Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 39(7): 3469-3485, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31262871

ABSTRACT

BACKGROUND/AIM: Isothiocyanates (ITCs) are phytochemicals with potential cancer-preventative properties derived from the breakdown of glucosinolates that exist in cruciferous vegetables. Studies, to date, have demonstrated that various ITCs possess the ability to act as anticancer agents in different cancer types. This study investigated the anticancer properties of dietary ITCs (allyl-ITC, benzyl-ITC, phenylethyl-ITC) and synthetic (phenylbutyl-ITC and phenylhexyl-ITC) on liver and prostate carcinoma cells in vitro. MATERIALS AND METHODS: The effects of ITCs on cellular viability, migration, invasion, clonogenicity, apoptosis induction and reactive oxygen species generation were assessed in HepG2, DU145 and 22Rv1 cells. RESULTS: All ITCs reduced metabolic activity in each cell line with the most significant being phenylethyl-ITC. Both dietary and synthetic ITCs suppressed the migratory and invasive potential of all cell lines, inhibited colony-forming capability and induced apoptosis. Phenylethyl-ITC exposure resulted in the significant generation of reactive oxygen species. CONCLUSION: These data highlight the potential advantages of utilizing ITCs to delay the carcinogenic process and the potential for dietary and synthetic ITCs to act as anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Isothiocyanates/pharmacology , Liver Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Humans , Liver Neoplasms/metabolism , Male , Prostatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism
2.
Cancer Biol Ther ; 17(3): 262-71, 2016.
Article in English | MEDLINE | ID: mdl-26785864

ABSTRACT

Triple negative breast cancers (TNBCs) are highly aggressive and grow in response to sex steroid hormones despite lacking expression of the classical estrogen (E2) and progesterone (P4) receptors. Since P4 receptor membrane component 1 (PGRMC1) is expressed in breast cancer tumors and is known to mediate P4-induced cell survival, this study was designed to determine the expression of PGRMC1 in TNBC tumors and the involvement of PGRMC1 in regulating proliferation and survival of TNBC cells in vitro and the growth of TNBC tumors in vivo. For the latter studies, the MDA-MB-231 (MDA) cell line derived from TNBC was used. These cells express PGRMC1 but lack expression of the classical P4 receptor. A lentiviral-based shRNA approach was used to generate a stably transfected PGRMC1-deplete MDA line for comparison to the PGRMC1-intact MDA line. The present studies demonstrate that PGRMC1: 1) is expressed in TNBC cells; 2) mediates the ability of P4 to suppress TNBC cell mitosis in vitro; 3) is required for P4 to reduce the apoptotic effects of doxorubicin in vitro; and 4) facilitates TNBC tumor formation and growth in vivo. Taken together, these findings indicate that PGRMC1 plays an important role in regulating the growth and survival of TNBC cells in vitro and ultimately in the formation and development of these tumors in vivo. Thus, PGRMC1 may be a therapeutic target for TNBCs.


Subject(s)
Membrane Proteins/biosynthesis , Receptors, Progesterone/biosynthesis , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Survival/physiology , Female , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Receptors, Progesterone/metabolism , Transfection , Triple Negative Breast Neoplasms/pathology
3.
Cancer Lett ; 356(2 Pt B): 434-42, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25304370

ABSTRACT

Endometrial cancer is the leading gynecologic cancer in women in the United States with 52,630 women predicted to be diagnosed with the disease in 2014. The objective of this study was to determine if progesterone (P4) receptor membrane component 1 (PGRMC1) influenced endometrial cancer cell viability in response to chemotherapy in vitro and in vivo. A lentiviral-based shRNA knockdown approach was used to generate stable PGRMC1-intact and PGRMC1-deplete Ishikawa endometrial cancer cell lines that also lacked expression of the classical progesterone receptor (PGR). Progesterone treatment inhibited mitosis of PGRMC1-intact, but not PGRMC1-deplete cells, suggesting that PGRMC1 mediates the anti-mitotic actions of P4. To test the hypothesis that PGRMC1 attenuates chemotherapy-induced apoptosis, PGRMC1-intact and PGRMC1-deplete cells were treated in vitro with vehicle, P4 (1 µM), doxorubicin (Dox, 2 µg/ml), or P4 + Dox for 48 h. Doxorubicin treatment of PGRMC1-intact cells resulted in a significant increase in cell death; however, co-treatment with P4 significantly attenuated Dox-induced cell death. This response to P4 was lost in PGRMC1-deplete cells. To extend these observations in vivo, a xenograft model was employed where PGRMC1-intact and PGRMC1-deplete endometrial tumors were generated following subcutaneous and intraperitoneal inoculation of immunocompromised NOD/SCID and nude mice, respectively. Tumors derived from PGRMC1-deplete cells grew slower than tumors from PGRMC1-intact cells. Mice harboring endometrial tumors were then given three treatments of vehicle (1:1 cremophor EL: ethanol + 0.9% saline) or chemotherapy [Paclitaxel (15 mg/kg, i.p.) followed after an interval of 30 minutes by CARBOplatin (50 mg/kg)] at five day intervals. In response to chemotherapy, tumor volume decreased approximately four-fold more in PGRMC1-deplete tumors when compared with PGRMC1-intact control tumors, suggesting that PGRMC1 promotes tumor cell viability during chemotherapeutic stress. In sum, these in vitro and in vivo findings demonstrate that PGRMC1 plays a prominent role in the growth and chemoresistance of human endometrial tumors.


Subject(s)
Apoptosis , Cell Proliferation , Drug Resistance, Neoplasm , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/prevention & control , Membrane Proteins/metabolism , Receptors, Progesterone/metabolism , Animals , Blotting, Western , Endometrial Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mitosis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Eur J Cancer ; 49(8): 1845-59, 2013 May.
Article in English | MEDLINE | ID: mdl-23453937

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) accounts for 15-20% of breast cancers but is responsible for a disproportionate number of deaths. We investigated the relevance, in TNBC, of nano-sized exosomes expelled from cells. Specifically, we compared effects of exosomes derived from the claudin-low TNBC cell line Hs578T and its more invasive Hs578Ts(i)8 variant, as well as exosomes from TNBC patient sera compared to normal sera. METHODS: Exosomes were isolated from conditioned media (CM) of Hs578T and Hs578Ts(i)8 cells and from sera by filtration and ultracentrifugation. Successful isolation was confirmed by transmission electron microscopy and immunoblotting. Subsequent analysis, of secondary/recipient cells in response to exosomes, included proliferation; motility/migration; invasion; anoikis assays and endothelial tubule formation assays. RESULTS: Hs578Ts(i)8-exosomes versus Hs578T-exosomes significantly increased the proliferation, migration and invasion capacity of all three recipient cell lines evaluated i.e. SKBR3, MDA-MB-231 and HCC1954. Exosomes from Hs578Ts(i)8 cells also conferred increased invasiveness to parent Hs578T cells. Hs578Ts(i)8-exosomes increased sensitivity of SKBR3, MDA-MB-231 and HCC1954 to anoikis when compared to the effects of Hs578T-exosomes reflecting the fact that Hs578Ts(i)8 cells are themselves innately more sensitive to anoikis. In relation to vasculogenesis and subsequent angiogenesis, Hs578Ts(i)8-exosomes versus Hs578T-exosomes stimulated significantly more endothelial tubules formation. Finally, our pilot translational study showed that exosomes from TNBC patients' sera significantly increased recipient cells' invasion when compared to those derived from age- and gender-matched healthy control sera. CONCLUSION: This study supports the hypothesis that TNBC exosomes may be involved in cancer cell-to-cell communication, conferring phenotypic traits to secondary cells that reflect those of their cells of origin.


Subject(s)
Anoikis , Breast Neoplasms/physiopathology , Cell Movement , Cell Proliferation , Exosomes/physiology , Aged , Blood Vessels/metabolism , Blood Vessels/physiopathology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Exosomes/genetics , Exosomes/ultrastructure , Female , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Microscopy, Confocal , Microscopy, Electron, Transmission , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction
5.
PLoS One ; 6(11): e28077, 2011.
Article in English | MEDLINE | ID: mdl-22140510

ABSTRACT

BACKGROUND: Recent evidence links aberrant activation of Hedgehog (Hh) signaling with the pathogenesis of several cancers including medulloblastoma, basal cell, small cell lung, pancreatic, prostate and ovarian. This investigation was designed to determine if inhibition of this pathway could inhibit serous ovarian cancer growth. METHODOLOGY: We utilized an in vivo pre-clinical model of serous ovarian cancer to characterize the anti-tumor activity of Hh pathway inhibitors cyclopamine and a clinically applicable derivative, IPI-926. Primary human serous ovarian tumor tissue was used to generate tumor xenografts in mice that were subsequently treated with cyclopamine or IPI-926. PRINCIPAL FINDINGS: Both compounds demonstrated significant anti-tumor activity as single agents. When IPI-926 was used in combination with paclitaxel and carboplatinum (T/C), no synergistic effect was observed, though sustained treatment with IPI-926 after cessation of T/C continued to suppress tumor growth. Hh pathway activity was analyzed by RT-PCR to assess changes in Gli1 transcript levels. A single dose of IPI-926 inhibited mouse stromal Gli1 transcript levels at 24 hours with unchanged human intra-tumor Gli1 levels. Chronic IPI-926 therapy for 21 days, however, inhibited Hh signaling in both mouse stromal and human tumor cells. Expression data from the micro-dissected stroma in human serous ovarian tumors confirmed the presence of Gli1 transcript and a significant association between elevated Gli1 transcript levels and worsened survival. CONCLUSIONS/SIGNIFICANCE: IPI-926 treatment inhibits serous tumor growth suggesting the Hh signaling pathway contributes to the pathogenesis of ovarian cancer and may hold promise as a novel therapeutic target, especially in the maintenance setting.


Subject(s)
Hedgehog Proteins/metabolism , Ovarian Neoplasms/pathology , Signal Transduction , Xenograft Model Antitumor Assays , Animals , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/genetics , Humans , Maintenance Chemotherapy , Mice , Neoplasms, Cystic, Mucinous, and Serous/drug therapy , Neoplasms, Cystic, Mucinous, and Serous/genetics , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Survival Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Veratrum Alkaloids/pharmacology , Veratrum Alkaloids/therapeutic use , Zinc Finger Protein GLI1
6.
Methods Mol Biol ; 784: 55-75, 2011.
Article in English | MEDLINE | ID: mdl-21898213

ABSTRACT

Current literature provides significant evidence to support the concept that there are limited subpopulations of cells within a solid tumour that have increased tumour-initiating potential relative to the total tumour population. Such tumour-initiating cells have been identified in leukaemia and in a variety of solid tumours using different combinations of cell surface markers, suggesting that a tumour-initiating cell heterogeneity exists for each specific tumour. These studies have been extended to endometrial cancer; and herein we present several experimental approaches, both in vitro and in vivo, that can be used to determine whether such populations exist, and if so, to characterize them. These methods are adaptable to the investigation of tumour-initiating cells from other tumour types.


Subject(s)
Biomarkers, Tumor/genetics , Endometrial Neoplasms/genetics , Neoplastic Cells, Circulating/metabolism , Neoplastic Stem Cells/metabolism , Animals , Antigens, Surface/analysis , Bone Marrow/metabolism , DNA, Neoplasm/isolation & purification , Female , Gene Expression Regulation, Neoplastic , Humans , Leukocytes, Mononuclear/cytology , Mice , Mice, SCID
7.
Clin Chem ; 57(1): 18-32, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21059829

ABSTRACT

BACKGROUND: Successful treatment of breast cancer is enhanced by early detection and, if possible, subsequent patient-tailored therapy. Toward this goal, it is essential to identify and understand the most relevant panels of biomarkers, some of which may also have relevance as therapeutic targets. METHODS: We critically reviewed published literature on microRNAs (miRNAs) as relevant to breast cancer. SUMMARY: Since the initial recognition of the association of miRNAs with breast cancer in 2005, studies involving cell lines, in vivo models, and clinical specimens have implicated several functions for miRNAs, including suppressing oncogenesis and tumors, promoting or inhibiting metastasis, and increasing sensitivity or resistance to chemotherapy and targeted agents in breast cancer. For example, miR-21 is overexpressed in both male and female breast tumors compared with normal breast tissue and has been associated with advanced stage, lymph node positivity, and reduced survival time. miR-21 knock-down in cell-line models has been associated with increased sensitivity to topotecan and taxol in vitro and the limitation of lung metastasis in vivo. Furthermore, the discovery of extracellular miRNAs (including miR-21), existing either freely or in exosomes in the systemic circulation, has led to the possibility that such molecules may serve as biomarkers for ongoing patient monitoring. Although additional investigations are necessary to fully exploit the use of miRNAs in breast cancer, there is increasing evidence that miRNAs have potential not only to facilitate the determination of diagnosis and prognosis and the prediction of response to treatment, but also to act as therapeutic targets and replacement therapies.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/metabolism , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms, Male/diagnosis , Breast Neoplasms, Male/drug therapy , Breast Neoplasms, Male/genetics , Drug Resistance, Neoplasm , Extracellular Space/metabolism , Female , Humans , Intracellular Space/metabolism , Male , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Oncogenes , Prognosis
8.
Reprod Biol Endocrinol ; 8: 147, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21122138

ABSTRACT

BACKGROUND: Recent data provide significant evidence to support the hypothesis that there are sub-populations of cells within solid tumors that have an increased tumor initiating potential relative to the total tumor population. CD133, a cell surface marker expressed on primitive cells of neural, hematopoietic, endothelial and epithelial lineages has been identified as a marker for tumor initiating cells in solid tumors of the brain, colon, pancreas, ovary and endometrium. Our objectives were to assess the relative level of CD133 expressing cells in primary human endometrial tumors, confirm their tumorigenic potential, and determine whether CD133 expression was epigenetically modified. METHODS: We assessed CD133 expression in primary human endometrial tumors by flow cytometry and analyzed the relative tumorigenicity of CD133+ and CD133- cells in an in vivo NOD/SCID mouse model. We assessed potential changes in CD133 expression over the course of serial transplantation by immunofluorescence and flow cytometry. We further examined CD133 promoter methylation and expression in normal endometrium and malignant tumors. RESULTS: As determined by flow cytometric analysis, the percentage of CD133+ cells in primary human endometrial cancer samples ranged from 5.7% to 27.4%. In addition, we confirmed the tumor initiating potential of CD133+ and CD133- cell fractions in NOD/SCID mice. Interestingly, the percentage of CD133+ cells in human endometrial tumor xenografts, as evidenced by immunofluorescence, increased with serial transplantation although this trend was not consistently detected by flow cytometry. We also determined that the relative levels of CD133 increased in endometrial cancer cell lines following treatment with 5-aza-2'-deoxycytidine suggesting a role for methylation in the regulation of CD133. To support this finding, we demonstrated that regions of the CD133 promoter were hypomethylated in malignant endometrial tissue relative to benign control endometrial tissue. Lastly, we determined that methylation of the CD133 promoter decreases over serial transplantation of an endometrial tumor xenograft. CONCLUSIONS: These findings support the hypotheses that CD133 expression in endometrial cancer may be epigenetically regulated and that cell fractions enriched for CD133+ cells may well contribute to endometrial cancer tumorigenicity, pathology and recurrence.


Subject(s)
Antigens, CD/genetics , Cell Transformation, Neoplastic/pathology , Endometrial Neoplasms/pathology , Epigenomics , Glycoproteins/genetics , Neoplastic Stem Cells/pathology , Peptides/genetics , AC133 Antigen , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Decitabine , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/metabolism , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplastic Stem Cells/immunology
9.
Front Biosci (Elite Ed) ; 2(3): 882-905, 2010 06 01.
Article in English | MEDLINE | ID: mdl-20515761

ABSTRACT

Uterine tumors, whether benign or malignant, are diagnosed in a significant portion of women and are associated with a number of co-morbidities that negatively impact quality of life. Uterine tumors can be derived from the epithelial (endometrial hyperplasia or carcinoma) and mesenchymal (leiomyoma, sarcoma) layers of the uterus. The exact etiologies of the various tumor types are yet to be defined. Collectively their development and progression often results from aberrant steroid hormone exposure or dysregulation of related growth factor signaling and apoptotic pathways, reflecting the role of steroid hormone-dependent signaling and survival pathways in the cycles of cell growth and involution that characterize normal uterine physiology. While molecular analyses of human tumors can identify candidate genetic and epigenetic lesions contributing to uterine tumor initiation and progression, in vivo genetic models are needed to establish the functional significance of such lesions and their contribution to tumorigenesis. For this purpose, genetically-engineered mouse models have proven valuable. Here we review genetically-modified mouse models that develop uterine tumors and compare their pathology, utility/feasibility, and discuss their clinical relevance.


Subject(s)
Disease Models, Animal , Uterine Neoplasms/pathology , Animals , Female , Mice
10.
Breast Cancer Res Treat ; 123(3): 613-25, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20549336

ABSTRACT

Early detection of cancer is vital to improved overall survival rates. At present, evidence is accumulating for the clinical value of detecting occult tumor cells in peripheral blood, plasma, and serum specimens from cancer patients. Both molecular and cellular approaches, which differ in sensitivity and specificity, have been used for such means. Circulating tumor cells and extracellular nucleic acids have been detected within blood, plasma, and sera of cancer patients. As the presence of malignant tumors are clinically determined and/or confirmed upon biopsy procurement-which in itself may have detrimental effects in terms of stimulating cancer progression/metastases-minimally invasive methods would be highly advantageous to the diagnosis and prognosis of breast cancer and the subsequent tailoring of targeted treatments for individuals, if reliable panels of biomarkers suitable for such an approach exist. Herein, we review the current advances made in the detection of such circulating tumor cells and nucleic acids, with particular emphasis on extracellular nucleic acids, specifically extracellular mRNAs and discuss their clinical relevance.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/diagnosis , Exosomes/metabolism , Genetic Markers , Neoplastic Cells, Circulating/metabolism , Nucleic Acids/blood , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Early Detection of Cancer , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/blood , Neoplastic Cells, Circulating/pathology , Predictive Value of Tests , Prognosis , RNA, Messenger/blood
11.
Stem Cells ; 27(12): 2875-83, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19816957

ABSTRACT

Evidence is accumulating that solid tumors contain a rare phenotypically distinct population of cells, termed cancer stem cells (CSC), which give rise to and maintain the bulk of the tumor. These CSC are thought to be resistant to current chemotherapeutic strategies due to their intrinsic stem-like properties and thus may provide the principal driving force behind recurrent tumor growth. Given the high frequency of recurrent metastasis associated with human ovarian cancer, we sought to determine whether primary human ovarian tumors contain populations of cells with enhanced tumor-initiating capacity, a characteristic of CSC. Using an in vivo serial transplantation model, we show that primary uncultured human ovarian tumors can be reliably propagated in NOD/SCID mice, generating heterogeneous tumors that maintain the histological integrity of the parental tumor. The observed frequency of tumor engraftment suggests only certain subpopulations of ovarian tumor cells have the capacity to recapitulate tumor growth. Further profiling of human ovarian tumors for expression of candidate CSC surface markers indicated consistent expression of CD133. To determine whether CD133 expression could define a tumor-initiating cell population in primary human ovarian tumors, fluorescence-activated cell sorting (FACS) methods were employed. Injection of sorted CD133(+) and CD133(-) cell populations into NOD/SCID mice established that tumor-derived CD133(+) cells have an increased tumorigenic capacity and are capable of recapitulating the original heterogeneous tumor. Our data indicate that CD133 expression defines a NOD/SCID tumor initiating subpopulation of cells in human ovarian cancer that may be an important target for new chemotherapeutic strategies aimed at eliminating ovarian cancer.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peptides/metabolism , AC133 Antigen , Animals , Biomarkers, Tumor/metabolism , Cell Count , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Xenograft Model Antitumor Assays
12.
Cancer Res ; 69(21): 8241-8, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19843861

ABSTRACT

Emerging evidence indicates that the highly regenerative human endometrium harbors rare populations of epithelial progenitor cells. In tumors of other regenerative epithelial tissues, rare cancer stem cells (CSC) have been identified that may have originated from normal epithelial stem/progenitor cells. We hypothesized that CSC are responsible for epithelial neoplasia associated with endometrial carcinoma, the most common gynecologic malignancy in women. Stem cell characteristics of single cells isolated from endometrial carcinoma tissues from women ages 62 +/- 11.8 years (n = 34) were assessed. Twenty-five of 28 endometrial carcinoma samples contained a small population of clonogenic cells [0.24% (0-1.84%)], with no significant difference in cloning efficiency between the three grades of endometrial carcinoma or between endometrial carcinoma and normal endometrial epithelial samples. Isolated endometrial carcinoma cells transplanted under the kidney capsule of immunocompromised mice in serial dilution (2 x 10(6)-1 x 10(4) cells) generated tumors in 8 of 9 samples with morphologies similar to the parent tumors. These tumors recapitulated cytokeratin, vimentin, estrogen receptor-alpha, and progesterone receptor expression of the parent tumor, indicating that tumor-initiating cells likely differentiated into cells comprising the endometrial carcinoma tissue. Individual clones underwent serial clonal subculture 2.5 to 4 times, with a trend of increasing number of subclonings with increasing tumor grade, indicating increasing self-renewal with greater malignancy. Clonally derived endometrial carcinoma cells also expressed the self-renewal genes BMI-1, NANOG, and SOX-2. Isolated cells from primary tumors were serially transplanted 3 to 5 times in nonobese diabetic/severe combined immunodeficient mice, showing self-renewal in vivo. This evidence of cells with clonogenic, self-renewing, differentiating, and tumorigenic properties suggests that a CSC population may be responsible for production of endometrial carcinoma tumor cells.


Subject(s)
Endometrial Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Lineage , Colony-Forming Units Assay , Endometrial Neoplasms/metabolism , Female , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/transplantation , Polymerase Chain Reaction , RNA, Neoplasm/isolation & purification , Transplantation, Heterologous , Tumor Cells, Cultured
13.
Hypertens Pregnancy ; 27(2): 169-81, 2008.
Article in English | MEDLINE | ID: mdl-18484422

ABSTRACT

OBJECTIVES: Preeclampsia is characterised by an abnormal vascular response to placentation and is associated with increased systemic vascular resistance and endothelial cell dysfunction. This study investigated the mRNA and protein expression of the beta(2) and beta(3)-adrenoceptors (beta-ARs) in placenta, and umbilical arteries, from preeclamptic and normotensive patients, to determine if the presence of preeclampsia altered the expression of either receptor. METHODS: RT-PCR was used to identify beta(2)-AR and beta(3)-AR mRNA transcripts in the human placenta and in human umbilical arteries. Real-time RT-PCR was performed on total RNA from normal and preeclamptic placentae and umbilical arteries. Western blotting using antibodies for beta(2)-AR, beta(3)-AR, and beta-actin was performed on total protein isolated from preeclamptic and normotensive placentae. RESULTS: There was no significant difference in mRNA expression levels of beta(2)-AR and beta(3)-AR between normal and preeclamptic tissues (p > 0.05). No significant difference was observed in protein levels of beta(2)-AR and beta(3)-AR between placentae from normal and preeclamptic patients (p > 0.05). CONCLUSIONS: Aberrations in the beta-adrenoceptor signalling systems, rather than in the regulation of expression of these receptors may occur in preeclampsia, as is the case in other hypertensive disorders.


Subject(s)
Placenta/metabolism , Pre-Eclampsia/metabolism , Receptors, Adrenergic, beta/biosynthesis , Umbilical Arteries/metabolism , Adult , Blotting, Western , Female , Gene Expression , Gene Expression Regulation , Humans , Pregnancy , Receptors, Adrenergic, beta-2/biosynthesis , Receptors, Adrenergic, beta-3/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
14.
Cell Cycle ; 7(2): 242-9, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18256549

ABSTRACT

Recent data suggest that rare stem cell populations with the capacity to self renew and drive tumor formation are a feature of solid tumors. Several investigators have identified putative stem cells from solid tumors and cancer cell lines following isolation of a side population (SP) defined by dye exclusion. We investigated this parameter in our efforts to identify an endometrial cancer (EnCa) stem cell population. Multiple EnCa cell lines were assessed and verapamil sensitive SP and non-SP cells were isolated from two human EnCa cell lines. The functional significance of the SP and non-SP derived from AN3CA was evaluated in vitro and in vivo. SP cells proliferated at a significantly slower rate than the non-SP fraction, and a larger proportion of the SP cells were in the G(1) phase of the cell cycle as compared to the non-SP fraction. The SP fraction was more resistant to the chemotherapeutic agent paclitaxel. The SP comprised -0.02% of the initial AN3CA cell population and this proportion of SP cells was maintained within the larger heterogeneous population following repeated passages of purified SP cells. These findings suggest that SP cells derived from the An3CA cell line have the stem cell properties of low proliferative activity, chemoresistance and self-renewal. We also tested relative tumor formation activity of the SP and non-SP fractions. Only the SP fraction was tumorigenic. Additionally, we identified SP fractions in primary EnCa. Together these results are consistent with the hypothesis that EnCa contain a subpopulation of tumor initiating cells with stem like properties.


Subject(s)
Endometrial Neoplasms/pathology , Neoplastic Stem Cells/cytology , Animals , Cell Line, Tumor , Cell Separation , Cell Transformation, Neoplastic , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Transplantation, Heterologous
15.
Reprod Sci ; 15(2): 179-88, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18089586

ABSTRACT

Preeclampsia is characterized by intense and prolonged vasoconstriction. Rho A-mediated calcium sensitization is central to prolonged contractility of vascular smooth muscle. The aims of this study are (1) to investigate mRNA expression levels of Rho A/Rho kinases in placental tissues from normotensive and preeclamptic women and (2) to investigate the effects of 2 isoprostanes, 8-iso prostaglandin F(2)( alpha) (8-iso PGF(2 alpha) ) and 8-iso prostaglandin E(2) (8-iso PGE(2)), on small placental and myometrial vessel resistance and to determine if their effects were mediated via the Rho kinase pathway. Real-time reverse transcription polymerase chain reaction for Rho A, ROCK I, and ROCK II was performed on total RNA from normotensive and preeclamptic placentae. The effects of 8- iso PGF(2 alpha) and 8-iso PGE(2) (alone and with the specific Rho kinase inhibitor Y-27632) on placental and myometrial vessels (<400 microm) were measured and compared with control recordings. Rho A mRNA expression levels were significantly higher in placentae from preeclamptic women than in placentae from normotensive women (P < .01). There was no significant difference in expression levels of ROCK I and ROCK II between both tissue types (P > .05). Both isoprostanes exerted a significant concentration-dependent vasocontractile effect on both vessel types (P < .001). This effect was antagonized by Y-27632 in placental arteries but not in myometrial arteries. Increased Rho A mRNA expression in placentae from preeclamptic women is suggestive of a role for the Rho kinase pathway in the modulation of the placental vasculature in this condition. Isoprostanes exert their vasocontractile effect, in placental vasculature, in part via the Rho kinase pathway.


Subject(s)
Isoprostanes/pharmacology , Myometrium/blood supply , Placenta/blood supply , Pre-Eclampsia/physiopathology , Vasoconstriction/drug effects , rho-Associated Kinases/biosynthesis , Dinoprost/analogs & derivatives , Dinoprost/antagonists & inhibitors , Dinoprost/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/antagonists & inhibitors , Dinoprostone/pharmacology , Female , Humans , Isoprostanes/antagonists & inhibitors , Pre-Eclampsia/metabolism , Pregnancy , RNA, Messenger/biosynthesis , Signal Transduction , rho-Associated Kinases/antagonists & inhibitors
16.
Cancer Biol Ther ; 7(2): 180-88, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18059193

ABSTRACT

Cables 1, a cyclin-dependent kinase binding protein, is primarily involved in cell cycle regulation. Loss of nuclear Cables 1 expression is observed in human colon, lung and endometrial cancers. We previously reported that loss of nuclear Cables 1 expression was also observed with high frequency in a limited sample set of human ovarian carcinomas, although the mechanisms underlying loss of nuclear Cables 1 expression remained unknown. Our present objective was to examine Cables 1 expression in ovarian cancer in greater detail, and determine the predominant mechanisms of Cables 1 loss. We assessed potential genetic and epigenetic modifications of the Cables 1 locus through analyses of mutation, polymorphisms, loss of heterozygosity and DNA methylation. We observed a marked loss of nuclear Cables 1 expression in serous and endometrioid ovarian carcinomas that correlated with decreased Cables 1 mRNA levels. Although we detected no Cables 1 mutations, there was evidence of LOH at the Cables 1 locus and epigenetic modification of the Cables 1 promoter region in a subset of ovarian carcinomas and established cancer cell lines. From a functional perspective, over-expression of Cables 1 induced apoptosis, whereas, knockdown of Cables 1 negated this effect. Together these findings suggest that multiple mechanisms underlie the loss of Cables 1 expression in ovarian cancer cells, supporting the hypothesis that Cables 1 is a tumor suppressor in human ovarian cancer.


Subject(s)
Carrier Proteins/genetics , Cyclins/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Loss of Heterozygosity , Ovarian Neoplasms/genetics , Phosphoproteins/genetics , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Chromosomes, Human, Pair 18 , DNA Methylation , Epigenesis, Genetic , Female , Humans , Immunohistochemistry , Ovarian Neoplasms/classification , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Promoter Regions, Genetic , Sequence Analysis, DNA , Tumor Suppressor Proteins
17.
Cell Cycle ; 6(21): 2678-84, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17912041

ABSTRACT

Recent studies have shown that cell cycle inhibitors encoded by the Ink4a gene locus constrain the self-renewing activity of adult stem cells of the hematopoietic and nervous systems. Here we report that knockout (KO) of the Cables1 [cyclin-dependent kinase (CDK)-5 and ABL enzyme substrate 1] cell cycle-regulatory gene in mice has minimal to no effect on hematopoietic stem cell (HSC) dynamics. However, female Cables1-null mice exhibit a significant expansion of germ cell (oocyte) numbers throughout adulthood. This is accompanied by a dramatic elevation in the number of atretic immature oocytes within the ovaries and an increase in the incidence of degenerating oocytes retrieved following superovulation of CABLES1-deficient females. These outcomes are not observed in mice lacking p16INK4a alone or both p16INK4a and p19ARF. These data support recent reports that adult female mice can generate new oocytes and follicles but the enhancement of postnatal oogenesis by Cables1 KO appears offset by a reduction in oocyte quality, as reflected by increased elimination of these additional germ cells via apoptosis. This work also reveals cell lineage specificity with respect to the role that specific CDK-interacting proteins play in restraining the activity of adult germline versus somatic stem cells.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle/physiology , Cyclin-Dependent Kinases/metabolism , Cyclins/deficiency , Cyclins/metabolism , Growth Inhibitors/deficiency , Growth Inhibitors/metabolism , Oocytes/growth & development , Oocytes/metabolism , Phosphoproteins/deficiency , Phosphoproteins/metabolism , Age Factors , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Count , Cyclin-Dependent Kinases/deficiency , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Cyclins/physiology , Female , Germ Cells/cytology , Germ Cells/growth & development , Growth Inhibitors/genetics , Growth Inhibitors/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Ovum/cytology , Ovum/growth & development , Ovum/metabolism , Phosphoproteins/genetics , Phosphoproteins/physiology
18.
Am J Obstet Gynecol ; 196(2): 169.e1-6, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17306668

ABSTRACT

OBJECTIVE: Little is known about the presence or functional effects of protease-activated receptor subtypes in human uterine tissues. The aims of this study were as follows: (1) to investigate for protease-activated receptor-4 messenger RNA and protein expression in human myometrium, (2) to evaluate the effects of a specific protease-activated receptor-4 activating peptide (AYPGKF-NH2) on spontaneous human myometrial contractility in vitro, and (3) to examine the effects of a protease-activated receptor-4 antagonist (tcYPGKF-NH2) on thrombin-mediated uterine contractility. STUDY DESIGN: Reverse transcriptase-polymerase chain reaction and Immunofluorescence studies were used to investigate for protease-activated receptor-4 messenger RNA and protein expression, respectively. Isometric tension recordings were used to examine the functional effects on contractility. RESULTS: Reverse transcriptase-polymerase chain reaction demonstrated messenger RNA expression for protease-activated receptor-4 in pregnant and non-pregnant myometrium. Immunofluorescence confocal microscopy demonstrated the presence of protease-activated receptor-4 protein in myometrial cells. With the use of isometric recordings, protease-activated receptor 4-activating peptide elicited a stimulatory effect on spontaneous human pregnant myometrial contractility (13.1% +/- 2.7 SEM; n = 6; P < .05). Protease-activated receptor-4 antagonism alone elicited a significant uterorelaxant effect (14.7% +/- 2.4; n = 6; P < .05). The observed thrombin-mediated uterotonic effect was similar in the absence (46.1% +/- 12.8; n = 6) and presence (48.8% +/- 12.6; n = 6) of the protease-activated receptor-4 antagonist (P = .91). CONCLUSIONS: This study outlines protease-activated receptor-4 messenger RNA and protein expression in human myometrium. Protease-activated receptor-4 activation exerts a mild uterotonic effect, whereas protease-activated receptor-4 antagonism results in a mild uterorelaxant effect. The potent human uterotonic effect of thrombin is not apparently mediated to any great extent by protease-activated receptor-4.


Subject(s)
Myometrium/physiology , Oligopeptides/physiology , Receptors, Proteinase-Activated/biosynthesis , Uterine Contraction/physiology , Adult , Female , Humans , In Vitro Techniques , Oligopeptides/biosynthesis , Pregnancy , Receptors, Proteinase-Activated/physiology
19.
Reproduction ; 132(1): 169-76, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16816342

ABSTRACT

Pre eclampsia represents a state of increased or prolonged vasoconstriction, partially linked to the potent vasocontractile effect of isoprostanes. The process of Rho A-mediated calcium sensitisation is inherent to a state of prolonged contractility in many smooth muscle types. The aim of this study was (1) to investigate mRNA expression levels of Rho A and Rho kinase isoforms (I and II) in the umbilical artery from normotensive and pre eclamptic women and (2) to determine whether the effects of two isoprostanes, 8-iso prostaglandin F(2alpha) (8-iso PGF2alpha) and 8-iso prostaglandin E(2) (8-iso PGE(2)), on umbilical artery tone, were mediated via the Rho kinase pathway. Real-time RT-PCR using primers for Rho A, ROCK I and ROCK II was performed on total RNA isolated from umbilical artery specimens obtained from normotensive and pre eclamptic women. The effects of both isoprostanes (n = 6) (in the absence and presence of the specific Rho kinase inhibitor Y-27632), on umbilical artery tone were measured, and compared with control recordings. Rho A mRNA expression levels were significantly lower in umbilical artery samples obtained from pre eclamptic women (n = 4) in comparison to those from normotensive women (n = 6) (P < 0.05). ROCK I and ROCK II mRNA levels were similar in both vessel types (P > 0.05). Both isoprostanes exerted a significant concentration-dependent vasocontractile effect (n = 7) (P < 0.001) on umbilical artery. For 8-iso PGE(2), this effect was antagonised by Y-27632 (n = 6) (P < 0.01). The significant reduction of Rho A mRNA levels in umbilical arteries from pregnancies complicated by pre eclampsia may serve to counteract the diminished perfusion associated with the pathophysiology of pre eclampsia. The vasocontractile effect of 8-iso PGE(2) in pre eclampsia may in part be mediated via the Rho kinase pathway.


Subject(s)
Acute-Phase Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Muscle, Smooth, Vascular/enzymology , Pre-Eclampsia/enzymology , Protein Isoforms/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/analysis , Amides/pharmacology , Analysis of Variance , Case-Control Studies , Dinoprost/analogs & derivatives , Dinoprost/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Female , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Isoprostanes/pharmacology , Muscle, Smooth, Vascular/drug effects , Pregnancy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Culture Techniques , Umbilical Arteries , Vasoconstrictor Agents/pharmacology , rho-Associated Kinases
20.
BJOG ; 112(8): 1034-42, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16045514

ABSTRACT

OBJECTIVE: PGF(2alpha) acts through its receptor, FP, as an important smooth muscle contractile agent. The aim of this study was to investigate the effects of specific FP antagonism, using the novel-specific FP non-competitive antagonist THG113.31, on spontaneous and agonist-elicited contractions in pregnant and non-pregnant human myometrium in vitro. DESIGN: Scientific study. SETTING: University hospital and laboratories. Population Women undergoing caesarean section or hysterectomy. METHODS: Biopsies of human myometrium were obtained at elective caesarean section (n= 22) and from hysterectomy specimens from premenopausal women (n= 8). Dissected strips were mounted in tissue baths under physiological conditions. The effects of THG113.31 on spontaneous and oxytocin-induced contractions, in pregnant myometrium, and on phenylephrine-induced contractions, in non-pregnant myometrium, were measured. The effects of PGF(2alpha) on spontaneous contractions, in pregnant tissue, in the presence and absence of THG113.31, were investigated. The integrals of contractile activity measured were compared with those from simultaneously run control experiments. The pD(2) and mean maximal effect observed for THG113.31, and for PGF(2alpha) in the presence and absence of THG113.31, were calculated. MAIN OUTCOME MEASURES: Changes in contractility. RESULTS: THG113.31 exerted a potent relaxant effect in both spontaneous and oxytocin-induced contractility in pregnant tissue (P < 0.001), and phenylephrine-induced contractility in non-pregnant tissue (P < 0.001), compared with control experiments. PGF(2alpha) exerted a significant contractile effect on spontaneous contractions in pregnant tissue and this effect was not significantly attenuated by THG113.31 (P > 0.05). CONCLUSION: THG113.31 exerted a significant relaxant effect on human spontaneous and oxytocin-induced contractility but did not alter PGF(2alpha)-elicited contractility. These data raise questions about the exact mechanism of effect of THG113.31 and its interaction with FP. The uterorelaxant potency of THG113.31 in human myometrium in vitro indicates that it may be of limited use as a tocolytic compound.


Subject(s)
Phenylephrine/pharmacology , Receptors, Prostaglandin/antagonists & inhibitors , Uterine Contraction/drug effects , Adult , Cesarean Section , Dose-Response Relationship, Drug , Female , Humans , Hysterectomy , In Vitro Techniques , Middle Aged , Oxytocin/pharmacology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...