Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters

Publication year range
1.
BMC Cancer ; 22(1): 387, 2022 Apr 10.
Article in English | MEDLINE | ID: mdl-35399074

ABSTRACT

BACKGROUND: Abnormal accumulation of acyl-CoA cholesterol acyltransferase-1 (ACAT1) and ACAT1-mediated cholesterol esterified with fatty acids (CE) contribute to cancer progression in various cancers. Our findings of increased CE and ACAT1 levels in epithelial ovarian cancer (EOC) cell lines prompted us to investigate whether such an increase occurs in primary clinical samples obtained from human subjects diagnosed with EOC. We evaluated the diagnostic/prognostic potential of ACAT1 and CE in EOC by: 1) assessing ACAT1 and CE levels in plasma, peritoneal fluid, and ovarian/tumor tissues; 2) assessing diagnostic performance by Receiver Operating Characteristic (ROC) analysis; and 3) comparing expression of ACAT1 and CE with that of tumor proliferation marker, Ki67. METHODS: ACAT1 protein levels in plasma, peritoneal fluid and tissue were measured via enzyme-linked immunosorbent assay. Tissue expression of ACAT1 and Ki67 proteins were confirmed by immunohistochemistry and mRNA transcript levels were evaluated using quantitative real-time polymerase chain reaction (qRT-PCR). CE levels were assessed in plasma, peritoneal fluid (colorimetric assay) and in tissue (thin layer chromatography). RESULTS: Preoperative levels of ACAT1 and CE on the day of surgery were significantly higher in tissue and peritoneal fluid from EOC patients vs. the non-malignant group, which included subjects with benign tumors and normal ovaries; however, no significant differences were observed in plasma. In tissue and peritoneal fluid, positive correlations were observed between CE and ACAT1 levels, as well as between ACAT1/CE and Ki67. CONCLUSIONS: ACAT1 and CE accumulation may be linked to the aggressive potential of EOC; therefore, these mediators may be useful biomarkers for EOC prognosis and target-specific treatments.


Subject(s)
Acetyl-CoA C-Acetyltransferase , Carcinoma, Ovarian Epithelial , Ovarian Neoplasms , Acetyl-CoA C-Acetyltransferase/genetics , Acyltransferases , Ascitic Fluid/pathology , Biomarkers, Tumor/genetics , Carcinoma, Ovarian Epithelial/diagnosis , Carcinoma, Ovarian Epithelial/genetics , Cholesterol , Fatty Acids , Female , Humans , Ki-67 Antigen , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Pilot Projects , Prognosis
2.
J Low Genit Tract Dis ; 25(2): 106-112, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33631781

ABSTRACT

OBJECTIVES: The aims of the study were to identify unmet basic needs (BNs) among women referred to colposcopy, to assess patient acceptability/satisfaction with assistance from a navigator to address unmet BNs, and to estimate adherence to colposcopy. METHODS: Women were recruited between September 2017 and January 2019 from 2 academic colposcopy centers, one serving a rural and one an urban area. Basic needs were assessed by phone before colposcopy appointments and considered unmet if unlikely to resolve in 1 month. Colposcopy adherence prestudy and poststudy implementation was abstracted over 4-6 months from administrative records. After a lead-in phase of 25 patients at each site, a BN navigator was offered to new participants with 1 or more unmet BNs. Primary outcome was adherence to initial appointment. RESULTS: Among 100 women, 59% had 1 or more unmet BNs, with similar prevalence between urban and rural sites. Adherence to initial colposcopy was 83% overall, 72% at the rural clinic, and 94% at the urban clinic (p = .006). These adherence rates were improved from 4 months before study launch (30/59 [51%] rural clinic and 68/137 [50%] urban clinic). Although acceptability of BN navigation was greater than 96% and women felt that it helped them get to their colposcopy visit, having a navigator was not associated with adherence. Women reporting no unmet BNs had the lowest adherence compared with women with 1 or more unmet BNs, regardless of navigator assistance (p = .03). CONCLUSIONS: Disadvantaged women who need colposcopy have unmet BNs and value navigator assistance for initial appointments. However, when appointment scheduling includes telephone reminders and inquiring about BNs, a navigator may not add value.


Subject(s)
Colposcopy/psychology , Colposcopy/statistics & numerical data , Health Knowledge, Attitudes, Practice , Patient Acceptance of Health Care/psychology , Patient Acceptance of Health Care/statistics & numerical data , Uterine Cervical Neoplasms/psychology , Adult , Cohort Studies , Early Detection of Cancer/methods , Early Detection of Cancer/psychology , Female , Health Services Accessibility , Humans , Middle Aged , Papanicolaou Test , Patient Compliance/psychology , Patient Compliance/statistics & numerical data , Pilot Projects , Professional-Patient Relations , Prospective Studies , Rural Population/statistics & numerical data , Urban Population/statistics & numerical data , Uterine Cervical Neoplasms/diagnosis , Vaginal Smears
3.
Biol Reprod ; 102(5): 1011-1019, 2020 04 24.
Article in English | MEDLINE | ID: mdl-31950153

ABSTRACT

Endometriosis is a common gynecological disease, which causes chronic pelvic pain and infertility in women of reproductive age. Due to limited efficacy of current treatment options, a critical need exists to develop new and effective treatments for endometriosis. Niclosamide is an efficacious and FDA-approved drug for the treatment of helminthosis in humans that has been used for decades. We have reported that niclosamide reduces growth and progression of endometriosis-like lesions via targeting STAT3 and NFĸB signaling in a mouse model of endometriosis. To examine the effects of niclosamide on macrophage-induced inflammation in endometriosis, a total of 29 stage III-IV endometrioma samples were used to isolate human endometriotic stromal cells (hESCs). M1 or M2 macrophages were isolated and differentiated from fresh human peripheral blood samples. Then, hESCs were cultured in conditioned media (CM) from macrophages with/without niclosamide. Niclosamide dose dependently reduced cell viability and the activity of STAT3 and NFκB signaling in hESCs. While macrophage CM stimulated cell viability in hESCs, niclosamide inhibited this stimulation. Macrophage CM stimulated the secretion of proinflammatory cytokines and chemokines from hESCs. Most of these secreted factors were inhibited by niclosamide. These results indicate that niclosamide is able to reduce macrophage-induced cell viability and cytokine/chemokine secretion in hESCs by inhibiting inflammatory mechanisms via STAT3 and/or NFκB signaling.


Subject(s)
Cell Survival/drug effects , Endometriosis/pathology , Macrophages/drug effects , Macrophages/metabolism , Niclosamide/pharmacology , Animals , Anticestodal Agents/pharmacology , Cells, Cultured , Endometriosis/drug therapy , Female , Gene Expression Regulation/drug effects , Humans , Inflammation/drug therapy , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Stromal Cells
4.
J Minim Invasive Gynecol ; 23(5): 798-803, 2016.
Article in English | MEDLINE | ID: mdl-27103374

ABSTRACT

STUDY OBJECTIVE: To determine whether the location of the superior and inferior epigastric vessels (deep epigastric vessels) change with abdominal insufflation. DESIGN: Descriptive study (Canadian Task Force classification III). SETTING: Tertiary care academic institution. PATIENTS: Patients undergoing gynecologic laparoscopic surgery were recruited. A total of 35 subjects were enrolled. INTERVENTIONS: Subjects underwent color Doppler ultrasound assessment of deep epigastric vessel location preoperatively and intraoperatively following abdominal insufflation. The deep epigastric vessels were identified at 5 points along the abdomen (pubic symphysis, anterior superior iliac spine [ASIS], umbilicus, xiphoid, and midpoint from umbilicus to xiphoid), with the distance from vessels to midline measured. Paired t tests and split-plot analysis of variance were used as appropriate. MEASUREMENTS AND MAIN RESULTS: The mean patient age was 45.6 ± 16.5 years, and mean BMI was 29.8 ± 7.2. A significant difference between vessel location in the resting abdomen and insufflated abdomen was noted bilaterally at the ASIS, umbilicus, and midpoint from the umbilicus to the xiphoid. At each of these points, the deep epigastric vessels were found more laterally after insufflation on average, ranging from 0.6 ± 0.9 cm (p < .001) more laterally at the midpoint between the umbilicus and xiphoid to 1.1 ± 0.8 cm (p < .001) more laterally at the umbilicus. The most lateral location of the deep vessels after insufflation was seen at the ASIS (10.6 cm) and the umbilicus (10.9 cm). In a subanalysis of subjects grouped by body mass index (obese vs nonobese), deep epigastric vessels were more lateral in the insufflated abdomen of obese subjects compared with that of nonobese subjects at the ASIS, umbilicus, and midpoint from umbilicus to xiphoid (p < .05 for each point bilaterally). CONCLUSION: The deep epigastric vessels shift laterally with abdominal insufflation, and may be found as far as 10.9 cm from the midline; this is more lateral than previously described and is clinically significant. Obesity is associated with a more lateral location of the deep epigastric vessels.


Subject(s)
Epigastric Arteries/anatomy & histology , Insufflation , Abdomen , Abdominal Cavity/blood supply , Adult , Aged , Body Mass Index , Epigastric Arteries/diagnostic imaging , Female , Gynecologic Surgical Procedures , Humans , Laparoscopy , Middle Aged , Obesity/pathology , Ultrasonography, Doppler, Color
5.
J Reprod Immunol ; 163: 104241, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38492533

ABSTRACT

Epithelial ovarian cancer (OC) is the deadliest female reproductive cancer; an estimated 13,270 women will die from OC in 2023. Platinum-based chemotherapy resistance mechanisms contribute to poor OC 5-year survival rates. Peripheral inflammation is linked to various disease states and we previously identified unique peritoneal microbial features predictive of OC. We hypothesized that unique peripheral immune profiles and peritoneal microbial features may be predictive of disease-free interval (time to recurrence) and response to chemotherapy in participants with OC. We also investigated self-rated health (SRH) scores in the context of peripheral inflammation as a potential screening tool for OC. Blood and peritoneal fluid were collected from participants with OC or a benign adnexal mass (BPM). Lymphocyte populations were analyzed using Fluorescence Activated Cell Sorting, serum cytokine levels were analyzed using the Human Th17 Magnetic Bead Panel assay and peritoneal fluid microbial features were analyzed using Next Generation Sequencing (NGS). Participants completed a standardized questionnaire on self-rated physical and emotional health. Participants were classified into three chemotherapy response categories: platinum-refractory, platinum-resistant or platinum-sensitive. A significant positive correlation was found between elevated inflammatory status on the day of surgery and longer disease-free interval. SRH measures did not correlate with immune status in participants with OC or a BPM. We identified a correlation between peritoneal microbial features and chemotherapy response. We conclude that immune dysbiosis may be useful in predicting OC recurrence. The immune findings reported here set the framework for additional studies utilizing immune profiles to predict platinum-based chemotherapy responsiveness in OC.


Subject(s)
Dysbiosis , Humans , Female , Middle Aged , Dysbiosis/immunology , Adult , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/drug therapy , Aged , Ovarian Neoplasms/immunology , Ovarian Neoplasms/drug therapy , Drug Resistance, Neoplasm/immunology , Prognosis , Microbiota/immunology , Microbiota/drug effects , Cytokines/metabolism , Cytokines/blood , Ascitic Fluid/immunology , Ascitic Fluid/microbiology
6.
Oncol Lett ; 25(6): 231, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37153054

ABSTRACT

Endometrial cancer (EC) is the most prevalent gynecological malignancy. Abnormal accumulation of sterol-O-acyl transferase 1 (SOAT1) and SOAT1-mediated cholesterol ester (CE) contributes to cancer progression in various malignancies, including ovarian cancer. Therefore, it was hypothesized that similar molecular changes may occur in EC. The present study aimed to evaluate the diagnostic and/or prognostic potential of SOAT1 and CE in EC by: i) Determining SOAT1 and CE levels in plasma, peritoneal fluid and endometrial tissue from patients with EC and control subjects; ii) performing receiver operating characteristic curve analysis to determine diagnostic performance; iii) comparing SOAT1 and CE expression to that of the tumor proliferation marker Ki67; and iv) assessing the association between SOAT1 expression and survival. Enzyme-linked immunosorbent assay was used to determine the levels of SOAT1 protein in tissue, plasma and peritoneal fluid. The mRNA and protein expression levels of SOAT1 and Ki67 in tissues were detected by reverse transcription-quantitative polymerase chain reaction and immunohistochemistry, respectively. CE levels were determined colorimetrically in plasma and peritoneal fluid. SOAT1-associated survival data from the cBioPortal cancer genomics database were used to assess prognostic relevance. The results revealed that SOAT1 and CE levels were significantly elevated in tumor tissue and peritoneal fluid samples collected from the EC group. By contrast, the plasma levels of SOAT1 and CE in the EC and control groups were similar. Significant positive associations between CE and SOAT1, SOAT1/CE and Ki67, and SOAT1/CE and poor overall survival in patients with EC suggested that SOAT1/CE may be associated with malignancy, aggressiveness and poor prognosis. In conclusion, SOAT1 and CE may serve as potential biomarkers for prognosis and target-specific treatment of EC.

7.
Life (Basel) ; 12(4)2022 Apr 09.
Article in English | MEDLINE | ID: mdl-35455053

ABSTRACT

Endometrial carcinoma (EC) is the most common type of gynecologic malignant epithelial tumor, with the death rate from this disease doubling over the past 20 years. Mitochondria provide cancer cells with necessary anabolic building blocks such as amino acids, lipids, and nucleotides, and EC samples have been shown to increase mitochondrial biogenesis. In cancer, mitochondrial DNA (mtDNA) heteroplasmy studies suggest that heteroplasmic variants encode predicted pathogenic proteins. We investigated the mtDNA genotypes within peri-normal and tumor specimens obtained from three individuals diagnosed with EC. DNA extracts from peri-normal and tumor tissues were used for mtDNA-specific next-generation sequencing and analyses of mtDNA content and topoisomers. The three tumors harbor heteroplasmic somatic mutations, and at least one mutation in each carcinoma is predicted to deleteriously alter a mtDNA-encoded protein. Somatic heteroplasmy linked to two mtDNA tRNA genes was found in separate tumors, and two heteroplasmic non-coding variants were identified in a single EC tumor. While two tumors had altered mtDNA content, all three displayed increased mtDNA catenanes. Our findings support that EC cells require wild-type mtDNA, but heteroplasmic mutations may alter mitochondrial metabolism to help promote cancer cell growth and proliferation.

8.
Am J Reprod Immunol ; 85(3): e13362, 2021 03.
Article in English | MEDLINE | ID: mdl-33070438

ABSTRACT

PROBLEM: Endometriosis is defined as growth of endometrial tissue in ectopic locations; it is associated with infertility and chronic pain and affects ~12% of reproductive-aged women. Although inflammation is known to play a key role in endometriosis, knowledge related to immune phenotypes associated with this disease is lacking. This study aimed to characterize immune profiles in patients with endometriosis, to assess inflammatory mediators, to and determine if surgical and/or hormonal therapies restore immune homeostasis. METHODS OF STUDY: Samples from nine controls and 20 histologically confirmed endometriosis patients were collected upon surgery and ~1-3 weeks post-surgical intervention. Subjects were either not utilizing hormonal suppression or were currently on monophasic hormonal therapy. Tolerant regulatory T cells (Tregs = natural [nTregs] +inducible [iTregs]) and inflammatory T helper 17 (Th17) cells were identified in peripheral blood via flow cytometry and within the eutopic/ectopic endometrial tissues via immunohistochemistry and real-time-qPCR. Cytokines were assessed via 10-plex-ELISA. RESULTS: Patients with endometriosis not utilizing hormonal therapy exhibited lower iTregs (tolerant), greater Th17 (inflammatory), and a reduction in Treg/Th17 ratio (P < .05), indicative of systemic inflammation. Treg and Th17 localizations were enhanced within the ectopic endometrial implant, which promotes lesion development. Hormonal therapy decreased systemic and local inflammation (eutopic/ectopic endometrium) via decreased iTregs and Th17 cells in patients with endometriosis (P < .05). Thus, imbalance within immune populations correlated with increased inflammation in patients with endometriosis, which was mitigated by hormonal therapy. CONCLUSIONS: Patients with endometriosis exhibited systemic and localized inflammation within ectopic and endometrial tissues. Hormonal therapy dampened inflammation caused by disease.


Subject(s)
Endometriosis/immunology , Hormones/therapeutic use , Inflammation/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Uterus/immunology , Adult , Endometriosis/drug therapy , Female , Humans , Immune Tolerance , Immunity, Cellular , Inflammation/drug therapy , Phenotype
9.
PLoS One ; 16(12): e0261362, 2021.
Article in English | MEDLINE | ID: mdl-34914785

ABSTRACT

Endometriosis is an estrogen dependent gynecological disease associated with altered microbial phenotypes. The association among endogenous estrogen, estrogen metabolites, and microbial dynamics on disease pathogenesis has not been fully investigated. Here, we identified estrogen metabolites as well as microbial phenotypes in non-diseased patients (n = 9) and those with pathologically confirmed endometriosis (P-EOSIS, n = 20), on day of surgery (DOS) and ~1-3 weeks post-surgical intervention (PSI). Then, we examined the effects of surgical intervention with or without hormonal therapy (OCPs) on estrogen and microbial profiles of both study groups. For estrogen metabolism analysis, liquid chromatography/tandem mass spectrometry was used to quantify urinary estrogens. The microbiome data assessment was performed with Next generation sequencing to V4 region of 16S rRNA. Surgical intervention and hormonal therapy altered gastrointestinal (GI), urogenital (UG) microbiomes, urinary estrogen and estrogen metabolite levels in P-EOSIS. At DOS, 17ß-estradiol was enhanced in P-EOSIS treated with OCPs. At PSI, 16-keto-17ß-estradiol was increased in P-EOSIS not receiving OCPs while 2-hydroxyestradiol and 2-hydroxyestrone were decreased in P-EOSIS receiving OCPs. GI bacterial α-diversity was greater for controls and P-EOSIS that did not receive OCPs. P-EOSIS not utilizing OCPs exhibited a decrease in UG bacterial α-diversity and differences in dominant taxa, while P-EOSIS utilizing OCPs had an increase in UG bacterial α-diversity. P-EOSIS had a strong positive correlation between the GI/UG bacteria species and the concentrations of urinary estrogen and its metabolites. These results indicate an association between microbial dysbiosis and altered urinary estrogens in P-EOSIS, which may impact disease progression.


Subject(s)
Endometriosis/microbiology , Estrogens/urine , Adult , Chromatography, Liquid/methods , Dysbiosis/metabolism , Dysbiosis/urine , Endometriosis/urine , Estradiol/analogs & derivatives , Estrogens/analysis , Estrogens/metabolism , Female , Humans , Hydroxyestrones , Microbiota/genetics , RNA, Ribosomal, 16S/genetics , Tandem Mass Spectrometry/methods
10.
PLoS One ; 15(1): e0228024, 2020.
Article in English | MEDLINE | ID: mdl-31978092

ABSTRACT

Abnormal accumulation of acyl-CoA cholesterol acyltransferase-1 (ACAT-1) mediated cholesterol ester has been shown to contribute to cancer progression in various cancers including leukemia, glioma, breast, pancreatic and prostate cancers. However, the significance of ACAT-1 and cholesterol esters (CE) is relatively understudied in ovarian cancer. In this in vitro study, we assessed the expression and contribution of ACAT-1 in ovarian cancer progression. We observed a significant increase in the expression of ACAT-1 and CE levels in a panel of ovarian cancer cell lines (OC-314, SKOV-3 and IGROV-1) compared to primary ovarian epithelial cells (normal controls). To confirm the tumor promoting capacity of ACAT-1, we inhibited ACAT-1 expression and activity by treating our cell lines with an ACAT inhibitor, avasimibe, or by stable transfection with ACAT-1 specific short hairpin RNA (shRNA). We observed significant suppression of cell proliferation, migration and invasion in ACAT-1 knockdown ovarian cancer cell lines compared to their respective controls (cell lines transfected with scrambled shRNA). ACAT-1 inhibition enhanced apoptosis with a concurrent increase in caspases 3/7 activity and decreased mitochondrial membrane potential. Increased generation of reactive oxygen species (ROS) coupled with increased expression of p53 may be the mechanism(s) underlying pro-apoptotic action of ACAT-1 inhibition. Additionally, ACAT-1 inhibited ovarian cancer cell lines displayed enhanced chemosensitivity to cisplatin treatment. These results suggest ACAT-1 may be a potential new target for the treatment of ovarian cancer.


Subject(s)
Disease Progression , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Sterol O-Acyltransferase/metabolism , Acetyl-CoA C-Acetyltransferase , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cholesterol Esters/metabolism , Cisplatin/pharmacology , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Neoplasm Invasiveness , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Sterol O-Acyltransferase/antagonists & inhibitors , Tumor Stem Cell Assay
11.
Autism ; 24(6): 1400-1410, 2020 08.
Article in English | MEDLINE | ID: mdl-32054311

ABSTRACT

LAY ABSTRACT: Oxytocin is a hormone naturally produced in the human body that can make the womb (uterus) contract during labor. Manufactured oxytocin is frequently given to mothers in labor to strengthen the contractions or in some cases to start labor. This study compared children with a diagnosis of autism and children without autism to see whether children with autism received more oxytocin during labor. The odds of a child having an autism diagnosis were significantly higher if the delivery was a first-time Cesarean section, if the mother had a body mass index of 35 or higher, or if the reason for delivery were a range of fetal problems that made delivery necessary. It was found that boys who were exposed to oxytocin for longer periods of time during labor and received higher total doses of oxytocin had significantly higher odds of developing autism. There were no significant associations of oxytocin dosing and autism noted in female children. As this is the first study to look at any relationship between the dose of oxytocin received during labor and the odds of developing autism, further study needs to be done to determine whether there is any cause and effect relationship. Thus, at this time, there is no recommended change in clinical practice.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Labor, Obstetric , Autism Spectrum Disorder/chemically induced , Autistic Disorder/chemically induced , Autistic Disorder/epidemiology , Cesarean Section , Child , Female , Humans , Male , Oxytocin/adverse effects , Pregnancy
12.
PLoS One ; 15(1): e0227707, 2020.
Article in English | MEDLINE | ID: mdl-31917801

ABSTRACT

Epithelial ovarian cancer (OC) is the most deadly cancer of the female reproductive system. To date, there is no effective screening method for early detection of OC and current diagnostic armamentarium may include sonographic grading of the tumor and analyzing serum levels of tumor markers, Cancer Antigen 125 (CA-125) and Human epididymis protein 4 (HE4). Microorganisms (bacterial, archaeal, and fungal cells) residing in mucosal tissues including the gastrointestinal and urogenital tracts can be altered by different disease states, and these shifts in microbial dynamics may help to diagnose disease states. We hypothesized that the peritoneal microbial environment was altered in patients with OC and that inclusion of selected peritoneal microbial features with current clinical features into prediction analyses will improve detection accuracy of patients with OC. Blood and peritoneal fluid were collected from consented patients that had sonography confirmed adnexal masses and were being seen at SIU School of Medicine Simmons Cancer Institute. Blood was processed and serum HE4 and CA-125 were measured. Peritoneal fluid was collected at the time of surgery and processed for Next Generation Sequencing (NGS) using 16S V4 exon bacterial primers and bioinformatics analyses. We found that patients with OC had a unique peritoneal microbial profile compared to patients with a benign mass. Using ensemble modeling and machine learning pathways, we identified 18 microbial features that were highly specific to OC pathology. Prediction analyses confirmed that inclusion of microbial features with serum tumor marker levels and control features (patient age and BMI) improved diagnostic accuracy compared to currently used models. We conclude that OC pathogenesis alters the peritoneal microbial environment and that these unique microbial features are important for accurate diagnosis of OC. Our study warrants further analyses of the importance of microbial features in regards to oncological diagnostics and possible prognostic and interventional medicine.


Subject(s)
Ascitic Fluid/microbiology , CA-125 Antigen/blood , Carcinoma, Ovarian Epithelial/diagnosis , Membrane Proteins/blood , Microbiota/genetics , Ovarian Neoplasms/diagnosis , WAP Four-Disulfide Core Domain Protein 2/analysis , Aged , Carcinoma, Ovarian Epithelial/blood , Carcinoma, Ovarian Epithelial/microbiology , Carcinoma, Ovarian Epithelial/surgery , Cross-Sectional Studies , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Female , Humans , Hysterectomy , Laparoscopy , Machine Learning , Middle Aged , Models, Biological , Ovarian Neoplasms/blood , Ovarian Neoplasms/microbiology , Ovarian Neoplasms/surgery , Ovariectomy , Pilot Projects , Preoperative Period , Prognosis , RNA, Ribosomal, 16S/genetics
13.
Reprod Sci ; 17(4): 339-49, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20228380

ABSTRACT

Promyelocytic leukemia (PML) protein is a nucleoprotein that can regulate a variety of cellular stress responses. The aim of this study was to determine qualitative and quantitative changes in PML expression in preeclamptic placentae. Immunoblot, quantitative reverse transcription polymerase chain reaction (qRT-PCR), and immunohistochemistry techniques were used to determine PML gene expression and localization in normal (n = 6) and preeclamptic (n = 6) placentae and primary cells. Promyelocytic leukemia protein was immunolocalized within nuclei of villus mesenchyme, but largely absent in trophoblast nuclei, with a trend for increased PML reactivity in preeclamptic placenta. Immunoblot analyses of nuclear extracts confirmed relative increases (approximately 3-fold) of PML expression in preeclamptic placentae (P < .05). Conversely, less PML messenger RNA (mRNA; approximately 2-fold) was detected in preeclamptic versus normal placental samples. In vitro, PML expression could be increased by hypoxia in cultured endothelial cells but not trophoblast. Increased PML protein expression in preeclamptic villi suggests it could contribute to decreased vascularity and placental growth and/or function.


Subject(s)
Nuclear Proteins/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adolescent , Adult , Female , Humans , Hypoxia/metabolism , Pregnancy , Promyelocytic Leukemia Protein , Trophoblasts/metabolism , Young Adult
14.
Biol Reprod ; 78(5): 841-51, 2008 May.
Article in English | MEDLINE | ID: mdl-18160678

ABSTRACT

Placental growth factor (PGF, previously known as PlGF) is prominently expressed by trophoblasts in human placenta, whereas most nontrophoblast cells express low levels of PGF mRNA under normal physiological conditions. We have shown that hypoxia decreases PGF expression in the trophoblast, but little is known about transcriptional regulation of PGF gene expression. We sought to determine promoter regions of the human PGF gene that contribute to its restricted high constitutive expression in the trophoblast. Overlapping putative promoter regions of human PGF gene encompassing 2-1.5 kb were cloned into reporter vectors and co-transfected into trophoblast and nontrophoblast cell lines. Promoter activity generated by a 2-1.5-kb clone was significantly higher in trophoblasts than in nontrophoblasts. Selective deletion mutants showed that a clone encompassing the PGF (2-828/++34) region generated promoter activity similar to the 2-1.5-kb region in the trophoblast. However, deletion of another 131 bp from this subclone (2-698/++34) resulted in significantly less promoter activity in the trophoblast. The (2-828/2-698) region significantly enhanced activity of a minimal promoter construct in trophoblast but not in nontrophoblast cells, suggesting that this region contributes to regulating PGF transcription in the trophoblast. Site-directed mutagenesis of a glial cell missing 1 (GCM1) motif in the 131-bp region significantly decreased enhancer activity in the trophoblast. Furthermore, overexpression of GCM1 significantly increased PGF 2-1.5-kb promoter activity and PGF mRNA expression in trophoblast and nontrophoblast cells. Forced overexpression of GCM1 restored PGF expression in the hypoxic trophoblast. These data support a functional role for GCM1 contributing to constitutively high trophoblast PGF expression and is the first direct evidence of an oxygen-responsive, trophoblast-specific transcription factor contributing to the regulation of PGF expression.


Subject(s)
Nuclear Proteins/metabolism , Pregnancy Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic/physiology , Trophoblasts/metabolism , Cell Line , Cell Line, Tumor , DNA-Binding Proteins , Gene Deletion , Gene Expression Regulation/physiology , Genes, Reporter/genetics , Humans , Mutation/genetics , Nuclear Proteins/genetics , Placenta Growth Factor , Pregnancy Proteins/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Transfection , Trophoblasts/cytology
15.
J Assist Reprod Genet ; 24(7): 303-15, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17616801

ABSTRACT

PROBLEM: Implantation failure and early pregnancy loss are common following natural conceptions and they are particularly important clinical hurdles to overcome following assisted reproduction attempts. The importance of adequate vascular development and maintenance during implantation has recently become a major focus of investigation. MATERIALS AND METHODS: Review of current published literature was undertaken to summerize the cells and cell products that regulate tissue vascularity during implantation. RESULTS: Vascular development at the maternal fetal interface can be regulated by a number of different cell types; two principal candidates are trophoblast and natural killer cells. A wide range of soluble factors, some with well established angiogenic functions as well as other more novel factors, can contribute to vascular development and maintenance at the maternal-fetal interface. CONCLUSIONS: Robust vascular development occurs during implantation and early placentation of normal pregnancies. Studies to define the extent and mechanisms by which defects in vascularity contribute to human implantation failure and early miscarriage need to be undertaken.


Subject(s)
Embryo Implantation/physiology , Endometrium/blood supply , Endometrium/metabolism , Female , Humans , Menstrual Cycle/metabolism , Neovascularization, Physiologic/physiology , Pregnancy , Vascular Endothelial Growth Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL