Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 3732, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35768444

RESUMO

Melanoma is commonly driven by activating mutations in the MAP kinase BRAF; however, oncogenic BRAF alone is insufficient to promote melanomagenesis. Instead, its expression induces a transient proliferative burst that ultimately ceases with the development of benign nevi comprised of growth-arrested melanocytes. The tumor suppressive mechanisms that restrain nevus melanocyte proliferation remain poorly understood. Here we utilize cell and murine models to demonstrate that oncogenic BRAF leads to activation of the Hippo tumor suppressor pathway, both in melanocytes in vitro and nevus melanocytes in vivo. Mechanistically, we show that oncogenic BRAF promotes both ERK-dependent alterations in the actin cytoskeleton and whole-genome doubling events, which independently reduce RhoA activity to promote Hippo activation. We also demonstrate that functional impairment of the Hippo pathway enables oncogenic BRAF-expressing melanocytes to bypass nevus formation and rapidly form melanomas. Our data reveal that the Hippo pathway enforces the stable arrest of nevus melanocytes and represents a critical barrier to melanoma development.


Assuntos
Melanoma , Nevo , Neoplasias Cutâneas , Animais , Melanócitos/metabolismo , Melanoma/patologia , Camundongos , Mutação , Nevo/genética , Nevo/metabolismo , Nevo/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Neoplasias Cutâneas/patologia
2.
Pigment Cell Melanoma Res ; 35(1): 52-65, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34468072

RESUMO

Yes-associated protein 1 (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are transcriptional coactivators that have been implicated in driving metastasis and progression in many cancers, mainly through their transcriptional regulation of downstream targets. Although YAP and TAZ have shown redundancy in many contexts, it is still unknown whether or not this is true in melanoma. Here, we show that while both YAP and TAZ are expressed in a panel of melanoma cell lines, depletion of YAP results in decreased cell numbers, focal adhesions, and the ability to invade matrigel. Using non-biased RNA-sequencing analysis, we find that melanoma cells depleted of YAP, TAZ, or YAP/TAZ exhibit drastically different transcriptomes. We further uncover the ARP2/3 subunit ARPC5 as a specific target of YAP but not TAZ and that ARPC5 is essential for YAP-dependent maintenance of melanoma cell focal adhesion numbers. Our findings suggest that in melanoma, YAP drives melanoma progression, survival, and invasion.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Melanoma/metabolismo , Neoplasias Cutâneas/metabolismo , Proteínas de Sinalização YAP/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Adesões Focais/genética , Adesões Focais/metabolismo , Adesões Focais/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/genética , Melanoma/patologia , Invasividade Neoplásica , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional/genética , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional/metabolismo , Transcriptoma , Proteínas de Sinalização YAP/genética
3.
Cancer Res ; 81(8): 2071-2085, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33526511

RESUMO

The failure of once promising target-specific therapeutic strategies often arises from redundancies in gene expression pathways. Even with new melanoma treatments, many patients are not responsive or develop resistance, leading to disease progression in terms of growth and metastasis. We previously discovered that the transcription factors ETS1 and PAX3 drive melanoma growth and metastasis by promoting the expression of the MET receptor. Here, we find that there are multiple ETS family members expressed in melanoma and that these factors have redundant functions. The small molecule YK-4-279, initially developed to target the ETS gene-containing translocation product EWS-FLI1, significantly inhibited cellular growth, invasion, and ETS factor function in melanoma cell lines and a clinically relevant transgenic mouse model, BrafCA;Tyr-CreERT2;Ptenf/f. One of the antitumor effects of YK-4-279 in melanoma is achieved via interference of multiple ETS family members with PAX3 and the expression of the PAX3-ETS downstream gene MET. Expression of exogenous MET provided partial rescue of the effects of YK-4-279, further supporting that MET loss is a significant contributor to the antitumor effects of the drug. This is the first study identifying multiple overlapping functions of the ETS family promoting melanoma. In addition, targeting all factors, rather than individual members, demonstrated impactful deleterious consequences in melanoma progression. Given that multiple ETS factors are known to have oncogenic functions in other malignancies, these findings have a high therapeutic impact. SIGNIFICANCE: These findings identify YK-4-279 as a promising therapeutic agent against melanoma by targeting multiple ETS family members and blocking their ability to act as transcription factors.


Assuntos
Indóis/farmacologia , Melanoma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-ets/antagonistas & inibidores , Neoplasias Cutâneas/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Progressão da Doença , Humanos , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Transgênicos , Invasividade Neoplásica , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Fator de Transcrição PAX3/antagonistas & inibidores , Fator de Transcrição PAX3/metabolismo , Proteína Proto-Oncogênica c-ets-1/antagonistas & inibidores , Proteína Proto-Oncogênica c-ets-1/metabolismo , Proteína Proto-Oncogênica c-fli-1/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Proteína EWS de Ligação a RNA/antagonistas & inibidores , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo
4.
Cancers (Basel) ; 14(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35008307

RESUMO

More options are needed for the effective treatment of melanoma. In a previous study, we discovered the small molecule drug YK-4-279 almost completely inhibited tumor progression in the BrafCA;Tyr-CreERT2;Ptenflox/flox transgenic mouse model. YK-4-279 had no effect on tumor initiation but blocked progression of invasive melanoma. Our current study was designed as a treatment model, where YK-4-279 was administered during pigmented lesion formation. The study design included the use of three groups: (1) a control group that received only DMSO without a drug (MOCK), (2) mice following our prior studies with YK-4-279 administered at the time of tumor induction (YK-4-279), and (3) mice treated during tumor initiation (YK-4-279 delay). While the MOCK mice had progression of tumors, both YK-4-279 and YK-4-279 delay groups had a significant block or delay of progression. The majority of mice in the YK-4-279 groups had a block of progression, while the YK-4-279 delay group had either a partial block (60% in male mice or 29% in females) or a delay in disease progression in females (28 days in controls to 50 days in YK-4-279 delay group). Here, we demonstrate that YK-4-279 has a significant impact on blocking or delaying tumor progression in a pre-clinical treatment model of melanoma.

6.
J Cancer ; 10(1): 1-10, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30662519

RESUMO

Yes Associated Protein 1 (YAP) and Transcriptional coactivator with PDZ-Binding Motif (TAZ) have gained notoriety for their ability to drive tumor initiation and progression in a wide variety of cancers, including melanoma. YAP and TAZ act as drivers of melanoma through its interaction with the TEAD family of transcription factors. Verteporfin is a benzoporphyrin derivative that is used clinically for photodynamic treatment of macular degeneration. Recently it has emerged as a potential inhibitor of YAP/TAZ-TEAD interaction independent of light activation. In this study we determine if verteporfin has clinical potential by testing this compound on human melanoma cell cultures and in a clinically significant mouse model, BrafCA; Tyr-CreERT2; Ptenf/f, which parallels human melanoma in terms of disease progression, genetics, and histopathology. In culture, Verteporfin treatment induces a rapid drop in YAP and TAZ protein levels and cell numbers. In the transgenic model, utilizing drug levels that correspond to previously determined safe doses in human patients and with a dosing regimen calculated in this study, Verteporfin did not inhibit melanoma initiation or progression in comparison to mock treated controls. Taken together, our study suggests that although Verteporfin induces YAP/TAZ degradation in melanoma cell lines, Verteporfin was not effective as a YAP/TAZ-TEAD specific inhibitor of melanoma in our studies that aimed to mimic conditions found in clinic in terms of treatment regimen and disease model.

7.
Oncol Lett ; 13(6): 4027-4034, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28588695

RESUMO

Pancreatic cancer is a lethal disease with a propensity for invading and metastasizing into the surrounding tissues, including the liver and intestines. A number of factors are aberrantly overexpressed in this tumor type and actively promote cancer progression and metastasis. The present study demonstrates that paired box transcription factor 6 (PAX6) and C-X-C chemokine receptor 4 (CXCR4) are frequently co-expressed in primary pancreatic adenocarcinoma tumors and established cell lines. Expression analysis methods used in the present study included evaluation of protein expression by western blot analysis and immunofluorescence, transcript expression levels by reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and luciferase assays utilizing regulatory elements from the CXCR4 gene locus. Canonical PAX6 and alternative splice variant PAX6(5a) proteins are expressed in pancreatic cancer and can drive gene expression through a conserved enhancer element within the first intron of the CXCR4 gene. As demonstrated by the introduction of an exogenous reporter construct with or without the intronic enhancer, loss of this element inhibited gene expression within numerous pancreatic cancer cell lines including Panc1, MIA-PaCa2 and BxPC3. All of the pancreatic cancer cell lines expressed the canonical CXCR4B transcript in addition to the alternatively spliced variant CXCR4A as determined by RT-qPCR experiments. The discovery of variant transcripts in pancreatic cancer cells may provide new candidates for future targeted therapies.

8.
J Cell Biochem ; 117(2): 533-41, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26252164

RESUMO

Several key transcription factors regulate cell growth, survival, and differentiation during neural crest and melanoblast development in the embryo, and these same pathways may be reactivated in tumors arising from the progenitors of these cells. The transcription factors PAX3 and FOXD3 have essential roles in melanoblasts and melanoma. In this study, we define a regulatory pathway where FOXD3 promotes the expression of PAX3. Both factors are expressed in melanoma cells and there is a positive correlation between the transcript levels of PAX3 and FOXD3. The PAX3 gene contains two FOX binding motifs within highly conserved enhancer regulatory elements that are essential for neural crest development. FOXD3 binds to both of these motifs in vitro but only one of these sites is preferentially utilized in melanoma cells. Overexpression of FOXD3 upregulates PAX3 levels while inhibition of FOXD3 function does not alter PAX3 protein levels, supporting that FOXD3 is sufficient but not necessary to drive PAX3 expression in melanoma cells. Here, we identify a molecular pathway where FOXD3 upregulates PAX3 expression and therefore contributes to melanoma progression.


Assuntos
Fatores de Transcrição Forkhead/fisiologia , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Box Pareados/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Sequência Conservada , Expressão Gênica , Humanos , Melanoma , Dados de Sequência Molecular , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/genética , Regiões Promotoras Genéticas , Ativação Transcricional
9.
J Biol Chem ; 290(36): 21901-14, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26205821

RESUMO

Metastatic melanoma is an aggressive and deadly disease. The chemokine receptor CXCR4 is active in melanoma metastasis, although the mechanism for the promotion and maintenance of CXCR4 expression in these cells is mostly unknown. Here, we find melanoma cells express two CXCR4 isoforms, the common version and a variant that is normally restricted to cells during development or to mature blood cells. CXCR4 expression is driven through a highly conserved intronic enhancer element by the transcription factors PAX3 and FOXD3. Inhibition of these transcription factors slows melanoma cell growth, migration, and motility, as well as reduces CXCR4 expression. Overexpression of these transcription factors drives the production of increased CXCR4 levels. Loss of PAX3 and FOXD3 transcription factor activity results in a reduction in cell motility, migration, and chemotaxis, all of which are rescued by CXCR4 overexpression. Here, we discover a molecular pathway wherein PAX3 and FOXD3 promote CXCR4 gene expression in melanoma.


Assuntos
Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Box Pareados/genética , Receptores CXCR4/genética , Células 3T3 , Animais , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/genética , Elementos Facilitadores Genéticos/genética , Fatores de Transcrição Forkhead/metabolismo , Células HEK293 , Humanos , Íntrons/genética , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/metabolismo , Ligação Proteica , Receptores CXCR4/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Clin Dermatol ; 31(2): 166-78, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23438380

RESUMO

Melanocyte stem cells differ greatly from melanoma stem cells; the former provide pigmented cells during normal tissue homeostasis and repair, and the latter play an active role in a lethal form of cancer. These 2 cell types share several features and can be studied by similar methods. Aspects held in common by both melanocyte stem cells and melanoma stem cells include their expression of shared biochemical markers, a system of similar molecular signals necessary for their maintenance, and a requirement for an ideal niche microenvironment for providing these factors. This review provides a perspective of both these cell types and discusses potential models of stem cell growth and propagation. Recent findings provide a strong foundation for the development of new therapeutics directed at isolating and manipulating melanocyte stem cells for tissue engineering or at targeting and eradicating melanoma specifically, while sparing nontumor cells.


Assuntos
Melanócitos/patologia , Melanoma/patologia , Neoplasias Cutâneas/patologia , Células-Tronco/patologia , Diferenciação Celular , Humanos , Melanócitos/metabolismo , Melanoma/metabolismo , Modelos Biológicos , Neoplasias Cutâneas/metabolismo , Células-Tronco/metabolismo , Engenharia Tecidual/métodos
11.
Mol Cancer Res ; 10(8): 1065-76, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22679108

RESUMO

GSK-3 is a serine/threonine kinase involved in a diverse range of cellular processes. GSK-3 exists in two isoforms, GSK-3α and GSK-3ß, which possess some functional redundancy but also play distinct roles depending on developmental and cellular context. In this article, we found that GSK-3 actively promoted cell growth and survival in melanoma cells, and blocking this activity with small-molecule inhibitor SB216763 or gene-specific siRNA decreased proliferation, increased apoptosis, and altered cellular morphology. These alterations coincided with loss of PAX3, a transcription factor implicated in proliferation, survival, and migration of developing melanoblasts. We further found that PAX3 directly interacted with and was phosphorylated in vitro on a number of residues by GSK-3ß. In melanoma cells, direct inhibition of PAX3 lead to cellular changes that paralleled the response to GSK-3 inhibition. Maintenance of PAX3 expression protected melanoma cells from the anti-tumor effects of SB216763. These data support a model wherein GSK-3 regulates proliferation and morphology of melanoma through phosphorylation and increased levels of PAX3.


Assuntos
Quinase 3 da Glicogênio Sintase , Melanoma , Fatores de Transcrição Box Pareados , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Melanoma/genética , Melanoma/metabolismo , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Interferência de RNA , Transdução de Sinais
12.
Pigment Cell Melanoma Res ; 23(2): 225-37, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20067553

RESUMO

Melanoma is a cancer with a poorly understood molecular pathobiology. We find the transcription factors PAX3, SOX10, MITF, and the tyrosine kinase receptor MET expressed in melanoma cell lines and primary tumors. Analysis for MET expression in primary tumor specimens showed 27/40 (68%) of the samples displayed an increased expression of MET, and this expression was highly correlated with parallel expression of PAX3, SOX10, and MITF. PAX3 and MITF bind to elements in the MET promoter independently, without evidence of either synergistic activation or inhibition. SOX10 does not directly activate the MET gene alone, but can synergistically activate MET expression with either PAX3 or MITF. In melanoma cells, there was evidence of two pathways for PAX3 mediated MET induction: (i) direct activation of the gene, and (ii) indirect regulation through MITF. SK-MEL23 melanoma cells have both of these pathways intact, while SK-MEL28 melanoma cells only have the first pathway. In summary, we find that PAX3, SOX10 and MITF play an active role in melanoma cells by regulating the MET gene. In consequence, MET promotes the melanoma cancer phenotype by promoting migration, invasion, resistance to apoptosis, and tumor cell growth.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Proto-Oncogênicas c-met/biossíntese , Fatores de Transcrição SOXE/metabolismo , Apoptose , Linhagem Celular Tumoral , Progressão da Doença , Humanos , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/biossíntese , Proteínas Proto-Oncogênicas c-met/metabolismo , RNA/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Fatores de Transcrição SOXE/biossíntese
13.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1214-22, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19779014

RESUMO

GATA5 is a member of the GATA zinc finger transcription factor family involved in tissue-specific transcriptional regulation during cell differentiation and embryogenesis. Previous reports indicate that null mutation of the zebrafish GATA5 gene results in embryonic lethality, whereas deletion of exon 1 from the mouse GATA5 gene causes only derangement of female urogenital development. Here, we have identified an alternate promoter within intron 1 of the mouse GATA5 gene that transcribes a 2.5-kb mRNA that lacks exon 1 entirely but includes 82 bp from intron 1 and all of exons 2-6. The alternative promoter was active during transient transfection in cultured airway myocytes and bronchial epithelial cells, and it drove reporter gene expression in gastric epithelial cells in transgenic mice. The 2.5-kb alternative transcript encodes an NH(2)-terminally truncated "short GATA5" comprising aa 226-404 with a single zinc finger, which retains ability to transactivate the atrial natriuretic factor promoter (albeit less efficiently than full-length GATA5). Another new GATA5 transcript contains all of exons 1-5 and the 5' portion of exon 6 but lacks the terminal 1143 bp of the 3'-untranslated region from exon 6. These findings extend current understanding of the tissue distribution of GATA5 expression and suggests that GATA5 expression and function are more complex than previously appreciated.


Assuntos
Processamento Alternativo , Fator de Transcrição GATA5/genética , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Regiões 3' não Traduzidas , Região 5'-Flanqueadora , Animais , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Northern Blotting , Cães , Células Epiteliais/metabolismo , Éxons , Fator de Transcrição GATA5/metabolismo , Mucosa Gástrica/metabolismo , Genes Reporter , Íntrons , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miócitos de Músculo Liso/metabolismo , Células NIH 3T3 , Isoformas de Proteínas , Técnica de Amplificação ao Acaso de DNA Polimórfico , Mucosa Respiratória/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sítio de Iniciação de Transcrição , Ativação Transcricional , Transfecção
14.
J Biol Chem ; 284(40): 27524-32, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19651775

RESUMO

Tumors of the exocrine pancreas have a poor prognosis. Several proteins are overexpressed in this cancer type, including the MET tyrosine kinase receptor and the transcription factor PAX6. In this report, we find that PAX6(5a), an alternately spliced variant form of PAX6, is expressed in pancreatic carcinoma cell lines at higher levels than the canonical PAX6 protein. Both protein forms of PAX6 bind directly to an enhancer element in the MET promoter and activate the expression of the MET gene. In addition, inhibition of PAX6 transcripts leads to a decline in cell growth and survival, differentiation, and a concurrent reduction of MET protein expression. These data support a model for a neoplastic pathway, where expression of a transcription factor from development activates the MET receptor, a protein that has been directly linked to protumorigenic processes of resisting apoptosis, tumor growth, invasion, and metastasis.


Assuntos
Progressão da Doença , Proteínas do Olho/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Fatores de Transcrição Box Pareados/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas/genética , Receptores de Fatores de Crescimento/genética , Proteínas Repressoras/genética , Ativação Transcricional , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Apoptose/genética , Sequência de Bases , Linhagem Celular Tumoral , Genes Reporter , Humanos , Camundongos , Dados de Sequência Molecular , Mutação , Metástase Neoplásica/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/deficiência , Fenótipo , Proteínas Proto-Oncogênicas c-met
15.
Lab Invest ; 89(3): 301-14, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19139719

RESUMO

PAX5 is a nuclear transcription factor required for B cell development, and its expression was evaluated in upper aerodigestive malignancies and pancreatic cancer by immunoblotting. The PAX5 protein expression was relatively strong in small-cell lung cancer (SCLC, 11/12); however, its expression was not detected in non-SCLC (NSCLC, n=13), mesothelioma (n=7), pancreatic (n=6), esophageal (n=6) and head and neck cancer cell lines (n=12). In comparison, PAX8 and PAX3 expressions were absent or non-detectable in SCLC cell lines; however, PAX8 was expressed in most of the tested NSCLC cell lines (13/13) and also frequently in all the other cell lines. We also detected frequent expressions of PAX2 and PAX9 protein in the various cell lines. Utilizing neuroendocrine tumor samples, we found that the frequency as well as the average intensity of the expression of PAX5 increased from pulmonary carcinoid (9%, moderate and strong PAX5 expression, n=44), to large-cell neuroendocrine carcinoma (LCNC, 27%, n=11) to SCLC (33%, n=76). FISH analysis revealed no translocations of the PAX5 gene, but polyploidy in some SCLC tumor tissues (6/37). We determined that PAX5 could regulate the transcription of c-Met using luciferase-coupled reporter and chromatin immunoprecipitation analysis. In addition, the phospho-c-Met (active form) and PAX5 were both localized to the same intra-nuclear compartment in hepatocyte growth factor treated SCLC cells and interacted with each other. Finally, we determined the therapeutic translational potential of PAX5 using PAX5 knockdown SCLC cells in conjunction with Topoisomerase 1 (SN38) and c-Met (SU11274) inhibitors. Loss of endogenous PAX5 significantly decreased the viability of SCLC cells, especially when combined with SN38 or SU11274, and maximum effect was seen when both inhibitors were used. Therefore, we propose that PAX5 could be an important regulator of c-Met transcription and a potential target for therapy in SCLC.


Assuntos
Neoplasias Pulmonares/metabolismo , Fator de Transcrição PAX5/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Carcinoma de Pequenas Células do Pulmão/metabolismo , Apoptose , Carcinoma Neuroendócrino/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Análise Mutacional de DNA , DNA Topoisomerases Tipo I/metabolismo , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Immunoblotting , Hibridização in Situ Fluorescente , Indóis/metabolismo , Neoplasias Pulmonares/genética , Neoplasias/metabolismo , Fator de Transcrição PAX5/genética , Fatores de Transcrição Box Pareados/metabolismo , Piperazinas/metabolismo , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , RNA Interferente Pequeno/metabolismo , Carcinoma de Pequenas Células do Pulmão/genética , Sulfonamidas/metabolismo , Inibidores da Topoisomerase I
16.
Pigment Cell Melanoma Res ; 21(6): 627-45, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18983540

RESUMO

Transcription factors initiate programs of gene expression and are catalysts in downstream molecular cascades that modulate a variety of cellular processes. Pax3 is a transcription factor that is important in the melanocyte and influences melanocytic proliferation, resistance to apoptosis, migration, lineage specificity and differentiation. In this review, we focus on Pax3 and the molecular pathways that Pax3 is a part of during melanogenesis and in the melanocyte stem cell. These roles of Pax3 are emphasized during the development of diseases and syndromes resulting from either too much or too little Pax3 function. Due to its key task in melanocyte stem cells and tumors, the Pax3 pathway may provide an ideal target for either stem cell or cancer therapies.


Assuntos
Melanócitos/metabolismo , Melanoma/metabolismo , Fatores de Transcrição Box Pareados/fisiologia , Pigmentação/fisiologia , Células-Tronco/metabolismo , Sequência de Aminoácidos , Diferenciação Celular , Regulação da Expressão Gênica , Humanos , Melanócitos/citologia , Melanoma/genética , Dados de Sequência Molecular , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/química , Células-Tronco/citologia
17.
Mod Pathol ; 21(5): 525-30, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18327212

RESUMO

Melanoma is responsible for an estimated 62,000 new American cancer diagnoses and is projected to cause nearly 8000 deaths in 2008 alone. Although the histogenesis of the tumor is not well understood, it is thought to originate from a rare melanocyte stem cell that resides in the skin. The transcription factor PAX3 has a well-established role in the development of melanocytes during embryogenesis, and has recently been characterized as a molecular switch in the mature melanocyte. Based on this function, PAX3 promotes a melanocytic phenotype but blocks terminal differentiation. This mechanism may also contribute to the uncontrolled cell growth and loss of terminal differentiation in melanomas. Here, we find PAX3 expression in 8/8 melanoma cell lines. We also find that PAX3 is commonly expressed in primary melanoma samples (21/58) but significantly less frequently in benign pigmented lesions (9/75). Further analysis of our melanoma set revealed that PAX3 expression is strongly correlated with younger patients with low or no evidence of sun damage. Our data suggest that PAX3-expressing melanomas may be less environmentally dependent and more genetically linked.


Assuntos
Biomarcadores Tumorais/análise , Melanoma/metabolismo , Nevo Pigmentado/metabolismo , Fatores de Transcrição Box Pareados/biossíntese , Neoplasias Cutâneas/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Melanoma/genética , Camundongos , Nevo Pigmentado/genética , Fator de Transcrição PAX3 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/genética , Luz Solar/efeitos adversos
18.
Mol Carcinog ; 47(2): 148-56, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17849422

RESUMO

Tumors of the exocrine pancreas are a major cause of cancer death and have among the poorest prognosis of any malignancy. Following the "cancer stem cell hypothesis," where tumors are believed to originate in tissue specific stem cells, we screened primary ductal pancreatic carcinomas and cell lines for the expression of possible stem cell factors. We find 32/46 (70%) of primary tumors and 9/10 (90%) of cell lines express PAX6. PAX6 is a transcription factor expressed throughout the pancreatic bud during embryogenesis but not in the mature exocrine pancreas. PAX proteins have also been implicated in maintaining stem cells in a committed but undifferentiated state but a role for PAX proteins in putative pancreas stem cells is not known. We induced a pancreatic carcinoma cell line, Panc-1, to differentiate by transfecting wild-type p53 and treating the cells with differentiation agents gastrin or butyrate. This treatment induces cells to terminally differentiate into a growth-arrested cell with neurite-like processes, express the terminal differentiation marker somatostatin and downregulate PAX6. This phenotype can be replicated by directly inhibiting PAX6 expression. These data support a model where PAX proteins are aberrantly expressed in tumors and downregulation leads to differentiation.


Assuntos
Adenocarcinoma/genética , Diferenciação Celular/genética , Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição Box Pareados/genética , Neoplasias Pancreáticas/genética , Proteínas Repressoras/genética , Células 3T3 , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Sequência de Bases , Divisão Celular , Linhagem Celular Tumoral , Primers do DNA , Camundongos , Fator de Transcrição PAX6 , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Ratos , Proteína Supressora de Tumor p53/metabolismo
19.
Mol Med ; 13(3-4): 160-5, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17592550

RESUMO

Angiomyolipomas are benign tumors of the kidney which express phenotypes of smooth muscle, fat, and melanocytes. These tumors appear with increased frequency in the autosomal dominant disorder tuberous sclerosis and are the leading cause of morbidity in adults with tuberous sclerosis. While benign, these tumors are capable of provoking life threatening hemorrhage and replacement of the kidney parenchyma, resulting in renal failure. The histogenesis of these tumors is currently unclear, although currently, we believe these tumors arise from "perivascular epithelioid cells" of which no normal counterpart has been convincingly demonstrated. Recently, stem cell precursors have been recognized that can give rise to smooth muscle and melanocytes. These precursors have been shown to express the neural stem cell marker NG2 and L1. In order to determine whether angiomyolipomas, which exhibit smooth muscle and melanocytic phenotypes, express NG2 and L1, we performed immunocytochemistry on a cell line derived from a human angiomyolipoma, and found that these cells are uniformly positive. Immunohistochemistry of human angiomyolipoma specimens revealed uniform staining of tumor cells, while renal cell carcinomas revealed positivity only of angiogenic vessels. These results support a novel histogenesis of angiomyolipoma as a defect in differentiation of stem cell precursors.


Assuntos
Angiomiolipoma/química , Angiomiolipoma/etiologia , Biomarcadores Tumorais/metabolismo , Neoplasias Renais/química , Neoplasias Renais/etiologia , Células-Tronco/metabolismo , Angiomiolipoma/patologia , Antígenos/metabolismo , Linhagem Celular Transformada , Transformação Celular Viral , Humanos , Imuno-Histoquímica , Neoplasias Renais/patologia , Complexo Antígeno L1 Leucocitário/metabolismo , Proteoglicanas/metabolismo , Vírus 40 dos Símios/fisiologia , Células-Tronco/citologia
20.
Biochem Pharmacol ; 73(1): 1-14, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16904651

RESUMO

PAX proteins function as transcription factors and play an essential role in organogenesis during embryonic development in regulating cell proliferation and self-renewal, resistance to apoptosis, migration of embryonic precursor cells, and the coordination of specific differentiation programs. Recent studies have also discovered a role for PAX proteins in specific stem cell or progenitor cell populations, including melanocytes, muscle, and B-cells. The normal functions of the PAX proteins, including apoptosis resistance and repression of terminal differentiation, may be subverted during the progression of a number of specific malignancies. This is supported by the fact that expression of PAX proteins is dysregulated in several different types of tumors, although the precise roles for PAX proteins in cancer are not clearly understood. An emerging hypothesis is that PAX proteins play an essential role in maintaining tissue specific stem cells by inhibiting terminal differentiation and apoptosis and that these functional characteristics may facilitate the development and progression of specific cancers. In this review, we provide a general background to the PAX protein family and focus on specific cells and tissues and the role PAX proteins play within these tissues in terms of development, mature tissue maintenance, and expression in tumors. Understanding the normal developmental pathways regulated by PAX proteins may shed light on potentially parallel pathways shared in tumors, and ultimately result in defining new molecular targets and signaling pathways for the development of novel anti-cancer therapies.


Assuntos
Desenvolvimento Embrionário/fisiologia , Neoplasias/fisiopatologia , Fatores de Transcrição Box Pareados/fisiologia , Progressão da Doença , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA