Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Mol Carcinog ; 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990091

ABSTRACT

Ovarian cancer (OCa) is the deadliest of all gynecological cancers. The standard treatment for OCa is platinum-based chemotherapy, such as carboplatin or cisplatin in combination with paclitaxel. Most patients are initially responsive to these treatments; however, nearly 90% will develop recurrence and inevitably succumb to chemotherapy-resistant disease. Recent studies have revealed that the epigenetic modifier lysine-specific histone demethylase 1A (KDM1A/LSD1) is highly overexpressed in OCa. However, the role of KDM1A in chemoresistance and whether its inhibition enhances chemotherapy response in OCa remains uncertain. Analysis of TCGA datasets revealed that KDM1A expression is high in patients who poorly respond to chemotherapy. Western blot analysis show that treatment with chemotherapy drugs cisplatin, carboplatin, and paclitaxel increased KDM1A expression in OCa cells. KDM1A knockdown (KD) or treatment with KDM1A inhibitors NCD38 and SP2509 sensitized established and patient-derived OCa cells to chemotherapy drugs in reducing cell viability and clonogenic survival and inducing apoptosis. Moreover, knockdown of KDM1A sensitized carboplatin-resistant A2780-CP70 cells to carboplatin treatment and paclitaxel-resistant SKOV3-TR cells to paclitaxel. RNA-seq analysis revealed that a combination of KDM1A-KD and cisplatin treatment resulted in the downregulation of genes related to epithelial-mesenchymal transition (EMT). Interestingly, cisplatin treatment increased a subset of NF-κB pathway genes, and KDM1A-KD or KDM1A inhibition reversed this effect. Importantly, KDM1A-KD, in combination with cisplatin, significantly reduced tumor growth compared to a single treatment in an orthotopic intrabursal OCa xenograft model. Collectively, these findings suggest that combination of KDM1A inhibitors with chemotherapy could be a promising therapeutic approach for the treatment of OCa.

2.
Int J Mol Sci ; 24(24)2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38139260

ABSTRACT

Endometrial cancer (ECa) is the most common female gynecologic cancer. When comparing the two histological subtypes of endometrial cancer, Type II tumors are biologically more aggressive and have a worse prognosis than Type I tumors. Current treatments for Type II tumors are ineffective, and new targeted therapies are urgently needed. LIFR and its ligand, LIF, have been shown to play a critical role in the progression of multiple solid cancers and therapy resistance. The role of LIF/LIFR in the progression of Type II ECa, on the other hand, is unknown. We investigated the role of LIF/LIFR signaling in Type II ECa and tested the efficacy of EC359, a novel small-molecule LIFR inhibitor, against Type II ECa. The analysis of tumor databases has uncovered a correlation between diminished survival rates and increased expression of leukemia inhibitory factor (LIF), suggesting a potential connection between altered LIF expression and unfavorable overall survival in Type II ECa. The results obtained from cell viability and colony formation assays demonstrated a significant decrease in the growth of Type II ECa LIFR knockdown cells in comparison to vector control cells. Furthermore, in both primary and established Type II ECa cells, pharmacological inhibition of the LIF/LIFR axis with EC359 markedly decreased cell viability, long-term cell survival, and invasion, and promoted apoptosis. Additionally, EC359 treatment reduced the activation of pathways driven by LIF/LIFR, such as AKT, mTOR, and STAT3. Tumor progression was markedly inhibited by EC359 treatment in two different patient-derived xenograft models in vivo and patient-derived organoids ex vivo. Collectively, these results suggest LIFR inhibitor EC359 as a possible new small-molecule therapeutics for the management of Type II ECa.


Subject(s)
Endometrial Neoplasms , Signal Transduction , Humans , Female , Receptors, OSM-LIF/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Endometrial Neoplasms/drug therapy
3.
Int J Mol Sci ; 23(13)2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35806169

ABSTRACT

Ovarian cancer (OCa) is the deadliest gynecologic cancer. Emerging studies suggest ovarian cancer stem cells (OCSCs) contribute to chemotherapy resistance and tumor relapse. Recent studies demonstrated estrogen receptor beta (ERß) exerts tumor suppressor functions in OCa. However, the status of ERß expression in OCSCs and the therapeutic utility of the ERß agonist LY500307 for targeting OCSCs remain unknown. OCSCs were enriched from ES2, OV90, SKOV3, OVSAHO, and A2780 cells using ALDEFLUOR kit. RT-qPCR results showed ERß, particularly ERß isoform 1, is highly expressed in OCSCs and that ERß agonist LY500307 significantly reduced the viability of OCSCs. Treatment of OCSCs with LY500307 significantly reduced sphere formation, self-renewal, and invasion, while also promoting apoptosis and G2/M cell cycle arrest. Mechanistic studies using RNA-seq analysis demonstrated that LY500307 treatment resulted in modulation of pathways related to cell cycle and apoptosis. Western blot and RT-qPCR assays demonstrated the upregulation of apoptosis and cell cycle arrest genes such as FDXR, p21/CDKN1A, cleaved PARP, and caspase 3, and the downregulation of stemness markers SOX2, Oct4, and Nanog. Importantly, treatment of LY500307 significantly attenuated the tumor-initiating capacity of OCSCs in orthotopic OCa murine xenograft models. Our results demonstrate that ERß agonist LY500307 is highly efficacious in reducing the stemness and promoting apoptosis of OCSCs and shows significant promise as a novel therapeutic agent in treating OCa.


Subject(s)
Estrogen Receptor beta , Ovarian Neoplasms , Animals , Cell Line, Tumor , Cell Proliferation , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogens/metabolism , Female , Humans , Mice , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology
4.
Int J Gynecol Cancer ; 26(6): 1129-36, 2016 07.
Article in English | MEDLINE | ID: mdl-27327152

ABSTRACT

OBJECTIVES: Approximately 3% to 5% of endometrial cancers (EC) are associated with Lynch syndrome (LS). The clinical characteristics and prevalence of LS have not been well studied in the US Hispanic population. Hispanics are the largest and fastest growing ethnic minority group in the United States. We sought to characterize the demographics, tumor characteristics, and prevalence of loss of mismatch repair (MMR) protein expression in a large Hispanic population with EC. METHODS: From January 1, 2005, to August 1, 2012, 83 women of Hispanic ethnicity diagnosed with EC 50 years and younger were identified. Clinical and pathologic data were abstracted from the electronic medical record. Tumor studies included immunohistochemistry of MLH1, MSH2, MSH6, and PMS2 and methylation of the MLH1 promoter. RESULTS: Ninety-five percent of patients were overweight or obese. The mean body mass index was 40.1 kg/m, 75% had irregular menses, 36% had diabetes, 46% were nulliparous, and 95% had endometrioid histology. Thirteen patients (15.7%) had tumor MMR deficiency due to a presumed germline mutation (9 MSH6, 3 MSH2, and 1 MLH1). The pattern of MMR protein loss was consistent with the expected binding properties of the MMR heterodimer complexes. No significant difference was found in clinical or pathological variables between patients with and without MMR deficient tumors. CONCLUSIONS: The prevalence of molecular findings consistent with LS was at least as high as other populations of varied geography, race, and ethnicity. We found no reliable factors to include body mass index, family history, synchronous tumors, or pathologic tumor features to serve as triage markers for which ECs should be screened for MMR protein loss. Our findings support a recommendation for universal screening of ECs utilizing 2-antibody testing with MLH1 promoter methylation testing as indicated up to 60 years or older. Our recommendations should be generalizable to other Hispanic populations in the Southern United States.


Subject(s)
DNA Mismatch Repair , Endometrial Neoplasms/ethnology , Endometrial Neoplasms/genetics , Adult , Cohort Studies , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA, Neoplasm/genetics , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/pathology , Female , Hispanic or Latino , Humans , Middle Aged , Obesity/ethnology , Obesity/genetics , Obesity/pathology
5.
NPJ Precis Oncol ; 8(1): 118, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789520

ABSTRACT

Of all gynecologic cancers, epithelial-ovarian cancer (OCa) stands out with the highest mortality rates. Despite all efforts, 90% of individuals who receive standard surgical and cytotoxic therapy experience disease recurrence. The precise mechanism by which leukemia inhibitory factor (LIF) and its receptor (LIFR) contribute to the progression of OCa remains unknown. Analysis of cancer databases revealed that elevated expression of LIF or LIFR was associated with poor progression-free survival of OCa patients and a predictor of poor response to chemotherapy. Using multiple primary and established OCa cell lines or tissues that represent five subtypes of epithelial-OCa, we demonstrated that LIF/LIFR autocrine signaling is active in OCa. Moreover, treatment with LIFR inhibitor, EC359 significantly reduced OCa cell viability and cell survival with an IC50 ranging from 5-50 nM. Furthermore, EC359 diminished the stemness of OCa cells. Mechanistic studies using RNA-seq and rescue experiments unveiled that EC359 primarily induced ferroptosis by suppressing the glutathione antioxidant defense system. Using multiple in vitro, ex vivo and in vivo models including cell-based xenografts, patient-derived explants, organoids, and xenograft tumors, we demonstrated that EC359 dramatically reduced the growth and progression of OCa. Additionally, EC359 therapy considerably improved tumor immunogenicity by robust CD45+ leukocyte tumor infiltration and polarizing tumor-associated macrophages (TAMs) toward M1 phenotype while showing no impact on normal T-, B-, and other immune cells. Collectively, our findings indicate that the LIF/LIFR autocrine loop plays an essential role in OCa progression and that EC359 could be a promising therapeutic agent for OCa.

6.
Cancers (Basel) ; 16(3)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38339252

ABSTRACT

Ovarian cancer (OCa) is the most lethal form of gynecologic cancer, and the tumor heterogeneities at the molecular, cellular, and tissue levels fuel tumor resistance to standard therapies and pose a substantial clinical challenge. Here, we tested the hypothesis that the heightened basal endoplasmic reticulum stress (ERS) observed in OCa represents an exploitable vulnerability and may overcome tumor heterogeneity. Our recent studies identified LIPA as a novel target to induce ERS in cancer cells using the small molecule ERX-41. However, the role of LIPA and theutility of ERX-41 to treat OCa remain unknown. Expression analysis using the TNMplot web tool, TCGA data sets, and immunohistochemistry analysis using a tumor tissue array showed that LIPA is highly expressed in OCa tissues, compared to normal tissues. ERX-41 treatment significantly reduced the cell viability and colony formation ability and promoted the apoptosis of OCa cells. Mechanistic studies revealed a robust and consistent induction of ERS markers, including CHOP, elF2α, PERK, and ATF4, upon ERX-41 treatment. In xenograft and PDX studies, ERX-41 treatment resulted in a significant reduction in tumor growth. Collectively, our results suggest that ERX-41 is a novel therapeutic agent that targets the LIPA with a unique mechanism of ERS induction, which could be exploited to treat heterogeneity in OCa.

7.
Mol Oncol ; 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37853941

ABSTRACT

Endometrial carcinoma (ECa) is the fourth most common cancer among women. The oncogene PELP1 is frequently overexpressed in a variety of cancers, including ECa. We recently generated SMIP34, a small-molecule inhibitor of PELP1 that suppresses PELP1 oncogenic signaling. In this study, we assessed the effectiveness of SMIP34 in treating ECa. Treatment of established and primary patient-derived ECa cells with SMIP34 resulted in a significant reduction of cell viability, colony formation ability, and induction of apoptosis. RNA-seq analyses showed that SMIP34-regulated genes were negatively correlated with ribosome biogenesis and eukaryotic translation pathways. Mechanistic studies showed that the Rix complex, which is essential for ribosomal biogenesis, is disrupted upon SMIP34 binding to PELP1. Biochemical assays confirmed that SMIP34 reduced ribosomal biogenesis and new protein synthesis. Further, SMIP34 enhanced the efficacy of mTOR inhibitors in reducing viability of ECa cells. SMIP34 is also effective in reducing cell viability in ECa organoids in vitro and explants ex vivo. Importantly, SMIP34 treatment resulted in a significant reduction of the growth of ECa xenografts. Collectively, these findings underscore the potential of SMIP34 in treating ECa.

8.
Cancer Lett ; 575: 216383, 2023 10 28.
Article in English | MEDLINE | ID: mdl-37714256

ABSTRACT

Ovarian cancer (OCa) is the most lethal gynecologic cancer. Emerging data indicates that estrogen receptor beta (ERß) functions as a tumor suppressor in OCa. Lysine-specific histone demethylase 1A (KDM1A) is an epigenetic modifier that acts as a coregulator for steroid hormone receptors. However, it remain unknown if KDM1A interacts with ERß and regulates its expression/functions in OCa. Analysis of TCGA data sets indicated KDM1A and ERß expression showed an inverse relationship in OCa. Knockout (KO), knockdown (KD), or inhibition of KDM1A increased ERß isoform 1 expression in established and patient-derived OCa cells. Further, KDM1A interacts with and functions as a corepressor of ERß, and its inhibition enhances ERß target gene expression via alterations of histone methylation marks at their promoters. Importantly, KDM1A-KO or -KD enhanced the efficacy of ERß agonist LY500307, and the combination of KDM1A inhibitor (KDM1Ai) NCD38 with ERß agonist synergistically reduced the cell viability, colony formation, and invasion of OCa cells. RNA-seq and DIA mass spectrometry analyses showed that KDM1A-KO resulted in enhanced ERß signaling and that genes altered by KDM1A-KO and ERß agonist were related to apoptosis, cell cycle, and EMT. Moreover, combination treatment significantly reduced the tumor growth in OCa orthotopic, syngeneic, and patient-derived xenograft models and proliferation in patient-derived explant models. Our results demonstrate that KDM1A regulates ERß expression/functions, and its inhibition improves ERß mediated tumor suppression. Overall, our findings suggest that KDM1Ai and ERß agonist combination therapy is a promising strategy for OCa.


Subject(s)
Estrogen Receptor beta , Ovarian Neoplasms , Humans , Female , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Cell Line, Tumor , Genes, Tumor Suppressor , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Estrogens , Histone Demethylases
9.
Cancer Lett ; 524: 219-231, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34673129

ABSTRACT

Endometrial cancer (EC) often exhibit aberrant activation of PI3K/Akt/mTOR signaling and targeted therapies using mTOR inhibitors showed limited success. The epigenetic modifier, lysine-specific histone demethylase-1A (KDM1A/LSD1) is overexpressed in EC, however, the mechanistic and therapeutic implications of KDM1A in EC are poorly understood. Here, using 119 FDA-approved drugs screen, we identified that KDM1A inhibition is highly synergistic with mTOR inhibitors. Combination therapy of KDM1A and mTOR inhibitors potently reduced the cell viability, survival, and migration of EC cells. Mechanistic studies demonstrated that KDM1A inhibition attenuated the activation of mTOR signaling cascade and abolished rapamycin induced feedback activation of Akt. RNA-seq analysis identified that KDM1A inhibition downregulated the expression of genes involved in rapamycin induced activation of Akt, including the mTORC2 complex. Chromatin immunoprecipitation experiments confirmed KDM1A recruitment to the promoter regions of mTORC2 complex genes and that KDM1A inhibition promoted enrichment of repressive H3K9me2 marks at their promoters. Combination therapy of KDM1A inhibitor and rapamycin reduced the tumor growth in EC xenograft and patient derived xenograft models in vivo and patient derived tumor explants ex vivo. Importantly, in silico analysis of TCGA EC patients data sets revealed that KDM1A expression positively correlated with the levels of PI3K/Akt/mTOR genes. Collectively, our results provide compelling evidence that KDM1A inhibition potentiates the activity of mTOR inhibitors by attenuating the feedback activation of Akt survival signaling. Furthermore, the use of concurrent KDM1A and mTOR inhibitors may be an attractive targeted therapy for EC patients.


Subject(s)
Endometrial Neoplasms/drug therapy , Histone Demethylases/genetics , MTOR Inhibitors/pharmacology , TOR Serine-Threonine Kinases/genetics , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Demethylases/antagonists & inhibitors , Humans , MTOR Inhibitors/chemistry , Male , Mice , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
10.
Cancers (Basel) ; 14(21)2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36358818

ABSTRACT

Endometrial cancer (EC) is the fourth most common cancer in women, and half of the endometrioid EC (EEC) cases are attributable to obesity. However, the underlying mechanism(s) of obesity-driven EEC remain(s) unclear. In this study, we examined whether LIF signaling plays a role in the obesity-driven progression of EEC. RNA-seq analysis of EEC cells stimulated by adipose conditioned medium (ADP-CM) showed upregulation of LIF/LIFR-mediated signaling pathways including JAK/STAT and interleukin pathways. Immunohistochemistry analysis of normal and EEC tissues collected from obese patients revealed that LIF expression is upregulated in EEC tissues compared to the normal endometrium. Treatment of both primary and established EEC cells with ADP-CM increased the expression of LIF and its receptor LIFR and enhanced proliferation of EEC cells. Treatment of EEC cells with the LIFR inhibitor EC359 abolished ADP-CM induced colony formation andcell viability and decreased growth of EEC organoids. Mechanistic studies using Western blotting, RT-qPCR and reporter assays confirmed that ADP-CM activated LIF/LIFR downstream signaling, which can be effectively attenuated by the addition of EC359. In xenograft assays, co-implantation of adipocytes significantly enhanced EEC xenograft tumor growth. Further, treatment with EC359 significantly attenuated adipocyte-induced EEC progression in vivo. Collectively, our data support the premise that LIF/LIFR signaling plays an important role in obesity-driven EEC progression and the LIFR inhibitor EC359 has the potential to suppress adipocyte-driven tumor progression.

11.
Am J Obstet Gynecol ; 205(4): 356.e1-5, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21689805

ABSTRACT

OBJECTIVE: The purpose of this study was to compare operative outcomes and complications for patients with endometrial cancer who underwent staging by laparoscopy vs laparotomy in a low-volume facility. STUDY DESIGN: Research was conducted with a retrospective cohort of surgical patients with clinical stage I endometrial cancer from 2004-2009. RESULTS: Eighty-six demographically similar patients (50 laparotomy and 36 laparoscopy) were identified. Laparoscopy had less estimated blood loss (339 vs 558 mL; P = .013) and lower rates of transfusion (5.6% vs 24%; P = .02). Laparoscopy was longer (281 vs 202 minutes; P < .0005) but required a shorter hospital stay (2.2 vs 5.5 days; P < .0005). Laparoscopy patients had fewer overall complications (16.7% vs 32%; P = .11). No differences in final surgical stage or lymph node yields between the groups were present. CONCLUSION: Although a longer procedure, laparoscopy had fewer complications and shorter hospital stays. Prolonged operative time, compared with published experience, is potentially the result of unique factors in our center.


Subject(s)
Endometrial Neoplasms/surgery , Laparoscopy , Laparotomy , Adult , Aged , Aged, 80 and over , Endometrial Neoplasms/pathology , Female , Humans , Internship and Residency , Laparoscopy/adverse effects , Laparotomy/adverse effects , Middle Aged , Military Medicine , Neoplasm Staging , Postoperative Complications/epidemiology , Retrospective Studies
12.
Gynecol Oncol Rep ; 36: 100772, 2021 May.
Article in English | MEDLINE | ID: mdl-34026998

ABSTRACT

•Rectovaginal septum mass in BRCA1 positive patient after risk reducing BSO years prior.•Papillary serous carcinoma presenting as a rectovaginal septum mass.•PAOLA-1 trial discussion for rectovaginal septum mass in BRCA1 positive patient.

13.
Cell Death Discov ; 7(1): 216, 2021 Aug 16.
Article in English | MEDLINE | ID: mdl-34400617

ABSTRACT

Endometrial cancer (EC) is the fourth most common cancer in women. Advanced-stage EC has limited treatment options with a poor prognosis. There is an unmet need for the identification of actionable drivers for the development of targeted therapies in EC. Leukemia inhibitory factor receptor (LIFR) and its ligand LIF play a major role in cancer progression, metastasis, stemness, and therapy resistance. However, little is known about the functional significance of the LIF/LIFR axis in EC progression. In this study using endometrial tumor tissue arrays, we identified that expression of LIF, LIFR is upregulated in EC. Knockout of LIFR using CRISPR/Cas9 in two different EC cells resulted in a significant reduction of their cell viability and cell survival. In vivo studies demonstrated that LIFR-KO significantly reduced EC xenograft tumor growth. Treatment of established and primary patient-derived EC cells with a novel LIFR inhibitor, EC359 resulted in the reduction of cell viability with an IC50 in the range of 20-100 nM and induction of apoptosis. Further, treatment with EC359 reduced the spheroid formation of EC cancer stem cells and reduced the levels of cancer stem cell markers SOX2, OCT4, NANOG, and Axin2. Mechanistic studies demonstrated that EC359 treatment attenuated the activation of LIF-LIFR driven pathways, including STAT3 and AKT/mTOR signaling in EC cells. Importantly, EC359 treatment resulted in a significant reduction of the growth of EC patient-derived explants ex vivo, EC cell line-derived xenografts, and patient-derived xenografts in vivo. Collectively, our work revealed the oncogenic potential of the LIF/LIFR axis in EC and support the utility of LIFR inhibitor, EC359, as a novel targeted therapy for EC via the inhibition of LIF/LIFR oncogenic signaling.

14.
Int Urogynecol J ; 21(1): 125-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19533004

ABSTRACT

We report on a case of a suburethral erosion of transobturator tape tracking into the right thigh with abscess formation 20 months from surgery resulting in necrotizing fasciitis. A 64-year-old woman presented with new onset of severe right thigh pain. Physical exam was concerning for a right thigh abscess and she was admitted to general surgery for evaluation. Gynecology was consulted secondary to a complaint of foul vaginal discharge. The patient was noted to have a vaginal mass on the anterior vaginal wall. A computed tomography scan showed a large right medial thigh abscess. The patient was taken for emergency exploratory surgery where an ObTape (Mentor, Santa Barbara, CA, USA) TOT was found eroding through the anterior vaginal wall, extending into the thigh abscess. During removal of the ObTape from her thigh, the entire gracilis muscle was removed due to necrotizing fasciitis and extensive tissue debridement was performed. A wound vac was placed and the patient had an uneventful recovery.


Subject(s)
Fasciitis, Necrotizing/etiology , Foreign-Body Migration/complications , Polypropylenes , Suburethral Slings/adverse effects , Device Removal , Fasciitis, Necrotizing/diagnosis , Female , Foreign-Body Migration/diagnosis , Humans , Middle Aged , Thigh/diagnostic imaging , Thigh/surgery , Tomography, X-Ray Computed , Treatment Outcome , Vaginal Discharge/etiology
15.
Gynecol Oncol Rep ; 34: 100635, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32984494

ABSTRACT

•A unique initial presentation of GTN as pulmonary arteriovenous malformations.•Metastatic GTN presenting as multiple visceral AVMs in the brain, liver, and lungs.•Management of metastatic GTN with brain metastases with induction chemotherapy.

16.
Cell Rep ; 33(2): 108253, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053339

ABSTRACT

While plasminogen activator inhibitor-1 (PAI-1) is known to potentiate cellular migration via proteolytic regulation, this adipokine is implicated as an oncogenic ligand in the tumor microenvironment. To understand the underlying paracrine mechanism, here, we conduct transcriptomic analysis of 1,898 endometrial epithelial cells (EECs) exposed and unexposed to PAI-1-secreting adipose stromal cells. The PAI-1-dependent action deregulates crosstalk among tumor-promoting and tumor-repressing pathways, including transforming growth factor ß (TGF-ß). When PAI-1 is tethered to lipoprotein receptor-related protein 1 (LRP1), the internalized signaling causes downregulation of SMAD4 at the transcriptional and post-translational levels that attenuates TGF-ß-related transcription programs. Repression of genes encoding the junction and adhesion complex preferentially occurs in SMAD4-underexpressed EECs of persons with obesity. The findings highlight a role of PAI-1 signaling that renders ineffective intercellular communication for the development of adiposity-associated endometrial cancer.


Subject(s)
Endometrial Neoplasms/metabolism , Junctional Adhesion Molecules/metabolism , Obesity/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Smad4 Protein/metabolism , Adipose Tissue/pathology , Down-Regulation/genetics , Endometrial Neoplasms/complications , Endometrial Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Obesity/complications , Protein Binding , Proteolysis , Proteomics , Smad4 Protein/genetics , Stromal Cells/metabolism , Transcription, Genetic , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Ubiquitin/metabolism
17.
Cancer Res ; 79(1): 196-208, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30389702

ABSTRACT

Emerging evidence indicates that adipose stromal cells (ASC) are recruited to enhance cancer development. In this study, we examined the role these adipocyte progenitors play relating to intercellular communication in obesity-associated endometrial cancer. This is particularly relevant given that gap junctions have been implicated in tumor suppression. Examining the effects of ASCs on the transcriptome of endometrial epithelial cells (EEC) in an in vitro coculture system revealed transcriptional repression of GJA1 (encoding the gap junction protein Cx43) and other genes related to intercellular communication. This repression was recapitulated in an obesity mouse model of endometrial cancer. Furthermore, inhibition of plasminogen activator inhibitor 1 (PAI-1), which was the most abundant ASC adipokine, led to reversal of cellular distribution associated with the GJA1 repression profile, suggesting that PAI-1 may mediate actions of ASC on transcriptional regulation in EEC. In an endometrial cancer cohort (n = 141), DNA hypermethylation of GJA1 and related loci TJP2 and PRKCA was observed in primary endometrial endometrioid tumors and was associated with obesity. Pharmacologic reversal of DNA methylation enhanced gap-junction intercellular communication and cell-cell interactions in vitro. Restoring Cx43 expression in endometrial cancer cells reduced cellular migration; conversely, depletion of Cx43 increased cell migration in immortalized normal EEC. Our data suggest that persistent repression by ASC adipokines leads to promoter hypermethylation of GJA1 and related genes in the endometrium, triggering long-term silencing of these loci in endometrial tumors of obese patients. SIGNIFICANCE: Studies reveal that adipose-derived stem cells in endometrial cancer pathogenesis influence epigenetic repression of gap junction loci, which suggests targeting of gap junction activity as a preventive strategy for obesity-associated endometrial cancer.


Subject(s)
Adipokines/pharmacology , Adipose Tissue/pathology , Cell Communication , Connexin 43/genetics , Endometrial Neoplasms/pathology , Epigenetic Repression , Obesity/complications , Adipose Tissue/metabolism , Animals , Cell Movement , Cells, Cultured , Connexin 43/metabolism , Diet, High-Fat/adverse effects , Endometrial Neoplasms/etiology , Endometrial Neoplasms/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gap Junctions , Humans , Male , Mice , Mice, Knockout , Obesity/physiopathology , Stromal Cells/metabolism , Stromal Cells/pathology
18.
Obstet Gynecol ; 109(2 Pt2): 507-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17267875

ABSTRACT

BACKGROUND: Extremity compartment syndrome is a rare surgical complication in which increased intracompartmental pressure threatens the viability of the muscular and nervous tissue. We report a case of a delivery complicated by postpartum hemorrhage and a lower extremity compartment syndrome. CASE: A multigravida in her late 20s was admitted for induction of labor at 41 weeks of gestation. She underwent a cesarean delivery for a nonreassuring fetal heart rate tracing. The delivery was complicated by severe postpartum hemorrhage due to uterine atony, resulting in a cesarean hysterectomy. The patient developed a lower extremity compartment syndrome and underwent an emergent anterior tibial fasciotomy. CONCLUSION: Extremity compartment syndrome is rarely encountered in obstetric care; however, there may be an association with severe postpartum hemorrhage.


Subject(s)
Compartment Syndromes/diagnosis , Hysterectomy/methods , Postoperative Complications/diagnosis , Postpartum Hemorrhage/surgery , Adult , Cesarean Section , Compartment Syndromes/surgery , Diagnosis, Differential , Female , Hemostatic Techniques , Humans , Labor, Induced , Leg/pathology , Postoperative Complications/surgery , Pregnancy , Pregnancy Trimester, Third
19.
Oncotarget ; 8(30): 50002-50014, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28654894

ABSTRACT

Ovarian cancer is the deadliest of all gynecologic cancers. Despite success with initial chemotherapy, the majority of patients relapse with an incurable disease. Development of chemotherapy resistance is a major factor for poor long-term survival in ovarian cancer. The biological effects of estrogens are mediated by estrogen receptor alpha (ERα) and estrogen receptor beta (ERß). Emerging evidence suggests that ovarian cancer cells express ERß that functions as a tumor suppressor; however, the clinical utility of ERß agonists in ovarian cancer remains elusive. We tested the utility of two natural ERß agonists liquiritigenin (Liq), which is isolated from Glycyrrhiza uralensis and S-equol, which is isolated from soy isoflavone daidzein, for treating ovarian cancer. Both natural ERß ligands had significant growth inhibition in cell viability and survival assays, reduced migration and invasion, and promoted apoptosis. Further, ERß agonists showed tumor suppressive functions in therapy-resistant ovarian cancer model cells and sensitized ovarian cancer cells to cisplatin and paclitaxel treatment. Global RNA-Seq analysis revealed that ERß agonists modulate several tumor suppressive pathways, including downregulation of the NF-κB pathway. Immunoprecipitation assays revealed that ERß interacts with p65 subunit of NF-κB and ERß overexpression reduced the expression of NF-κB target genes. In xenograft assays, ERß agonists reduced tumor growth and promoted apoptosis. Collectively, our findings demonstrated that natural ERß agonists have the potential to significantly inhibit ovarian cancer cell growth by anti-inflammatory and pro-apoptotic actions, and natural ERß agonists represent novel therapeutic agents for the management of ovarian cancer.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Biological Products/pharmacology , Estrogen Receptor beta/agonists , Estrogens/pharmacology , Ovarian Neoplasms/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , NF-kappa B/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
20.
Gynecol Oncol Rep ; 17: 45-8, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27355001

ABSTRACT

•Management of cervical leiomyosarcoma in pregnancy requires a multidisciplinary approach.•Ovarian preservation is preferred in young patients with early stage cervical leiomyosarcoma.•Routine lymphadenectomy in patients with early stage cervical leiomyosarcoma is not useful.

SELECTION OF CITATIONS
SEARCH DETAIL