role of Id1 in a more clinically relevant tumor model system using a two-step chemical carcinogenesis protocol. Remarkably, we find that Id1-/- mice are more susceptible to skin tumorigenesis compared to their wild-type counterparts. Cutaneous neoplasms in Id1-/- mice show increased proliferation without alterations in tumor angiogenesis; however, Id1-/- mice possess 50% fewer cutaneous gammadelta T cells than their wild-type counterparts due to an intrinsic migration defect associated with loss of expression of the chemokine receptor, CXCR4. We suggest that there are important differences between the mechanisms of angiogenesis in transplanted and autochthonous tumors and that these findings will have significant implications for the potential utility of antiangiogenic therapies in cancer.


Assuntos
Melanoma/metabolismo , Neovascularização Patológica/fisiopatologia , Proteínas Repressoras , Neoplasias Cutâneas/fisiopatologia , Fatores de Transcrição/metabolismo , Transplante Heterólogo , Animais , Carcinógenos/toxicidade , Endotélio Vascular/fisiopatologia , Citometria de Fluxo , Proteína 1 Inibidora de Diferenciação , Melanoma/induzido quimicamente , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/fisiopatologia , Receptores CXCR4/metabolismo , Neoplasias Cutâneas/induzido quimicamente , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Transcrição/genética
18.
Cancer Cell ; 2(6): 473-83, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12498716

RESUMO

Id proteins are helix-loop-helix transcription factors that regulate tumor angiogenesis. In order to identify downstream effectors of Id1 involved in the regulation of angiogenesis, we performed PCR-select subtractive hybridization on wild-type and Id1 knockout mouse embryo fibroblasts (MEFs). Here we demonstrate that thrombospondin-1 (TSP-1), a potent inhibitor of angiogenesis, is a target of transcriptional repression by Id1. We also show that Id1-null MEFs secrete an inhibitor of endothelial cell migration, which is completely inactivated by depletion of TSP-1. Furthermore, in vivo studies revealed decreased neovascularization in matrigel assays in Id1-null mice compared to their wild-type littermates. This decrease was completely reversed by a TSP-1 neutralizing antibody. We conclude that TSP-1 is a major target for Id1 effects on angiogenesis.


Assuntos
Regulação da Expressão Gênica , Neovascularização Patológica/genética , Proteínas Repressoras , Trombospondina 1/genética , Fatores de Transcrição/fisiologia , Animais , Bovinos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Elementos E-Box , Embrião de Mamíferos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Fibroblastos/fisiologia , Humanos , Imuno-Histoquímica , Proteína 1 Inibidora de Diferenciação , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Trombospondina 1/antagonistas & inibidores , Trombospondina 1/biossíntese , Trombospondina 1/farmacologia , Fatores de Transcrição/deficiência , Transcrição Gênica , Regulação para Cima
19.
Clin Rev Allergy Immunol ; 63(3): 447-471, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36346551

RESUMO

Epigenetics is the study of heritable, reversible gene expression patterns that do not originate from alterations in the DNA sequence. Epigenetic modifications influence gene expression patterns and include DNA methylation, histone modifications, and gene regulation via non-coding RNAs. While the study of epigenetics has been most broadly applied to neoplastic diseases, the role of the epigenome in a wide range of disease processes including autoimmune, allergic, and inflammatory processes is increasingly being recognized. Recent advances in the study of the epigenome have led to novel insights into the pathogenesis and potential therapeutic targets of various pathologic entities including inflammatory diseases. In this review, we examine the nature of epigenetic modifications in several well-studied autoimmune, allergic, and/or inflammatory disorders of the skin including systemic lupus erythematosus, vitiligo, systemic sclerosis, alopecia areata, pemphigus, psoriasis, atopic dermatitis, keloidal scarring, and hidradenitis suppurativa with the aim to determine how such epigenetic changes may be targeted for therapeutic benefit.


Assuntos
Alopecia em Áreas , Psoríase , Humanos , Epigenômica , Epigênese Genética , Pele , Psoríase/genética
20.
JID Innov ; 2(2): 100090, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35199090

RESUMO

This past decade has seen tremendous advances in understanding the molecular pathogenesis of melanoma and the development of novel effective therapies for melanoma. Targeted therapies and immunotherapies that extend survival of patients with advanced disease have been developed; however, the vast majority of patients experience relapse and therapeutic resistance over time. Moreover, cellular plasticity has been demonstrated to be a driver of therapeutic resistance mechanisms in melanoma and other cancers, largely functioning through epigenetic mechanisms, suggesting that targeting of the cancer epigenetic landscape may prove a worthwhile endeavor to ensure durable treatment responses and cures. Here, we review the epigenetic alterations that characterize melanoma development, progression, and resistance to targeted therapies as well as epigenetic therapies currently in use and under development for melanoma and other cancers. We further assess the landscape of epigenetic therapies in clinical trials for melanoma and provide a framework for future advances in epigenetic therapies to circumvent the development of therapeutic resistance in melanoma.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA