Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
2.
Confl Health ; 18(1): 42, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822366

ABSTRACT

In November 2023, a variety of disparate health organizations formed an international coalition to consolidate efforts and develop collaborative strategies in response to the increasing critical healthcare challenges caused by the recent war in Gaza. The coalition includes medical and public health experts, humanitarian practitioners, academics, and health policy-makers from across the world. Their membership has not much to do with mainstream humanitarian organisations. It is lead by the diaspora from the region. Their vision is the long-term reconstruction of the health system in Gaza while responding the most urgent needs. This collective effort will require explicit efforts to speak with one voice and avoid duplication. This collective movement may be an orginal initiative that may be able to beat the expected international donor fatigue.

3.
Expert Rev Anticancer Ther ; 24(7): 485-491, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38712572

ABSTRACT

INTRODUCTION: The link between talcum powder use and cancer, particularly ovarian cancer, has been a topic of scientific research and legal debate for several years. Studies have suggested a potential association between long-term talcum powder use in the genital area and an increased risk of ovarian cancer. AREAS COVERED: The following report includes up-to-date evidence to support the potential link between talcum powder use and the risk of developing ovarian cancer. The International Agency for Research on Cancer, which is part of the World Health Organization, classified talc-based body powder as possibly carcinogenic to humans when used in the female genital area. However, other studies have not consistently supported this association, and thus more research is needed to establish a clear and definitive link between talcum powder use and cancer. Despite this, recent molecular-level data have linked talc to alterations in redox balance, gene mutations, and inflammatory responses. Specifically, we have identified a role for talc to induce the pro-oxidant state, inhibit apoptosis, and more importantly induced cellular transformation in normal ovarian cells. EXPERT OPINION: We presented unequivocal evidence to support our opinion that talc is not biologically inert and induces molecular changes that mimic the hallmarks of cancer.


Subject(s)
Ovarian Neoplasms , Oxidative Stress , Talc , Talc/adverse effects , Talc/administration & dosage , Humans , Female , Ovarian Neoplasms/pathology , Animals , Apoptosis , Powders , Cell Transformation, Neoplastic/chemically induced , Risk , Carcinogens/toxicity
4.
Biomarkers ; 28(7): 663-671, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37982229

ABSTRACT

Background: Ovarian cancer cells are known to express myeloperoxidase (MPO), an oxidant-producing enzyme with a 150 kDa homodimer, consisting of two identical monomers connected by a disulfide bond. Here, we aim to validate monomeric MPO (mMPO) as a biomarker for the early detection of ovarian cancer.Methods: Human ovarian cancer cells, sera from patients at various stages, sera from non-cancer inflammatory gynecological diseases, and healthy volunteers were used. Monomeric and dimeric MPO were measured by ELISA. Receiver operating curves were used to compare the predictive powers of serum dimeric and monomeric MPO to discriminate between samples.Results: The expression of MPO was unique to ovarian cancer cells. Specifically, mMPO was found to be the only form of MPO in all ovarian cancer cell lines. Intriguingly, mMPO was detected in the sera from all patients with ovarian cancer at various stages, but not from healthy individuals. Serum mMPO discriminated between early-stage ovarian cancer, healthy controls, and benign inflammatory gynecologic disorders. In addition, mMPO discriminated between the early and late stages of the disease.Conclusion: This work highlights mMPO as a potential biomarker for early detection of ovarian cancer, which is critically needed.


Subject(s)
Ovarian Neoplasms , Female , Humans , Biomarkers, Tumor , Ovarian Neoplasms/diagnosis , Peroxidase/metabolism
5.
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
6.
Minerva Obstet Gynecol ; 75(3): 219-226, 2023 Jun.
Article in English | MEDLINE | ID: mdl-34282611

ABSTRACT

BACKGROUND: The aim of this study was to explore the organic features of redundant endometrium (RE), we examined the expression of different endometrial hormone receptors, oncogenes, and cell replication markers, in normal endometrium (NE), endometrial polyps (EP) and RE specimens. METHODS: This was an experimental study examining endometrial tissue expression of estrogen receptors (ER1 and 2), progesterone receptors (PR-A+B), androgen receptor (AR), insulin receptor (Insulin-R), insulin-like growth factor receptor 1 (IGFR-1), thyroid hormone receptor (TH-RB), B-cell lymphoma 2 (Bcl-2), Ki67, HOXA10, in women with NE, EP and RE, of women undergoing hysteroscopy for benign gynecologic pathology. Specimens were separated in 3 groups: NE, EP, RE. Endometrial samples were processed for real-time RT-PCR analyses. Main outcome measure was tissue expression of the markers in the three groups. RESULTS: Of the 16 patients, 2 had NE, 8 had RE, 5 had EP, 1 had both, RE and EP. Compared to NE, RE and EP showed significantly increased Bcl-2, Insulin-R, ER-ß, PR-A+B, and TRB expression (P<0.044), with EP showing significantly increased PR-A+B, compared to RE (3.29±0.47 fg/µg RNA versus 1.86±0.34 fg/µg RNA; P=0.023). The other markers were not significantly different across the three groups: Ki67 appeared non-significantly decreased, while HOXA10, IGF-R1, AR, and ER-α, were non-significantly increased. CONCLUSIONS: RE showed biochemical characteristics different from NE. Similar to endometrial polyps, RE showed enhanced cell differentiation, but not cell replication. These changes in RE could be detrimental for embryo implantation and should be of consideration in women undergoing fertility treatments.


Subject(s)
Insulins , Polyps , Female , Humans , Endometrium/chemistry , Endometrium/metabolism , Endometrium/pathology , Insulins/metabolism , Ki-67 Antigen/genetics , Ki-67 Antigen/analysis , Ki-67 Antigen/metabolism , Pilot Projects , Polyps/genetics , Polyps/metabolism , Polyps/pathology , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
7.
Reprod Sci ; 30(1): 291-300, 2023 01.
Article in English | MEDLINE | ID: mdl-35799017

ABSTRACT

We were the first to report that epithelial ovarian cancer (EOC) cells and tissues express myeloperoxidase (MPO) that is known to play a role in immune surveillance and inflammation by myeloid cells. Additionally, we reported that MPO is colocalized with inducible nitric oxide synthase (iNOS), a key pro-oxidant enzyme, and plays a key role in regulating apoptosis in EOC cells. Whereas myeloid cells express MPO in a dimeric form, intriguingly, here we report the unique expression of only the monomeric form of MPO in EOC cells, tissues, and blood of an ovarian cancer patient. Additionally, we have identified a cell membrane receptor, αV/ß1 integrin, that is uniquely expressed by both chemosensitive and chemoresistant EOC cells with significantly higher expression in chemoresistant EOC cells. More importantly, we have demonstrated that monoclonal antibodies against αV/ß1 integrin induced cytotoxicity in EOC cells, but not in normal cells, that is also synergistic with conventional chemotherapies. Cytotoxicity of αV/ß1 antibodies is due to conformational changes in αV/ß1 integrin which prevents monomeric MPO binding to αV/ß1 integrin inhibiting the activation of MPO, leading to increased apoptosis. Since normal epithelial cells and macrophages lack monomeric MPO and αV/ß1 integrin system, targeting this unique MPO-dependent survival mechanism will selectively eliminate EOC cells and will be the target for developing specific ovarian cancer therapies.


Subject(s)
Ovarian Neoplasms , Receptors, Vitronectin , Female , Humans , Carcinoma, Ovarian Epithelial , Epithelial Cells/metabolism , Ovarian Neoplasms/metabolism , Peroxidase/metabolism , Receptors, Vitronectin/metabolism
8.
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.

9.
Article in English | MEDLINE | ID: mdl-36222785

ABSTRACT

BACKGROUND: The link between post-operative adhesion development and epigenetic modifications is important in understanding the mechanism behind their formation. The purpose of this study was to determine whether epigenetic differences exist between primary fibroblasts of normal peritoneum and adhesion tissues isolated from the same patient(s). METHODS: DNA from fibroblasts isolated from normal peritoneum and adhesion tissues was isolated using Qiagen's EZ1 Advanced Kit. Methylation patterns of genes were quantified and compared in both cell lines using the Infinium Human Methylation 27 Beadchip system. RESULTS: A total of 7364 genes had been found to manifest significantly different DNA methylation levels in adhesion fibroblasts as compared to normal peritoneal fibroblasts (p<0.01). A total of 1685 genes were found to have increased DNA methylation by 50% in adhesion compared to peritoneal fibroblasts, and were enriched in Gene Ontology categories, Glycoprotein, and Defense Response. Furthermore, 1287 genes were found to have decreased DNA methylation patterns with enriched Gene Ontology categories, "Homeobox", and Transcription Factor Activity in adhesion fibroblasts. CONCLUSIONS: Epigenetic differences in fibroblasts isolated from normal peritoneum and adhesion tissues were observed. Future studies focusing on the precise role of these genes in the development of post operative adhesions will allow us to more fully appreciate regulatory mechanisms leading to adhesion development, thereby establishing targets for therapeutic interventions to prevent or limit adhesion development.

10.
Reprod Sci ; 29(11): 3055-3077, 2022 11.
Article in English | MEDLINE | ID: mdl-34515982

ABSTRACT

Intraperitoneal adhesions complicate over half of abdominal-pelvic surgeries with immediate, short, and long-term sequelae of major healthcare concern. The pathogenesis of adhesion development is similar to the pathogenesis of wound healing in all tissues, which if unchecked result in production of fibrotic conditions. Given the similarities, we explore the published literature to highlight the similarities in the pathogenesis of intra-abdominal adhesion development (IPAD) and other fibrotic diseases such as keloids, endometriosis, uterine fibroids, bronchopulmonary dysplasia, and pulmonary, intraperitoneal, and retroperitoneal fibrosis. Following a literature search using PubMed database for all relevant English language articles up to November 2020, we reviewed relevant articles addressing the genetic and epidemiological similarities and differences in the pathogenesis and pathobiology of fibrotic diseases. We found genetic and epidemiological similarities and differences between the pathobiology of postoperative IPAD and other diseases that involve altered fibroblast-derived cells. We also found several genes and single nucleotide polymorphisms that are up- or downregulated and whose products directly or indirectly increase the propensity for postoperative adhesion development and other fibrotic diseases. An understanding of the similarities in pathophysiology of adhesion development and other fibrotic diseases contributes to a greater understanding of IPAD and these disease processes. At a very fundamental level, blocking changes in the expression or function of genes necessary for the transformation of normal to altered fibroblasts may curtail adhesion formation and other fibrotic disease since this is a prerequisite for their development. Similarly, applying measures to induce apoptosis of altered fibroblast may do the same; however, apoptosis should be at a desired level to simultaneously ameliorate development of fibrotic diseases while allowing for normal healing. Scientists may use such information to develop pharmacologic interventions for those most at risk for developing these fibrotic conditions.


Subject(s)
Endometriosis , Female , Humans , Endometriosis/metabolism , Fibroblasts/metabolism , Fibrosis , Tissue Adhesions/metabolism , Wound Healing
11.
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
12.
Reprod Sci ; 28(8): 2076-2086, 2021 08.
Article in English | MEDLINE | ID: mdl-33090376

ABSTRACT

Adhesions are permanent fibrovascular bands between peritoneal surfaces, which develop following virtually all body cavity surgeries. The susceptibility to develop, and the severity, of adhesions following intra-abdominal surgery varies within and between individuals, suggesting that heritable factors influence adhesion development. In this manuscript, we discuss the pathophysiology of adhesion development from the perspective of genetic susceptibility. We restrict our discussion to genes and single-nucleotide polymorphisms (SNPs) that are specifically involved in, or that cause modification of, the adhesion development process. We performed a literature search using the PubMed database for all relevant English language articles up to March 2020 (n = 186). We identified and carefully reviewed all relevant articles addressing genetic mutations or single-nucleotide polymorphisms (SNPs) that impact the risk for adhesion development. We also reviewed references from these articles for additional information. We found several reported SNPs, genetic mutations, and upregulation of messenger RNAs that directly or indirectly increase the propensity for postoperative adhesion development, namely in genes for transforming growth factor beta, vascular endothelial growth factor, interferon-gamma, matrix metalloproteinase, plasminogen activator inhibitor-1, and the interleukins. An understanding of genetic variants could provide insight into the pathophysiology of adhesion development. The information presented in this review contributes to a greater understanding of adhesion development at the genetic level and may allow modification of these genetic risks, which may subsequently guide management in preventing and treating this challenging complication of abdominal surgery. In particular, the information could help identify patients at greater risk for adhesion development, which would make them candidates for anti-adhesion prophylaxis. Currently, agents to reduce postoperative adhesion development exist, and in the future, development of agents, which specifically target individual genetic profile, would be more specific in preventing intraperitoneal adhesion development.


Subject(s)
Genetic Predisposition to Disease , Peritoneal Diseases/genetics , Polymorphism, Single Nucleotide , Postoperative Complications/genetics , Humans , Peritoneal Diseases/etiology , Tissue Adhesions/etiology , Tissue Adhesions/genetics
14.
Reprod Sci ; 27(10): 1873-1878, 2020 10.
Article in English | MEDLINE | ID: mdl-32617880

ABSTRACT

To investigate whether recombinant AMH (rAMH) is able to decrease cellular proliferation/apoptosis in luteinized granulosa cells (GCs) through hormonal regulation, a primary culture of GCs was established from GCs obtained at time of oocyte retrieval from follicular fluid of 3 patients. Cells were seeded in well cell culture plates at a density of 100,000 cells/well in medium and treated with rAMH 20 ng/ml (rAMH group), or phosphate-buffered saline (PBS-control group), for 24 h. Total RNA was extracted from all cells, followed by cDNA synthesis and real-time RT-PCR to quantify the expression levels of AMH, AMH-R2, FSH-R, inhibin B, cell proliferation (Ki67), and apoptosis (Caspase 3). We used independent sample t test (SPSS v25) and a p < 0.05 significance. Cellular expressions of AMH, AMH-R2, FSH-R, and inhibin B were reduced greater than 50% in the rAMH group, compared with that of the the control group (p ≤ 0.005 for all). Ki67 and Caspase3 were also reduced greater than 30% in the rAMH group (p ≤ 0.001 for both). Our findings show a direct inhibitory effect of AMH on luteinized GCs' expression of the major regulatory hormones, in addition to a significant decrease in markers of cell proliferation and apoptosis. These results confirm the inhibitory effects of AMH on follicular development.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Granulosa Cells/drug effects , Recombinant Proteins/pharmacology , Anti-Mullerian Hormone/metabolism , Caspase 3/metabolism , Female , Follicular Fluid , Granulosa Cells/cytology , Humans , Inhibins/metabolism , Oocyte Retrieval , Receptors, FSH/metabolism , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism
15.
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
16.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL