Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Clin Cancer Res ; 23(7): 1862-1874, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27663592

RESUMO

Purpose: Cyclin E1 (CCNE1) amplification is associated with primary treatment resistance and poor outcome in high-grade serous ovarian cancer (HGSC). Here, we explore approaches to target CCNE1-amplified cancers and potential strategies to overcome resistance to targeted agents.Experimental Design: To examine dependency on CDK2 in CCNE1-amplified HGSC, we utilized siRNA and conditional shRNA gene suppression, and chemical inhibition using dinaciclib, a small-molecule CDK2 inhibitor. High-throughput compound screening was used to identify selective synergistic drug combinations, as well as combinations that may overcome drug resistance. An observed relationship between CCNE1 and the AKT pathway was further explored in genomic data from primary tumors, and functional studies in fallopian tube secretory cells.Results: We validate CDK2 as a therapeutic target by demonstrating selective sensitivity to gene suppression. However, we found that dinaciclib did not trigger amplicon-dependent sensitivity in a panel of HGSC cell lines. A high-throughput compound screen identified synergistic combinations in CCNE1-amplified HGSC, including dinaciclib and AKT inhibitors. Analysis of genomic data from TCGA demonstrated coamplification of CCNE1 and AKT2 Overexpression of Cyclin E1 and AKT isoforms, in addition to mutant TP53, imparted malignant characteristics in untransformed fallopian tube secretory cells, the dominant site of origin of HGSC.Conclusions: These findings suggest a specific dependency of CCNE1-amplified tumors for AKT activity, and point to a novel combination of dinaciclib and AKT inhibitors that may selectively target patients with CCNE1-amplified HGSC. Clin Cancer Res; 23(7); 1862-74. ©2016 AACR.


Assuntos
Ciclina E/genética , Quinase 2 Dependente de Ciclina/genética , Proteína Oncogênica v-akt/genética , Proteínas Oncogênicas/genética , Neoplasias Ovarianas/tratamento farmacológico , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Linhagem Celular Tumoral , Óxidos N-Cíclicos , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Indolizinas , Proteína Oncogênica v-akt/antagonistas & inibidores , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Compostos de Piridínio/administração & dosagem , RNA Interferente Pequeno/genética
2.
Oncotarget ; 6(36): 38983-98, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26472020

RESUMO

High grade serous ovarian carcinoma (HGSC) is a DNA instable tumor and its precursor is commonly found originating from the fimbriated end of the fallopian tube secretory epithelial (FTSE) cells. The local stresses via ovulation and related inflammation are risks for HGSC. In this study, we examined the cellular and molecular responses of FTSE cells to stress. We found that excess intracellular reactive oxygen species (ROS) in normal FTSE cells upregulated a subset of microRNA expression (defined as ROSmiRs). Most ROSmiRs' expression and function were influenced and regulated by p53, and together they drove the cells into stress-induced premature senescence (SIPS). However, ROS-induced miR-182 is regulated by ß-catenin, not by p53. In normal FTSE cells, miR-182 overexpression triggers cellular senescence by p53-mediated upregulation of p21. Conversely, in cells with p53 mutations, miR-182 overexpression no longer enhances p21 but functions as an "Onco-miR". p53 dysfunction is a prerequisite for miR-182-mediated tumorigenesis. In addition, we found that human follicular fluid could significantly induce intracellular ROS in normal FTSE cells. These findings suggest that ROS and p53 mutations may trigger a series of events, beginning with overexpressing miR-182 by ROS and ß-catenin, impairing the DNA damage response, promoting DNA instability, bypassing senescence and eventually leading to DNA instable tumors in FTSE cells.


Assuntos
Tubas Uterinas/citologia , Tubas Uterinas/metabolismo , MicroRNAs/metabolismo , Estresse Oxidativo/genética , Espécies Reativas de Oxigênio/metabolismo , Animais , Carcinogênese , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Senescência Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , MicroRNAs/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
Gynecol Oncol ; 139(1): 104-11, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26206555

RESUMO

OBJECTIVE: To credential Stathmin 1 (STMN1) and p16(INK4A) (p16) as adjunct markers for the diagnosis of serous tubal intraepithelial carcinoma (STIC), and to compare STMN1 and p16 expression in p53-positive and p53-negative STIC and invasive high-grade serous carcinoma (HGSC). METHODS: Immunohistochemistry (IHC) was used to examine STMN1 and p16 expression in fallopian tube specimens (n=31) containing p53-positive and p53-negative STICs, invasive HGSCs, and morphologically normal FTE (fallopian tube epithelium). STMN1 and p16 expression was scored semiquantitatively by four individuals. The semiquantitative scores were dichotomized, and reported as positive or negative. Pooled siRNA was used to knockdown p53 in a panel of cell lines derived from immortalized FTE and HGSC. RESULTS: STMN1 and p16 were expressed in the majority of p53-positive and p53-negative STICs and concomitant invasive HGSCs, but only scattered positive cells were present in morphologically normal FTE. Both proteins were expressed consistently across multiple STICs from the same patient and in concomitant invasive HGSC. Knockdown of p53 in immortalized FTE cells and in four HGSC-derived cell lines expressing different missense p53 mutations did not affect STMN1 protein levels. CONCLUSIONS: This study demonstrates that STMN1 and p16 are sensitive and specific adjunct biomarkers that, when used with p53 and Ki-67, improve the diagnostic accuracy of STIC. The addition of STMN1 and p16 helps to compensate for practical limitations of p53 and Ki-67 that complicate the diagnosis in up to one third of STICs.


Assuntos
Biomarcadores Tumorais/biossíntese , Cistadenocarcinoma Seroso/metabolismo , Neoplasias das Tubas Uterinas/metabolismo , Proteínas de Neoplasias/biossíntese , Neoplasias Ovarianas/metabolismo , Estatmina/biossíntese , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Neoplasias das Tubas Uterinas/genética , Neoplasias das Tubas Uterinas/patologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Mutação , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
4.
Proc Natl Acad Sci U S A ; 111(3): 1102-7, 2014 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-24385586

RESUMO

High-grade serous ovarian cancers are characterized by widespread recurrent copy number alterations. Although some regions of copy number change harbor known oncogenes and tumor suppressor genes, the genes targeted by the majority of amplified or deleted regions in ovarian cancer remain undefined. Here we systematically tested amplified genes for their ability to promote tumor formation using an in vivo multiplexed transformation assay. We identified the GRB2-associated binding protein 2 (GAB2) as a recurrently amplified gene that potently transforms immortalized ovarian and fallopian tube secretory epithelial cells. Cancer cell lines overexpressing GAB2 require GAB2 for survival and show evidence of phosphatidylinositol 3-kinase (PI3K) pathway activation, which was required for GAB2-induced transformation. Cell lines overexpressing GAB2 were as sensitive to PI3K inhibition as cell lines harboring mutant PIK3CA. Together, these observations nominate GAB2 as an ovarian cancer oncogene, identify an alternative mechanism to activate PI3K signaling, and underscore the importance of PI3K signaling in this cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Feminino , Genômica , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Fases de Leitura Aberta , Neoplasias Ovarianas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais
5.
Cancer Res ; 74(4): 1141-52, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24366882

RESUMO

The fallopian tube is now generally considered the dominant site of origin for high-grade serous ovarian carcinoma. However, the molecular pathogenesis of fallopian tube-derived serous carcinomas is poorly understood and there are few experimental studies examining the transformation of human fallopian tube cells. Prompted by recent genomic analyses that identified cyclin E1 (CCNE1) gene amplification as a candidate oncogenic driver in high-grade serous ovarian carcinoma, we evaluated the functional role of cyclin E1 in serous carcinogenesis. Cyclin E1 was expressed in early- and late-stage human tumor samples. In primary human fallopian tube secretory epithelial cells, cyclin E1 expression imparted malignant characteristics to untransformed cells if p53 was compromised, promoting an accumulation of DNA damage and altered transcription of DNA damage response genes related to DNA replication stress. Together, our findings corroborate the hypothesis that cyclin E1 dysregulation acts to drive malignant transformation in fallopian tube secretory cells that are the site of origin of high-grade serous ovarian carcinomas.


Assuntos
Transformação Celular Neoplásica/genética , Ciclina E/genética , Cistadenocarcinoma Seroso/genética , Neoplasias das Tubas Uterinas/genética , Tubas Uterinas/patologia , Proteínas Oncogênicas/genética , Neoplasias Ovarianas/genética , Proliferação de Células , Ciclina E/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/secundário , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Neoplasias das Tubas Uterinas/patologia , Tubas Uterinas/metabolismo , Feminino , Amplificação de Genes/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Oncogênicas/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/secundário , Análise Serial de Tecidos , Células Tumorais Cultivadas
6.
Cancer Cell ; 24(6): 751-65, 2013 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-24332043

RESUMO

High-grade serous ovarian carcinoma presents significant clinical and therapeutic challenges. Although the traditional model of carcinogenesis has focused on the ovary as a tumor initiation site, recent studies suggest that there may be additional sites of origin outside the ovary, namely the secretory cells of the fallopian tube. Our study demonstrates that high-grade serous tumors can originate in fallopian tubal secretory epithelial cells and also establishes serous tubal intraepithelial carcinoma as the precursor lesion to high-grade serous ovarian and peritoneal carcinomas in animal models targeting the Brca, Tp53, and Pten genes. These findings offer an avenue to address clinically important questions that are critical for cancer prevention and early detection in women carrying BRCA1 and BRCA2 mutations.


Assuntos
Transformação Celular Neoplásica , Cistadenocarcinoma Seroso/etiologia , Neoplasias das Tubas Uterinas/patologia , Genes BRCA1 , Genes BRCA2 , Neoplasias Ovarianas/etiologia , Lesões Pré-Cancerosas/patologia , Animais , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Epitélio/patologia , Feminino , Genes p53 , Integrases/genética , Camundongos , Camundongos Endogâmicos C57BL , Gradação de Tumores , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fator de Transcrição PAX8 , PTEN Fosfo-Hidrolase/genética , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/fisiologia
7.
Nat Protoc ; 7(9): 1755-64, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22936217

RESUMO

Primary human fallopian tube secretory epithelial cell (FTSEC) cultures are useful for studying normal fallopian tube epithelial biology, as well as for developing models of fallopian tube disease, such as cancer. Because of the limited ability of primary human FTSECs to proliferate in vitro, it is necessary to immortalize them in order to establish a cell line that is suitable for long-term culture and large-scale in vitro experimentation. This protocol describes the isolation of FTSECs from human fallopian tube tissue, conditions for primary FTSEC culture and techniques for establishing immortal FTSEC lines. The entire process, from primary cell isolation to establishment of an immortal cell line, may take up to 2 months. Once established, immortal FTSECs can typically be maintained for at least 30 passages.


Assuntos
Técnicas de Cultura de Células/métodos , Células Epiteliais/citologia , Tubas Uterinas/citologia , Linhagem Celular Transformada , Transformação Celular Neoplásica , Feminino , Humanos
8.
Sci Transl Med ; 4(147): 147ra112, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22896676

RESUMO

The comprehensive characterization of a large number of cancer genomes will eventually lead to a compendium of genetic alterations in specific cancers. Unfortunately, the number and complexity of identified alterations complicate endeavors to identify biologically relevant mutations critical for tumor maintenance because many of these targets are not amenable to manipulation by small molecules or antibodies. RNA interference provides a direct way to study putative cancer targets; however, specific delivery of therapeutics to the tumor parenchyma remains an intractable problem. We describe a platform for the discovery and initial validation of cancer targets, composed of a systematic effort to identify amplified and essential genes in human cancer cell lines and tumors partnered with a novel modular delivery technology. We developed a tumor-penetrating nanocomplex (TPN) that comprised small interfering RNA (siRNA) complexed with a tandem tumor-penetrating and membrane-translocating peptide, which enabled the specific delivery of siRNA deep into the tumor parenchyma. We used TPN in vivo to evaluate inhibitor of DNA binding 4 (ID4) as a novel oncogene. Treatment of ovarian tumor-bearing mice with ID4-specific TPN suppressed growth of established tumors and significantly improved survival. These observations not only credential ID4 as an oncogene in 32% of high-grade ovarian cancers but also provide a framework for the identification, validation, and understanding of potential therapeutic cancer targets.


Assuntos
Proteínas Inibidoras de Diferenciação/genética , Nanopartículas/química , Oncogenes/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , RNA Interferente Pequeno/metabolismo , Animais , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Nanopartículas/efeitos adversos , Tela Subcutânea/patologia , Transcrição Gênica
9.
F1000 Med Rep ; 3: 22, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22076125

RESUMO

Ovarian cancer has a disproportionately high mortality rate because patients typically present with late-stage metastatic disease. The vast majority of these deaths are from high-grade serous carcinoma. Recent studies indicate that many of these tumors arise from the fallopian tube and subsequently metastasize to the ovary. This may explain why such tumors have not been detected at early stage as detection efforts have been focused purely on the ovary. In keeping with this leap in understanding other advances such as the development of ex-vivo models and immortalization of human fallopian tube epithelial cells, and the use of integrated genomic analyses to identify hundreds of novel candidate oncogenes and tumor suppressors potentially involved in tumorigenesis now engender hope that we can begin to truly define the differences in pathogenesis between fallopian tube and ovarian-derived tumors. In doing so, we can hopefully improve early detection, treatment, and outcome.

10.
Gynecol Oncol ; 123(1): 5-12, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21683992

RESUMO

BACKGROUND: Most high-grade pelvic serous carcinomas (HGPSCs) arise from fallopian tube epithelium (FTE). To date, few markers have been shown to characterize FTE transformation. Stathmin 1 (STMN1) is a candidate oncogene whose activity is influenced by p53, p27Kip1 (p27), and PI3K/Akt pathway activation. As a microtubule destabilizing protein, STMN1 regulates cytoskeletal dynamics, cell cycle progression, mitosis, and cell migration. This study examines the expression of STMN1 and its negative regulator p27 along the morphologic continuum from normal FTE to invasive carcinoma. METHODS: STMN1 and p27 expression were examined by immunohistochemistry (IHC) in benign (n=12) and malignant (n=13) fallopian tubes containing normal epithelium, morphologically benign putative precursor lesions ("p53 signatures"), potential transitional precursor lesions ("proliferative p53 signatures"), tubal intraepithelial carcinoma (TIC), and/or invasive serous carcinoma. STMN1 expression was further assessed in 131 late-stage HGPSCs diagnosed as primary ovarian and in 6 ovarian cancer cell lines by IHC and Western blot, respectively. RESULTS: STMN1 expression was absent in benign FTE and infrequently detected in p53 signatures. However, it was weakly expressed in proliferative p53 signatures and robustly induced upon progression to TIC and invasive carcinoma, typically accompanied by decreased p27 levels. STMN1 was expressed in >80% of high-grade serous ovarian carcinomas and cell lines. CONCLUSIONS: STMN1 is a novel marker of early serous carcinoma that may play a role in FTE tumor initiation. Our data are consistent with a model by which STMN1 overexpression, resulting from loss of p27-mediated regulation, may potentiate aberrant cell proliferation, migration, and/or loss of polarity during early tumorigenesis.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias das Tubas Uterinas/metabolismo , Microtúbulos/metabolismo , Estatmina/biossíntese , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Cistadenocarcinoma Seroso/enzimologia , Cistadenocarcinoma Seroso/metabolismo , Neoplasias das Tubas Uterinas/enzimologia , Neoplasias das Tubas Uterinas/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Estatmina/metabolismo , Proteína Supressora de Tumor p53/metabolismo
11.
Proc Natl Acad Sci U S A ; 108(18): 7547-52, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21502498

RESUMO

High-grade serous ovarian carcinoma (HGSOC) is a lethal disease for which improved screening and treatment strategies are urgently needed. Progress in these areas is impeded by our poor understanding of HGSOC pathogenesis. Most ovarian cancer research is based on the hypothesis that HGSOC arises from ovarian surface epithelial cells. However, recent studies suggest that >50% of high-grade serous carcinomas involving the ovary likely arise from fallopian tube epithelium. Therefore, limiting HGSOC research to modeling based on ovarian surface epithelium alone is inadequate. To address the need for a fallopian tube-based model of HGSOC, we have developed a system for studying human fallopian tube secretory epithelial cell (FTSEC) transformation. Our model is based on (i) immortalization of FTSECs isolated from primary samples of normal, nondiseased human fallopian tubes, (ii) transformation of FTSECs with defined genetic elements, and (iii) xenograft-based tumorigenic assays. We use our model to show that FTSECs immortalized with human telomerase reverse transcriptase (hTERT) plus SV40 large T and small T antigens are transformed by either oncogenic Ras (H-Ras(V12)) or c-Myc expression, leading to increased proliferation, clonogenicity, and anchorage-independent growth. Additionally, we demonstrate that FTSECs remain susceptible to c-Myc-mediated transformation in the absence of viral oncoproteins, by replacing SV40 large T and small T antigens with sh-p53, mutant CDK4 (CDK4(R24C)), and sh-PP2A-B56γ. Importantly, all transformed FTSECs gave rise to high-grade Müllerian carcinomas that were grossly, histologically, immunophenotypically, and genomically similar to human HGSOC. With this model, we will now be able to assess the transformative effects of specific genetic alterations on FTSECs in order to characterize their respective roles in HGSOC development.


Assuntos
Carcinoma/fisiopatologia , Linhagem Celular Tumoral , Células Epiteliais/citologia , Tubas Uterinas/citologia , Neoplasias Ovarianas/fisiopatologia , Antígenos Transformantes de Poliomavirus , Western Blotting , Testes de Carcinogenicidade , Transformação Celular Neoplásica , Hibridização Genômica Comparativa , Feminino , Humanos , Imuno-Histoquímica , Microscopia de Fluorescência , Telomerase , Ensaio Tumoral de Célula-Tronco
12.
J Oncol ; 2010: 932371, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19746182

RESUMO

Ovarian cancer is a deadly disease for which there is no effective means of early detection. Ovarian carcinomas comprise a diverse group of neoplasms, exhibiting a wide range of morphological characteristics, clinical manifestations, genetic alterations, and tumor behaviors. This high degree of heterogeneity presents a major clinical challenge in both diagnosing and treating ovarian cancer. Furthermore, the early events leading to ovarian carcinoma development are poorly understood, thus complicating efforts to develop screening modalities for this disease. Here, we provide an overview of the current models of ovarian cancer pathogenesis, highlighting recent findings implicating the fallopian tube fimbria as a possible site of origin of ovarian carcinomas. The ovarian cancer model will continue to evolve as we learn more about the genetics and etiology of this disease.

13.
Int J Cancer ; 124(2): 494-501, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18942706

RESUMO

Nuclear factor kappa B (NF-kappaB) signaling is deregulated in many tumor types, resulting in aberrant expression and/or activation of NF-kappaB transcriptional complexes. We have previously reported that nuclear expression of the NF-kappaB subunit p50 is strongly correlated with melanoma progression and poor 5-year patient survival. In this study, we used cDNA microarray to analyze the gene expression profiles of melanoma cells overexpressing NF-kappaB p50. We found that NF-kappaB p50 expression strongly induced interleukin-6 (IL-6) upregulation in melanoma cells at both the transcriptional and translational levels and that IL-6 production by melanoma cells enhanced the growth of endothelial cells in vitro. Expression of activating transcription factor 3 (ATF3), a negative regulator of IL-6 gene transcription, inhibited p50-mediated IL-6 upregulation. Knockdown of p50 expression using lentiviral-based shRNA abrogated IL-6 induction in melanoma cells and inhibited its effects on endothelial cell growth. Finally, we used an in vivo matrigel plug assay to show that NF-kappaB p50 overexpression promotes angiogenesis, while silencing NF-kappaB p50 inhibits blood vessel formation. Our results demonstrate for the first time that the NF-kappaB p50 subunit mediates melanoma angiogenesis by specifically upregulating IL-6, highlighting a novel and important role for the NF-kappaB p50/IL-6 signaling axis in melanoma progression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Interleucina-6/biossíntese , Subunidade p50 de NF-kappa B/fisiologia , Neovascularização Patológica , Fator 3 Ativador da Transcrição/biossíntese , Animais , Linhagem Celular Tumoral , Proliferação de Células , Colágeno/química , Combinação de Medicamentos , Humanos , Laminina/química , Masculino , Camundongos , Camundongos Nus , Subunidade p50 de NF-kappa B/metabolismo , Transplante de Neoplasias , Proteoglicanas/química , Regulação para Cima
14.
Cancer Res ; 66(18): 9221-6, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16982766

RESUMO

Malignant melanoma is an aggressive and chemoresistant form of skin cancer characterized by rapid metastasis and poor patient prognosis. The development of innovative therapies with improved efficacy is critical to treatment of this disease. Here, we show that aberrant expression of two proteins, p53 up-regulated modulator of apoptosis (PUMA) and phosphorylated Akt (p-Akt), is associated with poor patient survival. Using tissue microarray analysis, we found that patients exhibiting both weak PUMA expression and strong p-Akt expression in their melanoma tumor tissue had significantly worse 5-year survival than patients with either weak PUMA or strong p-Akt expression alone (P < 0.001). Strikingly, no patients exhibiting strong PUMA expression and weak p-Akt expression in primary tumor tissue died within 5 years of diagnosis. We propose a two-pronged therapeutic strategy of (a) boosting PUMA expression and (b) inhibiting Akt phosphorylation in melanoma tumor tissue. Here, we report that a recombinant adenovirus containing human PUMA cDNA (ad-PUMA) efficiently inhibits human melanoma cell survival in vitro, rapidly induces apoptosis, and dramatically suppresses human melanoma tumor growth in a severe combined immunodeficient mouse xenograft model. In melanoma cells strongly expressing p-Akt, we show that Akt/protein kinase B signaling inhibitor-2 (API-2; a small-molecule Akt inhibitor) reduces cell survival in a dose- and time-dependent manner and enhances ad-PUMA-mediated growth inhibition of melanoma cells. Finally, we show that, by combining ad-PUMA and API-2 treatments, human melanoma tumor growth can be inhibited by >80% in vivo compared with controls. Our results suggest that a strategy to correct dysregulated PUMA and p-Akt expression in malignant melanoma may be an effective therapeutic option.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Melanoma/patologia , Melanoma/terapia , Proteína Oncogênica v-akt/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia , Adenoviridae/genética , Animais , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/biossíntese , Proteínas Reguladoras de Apoptose/genética , Processos de Crescimento Celular/fisiologia , DNA Complementar/genética , Humanos , Masculino , Melanoma/enzimologia , Camundongos , Camundongos SCID , Mitocôndrias/fisiologia , Proteína Oncogênica v-akt/biossíntese , Fosforilação , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Distribuição Aleatória , Neoplasias Cutâneas/enzimologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Oncogene ; 24(6): 1111-6, 2005 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-15690057

RESUMO

Cutaneous malignant melanoma is an aggressive form of skin cancer, characterized by strong chemoresistance and poor patient prognosis. The molecular mechanisms underlying its resistance to chemotherapy remain unclear but are speculated to involve the dysregulation of apoptotic pathways. In this study, we sought to determine whether PUMA (p53 upregulated modulator of apoptosis) contributes to human melanoma formation, tumor progression, and survival. We used tissue microarray and immunohistochemistry to examine PUMA expression in 107 primary melanomas, 51 metastatic melanomas, and 64 dysplastic nevi. Here we report that PUMA expression is significantly weaker in primary melanomas compared to dysplastic nevi (P<0.0001), and is further reduced in metastatic melanomas compared to primary tumors (P=0.001). We show that weak PUMA expression in melanoma correlates with poorer overall and disease-specific 5-year survival (P<0.005 and P<0.001, respectively) of melanoma patients and that PUMA expression in tumor tissue is an independent predictor of both overall and disease-specific 5-year survival (P=0.05). Additionally, we show that exogenous PUMA expression in human melanoma cell lines (both wild type and mutant p53) results in significant apoptotic cell death. Our results suggest that PUMA expression may be an important prognostic marker for human melanoma and that adenoviral delivery of PUMA sensitizes melanoma cells to apoptosis.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma/genética , Melanoma/patologia , Proteínas Proto-Oncogênicas/biossíntese , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Proteína Supressora de Tumor p53/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Proteínas Reguladoras de Apoptose , Biomarcadores Tumorais , Progressão da Doença , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...