Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Minerva Obstet Gynecol ; 75(2): 150-157, 2023 Apr.
Article in English | MEDLINE | ID: mdl-34825792

ABSTRACT

BACKGROUND: Several studies have linked perineal use of talcum powder to increased risk of ovarian cancer (OC). Here, we determined that exposure to talcum powder induces malignant transformation in human normal ovarian cells. METHODS: Human primary ovarian epithelial cells (HPOE), ovarian epithelial cells (HOSEpiC), and primary fibroblasts (NF) were treated with either 100 or 500 µg/mL of talcum powder or titanium dioxide (TiO2) as a particulate control for 72 hours before assessment with a cell transformation assay and p53 and Ki-67 immunohistochemistry. RESULTS: Treatment with talcum powder resulted in formation of colonies, indicating cell malignant transformation in a dose dependent manner in ovarian cell lines. No colonies formed in the untreated ovarian cells or control ovarian cells (TiO2 treated) at either dose. There were no colonies formed in talc treated NF cells. Transformed ovarian cells were increased by 11% and 20% in HPOE and 24% and 40% in HOSEpic cells for talcum powder 100 and 500 µg/mL doses, respectively (P<0.05). There were no detectible transformed cells when cells were treated with TiO2. Importantly, p53 mutant type as well as increased expression of Ki-67 were detected in HPOE and HOSEpic cells when exposed to talcum powder. CONCLUSIONS: Exposure to talcum powder induces malignant transformation in ovarian epithelial cells but not in NF cells. These findings represent a direct effect of talcum powder exposure that is specific to normal ovarian cells and further supports previous studies demonstrating an association between the genital use of talcum powder and an increased risk of OC.


Subject(s)
Ovarian Neoplasms , Talc , Female , Humans , Talc/toxicity , Ki-67 Antigen/genetics , Tumor Suppressor Protein p53/genetics , Ovarian Neoplasms/chemically induced , Epithelial Cells
2.
Article in English | MEDLINE | ID: mdl-36255168

ABSTRACT

BACKGROUND: 2,4-dinitrophenol (DNP), an uncoupling mitochondrial agent, has been identified as a source of oxidative stress and linked to the pathogenesis of ovarian cancer. In this study, we determine the cytotoxic effect of DNP alone or in combination with chemotherapies in ovarian cancer cells. METHODS: We utilized human ovarian cancer cell lines SKOV-3 and MDAH-2774 with their chemoresistant counterparts. Cancer stem cells (CSCs) were isolated from SKOV-3 utilizing magnetic-activated cell sorting technique for CD44+/CD117+ cells. Human normal primary ovarian epithelial (NOEC) and HOSEpiC cell lines were used as a control. Cells were treated with and without chemotherapy (Taxotere 0.3µM or cisplatin 50 µM), with or without increasing doses of DNP (0.125, 0.25, or 0.5 mM) for 24 hours followed by evaluation of cell viability and IC50 utilizing MTT assay. For determination of synergism, Facombination index plots were created using the CompuSyn software. All data were run in triplicates and analyzed by t-test. RESULTS: DNP treatment of ovarian cancer and chemoresistant ovarian cancer cell lines as well as CSCs resulted in decreased cell viability in a dose dependent manner with no effect on normal cells. Combination of DNP with chemotherapy synergistically enhances cytotoxicity of chemotherapeutics in all ovarian cancer cells as compared to chemotherapy alone. CONCLUSIONS: Our data indicates the potential of the addition of DNP to the arsenal of drugs available to treat ovarian cancer, whether alone or in combination with chemotherapies. The synergistic effects of DNP in reducing the required amount of chemotherapy, is critical for the alleviation of harmful side effects.

3.
Am J Physiol Heart Circ Physiol ; 323(5): H1019-H1036, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36149771

ABSTRACT

Historically, capillaries have been viewed as the microvascular site for flux of nutrients to cells and removal of waste products. Capillaries are the most numerous blood vessel segment within the tissue, whose vascular wall consists of only a single layer of endothelial cells and are situated within microns of each cell of the tissue, all of which optimizes capillaries for the exchange of nutrients between the blood compartment and the interstitial space of tissues. There is, however, a growing body of evidence to support that capillaries play an important role in sensing the tissue environment, coordinating microvascular network responses, and controlling blood flow. Much of our growing understanding of capillaries stems from work in skeletal muscle and more recent work in the brain, where capillaries can be stimulated by products released from cells of the tissue during increased activity and are able to communicate with upstream and downstream vascular segments, enabling capillaries to sense the activity levels of the tissue and send signals to the microvascular network to coordinate the blood flow response. This review will focus on the emerging role that capillaries play in communication between cells of the tissue and the vascular network required to direct blood flow to active cells in skeletal muscle and the brain. We will also highlight the emerging central role that disruptions in capillary communication may play in blood flow dysregulation, pathophysiology, and disease.


Subject(s)
Capillaries , Endothelial Cells , Capillaries/physiology , Muscle, Skeletal/blood supply , Regional Blood Flow/physiology , Communication
4.
Eur J Appl Physiol ; 121(5): 1241-1254, 2021 May.
Article in English | MEDLINE | ID: mdl-33538852

ABSTRACT

PURPOSE: We explore the motor unit recruitment and control of perfusion of microvascular units in skeletal muscle to determine whether they coordinate to match blood flow to metabolic demand. METHODS: The PubMed database was searched for historical, current and relevant literature. RESULTS: A microvascular, or capillary unit consists of 2-20 individual capillaries. Individual capillaries within a capillary unit cannot increase perfusion independently of other capillaries within the unit. Capillary units perfuse a short segment of approx. 12 muscle fibres located beside each other. Motor units consist of muscle fibres that can be dispersed widely within the muscle volume. During a contraction, where not all motor units are recruited, muscle fibre contraction will result in increased perfusion of associated capillaries as well as all capillaries within that capillary unit. Perfusion of the entire capillary unit will result in an increased blood flow delivery to muscle fibres associated with active motor unit plus approximately 11 other inactive muscle fibres within the same region. This will result in an overperfusion of the muscle resulting in blood flow in excess of the muscle fibre needs. CONCLUSIONS: Given the architecture of the capillary units and the dispersed nature of muscle fibres within a motor unit, during submaximal contractions, where not all motor units are recruited, there will be a greater perfusion to the muscle than that predicted by the number of active muscle fibres. Such overperfusion brings into question if blood flow and metabolic demand are as tightly matched as previously assumed.


Subject(s)
Microcirculation/physiology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/blood supply , Muscle, Skeletal/innervation , Recruitment, Neurophysiological/physiology , Hemodynamics/physiology , Humans , Muscle Contraction/physiology , Regional Blood Flow/physiology
5.
Gynecol Endocrinol ; 37(4): 349-352, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33084436

ABSTRACT

OBJECTIVE: To test whether recombinant anti-Müllerian hormone (rAMH) could exert an inhibitory function on BRCA1/2 expression in human ovarian cortex. METHODS: Pilot study on ovariectomized nude mice xenotransplanted with human vitrified/warmed ovarian cortex and treated with rAMH via infusion pump. Twelve nude mice were ovariectomized and Alzet pumps delivering 1.23 mcg rAMH/day to reach a serum concentration of 17.5 ng/mL, or placebo (controls), were inserted intraabdominally. Previously vitrified/warmed 2x2 mm ovarian cortex fragments were transplanted on day 7 and then harvested on day 14 after pump placement. PCR analyses determined mRNA levels for BRCA1 and BRCA2 in the human ovarian cortex. RESULTS: In mice treated with rAMH, BRCA1 expression was significantly lower (0.196 fg/µg RNA, IQR 0.158, 0.236) than in controls (0.544 fg/µg RNA, IQR 0.458, 0.554; p = .030), while BRCA2 expression remained similar in rAMH mice (5.355 fg/µg RNA, IQR 4.479, 6.230) and in controls (4.011 fg/µg RNA, IQR 3.650, 4.182; p = .327). CONCLUSION: Administration of rAMH in the peri-transplant period caused downregulation of BRCA1, but not of BRCA2 expression, in human ovarian cortex. These results help our understanding of DNA repair mechanism in the ovarian cortex and identify AMH's possible protective effect on ovarian reserve in BRCA1 mutation carriers.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Gene Expression Regulation/drug effects , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Ovary/drug effects , RNA, Messenger/drug effects , Adolescent , Animals , Female , Humans , Mice , Mice, Nude , Ovary/transplantation , Pilot Projects , RNA, Messenger/metabolism
7.
Reprod Sci ; 27(4): 1030-1036, 2020 04.
Article in English | MEDLINE | ID: mdl-32124395

ABSTRACT

HSP60 is a mitochondrial chaperone protein that is associated with decreased overall survival of ovarian cancer patients. We determined whether targeting HSP60 with its monoclonal antibody would induce cytotoxicity in sensitive and chemoresistant ovarian cancer cells and whether it is synergistic when combined with chemotherapeutic drugs. Epithelial ovarian cancer (EOC) cells and their docetaxel- or cisplatin-resistant counterparts were utilized. HSP60 mRNA levels were determined by real-time RT-PCR. Cytotoxicity of HSP60 antibody (0.5 or 1.5 µg/ml) alone and in combination with chemotherapy were assessed by MTT Cell Proliferation Assay. Unpaired t tests were used to compare groups for real-time RT-PCR. One-way ANOVA followed by Tukey's post hoc tests with Bonferroni correction was performed for cytotoxicity comparisons. Significant synergistic effects of the antibody combined with chemotherapy were determined by the CompuSyn Software. Basal HSP60 mRNA levels were increased in chemoresistant EOC cells as compared with their sensitive counterparts (p < 0.05). There was no significant difference in cytotoxicity between EOC cell types; however, treatment with the HSP60 antibody for 24 h showed a dose response (0.5 and 1.5 µg/ml) cytotoxic effect to both sensitive and chemoresistant EOC cells as compared with the isotype control (p < 0.05). Importantly, treatment with both doses of HSP60 antibody was not cytotoxic to normal macrophages. Combination of the HSP60 antibody with docetaxel or cisplatin was significantly synergistic in both sensitive and chemoresistant EOC cells. Here, we identify a novel target that may serve not only for ovarian cancer treatment but also for sensitization of patients to chemotherapy. The cytotoxic effect of HSP60 monoclonal antibody and its synergism with chemotherapeutic agents highlight HSP60 as a promising target for therapy and chemosensitization in ovarian cancer treatment.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Carcinoma, Ovarian Epithelial/drug therapy , Chaperonin 60/administration & dosage , Drug Resistance, Neoplasm/drug effects , Mitochondrial Proteins/administration & dosage , Ovarian Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Chaperonin 60/immunology , Chaperonin 60/metabolism , Cisplatin/administration & dosage , Docetaxel/administration & dosage , Drug Therapy, Combination/methods , Female , Humans , Mitochondrial Proteins/immunology , Mitochondrial Proteins/metabolism , RNA, Messenger/metabolism
8.
Reprod Sci ; 26(12): 1603-1612, 2019 12.
Article in English | MEDLINE | ID: mdl-30819054

ABSTRACT

Genital use of talcum powder and its associated risk of ovarian cancer is an important controversial topic. Epithelial ovarian cancer (EOC) cells are known to manifest a persistent prooxidant state. Here we demonstrated that talc induces significant changes in key redox enzymes and enhances the prooxidant state in normal and EOC cells. Using real-time reverse transcription polymerase chain reaction and enzyme-linked immunosorbent assay, levels of CA-125, caspase-3, nitrate/nitrite, and selected key redox enzymes, including myeloperoxidase (MPO), inducible nitric oxide synthase (iNOS), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPX), and glutathione reductase (GSR), were determined. TaqMan genotype analysis utilizing the QuantStudio 12K Flex was used to assess single-nucleotide polymorphisms in genes corresponding to target enzymes. Cell proliferation was determined by MTT proliferation assay. In all talc-treated cells, there was a significant dose-dependent increase in prooxidant iNOS, nitrate/nitrite, and MPO with a concomitant decrease in antioxidants CAT, SOD, GSR, and GPX (P < .05). Remarkably, talc exposure induced specific point mutations that are known to alter the activity in some of these key enzymes. Talc exposure also resulted in a significant increase in inflammation as determined by increased tumor marker CA-125 (P < .05). More importantly, talc exposure significantly induced cell proliferation and decreased apoptosis in cancer cells and to a greater degree in normal cells (P < .05). These findings are the first to confirm the cellular effect of talc and provide a molecular mechanism to previous reports linking genital use to increased ovarian cancer risk.


Subject(s)
Carcinoma, Ovarian Epithelial/etiology , Ovarian Neoplasms/etiology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Talc/adverse effects , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Catalase/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Glutathione Peroxidase/metabolism , Humans , Mutation , Nitric Oxide Synthase Type II/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Peroxidase/metabolism , Risk Factors , Superoxide Dismutase/metabolism , Talc/administration & dosage
10.
Reprod Sci ; 26(6): 724-733, 2019 06.
Article in English | MEDLINE | ID: mdl-30185141

ABSTRACT

AIMS: Hypoxia and the resulting oxidative stress play a major role in postoperative tissue fibrosis. The objective of this study was to determine the effect of l-alanyl-l-glutamine (Ala-Gln) on key markers of postoperative tissue fibrosis: hypoxia-inducible factor (HIF) 1α and type I collagen. METHODS: Primary cultures of human normal peritoneal fibroblasts (NPF) established from normal peritoneal tissue were treated with increasing doses of Ala-Gln (0, 1, 2, or 10 mM) with hypoxia ([2% O2] 0-48 hours; continuous hypoxia) or after hypoxia (0.5, 1, 2, 4 hours) and restoration of normoxia (episodic hypoxia) with immediate treatment with Ala-Gln. Hypoxia-inducible factor 1α and type 1 collagen levels were determined by enzyme-linked immunosorbent assay. Data were analyzed with 1-way analysis of variance followed by Tukey tests with Bonferroni correction. RESULTS: Hypoxia-inducible factor 1α and type I collagen levels increased in untreated controls by 3- to 4-fold in response to continuous and episodic hypoxia in human NPF. Under continuous hypoxia, HIF-1α and type I collagen levels were suppressed by Ala-Gln in a dose-dependent manner. l-alanyl-l-glutamine treatment after episodic hypoxia also suppressed HIF-1α and type I collagen levels for up to 24 hours for all doses and up to 48 hours at the highest dose, regardless of exposure time to hypoxia. CONCLUSIONS: l-alanyl-l-glutamine significantly suppressed hypoxia-induced levels of key tissue fibrosis (adhesion) phenotype markers under conditions of continuous as well as episodic hypoxia in vitro. This effect of glutamine on molecular events involved in the cellular response to insult or injury suggests potential therapeutic value for glutamine in the prevention of postoperative tissue fibrosis.


Subject(s)
Dipeptides/pharmacology , Fibrosis/metabolism , Postoperative Complications/metabolism , Signal Transduction/drug effects , Tissue Adhesions/prevention & control , Biomarkers/analysis , Cell Hypoxia , Cells, Cultured , Collagen Type I/analysis , Dipeptides/administration & dosage , Fibroblasts/chemistry , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Peritoneum/cytology
11.
J Assist Reprod Genet ; 35(10): 1831-1841, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30043336

ABSTRACT

OBJECTIVE: To determine whether recombinant AMH (rAMH) could prevent post-transplant follicular depletion by acting on the stemness markers Oct-4, Sox2, and NANOG. MATERIALS AND METHODS: This was an experimental study where 12 ovariectomized nude mice were xenotransplanted with vitrified/warmed ovarian cortex obtained from a pre-pubertal girl and Alzet pumps delivering rAMH, or placebo (control), were inserted intra-abdominally. Previously vitrified/warmed ovarian cortex fragments were transplanted after 7 days and then harvested after 14 days from pump placement. We performed real-time RT-PCR analyses, ELISA for AMH, FSH, and estradiol, histologic measurement of ovarian follicles, and immunohistochemistry for Ki67 and TUNEL. The main outcome measures were serum levels and tissue expression of the parameters under investigation and follicle count. RESULTS: Serum AMH, FSH, and estradiol reflected post-ovariectomy profiles and were mildly influenced by rAMH administration. Ovarian cortex expression of AMH, AMH-R2, VEGF, GDF9, Oct-4, and Sox2 was lower in rAMH mice than in controls, while NANOG was upregulated. There was a non-significant decrease in primordial follicles after vitrification-warming, and xenotransplantation further decreased this number. There were lower cell replication and depressed apoptosis in the rAMH group. CONCLUSIONS: Administration of recombinant AMH in the peri-transplant period did not protect the initial follicular depletion but decreased apoptosis and cellular activation and regulated stem cell markers' tissue expression. These results aid our understanding of the inhibitory effects of AMH on follicular development and show the benefit of administering exogenous AMH at the time of pre-pubertal ovarian cortex transplant to protect the follicles from pre-activation and premature depletion.


Subject(s)
Anti-Mullerian Hormone/genetics , Heterografts/metabolism , Ovarian Follicle/transplantation , Ovary/transplantation , Animals , Anti-Mullerian Hormone/administration & dosage , Anti-Mullerian Hormone/blood , Apoptosis/genetics , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Gene Expression Regulation, Developmental , Heterografts/drug effects , Heterografts/growth & development , Humans , Mice , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/genetics , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovariectomy , Ovary/drug effects , Ovary/growth & development , Ovary/metabolism , SOXB1 Transcription Factors/genetics , Transplantation, Heterologous , Vitrification
12.
Gynecol Oncol ; 148(3): 567-575, 2018 03.
Article in English | MEDLINE | ID: mdl-29329880

ABSTRACT

OBJECTIVE: The objective of this study was to determine the expression, and effect of targeting CD11b with a monoclonal antibody in ovarian cancer cells. METHODS: CD11b expression was determined in epithelial ovarian cancer (EOC) cell lines and tissues by immunofluorescence and flow cytometry. Cytotoxicity of the CD11b antibody and synergism with chemothearapeutic drugs were determined by the MTT Cell Proliferation Assay in human macrophages, normal ovarian epithelial cells, and in both sensitive and chemoresistant EOC cell lines. Cell migration was assessed with a scratch assay and in vivo effects of the CD11b antibody was assessed with a nude mouse ovarian cancer xenograft model. Data was analyzed with either t-tests or one-way ANOVA. RESULTS: CD11b was unexpectedly expressed in several EOC lines and tissues, but not normal tissues. Targeting CD11b with its monoclonal antibody resulted in intriguing cytotoxic effects in sensitive and chemoresistant EOC lines, while surprisingly not affecting normal cells. More importantly, the cytotoxicity of the CD11b antibody when combined with chemotherapeutic drugs (cisplatin or docetaxel) was significantly synergistic, in both sensitive and chemoresistant EOC cells. The anti-tumorigenic effect of the CD11b antibody was confirmed in an ovarian cancer nude mouse xenograft model. CONCLUSION: Here we identify CD11b as a novel target, which selectively induces cytotoxicity in ovarian cancer cells.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents/pharmacology , CD11b Antigen/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cisplatin/pharmacology , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Taxoids/pharmacology , Animals , Antineoplastic Agents, Immunological/immunology , Apoptosis/drug effects , CD11b Antigen/immunology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel , Drug Resistance, Neoplasm , Drug Synergism , Epithelial Cells/drug effects , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , In Vitro Techniques , Mice , Mice, Nude , Molecular Targeted Therapy , Neoplasm Transplantation , Neoplasms, Glandular and Epithelial/immunology , Ovarian Neoplasms/immunology , Ovary/drug effects , Oxidative Stress/drug effects , Peroxidase/drug effects , Peroxidase/metabolism , Xenograft Model Antitumor Assays
13.
Reprod Sci ; 25(8): 1218-1223, 2018 08.
Article in English | MEDLINE | ID: mdl-29141508

ABSTRACT

OBJECTIVE: To test whether recombinant anti-Müllerian hormone (AMH) can inhibit ovarian cortex function by modulating the expression of other hormone receptors. MATERIALS AND METHODS: Pilot experimental study with ovarian cortex obtained from 5 patients. Immediately after explant, the ovarian cortex specimens were divided into 5 equal fragments. One fragment was flash-frozen (uncultured) and 4 were incubated for 48 hours at 37°C in a pH-adjusted gamete buffer medium with increasing AMH concentrations of 0, 5, 25, and 50 ng/mL. After incubation, all specimens were rinsed and flash-frozen for polymerase chain reaction (PCR) executed in triplicates. We utilized real-time reverse transcription-polymerase chain reaction (RT-PCR) to determine messenger RNA (mRNA) levels of AMH and its receptor Anti-Müllerian Hormone-Receptor 2 (AMH-R2), follicle stimulating hormone receptor (FSH-R), luteinizing hormone receptor (LH-R), inhibin B, and insulin-like growth factor 1 receptor 1 (IGF1-R1) in ovarian cortex tissue. In addition, we performed Ki-67 immunostaining to evaluate cell proliferation in the treatment groups. RESULTS: Absence of recombinant human AMH (rAMH) caused upregulation of all markers. Exposure to increasing rAMH concentrations caused tissue AMH expression downregulation ( P = .024), while AMH-R2 ( P = .005), FSH-R ( P = .009), LH-R ( P = .003), and inhibin B ( P = .001) mRNA expression followed a bell-shaped response with an increased expression at low dose, followed by a decreased expression at higher doses. Expression of IGF1-R1 was independent ( P = .039) of rAMH exposure. The Ki-67 immunostaining showed an increased cell proliferation in the media control compared to the uncultured and the tissue cultured with rAMH. CONCLUSIONS: Culture with increasing rAMH concentrations caused downregulation of its own, as well as other hormone receptors, and a decreased ovarian cortex cell proliferation. These results help understanding the inhibitory effects of AMH on follicular development.


Subject(s)
Anti-Mullerian Hormone/metabolism , Ovary/metabolism , Receptors, Peptide/metabolism , Receptors, Pituitary Hormone/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Adult , Anti-Mullerian Hormone/administration & dosage , Female , Gene Expression Regulation , Humans , Inhibins/metabolism , Ovary/drug effects , Pilot Projects , Premenopause , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism , Receptors, FSH/metabolism , Receptors, LH/metabolism , Receptors, Somatomedin/metabolism , Recombinant Proteins
14.
Fertil Steril ; 107(6): 1387-1394.e1, 2017 06.
Article in English | MEDLINE | ID: mdl-28483502

ABSTRACT

OBJECTIVE: To determine the effects of attenuating oxidative stress with the use of dichloroacetate (DCA) on the expression of key redox enzymes myeloperoxidase (MPO) and inducible nitric oxide synthase (iNOS) as well as on apoptosis. DESIGN: Prospective experimental study. SETTING: University medical center. PATIENT(S): Cells established from myometrium and uterine fibroid from the same patients. INTERVENTION(S): Cells were exposed to normal (20% O2) or hypoxic (2% O2) conditions for 24 hours with or without DCA (20 µg/mL), a metabolic modulator that shifts anaerobic to aerobic metabolism. MAIN OUTCOME MEASURE(S): Nitrate/nitrite (iNOS activity indicator), iNOS, Bcl-2/Bax ratio, MPO, and caspase-3 activities and levels were determined by means of Greiss assay, real-time reverse-transcription polymerase chain reaction, and ELISA. Data were analyzed with the use of SPSS by means of one-way analysis of variance with Tukey post hoc analysis and independent t tests. RESULT(S): MPO, iNOS, and nitrate/nitrite expression were higher in leiomyoma than in myometrial cells, and they were further enhanced by hypoxia in myometrial cells. Treatment with the use of DCA decreased MPO, iNOS, and nitrate/nitrite levels and negated the effect of hypoxia in both types of cells. Leiomyoma cells showed less apoptosis, as indicated by both caspase-3 activity and the Bcl-2/Bax ratio, than myometrial cells. Hypoxia further decreased apoptosis in myometrial cells with no further effect on leiomyoma cells. Treatment with DCA resulted in increased apoptosis in both types of cells, even in the presence of hypoxia. CONCLUSION(S): Shifting anaerobic to aerobic metabolism with the use of DCA resulted in an increase in apoptosis in leiomyoma cells and protected myometrial cells from the acquisition of the leiomyoma-like phenotype.


Subject(s)
Cell Survival/immunology , Leiomyoma/immunology , Oxidative Stress/immunology , Oxidoreductases/immunology , Tumor Hypoxia/immunology , Uterine Neoplasms/immunology , Female , Humans , Leiomyoma/pathology , Nitric Oxide Synthase Type II/immunology , Oxygen/immunology , Peroxidase/immunology , Reactive Oxygen Species/immunology , Tumor Cells, Cultured , Uterine Neoplasms/pathology
15.
Gynecol Oncol ; 145(3): 595-602, 2017 06.
Article in English | MEDLINE | ID: mdl-28237618

ABSTRACT

Clinical and epidemiological investigations have provided evidence supporting the role of reactive oxygen species (ROS) and reactive nitrogen species (RNS), collectively known as oxidative stress, in the etiology of cancer. Exogenous factors such as chronic inflammation, infection and hypoxia are major sources of cellular oxidative stress. Specifically, oxidative stress plays an important role in the pathogenesis, neoangiogenesis, and dissemination of local or distant ovarian cancer, as it is known to induce phenotypic modifications of tumor cells by cross talk between tumor cells and the surrounding stroma. Subsequently, the biological significance of the relationship between oxidative stress markers and various stages of epithelial ovarian cancer highlights potential therapeutic interventions as well as provides urgently needed early detection biomarkers. In the light of our scientific research and the most recent experimental and clinical observations, this review provides the reader with up to date most relevant findings on the role of oxidative stress in the pathogenesis of ovarian cancer and the possible therapeutic implications.


Subject(s)
Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Oxidative Stress/physiology , Carcinogenesis , Female , Humans
16.
Free Radic Biol Med ; 102: 122-132, 2017 01.
Article in English | MEDLINE | ID: mdl-27890641

ABSTRACT

Oxidative stress plays an important role in the pathophysiology of ovarian cancer. Resistance to chemotherapy presents a significant challenge for ovarian cancer treatment. Specific single nucleotide polymorphisms (SNPs) in key redox enzymes have been associated with ovarian cancer survival and progression. The objective of this study was to determine whether chemotherapy induces point mutations in key redox enzymes that lead to the acquisition of chemoresistance in epithelial ovarian cancer (EOC). Human EOC cell lines and their chemoresistant counterpart were utilized for this study. Specific SNPs in key redox enzymes were analyzed by TaqMan SNP Genotyping. Activities and levels of key redox enzymes were determined by real-time RT-PCR, ELISA and a greiss assay. Point mutations in key redox enzymes were introduced into sensitive EOC cells via the CRISPR/Cas9 system. Cell viability and IC50 for cisplatin were determined by the MTT Cell Proliferation Assay. Data was analyzed with SPSS using Student's two-tailed t-tests and One-way ANOVA followed by Dunnett's or Tukey's post hoc tests, p<0.05. Here, we demonstrate that chemoresistant EOC cells are characterized by a further enhancement in oxidative stress as compared to sensitive counterparts. Additionally, chemoresistant EOC cells manifested specific point mutations, which are associated with altered enzymatic activity, in key redox enzymes that are not detected in sensitive counterparts. Supplementation of an antioxidant was able to successfully sensitize EOC cells to chemotherapeutics. Causality was established by the induction of these point mutations in sensitive EOC cells, which resulted in a significant increase in the level of chemoresistance. These findings indicate that chemotherapy induces specific point mutations in key redox enzymes that contribute to the acquisition of chemoresistance in EOC cells, highlighting a potential novel mechanism. Identification of targets for chemoresistance with either biomarker and/or screening potential will have a significant impact for the treatment of this disease.


Subject(s)
Cisplatin/administration & dosage , Drug Resistance, Neoplasm/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Oxidative Stress/genetics , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Survival/drug effects , Female , Genotype , Humans , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Oxidation-Reduction , Point Mutation/genetics , Polymorphism, Single Nucleotide/genetics
17.
Reprod Sci ; 23(11): 1484-1492, 2016 11.
Article in English | MEDLINE | ID: mdl-27122375

ABSTRACT

OBJECTIVE: Chemoresistance remains a major challenge in the treatment of ovarian cancer. As part of a survival mechanism, tumor cells have been shown to release proangiogenic factors, such as vascular endothelial growth factor (VEGF), through a mechanism that involves the upregulation of hypoxia-induced factor (HIF)-1α. The objective of this study was to compare the expression of VEGF and its receptors (R1 and R2) as well as HIF-1α in chemoresistant epithelial ovarian cancer (EOC) cells to their chemosensitive counterparts and determine their impact on angiogenesis. METHODS: Two human EOC cell lines, MDAH-2774 and SKOV-3, and their cisplatin- or taxotere-resistant counterparts were used. Total RNA and protein were subjected to real-time reverse transcriptase-polymerase chain reaction, immunoprecipitation/Western blot and enzyme-linked immunosorbent assay to evaluate the expression of VEGF, VEGF receptors (R1 and R2), and HIF-1α. Angiogenesis was assessed with an in vitro angiogenesis assay. Data were analyzed using independent Student t tests and chi-square. RESULTS: Both taxotere- and cisplatin-resistant MDAH-2774 and SKOV-3 EOC cell lines manifested a significant decrease in VEGF, VEGF receptors, HIF-1α messenger RNA, and protein levels as compared to their chemosensitive counterparts. There was a significant decrease in the number and thickness of polygon blood vessel formation in chemoresistant EOC cells compared to chemosensitive counterparts. CONCLUSION: Cisplatin- and taxotere-resistant EOC cells are characterized by lower VEGF, VEGF receptors, and HIF-1α, and decreased angiogenesis. These findings may indicate a decrease in drug delivery at the tumor site, hence allowing the persistence of chemoresistant EOC cells.


Subject(s)
Drug Resistance , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Antineoplastic Agents/administration & dosage , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cisplatin/administration & dosage , Docetaxel , Humans , Taxoids/administration & dosage
18.
Fertil Steril ; 105(6): 1628-1637.e1, 2016 06.
Article in English | MEDLINE | ID: mdl-26920255

ABSTRACT

OBJECTIVE: To determine whether metabolic markers are differentially expressed in normal and adhesion fibroblasts with and without hypoxia exposure. DESIGN: Prospective experimental study. SETTING: University research laboratory. PATIENT(S): Fibroblasts established from normal peritoneum and adhesion tissues from the same patients. INTERVENTION(S): In vitro experiments on normal peritoneal and adhesion fibroblasts under normal and hypoxic (2% O2) conditions. MAIN OUTCOME MEASURE(S): Expression of metabolic markers, glyceraldehyde 3-phosphate dehydrogenase (GAPDH), glucose transporter 1 (GLUT1), hypoxia inducible factor (HIF)-1α, hexokinase 2 (HK2), lactose dehydrogenase A (LDHA), and pyruvate dehydrogenase alpha 1 (PDHA1) were measured using real-time reverse transcription polymerase chain reaction; adenosine triphosphate (ATP), HIF-1α, and lactate levels were assessed with ELISAs. RESULT(S): Baseline mRNA levels of GAPDH and HIF-1α were increased, while GLUT1 and PDHA1 were decreased in adhesion as compared with in normal peritoneal fibroblasts. There was no change in baseline levels of HK2 or LDHA between the cell lines. Hypoxia increased protein levels of HIF-1α and mRNA levels of GAPDH, GLUT1, and HK2 and decreased levels of PDHA1 in both cell lines. Hypoxia increased LDHA mRNA levels in normal peritoneal fibroblasts. Baseline levels of lactate and ATP were lower in adhesion as compared with in normal peritoneal fibroblasts. In response to hypoxia, there was an increase in lactate in both cell lines and a decrease in ATP in normal fibroblasts. CONCLUSION(S): Adhesion fibroblasts manifested an altered metabolic profile, which favors the glycolytic pathway, and is further altered by hypoxia. Targeting these specific metabolic markers during surgery can be an important therapeutic intervention minimizing the development of postoperative adhesions.


Subject(s)
Fibroblasts/metabolism , Glycolysis/physiology , Metabolome/physiology , Phenotype , Cells, Cultured , Female , Humans , Peritoneum/metabolism , Prospective Studies , Tissue Adhesions/metabolism
19.
Reprod Sci ; 23(5): 610-2, 2016 May.
Article in English | MEDLINE | ID: mdl-26408397

ABSTRACT

BACKGROUND: Postoperative adhesions occur in the overwhelming majority of patients after laparotomy and laparoscopy. The clinical consequences of adhesions can be devastating. Adhesions are believed to contribute to infertility and are the most common cause of small bowel obstruction and abdominal pelvic pain. Our objective is to develop a model for ex vivo development of postoperative adhesions. METHODS: Peritoneal tissue strips were obtained from Sprague Dawley rats and were abraded by scraping the tissue surface with a scalpel. Tissues were incubated in a special medium, and visible adhesion bands were assessed. Experiments were performed in a university setting. RESULTS: Adhesion bands were observed as early as 24 hours after abrasion. No adhesion bands were observed in the control tissue strips. CONCLUSIONS: This is a model system for the creation of actual adhesive bands utilizing an ex vivo culture of peritoneal strips.


Subject(s)
Models, Animal , Peritoneum/pathology , Postoperative Complications/pathology , Tissue Adhesions/pathology , Animals , Female , Organ Culture Techniques/methods , Peritoneum/surgery , Postoperative Complications/etiology , Rats , Rats, Sprague-Dawley , Tissue Adhesions/etiology
20.
PLoS One ; 10(8): e0135739, 2015.
Article in English | MEDLINE | ID: mdl-26301412

ABSTRACT

Ovarian cancer is the deadliest of all gynecologic cancers. Recent evidence demonstrates an association between enzymatic activity altering single nucleotide polymorphisms (SNP) with human cancer susceptibility. We sought to evaluate the association of SNPs in key oxidant and antioxidant enzymes with increased risk and survival in epithelial ovarian cancer. Individuals (n = 143) recruited were divided into controls, (n = 94): healthy volunteers, (n = 18), high-risk BRCA1/2 negative (n = 53), high-risk BRCA1/2 positive (n = 23) and ovarian cancer cases (n = 49). DNA was subjected to TaqMan SNP genotype analysis for selected oxidant and antioxidant enzymes. Of the seven selected SNP studied, no association with ovarian cancer risk (Pearson Chi-square) was found. However, a catalase SNP was identified as a predictor of ovarian cancer survival by the Cox regression model. The presence of this SNP was associated with a higher likelihood of death (hazard ratio (HR) of 3.68 (95% confidence interval (CI): 1.149-11.836)) for ovarian cancer patients. Kaplan-Meier survival analysis demonstrated a significant median overall survival difference (108 versus 60 months, p<0.05) for those without the catalase SNP as compared to those with the SNP. Additionally, age at diagnosis greater than the median was found to be a significant predictor of death (HR of 2.78 (95% CI: 1.022-7.578)). This study indicates a strong association with the catalase SNP and survival of ovarian cancer patients, and thus may serve as a prognosticator.


Subject(s)
Catalase/genetics , Ovarian Neoplasms/mortality , Polymorphism, Single Nucleotide/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Catalase/physiology , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Prognosis , Proportional Hazards Models , Survival Analysis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...