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













Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1295208, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38235131

RESUMEN

Introduction: Epithelial ovarian cancer (OC) stands as one of the deadliest gynecologic malignancies, urgently necessitating novel therapeutic strategies. Approximately 60% of ovarian tumors exhibit reduced expression of major histocompatibility complex class I (MHC I), intensifying immune evasion mechanisms and rendering immunotherapies ineffective. NOD-like receptor CARD domain containing 5 (NLRC5) transcriptionally regulates MHC I genes and many antigen presentation machinery components. We therefore explored the therapeutic potential of NLRC5 in OC. Methods: We generated OC cells overexpressing NLRC5 to rescue MHC I expression and antigen presentation and then assessed their capability to respond to PD-L1 blockade and an infected cell vaccine. Results: Analysis of microarray datasets revealed a correlation between elevated NLRC5 expression and extended survival in OC patients; however, NLRC5 was scarcely detected in the OC tumor microenvironment. OC cells overexpressing NLRC5 exhibited slower tumor growth and resulted in higher recruitment of leukocytes in the TME with lower CD4/CD8 T-cell ratios and increased activation of T cells. Immune cells from peripheral blood, spleen, and ascites from these mice displayed heightened activation and interferon-gamma production when exposed to autologous tumor-associated antigens. Finally, as a proof of concept, NLRC5 overexpression within an infected cell vaccine platform enhanced responses and prolonged survival in comparison with control groups when challenged with parental tumors. Discussion: These findings provide a compelling rationale for utilizing NLRC5 overexpression in "cold" tumor models to enhance tumor susceptibility to T-cell recognition and elimination by boosting the presentation of endogenous tumor antigens. This approach holds promise for improving antitumoral immune responses in OC.


Asunto(s)
Neoplasias Ováricas , Vacunas , Humanos , Femenino , Animales , Ratones , Proteínas NLR , Dominio de Reclutamiento y Activación de Caspasas , Microambiente Tumoral , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Antígenos de Histocompatibilidad Clase I , Neoplasias Ováricas/genética , Antígenos de Neoplasias
2.
Exp Ther Med ; 23(6): 412, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35601066

RESUMEN

In adult tissue, the paired box 2 (PAX2) protein is expressed in healthy oviductal, but not normal ovarian surface epithelial cells. PAX2 is expressed in a subset of cases of serous ovarian carcinoma; however, the role of PAX2 in the initiation and progression of ovarian cancer remains unknown. The aim of the present study was to determine the biological effects of PAX2 expression in normal and cancerous epithelial cells. By culturing the normal and cancerous ovarian cells that express PAX2 in 3D culture and staining the cells with vasculogenic mimicry markers such as CD31 and PAS, it was shown that PAX2 overexpression in both normal and cancerous ovarian epithelial cells induced formation of vascular-like structures both in vitro and in vivo. These results indicated a potential role of PAX2 in ovarian cancer progression by increasing the presence of vascular-like structures to promote the supply of nutrients to tumor cells and facilitate cancer cell proliferation and invasion.

3.
Int J Oncol ; 56(3): 697-708, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31922217

RESUMEN

Ovarian cancer is the fifth most common type of cancer afflicting women and frequently presents at a late stage with a poor prognosis. While paired box 2 (PAX2) expression is frequently lost in high­grade serous ovarian cancer, it is expressed in a subset of ovarian tumors and may play a role in tumorigenesis. This study investigated the expression of PAX2 in ovarian cancer. The expression of PAX2 in a murine allograft model of ovarian cancer, the RM model, led to a more rapidly growing cell line both in vitro and in vivo. This finding was in accordance with the shorter progression­free survival observed in patients with a higher PAX2 expression, as determined in this study cohort by immunohistochemistry. iTRAQ­based proteomic profiling revealed that proteins involved in fatty acid metabolism and oxidative phosphorylation were found to be upregulated in RM tumors expressing PAX2. The expression of two key fatty acid metabolic genes was also found to be upregulated in PAX2­expressing human ovarian cancer samples. The analysis of existing datasets also indicated that a high expression of key enzymes in fatty acid metabolism was associated with a shorter progression­free survival time in patients with serous ovarian cancer. Thus, on the whole, the findings of this study indicate that PAX2 may promote ovarian cancer progression, involving fatty acid metabolic reprograming.


Asunto(s)
Carcinoma Epitelial de Ovario/patología , Ácidos Grasos/metabolismo , Neoplasias Ováricas/patología , Factor de Transcripción PAX2/metabolismo , Animales , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/mortalidad , Línea Celular , Proliferación Celular , Transformación Celular Neoplásica , Progresión de la Enfermedad , Femenino , Humanos , Ratones , Ratones SCID , Mitocondrias/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/mortalidad , Fosforilación Oxidativa , Factor de Transcripción PAX2/genética , Pronóstico , Supervivencia sin Progresión
4.
Oncogene ; 37(44): 5873-5886, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29973689

RESUMEN

Estrogenic hormone replacement therapy increases the risk of developing ovarian cancer, and estrogen promotes tumour initiation and growth in mouse models of this disease. GREB1 (Growth regulation by estrogen in breast cancer 1) is an ESR1 (estrogen receptor 1)-upregulated protein which may mediate estrogen action. GREB1 knockdown prevents hormone-driven proliferation of several breast and prostate cancer cell lines and prolongs survival of mice engrafted with ovarian cancer cells, but its mechanism of action remains unclear. In this study, we explored GREB1 function in ovarian cancer. GREB1 overexpression in ovarian cancer cell lines increased cell proliferation and migration and promoted a mesenchymal morphology associated with increased Col1a2, which encodes a collagen I subunit. GREB1 knockdown inhibited proliferation and promoted an epithelial morphology associated with decreased Col1a2. In human tissues, GREB1 was expressed in all ESR1-expressing tissues throughout the normal female reproductive tract, in addition to several tissues that did not show ESR1 expression. In a TMA of ovarian cancer cases, GREB1 was expressed in 75-85% of serous, endometrioid, mucinous, and clear cell carcinomas. Serous, endometrioid, and mucinous ovarian cancers were almost always positive for either ESR1 or GREB1, suggesting a possible reliance on signalling through ESR1 and/or GREB1. Targeting GREB1 may inhibit tumour-promoting pathways both downstream and independent of ESR1 and is therefore a possible treatment strategy worthy of further investigation.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/metabolismo , Animales , Línea Celular Tumoral , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Transición Epitelial-Mesenquimal , Receptor alfa de Estrógeno/genética , Estrógenos/genética , Estrógenos/fisiología , Femenino , Humanos , Ratones , Ratones Mutantes , Ratones SCID , Proteínas de Neoplasias/genética , Neoplasias Ováricas/genética
5.
Oncotarget ; 8(44): 76881-76897, 2017 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-29100356

RESUMEN

Recent studies have provided evidence that the secretory cells of the fallopian tube (oviduct) are a probable origin for high-grade serous ovarian carcinoma. In addition to secretory cells, the fallopian tube epithelium consists of ciliated cells and CD44+ undifferentiated stem-like cells. Loss of PAX2 expression is recognized as an early event in epithelial transformation, but the specific role of PAX2 in this transition is unknown. The aim of this study was to define the role of PAX2 in oviductal epithelial (OVE) cells and its response to transforming growth factor ß1 (TGFß), characterizing specifically its potential involvement in regulating stem cell-like behaviors that may contribute to formation of cancer-initiating cells. Treatment of primary cultures of mouse OVE cells with TGFß induced an epithelial-mesenchymal transition (EMT) associated with decreased expression of PAX2 and an increase in the fraction of cells expressing CD44. PAX2 knockdown in OVE cells and overexpression in ovarian epithelial cells confirmed that PAX2 inhibits stem cell characteristics and regulates the degree of epithelial differentiation of OVE cells. These results suggest that loss of PAX2, as occurs in serous tubal intraepithelial carcinomas, may shift secretory cells to a more mesenchymal phenotype associated with stem-like features.

6.
J Nucl Med ; 58(2): 221-227, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27635026

RESUMEN

Oncolytic virus (OV) therapy has emerged as a novel tool in our therapeutic arsenals for fighting cancer. As a live biologic agent, OV has the ability to target and selectively amplify at the tumor sites. We have reported that a vaccinia-based OV (Pexa-Vec) has shown good efficacy in preclinical models and in clinical trials. To give an additional tool to clinicians to allow both treatment of the tumor and improved visualization of tumor margins, we developed new viral-based platforms with 2 specific gene reporters. METHODS: We incorporated the human sodium iodide symporter (hNIS) and the human somatostatin receptor 2 (hSSR2) in the vaccinia-based OV and tested viral constructs for their abilities to track and treat tumor development in vivo. RESULTS: Early and high-level expression of hNIS is detrimental to the recombinant virus, leading to the aggregation of hNIS protein and early cell death. Putting hNIS under a late synthetic promoter allowed a higher functional expression of the protein and much stronger 123I or 99Tc uptake. In vivo, the hNIS-containing virus infected and amplified in the tumor site, showing a better efficacy than the parental virus. The hNIS expression at the tumor site allowed for the imaging of viral infection and tumor regression. Similarly, hSSR2-containing OV vaccinia infected and lysed cancer cells. CONCLUSION: When tumor-bearing mice were given hNIS- and hSSR2-containing OV, 99Tc and 111In signals coalesced at the tumor, highlighting the power of using these viruses for tumor diagnosis and treatment.


Asunto(s)
Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/terapia , Viroterapia Oncolítica/métodos , Receptores de Somatostatina/genética , Simportadores/genética , Virus Vaccinia/fisiología , Animales , Línea Celular Tumoral , Femenino , Genes Reporteros/genética , Humanos , Ratones , Ratones Desnudos , Neoplasias Experimentales/virología , Virus Oncolíticos/fisiología , Tomografía de Emisión de Positrones/métodos , Recombinación Genética/genética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Nanomedicina Teranóstica/métodos , Resultado del Tratamiento , Regulación hacia Arriba/genética
7.
Cancer Prev Res (Phila) ; 8(12): 1163-73, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26373819

RESUMEN

PAX2 is an essential transcription factor for development. Aberrant PAX2 expression in adult tissues is associated with carcinogenesis and experimental evidence shows that PAX2 generally exhibits oncogenic properties. Although PAX2 is not expressed in normal ovaries, it is highly expressed in low malignant potential and low-grade epithelial ovarian tumors, suggesting that PAX2 induction in ovarian surface epithelium (OSE) may contribute to transformation. Herein, we provide evidence that expression of PAX2 in normal murine OSE cells (mOSE) enhances their proliferation and survival and, with loss of p53, induces tumorigenicity. PAX2 expression in murine ovarian cancer cells enhanced or inhibited tumorigenicity, depending on the model system. In RM cells (mOSE transformed by K-RAS and c-MYC), PAX2 expression inhibited p53 and induced pERK1/2 and COX2, resulting in enhanced angiogenesis and decreased apoptosis of tumors arising from these cells. However, in a murine model of high-grade serous ovarian cancer (STOSE), PAX2 expression improved animal survival by reducing proliferation and metastasis, which correlated with increased Htra1 and decreased COX2. Thus, PAX2 may not be a classical oncogene or tumor suppressor but instead can act in either role by differential regulation of COX2 and/or HTRA1.


Asunto(s)
Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Factor de Transcripción PAX2/metabolismo , Animales , Western Blotting , Carcinoma Epitelial de Ovario , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Inmunohistoquímica , Ratones , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , ARN Interferente Pequeño , Transcriptoma , Transfección
8.
Nat Commun ; 6: 6410, 2015 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-25817275

RESUMEN

In this study, we show that several microtubule-destabilizing agents used for decades for treatment of cancer and other diseases also sensitize cancer cells to oncolytic rhabdoviruses and improve therapeutic outcomes in resistant murine cancer models. Drug-induced microtubule destabilization leads to superior viral spread in cancer cells by disrupting type I IFN mRNA translation, leading to decreased IFN protein expression and secretion. Furthermore, microtubule-destabilizing agents specifically promote cancer cell death following stimulation by a subset of infection-induced cytokines, thereby increasing viral bystander effects. This study reveals a previously unappreciated role for microtubule structures in the regulation of the innate cellular antiviral response and demonstrates that unexpected combinations of approved chemotherapeutics and biological agents can lead to improved therapeutic outcomes.


Asunto(s)
Efecto Espectador/efectos de los fármacos , Citocinas/efectos de los fármacos , Interferón Tipo I/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Viroterapia Oncolítica , Virus Oncolíticos , ARN Mensajero/efectos de los fármacos , Infecciones por Rhabdoviridae/inmunología , Moduladores de Tubulina/farmacología , Albendazol/farmacología , Animales , Bencimidazoles/farmacología , Efecto Espectador/inmunología , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Colchicina/farmacología , Citocinas/inmunología , Células HT29 , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ratones , Nocodazol/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mensajero/metabolismo , Rhabdoviridae , Células Vero , Vinblastina/análogos & derivados , Vinblastina/farmacología , Vinorelbina
9.
Reproduction ; 149(2): R59-70, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25301968

RESUMEN

The lack of significant progress in the treatment of epithelial ovarian cancer (EOC) underscores the need to gain a better understanding of the processes that lead to chemoresistance and recurrence. The cancer stem cell (CSC) hypothesis offers an attractive explanation of how a subpopulation of cells within a patient's tumour might remain refractory to treatment and subsequently form the basis of recurrent chemoresistant disease. This review examines the literature defining somatic stem cells of the ovary and fallopian tube, two tissues that give rise to EOC. In addition, considerable research has been reviewed, that has identified subpopulations of EOC cells, based on marker expression (CD133, CD44, CD117, CD24, epithelial cell adhesion molecule, LY6A, ALDH1 and side population (SP)), which are enriched for tumour initiating cells (TICs). While many studies identified either CD133 or CD44 as markers useful for enriching for TICs, there is little consensus. This suggests that EOC cells may have a phenotypic plasticity that may preclude the identification of universal markers defining a CSC. The assay that forms the basis of quantifying TICs is the xenograft assay. Considerable controversy surrounds the xenograft assay and it is essential that some of the potential limitations be examined in this review. Highlighting such limitations or weaknesses is required to properly evaluate data and broaden our interpretation of potential mechanisms that might be contributing to the pathogenesis of ovarian cancer.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Células Madre , Antígeno AC133 , Familia de Aldehído Deshidrogenasa 1 , Animales , Antígenos CD/análisis , Biomarcadores de Tumor/análisis , Carcinoma Epitelial de Ovario , Trompas Uterinas/patología , Femenino , Glicoproteínas/análisis , Humanos , Receptores de Hialuranos/análisis , Isoenzimas/análisis , Recurrencia Local de Neoplasia , Neoplasias Glandulares y Epiteliales/química , Neoplasias Ováricas/química , Ovario/patología , Péptidos/análisis , Retinal-Deshidrogenasa/análisis , Células Madre/química , Células Madre/efectos de los fármacos , Células Madre/patología
10.
Oncol Lett ; 9(1): 55-62, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25435933

RESUMEN

The aim of the present study was to optimize the polyethylenimine (PEI)-mediated transfection method in order to simplify the efficient production of lentiviral vectors (LvVs), and to compare the CaPO4- and PEI-mediated transfection methods for producing LvVs. Different titration methods of LvV stocks, as well as different culture media, culture durations, cell densities and DNA quantities were compared to obtain an optimized procedure for the production of LvVs. Optimization of the production method for LvVs was achieved using PEI-mediated transient transfections. Serum-free Opti-MEM® was used to directly produce LvVs that could be harvested 48 h after transfection. Furthermore, a cell density of 15×106 cells/10-cm plate and a DNA concentration of 1X were selected for the optimum production of LvVs. The optimized LvV titration method was simple and direct; it involved LvVs carrying fluorescent reporters, which proved to be faster than the standard methods but equally as sensitive. In conclusion, a scalable process for production of LvVs by PEI-mediated transfection was established and optimized. The optimized PEI-mediated transfection method was easy to use, as well as providing greater reliability with a higher degree of reproducibility and consistency. Despite using less DNA, the PEI-mediated transfection method resulted in viral titers that were the same as those achieved using the CaPO4-mediated method.

11.
Front Oncol ; 4: 53, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24672774

RESUMEN

Improving screening and treatment options for patients with epithelial ovarian cancer has been a major challenge in cancer research. Development of novel diagnostic and therapeutic approaches, particularly for the most common subtype, high-grade serous ovarian cancer (HGSC), has been hampered by controversies over the origin of the disease and a lack of spontaneous HGSC models to resolve this controversy. Over long-term culture in our laboratory, an ovarian surface epithelial (OSE) cell line spontaneously transformed OSE (STOSE). The objective of this study was to determine if the STOSE cell line is a good model of HGSC. STOSE cells grow faster than early passage parental M0505 cells with a doubling time of 13 and 48 h, respectively. STOSE cells form colonies in soft agar, an activity for which M0505 cells have negligible capacity. Microarray analysis identified 1755 down-regulated genes and 1203 up-regulated genes in STOSE compared to M0505 cells, many associated with aberrant Wnt/ß-catenin and Nf-κB signaling. Upregulation of Ccnd1 and loss of Cdkn2a in STOSE tumors is consistent with changes identified in human ovarian cancers by The Cancer Genome Atlas. Intraperitoneal injection of STOSE cells into severe combined immunodeficient and syngeneic FVB/N mice produced cytokeratin+, WT1+, inhibin-, and PAX8+ tumors, a histotype resembling human HGSC. Based on evidence that a SCA1+ stem cell-like population exists in M0505 cells, we examined a subpopulation of SCA1+ cells that is present in STOSE cells. Compared to SCA1- cells, SCA1+ STOSE cells have increased colony-forming capacity and form palpable tumors 8 days faster after intrabursal injection into FVB/N mice. This study has identified the STOSE cells as the first spontaneous murine model of HGSC and provides evidence for the OSE as a possible origin of HGSC. Furthermore, this model provides a novel opportunity to study how normal stem-like OSE cells may transform into tumor-initiating cells.

12.
J Ovarian Res ; 5(1): 39, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-23190474

RESUMEN

The development of genetically engineered models (GEM) of epithelial ovarian cancer (EOC) has been very successful, with well validated models representing high grade and low grade serous adenocarcinomas and endometrioid carcinoma (EC). Most of these models were developed using technologies intended to target the ovarian surface epithelium (OSE), the cell type long believed to be the origin of EOC. More recent evidence has highlighted what is likely a more prevalent role of the secretory cell of the fallopian tube in the ontogeny of EOC, however none of the GEM of EOC have demonstrated successful targeting of this important cell type.The precise technologies exploited to develop the existing GEM of EOC are varied and carry with them advantages and disadvantages. The use of tissue specific promoters to model disease has been very successful, but the lack of any truly specific OSE or oviductal secretory cell promoters makes the outcomes of these models quite unpredictable. Effecting genetic change by the administration of adenoviral vectors expressing Cre recombinase may alleviate the perceived need for tissue specific promoters, however the efficiencies of infection of different cell types is subject to numerous biological parameters that may lead to preferential targeting of certain cell populations.One important future avenue of GEM of EOC is the evaluation of the role of genetic modifiers. We have found that genetic background can lead to contrasting phenotypes in one model of ovarian cancer, and data from other laboratories have also hinted that the exact genetic background of the model may influence the resulting phenotype. The different genetic backgrounds may modify the biology of the tumors in a manner that will be relevant to human disease, but they may also be modifying parameters which impact the response of the host to the technologies employed to develop the model.

13.
Mol Ther ; 20(9): 1791-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22760544

RESUMEN

Treatment of permissive tumors with the oncolytic virus (OV) VSV-Δ51 leads to a robust antitumor T-cell response, which contributes to efficacy; however, many tumors are not permissive to in vivo treatment with VSV-Δ51. In an attempt to channel the immune stimulatory properties of VSV-Δ51 and broaden the scope of tumors that can be treated by an OV, we have developed a potent oncolytic vaccine platform, consisting of tumor cells infected with VSV-Δ51. We demonstrate that prophylactic immunization with this infected cell vaccine (ICV) protected mice from subsequent tumor challenge, and expression of granulocyte-monocyte colony stimulating factor (GM-CSF) by the virus (VSVgm-ICV) increased efficacy. Immunization with VSVgm-ICV in the VSV-resistant B16-F10 model induced maturation of dendritic and natural killer (NK) cell populations. The challenge tumor is rapidly infiltrated by a large number of interferon γ (IFNγ)-producing T and NK cells. Finally, we demonstrate that this approach is robust enough to control the growth of established tumors. This strategy is broadly applicable because of VSV's extremely broad tropism, allowing nearly all cell types to be infected at high multiplicities of infection in vitro, where the virus replication kinetics outpace the cellular IFN response. It is also personalized to the unique tumor antigen(s) displayed by the cancer cell.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Melanoma Experimental/prevención & control , Melanoma Experimental/terapia , Neoplasias Cutáneas/prevención & control , Neoplasias Cutáneas/terapia , Vesiculovirus/inmunología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Terapia Genética/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Inmunización , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Neoplasias Cutáneas/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Vero , Vesiculovirus/genética , Replicación Viral
14.
PLoS One ; 6(9): e24643, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21931792

RESUMEN

BACKGROUND: Genetic manipulation of poxvirus genomes through attenuation, or insertion of therapeutic genes has led to a number of vector candidates for the treatment of a variety of human diseases. The development of recombinant poxviruses often involves the genomic insertion of a selectable marker for purification and selection purposes. The use of marker genes however inevitably results in a vector that contains unwanted genetic information of no therapeutic value. METHODOLOGY/PRINCIPAL FINDINGS: Here we describe an improved strategy that allows for the creation of marker-free recombinant poxviruses of any species. The Selectable and Excisable Marker (SEM) system incorporates a unique fusion marker gene for the efficient selection of poxvirus recombinants and the Cre/loxP system to facilitate the subsequent removal of the marker. We have defined and characterized this new methodological tool by insertion of a foreign gene into vaccinia virus, with the subsequent removal of the selectable marker. We then analyzed the importance of loxP orientation during Cre recombination, and show that the SEM system can be used to introduce site-specific deletions or inversions into the viral genome. Finally, we demonstrate that the SEM strategy is amenable to other poxviruses, as demonstrated here with the creation of an ectromelia virus recombinant lacking the EVM002 gene. CONCLUSION/SIGNIFICANCE: The system described here thus provides a faster, simpler and more efficient means to create clinic-ready recombinant poxviruses for therapeutic gene therapy applications.


Asunto(s)
Marcadores Genéticos/genética , Poxviridae/genética , Recombinación Genética/genética , Animales , Bovinos , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Vectores Genéticos/genética , Humanos
15.
Endocrinology ; 151(3): 929-38, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20056833

RESUMEN

Epithelial ovarian cancer is thought to be derived from the ovarian surface epithelium (OSE) but often goes undetected in the early stages, and as a result, the factors that contribute to its initiation and progression remain poorly understood. Epidemiological studies have suggested that the female steroid hormones are involved in ovarian carcinogenesis and that women who use hormone replacement therapy are at increased risk of developing the disease. A novel transgenic mouse model of ovarian cancer (tgCAG-LS-TAg) was developed to examine the role of the female reproductive steroid hormones [17beta-estradiol (E(2)) and progesterone (P(4))] on the initiation, progression, and pathology of ovarian cancer. The mouse model uses the Cre-LoxP system to induce expression of the simian virus 40 large and small T antigens (SV40 TAg). After targeted induction of the oncogene in the OSE, mice develop poorly differentiated ovarian tumors, tumor dissemination to tissues within the abdominal cavity, and a subset develops hemorrhagic ascites. Treatment with P(4) had no impact on the disease, but E(2) altered the pathophysiology, resulting in an earlier onset of tumors, decreased overall survival time, and a distinctive papillary histology. Normal ovaries collected from mice treated with E(2), but lacking expression of SV40 TAg, displayed an increase in the areas of columnar and hyperplastic OSE cells compared to placebo-treated controls. A better understanding of the mechanisms by which E(2) alters the morphology of normal OSE cells and reduces survival in this mouse model may translate into improved prevention and treatment options for women using hormone replacement therapy.


Asunto(s)
Estradiol/efectos adversos , Neoplasias Ováricas/inducido químicamente , Lesiones Precancerosas/inducido químicamente , Progesterona/efectos adversos , Animales , Antígenos Transformadores de Poliomavirus/genética , Femenino , Ratones , Ratones Transgénicos , Neoplasias Experimentales , Oncogenes , Neoplasias Ováricas/genética , Fenotipo
16.
Exp Cell Res ; 313(1): 133-45, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17070800

RESUMEN

Epithelial ovarian cancer (EOC) is thought to arise from the ovarian surface epithelium (OSE); however, the molecular events underlying this transformation are poorly understood. Germline mutations in the BRCA1 tumor suppressor gene result in a significantly increased risk of developing EOC and a large proportion of sporadic EOCs display some sort of BRCA1 dysfunction. Using mice with conditional expression of Brca1, we inactivated Brca1 in the murine OSE and demonstrate that this inactivation results in the development of preneoplastic changes, such as hyperplasia, epithelial invaginations, and inclusion cysts, which arise earlier and are more numerous than in control ovaries. These changes resemble the premalignant lesions that have been reported in human prophylactic oophorectomy specimens from women with BRCA1 germline mutation. We also report that inactivation of Brca1 in primary cultures of murine OSE cells leads to a suppression of proliferation due to increased apoptosis that can be rescued by concomitant inactivation of p53. These observations, along with our finding that these cells display an increased sensitivity to the DNA-damaging agent cisplatin, indicate that loss of function of Brca1 in OSE cells impacts both cellular growth control and DNA-damage repair which results in altered cell behavior manifested as morphological changes in vivo that arise earlier and are more numerous than what can be attributed to ageing.


Asunto(s)
Genes BRCA1 , Neoplasias Ováricas/genética , Ovario/metabolismo , Lesiones Precancerosas/genética , Animales , Apoptosis , Secuencia de Bases , Proliferación Celular , Células Cultivadas , Cisplatino/farmacología , Daño del ADN , Cartilla de ADN/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Marcación de Gen , Genes p53 , Humanos , Hiperplasia , Ratones , Ratones Noqueados , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Ovario/efectos de los fármacos , Ovario/patología , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología
17.
Zhonghua Fu Chan Ke Za Zhi ; 41(5): 333-8, 2006 May.
Artículo en Chino | MEDLINE | ID: mdl-16762191

RESUMEN

OBJECTIVE: To study the inhibition of OPCML on ovarian cancer cell lines using a lentiviral vector system for efficient gene transduction. METHODS: The murine OPCML cDNA was amplified by PCR from CD1 murine brain cDNA using gene specific primers, and subcloned into the lentiviral vector, pWPI-GFP, to generate the lentiviral expression vector, pWPI-OPCML. Recombinant lentiviruses were produced by 293T cells following the co-transfection of pWPI-OPCML, with the packaging plasmids pCMV-dR8.74 and pMDG. The resulting recombinant lentiviruses which carried OPCML or control viruses (only carrying GFP), were then used to infect the human ovarian cancer cell lines A2780 and OCC1 in addition to normal CD1 mouse ovarian surface epithelial cells. The infected cells were then characterized by cell proliferation assays, cell aggregation assays, cell cycle analysis by flow cytometry and tumorigenicity assays following injection into nude mice. RESULTS: (1) The efficiency of infection of the cell lines using the lentiviral vectors was almost 100% allowing the stable expression of OPCML in nearly all cells. Stable expression of OPCML (60 000) and GFP (27 000) proteins was confirmed by western blot analysis. (2) A2780 cells expressing OPCML [(7.6 +/- 1.0) x 10(5)] grew slowly compared to A2780 parental [(20.0 +/- 2.6) x 10(5)] or control virus infected cells [(18.1 +/- 1.7) x 10(5), P < 0.01], but the expression of OPCML had no effect on the proliferation rates of OCC1 and the normal CD1 cells when compared to their respective parental or controls (P > 0.05). (3) Flow cytometry based cell cycle assays showed that the expression of OPCML could arrest A2780 cells (G(0) approximately G(1) 67% vs 75%, P < 0.05); but not OCC1, CD1 cells. (4) The rate of aggregation of single cell suspensions was measured and found to be increased in all cell lines expressing OPCML indicating the increased cell surface adhesion mediated by OPCML. (5) A2780 cells expressing OPCML only formed a single tumor in 1/4 mice (10 mg) which was significantly smaller than controls [4/4; A2780 (280 +/- 53) mg and A2780/pWPI (677 +/- 323) mg; P < 0.01]. Expression of OPCML in tumor was confirmed by immunohistochemistry. CONCLUSIONS: The use of lentiviral vectors allowed the efficient expression of OPCML in nearly 100% of target cells. Expression of the OPCML cDNA resulted in an increase of cell adhesion in all cell lines tested, and decreased the proliferation and tumorigenicity of the A2780 ovarian cancer cell line. This indicates that the OPCML may be a new tumor suppressor gene.


Asunto(s)
Moléculas de Adhesión Celular/genética , Terapia Genética/métodos , Lentivirus/genética , Neoplasias Ováricas/terapia , Animales , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Ciclo Celular , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Citometría de Flujo , Proteínas Ligadas a GPI , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Microscopía Fluorescente , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
18.
Zhonghua Zhong Liu Za Zhi ; 28(12): 881-5, 2006 Dec.
Artículo en Chino | MEDLINE | ID: mdl-17533735

RESUMEN

OBJECTIVE: To study the function of c-myc and K-ras in tumorigenesis of ovarian cancer. METHODS: K-ras and/or c-myc cDNAs were introduced into mouse ovarian surface epithelium cells (MOSE) using recombinant Moloney retroviral vectors. The resulting MOSE cells were studied by cell proliferation assays, the ability to form colonies in soft agarose, matrigel invasion assays and tumorigenicity assays in nude mice. RESULTS: K-ras and c-myc can be easily delivered to the normal MOSE cells by recombinant retroviruses. mRNA and protein of the target genes can be detected by RT-PCR and Western blot. Cell proliferation assays showed that MOSE-Ras cells and MOSE-RM cells (MOSE-Ras/Myc) grew more rapidly than parental cells (MOSE) and MOSE-Myc cells (P <0.01). In addtition, MOSE-RM cells grew more rapidly than MOSE-Ras cells (P <0. 05). Cell colony formation assays showed that while MOSE-Ras and MOSE-RM cells can form colonies in soft-agarose, the MOSE-Myc and MOSE cells did not. Matrigel invasion assays showed that MOSE-Ras and MOSE-RM cells have invasion ability, but not MOSE-Myc ascites and the control MOSE cells. Xenograft experiments showed that MOSE-Ras and MOSE-RM cells were able to form tumors in nude mice following intraperitoneal injection. Tumors were not observed in animals injected with either MOSE-Myc or MOSE cells. CONCLUSION: The recombinant Moloney retroviral system is a highly efficient and convenient method for introducing and expressing foreign genes in murine surface epithelial cell cultures. In this model, expression of K-ras alone is sufficient to generate tumorigenic MOSE, however expression of c-myc in conjunction with K-ras results in cells with a higher index of malignancy. Based on the assays described in this report, expression of c-myc alone can not transform MOSE cultures although it does play a role in cooperation with K-ras.


Asunto(s)
Transformación Celular Neoplásica , Células Epiteliales/metabolismo , Proteína Oncogénica p21(ras)/biosíntesis , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Animales , Western Blotting , Movimiento Celular , Proliferación Celular , Células Cultivadas , Células Epiteliales/citología , Femenino , Inmunohistoquímica , Ratones , Ratones Desnudos , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteína Oncogénica p21(ras)/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Ovario/citología , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes/biosíntesis , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
19.
Mol Cell Endocrinol ; 239(1-2): 15-26, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15955618

RESUMEN

Ovarian cancer is the most lethal of all gynecological cancers and arises most commonly from the surface epithelium. Successful clinical management of patients with epithelial ovarian cancer is limited by the lack of a reliable and specific method for early detection, and the frequent recurrence of chemoresistant disease. Experimental models are of crucial importance not only to understand the biological and genetic factors that influence the phenotypic characteristics of the disease but also to utilize as a basis for developing rational intervention strategies. Ovarian cancer cell lines derived from ascites or primary ovarian tumors have been used extensively and can be very effective for studying the processes controlling growth regulation and chemosensitivity or evaluating novel therapeutics, both in vitro and in xenograft models. While our limited knowledge of the initiating events of ovarian cancer has restricted the development of models in which the early pathogenic events can be studied, recent advances in the ability to manipulate gene expression in ovarian surface epithelial cells in vitro and in vivo have begun to provide insights into the molecular changes that may contribute to the development of ovarian cancer. This review highlights the strengths and weaknesses of some of the current models of ovarian cancer, with special consideration of the recent progress in modeling ovarian cancer using genetically engineered mice.


Asunto(s)
Modelos Animales de Enfermedad , Neoplasias Ováricas , Ovario , Animales , Epitelio/patología , Femenino , Humanos , Ratones , Neoplasias Experimentales/patología , Neoplasias Ováricas/patología , Ovario/patología , Lesiones Precancerosas
20.
J Soc Gynecol Investig ; 10(4): 244-50, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12759154

RESUMEN

OBJECTIVE: The ovarian-specific promoter, OSP-1, which was cloned from the transcript of a rat retrovirus-like element specifically expressed in ovarian tissue, was tested for its ability to drive ovary-specific transcription in transgenic mice. METHODS: Transgenic mice were generated with the lacZ reporter gene (OSP-lacZ) or the early region of SV40 virus (OSP-TAg) placed under the control of the OSP-1 promoter. OSP-lacZ and OSP-TAg transgenic animals were examined, respectively, for the expression of lacZ (OSP-lacZ) or the development of tumors (OSP-TAg). RESULTS: The expression of lacZ in the resulting OSP-lacZ mice was restricted to the ovary as determined by X-gal staining of multiple organs. Immunohistochemical detection of beta-galactosidase showed lacZ expression mainly in the granulosa cells and ovarian surface epithelial cells. OSP-TAg mice developed tumors in a variety of tissues, including unilateral granulosa cell tumors in two of three female founder mice. In the contralateral ovary of one mouse with a granulosa cell tumor, there were alterations in the ovarian surface epithelial cells suggestive of preneoplasia. CONCLUSIONS: Although the OSP-1 promoter was able to restrict reporter gene expression to the ovary in transgenic mice, the expression of TAg in the OSP-TAg mice resulted in ovarian tumors as well as tumors in numerous other organs. This indicated that although transcription from the OSP-1 promoter occurs predominantly in the ovary, this promoter is sufficiently leaky in cells in other tissues to permit their tumorigenic conversion by SV40 TAg.


Asunto(s)
Neoplasias Ováricas/genética , Regiones Promotoras Genéticas/genética , Animales , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/fisiología , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Inmunohistoquímica , Operón Lac/fisiología , Masculino , Ratones , Ratones Transgénicos , Hibridación de Ácido Nucleico , ARN Mensajero/química , ARN Mensajero/genética , Virus 40 de los Simios/genética , Virus 40 de los Simios/fisiología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA