Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 15(6): e0233962, 2020.
Article in English | MEDLINE | ID: mdl-32525899

ABSTRACT

High grade serous ovarian cancer (HGSC) is the most common and deadly type of ovarian cancer, largely due to difficulties in early diagnosis and rapid metastasis throughout the peritoneal cavity. Previous studies have shown that expression of Notch3 correlates with worse prognosis and increased tumorigenic cell behaviors in HGSC. We investigated the mechanistic role of Notch3 in a model of metastatic ovarian cancer using the murine ovarian surface epithelial cell line, ID8 IP2. Notch3 was activated in ID8 IP2 cells via expression of the Notch3 intracellular domain (Notch3IC). Notch3IC ID8 IP2 cells injected intraperitoneally caused accelerated ascites and reduced survival compared to control ID8 IP2, particularly in early stages of disease. We interrogated downstream targets of Notch3IC in ID8 IP2 cells by RNA sequencing and found significant induction of genes that encode adhesion and extracellular matrix proteins. Notch3IC ID8 IP2 showed increased expression of ITGA1 mRNA and cell-surface protein. Notch3IC-mediated increase of ITGA1 was also seen in two human ovarian cancer cells. Notch3IC ID8 IP2 cells showed increased adhesion to collagens I and IV in vitro. We propose that Notch3 activation in ovarian cancer cells causes increased adherence to collagen-rich peritoneal surfaces. Thus, the correlation between increased Notch3 signaling and poor prognosis may be influenced by increased metastasis of HGSC via increased adherence of disseminating cells to new metastatic sites in the peritoneum.


Subject(s)
Carcinoma, Ovarian Epithelial/secondary , Cystadenocarcinoma, Serous/secondary , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Receptor, Notch3/metabolism , Animals , Carcinogenesis/metabolism , Carcinoma, Ovarian Epithelial/metabolism , Cell Adhesion , Cell Line, Tumor , Cystadenocarcinoma, Serous/metabolism , Disease Progression , Extracellular Matrix Proteins/metabolism , Female , Humans , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/metabolism , Receptor, Notch3/genetics
2.
Front Pediatr ; 8: 35, 2020.
Article in English | MEDLINE | ID: mdl-32117840

ABSTRACT

Background: Optimal timing for resection of asymptomatic congenital lung malformations (CLMs) remains controversial. The aim of this study is to define optimal timing for surgical intervention of patients with CLMs and define clinical variables that affect surgical outcomes. Methods: An IRB-approved retrospective analysis was conducted for patients undergoing surgery for CLMs between 2012 and 2017. Subjects were divided into cohorts based on timing of operative intervention. "Early intervention" was defined as surgery within 4 months of birth; "intermediate intervention"-between 4 and 6 months; and "late intervention"-6-12 months. Surgical outcomes including intraoperative estimated blood loss (EBL), surgical time, post-operative pneumothorax, length of time chest tube stayed in, and hospital length of stay were compared among the three groups using Fisher's exact test or Chi-squared test for categorical variables and one-way analysis of variance test for continuous variables. Results: We analyzed 63 patients who underwent surgery for CLM. There were no significant differences in baseline characteristics. Timing of surgery did not significantly correlate with post-operative outcomes. Specifically, there was no difference in operative time, EBL, post-operative pneumothorax, or length of hospital stay among the early, intermediate, and late intervention groups. Even after controlling for cyst-volume ratio (CVR), timing of surgery still did not affect post-operative outcomes. Conclusions: Surgical outcomes for resection of CLMs are not significantly affected by timing of surgery. We advocate for early intervention to decrease the incidence of associated complications that can occur with later intervention.

3.
BMC Cancer ; 14: 884, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25427824

ABSTRACT

BACKGROUND: Endometrial cancer (EC) is the 8th leading cause of cancer death amongst American women. Most ECs are endometrioid, serous, or clear cell carcinomas, or an admixture of histologies. Serous and clear ECs are clinically aggressive tumors for which alternative therapeutic approaches are needed. The purpose of this study was to search for somatic mutations in the tyrosine kinome of serous and clear cell ECs, because mutated kinases can point to potential therapeutic targets. METHODS: In a mutation discovery screen, we PCR amplified and Sanger sequenced the exons encoding the catalytic domains of 86 tyrosine kinases from 24 serous, 11 clear cell, and 5 mixed histology ECs. For somatically mutated genes, we next sequenced the remaining coding exons from the 40 discovery screen tumors and sequenced all coding exons from another 72 ECs (10 clear cell, 21 serous, 41 endometrioid). We assessed the copy number of mutated kinases in this cohort of 112 tumors using quantitative real time PCR, and we used immunoblotting to measure expression of these kinases in endometrial cancer cell lines. RESULTS: Overall, we identified somatic mutations in TNK2 (tyrosine kinase non-receptor, 2) and DDR1 (discoidin domain receptor tyrosine kinase 1) in 5.3% (6 of 112) and 2.7% (3 of 112) of ECs. Copy number gains of TNK2 and DDR1 were identified in another 4.5% and 0.9% of 112 cases respectively. Immunoblotting confirmed TNK2 and DDR1 expression in endometrial cancer cell lines. Three of five missense mutations in TNK2 and one of two missense mutations in DDR1 are predicted to impact protein function by two or more in silico algorithms. The TNK2(P761Rfs*72) frameshift mutation was recurrent in EC, and the DDR1(R570Q) missense mutation was recurrent across tumor types. CONCLUSIONS: This is the first study to systematically search for mutations in the tyrosine kinome in clear cell endometrial tumors. Our findings indicate that high-frequency somatic mutations in the catalytic domains of the tyrosine kinome are rare in clear cell ECs. We uncovered ten new mutations in TNK2 and DDR1 within serous and endometrioid ECs, thus providing novel insights into the mutation spectrum of each gene in EC.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Cystadenocarcinoma, Serous/genetics , Endometrial Neoplasms/genetics , Mutation , Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adenocarcinoma, Clear Cell/pathology , Cell Line, Tumor , Cystadenocarcinoma, Serous/pathology , DNA Copy Number Variations , DNA Mutational Analysis , DNA Polymerase II/genetics , Discoidin Domain Receptor 1 , Endometrial Neoplasms/pathology , Female , Humans , Microsatellite Instability , Neoplasm Grading , Poly-ADP-Ribose Binding Proteins , Protein Interaction Domains and Motifs/genetics
4.
PLoS One ; 8(6): e63313, 2014.
Article in English | MEDLINE | ID: mdl-23755103

ABSTRACT

Most endometrial cancers can be classified histologically as endometrioid, serous, or clear cell. Non-endometrioid endometrial cancers (NEECs; serous and clear cell) are the most clinically aggressive of the three major histotypes and are characterized by aneuploidy, a feature of chromosome instability. The genetic alterations that underlie chromosome instability in endometrial cancer are poorly understood. In the present study, we used Sanger sequencing to search for nucleotide variants in the coding exons and splice junctions of 21 candidate chromosome instability genes, including 19 genes implicated in sister chromatid cohesion, from 24 primary, microsatellite-stable NEECs. Somatic mutations were verified by sequencing matched normal DNAs. We subsequently resequenced mutated genes from 41 additional NEECs as well as 42 endometrioid ECs (EECs). We uncovered nonsynonymous somatic mutations in ESCO1, CHTF18, and MRE11A in, respectively, 3.7% (4 of 107), 1.9% (2 of 107), and 1.9% (2 of 107) of endometrial tumors. Overall, 7.7% (5 of 65) of NEECs and 2.4% (1 of 42) of EECs had somatically mutated one or more of the three genes. A subset of mutations are predicted to impact protein function. The co-occurrence of somatic mutations in ESCO1 and CHTF18 was statistically significant (P = 0.0011, two-tailed Fisher's exact test). This is the first report of somatic mutations within ESCO1 and CHTF18 in endometrial tumors and of MRE11A mutations in microsatellite-stable endometrial tumors. Our findings warrant future studies to determine whether these mutations are driver events that contribute to the pathogenesis of endometrial cancer.


Subject(s)
Acetyltransferases/genetics , Carrier Proteins/genetics , Chromosomal Instability/genetics , DNA-Binding Proteins/genetics , Endometrial Neoplasms/genetics , Genetic Association Studies , Mutation/genetics , Nuclear Proteins/genetics , ATPases Associated with Diverse Cellular Activities , Cell Line, Tumor , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Humans , MRE11 Homologue Protein
5.
Nat Genet ; 44(12): 1310-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23104009

ABSTRACT

Endometrial cancer is the sixth most commonly diagnosed cancer in women worldwide, causing ~74,000 deaths annually. Serous endometrial cancers are a clinically aggressive subtype with a poorly defined genetic etiology. We used whole-exome sequencing to comprehensively search for somatic mutations within ~22,000 protein-encoding genes in 13 primary serous endometrial tumors. We subsequently resequenced 18 genes, which were mutated in more than 1 tumor and/or were components of an enriched functional grouping, from 40 additional serous tumors. We identified high frequencies of somatic mutations in CHD4 (17%), EP300 (8%), ARID1A (6%), TSPYL2 (6%), FBXW7 (29%), SPOP (8%), MAP3K4 (6%) and ABCC9 (6%). Overall, 36.5% of serous tumors had a mutated chromatin-remodeling gene, and 35% had a mutated ubiquitin ligase complex gene, implicating frequent mutational disruption of these processes in the molecular pathogenesis of one of the deadliest forms of endometrial cancer.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Carcinoma, Endometrioid/genetics , Chromatin Assembly and Disassembly/genetics , Endometrial Neoplasms/genetics , Exome/genetics , Ubiquitin-Protein Ligase Complexes/genetics , ATP-Binding Cassette Transporters/genetics , Adult , Autoantigens/genetics , Base Sequence , Cell Cycle Proteins/genetics , DNA-Binding Proteins , E1A-Associated p300 Protein/genetics , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Female , Gene Frequency , Humans , MAP Kinase Kinase Kinase 4/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Repressor Proteins/genetics , Sequence Analysis, DNA , Sulfonylurea Receptors , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics
6.
PLoS Genet ; 7(8): e1002245, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21901109

ABSTRACT

ATAD5, the human ortholog of yeast Elg1, plays a role in PCNA deubiquitination. Since PCNA modification is important to regulate DNA damage bypass, ATAD5 may be important for suppression of genomic instability in mammals in vivo. To test this hypothesis, we generated heterozygous (Atad5(+/m)) mice that were haploinsuffficient for Atad5. Atad5(+/m) mice displayed high levels of genomic instability in vivo, and Atad5(+/m) mouse embryonic fibroblasts (MEFs) exhibited molecular defects in PCNA deubiquitination in response to DNA damage, as well as DNA damage hypersensitivity and high levels of genomic instability, apoptosis, and aneuploidy. Importantly, 90% of haploinsufficient Atad5(+/m) mice developed tumors, including sarcomas, carcinomas, and adenocarcinomas, between 11 and 20 months of age. High levels of genomic alterations were evident in tumors that arose in the Atad5(+/m) mice. Consistent with a role for Atad5 in suppressing tumorigenesis, we also identified somatic mutations of ATAD5 in 4.6% of sporadic human endometrial tumors, including two nonsense mutations that resulted in loss of proper ATAD5 function. Taken together, our findings indicate that loss-of-function mutations in mammalian Atad5 are sufficient to cause genomic instability and tumorigenesis.


Subject(s)
Adenosine Triphosphatases/genetics , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Neoplasms/genetics , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/metabolism , Aneuploidy , Animals , Cell Line , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Endometrial Neoplasms/genetics , Female , Genetic Predisposition to Disease , Genomic Instability , Humans , Male , Mice , Mutation/genetics , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination
7.
Clin Cancer Res ; 17(6): 1331-40, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21266528

ABSTRACT

PURPOSE: The goal of this study was to comprehensively define the incidence of mutations in all exons of PIK3CA in both endometrioid endometrial cancer (EEC) and nonendometrioid endometrial cancer (NEEC). EXPERIMENTAL DESIGN: We resequenced all coding exons of PIK3CA and PTEN, and exons 1 and 2 of KRAS, from 108 primary endometrial tumors. Somatic mutations were confirmed by sequencing matched normal DNAs. The biochemical properties of a subset of novel PIK3CA mutations were determined by exogenously expressing wild type and mutant constructs in U2OS cells and measuring levels of AKT(Ser473) phosphorylation. RESULTS: Somatic PIK3CA mutations were detected in 52.4% of 42 EECs and 33.3% of 66 NEECs. Half (29 of 58) of all nonsynonymous PIK3CA mutations were in exons 1-7 and half were in exons 9 and 20. The exons 1-7 mutations localized to the ABD, ABD-RBD linker and C2 domains of p110α. Within these regions, Arg88, Arg93, Gly106, Lys111, Glu365, and Glu453, were recurrently mutated; Arg88, Arg93, and Lys111 formed mutation hotspots. The p110α-R93W, -G106R, -G106V, -K111E, -delP449-L455, and -E453K mutants led to increased levels of phospho-AKT(Ser473) compared to wild-type p110α. Overall, 62% of exons 1-7 PIK3CA mutants and 64% of exons 9-20 PIK3CA mutants were activating; 72% of exon 1-7 mutations have not previously been reported in endometrial cancer. CONCLUSIONS: Our study identified a new subgroup of endometrial cancer patients with activating mutations in the amino-terminal domains of p110α; these patients might be appropriate for consideration in clinical trials of targeted therapies directed against the PI3K pathway.


Subject(s)
Carcinoma/genetics , Endometrial Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/physiology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , DNA Mutational Analysis , Exons , Female , Humans , PTEN Phosphohydrolase/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras) , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...