Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 107(44): 18874-9, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20952655

ABSTRACT

Cancer stem cells are proposed to be tumor-initiating cells capable of tumorigenesis, recurrence, metastasis, and drug resistance, and, like somatic stem cells, are thought to be capable of unlimited self-renewal and, when stimulated, proliferation and differentiation. Here we select cells by expression of a panel of markers to enrich for a population with stem cell-like characteristics. A panel of eight was initially selected from 95 human cell surface antigens as each was shared among human ovarian primary cancers, ovarian cancer cell lines, and normal fimbria. A total of 150 combinations of markers were reduced to a panel of three--CD44, CD24, and Epcam--which selected, in three ovarian cancer cell lines, those cells which best formed colonies. Cells expressing CD44, CD24, and Epcam exhibited stem cell characteristics of shorter tumor-free intervals in vivo after limiting dilution, and enhanced migration in invasion assays in vitro. Also, doxorubicin, cisplatin, and paclitaxel increased this enriched population which, conversely, was significantly inhibited by Müllerian inhibiting substance (MIS) or the MIS mimetic SP600125. These findings demonstrate that flow cytometry can be used to detect a population which shows differential drug sensitivity, and imply that treatment of patients can be individualized to target both stem/progenitor cell enriched and nonenriched subpopulations. The findings also suggest that this population, amenable to isolation by flow cytometry, can be used to screen for novel treatment paradigms, including biologic agents such as MIS, which will improve outcomes for patients with ovarian cancer.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , CD24 Antigen/metabolism , Cell Adhesion Molecules/metabolism , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Anthracenes/pharmacology , Anti-Mullerian Hormone/agonists , Antineoplastic Agents/agonists , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Epithelial Cell Adhesion Molecule , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans
2.
Reprod Sci ; 17(2): 158-67, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19805552

ABSTRACT

Uterine leiomyomas (also known as uterine fibroids) are the most common benign tumors of female reproductive tract and are the single most common indication for hysterectomies. Despite their high prevalence, the exact pathogenesis of these benign tumors is still unknown. One possible mechanism for leiomyoma formation is dysregulation of mesenchymal stem cell activity. Mesenchymal stem cells have been identified in both human and murine uteri and cancer stem cells have been identified in female reproductive malignancies. We compared stem/progenitor cell characteristics in both normal myometrium and the corresponding leiomyoma of patient's undergoing hysterectomies. We found that leiomyoma cells form fewer mesenchymal stem cell colonies and exhibit less Hoechst dye-excluding side population (SP) activity, which is a function associated with progenitor cells in other tissues, than cells isolated from normal myometrium. Whereas in normal myometrium, we observed heterogeneous expression of CD90, a cell surface marker associated the with differentiation potential of uterine fibroblasts, in leiomyomas, we observed homogenous expression of CD90, suggesting leiomyoma cells are more terminally differentiated. Furthermore, we found that while leiomyoma cells could only produce CD90 expressing cells, both CD90+ and CD90- myometrial cells could reestablish their original heterogeneous CD90 profile when expanded in vitro. These results suggest that normal myometrium contains cells with stem/progenitor cell activities that are absent in leiomyomas.


Subject(s)
Leiomyomatosis/pathology , Mesenchymal Stem Cells/pathology , Myometrium/cytology , Uterine Neoplasms/pathology , Cell Differentiation/physiology , Cell Division/physiology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Humans , In Vitro Techniques , Mesenchymal Stem Cells/metabolism , Thy-1 Antigens/metabolism
3.
Proc Natl Acad Sci U S A ; 105(34): 12469-73, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18711140

ABSTRACT

Ovulation induces cyclic rupture and regenerative repair of the ovarian coelomic epithelium. This process of repeated disruption and repair accompanied by complex remodeling typifies a somatic stem/progenitor cell-mediated process. Using BrdU incorporation and doxycycline inducible histone2B-green fluorescent protein pulse-chase techniques, we identify a label-retaining cell population in the coelomic epithelium of the adult mouse ovary as candidate somatic stem/progenitor cells. The identified population exhibits quiescence with asymmetric label retention, functional response to estrous cycling in vivo by proliferation, enhanced growth characteristics by in vitro colony formation, and cytoprotective mechanisms by enrichment for the side population. Together, these characteristics identify the label-retaining cell population as a candidate for the putative somatic stem/progenitor cells of the coelomic epithelium of the mouse ovary.


Subject(s)
Epithelial Cells/cytology , Ovary/cytology , Regeneration , Stem Cells/cytology , Animals , Bromodeoxyuridine , Cell Proliferation , Estrous Cycle , Female , Green Fluorescent Proteins , Mice
4.
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
5.
Stem Cells ; 25(5): 1317-25, 2007 May.
Article in English | MEDLINE | ID: mdl-17289934

ABSTRACT

Conditional deletion of beta-catenin in the Müllerian duct mesenchyme results in a degenerative uterus characterized by replacement of the myometrial smooth muscle with adipose tissue. We hypothesized that the mouse myometrium houses somatic smooth muscle progenitor cells that are hormonally responsive and necessary for remodeling and regeneration during estrous cycling and pregnancy. We surmise that the phenotype observed in beta-catenin conditionally deleted mice is the result of dysregulation of these progenitor cells. The objective of this study was to identify the mouse myometrial smooth muscle progenitor cell and its niche, define the surface marker phenotype, and show a functional response of these cells to normal myometrial cycling. Uteri were labeled with 5-bromo-2'-deoxyuridine (BrdU) and chased for up to 14 weeks. Myometrial label-retaining cells (LRCs) were observed in the myometrium and stroma throughout the chase period. After 12 weeks, phenotypic analysis of the LRCs by immunofluorescence demonstrated that the majority of LRCs colocalized with alpha-smooth muscle actin, estrogen receptor-alpha, and beta-catenin. Flow cytometry of myometrial cells identified a myometrial Hoechst 33342 effluxing "side population" that expresses MISRII-Cre-driven YFP. Functional response of LRCs was investigated by human chorionic gonadotropin stimulation of week 12 chase mice and demonstrated sequential proliferation of LRCs in the endometrial stroma, followed by the myometrium. These results suggest that conventional myometrial regeneration and repair is executed by hormonally responsive stem or progenitor cells derived from the Müllerian duct mesenchyme. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Aging/physiology , Bromodeoxyuridine/metabolism , Chorionic Gonadotropin/pharmacology , Myometrium/cytology , Myometrium/drug effects , Adipocytes/cytology , Adipocytes/drug effects , Animals , Bacterial Proteins/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Proliferation/drug effects , Female , Humans , Integrases/metabolism , Luminescent Proteins/metabolism , Mice , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Phenotype , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta , Stromal Cells/cytology , Stromal Cells/drug effects , beta Catenin/deficiency
6.
Proc Natl Acad Sci U S A ; 103(30): 11154-9, 2006 Jul 25.
Article in English | MEDLINE | ID: mdl-16849428

ABSTRACT

The recent identification of "side population" (SP) cells in a number of unrelated human cancers and their normal tissue sources has renewed interest in the hypothesis that cancers may arise from somatic stem/progenitor cells. The high incidence of recurrence attributable to multidrug resistance and the multiple histologic phenotypes indicative of multipotency suggests a stem cell-like etiology of ovarian cancer. Here we identify and characterize SP cells from two distinct genetically engineered mouse ovarian cancer cell lines. Differential efflux of the DNA-binding dye Hoechst 33342 from these cell lines defined a human breast cancer-resistance protein 1-expressing, verapamil-sensitive SP of candidate cancer stem cells. In vivo, mouse SP cells formed measurable tumors sooner than non-SP (NSP) cells when equal numbers were injected into the dorsal fat pad of nude mice. The presence of Mullerian Inhibiting Substance (MIS) signaling pathway transduction molecules in both SP and NSP mouse cells led us to investigate the efficacy of MIS against these populations in comparison with traditional chemotherapies. MIS inhibited the proliferation of both SP and NSP cells, whereas the lipophilic chemotherapeutic agent doxorubicin more significantly inhibited the NSP cells. Finally, we identified breast cancer-resistance protein 1-expressing verapamil-sensitive SPs in three of four human ovarian cancer cell lines and four of six patient primary ascites cells. In the future, individualized therapy must incorporate analysis of the stem cell-like subpopulation of ovarian cancer cells when designing therapeutic strategies for ovarian cancer patients.


Subject(s)
Gene Expression Regulation, Neoplastic , Glycoproteins/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Stem Cells/cytology , Testicular Hormones/pharmacology , Animals , Anti-Mullerian Hormone , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , CHO Cells , Cell Line, Tumor , Cricetinae , Female , Fluorescent Dyes/pharmacology , Humans , Mice , Signal Transduction , Verapamil/pharmacology
7.
Development ; 133(12): 2359-69, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16687449

ABSTRACT

Examination of Müllerian inhibiting substance (MIS) signaling in the rat in vivo and in vitro revealed novel developmental stage- and tissue-specific events that contributed to a window of MIS responsiveness in Müllerian duct regression. The MIS type II receptor (MISRII)-expressing cells are initially present in the coelomic epithelium of both male and female urogenital ridges, and then migrate into the mesenchyme surrounding the male Müllerian duct under the influence of MIS. Expression of the genes encoding MIS type I receptors, Alk2 and Alk3, is also spatiotemporally controlled; Alk2 expression appears earlier and increases predominantly in the coelomic epithelium, whereas Alk3 expression appears later and is restricted to the mesenchyme, suggesting sequential roles in Müllerian duct regression. MIS induces expression of Alk2, Alk3 and Smad8, but downregulates Smad5 in the urogenital ridge. Alk2-specific small interfering RNA (siRNA) blocks both the transition of MISRII expression from the coelomic epithelium to the mesenchyme and Müllerian duct regression in organ culture. Müllerian duct regression can also be inhibited or accelerated by siRNA targeting Smad8 and Smad5, respectively. Thus, the early action of MIS is to initiate an epithelial-to-mesenchymal transition of MISRII-expressing cells and to specify the components of the receptor/SMAD signaling pathway by differentially regulating their expression.


Subject(s)
Epithelial Cells/physiology , Glycoproteins/metabolism , Mesoderm/physiology , Mullerian Ducts/physiology , Receptors, Peptide/metabolism , Signal Transduction/physiology , Smad Proteins/metabolism , Testicular Hormones/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Animals , Anti-Mullerian Hormone , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Movement/physiology , Embryonic Structures/anatomy & histology , Embryonic Structures/physiology , Epithelial Cells/cytology , Female , Glycoproteins/genetics , Humans , In Situ Hybridization , Male , Mesoderm/cytology , Mice , Mullerian Ducts/anatomy & histology , Pregnancy , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta , Smad Proteins/genetics , Testicular Hormones/genetics
8.
Dev Biol ; 288(1): 276-83, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16256976

ABSTRACT

Precise cell fate decisions during differentiation of uterine tissues from the embryonic Müllerian duct are critical for normal fertility. Wnt-7a, a member of the Wnt family of secreted signaling molecules that can signal through a canonical beta-catenin pathway, is necessary for the correct differentiation of both anterior/posterior and radial axes of the uterus. In order to investigate the role of beta-catenin directly in mouse uterine development, we have generated mice that are deficient in beta-catenin expression in the embryonic Müllerian duct. We have found that conditional deletion of beta-catenin in the Müllerian duct mesenchyme before postnatal differentiation of the uterine layers results in a phenotype that is distinct from the phenotype observed by deletion of Wnt-7a. Shortly after birth, the uteri of the conditional mutants appear smaller and less organized. The uteri of adult conditional beta-catenin mutants are grossly deficient in smooth muscle of the myometrium, which has been replaced by adipose, a phenotype resembling human lipoleiomyoma. We also show that the adipocytes in the uteri of mice conditionally deleted for beta-catenin are derived from Müllerian inhibiting substance type II receptor-expressing cells suggesting that they share a common origin with the uterine smooth muscle cells. These results describe the first molecular evidence linking disruption of beta-catenin expression in mesenchymal cells with a switch from myogenesis to adipogenesis in vivo.


Subject(s)
Adipogenesis/genetics , Gene Deletion , Mesoderm/physiology , Myometrium/embryology , Uterus/embryology , beta Catenin/deficiency , Adipocytes/physiology , Adipose Tissue/physiology , Animals , Female , Integrases/biosynthesis , Integrases/genetics , Mice , Mice, Transgenic , Muscle, Smooth/metabolism , Myometrium/growth & development , Uterus/growth & development , beta Catenin/genetics , beta Catenin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...