Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
ACS Appl Bio Mater ; 7(3): 1588-1599, 2024 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-38437727

RESUMO

The synthesis of copper nanoparticles (CuNPs) was accomplished by using a rapid, green, and versatile argon plasma reduction method that involves solvent extraction. With this method, a plasma-solid state interaction forms and CuNPs can be synthesized from copper(II) sulfate using a low-pressure, low-temperature argon plasma. Characterization studies of the CuNPs revealed that when a metal precursor is treated under optimal experimental conditions of 80 W of argon plasma for 300 s, brown CuNPs are synthesized. However, when those same brown CuNPs are placed in Milli-Q water for a period of 10 days, oxidation occurs and green CuNPs are formed. Confirmation of the chemical identity of the CuNPs was performed by using X-ray photoelectron spectroscopy. The results reveal that the brown CuNPs are predominantly Cu0 or what we refer to as CuNPs, while the green CuNPs are a mixture of Cu0 and Cu(OH)2 NPs. Upon further characterization of both brown and green CuNPs with scanning electron microscopy (SEM), the results depict brown CuNPs with a rod-like shape and approximate dimensions of 40 nm × 160 nm, while the green CuNPs were smaller in size, with dimensions of 40-80 nm, and more of a round shape. When testing the antibacterial activity of both brown and green CuNPs, our findings demonstrate the effectiveness of both CuNPs against Escherichia coli and Staphylococcus aureus bacteria at a concentration of 17 µg/mL. The inactivation of S. aureus and E. coli 7-day-old biofilms required CuNP concentrations of 99 µg/mL. SEM images of treated 7-day-old S. aureus and E. coli biofilms depict cell membranes that are completely damaged, suggesting a physical killing mechanism. In addition, when the same concentration of CuNPs used to inactivate biofilms were tested with human fibroblasts, both brown and green CuNPs were found to be biocompatible.


Assuntos
Anti-Infecciosos , Nanopartículas , Gases em Plasma , Humanos , Cobre/farmacologia , Gases em Plasma/farmacologia , Escherichia coli , Staphylococcus aureus , Anti-Infecciosos/farmacologia
3.
JCI Insight ; 9(5)2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38271085

RESUMO

High-grade serous carcinoma (HGSC) is the most lethal gynecological malignancy in the United States. Late diagnosis and the emergence of chemoresistance have prompted studies into how the tumor microenvironment, and more recently tumor innervation, may be leveraged for HGSC prevention and interception. In addition to stess-induced sources, concentrations of the sympathetic neurotransmitter norepinephrine (NE) in the ovary increase during ovulation and after menopause. Importantly, NE exacerbates advanced HGSC progression. However, little is known about the role of NE in early disease pathogenesis. Here, we investigated the role of NE in instigating anchorage independence and micrometastasis of preneoplastic lesions from the fallopian tube epithelium (FTE) to the ovary, an essential step in HGSC onset. We found that in the presence of NE, FTE cell lines were able to survive in ultra-low-attachment (ULA) culture in a ß-adrenergic receptor-dependent (ß-AR-dependent) manner. Importantly, spheroid formation and cell viability conferred by treatment with physiological sources of NE were abrogated using the ß-AR blocker propranolol. We have also identified that NE-mediated anoikis resistance may be attributable to downregulation of colony-stimulating factor 2. These findings provide mechanistic insight and identify targets that may be regulated by ovary-derived NE in early HGSC.


Assuntos
Cistadenocarcinoma Seroso , Neoplasias Ovarianas , Feminino , Humanos , Neoplasias Ovarianas/metabolismo , Cistadenocarcinoma Seroso/tratamento farmacológico , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Tubas Uterinas/metabolismo , Tubas Uterinas/patologia , Anoikis , Norepinefrina/farmacologia , Norepinefrina/metabolismo , Microambiente Tumoral
4.
bioRxiv ; 2023 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-36909636

RESUMO

Loss of treatment-induced ovarian carcinoma (OC) growth suppression poses a major clinical challenge because it leads to disease recurrence. Therefore, there is a compelling need for well- -tolerated approaches that can support tumor growth-suppression after therapy is stopped. We have profiled ascites as OC tumor microenvironments to search for potential non-toxic soluble components that would activate tumor suppressor pathways in OC cells. Our investigations revealed that low levels of taurine, a non-proteogenic sulfonic amino acid, were present within OC ascites. Taurine supplementation, beyond levels found in ascites, induced growth suppression without causing cytotoxicity in various OC cells, including chemotherapy-resistant cell clones and patient-derived organoids representing primary or chemotherapy recovered disease. Inhibition of proliferation by taurine was linked to increased mutant or wild-type p53 proteins binding to DNA, induction of p21, and independently of p53, TIGAR expression. Taurine-induced activation of p21 and TIGAR was associated with suppression of cell-cycle progression, glycolysis, and mitochondrial respiration. Expression of p21 or TIGAR in OC cells mimicked taurine-induced growth suppression. Our studies support the potential therapeutic value of taurine supplementation in OC.

5.
Commun Biol ; 5(1): 1362, 2022 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-36509990

RESUMO

Most ovarian high-grade serous carcinomas (HGSC) arise from Serous Tubal Intraepithelial Carcinoma (STIC) lesions in the distal end of the fallopian tube (FT). Formation of STIC lesions from FT secretory cells leads to seeding of the ovarian surface, with rapid tumor dissemination to other abdominal structures thereafter. It remains unclear how nascent malignant cells leave the FT to colonize the ovary. This report provides evidence that the L1 cell adhesion molecule (L1CAM) contributes to the ability of transformed FT secretory cells (FTSEC) to detach from the tube, survive under anchorage-independent conditions, and seed the ovarian surface. L1CAM was highly expressed on the apical cells of STIC lesions and contributed to ovarian colonization by upregulating integrins and fibronectin in malignant cells and activating the AKT and ERK pathways. These changes increased cell survival under ultra-low attachment conditions that mimic transit from the FT to the ovary. To study dissemination to the ovary, we developed a tumor-ovary co-culture model. We showed that L1CAM expression was important for FT cells to invade the ovary as a cohesive group. Our results indicate that in the early stages of HGSC development, transformed FTSECs disseminate from the FT to the ovary in a L1CAM-dependent manner.


Assuntos
Cistadenocarcinoma Seroso , Neoplasias das Tubas Uterinas , Molécula L1 de Adesão de Célula Nervosa , Neoplasias Ovarianas , Feminino , Humanos , Tubas Uterinas/metabolismo , Tubas Uterinas/patologia , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias das Tubas Uterinas/metabolismo , Neoplasias das Tubas Uterinas/patologia , Cistadenocarcinoma Seroso/metabolismo
6.
Adv Biol (Weinh) ; 6(12): e2200197, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36084257

RESUMO

Ovarian carcinoma (OC) forms outgrowths that extend from the outer surface of an afflicted organ into the peritoneum. OC outgrowth formation is poorly understood due to the limited availability of cell culture models examining the behavior of cells that form outgrowths. Prompted by immunochemical evaluation of extracellular matrix (ECM) components in human tissues, laminin and collagen-rich ECM-reconstituted cell culture models amenable to studies of cell clusters that can form outgrowths are developed. It is demonstrated that ECM promotes outgrowth formation in fallopian tube non-ciliated epithelial cells (FNE) expressing mutant p53 and various OC cell lines. Outgrowths are initiated by cells that underwent outward translocation and retained the ability to intercalate into mesothelial cell monolayers. Electron microscopy, optical coherence tomography, and small amplitude oscillatory shear experiments reveal that increased ECM levels led to increased fibrous network thickness and high shear elasticity of the microenvironment. These physical characteristics are associated with outgrowth suppression. The low ECM microenvironment mimicks the viscoelasticity of malignant peritoneal fluid (ascites) and supports cell proliferation, cell translocation, and outgrowth formation. These results highlight the importance of the ECM microenvironment in modulating OC growth and can provide additional insights into the mode of dissemination of primary and recurrent ovarian tumors.


Assuntos
Carcinoma , Neoplasias Ovarianas , Humanos , Feminino , Recidiva Local de Neoplasia/metabolismo , Matriz Extracelular/metabolismo , Neoplasias Ovarianas/genética , Carcinoma Epitelial do Ovário/metabolismo , Laminina/genética , Carcinoma/metabolismo , Microambiente Tumoral
7.
Mol Cancer Res ; 19(7): 1168-1181, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33753553

RESUMO

High-grade serous ovarian cancer (HGSOC) is characterized by chromosomal instability, DNA damage, oxidative stress, and high metabolic demand that exacerbate misfolded, unfolded, and damaged protein burden resulting in increased proteotoxicity. However, the underlying mechanisms that maintain protein homeostasis to promote HGSOC growth remain poorly understood. This study reports that the neuronal deubiquitinating enzyme, ubiquitin carboxyl-terminal hydrolase L1 (UCHL1), is overexpressed in HGSOC and maintains protein homeostasis. UCHL1 expression was markedly increased in HGSOC patient tumors and serous tubal intraepithelial carcinoma (HGSOC precursor lesions). High UCHL1 levels correlated with higher tumor grade and poor patient survival. UCHL1 inhibition reduced HGSOC cell proliferation and invasion, as well as significantly decreased the in vivo metastatic growth of ovarian cancer xenografts. Transcriptional profiling of UCHL1-silenced HGSOC cells revealed downregulation of genes implicated with proteasome activity along with upregulation of endoplasmic reticulum stress-induced genes. Reduced expression of proteasome subunit alpha 7 (PSMA7) and acylaminoacyl peptide hydrolase (APEH), upon silencing of UCHL1, resulted in a significant decrease in proteasome activity, impaired protein degradation, and abrogated HGSOC growth. Furthermore, the accumulation of polyubiquitinated proteins in the UCHL1-silenced cells led to attenuation of mTORC1 activity and protein synthesis, and induction of terminal unfolded protein response. Collectively, these results indicate that UCHL1 promotes HGSOC growth by mediating protein homeostasis through the PSMA7-APEH-proteasome axis. IMPLICATIONS: This study identifies the novel links in the proteostasis network to target protein homeostasis in HGSOC and recognizes the potential of inhibiting UCHL1 and APEH to sensitize cancer cells to proteotoxic stress in solid tumors.


Assuntos
Cistadenocarcinoma Seroso/genética , Neoplasias Ovarianas/genética , Peptídeo Hidrolases/genética , Complexo de Endopeptidases do Proteassoma/genética , Proteostase/genética , Ubiquitina Tiolesterase/genética , Animais , Linhagem Celular Tumoral , Cistadenocarcinoma Seroso/tratamento farmacológico , Cistadenocarcinoma Seroso/metabolismo , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Humanos , Indóis/farmacologia , Estimativa de Kaplan-Meier , Camundongos Nus , Gradação de Tumores , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Oximas/farmacologia , Peptídeo Hidrolases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina Tiolesterase/antagonistas & inibidores , Ubiquitina Tiolesterase/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
8.
Nat Med ; 26(8): 1271-1279, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32572264

RESUMO

Malignant abdominal fluid (ascites) frequently develops in women with advanced high-grade serous ovarian cancer (HGSOC) and is associated with drug resistance and a poor prognosis1. To comprehensively characterize the HGSOC ascites ecosystem, we used single-cell RNA sequencing to profile ~11,000 cells from 22 ascites specimens from 11 patients with HGSOC. We found significant inter-patient variability in the composition and functional programs of ascites cells, including immunomodulatory fibroblast sub-populations and dichotomous macrophage populations. We found that the previously described immunoreactive and mesenchymal subtypes of HGSOC, which have prognostic implications, reflect the abundance of immune infiltrates and fibroblasts rather than distinct subsets of malignant cells2. Malignant cell variability was partly explained by heterogeneous copy number alteration patterns or expression of a stemness program. Malignant cells shared expression of inflammatory programs that were largely recapitulated in single-cell RNA sequencing of ~35,000 cells from additionally collected samples, including three ascites, two primary HGSOC tumors and three patient ascites-derived xenograft models. Inhibition of the JAK/STAT pathway, which was expressed in both malignant cells and cancer-associated fibroblasts, had potent anti-tumor activity in primary short-term cultures and patient-derived xenograft models. Our work contributes to resolving the HSGOC landscape3-5 and provides a resource for the development of novel therapeutic approaches.


Assuntos
Ascite/genética , Cistadenoma Seroso/genética , Neoplasias Ovarianas/genética , Análise de Célula Única , Ascite/patologia , Linhagem Celular Tumoral , Cistadenoma Seroso/patologia , Variações do Número de Cópias de DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Janus Quinase 1/genética , Gradação de Tumores , Proteínas de Neoplasias/genética , Neoplasias Ovarianas/patologia , Prognóstico , Fatores de Transcrição STAT/genética , Análise de Sequência de RNA , Transdução de Sinais/genética
9.
Trends Pharmacol Sci ; 41(3): 183-198, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32014341

RESUMO

The tumor microenvironment (TME) is a complex neighborhood that consists of immune cells, fibroblasts, pericytes, adipocytes, endothelial and neuronal cells, and the extracellular matrix proteins. TME also consists of physical factors, such as oxygen availability, changing pH, interstitial fluid pressure, and tissue stiffness. As cancer progresses, the physical properties and the cells in the TME change significantly, impacting the efficacy of the therapies and modulating drug resistance. This has led to the development of several new treatments targeting the TME. This review focuses on recent advances on the role of TME in drug resistance, with a particular focus on the ongoing clinical trials aiming at disrupting the TME- and the extracellular matrix-mediated protection against therapies.


Assuntos
Neoplasias , Preparações Farmacêuticas , Matriz Extracelular , Humanos , Neoplasias/tratamento farmacológico , Células Estromais , Microambiente Tumoral
10.
Cancers (Basel) ; 11(12)2019 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-31817625

RESUMO

Epithelial ovarian cancer (EOC) comprises multiple disease states representing a variety of distinct tumors that, irrespective of tissue of origin, genetic aberrations and pathological features, share common patterns of dissemination to the peritoneal cavity. EOC peritoneal dissemination is a stepwise process that includes the formation of malignant outgrowths that detach and establish widespread peritoneal metastases through adhesion to serosal membranes. The cell biology associated with outgrowth formation, detachment, and de novo adhesion is at the nexus of diverse genetic backgrounds that characterize the disease. Development of treatment for metastatic disease will require detailed characterization of cellular processes involved in each step of EOC peritoneal dissemination. This article offers a review of the literature that relates to the current stage of knowledge about distinct steps of EOC peritoneal dissemination, with emphasis on the cell biology aspects of the process.

11.
Cell Rep ; 27(1): 255-268.e6, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30943406

RESUMO

Analysis of cancer-derived extracellular vesicles (EVs) in biofluids potentially provides a source of disease biomarkers. At present there is no procedure to systematically identify which antigens should be targeted to differentiate cancer-derived from normal host cell-derived EVs. Here, we propose a computational framework that integrates information about membrane proteins in tumors and normal tissues from databases: UniProt, The Cancer Genome Atlas, the Genotype-Tissue Expression Project, and the Human Protein Atlas. We developed two methods to assess capture of EVs from specific cell types. (1) We used palmitoylated fluorescent protein (palmtdTomato) to label tumor-derived EVs. Beads displaying antibodies of interest were incubated with conditioned medium from palmtdTomato-expressing cells. Bound EVs were quantified using flow cytometry. (2) We also showed that membrane-bound Gaussia luciferase allows the detection of cancer-derived EVs in blood of tumor-bearing animals. Our analytical and validation platform should be applicable to identify antigens on EVs from any tumor type.


Assuntos
Biomarcadores Tumorais/metabolismo , Vesículas Extracelulares/metabolismo , Citometria de Fluxo/métodos , Proteínas de Membrana/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Linhagem Celular Tumoral , Feminino , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoensaio/métodos , Luciferases/metabolismo , Camundongos , Camundongos Nus , Pessoa de Meia-Idade
12.
Mol Cell Biol ; 37(24)2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-28993478

RESUMO

TP53 is one of the most commonly mutated genes in human cancers. Unlike other tumor suppressors that are frequently deleted or acquire loss-of-function mutations, the majority of TP53 mutations in tumors are missense substitutions, which lead to the expression of full-length mutant proteins that accumulate in cancer cells and may confer unique gain-of-function (GOF) activities to promote tumorigenic events. Recently, mutant p53 proteins have been shown to mediate metabolic changes as a novel GOF to promote tumor development. There is a strong rationale that the GOF activities, including alterations in cellular metabolism, might vary between the different p53 mutants. Accordingly, the effect of different mutant p53 proteins on cancer cell metabolism is largely unknown. In this study, we have metabolically profiled several individual frequently occurring p53 mutants in cancers, focusing on glycolytic and mitochondrial oxidative phosphorylation pathways. Our investigation highlights the diversity of different p53 mutants in terms of their effect on metabolism, which might provide a foundation for the development of more effective targeted pharmacological approaches toward variants of mutant p53.


Assuntos
Mitocôndrias/genética , Mitocôndrias/metabolismo , Mutação de Sentido Incorreto , Neoplasias/genética , Neoplasias/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Glicólise/genética , Células HCT116 , Humanos , Fosforilação Oxidativa
13.
Cell Rep ; 18(10): 2343-2358, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28273451

RESUMO

The degree of genetic aberrations characteristic of high-grade serous ovarian cancer (HGSC) makes identification of the molecular features that drive tumor progression difficult. Here, we perform genome-wide RNAi screens and comprehensive expression analysis of cell-surface markers in a panel of HGSC cell lines to identify genes that are critical to their survival. We report that the tetraspanin CD151 contributes to survival of a subset of HGSC cell lines associated with a ZEB transcriptional program and supports the growth of HGSC tumors. Moreover, we show that high CD151 expression is prognostic of poor clinical outcome. This study reveals cell-surface vulnerabilities associated with HGSC, provides a framework for identifying therapeutic targets, and reports a role for CD151 in HGSC.


Assuntos
Biomarcadores Tumorais/metabolismo , Membrana Celular/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Tetraspanina 24/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Células Epiteliais/metabolismo , Feminino , Redes Reguladoras de Genes , Humanos , Gradação de Tumores , Fenótipo , Prognóstico , Ensaios Antitumorais Modelo de Xenoenxerto , Homeobox 2 de Ligação a E-box com Dedos de Zinco/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
14.
Nat Commun ; 8: 13989, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28071763

RESUMO

Extracellular matrix adhesion is required for normal epithelial cell survival, nutrient uptake and metabolism. This requirement can be overcome by oncogene activation. Interestingly, inhibition of PI3K/mTOR leads to apoptosis of matrix-detached, but not matrix-attached cancer cells, suggesting that matrix-attached cells use alternate mechanisms to maintain nutrient supplies. Here we demonstrate that under conditions of dietary restriction or growth factor starvation, where PI3K/mTOR signalling is decreased, matrix-attached human mammary epithelial cells upregulate and internalize ß4-integrin along with its matrix substrate, laminin. Endocytosed laminin localizes to lysosomes, results in increased intracellular levels of essential amino acids and enhanced mTORC1 signalling, preventing cell death. Moreover, we show that starved human fibroblasts secrete matrix proteins that maintain the growth of starved mammary epithelial cells contingent upon epithelial cell ß4-integrin expression. Our study identifies a crosstalk between stromal fibroblasts and epithelial cells under starvation that could be exploited therapeutically to target tumours resistant to PI3K/mTOR inhibition.


Assuntos
Células Epiteliais/fisiologia , Matriz Extracelular/metabolismo , Integrina beta4/metabolismo , Laminina/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Fibroblastos/metabolismo , Humanos , Integrina beta4/genética , Laminina/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos Endogâmicos , Fosfatidilinositol 3-Quinases/metabolismo , Inanição
15.
JCI Insight ; 1(10)2016 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-27482544

RESUMO

High-grade serous ovarian carcinoma (HGS-OvCa) harbors p53 mutations and can originate from the epithelial cell compartment of the fallopian tube fimbriae. From this site, neoplastic cells detach, survive in the peritoneal cavity, and form cellular clusters that intercalate into the mesothelium to form ovarian and peritoneal masses. To examine the contribution of mutant p53 to phenotypic alterations associated with HGS-OvCA, we developed live-cell microscopy assays that recapitulate these early events in cultured fallopian tube nonciliated epithelial (FNE) cells. Expression of stabilizing mutant variants of p53, but not depletion of endogenous wild-type p53, in FNE cells promoted survival and cell-cell aggregation under conditions of cell detachment, leading to the formation of cell clusters with mesothelium-intercalation capacity. Mutant p53R175H-induced phenotypes were dependent on fibronectin production, α5ß1 fibronectin receptor engagement, and TWIST1 expression. These results indicate that FNE cells expressing stabilizing p53 mutants acquire anchorage independence and subsequent mesothelial intercalation capacity through a mechanism involving mesenchymal transition and matrix production. These findings provide important new insights into activities of mutant p53 in the cells of origin of HGS-OvCa.

16.
Genes Dev ; 30(7): 870, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27036968

RESUMO

In the above-mentioned article, it has come to the authors' attention that, during the preparation of Figure 5C and Supplemental Figure S2C for the final version of this article, the authors unintentionally assembled incorrect tubulin immunoblots due to similarities in the markings or names, such as FLT3 versus FT, between two similar experiments. The amended versions of these figures are shown below. Neither the quantitative determinations nor the conclusions of this article are altered. The authors apologize for these errors.

17.
Nat Rev Cancer ; 15(11): 668-79, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26493647

RESUMO

High-grade serous ovarian cancer (HGSOC) accounts for 70-80% of ovarian cancer deaths, and overall survival has not changed significantly for several decades. In this Opinion article, we outline a set of research priorities that we believe will reduce incidence and improve outcomes for women with this disease. This 'roadmap' for HGSOC was determined after extensive discussions at an Ovarian Cancer Action meeting in January 2015.


Assuntos
Cistadenocarcinoma Seroso/mortalidade , Cistadenocarcinoma Seroso/prevenção & controle , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/prevenção & controle , Cistadenocarcinoma Seroso/patologia , Feminino , Humanos , Gradação de Tumores , Neoplasias Ovarianas/patologia , Prognóstico , Taxa de Sobrevida
18.
J Clin Invest ; 124(6): 2611-25, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24762435

RESUMO

Metastatic dissemination of ovarian tumors involves the invasion of tumor cell clusters into the mesothelial cell lining of peritoneal cavity organs; however, the tumor-specific factors that allow ovarian cancer cells to spread are unclear. We used an in vitro assay that models the initial step of ovarian cancer metastasis, clearance of the mesothelial cell layer, to examine the clearance ability of a large panel of both established and primary ovarian tumor cells. Comparison of the gene and protein expression profiles of clearance-competent and clearance-incompetent cells revealed that mesenchymal genes are enriched in tumor populations that display strong clearance activity, while epithelial genes are enriched in those with weak or undetectable activity. Overexpression of transcription factors SNAI1, TWIST1, and ZEB1, which regulate the epithelial-to-mesenchymal transition (EMT), promoted mesothelial clearance in cell lines with weak activity, while knockdown of the EMT-regulatory transcription factors TWIST1 and ZEB1 attenuated mesothelial clearance in ovarian cancer cell lines with strong activity. These findings provide important insights into the mechanisms associated with metastatic progression of ovarian cancer and suggest that inhibiting pathways that drive mesenchymal programs may suppress tumor cell invasion of peritoneal tissues.


Assuntos
Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Transição Epitelial-Mesenquimal , Epitélio/metabolismo , Epitélio/patologia , Feminino , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Mesoderma/metabolismo , Mesoderma/patologia , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias Ovarianas/secundário , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/secundário , Análise Serial de Proteínas , Fatores de Transcrição da Família Snail , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma , Células Tumorais Cultivadas , Proteína 1 Relacionada a Twist/antagonistas & inibidores , Proteína 1 Relacionada a Twist/genética , Proteína 1 Relacionada a Twist/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco
19.
Cell Adh Migr ; 8(1): 42-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24589621

RESUMO

Nucleus movement, positioning, and orientation is precisely specified and actively regulated within cells, and it plays a critical role in many cellular and developmental processes. Mutation of proteins that regulate the nucleus anchoring and movement lead to diverse pathologies, laminopathies in particular, suggesting that the nucleus correct positioning and movement is essential for proper cellular function. In motile cells that polarize toward the direction of migration, the nucleus undergoes controlled rotation promoting the alignment of the nucleus with the axis of migration. Such spatial organization of the cell appears to be optimal for the cell migration. Nuclear reorientation requires the cytoskeleton to be anchored to the nuclear envelope, which exerts pulling or pushing torque on the nucleus. Here we discuss the possible molecular mechanisms regulating the nuclear rotation and reorientation and the significance of this type of nuclear movement for cell migration.

20.
PLoS One ; 8(11): e80314, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24303006

RESUMO

The potential role of the cell-of-origin in determining the tumor phenotype has been raised, but not adequately examined. We hypothesized that distinct cells-of-origin may play a role in determining ovarian tumor phenotype and outcome. Here we describe a new cell culture medium for in vitro culture of paired normal human ovarian (OV) and fallopian tube (FT) epithelial cells from donors without cancer. While these cells have been cultured individually for short periods of time, to our knowledge this is the first long-term culture of both cell types from the same donors. Through analysis of the gene expression profiles of the cultured OV/FT cells we identified a normal cell-of-origin gene signature that classified primary ovarian cancers into OV-like and FT-like subgroups; this classification correlated with significant differences in clinical outcomes. The identification of a prognostically significant gene expression signature derived solely from normal untransformed cells is consistent with the hypothesis that the normal cell-of-origin may be a source of ovarian tumor heterogeneity and the associated differences in tumor outcome.


Assuntos
Células Epiteliais/metabolismo , Tubas Uterinas/metabolismo , Perfilação da Expressão Gênica , Neoplasias Ovarianas/genética , Ovário/metabolismo , Transcriptoma , Animais , Linhagem Celular Transformada , Modelos Animais de Doenças , Feminino , Xenoenxertos , Humanos , Camundongos , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Cultura Primária de Células , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA