Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Oncotarget ; 7(30): 47343-47365, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27329838

ABSTRACT

Epithelial ovarian cancer (EOC) is the most lethal gynecological cancer and often is not detected until late stages when cancer cells transcoelomically metastasize to the abdomen and typically become resistant to therapy resulting in very low survival rates. We utilize an orthotopic, syngeneic mouse model to study late stage disease and have discovered that the tumor cells within the abdominal ascites are irreversibly re-programmed, with an increased tumorigenicity and resistance to apoptosis. The goal of this study was to characterize the reprogramming that occurred in the aggressive ascites-derived cells (28-2 cells) compared to the original cell line used for tumor induction (ID8 cells). Microarray experiments showed that the majority of genes upregulated in the 28-2 cells belonged to the mevalonate pathway, which is involved in cholesterol biosynthesis, protein prenylation, and activation of small GTPases. Upregulation of mevalonate appeared to be associated with the acquisition of a p53 mutation in the ascites-derived cells. Treatment with simvastatin to inhibit HMG CoA reductase, the rate limiting enzyme of this pathway, induced apoptosis in the 28-2 cell line. Rescue experiments revealed that mevalonate, but not cholesterol, could inhibit the simvastatin-mediated effects. In vivo, daily intraperitoneal simvastatin treatment significantly regressed advanced stage disease and induced death of metastatic tumor cells. These data suggest that ovarian cancer cells become reprogrammed, with genetic mutations, and upregulation of the mevalonate pathway, which facilitates the development of advanced stage disease. The use of statins to inhibit HMGCR may provide novel therapeutic opportunities for the treatment of advanced stage EOC.


Subject(s)
Mevalonic Acid/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Animals , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Genomic Instability , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Polyisoprenyl Phosphates/pharmacology , Simvastatin/pharmacology , Simvastatin/therapeutic use , Tumor Microenvironment
2.
Methods Mol Biol ; 1049: 409-23, 2013.
Article in English | MEDLINE | ID: mdl-23913234

ABSTRACT

One of the difficulties in studying ovarian cancer historically has been the lack of a suitable animal model that replicates the human disease. Mouse models that utilize intraperitoneal implantation of tumorigenic cells lack interaction between the transformed ovarian epithelial cells and the ovarian stroma, which we have shown to be an integral component in replicating the etiology seen in human epithelial ovarian cancer (Greenaway, Gynecol Oncol 108:385-394, 2008). Xenograft models generally require the use of immunocompromised hosts, which then eliminates the influence of the immune system in disease progression, which also has been shown to be an important part of the progression of epithelial ovarian cancer (EOC). In this chapter, we describe the generation and optimization of an orthotopic, syngeneic mouse model and illustrate the importance of facilitating epithelial-stromal cell interaction to more closely replicate human EOC.


Subject(s)
Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Stromal Cells/pathology , Animals , Carcinoma, Ovarian Epithelial , Cell Transformation, Neoplastic/genetics , Female , Humans , Mice , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Ovary/pathology , Transplantation, Heterologous
3.
Biochem Cell Biol ; 90(1): 96-107, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22003835

ABSTRACT

A lack of host-derived SPARC promotes disease progression in an intraperitoneal (IP) ID8 mouse model of epithelial ovarian cancer (EOC). Since orthotopic injection (OT) of ID8 cells better recapitulates high-grade serous cancer, we examined the impact of host-derived SPARC following OT injection. Sparc(-/-) and wild-type (WT) mice were injected with ID8 cells either OT or IP and tumors were analyzed at the moribund stage. Sparc(-/-) mice had reduced survival and fewer well-defined abdominal lesions compared with WT controls after IP injection, whereas no differences were observed in survival or abdominal lesions between Sparc(-/-) and WT mice after OT injection. No differences in mass or collagen content were observed in ovarian tumors between OT-injected Sparc(-/-) and WT mice. The abdominal wall of the IP-injected Sparc(-/-) mice exhibited immature and less abundant collagen fibrils compared with WT mice both in injected and non-injected controls. In contrast to human EOC, SPARC was expressed by the tumor cells but was absent in reactive stroma of WT mice. Exposure to the ovarian microenvironment through OT injections alters the metastatic behaviour of ID8 cells, which is not affected by the absence of host-derived SPARC.


Subject(s)
Osteonectin/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Tumor Microenvironment , Animals , Disease Models, Animal , Disease Progression , Female , Humans , Mice , Mice, Inbred C57BL , Osteonectin/deficiency
4.
Mol Cancer Ther ; 10(10): 1876-85, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21844212

ABSTRACT

Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and is often not diagnosed until late stages due to its asymptomatic nature. Women diagnosed with EOC typically undergo surgical debulking followed by chemotherapy; however, disease recurrence often occurs. In this study, we evaluated the ability of the thrombospondin-1 mimetic peptide, ABT-898, to regress established, late-stage tumors in a mouse model of human EOC. Ovarian tumors were induced and ABT-898 treatment was initiated at time points that were representative of late stages of the disease to study tumor regression. ABT-898 induced tumor regression and reduced the morbidity of treated animals compared with controls. Analysis of tumors from ABT-898-treated animals showed reduced abnormal tumor vasculature, decreased expression of the proangiogenic compound VEGF, and reduced tumor tissue hypoxia. ABT-898 treatment initiated at late-stage disease also significantly prolonged disease-free survival compared with control animals. Results from this study show that ABT-898 is capable of regressing established ovarian tumors in an animal model of the disease. As most women are detected at advanced stage EOC, ABT-898 may improve our treatment of ovarian cancer.


Subject(s)
Apoptosis/drug effects , Neoplasms, Glandular and Epithelial/drug therapy , Oligopeptides/pharmacology , Ovarian Neoplasms/drug therapy , Animals , Carcinoma, Ovarian Epithelial , Disease Models, Animal , Disease-Free Survival , Endothelial Cells/drug effects , Epithelial Cells/drug effects , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neoplasms, Glandular and Epithelial/blood supply , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/pathology , Survival Rate
5.
J Pathol ; 225(1): 106-17, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21744340

ABSTRACT

The discovery of occult invasive and intra-epithelial tubal carcinomas in BRCA1 mutation carriers undergoing prophylactic surgery has implicated the fallopian tube epithelium as the source of serous cancer. However, little is known of the early molecular events of serous oncogenesis, or why cancers in BRCA1 mutation carriers are found preferentially in tissues which are responsive to reproductive hormones. We hypothesize that molecular alterations present in morphologically normal tubal epithelium from BRCA1 heterozygotes reflect the earliest events in serous carcinogenesis and may be markers of increased cancer risk as well as targets for risk reduction. Genetic profiling of microdissected tubal epithelium from histologically normal BRCA1 mutation carriers and controls was performed. We sought to define a signature which differentiated BRCA1 mutant tubal epithelium from women with low risk of developing ovarian cancer. Molecular differences between the follicular and luteal phases were prominent and, by using filtering techniques and a two-way ANOVA without a False Discovery Rate correction, we identified 440 probe sets with a more than two-fold change in gene expression related to BRCA1 mutation status. Using gene ontology and known associations to cancer pathways, we selected five genes for further analysis by qPCR and immunohistochemistry, and were able to demonstrate statistically significant differentiation of BRCA1 and control cases in an independent set of cases. The altered expression profiles in histologically normal tubal epithelium from BRCA1 heterozygotes suggest that these cells may respond differently to microenvironmental stresses.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Fallopian Tube Neoplasms/genetics , Genes, BRCA1 , Mutation , Precancerous Conditions/genetics , Adult , Case-Control Studies , Cluster Analysis , Cystadenocarcinoma, Serous/metabolism , Epithelium/metabolism , Fallopian Tube Neoplasms/metabolism , Fallopian Tubes/metabolism , Female , Follicular Phase/physiology , Gene Expression Profiling/methods , Gene Expression Regulation , Heterozygote , Humans , Luteal Phase/physiology , Microdissection/methods , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Precancerous Conditions/metabolism , Signal Transduction/genetics
6.
Am J Pathol ; 177(3): 1320-32, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20651246

ABSTRACT

Although osteopontin (OPN) is up-regulated in inflammatory bowel diseases, its role in disease pathogenesis remains controversial. The objective of this study was to determine the role of OPN in host responses to a non-invasive bacterial pathogen, Citrobacter rodentium, which serves as a murine infectious model of colitis. OPN gene knockout and wild-type mice were infected orogastrically with either C. rodentium or Luria-Bertani (LB) broth. Mouse-derived OPN(+/+) and OPN(-/-) fibroblasts were incubated with C. rodentium and attaching-effacing lesions were demonstrated using transmission electron microscopy and immunofluorescence. Colonic expression of OPN was increased by C. rodentium infection of wild-type mice. Furthermore, colonic epithelial cell hyperplasia, the hallmark of C. rodentium infection, was reduced in OPN(-/-) mice, and spleen enlargement by infection was absent in OPN(-/-) mice. Rectal administration of OPN to OPN(-/-) mice restored these effects. There was an 8- to 17-fold reduction in bacterial colonization in OPN(-/-) mice, compared with wild-type mice, which was accompanied by reduced attaching-effacing lesions, both in infected OPN(-/-) mice and OPN(-/-) mouse fibroblasts. Moreover, adhesion pedestals were restored in OPN(-/-) cells complemented with human OPN. Therefore, lack of OPN results in decreased pedestal formation, colonization, and colonic epithelial cell hyperplasia responses to C. rodentium infection, indicating that OPN impacts disease pathogenesis through bacterial attachment and altered host immune responses.


Subject(s)
Citrobacter rodentium/metabolism , Colon/microbiology , Enterobacteriaceae Infections/metabolism , Epithelial Cells/microbiology , Osteopontin/metabolism , Animals , Colitis/metabolism , Colitis/microbiology , Colitis/pathology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Hyperplasia/metabolism , Hyperplasia/microbiology , Hyperplasia/pathology , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Knockout
7.
Neoplasia ; 12(3): 275-83, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20234821

ABSTRACT

Epithelial ovarian cancer (EOC) comprises approximately 90% of ovarian cancers and arises from the surface epithelium. Typical treatment of EOC involves cytoreductive surgery combined with chemotherapy. More recent therapies have targeted the tumor vasculature using antiangiogenic compounds such as thrombospondin-1 (TSP-1). TSP-1 mimetic peptides such as ABT-510 have been created and have been in various clinical trials. We have previously shown that ABT-510 reduces abnormal vasculature associated with tumor tissue and increases the presence of mature blood vessels. It has been hypothesized that treatment with antiangiogenic compounds would allow increased delivery of cytotoxic agents and enhance treatment. In this study, we evaluated the potential role of ABT-510 and various chemotherapeutics (cisplatin and paclitaxel) on tumor progression, angiogenesis, and the benefits of combinational treatments on tissue uptake and perfusion using an orthotopic syngeneic mouse model of EOC. Animals were treated with ABT-510 (100 mg/kg per day) alone or in combination with cisplatin (2 mg/kg per 3 days) or paclitaxel (10 mg/kg per 2 days) at 60 days after tumor induction. Radiolabeled and fluorescently labeled paclitaxel demonstrated a significant increase in tumor uptake after ABT-510 treatment. Combined treatment with ABT-510 and cisplatin or paclitaxel resulted in a significant increase in tumor cell and tumor endothelial cell apoptosis and a resultant decrease in ovarian tumor size. Combined treatment also regressed secondary lesions and eliminated the presence of abdominal ascites. The results from this study show that through vessel normalization, ABT-510 increases uptake of chemotherapy drugs and can induce regression of advanced ovarian cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Neoplasms, Glandular and Epithelial/metabolism , Oligopeptides/pharmacology , Ovarian Neoplasms/metabolism , Thrombospondin 1/metabolism , Animals , Biomimetics , Cell Line , Cisplatin/pharmacokinetics , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Immunoblotting , Mice , Mice, Inbred C57BL , Morbidity , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Paclitaxel/pharmacokinetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Survival Rate , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 8(1): 64-74, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19139114

ABSTRACT

Epithelial ovarian cancer (EOC) is the fifth most common cancer in women and is characterized by a low 5-year survival rate. One strategy that can potentially improve the overall survival rate in ovarian cancer is the use of antitumor agents such as ABT-510. ABT-510 is a small mimetic peptide of the naturally occurring antiangiogenic compound thrombospondin-1 and has been shown to significantly reduce tumor growth and burden in preclinical mouse models and in naturally occurring tumors in dogs. This is the first evaluation of ABT-510 in a preclinical model of human EOC. Tumorigenic mouse surface epithelial cells were injected into the bursa of C57BL/6 mice that were treated with either 100 mg/kg ABT-510 or an equivalent amount of PBS. ABT-510 caused a significant reduction in tumor size, ascites fluid volume, and secondary lesion dissemination when compared with PBS controls. Analysis of the vasculature of ABT-510-treated mice revealed vascular remodeling with smaller diameter vessels and lower overall area, increased number of mature vessels, and decreased tissue hypoxia. Tumors of ABT-510-treated mice had a significantly higher proportion of apoptotic tumor cells compared with the PBS-treated controls. Immunoblot analysis of cell lysates revealed a reduction in vascular endothelial growth factor, vascular endothelial growth factor receptor-2, and proliferating cell nuclear antigen protein expression as well as expression of members of the phosphatidylinositol 3-kinase and mitogen-activated protein kinase survival pathways. In vitro, ABT-510 induced tumor cell apoptosis in mouse and human ovarian cancer cells. This study shows ABT-510 as a promising candidate for inhibiting tumor growth and ascites formation in human EOC.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers/metabolism , CD36 Antigens/metabolism , Disease Models, Animal , Disease Progression , Epithelial Cells/metabolism , Fas Ligand Protein/metabolism , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/metabolism , Thrombospondin 1/deficiency , Thrombospondin 1/genetics , Thrombospondin 1/metabolism
9.
Gynecol Oncol ; 108(2): 385-94, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18036641

ABSTRACT

OBJECTIVE: Ovarian cancer is the fourth leading cause of cancer-related deaths among women and is among the least understood of all cancers. The objective of this study was to determine the effect of ovarian epithelial and stromal cell interaction in a mouse model of epithelial ovarian cancer (EOC) that closely resembled the human disease. METHODS: A mouse model of EOC was generated by orthotopic injection of an ID8 mouse ovarian surface epithelial cell line (MOSEC) under the ovarian bursa of syngeneic mice and tissue was collected to evaluate factors contributing to the formation and development of ovarian tumors. RESULTS: By 90 days post-injection, mice were moribund and had developed large primary ovarian tumors, secondary tumors within the peritoneal cavity, and extensive ascites fluid production. Tumors were hypervascularized and were characterized as serous epithelial carcinomatosis, which replicates the most common form of human ovarian cancer. Cells isolated from ascites fluid were more proliferative with increased expression of survival factors compared to original MOSEC cells and cells obtained from the abdomen following intraperitoneal injection. Orthotopic injection of these cells under the ovarian bursa resulted in more aggressive tumorigenesis, with mice becoming moribund at 60 days post-injection compared with 90 days post-injection with the original ID8-MOSEC cell line. DISCUSSION: This study describes the generation of an orthotopic, syngeneic model of ovarian cancer, which replicates the phenotype of the human disease. Expression of angiogenic and proliferative factors, and the interaction of epithelial cells with the ovarian stroma are important factors in ovarian tumorigenesis.


Subject(s)
Cell Communication/physiology , Ovarian Neoplasms/pathology , Animals , Biomarkers, Tumor/biosynthesis , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Disease Models, Animal , Epithelial Cells/pathology , Female , Humans , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/metabolism , Stromal Cells/pathology , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/metabolism
10.
J Cell Physiol ; 210(3): 807-18, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17154366

ABSTRACT

VEGF is a potent pro-angiogenic factor whose effects are opposed by a host of anti-angiogenic proteins, including thrombospondin-1 (TSP-1). We have previously shown that VEGF has important extravascular roles in the ovary and that VEGF and TSP-1 are inversely expressed throughout the ovarian cycle. To date, however, a causal interaction between TSP-1 and VEGF has not been identified. Here, we show that TSP-1 has a direct inhibitory effect on VEGF by binding the growth factor and internalizing it via LRP-1. Mice lacking TSP-1 are subfertile and exhibited ovarian hypervascularization and altered ovarian morphology. Treatment of ovarian cells with TSP-1 decreased VEGF levels and rendered the cells more susceptible to TNFalpha-induced apoptosis. Knockdown of TSP-1, through RNA interference, resulted in overexpression of VEGF and reduced cytokine-induced apoptosis. In conclusion, we demonstrate a direct inhibitory effect of TSP-1 on VEGF in the ovary. TSP-1's regulation of VEGF appears to be an important mediator of ovarian angiogenesis and follicle development.


Subject(s)
Ovary/metabolism , Receptors, LDL/metabolism , Thrombospondin 1/physiology , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis/drug effects , CHO Cells , Cell Line , Cell Proliferation , Cricetinae , Cricetulus , Female , Gene Expression Regulation , Granulosa Cells/cytology , Granulosa Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Mice, Knockout , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Ovary/blood supply , Ovary/cytology , Protein Binding , RNA Interference , Rats , Receptors, LDL/genetics , Thrombospondin 1/genetics , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor A/genetics
11.
Article in English | MEDLINE | ID: mdl-16515871

ABSTRACT

Real time RT-PCR was used to measure the changes in the rates of synthesis of mRNA encoding for growth hormone-1 (GH1) and -2 (GH2) and insulin-like growth factor-1 (IGF-1) and -2 (IGF-2), and whole embryo GH content was measured in early stage rainbow trout (Oncorhynchus mykiss) embryos reared at two incubation temperatures (8.5 and 6.0 degrees C). Particular attention was paid to the phase of embryo development that preceded the appearance of the pituitary gland. GH was present in zygotes, and there were no significant changes in whole embryo GH content of the two temperature treatment groups from fertilization (t0) until the time at which GH was detectable in the pituitary gland by immunostaining. The expression of the two GH genes decreased during the first 24 h post-fertilization, and then increased significantly by 17 dpf in embryos reared at both temperatures. There was a subsequent steep increase in the number of copies of GH1 and GH2 mRNA associated with the formation of the pituitary gland evident at 23 and 34 dpf in the 8.5 and 6.0 degrees C groups, respectively. The number of copies of mRNA encoding for IGF-1 and IGF-2 did not change during the first 24 h post-fertilization; however, there was a significant increase in the numbers of transcripts for both genes evident by 13 dpf in embryos reared at the two incubation temperatures. The differences in the timing of the increases in GH and IGF mRNA may suggest that IGF gene expression is not GH-dependent at that stage. Moreover, the increased expression of the GH genes prior to the formation of the pituitary gland suggests that tissues other than the pituitary are expressing these genes in early embryos. The pattern of changes in GH content was similar to the pattern of GH gene expression in embryos reared at the two incubation temperatures when the age of embryos was plotted using degree-days. There were no apparent compensatory responses in GH1, GH2, IGF-1 or IGF-2 gene expression related to altered growth rates. The number of copies of IGF-2 mRNA was higher than that of IGF-1 mRNA during the early developmental period; this is consistent with the hypothesis that IGF-2 predominates during embryonic development. A differential expression of GH2 and GH1 was also observed with the overall copy numbers of GH2 mRNA being consistently higher than those of GH1.


Subject(s)
Growth Hormone/genetics , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor I/genetics , Oncorhynchus mykiss/embryology , Pituitary Gland/embryology , Animals , Embryo, Nonmammalian , Fish Proteins/genetics , Gene Expression Regulation, Developmental , Growth Hormone/metabolism , Oncorhynchus mykiss/genetics , Protein Isoforms/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Temperature
12.
Biol Reprod ; 72(5): 1071-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15616224

ABSTRACT

Angiogenesis does not normally occur in most adult tissues. However, in the ovary, there are cyclical vascular changes including angiogenesis that involve the interaction of numerous cytokines and growth factors. Angiogenic processes are regulated by a balance between pro- and antiangiogenic factors. The purpose of this study was to determine the expression of the antiangiogenic thrombospondin family and proangiogenic vascular endothelial growth factor (VEGF) in various sizes of healthy bovine follicles. Ovaries were collected from slaughterhouse animals and healthy follicles were sorted based on size (< 0.5 cm, small; 0.5-1.0 cm, medium; >1.0 cm, large). Thrombospondin (TSP) protein levels were significantly higher in small follicles. Immunohistochemistry confirmed the granulosa layer as the primary area within the follicle involved in TSP generation and that small follicles had the highest proportion of immunopositive cells. TSP-1 and -2 mRNA levels were significantly higher in small follicles than either medium or large follicles. TSP colocalized with CD36 on granulosa cells (GC) in the follicle and in cultured cells. In contrast with TSP, VEGF expression increased during growth and development of the follicle. FSH stimulated GC expression of TSP, while LH had no effect. In summary, TSP-1 and -2 were coordinately expressed in the extravascular compartment of the ovary during early follicle development. VEGF was inversely expressed, with expression increasing as follicles developed. Regulated expression and localization of these proteins suggests that they may be involved in regulating growth and development of the follicle in a novel fashion.


Subject(s)
Ovarian Follicle/metabolism , Thrombospondins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , CD36 Antigens/metabolism , Cattle , Female , Follicular Fluid/metabolism , Gene Expression Regulation , Granulosa Cells/metabolism , In Vitro Techniques , Neovascularization, Physiologic , Ovarian Follicle/physiology , Ovary/blood supply , Ovary/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Thrombospondins/genetics , Vascular Endothelial Growth Factor A/genetics
13.
Endocrinology ; 145(6): 2896-905, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14988387

ABSTRACT

Vascular endothelial growth factor (VEGF) is a potent mitogen and cytoprotective factor for vascular endothelial cells. Although VEGF is ubiquitously expressed, its role in nonvascular tissues is poorly understood. VEGF interacts with various cell surface receptors to mediate its cellular effects. It previously has been thought that the VEGF receptor Flk-1/KDR, its main signaling receptor, was expressed exclusively by endothelial cells. However, in the present study using bovine and rodent models, we demonstrate that VEGF and Flk-1/KDR are coexpressed in ovarian granulosa cells. VEGF and Flk-1/KDR mRNA and protein were both detectable in follicle tissue sections and in vitro cultured granulosa cells. Expression of both ligand and receptor increased in healthy follicles throughout follicular development. VEGF treatment of serum-starved and cytokine-exposed granulosa cells resulted in enhanced survival, and this cytoprotection was ameliorated when Flk-1/KDR signaling was inhibited. Reduced expression of Flk-1/KDR was also associated with the onset and progression of follicle atresia, suggesting involvement in follicular health in vivo. The results of this study demonstrate for the first time expression of Flk-1/KDR in ovarian granulosa cells and identify a novel extravascular role for VEGF and its receptor in ovarian function.


Subject(s)
Cytoprotection , Ovarian Follicle/physiology , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology , Animals , Caspase 3 , Caspase Inhibitors , Cattle , Cells, Cultured , Female , Granulosa Cells/metabolism , Humans , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL