Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Oncogene ; 34(7): 932-8, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-24531709

ABSTRACT

Recent studies suggest that physiological and tumorigenic proliferation of mammalian cells is controlled by multiple cyclin-dependent kinases (CDKs) largely in tissue-specific manners. We and others previously demonstrated that adult mice deficient for the Cyclin D partner CDK4 (Cdk4(-/-) mice) exhibit hypoplasia in the pituitary and pancreatic islet due to primary postnatal defects in proliferation. Intriguingly, those neuroendocrine tissues affected in Cdk4(-/-) mice are the primary targets of tumorigenesis in the syndrome of multiple endocrine neoplasia type-1 (MEN1). Mice with heterozygous disruption of the tumor suppressor Men1 gene (Men1(+/-)) develop tumors in the pituitary, pancreatic islets and other neuroendocrine tissues, which is analogous to humans with MEN1 mutations. To explore the genetic interactions between loss of Men1 and activation of CDKs, we examined the impact of Cdk4 or Cdk2 disruption on tumorigenesis in Men1(+/-) mice. A majority of Men1(+/-) mice with wild-type CDKs developed pituitary and islet tumors by 15 months of age. Strikingly, Men1(+/-); Cdk4(-/-) mice did not develop any tumors, and their islets and pituitaries remained hypoplastic with decreased proliferation. In contrast, Men1(+/-); Cdk2(-/-) mice showed pituitary and islet tumorigenesis comparable to those in Men1(+/-) mice. Pituitaries of Men1(+/-); Cdk4(-/-) mice showed no signs of loss of heterozygosity (LOH) in the Men1 locus, whereas tumors in Men1(+/-) mice and Men1(+/-); Cdk2(-/-) mice exhibited LOH. Consistently, CDK4 knockdown in INS-1 insulinoma cells inhibited glucose-stimulated cell cycle progression with a significant decrease in phosphorylation of retinoblastoma protein (RB) at specific sites including Ser780. CDK2 knockdown had minimum effects on RB phosphorylation and cell cycle progression. These data suggest that CDK4 is a critical downstream target of MEN1-dependent tumor suppression and is required for tumorigenic proliferation in the pituitary and pancreatic islet, whereas CDK2 is dispensable for tumorigenesis in these neuroendocrine cell types.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Insulinoma/metabolism , Pancreatic Neoplasms/metabolism , Pituitary Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 4/genetics , Female , Humans , Insulinoma/genetics , Insulinoma/pathology , Male , Mice , Mice, Knockout , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation/genetics , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Rats , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Transcription Factors/genetics
2.
Br J Cancer ; 108(12): 2495-504, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23736031

ABSTRACT

BACKGROUND: A 28 amino-acid (aa) cell-penetrating peptide (p28) derived from azurin, a redox protein secreted from the opportunistic pathogen Pseudomonas aeruginosa, produces a post-translational increase in p53 in cancer cells by inhibiting its ubiquitination. METHODS: In silico computational simulations were used to predict motifs within the p53 DNA-binding domain (DBD) as potential sites for p28 binding. In vitro direct and competitive pull-down studies as well as western blot and RT-PCR analyses were used to validate predictions. RESULTS: The L1 loop (aa 112-124), a region within the S7-S8 loop (aa 214-236) and T140, P142, Q144, W146, R282 and L289 of the p53DBD were identified as potential sites for p28 binding. p28 decreased the level of the E3 ligase COP1 >80%, in p53wt and p53mut cells with no decrease in COP1 in p53dom/neg or p53null cells. Brief increases in the expression of the E3 ligases, TOPORS, Pirh2 and HDM2 (human double minute 2) in p53wt and p53mut cells were in response to sustained increases in p53. CONCLUSION: These data identify the specific motifs within the DBD of p53 that bind p28 and suggest that p28 inhibition of COP1 binding results in the sustained, post-translational increase in p53 levels and subsequent inhibition of cancer cell growth independent of an HDM2 pathway.


Subject(s)
Azurin/pharmacology , Peptide Fragments/pharmacology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Animals , Azurin/chemistry , Azurin/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Mice , Mice, Nude , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding/drug effects , Protein Interaction Domains and Motifs/drug effects , Protein Interaction Domains and Motifs/physiology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/chemistry , Ubiquitin-Protein Ligases/antagonists & inhibitors , Xenograft Model Antitumor Assays
3.
Br J Cancer ; 108(5): 1061-70, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23449360

ABSTRACT

BACKGROUND: This first-in-human, phase I clinical trial of p28 (NSC745104), a 28-amino-acid fragment of the cupredoxin azurin, investigated the safety, tolerability, pharmacokinetics and preliminary activity of p28 in patients with p53(+) metastatic solid tumours. METHODS: A total of 15 patients were administered p28 i.v. as a short infusion three times per week for 4 weeks followed by a 2-week rest under an accelerated titration 3+3 dose escalation design until either a grade 3-related adverse event occurred or the maximum tolerated dose (MTD) was reached. Single-dose and steady-state serum pharmacokinetics were characterised. Assessments included toxicity, best objective response by RECIST 1.1 Criteria, and overall survival. RESULTS: No patients exhibited any dose-limiting toxicities (DLTs), significant adverse events or exhibited an immune response (IgG) to the peptide. The No Observed Adverse Effect Level (NOAEL) and MTD were not reached. Seven patients demonstrated stable disease for 7-61 weeks, three a partial response for 44-125 weeks, and one a complete response for 139 weeks. Three patients are still alive at 158, 140, and 110 weeks post therapy completion. CONCLUSION: p28 was tolerated with no significant adverse events. An MTD was not reached. Evidence of anti-tumour activity indicates a highly favourable therapeutic index and demonstrates proof of concept for this new class of non-HDM2-mediated peptide inhibitors of p53 ubiquitination.


Subject(s)
Antineoplastic Agents/therapeutic use , Azurin/adverse effects , Azurin/therapeutic use , Peptide Fragments/adverse effects , Peptide Fragments/therapeutic use , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Azurin/pharmacokinetics , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , No-Observed-Adverse-Effect Level , Peptide Fragments/pharmacokinetics , Tumor Suppressor Protein p53/metabolism , Ubiquitination
4.
J BUON ; 14(2): 189-96, 2009.
Article in English | MEDLINE | ID: mdl-19650165

ABSTRACT

Antitumor agents can inhibit tumor growth by 4 major cellular mechanisms; suppressing proliferation, inducing differentiation, killing the cells or forcing them to senescence. Senescent cells (CS) are in permanent paralysis because they are unable to divide, penetrate the surrounding tissues, metastasize, and respond to treatment. In this short review, we will focus on cellular senescence (CS) induced by retinoids in mammary pre-malignant and tumor cells and its potential clinical implication. Novel information is provided about the role of retinoic acid receptor beta 5 (RARbeta5) in mediating the retinoid-induced senescent program.


Subject(s)
Cell Differentiation/drug effects , Cellular Senescence/drug effects , Neoplasms/pathology , Animals , Humans , Neoplasms/drug therapy , Receptors, Retinoic Acid/metabolism , Retinoids/pharmacology
5.
Eur J Cancer ; 40(9): 1404-11, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15177500

ABSTRACT

In most prostate chemoprevention studies conducted with animal models, the incidence and multiplicity of tumours have been used as endpoints for efficacy. However, the latency of tumours is usually over 1 year, making these studies costly and time consuming. The main purpose of this study was to assess the utility of prostate intraepithelial neoplasia (PIN), induced in Noble rats by continuous testosterone + oestradiol (T + E) administration, as a potential intermediate endpoint biomarker of efficacy in chemoprevention studies. Noble rats at the age of 12 weeks were treated for 36 weeks with T + E given subcutaneously via Silastic capsules. The incidence and multiplicity of PIN were assessed in various prostate glands by serial sections generated at three separate tissue levels. The efficacy of dehydroepiandrosterone (DHEA) and DHEA 8354 (1000 and 2000 mg/kg diet), difluoromethylornithine (DFMO) (1000 and 2000 mg/kg diet) and oltipraz (125 and 250 mg/kg diet) to inhibit PIN was assessed in two independent sets of experiments. T + E induced multiple PIN in the dorsolateral prostate (DLP) of 80-100% of the animals. DHEA and DHEA 8354 did not affect the incidence but decreased the multiplicity of PIN in the DLP, from 3.2 +/- 1.0 in control group to 1.5 +/- 1.0 in the low-dose and to 1.6 +/- 0.6 in the high-dose group for DHEA (P<0.05 and P<0.02, respectively), and to 1.9 +/- 0.8 in the high-dose (P<0.05) DHEA 8354. Both agents did not affect PIN in anterior prostate, seminal vesicles or ventral prostate. In a second experiment, DFMO and oltipraz were found not effective in inhibiting PIN. In this study, we provide new evidence that PIN in Noble rats, induced by continuous T + E treatment, is a useful intermediate endpoint for determining the efficacy of DHEA and other potential chemopreventive agents. The hormonal pathogenesis, high multiplicity, short latency, preferential location in the DLP, similarity in morphology and biology to PIN of human prostate, and the sensitivity to agents that suppress prostate carcinogenesis, makes PIN in Noble rats a promising intermediate endpoint for chemoprevention studies.


Subject(s)
Dehydroepiandrosterone/analogs & derivatives , Models, Animal , Prostatic Intraepithelial Neoplasia , Prostatic Neoplasms , Animals , Antineoplastic Agents/therapeutic use , Dehydroepiandrosterone/therapeutic use , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Eflornithine/therapeutic use , Estradiol , Male , Prostatic Intraepithelial Neoplasia/chemically induced , Prostatic Intraepithelial Neoplasia/drug therapy , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Pyrazines/therapeutic use , Rats , Rats, Inbred Strains , Testosterone , Thiones , Thiophenes , Time Factors
6.
Eur J Cancer ; 39(2): 230-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12509956

ABSTRACT

Terminal replicative senescence (TRS) is a physiological process associated with terminal differentiation, shortening of the telomere, and lack of proliferative activity. Immortalised and tumour cells have lost their differentiation potential and the ability to develop a senescence phenotype. Recently, others and we [11] have observed that some antitumour agents and radiation induce a senescence-like phenotype (SLP) in human immortalized and tumour cell lines. The main purpose of this study was to identify senescence-like cells (SLC) in mammary tumours of rats and assess whether chemopreventive agents that have been used for the prevention and/or treatment of breast cancer can induce a SLP in tumour cells. Sprague-Dawley rats with N-methyl-N-nitrosourea (MNU)-induced mammary tumours were randomised and treated with tamoxifen, vorozole, 4-(hydroxyphenyl)retinamide (4-HPR), or 9-cis-retinoic acid (9cRA). The SLC in mammary tumours were identified and characterised by: (a) SA-beta-Gal staining method, which has been considered specific for human cells in TRS (b) staining for lipofuscin, which, although not specific, accumulates in the cytoplasm of cells in senescence; (c) lack of 5-Bromodeoxyuridine (BrdU) labelling after continuous (7 days) infusion of BrdU via osmotic pumps; (d) 90 degrees side light scatter (9OLS) as evaluated by flow cytometry; and (e) decreased telomerase activity. We found that in control tumours, SA-beta-Gal-positive cells were rare (below 1.0%) among the tumour cells, stroma fibroblast, myoepithelial and endothelial cells. SA-beta-Gal-positive cells increased significantly in the tumours treated with chemopreventive agents and this was associated with a lack of proliferative activity, increased cell granularity, lipofuscin accumulation, and decreased telomerase activity. Thus, in this study we provide for the first time evidence that cells in replicative senescence are present in mammary tumours of rats and that chemopreventive agents can suppress tumor growth by a novel cellular mechanism, inducing a SLP in the tumor cells.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Cellular Senescence , Mammary Neoplasms, Experimental/prevention & control , Tamoxifen/therapeutic use , Animals , Cytoplasmic Granules , Female , Flow Cytometry , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Phenotype , Rats , Rats, Sprague-Dawley , Telomerase/metabolism
7.
Cancer Res ; 61(20): 7456-63, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11606380

ABSTRACT

Trans-3,4',5-trihydroxystilbene (resveratrol), a phytoalexin present in grapes and grape products such as wine, has been identified as a chemopreventive agent. Recent studies performed with MCF-7 human breast cancer cells have demonstrated superestrogenic effects with resveratrol. In contrast, studies performed using estrogen receptor-transfected cell lines have shown that resveratrol acts as a mixed agonist/antagonist. The major objective of this study was to characterize the estrogen-modulatory effects of resveratrol in a variety of in vitro and in vivo mammary models. Thus, the effect of resveratrol alone and in combination with 17beta-estradiol (E2) was assessed with MCF-7, T47D, LY2, and S30 mammary cancer cell lines. With cells transfected with reporter gene systems, the activation of estrogen response element-luciferase was studied, and using Western blot analysis, the expression of E2-responsive progesterone receptor (PR) and presnelin 2 protein was monitored. Furthermore, the effect of resveratrol on formation of preneoplastic lesions (induced by 7,12-dimethylbenz(a)anthracene) and PR expression (with or without E2) was evaluated with mammary glands of BALB/c mice placed in organ culture. Finally, the effect of p.o. administered resveratrol on N-methyl-N-nitrosourea-induced mammary tumors was studied in female Sprague Dawley rats. As a result, in transient transfection studies with MCF-7 cells, resveratrol showed a weak estrogenic response, but when resveratrol was combined with E2 (1 nM), a clear dose-dependent antagonism was observed. Similar mixed estrogenic/antiestrogenic effects were noted with S30 cells, whereas resveratrol functioned as a pure estrogen antagonist with T47D and LY2 cells. Furthermore, in MCF-7 cells, resveratrol induced PR protein expression, but when resveratrol was combined with E2, expression of PR was suppressed. With T47D cells, resveratrol significantly down-regulated steady-state and E2-induced protein levels of PR. With LY2 and S30 cells, resveratrol down-regulated presnelin 2 protein expression. Using the mouse mammary organ culture model, resveratrol induced PR when administered alone, but expression was suppressed in the presence of E2 (1 nM). Furthermore, resveratrol inhibited the formation of estrogen-dependent preneoplastic ductal lesions induced by 7,12-dimethylbenz(a)anthracene in these mammary glands (IC50 = 3.2 microM) and reduced N-methyl-N-nitrosourea-induced mammary tumorigenesis when administered to female Sprague Dawley rats by gavage. Therefore, in the absence of E2, resveratrol exerts mixed estrogen agonist/antagonist activities in some mammary cancer cell lines, but in the presence of E2, resveratrol functions as an antiestrogen. In rodent models, carcinogen-induced preneoplastic lesions and mammary tumors are inhibited. These data suggest that resveratrol may have beneficial effects if used as a chemopreventive agent for breast cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Ductal, Breast/chemically induced , Mammary Neoplasms, Experimental/prevention & control , Proteins , Selective Estrogen Receptor Modulators/pharmacology , Stilbenes/pharmacology , Animals , Carcinogens , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/prevention & control , Estrogens/physiology , Female , Humans , Luciferases/genetics , Luciferases/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Methylnitrosourea , Mice , Mice, Inbred BALB C , Organ Culture Techniques , Protein Biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Receptors, Progesterone/biosynthesis , Response Elements/physiology , Resveratrol , Trefoil Factor-1 , Tumor Cells, Cultured , Tumor Suppressor Proteins
9.
Cancer Res ; 60(23): 6601-6, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11118041

ABSTRACT

The antiestrogen tamoxifen (TAM) is extensively metabolized by cytochrome P-450 in humans and rodents. The active, estrogen receptor-binding metabolites, 4-hydroxy TAM (OHT) and N-desmethyl TAM (DMT) have been well characterized. We showed that the s.c. injection of 1 mg/kg TAM in adult female Sprague Dawley rats bearing carcinogen-induced mammary tumors resulted in rapid serum decline of parent TAM but higher exposure of the metabolites, OHT and DMT. We found for the first time that the administration of TAM for a short time resulted in a delayed induction of caspase activity and apoptosis within the mammary tumors. When TAM, OHT, or DMT was added to human breast cancer cell lines in culture, each elicited a time- and dose-dependent induction of caspase activity, preceding apoptosis. Importantly, pretreatment of the cells with a pharmacological inhibitor of caspases [benzyloxy Val-Ala-Asp-fluoromethyl ketone (z-VAD-fmk)] blocked apoptosis induced by all three of the compounds, implicating a critical role of caspases in TAM-, OHT-, or DMT-induced apoptosis. The results obtained from these studies suggest that one possible mechanism of inhibition of mammary carcinogenesis and tumor growth in vivo may be the induction of caspase-dependent apoptosis, and that the metabolites OHT and DMT may contribute to the antitumor effect of TAM.


Subject(s)
Breast Neoplasms/metabolism , Caspases/metabolism , Estrogen Receptor Modulators/pharmacology , Mammary Neoplasms, Experimental/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Animals , Apoptosis/drug effects , Biotransformation , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Caspases/biosynthesis , Enzyme Activation/drug effects , Enzyme Induction , Estrogen Receptor Modulators/blood , Estrogen Receptor Modulators/pharmacokinetics , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Rats , Rats, Sprague-Dawley , Tamoxifen/blood , Tamoxifen/pharmacokinetics , Tumor Cells, Cultured
10.
J Natl Cancer Inst ; 92(22): 1836-40, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11078761

ABSTRACT

BACKGROUND: Although the active form of vitamin D, i.e., 1,25-dihydroxyvitamin D(3), is a potent cell-differentiating agent, its use in cancer prevention or therapy is precluded because it induces excessive blood calcium levels (hypercalcemia). However, less calcemic or noncalcemic synthetic analogues of vitamin D(3) are poorly effective against mammary carcinogenesis. We synthesized an analogue of vitamin D(5), 1alpha-hydroxy-24-ethylcholecalciferol (1alpha-hydroxyvitamin D(5)), which was less calcemic than 1,25-dihydroxyvitamin D(3) and prevented the development of precancerous lesions in mammary glands. Here, we evaluate its efficacy in an experimental rat mammary carcinogenesis model. METHODS: Sprague-Dawley rats were treated with 1alpha-hydroxyvitamin D(5) beginning 2 weeks before carcinogen treatment. Animals received an intravenous injection of N-methyl-N-nitrosourea at 80 days of age and continued to receive dietary 1alpha-hydroxyvitamin D(5) for an additional 105 days. Tumor incidence and multiplicity were determined, and plasma concentrations of calcium and phosphorus were measured. The efficacy of 1alpha-hydroxyvitamin D(5) at different stages of carcinogenesis was determined in mouse mammary gland organ culture. All statistical tests were two-sided. RESULTS: The tumor incidence was reduced from 80% (95% confidence interval [CI] = 51.9%-95.7%) in control rats to 53.3% (95% CI = 26.6%-78.8%) and 46.6% (95% CI = 21.3%-73.4%) in rats treated with 1alpha-hydroxyvitamin D(5) at 25 microg/kg diet and 50 microg/kg diet, respectively. The tumor multiplicity was reduced from 1.6 tumors per rat to 1.2 (95% CI for the difference = -0.45 to 1.25; P=.34) and 0.8 (95% CI for the difference = 0.14-1.46; P =.02), respectively. There was no statistically significant increase in the plasma calcium or phosphorus concentration at either dose level. The vitamin D(5) analogue was effective during both the initiation and the promotion stages of mammary lesion formation in organ culture. CONCLUSION: Our findings indicate that 1alpha-hydroxyvitamin D(5) reduces the incidence of mammary carcinogenesis in vivo. This analogue appears to be a good candidate for further development as a chemopreventive agent.


Subject(s)
Anticarcinogenic Agents/pharmacology , Hydroxycholecalciferols/pharmacology , Mammary Neoplasms, Experimental/prevention & control , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/adverse effects , Calcium/blood , Carcinogens , Confidence Intervals , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hydroxycholecalciferols/administration & dosage , Hydroxycholecalciferols/adverse effects , Incidence , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/chemically induced , Methylnitrosourea , Phosphorus/blood , Rats , Rats, Sprague-Dawley
11.
Oncogene ; 19(33): 3791-8, 2000 Aug 03.
Article in English | MEDLINE | ID: mdl-10949934

ABSTRACT

Mammalian tissues differ dramatically in their sensitivity to genotoxic stress, although the mechanisms determining these differences remain largely unknown. To analyse the role of p53 and p21 in determination of tissue specificity to DNA damage in vivo, we compared the effects of gamma radiation on DNA synthesis on whole-body sections of wild type, p53-deficient and p21-deficient mice. A dramatic reduction in 14C-thymidine incorporation after gamma irradiation was observed in the majority of rapidly proliferating tissues of wild type and p21-/- but not in p53-/- mice, confirming the key role of p53 in determination of tissue response to genotoxic stress in vivo and suggesting that p53-mediated inhibition of DNA synthesis does not depend on p21. Rapid radiation induced p53-dependent apoptosis was mapped to the areas of high levels of p53 mRNA in radiation sensitive tissues analysed (white pulp in the spleen and bases of crypts in small intestine), indicating that p53 regulation at the mRNA level is a determinant of cellular sensitivity to genotoxic stress. High p53 mRNA expression is inherited as a recessive trait in cell-cell hybrids suggesting the involvement of a negative control mechanism in the regulation of p53 gene expression.


Subject(s)
Cyclins/physiology , Tumor Suppressor Protein p53/physiology , 3T3 Cells , Animals , Apoptosis/radiation effects , Cell Division/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Gamma Rays , Gene Expression/radiation effects , Genes, Dominant , Genes, Recessive , Intestine, Small/cytology , Intestine, Small/pathology , Intestine, Small/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA, Messenger , Spleen/cytology , Spleen/pathology , Spleen/radiation effects , Tumor Suppressor Protein p53/genetics , Whole-Body Irradiation
12.
Cell Growth Differ ; 11(8): 437-45, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10965848

ABSTRACT

Previously, we found that vorozole (Vz), a nonsteroidal aromatase inhibitor, suppresses the development and progression of mammary tumors in rats. Here we evaluated for the first time the expression of cell death-related proteins Bcl-2 and Bax in hyperplastic, premalignant (carcinoma in situ), or malignant (carcinoma) lesions of mammary carcinogenesis; we also assessed whether these proteins are involved in mediating Vz-induced cell death in tumors. We found that Bcl-2 and Bax were equally expressed in epithelial cells of terminal end buds, ducts, and alveoli. However, in myoepithelial cells, the level of Bax expression was much higher than the level of Bcl-2 expression. Bcl-2 and Bax levels in hyperplastic lesions were similar to those of normal mammary epithelial cells but lower in most carcinomas in situ and carcinomas. In animals with established mammary tumors, Vz induced apoptotic cell death, which was primarily associated with a decrease in Bcl-2 and, to a lesser extent, with a decrease in Bax. These data support the hypothesis that Bcl-2 loss is more potent than Bax gain in regulating apoptotic cell death in mammary tumors.


Subject(s)
Carcinoma/metabolism , Epithelial Cells/metabolism , Hyperplasia/metabolism , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/metabolism , Precancerous Conditions/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis , Aromatase Inhibitors , Carcinoma/pathology , Female , Hyperplasia/pathology , Mammary Glands, Animal/anatomy & histology , Mammary Glands, Animal/cytology , Mammary Neoplasms, Animal/pathology , Precancerous Conditions/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Triazoles/pharmacology , bcl-2-Associated X Protein
13.
Breast Cancer Res Treat ; 60(2): 117-28, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10845274

ABSTRACT

Vorozole (Vz) is a competitive non-steroidal inhibitor of aromatase, which has been used to treat breast cancer in postmenopausal women and in various chemoprevention pre-clinical studies. Recently, we assessed the inhibitory effect of Vz on MNU-induced mammary carcinogenesis (Lubet et al., 1994), as well as on the progression of mammary tumors (Lubet et al., 1998). In this study we evaluated the effects of Vz on tumor growth, serum estradiol, cell proliferation, apoptotic and non-apoptotic cell death to determine whether any of these 'surrogate' markers might reflect the efficacy of various doses of Vz. Vz at doses of 2.5 (Hi), 0.32 (Md), and 0.08 (Lo) mg/kg body weight induced complete (100%), 60%, and 20% regression of mammary tumors, respectively. Vz at Hi and Md doses caused a decrease in serum estradiol within the first two days of treatment, and the estradiol values remained low with additional treatment for 4 and 10 days. When Vz was administered to animals bearing palpable tumors a time and dose-dependent decrease in the proliferating cells (BrdU-L1) was observed. The percentage of apoptotic cells (A1) sharply increased 2 days after initiation of Vz treatment and then decreased followed by an increase in non-apoptotic dead cells. Interestingly even the Lo dose of Vz, which was only moderately effective in suppressing tumor growth, decreased cell proliferation and increased cell death in the peripheral tumor areas at 4 and 10 days after initiation of treatment. The time- and dose-dependent alterations in various cell parameters suggest two different phases of Vz-induced cellular responses: (1) an early phase (2-4 days of treatment) with a sharp increase in apoptotic cells and decrease in proliferating cells, and (2) a later phase (10 days) with disintegration of tumor parenchyma, increase in non-apoptotic dead cells, and decrease in apoptotic cells. The dose-dependent decrease in proliferating cells and increase in apoptotic and non-apoptotic cell death in Vz-treated animals suggest that these biomarkers might be used as potential surrogate endpoints for efficacy in breast cancer chemoprevention and therapy studies with aromatase inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aromatase Inhibitors , Enzyme Inhibitors/pharmacology , Mammary Neoplasms, Experimental/pathology , Triazoles/pharmacology , Animals , Biomarkers, Tumor/analysis , Cell Division/drug effects , Dose-Response Relationship, Drug , Estradiol/blood , Female , Rats , Rats, Sprague-Dawley
14.
Oncogene ; 19(15): 1875-84, 2000 Apr 06.
Article in English | MEDLINE | ID: mdl-10773877

ABSTRACT

p27Kip1 (p27) controls cell cycle progression by binding to and inhibiting the activity of cyclin dependent kinases. Disruption of the p27 gene in mice (p27-/-) results in increased body growth with a disproportionate enlargement of the spleen, thymus, testis, ovary and pituitary. The increase in pituitary size is due to selective hyperplasia of the intermediate lobe (IL) while the anterior lobe (AL) is not overtly affected. p27 heterozygous mice (p27+/-), as well as p27-/- mice, are hypersensitive to radiation- and chemical-induced tumors compared to wildtype (p27+/+) littermates. Therefore, unlike classical tumor suppressors, only a reduction in p27 levels is necessary to predispose tissues to secondary tumor promoters. Consistent with these studies is the fact that the p27 gene sequence and mRNA levels appear normal in human pituitary adenomas while p27 protein levels are decreased. Therefore, a reduction in p27 levels could be sufficient to sensitize pituitary cells to tumorigenic factors. To test this hypothesis, metallothionein promoter-driven, human growth hormone-releasing hormone (MT-hGHRH) transgenic mice, that exhibit somatotrope hyperplasia before 9 months of age and subsequent adenoma formation with 30 - 40% penetrance, were crossbred with p27+/- mice for two successive generations to produce p27+/+, p27+/- and p27-/- mice that expressed the hGHRH transgene. At 10 - 12 weeks of age, p27-/- and p27+/+, hGHRH mice were larger than their p27+/+ littermates and displayed characteristic hyperplasia of the IL and AL, respectively. Expression of the hGHRH transgene in both p27+/- and p27-/- mice selectively expanded the population of somatotropes within the AL, where pituitaries of p27+/-, hGHRH and p27-/-, hGHRH mice were two- and fivefold larger than p27+/+, hGHRH pituitaries, respectively. There was also a synergistic effect of hGHRH transgene expression and p27-deficiency on liver, spleen and ovarian growth. At 6 - 8 months of age, 83% of p27+/-, hGHRH mice displayed macroscopic AL adenomas (>100 mg), while all pituitaries from p27+/+, hGHRH mice remained hyperplastic (<20 mg). In contrast to the dramatic effects of p27-deficiency on hGHRH-induced organ growth, elimination of p53, by crossbreeding MT-hGHRH mice to p53-deficient mice, did not augment the hyperplastic/tumorigenic effects of hGHRH transgene expression. Taken together these results demonstrate that a reduction in p27 expression is sufficient to sensitize somatotropes to the proliferative actions of excess GHRH, resulting in the earlier appearance and increased penetrance of hGHRH-induced pituitary tumors.


Subject(s)
Adenoma/genetics , Cell Cycle Proteins , Cyclin-Dependent Kinases/antagonists & inhibitors , Growth Hormone-Releasing Hormone/pharmacology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Pituitary Neoplasms/genetics , Tumor Suppressor Proteins , Age Factors , Animals , Cyclin-Dependent Kinase Inhibitor p27 , Genes, Tumor Suppressor , Humans , Hyperplasia , Liver/pathology , Mice , Mice, Transgenic , Pituitary Gland/pathology , Spleen/pathology
15.
Carcinogenesis ; 21(2): 227-33, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10657962

ABSTRACT

Previous studies have shown that terminal end buds (TEBs) in the murine mammary gland have high proliferative activity and demonstrate apoptotic cell death (ACD). Since TEBs are considered the place of origin of most chemically induced mammary carcinomas, we hypothesized that the development of hyperplastic and premalignant (carcinoma in situ, CIS) lesions in TEBs is associated with either a further increase in cell proliferation and/or with a decrease in ACD. To test this hypothesis we used the N-methyl-N-nitorosourea (MNU) carcinogenesis model in rats, where the occurrence of mammary tumors is preceded by hyperplastic and premalignant lesions arising mostly in TEBs, as well as in ducts and alveoli. The percentage of proliferating cells, as evaluated by 5-bromodeoxyuridine labeling (BrdU-LI), was similar in TEBs to those in terminal endbud hyperplasia (TEBH), CIS, and carcinomas (CA), whereas the percentage of apoptotic cells (apoptotic index, AI) was relatively high in TEBs and decreased in TEBH, CIS, and CA. This indicates that neoplastic transformation of mammary epithelial cells in TEBs is not associated with an increase in cell proliferation, but with a decrease in ACD. In addition to TEBH, hyperplastic lesions developed in ductal branching areas (ductal hyperplasia, DH) and alveolar structures (alveolar hyperplasia, AH). However, BrdU-LI in both DH and AH was lower than in TEBH, whereas the AI values were similar, suggesting that TEBH has a higher potential for progression and malignant transformation than DH and AH. In mammary tumors apoptotic cells were rare in the peripheral, proliferative areas, but frequent close to the necrotic areas, suggesting that intratumoral factors may significantly affect ACD. Thus, it appears that dissociation between cell proliferation and apoptosis occurs in the hyperplastic stages of mammary carcinogenesis and that neoplastic transformation of mammary epithelial cells is associated with decreased ACD but not with increased cell proliferation.


Subject(s)
Apoptosis , Carcinoma in Situ/pathology , Carcinoma, Ductal, Breast/pathology , Cell Transformation, Neoplastic/pathology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Animals , Carcinogens/toxicity , Carcinoma in Situ/chemically induced , Carcinoma, Ductal, Breast/chemically induced , Cell Division , Cell Transformation, Neoplastic/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Hyperplasia , Mammary Glands, Animal/drug effects , Mammary Neoplasms, Experimental/chemically induced , Methylnitrosourea/toxicity , Necrosis , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology , Rats , Rats, Sprague-Dawley
16.
Cancer Res ; 59(15): 3761-7, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10446993

ABSTRACT

Exposure of human tumor cell lines to different chemotherapeutic drugs, ionizing radiation, and differentiating agents induced morphological, enzymatic, and ploidy changes resembling replicative senescence of normal cells. Moderate doses of doxorubicin induced this senescence-like phenotype (SLP) in 11 of 14 tested cell lines derived from different types of human solid tumors, including all of the lines with wild-type p53 and half of p53-mutated cell lines. SLP induction seemed to be independent from mitotic cell death, the other major effect of drug treatment. Among cells that survived drug exposure, SLP markers distinguished those cells that became terminally growth-arrested within a small number of cell divisions from the cells that recovered and resumed proliferation. SLP induction in breast carcinoma cells treated with retinoids in vitro or in vivo was found to correlate with permanent growth inhibition under the conditions of minimal cytotoxicity, suggesting that this response may be particularly important for the antiproliferative effect of differentiating agents. The senescence-like program of terminal proliferation arrest may provide an important determinant of treatment outcome and a target for augmentation in cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cellular Senescence/drug effects , Neoplastic Stem Cells/drug effects , Adenocarcinoma/pathology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/pathology , Doxorubicin/pharmacology , Female , Fibrosarcoma/pathology , Gamma Rays , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Phenotype , Ploidies , Tretinoin/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology , Tumor Cells, Cultured/radiation effects
17.
Carcinogenesis ; 20(8): 1535-40, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10426803

ABSTRACT

In most previous chemoprevention studies on inhibition of mammary carcinogenesis, the formation of palpable tumors has been used as an end-point. Little is known about whether chemopreventive agents may similarly or selectively suppress hyperplastic and premalignant stages of the neoplastic process. In this study, we evaluated the effect of 4-(hydroxyphenyl)retinamide (4-HPR) on the development and progression of hyperplastic lesions and carcinoma in situ (CIS) in the N-methyl-N-nitrosourea (MNU) mammary carcinogenesis model in rats. 4-HPR was used as the chemopreventive agent because of its proven inhibitory effect on both the early and late phases of mammary carcinogenesis. Treatment with 4-HPR (2.0 mM/kg diet), beginning 2 days after MNU administration and administered continuously for 10 weeks, suppressed all mammary gland lesions (hyperplasia, CIS and invasive carcinoma) in 35% of animals. In the remaining 65%, 4-HPR allowed the development of hyperplastic lesions, alone or combined with CIS, and/or invasive carcinomas (CA). 4-HPR also increased by 2-fold the ratio between CIS and CA (0.75 per animal in control versus 1.5 in 4-HPR-treated animals), suggesting that it may also suppress the transition of CIS into CA. 4-HPR, when administered beginning 4 weeks after MNU administration [when hyperplastic and premalignant (CIS) lesions are present in the mammary gland], inhibited the frequency of terminal end bud hyperplasia (TEBH) and CA but did not significantly suppress ductal hyperplasia, ductal alveolar hyperplasia, alveolar hyperplasia and CIS. In these animals, 4-HPR induced partial disintegration of mostly peripheral areas of lesions, including carcinomas. Taken together, our data indicate that 4-HPR selectively suppresses the development and progression of hyperplastic lesions and CIS in TEBs. Furthermore, it appears that, in addition to mammary carcinomas, TEBH and CIS could also be used as end-point biomarkers in breast cancer chemoprevention studies.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Carcinoma in Situ/prevention & control , Fenretinide/therapeutic use , Mammary Glands, Animal/drug effects , Mammary Neoplasms, Experimental/prevention & control , Precancerous Conditions/prevention & control , Animals , Carcinogens , Carcinoma in Situ/pathology , Disease Progression , Drug Screening Assays, Antitumor , Female , Hyperplasia/chemically induced , Hyperplasia/pathology , Hyperplasia/prevention & control , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Methylnitrosourea , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology , Rats , Rats, Sprague-Dawley
18.
Carcinogenesis ; 20(5): 879-83, 1999 May.
Article in English | MEDLINE | ID: mdl-10334206

ABSTRACT

The detection of telomerase activity has been proposed as a biomarker of breast cancer development and progression. In this study, we used cell proliferation and telomerase in MNU (N-methyl-N-nitrosourea)-induced mammary carcinomas as targets for assessing the response of tumor cells to 4-(hydroxyphenyl)retinamide (4-HPR), a known inhibitor of mammary carcinogenesis in animal models and premenopausal women. In mammary tumors of rats treated for 1, 2, 4 or 6 weeks with 4-HPR, we observed that telomerase activity decreased progressively with the extension of 4-HPR administration. A marked reduction in telomerase activity was already observed by 2 weeks after treatment and the lowest level was found at 6 weeks after initiation of 4-HPR treatment. The changes in telomerase activity were preceded and accompanied by a significant decrease in the percentage of proliferating cells as evaluated by 5-bromodeoxyuridine (BrdU)-labeling. However, when the values of telomerase activity in the individual tumors were compared with the percentage of proliferating cells, no significant correlation was found. These data suggest that the decreased telomerase activity in the animals treated with 4-HPR is not a simple consequence of the changes in cell proliferation, but a more complex phenomenon involving different cellular mechanisms and pathways. The time-dependent and consistent decrease of telomerase activity in the tumors treated with 4-HPR suggests that, in addition to the percentage of proliferating cells, telomerase activity could also be used as an endpoint in breast cancer chemotherapy studies.


Subject(s)
Antineoplastic Agents/pharmacology , Fenretinide/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Telomerase/drug effects , Animals , Antineoplastic Agents/therapeutic use , Cell Division/drug effects , Female , Fenretinide/therapeutic use , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Rats , Rats, Sprague-Dawley , Telomerase/metabolism , Time Factors
19.
Carcinogenesis ; 17(8): 1741-6, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8761435

ABSTRACT

In the present study, pituitary isografted animals serve as a model for evaluating the changes in differentiation, cell proliferation and programmed cell death (apoptosis) in mammary epithelial cells during carcinogenesis. The percentage of bromodeoxyuridine (BrdU)-labeled ductal and alveolar cells was significantly higher in pituitary isografted animals than in non-isografted control animals. BrdU-labeled cells increased in lobular hyperplastic nodules, keratinized nodules and mammary carcinomas; similar changes were observed with apoptotic cells, which were rare in mammary glands of adult non-isografted animals (one to three apoptotic cells per 2000 mammary epithelial cells), but their number increased in hyperplastic lesions and mammary carcinomas. Among hyperplastic nodular lesions, variants with high, moderate and low proliferative activity and/or apoptotic cell death were identified, which suggests that they may have different growth potentials and different propensities for malignant transformation. After removing pituitary isografts, apoptosis occurs in hyperplastic lesions but not in mammary carcinomas-implying that malignant tumors are hormone-independent. The dynamics of the changes in apoptotic cell death among various hyperplastic lesions after removal of pituitary isografts suggests that these lesions are composed of heterogeneous cell populations, as far as the initiation of apoptosis is concerned. Our data indicate that apoptosis can be used together with cell proliferation as a potential marker in characterizing the growth potential and phenotypic diversity of hyperplastic, premalignant and malignant mammary gland lesions.


Subject(s)
Apoptosis , Mammary Neoplasms, Experimental/pathology , Pituitary Gland, Anterior/transplantation , Animals , Cell Division , Estradiol/blood , Female , Hyperplasia , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/blood , Mice , Mice, Inbred BALB C , Precancerous Conditions/blood , Precancerous Conditions/pathology , Progesterone/blood , Prolactin/blood , Transplantation, Isogeneic
20.
Acta Anat (Basel) ; 152(3): 195-204, 1995.
Article in English | MEDLINE | ID: mdl-7572029

ABSTRACT

The goal of this research was to establish methods for inducing mammary epithelial differentiation from nonmammary epithelium. For this purpose, mid-ventral or dorsal epidermis (skin epithelium; SKE) from 13-day rat or mouse embryos was associated with 13-day embryonic mouse mammary mesenchyme (mammary gland mesenchyme; MGM) (mouse MGM+rat or mouse SKE). The resultant MGM+SKE recombinants as well as controls (homotypic mouse mammary recombinants, homotypic mouse skin recombinants and mouse mammary mesenchyme by itself) were grafted under the renal capsule of syngeneic or athymic female nude mouse hosts. Most female hosts were induced to undergo lactogenesis by grafting an adult pituitary which elicited a state of hyperprolactinemia. Tissue recombinants of mouse MGM+rat or mouse SKE grown for 1 month in vivo formed a hair-bearing keratinized skin from which mammary ductal structures extended into the mesenchyme. The ducts were composed of columnar luminal epithelial cells as well as basal, actin-positive myoepithelial cells. When grown in pituitary-grafted hosts, the ductal epithelial cells expressed casein and alpha-lactalbumin as judged by immunocytochemistry. The expression of caseins in MGM+SKE recombinants was confirmed by Western blot. The epithelial cells in mouse MGM+rat SKE recombinants expressing milk proteins were shown to be rat cells while the surrounding connective tissue was composed of mouse cells based upon staining with Hoechst dye 33258. Using mammary-specific markers, these studies confirmed the earlier morphological studies of Propper and unequivocally demonstrated for the first time that embryonic mammary mesenchyme can induce morphological and functional mammary differentiation from nonmammary epithelium.


Subject(s)
Embryonic Induction , Epidermis/embryology , Mammary Glands, Animal/embryology , Mesoderm/physiology , Animals , Blotting, Western , Cell Differentiation , Chimera , Epidermal Cells , Epidermis/metabolism , Female , Lactation , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred BALB C , Milk Proteins/biosynthesis , Morphogenesis , Phenotype , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL