Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Cancer Chemother Pharmacol ; 59(2): 235-49, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16972069

ABSTRACT

PURPOSE: Paclitaxel (PTX) is a widely used chemotherapy agent and may cause cell death by apoptosis subsequent to microtubule (MT) disruption. In this paper, we have investigated whether cell cycle transit and or Cdc2 (Cdk1) activity is required for the apoptosis induced by PTX. METHODS: Cell cycle was analyzed by flow cytometry, Cdc2 was assayed bio chemically. Cdc2 activity was decreased by siRNA and dominant negative (dn) Cdc2 expression. Cells were arrested by chemical or biological inhibitors in a G1 or S phase. Apoptosis was measured by DNA fragmentation and examination of nuclei by microscopy. JNK and AKT activations were assessed as well. RESULTS: Cell cycle inhibition was highly effective in decreasing PTX induced apoptosis. MT morphology was not altered by these inhibitors. PTX induced JNK activity or AKT mediated BAD phosphorylation was unaffected by cell cycle inhibitors. Abrogation of Cdc 2 activity was without effect on PTX induced apoptosis. CONCLUSIONS: While cell cycle transit is required for PTX induced apoptosis; Cdc2 activity is not required.


Subject(s)
Apoptosis/drug effects , CDC2 Protein Kinase/metabolism , Cell Cycle/drug effects , Paclitaxel/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , CDC2 Protein Kinase/genetics , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , DNA Fragmentation/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Flow Cytometry , Fulvestrant , Humans , Hydroxyurea/pharmacology , Inhibitor of Apoptosis Proteins , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/drug effects , Neoplasm Proteins/metabolism , Purines/pharmacology , RNA, Small Interfering/genetics , Roscovitine , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Survivin , Thymidine/pharmacology , Transfection , bcl-Associated Death Protein/metabolism
2.
Trends Endocrinol Metab ; 12(7): 320-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11504672

ABSTRACT

Clinical and experimental data have established that the leading cause of sporadic female breast cancer is exposure to estrogens, predominantly 17beta-estradiol. Recent advances in the understanding of cell-cycle control mechanisms have been applied to outline the molecular mechanisms through which estrogens regulate the cell cycle in cultured breast cancer cells, in particular, in MCF-7 cells. Here, we discuss how estrogens exert control over several key G1 phase cell-cycle regulators, namely cyclin D1, Myc, Cdk2, Cdk4, Cdk inhibitors and Cdc25A. Although the molecular mechanisms underlying estrogenic regulation of G1 phase regulators are far from clear, current evidence indicates that estrogens might regulate several key molecules required for S phase entry, this regulation being independent of cell-cycle transit per se.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , CDC2-CDC28 Kinases , Cell Cycle/physiology , Estrogens/physiology , Cell Cycle/drug effects , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/physiology , Estradiol/pharmacology , Female , Humans , Protein Serine-Threonine Kinases/metabolism
3.
BMC Dev Biol ; 1: 11, 2001.
Article in English | MEDLINE | ID: mdl-11439174

ABSTRACT

BACKGROUND: Mesenchymal-epithelial interactions play an important role in the physiology and pathology of epithelial tissues. Mesenchymal cells either associate with epithelium basement membrane [pericytes and perivascular monocyte-derived cells (MDC)] or reside within epithelium (MDC and T cells). Although intraepithelial mesenchymal cells were suggested to contribute to the epithelium physiology, their association with particular steps in differentiation of epithelial cells, interactions among themselves, and their fate remain unclear. We studied epitopes of mesenchymal cells and their products (immunoglobulins) in stratified epithelium of uterine ectocervix, which is one of the prototypes of complete cellular differentiation from stem into the aged cells. RESULTS: Perivascular CD14 primitive MDC associated with basal (stem) epithelial cells. Thy-1 pericytes of microvasculature secreted intercellular vesicles, which associated with Ki67 postmitotic epithelial cells expressing MHC class I. Intraepithelial T cells showed an association with veiled type MDC [dendritic cell (DC) precursors] among parabasal cells, and exhibited fragmentation after entering intermediate (mature) epithelial layers. Mature DC secreted CD68 and exhibited fragmentation after reaching mid intermediate layers. Binding of IgM was detected at the top of each layer: in the upper parabasal, upper intermediate, and most surface epithelial cells. IgG was confined to the entire superficial layer. CONCLUSIONS: These data suggest that the phylogenetically and ontogenetically developed hierarchy of mesenchymal cells (MDC, pericytes, T cells) and immunoglobulins (IgM, IgG) accompanies differentiation of epithelial cells from immature into the mature and aged phenotype. Further studies of an involvement of mesenchymal cells in the regulation of tissue homeostasis may bring novel approaches to the prevention and therapy of tissue dysfunctions characterized by permanent tissue immaturity (muscular dystrophy) or accelerated aging (degenerative diseases).


Subject(s)
Epithelial Cells/physiology , Immunoglobulins/physiology , Mesoderm/physiology , Apoptosis/physiology , CD3 Complex/analysis , CD8 Antigens/analysis , Cell Differentiation/physiology , Cervix Uteri/blood supply , Cervix Uteri/cytology , Epithelial Cells/cytology , Epithelium/blood supply , Epithelium/physiology , Female , Humans , Immunoglobulins/blood , Immunohistochemistry , Mesoderm/cytology
4.
Mol Carcinog ; 30(2): 88-98, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11241756

ABSTRACT

Zearalenone is a naturally occurring estrogenic contaminant of moldy feeds and is present in high concentrations in dairy products and cereals. Zearalenone was postulated to contribute to the overall estrogen load of women, but the mechanisms of its action are not known. We demonstrated that zearalenone could stimulate the growth of estrogen receptor-positive human breast carcinoma cell line MCF-7. In addition, zearalenone functioned as an antiapoptotic agent by increasing the survival of MCF-7 cell cultures undergoing apoptosis caused by serum withdrawal. Treatment of these cells with 100 nM zearalenone induced cell-cycle transit after increases in the expression of c-myc mRNA and cyclins D1, A, and B1 and downregulation of p27(Kip-1). G(1)/G(2)-phase kinase activity and phosphorylation of the retinoblastoma gene product was also evident. Flow cytometric analysis demonstrated entry of cells into the S and G(2)/M phases of the cell cycle, and phosphorylation of histone H3 occurred 36 h after zearalenone treatment. Ectopic expression of a dominant-negative p21(ras) completely abolished the zearalenone-induced DNA synthesis in these cells, and the specific inhibitor PD98059 for mitogen/extracellular-regulated protein kinase kinase arrested S-phase entry induced by zearalenone. These data suggest that the mitogen-activated protein kinase signaling cascade is required for zearalenone's effects on cell-cycle progression in MCF-7 cells. Given the presence of this mycotoxin in cereals, milk, and meat, the possibility that zearalenone is a potential promoter of breast cancer tumorigenesis should be investigated further. Mol. Carcinog. 30:88-98, 2001.


Subject(s)
Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Cycle/drug effects , Estrogens, Non-Steroidal/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Tumor Suppressor Proteins , Zearalenone/pharmacology , Adenoviridae/genetics , Annexin A5/metabolism , Blotting, Northern , Blotting, Western , Breast Neoplasms/metabolism , Bromodeoxyuridine , Cell Survival/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Down-Regulation , Female , Flow Cytometry , Formazans , Genes, myc/drug effects , Humans , Luciferases/metabolism , Microtubule-Associated Proteins/antagonists & inhibitors , Mitosis/drug effects , Phosphorylation , Tetrazolium Salts , Tumor Cells, Cultured/drug effects
5.
Mol Cell Biol ; 21(3): 794-810, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11154267

ABSTRACT

Estrogens induce proliferation of estrogen receptor (ER)-positive MCF-7 breast cancer cells by stimulating G(1)/S transition associated with increased cyclin D1 expression, activation of cyclin-dependent kinases (Cdks), and phosphorylation of the retinoblastoma protein (pRb). We have utilized blockade of cyclin D1-Cdk4 complex formation through adenovirus-mediated expression of p16(INK4a) to demonstrate that estrogen regulates Cdk inhibitor expression and expression of the Cdk-activating phosphatase Cdc25A independent of cyclin D1-Cdk4 function and cell cycle progression. Expression of p16(INK4a) inhibited G(1)/S transition induced in MCF-7 cells by 17-beta-estradiol (E(2)) with associated inhibition of both Cdk4- and Cdk2-associated kinase activities. Inhibition of Cdk2 activity was associated with delayed removal of Cdk-inhibitory activity in early G(1) and decreased cyclin A expression. Cdk-inhibitory activity and expression of both p21(Cip1) and p27(Kip1) was decreased, however, in both control and p16(INK4a)-expressing cells 20 h after estrogen treatment. Expression of Cdc25A mRNA and protein was induced by E(2) in control and p16(INK4a)-expressing MCF-7 cells; however, functional activity of Cdc25A was inhibited in cells expressing p16(INK4a). Inhibition of Cdc25A activity in p16(INK4a)-expressing cells was associated with depressed Cdk2 activity and was reversed in vivo and in vitro by active Cdk2. Transfection of MCF-7 cells with a dominant-negative Cdk2 construct inhibited the E(2)-dependent activation of ectopic Cdc25A. Supporting a role for Cdc25A in estrogen action, antisense CDC25A oligonucleotides inhibited estrogen-induced Cdk2 activation and DNA synthesis. In addition, inactive cyclin E-Cdk2 complexes from p16(INK4a)-expressing, estrogen-treated cells were activated in vitro by treatment with recombinant Cdc25A and in vivo in cells overexpressing Cdc25A. The results demonstrate that functional association of cyclin D1-Cdk4 complexes is required for Cdk2 activation in MCF-7 cells and that Cdk2 activity is, in turn, required for the in vivo activation of Cdc25A. These studies establish Cdc25A as a growth-promoting target of estrogen action and further indicate that estrogens independently regulate multiple components of the cell cycle machinery, including expression of p21(Cip1) and p27(Kip1).


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , Cell Cycle/physiology , Cyclin-Dependent Kinases/antagonists & inhibitors , Estrogens/metabolism , Proto-Oncogene Proteins , Tumor Suppressor Proteins , cdc25 Phosphatases/metabolism , Adenoviridae/genetics , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Female , Humans , Microtubule-Associated Proteins/metabolism , Models, Biological , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Oligodeoxyribonucleotides, Antisense/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptors, Estrogen/metabolism , Retinoblastoma Protein/metabolism , Transduction, Genetic , Tumor Cells, Cultured , cdc25 Phosphatases/genetics
6.
Med Hypotheses ; 57(6): 729-35, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11918436

ABSTRACT

Available data indicate that growth of invasive tumors is enhanced by homeostatic mechanisms of the host involved in normal tissue regeneration and repair. To achieve this, malignant cells may (i) induce degeneration of normal cells at the host-tumor interface, (ii) hybridize in situ with activated host stem cells, required for replacement of lost mature tissue cells, (iii) the resulting malignant/normal cell hybrids may exhibit an antigenic similarity to normal cells, (iv) thereby preventing recognition by the immune system, (v) and exploiting normal mechanisms of tissue regeneration by the host. In addition, primary cancers with allotypic determinants may utilize other homeostatic mechanisms evolved in mammals to promote fetal allograft survival. They may have a potential to grow in another (secondary) host. Novel approaches to cancer prevention and control may depend on a better understanding of the mechanisms by which normal cellular growth are controlled, and hybridization prevented.


Subject(s)
Neoplasm Invasiveness/physiopathology , Animals , Antigens, Neoplasm , Female , Fetus/immunology , Homeostasis , Humans , Hybrid Cells/immunology , Hybrid Cells/physiology , Isoantigens , Male , Mesoderm/immunology , Mesoderm/physiology , Models, Biological , Neoplasm Invasiveness/immunology , Neoplasms/etiology , Neoplasms/prevention & control , Neoplasms/therapy , Pregnancy
7.
Steroids ; 65(4): 190-205, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10713307

ABSTRACT

In the present paper, we report that ovaries of adult rats treated with testosterone propionate (TP) on a critical postnatal Day 5 exhibit histologic and immunohistochemical findings which resemble those of the anovulatory ovaries in middle-aged female rats. The sterile rat model has been long known whereas ovarian failure seems to be a reason for anovulation with normal hypothalamo-pituitary-gonadotropin background. Appropriate function of ovarian steroidogenic cells is also regulated by mesenchymal cells. To characterize the ovarian failure, we studied the histology, luteinizing hormone receptor (LHR) expression, and characterized changes of vascular pericytes, T cells, and dendritic cells in ovarian steroidogenic compartments consisting of interstitial cells (ISC) of ovarian interstitial glands, and granulosa and theca interna cells of ovarian follicles. Normal adult ovaries contained 63% of mature interstitial glands. The mature ISC exhibited moderate cytoplasmic and strong surface LHR expression and fine (<5 micrometer) cytoplasmic vacuoles (ISC of 'luteal type'). They originated from young ISC of 'thecal type,' which exhibited strong cytoplasmic LHR expression. Remaining 37% were aged interstitial glands, which consisted of aged ISC (increased cytoplasmic vacuolization, nuclear pyknosis, and reduced surface LHR expression) and regressing ISC (weak cytoplasmic and no surface LHR expression). However, no mature ISC of 'luteal type' were detected in anovulatory ovaries of adult rats (45- and 60-day-old) injected with TP (100 or 500 microgram) on postnatal Day 5 (TP rats). Their ovaries contained 96% of aged interstitial glands with aged and regressing ISC. Remaining 4% were abnormal interstitial glands with direct transition of young ISC of 'thecal type' into aged ISC (young/aged glands). Lack of mature ISC, and similar amount of aged (96%) and young/aged interstitial glands (4%) was also detected in anovulatory ovaries of untreated persistently estrous middle-aged (10-month-old) females (aging PE rats). The aging process in TP and aging PE rats was accompanied by regression of vascular pericytes, T cells, and dendritic cells within the interstitial glands. In addition, anovulatory ovaries of TP rats and aging PE females contained mature follicles exhibiting LHR overexpression by granulosa cells, and aged (cystic) follicles with reduced layers of granulosa cells lacking LHR expression. In contrast, when the rats were injected with 500 microgram of TP later, on postnatal Day 10, the adult females exhibited estrous cycles and normal ovaries with corpora lutea. These results show that injection of TP during the critical postnatal period causes a lack of mature and preponderance of aged ISC in adult ovaries, accompanied by degeneration of mesenchymal cells. We suggest that mesenchymal cells regulate qualitative aspects of tissue-specific cells, and this function of mesenchymal cells is programmed during the critical period of development.


Subject(s)
Ovary/cytology , Ovary/physiology , Testosterone/pharmacology , Androgens/metabolism , Androgens/pharmacology , Animals , Animals, Newborn , Anovulation , Cellular Senescence/physiology , Dendritic Cells/drug effects , Female , Mesoderm/cytology , Mesoderm/drug effects , Ovary/drug effects , Pericytes/cytology , Pericytes/drug effects , Rats , Rats, Inbred Strains , Receptors, LH/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Vacuoles
8.
Am J Reprod Immunol ; 42(3): 135-52, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10517174

ABSTRACT

PROBLEM: The classification of placental villi was reviewed, and regeneration of villous trees in mature human placentae was examined. METHOD OF STUDY: Expression of Thy-1 by placental fibroblasts and pericytes, and markers of endothelial cells and monocyte-derived cells were studied by immunohistochemistry and image analysis. RESULTS: Villous regeneration consists of: (i) dedifferentiation of mature ramuli into young stem villi producing mesenchymal villi; (ii) differentiation of mesenchymal villi into immature intermediate villi; and (iii) differentiation of immature intermediate villi into transitory intermediate villi, branching into the precursors of mature intermediate and terminal villi. These processes are associated with dedifferentiation and redifferentiation of placental monocyte-derived cells. Significant changes of Thy-1 expression by fibroblasts and pericytes accompany aging and degeneration, as well as regeneration of placental villi. CONCLUSIONS: Villous aging and degeneration in normal mature human placenta is compensated by regeneration of villous trees. Lack of villous regeneration may cause chronic fetal distress, due to the increasing demands of the growing fetus on the remaining terminal villi.


Subject(s)
Cellular Senescence/immunology , Chorionic Villi/growth & development , Chorionic Villi/physiology , Monocytes/physiology , Regeneration/immunology , Thy-1 Antigens/biosynthesis , Chorionic Villi/metabolism , Endothelium/cytology , Endothelium/metabolism , Endothelium/physiology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Immunohistochemistry , Male , Monocytes/cytology , Monocytes/metabolism
9.
J Biol Chem ; 274(12): 8208-16, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10075725

ABSTRACT

The antineoplastic agent paclitaxel (TaxolTM), a microtubule stabilizing agent, is known to arrest cells at the G2/M phase of the cell cycle and induce apoptosis. We and others have recently demonstrated that paclitaxel also activates the c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) signal transduction pathway in various human cell types, however, no clear role has been established for JNK/SAPK in paclitaxel-induced apoptosis. To further examine the role of JNK/SAPK signaling cascades in apoptosis resulting from microtubular dysfunction induced by paclitaxel, we have coexpressed dominant negative (dn) mutants of signaling proteins of the JNK/SAPK pathway (Ras, ASK1, Rac, JNKK, and JNK) in human ovarian cancer cells with a selectable marker to analyze the apoptotic characteristics of cells expressing dn vectors following exposure to paclitaxel. Expression of these dn signaling proteins had no effect on Bcl-2 phosphorylation, yet inhibited apoptotic changes induced by paclitaxel up to 16 h after treatment. Coexpression of these dn signaling proteins had no protective effect after 48 h of paclitaxel treatment. Our data indicate that: (i) activated JNK/SAPK acts upstream of membrane changes and caspase-3 activation in paclitaxel-initiated apoptotic pathways, independently of cell cycle stage, (ii) activated JNK/SAPK is not responsible for paclitaxel-induced phosphorylation of Bcl-2, and (iii) apoptosis resulting from microtubule damage may comprise multiple mechanisms, including a JNK/SAPK-dependent early phase and a JNK/SAPK-independent late phase.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Microtubules/drug effects , Mitogen-Activated Protein Kinases , Ovarian Neoplasms/physiopathology , Paclitaxel/pharmacology , Female , Humans , JNK Mitogen-Activated Protein Kinases , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Transfection , Tumor Cells, Cultured
10.
Biol Reprod ; 58(5): 1283-9, 1998 May.
Article in English | MEDLINE | ID: mdl-9603265

ABSTRACT

Cyclin-dependent kinases (Cdks) and their cyclin partners regulate mammalian cell proliferation and withdrawal from the cell cycle and, as such, control differentiation in many tissues. Studies were undertaken to examine the roles of cell cycle proteins in differentiating cytotrophoblasts. Cyclin E gene and protein expression was down-regulated after 24 h in cultured trophoblasts. Cdk2-associated kinase activity was decreased after 96 h in culture as was the amount of cyclin E in complexes with Cdk2; however, levels of the Cdk inhibitor, p27Kip1, were significantly increased. In freshly isolated trophoblasts and in 24-h cultures, the retinoblastoma gene product (pRb) was found in both the active and inactive forms, yet only hypophosphorylated, active pRb was present in syncytiotrophoblast. Thus, inactivation of Cdk2 through cyclin E down-regulation and increased p27Kip1 expression leads to an accumulation of active pRb in syncytiotrophoblast. Prevention of entry into S phase by hypophosphorylated pRb may allow trophoblasts to respond to signals that potentiate differentiation. Our studies suggest that regulation of G1-phase Cdk activity may be involved in the terminal differentiation process of cytotrophoblasts.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , Cyclin-Dependent Kinases , Cyclins/biosynthesis , Protein Serine-Threonine Kinases , Trophoblasts/metabolism , Tumor Suppressor Proteins , Blotting, Northern , Blotting, Western , Cell Differentiation , Cells, Cultured , Cyclin G , Cyclin G1 , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/biosynthesis , Humans , Immunohistochemistry , Microtubule-Associated Proteins/biosynthesis , Placenta/cytology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/biosynthesis
11.
J Biol Chem ; 273(9): 4928-36, 1998 Feb 27.
Article in English | MEDLINE | ID: mdl-9478937

ABSTRACT

The essential cellular functions associated with microtubules have led to a wide use of microtubule-interfering agents in cancer chemotherapy with promising results. Although the most well studied action of microtubule-interfering agents is an arrest of cells at the G2/M phase of the cell cycle, other effects may also exist. We have observed that paclitaxel (Taxol), docetaxel (Taxotere), vinblastine, vincristine, nocodazole, and colchicine activate the c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) signaling pathway in a variety of human cells. Activation of JNK/SAPK by microtubule-interfering agents is dose-dependent and time-dependent and requires interactions with microtubules. Functional activation of the JNKK/SEK1-JNK/SAPK-c-Jun cascade (where JNKK/SEK1 is JNK kinase/SAPK kinase) was demonstrated by activation of a 12-O-tetradecanoylphorbol-13-acetate response element (TRE) reporter construct in a c-Jun dependent fashion. Microtubule-interfering agents also activated both Ras and apoptosis signal-regulating kinase (ASK1) and coexpression of dominant negative Ras and dominant negative apoptosis signal-regulating kinase exerted individual and additive inhibition of JNK/SAPK activation by microtubule-interfering agents. These findings suggest that multiple signal transduction pathways are involved with cellular detection of microtubular disarray and subsequent activation of JNK/SAPK.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Microtubules/drug effects , Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases/metabolism , ras Proteins/metabolism , Colchicine/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Humans , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase Kinases , Models, Biological , Nocodazole/pharmacology , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Regulatory Sequences, Nucleic Acid , Signal Transduction , Transcription, Genetic , Tumor Cells, Cultured , Vinca Alkaloids/pharmacology
12.
Mol Hum Reprod ; 3(7): 569-78, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9268134

ABSTRACT

We examined the immunohistochemical localization of luteinizing hormone (LH)/human chorionic gonadotrophin (HCG) receptor (LH-R) in the human ovary using the anti-human LH-R monoclonal antibody, 3B5. In the antral follicles, LH-R was detected on theca interna cells. In pre-ovulatory follicles, granulosa cells also expressed LH-R. During corpus luteum formation, granulosa cells seemed to increase the expression of LH-R, and in corpus luteum of mid-luteal phase, large luteal cells expressed LH-R more intensely than small luteal cells. In the regressing corpus luteum, LH-R was almost undetectable on both luteal cells, whereas in the corpus luteum of early pregnancy, LH-R continued to be expressed on large luteal cells. The granulosa cells obtained from the patients undergoing in-vitro fertilization therapy were cultured for 3 days in serum-free medium, without or with HCG (10 IU/ml) and tumour necrosis factor (TNF)alpha (10 ng/ml). Flow cytometry showed that the expression of LH-R on the cell surface of luteinizing granulosa cells was enhanced by HCG, but was unaffected by TNFalpha. These results suggest that the main target cells for LH/HCG change from theca interna cells/small luteal cells to granulosa cells/large luteal cells during ovulation, corpus luteum formation, and differentiation into the corpus luteum of pregnancy, probably under the influence of LH/HCG.


Subject(s)
Chorionic Gonadotropin/pharmacology , Granulosa Cells/chemistry , Ovary/chemistry , Receptors, LH/analysis , Adolescent , Adult , Animals , Antibodies, Monoclonal , Cells, Cultured , Corpus Luteum/chemistry , Female , Flow Cytometry , Humans , Menstrual Cycle/physiology , Pregnancy , Rats , Tumor Necrosis Factor-alpha/pharmacology
13.
Environ Health Perspect ; 105 Suppl 3: 633-6, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9168007

ABSTRACT

It has been suggested that dietary estrogens neutralize the effect of synthetic chemicals that mimic the effects of estrogen (i.e., xenoestrogens, environmental estrogens). Genistein, a dietary estrogen, inhibits the growth of breast cancer cells at high doses but additional studies have suggested that at low doses, genistein stimulates proliferation of breast cancer cells. Therefore, if dietary estrogens are estrogenic at low doses, one would predict that they stimulate estrogen-receptor positive breast cancer cells to enter the cell cycle. Genistein and the fungal toxin zearalenone were found to increase the activity of cyclin dependent kinase 2 (Cdk2) and cyclin D1 synthesis and stimulate the hyperphosphorylation of the retinoblastoma susceptibility gene product pRb105 in MCF-7 cells. The steroidal antiestrogen ICI 182,780 suppressed dietary estrogen-mediated activation of Cdk2. Dietary estrogens not only failed to suppress DDT-induced Cdk2 activity, but were found to slightly increase enzyme activity. Both zearalenone and genistein were found to stimulate the expression of a luciferase reporter gene under the control of an estrogen response element in MVLN cells. Our findings are consistent with a conclusion that dietary estrogens at low concentrations do not act as antiestrogens, but act like DDT and estradiol to stimulate human breast cancer cells to enter the cell cycle.


Subject(s)
Breast/cytology , Breast/drug effects , CDC2-CDC28 Kinases , Cell Cycle/drug effects , Diet/adverse effects , Estrogens, Non-Steroidal/toxicity , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Carcinogens/toxicity , Cyclin D1 , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Cyclins/biosynthesis , Environmental Health , Enzyme Activation/drug effects , Female , Humans , Luciferases/biosynthesis , Luciferases/genetics , Neoplasms, Hormone-Dependent/etiology , Neoplasms, Hormone-Dependent/metabolism , Oncogene Proteins/biosynthesis , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Retinoblastoma Protein/metabolism , Risk Factors , Tumor Cells, Cultured
14.
Mol Carcinog ; 18(2): 107-14, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9049186

ABSTRACT

Estrogens play a critical role in the etiology of found breast cancer. Estradiol promotes the growth of breast cancer cells in vivo and in vitro. Exogenous estrogens in both the environment and in the human diet increase the growth of breast cancer cells in vitro. A role for xenoestrogens in breast cancer etiology has been proposed but remains controversial. We examined the effects of the xenoestrogenic pesticide 1,1,1-trichloro-2,2-bis(chlorophenyl)ethane (DDT) on estrogen-receptor (ER)-positive MCF-7 and T-47D human breast cancer cells as well as on ER-negative HS 578Bst breast cancer cells and rat liver cells. Estradiol and DDT were found to increase the growth of MCF-7 cells in the presence of insulin. The activity of cyclin-dependent kinase (Cdk)2 increased in growth-arrested T-47D and MCF-7 cells treated with beta-estradiol or DDT. The steroidal antiestrogen ICI 182,780 prevented both growth and Cdk2 activation induced by estradiol or DDT. Increased phosphorylation of Cdk2 and the retinoblastoma protein (pRb1O5) was observed in ER-positive cells treated with DDT or estradiol. Cdk2 activity was not affected by DDT or estradiol in ER-negative HS 578Bst breast cancer cells or in rat liver epithelial cells. Cyclin D1 protein synthesis was increased by DDT and estradiol in MCF-7 cells. DDT and estradiol-induced ER-dependent transcriptional activation of estrogen response elements (EREs) in stably transfected MVLN cells, and ERE activation by low doses of DDT was increased by insulin. These findings suggest that DDT can stimulate breast cancer cells to enter into the cell cycle by directly affecting key regulatory elements. The relative potency of DDT in inducing cell-cycle progression appears to be only 100-300 times less than that of estradiol when measured in the presence of insulin. Therefore, the cancer risks associated with DDT exposure may be greater than first thought, especially when additional mitogenic stimuli are present.


Subject(s)
Breast Neoplasms/pathology , Carcinogens/pharmacology , Cell Cycle/drug effects , DDT/pharmacology , Estradiol/pharmacology , Animals , Breast Neoplasms/chemically induced , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic , Cyclin D1 , Cyclin-Dependent Kinases/metabolism , Cyclins , Flow Cytometry , Humans , Insulin/pharmacology , Liver/cytology , Liver/drug effects , Oncogene Proteins , Phosphorylation , Rats , Receptors, Estrogen/metabolism , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured
15.
Am J Reprod Immunol ; 36(6): 327-41, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8985508

ABSTRACT

PROBLEM: We have recently observed that the regression of corpora lutea (CL) in women during the reproductive period of life is accompanied by a diminution of Thy-1 differentiation protein release from vascular pericytes and an accumulation of T lymphocytes and activated macrophages among both degenerating granulosa lutein cells (GLC) and theca lutein cells. These data suggest that the immune system and other stromal factors, representing components of the "tissue control system," may play a role in regression of the CL. We investigated degenerating CL from climacteric women to address the possibility that the decline of immune functions with advancing age may result in incomplete regression of luteal tissue. This could contribute to the altered hormonal profiles and abnormal uterine bleeding that frequently occur during the climacteric. METHOD: Immunoperoxidase staining and image analysis were used to localize Thy-1 differentiation protein of vascular pericytes, cytokeratin staining of GLC, neural cell adhesion molecule expression by theca lutein cells, CD15 of neutrophils, CD4, CD14, CD68, and leukocyte common antigens of macrophages, and CD3 and CD8 determinants of T lymphocytes. We also investigated the expression of luteinizing hormone receptor (LH receptor) and mitogen activated protein kinases (MAP kinases) in luteal cells. Samples of regressing luteal tissue were obtained during the follicular phase from perimenopausal women (age 45-50) who exhibited prolonged or irregular cycles. For comparison, luteal tissues from women with regular cycles (age 29-45) and CL of pregnancy were also investigated. RESULTS: Corpora lutea of the climacteric women exhibited irregular regression of luteal tissue characterized by a lack of cytoplasmic vacuolization and nuclear pyknosis in GLC, and by a persistence of theca lutein cells exhibiting hyperplasia and adjacent theca externa layers. This was accompanied by a continuing release of Thy-1 differentiation protein from vascular pericytes. Persisting GLC lacked surface expression of macrophage markers (CD4, CD14, CD68 and leukocyte common antigen) as well as nuclear granules exhibiting CD15 of neutrophils, detected in regularly regressing GLC. In addition, such persisting GLC showed weak or no LH receptor expression, and retained the expression of cytokeratin. They also exhibited enhanced staining for MAP kinases. Strong cytoplasmic MAP kinase expression with occasional nuclear translocation was also detected in persisting theca lutein cells, indicating high metabolic activity of these cells. T lymphocytes, although occasionally present in luteal stroma within luteal convolutions, did not invade among persisting GLC and were virtually absent from layers of theca externa and theca lutein cells. CONCLUSIONS: These data indicate that the regressing CL in climacteric women may exhibit persistence of luteal cells, perhaps because of age-induced alterations of the immune system and other local stromal homeostatic mechanisms involved in the elimination of luteal cells. Persisting GLC and/or theca lutein cells may exhibit abnormal hormonal secretion that contributes to the alteration of target tissues, such as the endometrium, resulting in abnormal uterine bleeding, hyperplasia, and neoplasia.


Subject(s)
Aging/immunology , Climacteric/immunology , Luteolysis/immunology , Organ Specificity/immunology , Adult , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Endometrium/immunology , Female , Humans , Immunohistochemistry , Keratins/analysis , Lewis X Antigen/analysis , Middle Aged , Neural Cell Adhesion Molecules/analysis , Ovary/immunology , Thy-1 Antigens/analysis
16.
Biol Reprod ; 55(1): 111-9, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8793065

ABSTRACT

A variety of growth factors and guanosine triphosphate (GTP) binding protein-linked receptors are known to activate mitogen activated protein kinases (MAPK); however, no evidence exists demonstrating activation of the MAPK pathway by glycoprotein hormones. Using porcine granulosa cells (PGC), we show that physiological concentrations of LH and FSH increase enzymatic activity 1) of p44MAPK extracellular regulated kinase 1 (ERK1) but not that of p42MAPK (ERK2) in the cytosol and 2) of both ERK1 and ERK2 in the nucleus. Cytosolic ERK1 was activated by LH more rapidly than by FSH. Cyclic AMP increased kinase activities of both ERK1 and ERK2 in the cytoplasm as well as in the nucleus. Activation of ERK1 by gonadotropins and cAMP was accompanied by increased tyrosine phosphorylation of the kinase. Immunohistochemical studies demonstrated predominantly cytoplasmic staining for MAPK in untreated PGC cultures whereas treatment with gonadotropins led to increased nuclear immunoreactivity indicating translocation of MAPK to the nucleus. The translocation as well as increase in nuclear ERK1 and ERK2 was delayed and coincided with a decrease in cytosolic ERK1 activity. Epidermal growth factor (EGF) increased ERK1 and ERK2-associated kinase activity 7-8-fold in the cytoplasm of PGC, while kinase activity of cytoplasmic ERK1 was enhanced 3-4-fold by LH, FSH, or cAMP. In summary, we have for the first time demonstrated that gonadotropins (and cAMP) can activate MAPK in appropriate target cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP/pharmacology , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/enzymology , Luteinizing Hormone/pharmacology , Mitogen-Activated Protein Kinases , Animals , Cell Nucleus/enzymology , Cells, Cultured , Cytosol/enzymology , Enzyme Activation/drug effects , Female , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Phosphotyrosine/metabolism , Swine
17.
Mol Endocrinol ; 10(5): 488-98, 1996 May.
Article in English | MEDLINE | ID: mdl-8732680

ABSTRACT

Cyclin-dependent kinases (Cdk) act to regulate G1- to S-phase transition in mammalian cells. We have studied the effects of estradiol and the steroidal antiestrogen ICI 182, 780 on induction of Cdk activity and the consequent phosphorylation of retinoblastoma protein (Rb) in estrogen-responsive MCF-7 breast cancer cells. Treatment of growth-arrested MCF-7 cells with physiological concentrations of estradiol led to a time-dependent increase in Cdk2-associated and cyclin E-dependent kinase activity, which was accompanied by hyperphosphorylation of Rb and S-phase entry. Induction of both Cdk2 activity and DNA synthesis by estradiol was dose dependent and was inhibited by coadministration of ICI 182,780. Elicitation of Cdk2 activity was found to require prolonged (> 8h) estradiol exposure. Levels of cyclins E and A were unchanged in MCF-7 cells undergoing G1- to S-transit; however, synthesis and steady state levels of cyclin D1 protein were increased by estradiol. Cdk4-associated Rb kinase activity was evident in MCF-7 cells by 6 h after estradiol exposure and was inhibited by antiestrogen. Cdk2 and Cdk4 protein levels were not altered by estrogen treatment; however, faster migrating, phosphorylated Cdk2 forms increased in estradiol-treated MCF-7 cells by 12 after release from growth arrest. Cdtk-inhibitory activities, associated with p27kip-1, were eliminated from growth-arrested MCF-7 cells after treatment with estradiol but were not eliminated from cells cotreated with estradiol and ICI 182,780. These findings suggest that estradiol regulates G1 progression in MCF-7 cells through direct effects upon Cdk activation, Rb phosphorylation, and by inducing elimination of Cdk inhibitors.


Subject(s)
Breast Neoplasms/metabolism , Cyclin B , Cyclin-Dependent Kinases/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Retinoblastoma Protein/metabolism , Cyclin B1 , Cyclin D1 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/biosynthesis , Cyclins/metabolism , DNA/biosynthesis , Enzyme Activation/drug effects , Enzyme Inhibitors/metabolism , Fulvestrant , G1 Phase/drug effects , Humans , Oncogene Proteins/biosynthesis , Phosphorylation , S Phase/drug effects , Tumor Cells, Cultured
18.
Biol Reprod ; 53(6): 1373-84, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8562694

ABSTRACT

Factors determining the life span of the human corpus luteum (CL) are not known. In addition to being determined by hormonal factors, such as hCG, the life of luteal cells may be determined by the preservation of luteal vascularization. Furthermore, the CL represents an immunologically unique tissue, as it is formed after menarche, long after adaptation of the immune system toward self. Thus, CL regression may be immunologically mediated. To determine what role the vasculature and immune system play in human CL development and regression, we examined immunohistochemically 1) the expression of Thy-1 differentiation protein by vascular pericytes, 2) the expression of major histocompatibility complex (MHC) class I and class II molecules in granulosa lutein cells (GLC), and 3) infiltration of the CL by macrophages and T lymphocytes. LH receptor (LHR) and cytokeratin 18 expression were also studied. In developing CL, the pericytes of luteal microvasculature released Thy-1 differentiation protein among the endothelial cells of proliferating vessels. In mature CL, Thy-1 released from vascular pericytes accumulated on the surface of GLC, and these cells exhibited LHR immunoreactivity (LHRI). Overall LHRI increased during the luteal phase and was strongest at the beginning of the late luteal phase. Although vascular pericytes showed strong LHRI, no staining of endothelium was detected during the luteal phase. GLC exhibited strong cytokeratin staining and moderate staining for MHC class I and MHC class II antigens; numerous macrophages were detected in luteal tissue. During pregnancy, the staining pattern was similar to that seen in the mature CL at the end of the midluteal phase. During the late luteal phase, surface expression of MHC class I and MHC class II antigens by GLC was substantially enhanced, and some T cells invaded among luteal cells. By the end of the cycle, an acute regression of vasculature and luteal tissue was observed along the fibrous septa. The remaining GLC showed only surface and no cytoplasmic LHRI. During the subsequent cycle, in the presence of numerous T cells, regressing GLC exhibited strong surface expression of various macrophage markers, such as CD4, CD14, CD68, and leukocyte common antigen, a feature not detected in the CL during the luteal phase nor described in other tissues. A complete loss of cytokeratin staining in GLC was observed. In regressing CL, strong LHRI was present in the endothelium of small and large luteal vessels. In conclusion, vascular pericytes and macrophages may stimulate the development and senescence of luteal tissue. The senescence of GLC may be inconsistent with preservation of luteal vasculature, and T lymphocytes appear to participate in terminal regression of the CL. Regression of luteal tissue therefore resembles immunologic rejection of a transplant. During pregnancy, the aging process of GLC appears to be interrupted, possibly due to the temporary acceptance of the CL "graft."


Subject(s)
Immunity , Luteolysis/immunology , Receptors, LH/analysis , Adult , Corpus Luteum/blood supply , Corpus Luteum/chemistry , Corpus Luteum/immunology , Endothelium, Vascular/chemistry , Female , Granulosa Cells/chemistry , Granulosa Cells/immunology , Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class II/analysis , Humans , Immunoenzyme Techniques , Keratins/analysis , Macrophages/immunology , Middle Aged , Pregnancy , T-Lymphocytes/immunology , Thy-1 Antigens/analysis
19.
Alcohol Clin Exp Res ; 19(6): 1382-8, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8749799

ABSTRACT

In this study, luteinized human granulosa cells (GC) obtained during in vitro fertilization procedures were used as a model system to evaluate the effects of ethanol (EtOH), a well-known reproductive toxin, on epidermal growth factor (EGF) and gonadotropin-stimulated steroidogenesis. Our results demonstrate that the basal progesterone (P4) and estradiol (E2) secretion by human GC in vitro was dependent on the ovarian stimulation protocol. EGF significantly enhanced P4, but not E2, secretion in human GC from clomiphene citrate (CC), human menopausal gonadotropin (hMG), and hMG/gonadotropin-releasing hormone agonist (GnRH-a)-treated patients. The effects of EGF plus luteinizing hormone (LH) were additive in cells from the CC group, but less than additive in hMG and hMG/GnRH-a groups. EtOH at 20 mM or more inhibited EGF stimulated P4 secretion in human GC from all three patient groups. EtOH inhibited P4 secretion stimulated by EGF and LH cotreatment in the CC and hMG/GnRH-a groups, but not in human GC from the hMG-treated patients. These results suggest that basal and EGF or LH-stimulated P4 secretion by human GC, as well as the effects of EtOH, are profoundly influenced by the follicle's hormonal milieu.


Subject(s)
Epidermal Growth Factor/antagonists & inhibitors , Ethanol/pharmacology , Granulosa Cells/drug effects , Progesterone/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Estradiol/metabolism , Female , Fertilization in Vitro , Granulosa Cells/metabolism , Humans , Ovulation Induction
20.
Biol Reprod ; 52(4): 776-92, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7780000

ABSTRACT

Ovarian follicular development is dependent on growth and differentiation of the oocyte, as well as the granulosa and theca cell layers. The majority of primary follicles in the adult human ovary are not growing, and most antral follicles undergo atresia. The mechanisms regulating follicular growth and differentiation are poorly understood. Expression of key regulatory proteins in cells of certain follicles may be involved. We have studied the distribution of retinoblastoma protein (pRb), a key cell cycle regulator, in human follicles and CL by quantitative immunohistochemistry. Recent studies suggest that high nuclear concentrations of pRb are associated with the arrest of cell proliferation and the beginning of differentiation; during advanced differentiation of cells pRb is markedly depleted or absent. We also studied follicular distribution of Thy-1 differentiation protein, a morpho-regulatory molecule associated with cell differentiation, and the presence of macrophages. Macrophages have been shown to stimulate steroidogenesis in granulosa cells in vitro, and they are required for release of Thy-1 differentiation protein from vascular pericytes among granulosa cells in vivo. Our results indicate that oocytes in resting follicles exhibit pRb in the nucleoli. During initiation of follicular growth, the pRb expression first extends over the oocyte nuclei and then diminishes from both nuclei and nucleoli in preantral follicles. When the oocytes reach maximum size in small antral follicles, the pRb expression is reestablished in oocyte nucleoli. In differentiating granulosa and theca cell layers of preantral and small antral follicles, pRb expression is high, but it is low in growing large antral follicles. During CL development and regression, pRb expression in the nuclei of granulosa lutein cells first increases and then decreases. Follicular development is accompanied by the presence of Thy-1 differentiation protein and macrophages under the follicular basement membrane. In growing large antral follicles, during the mid-follicular phase, larger macrophages exhibit physical contacts with granulosa cells through the follicular basement membrane, and, during the late follicular phase, small dendritic macrophages can be detected among granulosa cells, but not within the follicular antrum. Large antral follicles undergoing atresia exhibit strong pRb expression in granulosa cells. This is accompanied by a lack of Thy-1 differentiation protein among granulosa cells and the occurrence of large phagocytic macrophages in the follicular antrum. This is the first report of pRb expression in the human ovary.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Cell Cycle , Corpus Luteum/physiology , Macrophages/physiology , Ovarian Follicle/physiology , Retinoblastoma Protein/analysis , Thy-1 Antigens/analysis , Cell Differentiation , Epithelium/chemistry , Female , Fluorescent Antibody Technique , Follicular Atresia , Granulosa Cells/chemistry , Humans , Immunoenzyme Techniques , Theca Cells/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...