Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
BMC Dev Biol ; 13: 39, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24168030

ABSTRACT

BACKGROUND: Disabled-2 (Dab2) is an endocytic adaptor protein involved in clathrin-mediated endocytosis and cargo trafficking. Since its expression is lost in several cancer types, Dab2 has been suggested to be a tumor suppressor. In vitro studies indicate that Dab2 establishes epithelial cell polarity and organization by directing endocytic trafficking of membrane glycoproteins. Dab2 also modulates cellular signaling pathways by mediating the endocytosis and recycling of surface receptors and associated signaling components. Previously, two independent gene knockout studies have been reported, with some discrepancies in the observed embryonic phenotypes. To further clarify the in vivo roles of Dab2 in development and physiology, we designed a new floxed allele to delete dab2 gene. RESULTS: The constitutive dab2 deleted embryos showed a spectrum in the degree of endoderm disorganization in E5.5 and no mutant embryos persisted at E9.5. However, the mice were grossly normal when dab2 deletion was restricted to the embryo proper and the gene was retained in extraembryonic tissues using Meox2-Cre and Sox2-Cre. Adult Dab2-deficient mice had a small but statistically significant increase in serum cholesterol levels. CONCLUSION: The study of the new dab2 mutant allele in embryos and embryoid bodies confirms a role for Dab2 in extraembryonic endoderm development and epithelial organization. Experimental results with embryoid bodies suggest that additional endocytic adaptors such as Arh and Numb could partially compensate for Dab2 loss. Conditional deletion indicates that Dab2 is dispensable for organ development, when the vast majority of the embryonic cells are dab2 null. However, Dab2 has a physiological role in the endocytosis of lipoproteins and cholesterol metabolism.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Embryonic Development , Endoderm/embryology , Endoderm/metabolism , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Cells, Cultured , Cholesterol/metabolism , Embryo, Mammalian , Embryoid Bodies/metabolism , Embryonic Stem Cells/metabolism , Endocytosis , Female , Gene Deletion , Genes, Lethal , Lipoproteins/metabolism , Male , Mice , Mice, Knockout , Phenotype , Recombination, Genetic , Signal Transduction
2.
Oncogene ; 42(6): 409-420, 2023 02.
Article in English | MEDLINE | ID: mdl-36482202

ABSTRACT

SHP1, a tyrosine phosphatase, negatively regulates B-cell receptor (BCR) signaling. Ibrutinib selectively inhibits BTK and has been approved for the treatment of several types of B-cell lymphomas, but not yet in diffuse large B-cell lymphoma (DLBCL). A phase 3 clinical trial of ibrutinib-containing regimen has been completed to evaluate its activity in subtypes or subsets of DLBCL patients. Although the subtype of activated B-cell like (ABC) DLBCL is characterized by chronic active BCR signaling, only a fraction of ABC-DLBCL patients seem to benefit from ibrutinib-containing regimen. New alternative predictive biomarkers are needed to identify patients who better respond. We investigated if SHP1 plays a role in defining the level of the BCR activity and impacts the response to ibrutinib. A meta-analysis revealed that lack of SHP1 protein expression as well as SHP1 promoter hypermethylation is strongly associated with NHL including DLBCL. On a tissue microarray of 95 DLBCL samples, no substantial difference in SHP1 expression was found between the GCB and non-GCB subtypes of DLBCL. However, we identified a strong reverse correlation between SHP1 expression and promoter methylation suggesting that promoter hypermethylation is responsible for SHP1 loss. SHP1 knockout in BCR-dependent GCB and ABC cell lines increased BCR signaling activities and sensitize lymphoma cells to the action of ibrutinib. Rescue of SHP1 in the knockout clones, on the other hand, restored BCR signaling and ibrutinib resistance. Further, pharmacological inhibition of SHP1 in both cell lines and patient-derived primary cells demonstrate that SHP1 inhibition synergized with ibrutinib in suppressing tumor cell growth. Thus, SHP1 loss may serve as an alternative biomarker to cell-of-origin to identify patients who potentially benefit from ibrutinib treatment. Our results further suggest that reducing SHP1 pharmacologically may represent a new strategy to augment tumor response to BCR-directed therapies. Schematic diagram summarizing the major findings. Left panel. When SHP1 is present and functional, it negatively regulates the activity of the BCR pathway. Right pane. When SHP1 is diminished or lost, cells depend more on the increased BCR signaling and making them vulnerable to BTK inhibitor, ibrutinib. Diagram was generated using BioRender.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Signal Transduction , Humans , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/therapeutic use , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Cell Line, Tumor , Biomarkers
3.
Dev Dyn ; 240(3): 566-76, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20925113

ABSTRACT

Gene knockouts in mice have showed that Grb2 and GATA6 are essential for the formation of primitive endoderm in blastocysts. Here, we confirmed that implanted Grb2-null blastocysts lack primitive or extraembryonic endoderm cells either at E4.5 or E5.5 stages. We analyzed the relationship between Grb2 and GATA6 in the differentiation of embryonic stem (ES) cells to primitive endoderm in embryoid body models. Upon transfection with GATA6 expression vector, Grb2-null ES cells underwent endoderm differentiation as indicated by the expression of the extraembryonic endoderm markers Dab2 and GATA4. Transfection of GATA4 expression vector also had the same differentiation potency. When GATA6- or GATA4-transfected Grb2-null ES cells were allowed to aggregate, fragments of an endoderm layer formed on the surface of the spheroids. The results suggest that GATA6 is downstream of Grb2 in the inductive signaling pathway and the expression of GATA6 is sufficient to compensate for the defects caused by Grb2 deficiency in the development of the primitive and extraembryonic endoderm.


Subject(s)
Endoderm/embryology , Endoderm/metabolism , GATA6 Transcription Factor/metabolism , GRB2 Adaptor Protein/metabolism , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , GATA6 Transcription Factor/genetics , GRB2 Adaptor Protein/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Male , Mice , Mice, Knockout , Microscopy, Fluorescence , Nanog Homeobox Protein , RNA, Small Interfering
4.
Clin Cancer Res ; 11(2 Pt 1): 632-7, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15701850

ABSTRACT

PURPOSE: The cyclin-dependent kinase inhibitor p27(kip1) regulates cellular progression from G(1) to S phase. Several studies have shown that loss of p27(kip1) protein expression is associated with disease progression in various malignancies. The purpose of this study was to evaluate the subcellular localization of this cyclin-dependent kinase inhibitor in a large cohort of primary ovarian carcinomas and compare the results with clinicopathologic variables and overall survival. EXPERIMENTAL DESIGN: Subcellular localization of p27(kip1) was first assessed by Western blotting in nuclear and cytoplasmic extract from 13 cases of ovarian carcinoma. Subcellular localization of the p27(kip1) protein was evaluated using tissue microarrays containing 421 cases of ovarian carcinoma. RESULTS: The presence of p27(kip1) in the cytoplasm regardless of the nuclear stain correlated strongly with late-stage disease (P < 0.03), extent of cytoreduction (P = 0.03), and shorter disease-specific survival (P < 0.0001). CONCLUSION: Cytoplasmic localization of p27(kip1) predicts poorer prognosis in ovarian carcinoma, particularly in late-stage disease.


Subject(s)
Cell Cycle Proteins/metabolism , Ovarian Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/mortality , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/mortality , Carcinoma, Endometrioid/pathology , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cyclin-Dependent Kinase Inhibitor p27 , Cytoplasm/metabolism , Cytoplasm/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Middle Aged , Neoplasm Staging , Neoplasms, Cystic, Mucinous, and Serous/metabolism , Neoplasms, Cystic, Mucinous, and Serous/mortality , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Prognosis , Subcellular Fractions/metabolism , Survival Rate
5.
Mol Cell Biol ; 36(18): 2418-30, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27354067

ABSTRACT

We modeled the etiology of postmenopausal biology on ovarian cancer risk using germ cell-deficient white-spotting variant (Wv) mice, incorporating oncogenic mutations. Ovarian cancer incidence is highest in peri- and postmenopausal women, and epidemiological studies have established the impact of reproductive factors on ovarian cancer risk. Menopause as a result of ovarian follicle depletion is thought to contribute to higher cancer risk. As a consequence of follicle depletion, female Wv mice develop ovarian tubular adenomas, a benign epithelial tumor corresponding to surface epithelial invaginations and papillomatosis frequently found in postmenopausal human ovaries. Lineage tracing using MISR2-Cre indicated that the tubular adenomas that developed in Wv mice were largely derived from the MISR2 lineage, which marked only a fraction of ovarian surface and oviduct epithelial cells in wild-type tissues. Deletion of p27, either heterozygous or homozygous, was able to convert the benign tubular adenomas into more proliferative tumors. Restricted deletion of p53 in Wv/Wv mice by either intrabursal injection of adenoviral Cre or inclusion of the MISR2-Cre transgene also resulted in augmented tumor growth. This finding suggests that follicle depletion provides a permissive ovarian environment for oncogenic transformation of epithelial cells, presenting a mechanism for the increased ovarian cancer risk in postmenopausal women.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Ovarian Follicle/pathology , Ovarian Neoplasms/etiology , Tumor Suppressor Protein p53/genetics , Animals , Female , Humans , Mice , Mutation , Neoplasms, Experimental , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Postmenopause
6.
Neoplasia ; 17(1): 89-100, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25622902

ABSTRACT

White spotting variant (Wv) mice are spontaneous mutants attributed to a point mutation in the c-Kit gene, which reduces the tyrosine kinase activity to around 1% and affects the development of melanocytes, mast cells, and germ cells. Homozygous mutant mice are sterile but can live nearly a normal life span. The female Wv mice have a greatly reduced ovarian germ cell and follicle reserve at birth, and the remaining follicles are largely depleted soon after the females reach reproductive stage at around 7 weeks of age. Consequently, ovarian epithelial tumors develop in 100% of Wv females by 3 to 4 months of age. These tumors, called tubular adenomas, are benign but can become invasive in older Wv mice. We tested if additional genetic mutation(s) could convert the benign ovarian epithelial tumors to malignant tumors by crossing the Wv mutant into the Trp53 knockout background. Surprisingly, we found that global deletion of Trp53 suppressed the development of ovarian tubular adenomas in Wv mice. The ovaries of Wv/Wv; Trp53 (-/-) mice were covered by a single layer of surface epithelium and lacked excessive epithelial proliferation. Rather, the ovaries contained a small number of follicles. The presence of ovarian follicles and granulosa cells, as indicated by Pgc7 and inhibin-alpha expression, correlated with the absence of epithelial lesions. A reduction of Pten gene dosage, as in Wv/Wv; Pten (+/-) mice, produced a similar, though less dramatic, phenotype. We conclude that deletion of Trp53 prolongs the survival of ovarian follicles in Wv mice and consequently prevents the proliferation of ovarian epithelial cells and development of ovarian tubular adenomas. The results suggest that various cell types within the ovary communicate and mutually modulate, and an intact tissue environment is required to ensure homeostasis of ovarian surface epithelial cells. Especially, the current finding emphasizes the importance of ovarian follicles in suppressing the hyperplastic growth of ovarian epithelial cells, dominating over the loss of p53.


Subject(s)
Gene Deletion , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phenotype , Tumor Suppressor Protein p53/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Cell Proliferation , Disease Models, Animal , Female , Gene Dosage , Gene Expression , Inhibins/genetics , Inhibins/metabolism , Mice , Mice, Knockout , Models, Biological , Mutation , Nitric Oxide/biosynthesis , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Ovarian Neoplasms/metabolism , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-kit/genetics , Signal Transduction
7.
Hum Pathol ; 35(5): 552-9, 2004 May.
Article in English | MEDLINE | ID: mdl-15138928

ABSTRACT

Microsatellite instability (MSI) is commonly seen in tumors associated with the hereditary nonpolyposis colorectal cancer syndrome and is caused by defects in the DNA mismatch repair genes. MSI has also been observed in various sporadic cancers, including colorectal, gastric, and endometrial. The role and incidence of MSI in ovarian clear cell carcinoma remain unknown. This study was conducted to evaluate the frequency of MSI in ovarian clear cell carcinomas and to evaluate the sensitivity and specificity of immunohistochemistry in predicting mismatch-repair gene deficiency. A total of 42 ovarian clear cell carcinomas were analyzed for MSI using a panel of 5 microsatellite markers (BAT25, BAT26, D5S346, D2S123, and D17S250). Alterations in the expression of hMLH1 and hMSH2 proteins in these tumors were examined. Of the 42 ovarian clear cell tumors analyzed, 6 demonstrated a high level of MSI (MSI-H), 3 demonstrated a low level of MSI (MSI-L), and the remaining 33 exhibited microsatellite stability (MSS). No correlation was found between MSI level and patient age or tumor stage or size (P >0.05). Loss of expression of either hMLH1 or hMSH2 was observed in 4 of the 6 (67.7%) MSI-H tumors, whereas 34 of the 36 (94.4%) MSI-L or MSS tumors expressed both the hMLH1 and hMSH2 gene products. Our results indicate that MSI-H is involved in the development of a subset of ovarian clear cell carcinomas. A strong correlation exists between alterations in the expression of hMLH1 and hMSH2 and the presence of MSI-H in these tumors. However, immunohistochemical testing alone may miss a small fraction of cases with MSI-H.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , DNA-Binding Proteins/genetics , Microsatellite Repeats/genetics , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing , Adult , Aged , Carrier Proteins , Female , Humans , Immunohistochemistry , Middle Aged , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Nuclear Proteins
9.
PLoS One ; 5(2): e9295, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-20174585

ABSTRACT

BACKGROUND: The MAPK/ERK1/2 serine kinases are primary mediators of the Ras mitogenic signaling pathway. Phosphorylation by MEK activates MAPK/ERK in the cytoplasm, and phospho-ERK is thought to enter the nucleus readily to modulate transcription. PRINCIPAL FINDINGS: Here, however, we observe that in primary cultures of breast and ovarian epithelial cells, phosphorylation and activation of ERK1/2 are disassociated from nuclear translocalization and transcription of downstream targets, such as c-Fos, suggesting that nuclear translocation is limited in primary cells. Accordingly, in import assays in vitro, primary cells showed a lower import activity for ERK1/2 than cancer cells, in which activated MAPK readily translocated into the nucleus and activated c-Fos expression. Primary cells express lower levels of nuclear pore complex proteins and the nuclear transport factors, importin B1 and importin 7, which may explain the limiting ERK1/2 import found in primary cells. Additionally, reduction in expression of nucleoporin 153 by siRNA targeting reduced ERK1/2 nuclear activity in cancer cells. CONCLUSION: ERK1/2 activation is dissociated from nuclear entry, which is a rate limiting step in primary cells and in vivo, and the restriction of nuclear entry is disrupted in transformed cells by the increased expression of nuclear pores and/or nuclear transport factors.


Subject(s)
Cell Nucleus/metabolism , Epithelial Cells/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Active Transport, Cell Nucleus , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Enzyme Activation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Immunoblotting , Immunohistochemistry , Mammary Glands, Human/cytology , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovary/cytology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA Interference , Tissue Array Analysis
10.
Clin Cancer Res ; 16(12): 3171-81, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20423983

ABSTRACT

PURPOSE: Aurora kinase A (Aurora-A) is known to regulate genomic instability and tumorigenesis in multiple human cancers. The underlying mechanism, however, is not fully understood. We examined the molecular mechanism of Aurora-A regulation in human ovarian cancer. EXPERIMENTAL DESIGN: Retrovirus-mediated small hairpin RNA (shRNA) was used to silence the expression of Aurora-A in the ovarian cancer cell lines SKOV3, OVCA432, and OVCA433. Immunofluorescence, Western blotting, flow cytometry, cytogenetic analysis, and animal assay were used to test centrosome amplification, cell cycle alteration, apoptosis, DNA damage response, tumor growth, and genomic instability. Immunostaining of BRCA2 and Aurora-A was done in ovarian, pancreatic, breast, and colon cancer samples. RESULTS: Knockdown of Aurora-A reduced centrosome amplification, malformation of mitotic spindles, and chromosome aberration, leading to decreased tumor growth. Silencing Aurora-A attenuated cell cycle progression and enhanced apoptosis and DNA damage response by restoring p21, pRb, and BRCA2 expression. Aurora-A was inversely correlated with BRCA2 in high-grade ovarian serous carcinoma, breast cancer, and pancreatic cancer. In high-grade ovarian serous carcinoma, positive expression of BRCA2 predicted increased overall and disease-free survival, whereas positive expression of Aurora-A predicted poor overall and disease-free survival (P < 0.05). Moreover, an increased Aurora-A to BRCA2 expression ratio predicted poor overall survival (P = 0.047) compared with a decreased Aurora-A to BRCA2 expression ratio. CONCLUSION: Aurora-A regulates genomic instability and tumorigenesis through cell cycle dysregulation and BRCA2 suppression. The negative correlation between Aurora-A and BRCA2 exists in multiple cancers, whereas the expression ratio of Aurora-A to BRCA2 predicts ovarian cancer patient outcome.


Subject(s)
BRCA2 Protein/antagonists & inhibitors , Cell Cycle , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Serine-Threonine Kinases/physiology , Apoptosis , Aurora Kinase A , Aurora Kinases , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , DNA Damage , Female , Gene Silencing , Genomic Instability , Humans , Protein Serine-Threonine Kinases/antagonists & inhibitors
11.
Dev Dyn ; 238(3): 514-23, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19191218

ABSTRACT

Disabled-2 (Dab2) is expressed in primitive endoderm cells as they are differentiating from the inner cell mass and dab2 deficiency in mice results in lethality at E5.5-E6.5 due to the disorganization of the endoderm layers. Here we show that Dab2 suppresses c-Fos expression in endoderm cells. A morphological normal primitive endoderm layer was observed in putative E5.5 dab2 (-/-):c-fos (-/-) embryos, indicating that the primitive endoderm defect due to the loss of Dab2 is rescued by deletion of the c-fos gene. The lethality of the double knockout embryos was delayed until E9.5-E10.5 and the defective embryos failed to undergo organogenesis. We conclude that Dab2 plays a role in epithelial organization by suppression of c-Fos expression and suggest that unsuppressed c-Fos can lead to disruption of primitive endoderm epithelial organization, yet an additional dab2 function is required for later organogenesis.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Endoderm/embryology , Endoderm/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Animals , Apoptosis Regulatory Proteins , Cell Differentiation , Down-Regulation , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Gene Deletion , Gene Expression Regulation, Developmental , Genotype , Mice , Mice, Knockout , Proto-Oncogene Proteins c-fos/deficiency , Proto-Oncogene Proteins c-fos/genetics , Stem Cells
12.
PLoS One ; 4(7): e6454, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19649254

ABSTRACT

BACKGROUND: The family of zinc finger-containing GATA transcription factors plays critical roles in cell lineage specification during early embryonic development and organ formation. GATA4 and GATA6 were found to be frequently lost in ovarian cancer, and the loss is proposed to account for dedifferentiation of the cancer cells. METHODOLOGY/PRINCIPAL FINDINGS: We further investigated the expression of GATA4 and GATA6 in ovarian surface epithelial lesions and histological subtypes of ovarian carcinomas by immunostaining. GATA4 and GATA6 were found to be absent in high percentages (80 to 90%) of serous, clear cell, and endometrioid ovarian cancer examined. In contrast, both were found positive in 11 out of 12 cases of mucinous carcinomas, suggesting the expression of the GATA factors can distinguish mucinous cancer from other histological subtypes. GATA4 was frequently lost in preneoplastic lesions such as morphologically normal inclusion cysts and epithelial hyperplasia adjacent to malignant cells. The loss of GATA6 correlates closely with neoplastic morphological transformation of ovarian surface epithelia. In culture, GATA4 expression was progressively reduced upon passaging primary ovarian surface epithelial cells, which correlated with changes in histone modification of the GATA4 locus. A reduced GATA6 gene dosage as in GATA6 (+/-) mice led to an increased pre-neoplastic changes and inclusion cysts in the ovaries, suggesting the loss of GATA6 contributes to ovarian cancer development. CONCLUSIONS/SIGNIFICANCE: This study suggests that the expression status of GATA4 and GATA6 may dictate distinct pathologic pathways leading to serous or mucinous ovarian carcinomas. The readily loss of GATA4 expression through changes in chromatin conformation suggests a potential non-phenotypic initiating event, leading to subsequent loss of GATA6, morphological transformation, and ultimate tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , GATA4 Transcription Factor/metabolism , GATA6 Transcription Factor/metabolism , Ovarian Neoplasms/metabolism , Ovary/cytology , Animals , Cell Line, Transformed , Chromatin/metabolism , Female , Humans , Immunohistochemistry , Mice , Ovarian Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction
13.
Cancer Res ; 68(2): 444-9, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18199538

ABSTRACT

Activation of the phosphatidylinositol-3-OH kinase (PI3K) signaling cascade is becoming increasingly recognized as a common feature of thyroid follicular neoplasms. We have recently shown that conditional loss of Pten in the mouse thyroid follicular cells is sufficient to stimulate continuous autonomous growth, leading to a homogeneously hyperplastic gland and to the development of follicular adenomas. Because the PI3K/AKT cascade can activate a plethora of different signaling pathways, it is still unclear which of these may represent the key mitogenic output of PI3K-initiated signaling. Here, we show that the in vivo proliferative response to chronic PI3K activation profoundly relies on the activation of the mammalian target of rapamycin (mTOR)/S6K1 axis, and that mTOR inhibition in Pten mutant mice and cells restores virtually normal proliferation rates, despite the presence of still elevated Akt activity, at least in part by down-regulating cyclins D1 and D3, and without affecting cell survival.


Subject(s)
Cell Proliferation , Epithelium/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/physiology , Thyroid Gland/physiology , Adenocarcinoma, Follicular/pathology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Epithelium/metabolism , Everolimus , Humans , Mice , Mice, Transgenic , PTEN Phosphohydrolase/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Signal Transduction/physiology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Thyroid Gland/metabolism , Thyroid Neoplasms/pathology
14.
Am J Pathol ; 170(4): 1325-36, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17392171

ABSTRACT

Menopausal ovaries undergo morphological changes, known as ovarian aging, which are implicated in the high incidence of ovarian cancer occurring during the perimenopausal and immediate postmenopausal periods. The germ cell-deficient Wv mice recapitulate these postmenopausal alterations in ovarian morphology and develop tubular adenomas. We demonstrate that a reduction of cyclooxygenase 2 gene dosage rescued the ovarian aging phenotype of the Wv mice, whereas homozygous deletion was accompanied by a compensatory increase in ovarian cyclooxygenase 1 expression and prostaglandin E(2) synthesis. Cyclooxygenase inhibitors also reduced the tumor phenotype in a preliminary study. These findings suggest that increased cyclooxygenase activity contributes to the preneoplastic morphological changes of the ovarian surface epithelium, which can be reversed by a reduction of gene dosage achieved by either genetic or pharmacological approaches.


Subject(s)
Aging/pathology , Cyclooxygenase 2/metabolism , Ovarian Neoplasms/pathology , Ovary/pathology , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Blotting, Western , Celecoxib , Cell Transformation, Neoplastic/drug effects , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Cyclooxygenase Inhibitors/administration & dosage , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/metabolism , Epithelium/metabolism , Epithelium/pathology , Female , Gene Dosage , Heterozygote , Homozygote , Humans , Inbreeding , Indomethacin/administration & dosage , Indomethacin/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovary/drug effects , Ovary/metabolism , Phenotype , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Sulfonamides/administration & dosage , Sulfonamides/pharmacology
15.
Mol Carcinog ; 46(2): 130-43, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17131304

ABSTRACT

The role for matrix metalloproteinases (MMPs) in tumor cells invasion and metastasis is well established, and expression of MMPs is recognized as an indication of tumor cell malignancy. Previous studies suggest that the degradation of the basement membrane is a crucial early step in epithelial transformation and ovarian tumorigenesis. Thus, MMPs may also express and exert a role in preneoplastic lesions of ovarian tissues. We investigated the expression of the major metalloproteinases, gelatinase A, 72 kDa type IV collagenase (MMP-2), and gelatinase B, 92 kDa type IV collagenase (MMP-9), and the presence of basement membrane in ovarian tumors and tissues from prophylactic oophorectomies using immunostaining. MMP expression was also characterized in a panel of ovarian cancer cell lines and several nontumorigenic ovarian surface epithelial primary cells by zymography, Northern, and Western blots. We found, surprisingly, that MMP-2 and MMP-9 are expressed more frequently in early lesions than in established carcinomas. No correlation was found between the expression of MMPs and tumor grades or stages. In preneoplastic lesions, MMP-2 or MMP-9 expression often associates with the absence of basement membrane and morphological alterations. MMP-2 is often expressed in nontumorigenic ovarian surface epithelial cells but reduced or absent in cancer cells. Thus, we conclude that MMPs expression does not correlate with the malignancy of ovarian epithelial cells as generally thought. Rather, increased metalloproteinase expression is an early event in ovarian tumorigenesis and associates with the loss of epithelial basement membrane and morphological transformation. We propose that the increased MMP activity is an etiological factor for ovarian cancer risk. We found that MMPs expression does not correlate with the malignancy of ovarian epithelial cells as generally thought. Rather, increased metalloproteinase expression is an early event in ovarian tumorigenesis. The finding suggests roles of MMP in tumor initiation in addition to invasion, and may impact on the strategy for use of MMP inhibitors in cancer prevention.


Subject(s)
Cell Transformation, Neoplastic , Matrix Metalloproteinases/metabolism , Ovarian Neoplasms/enzymology , Blotting, Northern , Blotting, Western , Cell Line, Transformed , Female , Humans , Immunohistochemistry , Ovarian Neoplasms/pathology , RNA, Small Interfering , Tissue Array Analysis
16.
Am J Pathol ; 169(1): 258-67, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16816378

ABSTRACT

Disabled-2 (Dab2) is a phosphoprotein involved in cellular signal transduction and endocytic trafficking. The expression of Dab2 is frequently lost or suppressed in several epithelial tumors, and studies of its cellular function and growth suppressive activity when re-expressed in cancer cells led to the suggestion that Dab2 is a tumor suppressor. A role for Dab2 in epithelial cell positioning organization was derived from study of knockout mice: homozygous deletion of dab2 results in early embryonic lethality due to the disorganization of the primitive endoderm, the first epithelium in early embryos. We now report that dab2 heterozygous mice develop uterine hyperplasia and ovarian preneoplastic morphological changes at a high frequency. Crossing into a p53(-/-) background unexpectedly produced few female dab2(+/-):p53(-/-) mice, while the male dab2(+/-):p53(-/-) were born at the expected Mendelian frequency. The tumor-prone phenotype of dab2(+/-) mice provides additional support for a role of human Dab2 as a tumor suppressor, and the sex-biased embryonic lethality suggests a genetic interaction between p53 and dab2 genes in female mice.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Adenocarcinoma/genetics , Endometrial Neoplasms/genetics , Genes, p53 , Ovarian Neoplasms/genetics , Precancerous Conditions/genetics , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Blotting, Western , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Embryo, Mammalian , Female , Genes, Tumor Suppressor , Heterozygote , Humans , Male , Mice , Mice, Knockout , Polymerase Chain Reaction , Precancerous Conditions/pathology , Sex Factors
17.
Gynecol Oncol ; 103(2): 719-28, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16698071

ABSTRACT

OBJECTIVE: From analysis of pre-cancer ovarian tissues obtained from prophylactic oophorectomies, several studies reported the increased ovarian morphological changes in high-risk ovaries, but whether these morphological changes are associated with BRCA1/BRCA2 genotypes or are cancer precursors is controversial. Here, we have investigated a recent collection of ovaries from prophylactic oophorectomies and control ovaries from surgeries due to other non-ovarian-related cancer or non-neoplastic diseases to determine if ovarian morphological changes relate to BRCA1/2 genotypes or reproductive history. METHODS: We assembled a panel of archived ovarian tissues: 52 ovarian tissue blocks were from prophylactic oophorectomies of a high-risk (HR) population; 66 ovaries were from surgeries due to non-ovarian-related diseases, referred to as normal-risk (NR) group. The morphology of ovarian tissues was examined, and morphological changes including papillomatosis, invaginations, inclusion cysts, and epithelial stratification were assessed in a blinded fashion. RESULTS: No statistically significant difference in frequency of these histolopathologic features was found between HR and NR groups. However, inclusion cysts and deep invaginations were found much more commonly in women age 45-54 of either HR or NR groups. CONCLUSIONS: This study suggests that no significant increase in the presence of non-neoplastic ovarian morphological changes is associated with BRCA1/BRCA2 mutations. Rather, the frequency of these histological features, especially inclusion cysts, may associate with age or menopausal status. We propose that ovulatory and perimenopausal gonadotropin stimulation produces ovarian morphological changes, and these histological features may promote the transformation of genetically compromised epithelial cells in the development of ovarian cancer.


Subject(s)
Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovary/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Age Factors , Aged , Female , Genes, BRCA1 , Genes, BRCA2 , Humans , Middle Aged , Mutation , Risk Factors
18.
J Biol Chem ; 279(6): 4339-45, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-14625284

ABSTRACT

The Her-2/neu oncogene is overexpressed in approximately 30% of breast and ovarian cancer cases and often indicates a poor prognosis. Therapeutic agents against Her-2/neu have been intensively sought over the past decade. Here we show that small interfering RNA (siRNA) can silence the expression of Her-2/neu in models of human breast or ovarian cancer through retrovirus-mediated transfer of an siRNA against Her-2/neu. Cells infected with retrovirus expressing anti-Her-2/neu siRNA exhibit slower proliferation, increased apoptosis, increased G0/G1 arrest, and decreased tumor growth. Changes in cell cycle-associated factors included decreased levels of phosphatidylinositol 3-kinase, pAkt, and cyclin D1 and increased levels of p27 and phosphorylated retinoblastoma protein. Knockdown of Her-2/neu expression by siRNA is also associated with increased expression of the anti-angiogenic factor thrombospondin-1 and decreased expression of the pro-angiogenic vascular endothelial growth factor, suggesting that Her-2/neu stimulates tumor growth at least in part by regulating angiogenesis. siRNA-mediated gene silencing of Her-2/neu and increasing the expression of thrombospondin-1 may be a useful therapeutic strategy for Her-2/neu-over-expressing breast or ovarian cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Genes, erbB-2 , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Small Interfering/genetics , Apoptosis , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Cycle Proteins/metabolism , Cell Division , Cell Line, Tumor , Female , Gene Expression , Gene Silencing , Genetic Vectors , Humans , Neovascularization, Pathologic/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/therapy , RNA, Small Interfering/pharmacology , Retroviridae/genetics , Signal Transduction , Thrombospondin 1/genetics , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL