Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Int J Mol Sci ; 24(22)2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-38003636

RESUMO

The omentum is the predominant site of ovarian cancer metastasis, but it is difficult to remove the omentum in its entirety. There is a critical need for effective approaches that minimize the risk of colonization of preserved omental tissues by occult cancer cells. Normal saline (0.9% sodium chloride) is commonly used to wash the peritoneal cavity during ovarian cancer surgery. The omentum has a prodigious ability to absorb fluid in the peritoneal cavity, but the impact of normal saline on the omentum is poorly understood. In this review article, we discuss why normal saline is not a biocompatible solution, drawing insights from clinical investigations of normal saline in fluid resuscitation and from the cytopathologic evaluation of peritoneal washings. We integrate these insights with the unique biology of the omentum and omental metastasis, highlighting the importance of considering the absorptive ability of the omentum when administering agents into the peritoneal cavity. Furthermore, we describe insights from preclinical studies regarding the mechanisms by which normal saline might render the omentum conducive for colonization by cancer cells. Importantly, we discuss the possibility that the risk of colonization of preserved omental tissues might be minimized by using balanced crystalloid solutions for peritoneal washing.


Assuntos
Neoplasias Ovarianas , Neoplasias Peritoneais , Humanos , Feminino , Solução Salina/uso terapêutico , Cavidade Peritoneal/patologia , Neoplasias Peritoneais/secundário , Lavagem Peritoneal , Neoplasias Ovarianas/patologia
2.
Int J Mol Sci ; 21(15)2020 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-32751440

RESUMO

The tumor vasculature is essential for tumor growth and metastasis, and is a prime target of several anti-cancer agents. Increasing evidence indicates that tumor angiogenesis is stimulated by extracellular vesicles (EVs) that are secreted or shed by cancer cells. These EVs encapsulate a variety of biomolecules with angiogenic properties, and have been largely thought to stimulate vessel formation by transferring this luminal cargo into endothelial cells. However, recent studies have revealed that EVs can also signal to recipient cells via proteins on the vesicular surface. This review discusses and integrates emerging insights into the diverse mechanisms by which proteins associate with the EV membrane, the biological functions of EV membrane-associated proteins in tumor angiogenesis, and the clinical significance of these proteins in anti-angiogenic therapy.


Assuntos
Proteínas Angiogênicas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Vesículas Extracelulares/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias , Neovascularização Patológica/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
3.
J Cell Sci ; 128(16): 3055-67, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26208636

RESUMO

Megakaryocyte and erythroid development are tightly controlled by a repertoire of cytokines, but it is not clear how cytokine-activated signaling pathways are controlled during development of these two lineages. Here, we identify that expression of DLX4, a transcription factor encoded by a homeobox gene, increases during megakaryopoiesis but decreases during erythropoiesis. Enforced expression of DLX4 in CD34(+) stem and progenitor cells and in bipotent K562 cells induced lineage markers and morphologic features of megakaryocytes and repressed erythroid marker expression and hemoglobin levels. Converse results were obtained when DLX4 was knocked down. Gene Ontology and Gene Set Enrichment Analyses of genome-wide changes in gene expression revealed that DLX4 induces a megakaryocytic transcriptional program and inhibits an erythroid transcriptional program. DLX4 also induced gene signatures that are associated with nuclear factor κB (NF-κB) signaling. The ability of DLX4 to promote megakaryocyte development at the expense of erythroid generation was diminished by blocking NF-κB activity or by repressing IL1B, a transcriptional target of DLX4. Collectively, our findings indicate that DLX4 exerts opposing effects on the megakaryocytic and erythroid lineages in part by inducing IL-1ß and NF-κB signaling.


Assuntos
Diferenciação Celular/genética , Eritropoese/genética , Proteínas de Homeodomínio/biossíntese , Interleucina-1beta/genética , Células Progenitoras de Megacariócitos e Eritrócitos/citologia , Fatores de Transcrição/biossíntese , Linhagem da Célula/genética , Eritrócitos/citologia , Eritrócitos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Humanos , Interleucina-1beta/antagonistas & inibidores , Células K562 , Megacariócitos/citologia , Megacariócitos/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética
4.
Am J Pathol ; 185(8): 2298-308, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26067154

RESUMO

Ovarian cancers often highly express inflammatory cytokines and form implants throughout the peritoneal cavity. However, the mechanisms that drive inflammatory signaling and peritoneal metastasis of ovarian cancer are poorly understood. We previously identified that high expression of DLX4, a transcription factor encoded by a homeobox gene, is associated with reduced survival of ovarian cancer patients. In this study, we identified that DLX4 stimulates attachment of ovarian tumor cells to peritoneal mesothelial cells in vitro and increases the numbers of peritoneal implants in xenograft models. DLX4 induced expression of the cell surface molecule CD44 in ovarian tumor cells, and inhibition of CD44 abrogated the ability of DLX4 to stimulate tumor-mesothelial cell interactions. The induction of CD44 by DLX4 was attributed to increased activity of NF-κB that was stimulated by the inflammatory cytokine IL-1ß, a transcriptional target of DLX4. The stimulatory effects of DLX4 on CD44 levels and tumor-mesothelial cell interactions were abrogated when IL-1ß or NF-κB was inhibited in tumor cells. Furthermore, DLX4 expression levels strongly correlated with NF-κB activation and disease stage in clinical specimens of ovarian cancer. Collectively, these findings indicate that DLX4 induces CD44 by stimulating IL-1ß-mediated NF-κB activity, thereby promoting peritoneal metastasis of ovarian cancer.


Assuntos
Células Epiteliais/metabolismo , Proteínas de Homeodomínio/metabolismo , Receptores de Hialuronatos/metabolismo , NF-kappa B/metabolismo , Neoplasias Ovarianas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Células Epiteliais/patologia , Feminino , Proteínas de Homeodomínio/genética , Humanos , Receptores de Hialuronatos/genética , Interleucina-1beta/metabolismo , Camundongos , Camundongos Nus , NF-kappa B/genética , Metástase Neoplásica/patologia , Neoplasias Ovarianas/patologia , Peritônio/metabolismo , Peritônio/patologia , Fosforilação , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética
5.
Mol Cancer ; 14: 97, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25924901

RESUMO

BACKGROUND: Homeobox genes encode transcription factors that control patterning of virtually all organ systems including the vasculature. Tumor angiogenesis is stimulated by several homeobox genes that are overexpressed in tumor cells, but the mechanisms of these genes are poorly understood. In this study, we investigated the mechanisms by which DLX4, a homeobox gene that is associated with increased tumor microvessel density, stimulates ovarian tumor angiogenesis. METHODS: Expression of DLX4 and nitric oxide synthases was analyzed in publicly available transcriptional profiles of ovarian cancer clinical specimens. Levels of inducible nitric oxide synthase (iNOS) were evaluated by quantitative RT-PCR, flow cytometry and nitric oxide assays using ovarian cancer cell lines in which DLX4 was overexpressed or knocked down. Signal Transducer and Activator of Transcription 1 (STAT1) expression and activity were evaluated by luciferase reporter assays, immunofluorescence staining, Western blot and immunoprecipitation. Endothelial cell growth and tumor angiogenesis were evaluated in in vitro assays and xenograft models. RESULTS: We identified that DLX4 induces expression of iNOS, an enzyme that stimulates angiogenesis by generating nitric oxide. Analysis of datasets of two independent patient cohorts revealed that high DLX4 expression in ovarian cancer is strongly associated with elevated expression of iNOS but not of other nitric oxide synthases. Studies using STAT1-expressing and STAT1-deficient cells revealed that DLX4 interacts with STAT1 and induces iNOS expression in part by stimulating STAT1 activity. Expression of DLX4 in ovarian cancer cells stimulated endothelial cell growth in vitro and increased microvessel density in xenograft models, and these stimulatory effects of DLX4 were abrogated when its induction of iNOS was inhibited. CONCLUSION: These findings indicate that DLX4 promotes ovarian tumor angiogenesis in part by stimulating iNOS expression.


Assuntos
Proteínas de Homeodomínio/metabolismo , Neovascularização Patológica/enzimologia , Óxido Nítrico Sintase Tipo II/metabolismo , Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/enzimologia , Fatores de Transcrição/metabolismo , Animais , Ascite/patologia , Linhagem Celular Tumoral , Proliferação de Células , Células Endoteliais/metabolismo , Indução Enzimática , Feminino , Humanos , Camundongos Nus , Neovascularização Patológica/patologia , Neoplasias Ovarianas/patologia , Fator de Transcrição STAT1/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Am J Pathol ; 184(1): 271-81, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24332016

RESUMO

Tumor-associated macrophages (TAMs) exhibit an M2 macrophage phenotype that suppresses anti-tumor immune responses and often correlates with poor outcomes in patients with cancer. Patients with ovarian cancer frequently present with peritoneal carcinomatosis, but the mechanisms that induce naïve peritoneal macrophages into TAMs are poorly understood. In this study, we found an increased abundance of TAMs in mouse i.p. xenograft models of ovarian cancer that expressed HOXA9, a homeobox gene that is associated with poor prognosis in patients with ovarian cancer. HOXA9 expression in ovarian cancer cells stimulated chemotaxis of peritoneal macrophages and induced macrophages to acquire TAM-like features. These features included induction of the M2 markers, CD163 and CD206, and the immunosuppressive cytokines, IL-10 and chemokine ligand 17, and down-regulation of the immunostimulatory cytokine, IL-12. HOXA9-mediated induction of TAMs was primarily due to the combinatorial effects of HOXA9-induced, tumor-derived transforming growth factor-ß2 and chemokine ligand 2 levels. High HOXA9 expression in clinical specimens of ovarian cancer was strongly associated with increased abundance of TAMs and intratumoral T-regulatory cells and decreased abundance of CD8(+) tumor-infiltrating lymphocytes. Levels of immunosuppressive cytokines were also elevated in ascites fluid of patients with tumors that highly expressed HOXA9. HOXA9 may, therefore, stimulate ovarian cancer progression by promoting an immunosuppressive microenvironment via paracrine effects on peritoneal macrophages.


Assuntos
Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/metabolismo , Macrófagos Peritoneais/metabolismo , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Animais , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Humanos , Imuno-Histoquímica , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Nus , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Mol Cancer ; 13: 170, 2014 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-25023983

RESUMO

BACKGROUND: Epithelial ovarian cancer (EOC) is a lethal disease that frequently involves the peritoneal cavity. Dissemination of EOC is a multi-step process in which exfoliated tumor cells survive in the peritoneal fluid as multi-cellular aggregates and then form invasive implants on peritoneal surfaces. The mechanisms that control this process are poorly understood. We previously identified that high expression of the developmental patterning gene HOXA9 is associated with poor survival in EOC patients. In this study, we investigated the significance and mechanisms of HOXA9 in controlling aggregation and implantation of floating EOC cells. METHODS: HOXA9 was inhibited by shRNAs or expressed in EOC cells that were propagated in suspension cultures and in the peritoneal cavity of mice. Cell death was assayed by flow cytometry and ELISA. Cell aggregation, attachment and migration were evaluated by microscopy, transwell chamber assays and histopathologic analysis. DNA-binding of HOXA9 and its effect on expression of the cell adhesion molecule P-cadherin were assayed by chromatin immunoprecipitation, quantitative RT-PCR and Western blot. HOXA9 and P-cadherin expression was evaluated in publicly available datasets of EOC clinical specimens. RESULTS: We identified that HOXA9 promotes aggregation and inhibits anoikis in floating EOC cells in vitro and in xenograft models. HOXA9 also stimulated the ability of EOC cells to attach to peritoneal cells and to migrate. HOXA9 bound the promoter of the CDH3 gene that encodes P-cadherin, induced CDH3 expression in EOC cells, and was associated with increased CDH3 expression in clinical specimens of EOC. Inhibiting P-cadherin in EOC cells that expressed HOXA9 abrogated the stimulatory effects of HOXA9 on cell aggregation, implantation and migration. Conversely, these stimulatory effects of HOXA9 were restored when P-cadherin was reconstituted in EOC cells in which HOXA9 was inhibited. CONCLUSION: These findings indicate that HOXA9 contributes to poor outcomes in EOC in part by promoting intraperitoneal dissemination via its induction of P-cadherin.


Assuntos
Caderinas/metabolismo , Proteínas de Homeodomínio/genética , Neoplasias Ovarianas/genética , Neoplasias Peritoneais/genética , Animais , Comunicação Celular/genética , Feminino , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Metástase Neoplásica , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/secundário , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Nat Rev Cancer ; 5(5): 355-66, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15864277

RESUMO

Despite considerable efforts to improve early detection, and advances in chemotherapy, metastasis remains a major challenge in the clinical management of ovarian cancer. Studies of new murine models are providing novel insights into the pathophysiology of ovarian cancer, but these models are not readily amenable to genetic screens. Genetic analysis of border-cell migration in the Drosophila melanogaster ovary provides clues that will improve our understanding of ovarian cancer metastasis at the molecular level, and also might lead to potential therapeutic targets.


Assuntos
Movimento Celular , Drosophila melanogaster/genética , Modelos Animais , Metástase Neoplásica/genética , Neoplasias Ovarianas/patologia , Animais , Ascite/etiologia , Caderinas/fisiologia , Adesão Celular , Feminino , Substâncias de Crescimento/fisiologia , Ovário/citologia , Transdução de Sinais
9.
Front Mol Biosci ; 11: 1356780, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38449696

RESUMO

Extracellular microRNAs (miRNAs) can be detected in body fluids and hold great potential as cancer biomarkers. Extracellular miRNAs are protected from degradation by binding various proteins and through their packaging into extracellular vesicles (EVs). There is evidence that the diagnostic performance of cancer-associated extracellular miRNAs can be improved by assaying EV-miRNA instead of total cell-free miRNA, but several challenges have hampered the advancement of EV-miRNA in liquid biopsy. Because almost all types of cells release EVs, cancer cell-derived EVs might constitute only a minor fraction of EVs in body fluids of cancer patients with low volume disease. Furthermore, a given cell type can release several subpopulations of EVs that vary in their cargo, and there is evidence that the majority of EVs contain low copy numbers of miRNAs. In this mini-review, we discuss the potential of several candidate EV membrane proteins such as CD147 to define cancer cell-derived EVs, and approaches by which subpopulations of miRNA-rich EVs in body fluids might be identified. By integrating these insights, we discuss strategies by which EVs that are both cancer cell-derived and miRNA-rich could be isolated to enhance the diagnostic performance of extracellular miRNAs.

10.
JCI Insight ; 8(12)2023 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-37345662

RESUMO

The omentum contains immune cell structures called milky spots that are niches for transcoelomic metastasis. It is difficult to remove the omentum completely, and there are no effective strategies to minimize the risk of colonization of preserved omental tissues by cancer cells that circulate in the peritoneal fluid. Normal saline is commonly administered into the peritoneal cavity for diagnostic and intraoperative lavage. Here we show that normal saline, when administered into the peritoneal cavity of mice, is prominently absorbed by the omentum, exfoliates its mesothelium, and induces expression of CX3CL1, the ligand for CX3CR1, within and surrounding the omental vasculature. Studies using CX3CR1-competent and CX3CR1-deficient mice showed that the predominant response in the omentum following saline administration is an accumulation of CX3CR1+ monocytes/macrophages that expand milky spots and promote neoangiogenesis within these niches. Moreover, saline administration promoted the implantation of cancer cells of ovarian and colorectal origin onto the omentum. By contrast, these deleterious effects were not observed following i.p. administration of lactated Ringer's solution. Our findings suggest that normal saline stimulates the receptivity of the omentum for cancer cells and that the risk of colonization can be minimized by using a biocompatible crystalloid for lavage procedures.


Assuntos
Omento , Solução Salina , Animais , Camundongos , Líquido Ascítico , Implantação do Embrião , Epitélio
11.
J Extracell Vesicles ; 12(4): e12318, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36973758

RESUMO

Extracellular vesicles (EVs) are ideal for liquid biopsy, but distinguishing cancer cell-derived EVs and subpopulations of biomarker-containing EVs in body fluids has been challenging. Here, we identified that the glycoproteins CD147 and CD98 define subpopulations of EVs that are distinct from classical tetraspanin+ EVs in their biogenesis. Notably, we identified that CD147+ EVs have substantially higher microRNA (miRNA) content than tetraspanin+ EVs and are selectively enriched in miRNA through the interaction of CD147 with heterogeneous nuclear ribonucleoprotein A2/B1. Studies using mouse xenograft models showed that CD147+ EVs predominantly derive from cancer cells, whereas the majority of tetraspanin+ EVs are not of cancer cell origin. Circulating CD147+ EVs, but not tetraspanin+ EVs, were significantly increased in prevalence in patients with ovarian and renal cancers as compared to healthy individuals and patients with benign conditions. Furthermore, we found that isolating miRNAs from body fluids by CD147 immunocapture increases the sensitivity of detecting cancer cell-specific miRNAs, and that circulating miRNAs isolated by CD147 immunocapture more closely reflect the tumor miRNA signature than circulating miRNAs isolated by conventional methods. Collectively, our findings reveal that CD147 defines miRNA-enriched, cancer cell-derived EVs, and that CD147 immunocapture could be an effective approach to isolate cancer-derived miRNAs for liquid biopsy.


Assuntos
MicroRNA Circulante , Vesículas Extracelulares , MicroRNAs , Neoplasias , Animais , Camundongos , Humanos , MicroRNAs/genética , Vesículas Extracelulares/genética , Biomarcadores , Biópsia Líquida
12.
Nat Med ; 11(5): 531-7, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15821746

RESUMO

Although epithelial ovarian cancers (EOCs) have been thought to arise from the simple epithelium lining the ovarian surface or inclusion cysts, the major subtypes of EOCs show morphologic features that resemble those of the müllerian duct-derived epithelia of the reproductive tract. We found that HOX genes, which normally regulate mullerian duct differentiation, are not expressed in normal ovarian surface epithelium (OSE), but are expressed in different EOC subtypes according to the pattern of mullerian-like differentiation of these cancers. Ectopic expression of Hoxa9 in tumorigenic mouse OSE cells gave rise to papillary tumors resembling serous EOCs. In contrast, Hoxa10 and Hoxa11 induced morphogenesis of endometrioid-like and mucinous-like EOCs, respectively. Hoxa7 showed no lineage specificity, but promoted the abilities of Hoxa9, Hoxa10 and Hoxa11 to induce differentiation along their respective pathways. Therefore, inappropriate activation of a molecular program that controls patterning of the reproductive tract could explain the morphologic heterogeneity of EOCs and their assumption of müllerian-like features.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Ductos Paramesonéfricos/fisiologia , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/metabolismo , Animais , Diferenciação Celular/fisiologia , Primers do DNA , DNA Complementar/genética , Feminino , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Ductos Paramesonéfricos/citologia , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
13.
Commun Biol ; 2: 386, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31646189

RESUMO

Cancer-derived small extracellular vesicles (sEVs) induce stromal cells to become permissive for tumor growth. However, it is unclear whether this induction solely occurs through transfer of vesicular cargo into recipient cells. Here we show that cancer-derived sEVs can stimulate endothelial cell migration and tube formation independently of uptake. These responses were mediated by the 189 amino acid isoform of vascular endothelial growth factor (VEGF) on the surface of sEVs. Unlike other common VEGF isoforms, VEGF189 preferentially localized to sEVs through its high affinity for heparin. Interaction of VEGF189 with the surface of sEVs profoundly increased ligand half-life and reduced its recognition by the therapeutic VEGF antibody bevacizumab. sEV-associated VEGF (sEV-VEGF) stimulated tumor xenograft growth but was not neutralized by bevacizumab. Furthermore, high levels of sEV-VEGF were associated with disease progression in bevacizumab-treated cancer patients, raising the possibility that resistance to bevacizumab might stem in part from elevated levels of sEV-VEGF.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Bevacizumab/farmacologia , Neovascularização Patológica/etiologia , Microambiente Tumoral/fisiologia , Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular Tumoral , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Vesículas Extracelulares/metabolismo , Feminino , Heparina/metabolismo , Humanos , Camundongos , Camundongos Nus , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Microambiente Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Exp Med ; 216(1): 176-194, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30567719

RESUMO

Ovarian cancer preferentially metastasizes to the omentum, a fatty tissue characterized by immune structures called milky spots, but the cellular dynamics that direct this tropism are unknown. Here, we identified that neutrophil influx into the omentum is a prerequisite premetastatic step in orthotopic ovarian cancer models. Ovarian tumor-derived inflammatory factors stimulated neutrophils to mobilize and extrude chromatin webs called neutrophil extracellular traps (NETs). NETs were detected in the omentum of ovarian tumor-bearing mice before metastasis and of women with early-stage ovarian cancer. NETs, in turn, bound ovarian cancer cells and promoted metastasis. Omental metastasis was decreased in mice with neutrophil-specific deficiency of peptidylarginine deiminase 4 (PAD4), an enzyme that is essential for NET formation. Blockade of NET formation using a PAD4 pharmacologic inhibitor also decreased omental colonization. Our findings implicate NET formation in rendering the premetastatic omental niche conducive for implantation of ovarian cancer cells and raise the possibility that blockade of NET formation prevents omental metastasis.


Assuntos
Armadilhas Extracelulares/metabolismo , Neoplasias Experimentais/metabolismo , Neutrófilos/metabolismo , Omento/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Peritoneais/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Hidrolases/metabolismo , Camundongos , Camundongos Nus , Metástase Neoplásica , Neoplasias Experimentais/patologia , Neutrófilos/patologia , Omento/patologia , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/secundário , Proteína-Arginina Desiminase do Tipo 4
15.
Cancer Res ; 66(2): 889-97, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16424022

RESUMO

Homeobox genes encode transcription factors that control cell differentiation and play essential roles in developmental patterning. Increasing evidence indicates that many homeobox genes are aberrantly expressed in cancers, and that their deregulation significantly contributes to tumor progression. The homeobox gene HOXA10 controls uterine organogenesis during embryonic development and functional endometrial differentiation in the adult. We investigated whether HOXA10 expression is deregulated in endometrial carcinomas, and how counteracting this aberrant expression modifies tumor behavior. We found that down-regulation of HOXA10 expression in endometrial carcinomas strongly correlates with increased tumor grade and is associated with methylation of the HOXA10 promoter. Enforced expression of HOXA10 in endometrial carcinoma cells inhibited invasive behavior in vitro and tumor dissemination in nude mice. The inhibitory effect of HOXA10 on invasive behavior was attributable at least in part to the ability of HOXA10 to induce expression of the epithelial cell adhesion molecule E-cadherin by down-regulating expression of Snail, a repressor of E-cadherin gene transcription. These findings reveal a novel role for HOXA10 deregulation in the progression of endometrial carcinoma by promoting epithelial-mesenchymal transition.


Assuntos
Carcinoma/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/fisiologia , Neoplasias do Endométrio/genética , Animais , Carcinoma/fisiopatologia , Adesão Celular , Progressão da Doença , Regulação para Baixo , Neoplasias do Endométrio/fisiopatologia , Epitélio/fisiologia , Feminino , Regulação da Expressão Gênica , Proteínas Homeobox A10 , Proteínas de Homeodomínio , Humanos , Mesoderma/fisiologia , Camundongos , Camundongos Nus , Invasividade Neoplásica , Células Tumorais Cultivadas
16.
Expert Rev Mol Med ; 9(13): 1-12, 2007 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-17477890

RESUMO

Epithelial ovarian cancer comprises several subtypes of tumours that exhibit diverse histopathological features. The intriguing assumption by many epithelial ovarian cancers of specialised features of nonovarian tissue lineages has promoted considerable debate as to whether these tumours arise from the deceptively simple surface epithelium of the ovary. This review focuses on recent molecular and pathological studies of epithelial ovarian cancers that support and challenge their surface-epithelial derivation, and discusses the findings in the context of current views of the 'cell-of-origin' of solid tumours.


Assuntos
Neoplasias Ovarianas/etiologia , Neoplasias Ovarianas/patologia , Linhagem da Célula , Células Epiteliais/patologia , Feminino , Humanos , Neoplasias Ovarianas/classificação
17.
Cancer Res ; 64(10): 3550-8, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15150111

RESUMO

Constitutive activation of the Janus-activated kinase/signal transducer and activator of transcription (STAT) pathway promotes the proliferation and survival of cancer cells in culture and is associated with various cancers, including those of the ovary. We found that constitutively activated STAT3 levels correlated with aggressive clinical behavior of ovarian carcinoma specimens. Furthermore, inhibition of STAT3 reduced the motility of ovarian cancer cells in vitro. Surprisingly, we found that activated STAT3 localized not only to nuclei but also to focal adhesions in these cells. Activated STAT3 coimmunoprecipitated with phosphorylated paxillin and focal adhesion kinase and required paxillin and Src for its localization to focal adhesions. These results suggest that Janus-activated kinase/STAT signaling may contribute to ovarian cancer cell invasiveness.


Assuntos
Movimento Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Adesões Focais/metabolismo , Neoplasias Ovarianas/metabolismo , Transativadores/fisiologia , Animais , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/patologia , Feminino , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Adesões Focais/patologia , Humanos , Camundongos , Células NIH 3T3 , Neoplasias Ovarianas/patologia , Paxilina , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Fator de Transcrição STAT3 , Transativadores/metabolismo
18.
Eur J Cancer ; 41(16): 2428-37, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16199152

RESUMO

Homeobox genes encode transcription factors that play essential roles in controlling cell growth and differentiation during embryonic development. Many homeobox genes are aberrantly expressed in a wide variety of solid tumours, and their deregulation appears to enhance cell survival and proliferation and to inhibit differentiation. In hematologic malignancies, deregulated homeobox genes profoundly perturb self-renewal and proliferation of hematopoietic stem cells and progenitors. It is increasingly recognised that solid tumours, like hematologic malignancies, could arise from cancer stem cells, and that targeting these cells could be the most effective means of inhibiting tumour progression and disease recurrence. Studying the biological effects and mechanisms of homeobox genes in cancers could provide valuable insights into identifying cancer stem cells and targeting the self-renewal pathways in these cell populations.


Assuntos
Genes Homeobox/fisiologia , Neoplasias/genética , Transformação Celular Neoplásica/genética , Expressão Gênica/genética , Genes Homeobox/genética , Genes Supressores de Tumor/fisiologia , Humanos , Células-Tronco/fisiologia
19.
Mol Cell Endocrinol ; 245(1-2): 77-85, 2005 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-16298470

RESUMO

Border cell migration is a process that occurs during Drosophila ovarian development in which cells derived from a simple epithelium migrate and invade neighboring tissue. This process resembles the behavior of cancerous cells that derive from the simple epithelium of the human ovary. One important regulator of border cell migration is Taiman, a homolog of steroid receptor coactivator-3 (SRC-3). Because increasing evidence indicates that similarities exist between the molecular control of migration of border cells and of cancer cells, we investigated whether SRC-3 controls ovarian cancer cell migration. Little or no SRC-3 expression was detected in normal ovarian surface epithelium, ovarian cysts and borderline ovarian tumors that lack stromal invasion. In contrast, SRC-3 was abundantly expressed in high-grade ovarian carcinomas. Inhibiting SRC-3 expression in ovarian cancer cells markedly reduced cell spreading and migration, and altered intracellular localization of focal adhesion kinase. This inhibitory effect on cell migration was independent of the estrogen receptor (ER) status of the cells. These studies reveal a novel role for SRC-3 in ovarian cancer progression by promoting cell migration, independently of its role in estrogen receptor signaling.


Assuntos
Acetiltransferases/fisiologia , Movimento Celular , Proteínas Oncogênicas/fisiologia , Neoplasias Ovarianas/patologia , Transativadores/fisiologia , Acetiltransferases/química , Acetiltransferases/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proteínas de Drosophila , Feminino , Proteína-Tirosina Quinases de Adesão Focal/análise , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Regulação Neoplásica da Expressão Gênica , Histona Acetiltransferases , Humanos , Coativador 3 de Receptor Nuclear , Proteínas Oncogênicas/química , Proteínas Oncogênicas/genética , Neoplasias Ovarianas/química , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/genética , Ovário/citologia , Ovário/fisiologia , RNA Interferente Pequeno , Receptores de Estrogênio/análise , Receptores de Estrogênio/fisiologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais/fisiologia , Transativadores/química , Transativadores/genética , Fatores de Transcrição/química
20.
Front Oncol ; 4: 18, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24567915

RESUMO

The majority of women who are diagnosed with epithelial ovarian cancer present with extensive peritoneal carcinomatosis and are rarely cured by conventional chemotherapy. Ovarian cancer cells typically disseminate by shedding into the peritoneal fluid and implant on the mesothelium-lined peritoneal surfaces that overlie connective and white adipose tissues. Emerging evidence indicates that ovarian tumor progression is orchestrated by dynamic interplay between tumor cells and a variety of stromal cells such as adipocytes, endothelial cells, fibroblasts, mesenchymal stem cells, macrophages, and other immune cells. This mini-review discusses the biological significance of the heterotypic cellular interactions in the ovarian tumor microenvironment and the therapeutic implications of targeting these interactions.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa