Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Acta Neurol Scand ; 117(4): 273-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17922892

ABSTRACT

BACKGROUND: Narcolepsy typically begins between adolescence and early adulthood causing severe neuropsychiatric impairments, but few prevalence studies are available on adolescent narcoleptics. In the present study, we investigated the prevalence of narcolepsy-cataplexy in adolescents. METHODS: In total 20,407 students, aged 14-19 years, participated in this study. Ullanlinna Narcolepsy Scale (UNS) was applied to all subjects and those with a UNS score of > or =14 were contacted by phone for semi-structured interview. Subjects then suspected of having narcolepsy participated in a laboratory investigation, which included polysomnography and HLA typing, or were interviewed in detail by telephone. RESULTS: Three subjects were finally diagnosed as narcolepsy with cataplexy and seven subjects might be diagnosed as narcolepsy without cataplexy. Among three narcoleptics with cataplexy, two subjects were HLA-DQB1*0602 and DRB1*1501 positive, but one subject had no test of HLA typing. The prevalence of narcolepsy with cataplexy in Korean adolescence was thus determined to be 0.015% (95% confidence interval = 0.0-0.0313%). CONCLUSION: This epidemiologic study is the first of its type on adolescent narcolepsy to use the International Classification of Sleep Disorders, 2nd edition (ICSD-2) diagnostic criteria. Considering those cases with an onset after adolescence were not included, the prevalence of narcolepsy with cataplexy determined in the present study is comparable with that of other studies in adults.


Subject(s)
Narcolepsy/diagnosis , Narcolepsy/epidemiology , Adolescent , Adult , Age of Onset , Asian People , Diagnosis, Differential , Female , Genotype , HLA Antigens/genetics , Humans , Interviews as Topic , Korea/epidemiology , Male , Narcolepsy/physiopathology , Polysomnography , Predictive Value of Tests , Prevalence , Surveys and Questionnaires
2.
Oncogene ; 26(38): 5655-61, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-17334390

ABSTRACT

Rapamycin, a natural product inhibitor of the Raptor-mammalian target of rapamycin complex (mTORC1), is known to induce Protein kinase B (Akt/PKB) Ser-473 phosphorylation in a subset of human cancer cell lines through inactivation of S6K1, stabilization of insulin receptor substrate (IRS)-1, and increased signaling through the insulin/insulin-like growth factor-I/phosphatidylinositol 3-kinase (PI3K) axis. We report that A-443654, a potent small-molecule inhibitor of Akt serine/threonine kinases, induces Akt Ser-473 phosphorylation in all human cancer cell lines tested, including PTEN- and TSC2-deficient lines. This phenomenon is dose-dependent, manifests coincident with Akt inhibition and likely represents an alternative, rapid-feedback pathway that can be functionally dissociated from mTORC1 inhibition. Experiments performed in TSC2-/- cells indicate that TSC2 and IRS-1 cooperate with, but are dispensable for, A-443654-mediated Akt phosphorylation. This feedback event does require PI3K activity, however, as it can be inhibited by LY294002 or wortmannin. Small interfering RNA-mediated knockdown of mTOR or Rictor, components of the rapamycin-insensitive mTORC2 complex, but not the mTORC1 component Raptor, also inhibited Akt Ser-473 phosphorylation induced by A-443654. Our data thus indicate that Akt phosphorylation and activity are coupled in a manner not previously appreciated and provide a novel mode of Akt regulation that is distinct from the previously described rapamycin-induced IRS-1 stabilization mechanism.


Subject(s)
Indazoles/pharmacology , Indoles/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Transcription Factors/metabolism , Cell Line, Tumor , Chromones/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Immunoblotting , Mechanistic Target of Rapamycin Complex 1 , Morpholines/pharmacology , Multiprotein Complexes , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proteins , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Serine/metabolism , TOR Serine-Threonine Kinases , Time Factors , Transcription Factors/genetics , Transfection , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
Neoplasia ; 3(5): 411-9, 2001.
Article in English | MEDLINE | ID: mdl-11687952

ABSTRACT

Checkpoint kinase 1 (Chk1) is a checkpoint gene that is activated after DNA damage. It phosphorylates and inactivates the Cdc2 activating phosphatase Cdc25C. This in turn inactivates Cdc2, which leads to G2/M arrest. We report that blocking Chk1 expression by antisense or ribozymes in mammalian cells induces apoptosis and interferes with the G2/M arrest induced by adriamycin. The Chk1 inhibitor UCN-01 also blocks the G2 arrest after DNA damage and renders cells more susceptible to adriamycin. These results indicate that Chk1 is an essential gene for the checkpoint mechanism during normal cell proliferation as well as in the DNA damage response.


Subject(s)
Apoptosis , Enzyme Inhibitors/pharmacology , G2 Phase/physiology , Oxazines , Protein Kinase Inhibitors , Xanthenes , Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Caspases/metabolism , Cell Cycle/drug effects , Checkpoint Kinase 1 , Coloring Agents , DNA Primers/chemistry , DNA, Antisense/pharmacology , Doxorubicin/pharmacology , Drug Resistance , Etoposide/pharmacology , Flow Cytometry , Humans , Lung Neoplasms/metabolism , Mitosis , Protein Kinases/metabolism , RNA, Catalytic/pharmacology , Staurosporine/analogs & derivatives , Tumor Cells, Cultured
4.
Neoplasia ; 3(4): 278-86, 2001.
Article in English | MEDLINE | ID: mdl-11571628

ABSTRACT

The serine/threonine kinases, Akt1/PKBalpha, Akt2/PKBbeta, and Akt3/PKBgamma, play a critical role in preventing cancer cells from undergoing apoptosis. However, the function of individual Akt isoforms in the tumorigenicity of cancer cells is still not well defined. In the current study, we used an Akt1 antisense oligonucleotide (AS) to specifically downregulate Akt1 protein in both cancer and normal cells. Our data indicate that Akt1 AS treatment inhibits the ability of MiaPaCa-2, H460, HCT-15, and HT1080 cells to grow in soft agar. The treatment also induces apoptosis in these cancer cells as demonstrated by FACS analysis and a caspase activity assay. Conversely, Akt1 AS treatment has little effect on the cell growth and survival of normal human cells including normal human fibroblast (NHF), fibroblast from muscle (FBM), and mammary gland epithelial 184B5 cells. In addition, Akt1 AS specifically sensitizes cancer cells to typical chemotherapeutic agents. Thus, Akt1 is indispensable for maintaining the tumorigenicity of cancer cells. Inhibition of Akt1 may provide a powerful sensitization agent for chemotherapy specifically in cancer cells.


Subject(s)
Apoptosis , Oxazines , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Tumor Cells, Cultured/pathology , Xanthenes , Antineoplastic Agents/pharmacology , Blotting, Western , Caspases/metabolism , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Colony-Forming Units Assay , Coloring Agents , Cytochrome c Group/metabolism , Down-Regulation , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Humans , Oligonucleotides, Antisense/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt , Tumor Cells, Cultured/metabolism
5.
Cancer Res ; 61(14): 5480-5, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454695

ABSTRACT

Drug resistance is a prevalent problem in the treatment of neoplastic disease, and the effectiveness of many clinically useful drugs is limited by the fact that they are substrates for the efflux pump, P-glycoprotein. Because there is a need for new compounds that are effective in treating drug-resistant tumors, we tested A-204197 (4-[4-acetyl-4,5-dihydro-5-(3,4,5-trimethoxyphenyl)-1,3,4-oxadiazol-2-yl]-N,N-dimethylbenzeneamine), a novel oxadiazoline derivative with antiproliferative properties, on cell lines that were either sensitive or resistant to known microtubule inhibitors. Cell lines that were resistant to paclitaxel, vinblastine, or colchicine were equally sensitive to A-204197 (proliferation IC50s ranging from 36 to 48 nM) despite their expression levels of P-glycoprotein. The effect of A-204197 on cell growth was associated with cell cycle arrest in G2-M, increased phosphorylation of select G2-M checkpoint proteins, and apoptosis. In competition-binding assays, A-204197 competed with [3H]-labeled colchicine for binding to tubulin (K(i) = 0.75 microM); however, it did not compete with [3H]-labeled paclitaxel. A-204197 prevented tubulin polymerization in a dose-dependent manner (IC50 = 4.5 microM) in vitro and depolymerized microtubules in a time-dependent manner in cultured cells. These findings indicate A-204197 is a promising new tubulin-binding compound with antimitotic activity that has potential for treating neoplastic diseases with greater efficacy than currently used antimitotic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Microtubules/drug effects , Oxadiazoles/pharmacology , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Binding Sites , Cell Cycle/drug effects , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Colchicine/metabolism , Colchicine/pharmacology , Drug Interactions , Drug Resistance, Multiple , G2 Phase/drug effects , Humans , Microtubules/metabolism , Mitosis/drug effects , Oxadiazoles/metabolism , Paclitaxel/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors , Tubulin/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Vinblastine/pharmacology
6.
Anticancer Res ; 21(1A): 23-8, 2001.
Article in English | MEDLINE | ID: mdl-11299740

ABSTRACT

BACKGROUND: Chkl is a checkpoint gene that is activated after DNA damage. It phosphorylates and inactivates Cdc25C at the late G2 phase. The inactivation of Cdc25C and consequently, the inactivation of Cdc2, are required for the G2 arrest induced by DNA damage. METHODS: We treated 184B5 cell line and its E6 transformed cell lines with adriamycin in the presence of staurosporine or UCNO1 and examined G2 arrest and cell death. RESULTS: We found that adriamycin induced a p53 and p21 response as well as a G1 arrest in 184B5 cells, but not in its E6 transformed cells. Staurosporine or UCNO1 abrogated the G2 arrest induced by adriamycin in both cell lines. In addition, staurosporine or UCNO1 specifically sensitized p53 incompetent cells to adriamycin. CONCLUSION: G2/M checkpoint abrogators can potentially enhance the cytotoxic effect of conventional chemotherapeutic reagents specifically to tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage/drug effects , Doxorubicin/pharmacology , Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Alkaloids/pharmacology , Apoptosis/drug effects , Cell Line, Transformed , Checkpoint Kinase 1 , Enzyme Inhibitors/pharmacology , G2 Phase , Humans , Neoplasms/metabolism , Protein Kinases/metabolism , Staurosporine/pharmacology , Tumor Cells, Cultured
7.
Cancer Res ; 61(4): 1486-92, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245455

ABSTRACT

A high-throughput screening assay was designed to select compounds that inhibit the growth of cultured mammalian cells. After screening more than 60,000 compounds, A-105972 was identified and selected for further testing. A-105972 is a small molecule that inhibits the growth of breast, central nervous system, colon, liver, lung, and prostate cancer cell lines, including multidrug-resistant cells. The cytotoxic IC50 values of A-105972 were between 20 and 200 nM, depending on the specific cell type. The potency of A-105972 is similar in cells expressing wild-type or mutant p53. A majority of cells treated with A-105972 were trapped in the G2-M phases, suggesting that A-105972 inhibits the progression of the cell cycle. Using [3H]A-105972, we found that A-105972 bound to purified tubulin. Unlabeled A-105972 competed with [3H]A-105972 binding with an IC50 value of 3.6 microL. Colchicine partially inhibited [3H]A-105972 binding with an IC50 value of approximately 90 microM, whereas paclitaxel and vinblastine had no significant effect. Tumor cells treated with A-105972 were observed to contain abnormal microtubule arrangement and apoptotic bodies. DNA ladder studies also indicated that A-105972 induced apoptosis. A-105972 caused a mobility shift of bcl-2 on SDS-PAGE, suggesting that A-105972 induced bcl-2 phosphorylation. A-105972 treatment increased the life span of mice inoculated with B16 melanoma, P388 leukemia, and Adriamycin-resistant P388. These results suggest that A-105972 is a small molecule that interacts with microtubules, arrests cells in G2-M phases, and induces apoptosis in both multidrug resistance-negative and multidrug resistance-positive cancer cells. A-105972 and its analogues may be useful for treating cell proliferative disorders such as cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Oxadiazoles/pharmacology , Animals , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Inhibitory Concentration 50 , Leukemia P388/drug therapy , Leukemia P388/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Microtubules/drug effects , Microtubules/metabolism , Oxadiazoles/metabolism , Phosphorylation/drug effects , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Tubulin/metabolism , Tumor Cells, Cultured/drug effects
8.
Eur J Cancer ; 36(12): 1565-71, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10930805

ABSTRACT

Beta(beta)-tubulin isotype variation has recently been implicated in the modulation of resistance to paclitaxel in human lung cancer cells and in primary human ovarian tumour samples. Whether alpha-tubulin is involved in drug resistance has not been reported. We have generated a paclitaxel-resistant cell line (H460/T800) from the sensitive human lung carcinoma parental cell line NCI-H460. The resistant cells are more than 1000-fold resistant to taxol and overexpress P-glycoprotein. Interestingly, H460/T800 cells also overexpress alpha- and beta-tubulin as detected by Western blot analysis. From Northern blot analysis, the mechanism of tubulin overexpression appears to be post-transcriptional. To understand whether alpha-tubulin plays a role in drug resistance, we transfected antisense human kalpha1 cDNA construct into the H460/T800 paclitaxel-resistant cells. The antisense clones displayed a reduced alpha-tubulin expression, and the cells were 45-51% more sensitive to paclitaxel and other known antimitotic drugs, compared with vector transfected controls. Complementary experiments of transfecting the sense kalpha1 cDNA into H460 cells conferred a 1.8- to 3.3-fold increase in the IC(50) of several antimitotic agents. Our study suggests that alpha-tubulin is one of the factors that contributes to drug resistance.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Lung Neoplasms/drug therapy , Paclitaxel/therapeutic use , Tubulin/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blotting, Northern , Blotting, Western , Cell Cycle , Cell Division , DNA, Antisense/genetics , DNA, Complementary/genetics , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Transfection , Tubulin/metabolism , Tumor Cells, Cultured
9.
Br J Cancer ; 83(1): 83-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10883672

ABSTRACT

A recurring problem with cancer therapies is the development of drug resistance. While investigating the protein profile of cells resistant to a novel antimitotic compound (A204197), we discovered an increase in annexin IV expression. When we examined the annexin IV protein expression level in a paclitaxel-resistant cell line (H460/T800), we found that annexin IV was also overexpressed. Interestingly a closely related protein, annexin II, was not overexpressed in H460/T800 cells. Immunostaining with either annexin II or IV antibody revealed that annexin IV was primarily located in the nucleus of paclitaxel-resistant H460/T800 cells. Short-term treatment of H460 cells with 10 nM paclitaxel for up to 4 days resulted in induction of annexin IV, but not annexin II expression. In addition, there was an increase in annexin IV staining in the nucleus starting at day 1. Furthermore, cells pretreated with 10 nM paclitaxel for 4 days resulted in cells becoming approximately fivefold more resistant to paclitaxel. Transfection of annexin IV cDNA into 293T cells revealed that there was a threefold increase in paclitaxel resistance. Thus our results indicate that annexin IV plays a role in paclitaxel resistance in this cell line and it is among one of the earliest proteins that is induced in cells in response to cytotoxic stress such as antimitotic drug treatment.


Subject(s)
Annexin A4/physiology , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm , Lung Neoplasms/pathology , Neoplasm Proteins/physiology , Paclitaxel/pharmacology , Annexin A4/biosynthesis , Annexin A4/genetics , Blotting, Western , Colchicine/pharmacology , Colonic Neoplasms/metabolism , DNA, Complementary/genetics , Drug Resistance, Multiple , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation, Neoplastic , Growth Inhibitors/pharmacology , Humans , Lung Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nocodazole/pharmacology , Recombinant Fusion Proteins/biosynthesis , Transfection , Tubulin Modulators , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
10.
Oncogene ; 18(48): 6658-66, 1999 Nov 18.
Article in English | MEDLINE | ID: mdl-10597271

ABSTRACT

To clarify the roles of specific isoforms of PKC in regulating growth and cell cycle progression of the HC11 mammary epithelial cell line, we investigated the effects of activating endogenous PKC isoforms with the phorbol ester tumor promoter TPA, and also the effects of TPA on genetically engineered cells containing increased levels of individual PKC isoforms. We found that TPA treatment of HC11 cells induced a transient cell cycle arrest in G0/G1. Western blot analyses of the TPA treated cells provided evidence that the endogenous PKC alpha present in these cells mediated these effects. Indeed, derivatives of the HC11 cell line that inducibly overexpress an exogenous PKC alpha or ectopic PKC beta 1 exhibited more marked growth inhibition by TPA than control cells. Immunohistochemical staining of cells following treatment with TPA revealed selective translocation of PKC alpha into the nucleus, whereas PKC beta 1 remained in the cytoplasm. The transient arrest of HC11 cells following treatment with TPA was associated with marked induction of both p21cip1 mRNA and protein. This induction was exaggerated in the derivatives that overexpressed either PKC alpha or PKC beta 1. Therefore, in mouse mammary epithelial cells activation of the endogenous PKC alpha can transiently arrest cells in G0/G1 which may be due, at least in part, to induction of the transcription of p21cip1.


Subject(s)
Breast/drug effects , Cell Division/drug effects , Cyclins/biosynthesis , Isoenzymes/metabolism , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Animals , Breast/cytology , Breast/enzymology , Breast/metabolism , Cell Cycle , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Humans , Mice , Protein Kinase C-alpha , Thymidine/metabolism
11.
Biochem Pharmacol ; 58(7): 1097-107, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10484067

ABSTRACT

We examined the activity of two metabolites of sulindac (a nonsteroidal anti-inflammatory drug), sulindac sulfide and sulindac sulfone (exisulind, Prevatec), and a novel highly potent analog of exisulind (CP248) on a series of human prostate epithelial cell lines. Marked growth inhibition was seen with the BPH-1, LNCaP, and PC3 cell lines with IC50 values of about 66 microM, 137 microM, and 64 nM for sulindac sulfide, exisulind, and CP248, respectively. DNA flow cytometry and 4',6'-diamido-2-phenylindole (DAPI) staining indicated that these three compounds also induced apoptosis in all of these cell lines. Similar growth inhibition also was seen with the PrEC normal human prostate epithelial cell line, but these cells were resistant to induction of apoptosis at concentrations up to 300 microM, 1 mM, and 750 nM of sulindac sulfide, exisulind, and CP248, respectively. Derivatives of LNCaP cells that stably overexpress bcl-2 remained sensitive to growth inhibition and induction of apoptosis by these compounds. In vitro enzyme assays indicated that despite its high potency in inhibiting growth and inducing apoptosis, CP248, like exisulind, lacked cyclooxygenase (COX-1 and COX-2) inhibitory activity even at concentrations up to 10 mM. Moreover, despite variations of COX-1 and COX-2 expression, the three benign and malignant prostate cell lines showed similar sensitivity to growth inhibition and induction of apoptosis by these three compounds. Therefore, sulindac derivatives can cause growth inhibition and induce apoptosis in human prostate cancer cells by a COX-1 and -2 independent mechanism, and this occurs irrespective of androgen sensitivity or increased expression of bcl-2. These compounds may be useful in the prevention and treatment of human prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Prostatic Neoplasms/drug therapy , Sulindac/pharmacology , Androgens/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Division/drug effects , Cyclooxygenase Inhibitors/pharmacology , Drug Screening Assays, Antitumor , Humans , Male , Prostaglandin-Endoperoxide Synthases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulindac/analogs & derivatives , Tumor Cells, Cultured
12.
Apoptosis ; 4(3): 213-9, 1999 Jun.
Article in English | MEDLINE | ID: mdl-14634283

ABSTRACT

It is now apparent that apoptosis is closely linked to the control of cell cycle progression. During the G1 to S progression, cyclin D1, p53, and the cyclin dependent kinase inhibitors p21WAF1 and p27kip1 can play roles in induction of apoptosis. During the G2 and M phases, premature activation of Cdk1 can cause cells to enter mitotic catastrophe, which results in apoptosis. In this review we focus on factors acting during G1 and S, particularly cyclin D1, and their effects on cell growth, senescence and apoptosis. We emphasize that cyclin D1 can have diverse effects on cells depending on its level of expression, the specific cell type, the cell context and other factors. Possible mechanisms by which cyclin D1 exerts these diverse effects, via cyclin dependent kinase-dependent and -independent pathways, are discussed.

14.
Prostate ; 35(2): 95-101, 1998 May.
Article in English | MEDLINE | ID: mdl-9568672

ABSTRACT

BACKGROUND: The cyclin D1 gene is amplified and/or overexpressed in several types of human cancer, including cancers of the breast, esophagus, head, and neck. However, the role of cyclin D1 in prostate cancer has not been previously studied in detail. METHODS: Six human prostate cancer cell lines and cultures of normal human prostate cells were examined by Western and Northern blot analyses for levels of expression of the cyclin D1 protein and mRNA, respectively. Southern blot analyses were performed to examine possible amplification of this gene. Immunostaining for cyclin D1 was performed on 50 primary prostate cancer samples. RESULTS: Cyclin D1 protein was expressed at relatively high levels in all of the six human prostate cancer cell lines examined, but was not detected in the cultures of normal human prostate cells. The ALVA 41 cell line expressed the highest level of this protein. Relatively high levels of cyclin D1 mRNA were also found in all of the prostate cancer cell lines. Nevertheless, none of these cell lines revealed amplification of the cyclin D1 gene. Twelve of the 50 primary prostate cancer samples (24%) revealed regions of moderate to strongly positive staining for cyclin D1. CONCLUSIONS: The increased expression of cyclin D1 in several prostate cancer cell lines and in a subset of primary prostate cancer samples suggests that further studies on the expression of this gene and related genes may be of interest in understanding the pathogenesis of prostate cancer.


Subject(s)
Cyclin D1/biosynthesis , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Blotting, Northern , Blotting, Southern , Blotting, Western , Cyclin D1/genetics , Humans , Immunohistochemistry , Male , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Tumor Cells, Cultured
15.
J Cell Physiol ; 175(3): 314-22, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9572476

ABSTRACT

In previous studies, our laboratory demonstrated that Rat 6 (R6) fibroblasts which stably overproduce high levels of PKCepsilon display abnormalities in growth control that are characteristic of malignant transformation (Cacace et al., 1993, Oncogene, 8:2095-2104). The R6-PKCepsilon overproducing cell lines also exhibited a decreased growth factor requirement. The present study demonstrates that conditioned medium (CM) from two individual clones, R6-PKCepsilon 10 and 30, stimulates DNA synthesis in control R6-C1 cells. Maximal DNA synthesis and morphologic transformation was achieved in control cells when they were treated with medium from R6-PKCepsilon cells grown in the presence of TPA (TPA-CM). Size fractionation of the TPA-CM from PKCepsilon 30 cells revealed that this activity is due to a factor(s) that has an apparent molecular weight in the range of 10-30 kD and is heat and acid stable. This factor, like TGFbeta1, stimulated anchorage-independent growth of NRK cells. Western blot analysis (under nonreducing conditions) of the TPA-CM from R6-PKCepsilon 30 and R6-PKCepsilon 10 cells revealed the presence of the 25 kD active forms of TGFbeta2 and 3. These active forms of TGFbeta were not found in the CM of control R6 cells, or R6 cells that overexpress PKCalpha or PKCbeta1. The addition of a pan-specific TGFbeta antibody to NRK cells treated with the 10-30 kD fraction of TPA-CM from PKCepsilon 30 cells blocked the ability of this material to stimulate thymidine incorporation. Taken together, these studies suggest that the oncogenic activity of PKCepsilon in R6 cells is due, at least in part, to its ability to induce production of the active forms of TGFbeta2 and 3.


Subject(s)
Cell Transformation, Neoplastic , Fibroblasts/metabolism , Isoenzymes/physiology , Protein Kinase C/physiology , Transforming Growth Factor beta/physiology , Animals , Cell Adhesion , Cell Division , Cell Line , Culture Media, Conditioned , DNA/biosynthesis , Fibroblasts/enzymology , Isoenzymes/genetics , Mitogens/pharmacology , Molecular Weight , Protein Kinase C/genetics , Protein Kinase C-epsilon , RNA, Messenger/analysis , Rats , Tetradecanoylphorbol Acetate/pharmacology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/genetics
16.
Breast Cancer Res Treat ; 48(3): 195-203, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9598866

ABSTRACT

Nonsteriodal anti-inflammatory drugs (NSAIDs) are among the most commonly used medications in the United States and elsewhere, mainly for the treatment of arthritis. The NSAID sulindac causes regression and prevents the recurrence of premalignant colonic polyps in patients with familial adenomatous polyposis and inhibits colon carcinogenesis in rodents. Sulindac and sulindac sulfone, a metabolite of sulindac that lacks cyclooxygenase (cox) inhibitory activity, also inhibit mammary carcinogenesis in rats. To obtain insights into the relevance of these findings to human breast cancer, we examined the mechanism of action of sulindac and its sulfide and sulfone metabolites on the normal human mammary epithelial cell line MCF-10F and the human breast cancer cell line MCF-7. Of the three compounds, the sulfide was the most potent inhibitor of cell growth, although the sulfone and sulfoxide were also active at higher concentrations. Treatment of MCF-10F and MCF-7 cells with 100 microM sulindac sulfide resulted in accumulation of cells in the G1 phase of the cell cycle and induction of apoptosis. Apoptosis occurred within 24 h as determined by the TUNEL assay and DNA laddering was observed at 72 h. The accumulation of cells in G1 was associated with decreased levels of expression of cyclin D1 but no effect was seen on the expression of CDK4 or the immediate early response gene c-jun. Treatment with sulindac sulfide caused a striking induction of the CDK inhibitor p21WAF1 in MCF-10F cells. The MCF-7 cell line expressed a high basal level of p21WAF1 which did not change significantly after drug treatment. The pro-apoptotic gene BAX was not induced in either MCF-10F or MCF-7 cells by sulindac sulfide. Stable overexpression of cyclin D1, which frequently occurs in breast cancers, did not protect mammary epithelial cells from inhibition by the sulfide. These studies suggest that this class of compounds warrants further study with respect to breast cancer prevention and treatment.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast/drug effects , Sulindac/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast/cytology , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Line , Cyclin D1/analysis , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/analysis , Epithelial Cells/drug effects , Female , Humans , Sulindac/analogs & derivatives , Sulindac/therapeutic use
17.
Gastroenterology ; 113(6): 1892-900, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9394728

ABSTRACT

BACKGROUND & AIMS: Mutations of c-K-ras occur commonly in colonic neoplasms. The aim of this study was to determine how c-K-ras mutations alter the responses to the chemopreventive agent sulindac. METHODS: The parental rat intestinal cell line IEC-18 and c-K-ras-transformed derivatives were treated with sulindac sulfide. Cell cycle distribution was determined by flow-cytometric analysis (fluorescence-activated cell sorter), apoptosis by DNA fragmentation (laddering), flow cytometry, and microscopy, and changes in gene expression by immunoblotting. RESULTS: Sulindac sulfide inhibited cell growth and induced apoptosis in a time- and dose-dependent manner more rapidly in and at lower concentrations in parental cells than ras-transformed cells. Expression of the sulindac sulfide arrested cells in G0/G1, but cells entered apoptosis throughout the cell cycle. Proapoptotic protein Bak was relatively high in untreated parental cells and increased markedly after sulindac sulfide but was low in untreated ras-transformed cells and did not increase after sulindac sulfide. Expression of other Bcl-2 family members was unchanged after sulindac sulfide. However, sulindac sulfide reduced levels of cyclin D1 protein and cyclin E- and cyclin D1-associated kinase activity. CONCLUSIONS: c-K-ras-transformed enterocytes are relatively resistant to sulindac sulfide-induced growth inhibition and apoptosis, which may result from specific reduction of bak expression.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/physiology , Genes, ras/physiology , Intestines/drug effects , Intestines/physiology , Sulindac/pharmacology , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Transformed , Drug Resistance/physiology , Gene Expression Regulation/drug effects , Intestines/cytology , Rats
18.
Exp Cell Res ; 236(1): 173-80, 1997 Oct 10.
Article in English | MEDLINE | ID: mdl-9344597

ABSTRACT

In previous studies (W. Jiang, S. M. Kahn, P. Zhou, Y. (J. Zhang, A. M. Cacace, A. S. Infance, Y. Doi, R. M. Santella, and I. B. Weinstein 1993, Oncogene 8, 3447-3457) we reported that stable overexpression of cyclin D1 in R6 rat embryo fibroblasts shortens the G1 phase and impairs growth control. In the present study we examined the effects of cyclin D1 overexpression on other events involved in the G1 to S progression, utilizing the overexpressor cell line R6-ccnD1. We found that when compared to R6 control cells, serum-starved quiescent R6-ccnD1 cells had not only increased levels of the cyclin D1 protein but also increased levels of the cyclin E protein. The latter protein was complexed to phosphorylated cyclin-dependent kinase 2 (CDK2). However, in quiescent serum-starved R6-ccnD1 cells this cyclin E-CKD2 complex lacked in vitro kinase activity due to the presence of a heat-stable inhibitory activity, apparently reflecting the inhibitory effects of the CDK inhibitors (CDKIs) p21WAF1 and p27KIP1. Serum stimulation of the quiescent R6-ccnD1 cells was associated with a loss of this inhibitory activity and a decrease in the levels of the latter two proteins, as the cells progressed through the G1 phase. On the other hand, serum stimulation of the control R6 cells was associated with both induction of cyclin E and increased levels of phosphorylated CDK2 proteins and decreased levels of p21WAF1 and p27KIP1, as the cells progressed through the G1 phase. Thus, even though overexpression of cyclin D1 can induce the expression of cyclin E and phosphorylated CDK2, premature activation of cyclin E-CDK2 kinase activity in quiescent cells or during progression through G1 appears to be blocked by CDKIs. Nevertheless, the R6ccnD1 cells have a shorter G1 phase than the control cells presumably due to the high levels of both cyclin D1 and cyclin E. Taken together, these results indicate that overexpression of cyclin D, which is frequently seen in human tumors, can have complex effects on the expression of other genes that control cell cycle progression.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , Cyclin D1/genetics , G1 Phase/physiology , Tumor Suppressor Proteins , Animals , Cell Division/physiology , Cells, Cultured , Cyclin D1/metabolism , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/genetics , Cyclins/metabolism , Enzyme Inhibitors/metabolism , Fetus/cytology , Fibroblasts/chemistry , Fibroblasts/cytology , Fibroblasts/enzymology , G2 Phase/physiology , Gene Expression/physiology , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitosis/physiology , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Retinoblastoma Protein/metabolism , S Phase/physiology
19.
Carcinogenesis ; 18(6): 1139-48, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9214595

ABSTRACT

In the present study we have characterized eight human esophageal squamous carcinoma cell lines for levels of expression of cyclins D1, E, A and B1; CDKs 1, 2 and 4; the CDK inhibitors p16INK4, p21WAF1 and p27KIP1; the retinoblastoma (Rb) protein; and in vitro CDK2- and CDK4-associated kinase activity; and also compared the growth properties of these cell lines. The level of the cyclin D1 protein varied by over 30-fold amongst the eight cell lines. The high level in two cell lines was associated with amplification of this gene, but in three cell lines it was due to post-transcriptional events. Amongst the eight cell lines there was a significant correlation between the levels of cyclin D1, Rb and p27KIP1 proteins, and CDK4-associated kinase activity. Furthermore, when an exogenous cyclin D1 cDNA was over-expressed in the EC109 cell line by transfection, this led to increased expression of both Rb and p27KIP1. There was, however, no correlation between the level of cyclin D1 expression and the cell doubling times, duration of the G1 phase, or colony-forming efficiency in agar. Two of the cell lines displayed a high level of the cyclin E protein, low levels of cyclin D1, lacked expression of the Rb protein and expressed high levels of the p16INK4 protein. One of these cell lines displayed amplification of the latter gene. There was no correlation between the levels of cyclins E or A and in vitro CDK2 kinase activity, but CDK2 kinase activity was inversely correlated with the duration of the G1 phase of the cell cycle. Taken together, these studies indicate marked heterogeneity in the expression of cell cycle-related proteins amongst a series of esophageal carcinoma cell lines. The correlation between the levels of the cyclin D1, Rb and p27Kip1 proteins suggest the existence of a homeostatic feedback loop between positive and negative acting components of the cell cycle machinery.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Cycle Proteins , Cyclins/biosynthesis , Esophageal Neoplasms/metabolism , Microtubule-Associated Proteins/biosynthesis , Oncogene Proteins/biosynthesis , Tumor Suppressor Proteins , Blotting, Northern , Blotting, Southern , Carcinoma, Squamous Cell/pathology , Carrier Proteins/biosynthesis , Carrier Proteins/metabolism , Cell Cycle/physiology , Cell Division/physiology , Cyclin D1 , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/biosynthesis , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Esophageal Neoplasms/pathology , Humans , Oncogene Proteins/metabolism , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured
20.
Cancer Res ; 57(8): 1569-74, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9108461

ABSTRACT

Cyclin D1 plays an important role in regulating the progression of cells through the G1 phase of the cell cycle. This gene is frequently overexpressed in human colon cancer. To address the role of cyclin D1 in growth control and tumorigenesis in this disease, we have overexpressed an antisense cyclin D1 cDNA construct in the human colon cancer cell line SW480E8, which expresses high levels of cyclin D1. The integration and expression of the antisense construct was verified by Southern and Northern blot analyses, respectively, and resulted in decreased expression of the cyclin D1 protein. This was associated with decreased levels of the Rb and p27Kip1 proteins. In addition, the hypophosphorylated form of Rb was increased in these cells. The SW480E8 antisense cyclin D1 cells displayed an increased doubling time, a decrease in saturation density, decreased plating efficiency and anchorage-independent growth, and a loss of tumorigenicity in nude mice. These findings provide direct evidence that increased expression of cyclin D1 in colon tumor cells contributes to their abnormal growth and tumorigenicity. The ability to revert the transformed phenotype of these cells with antisense cyclin D1 suggests that cyclin D1 or its associated cyclin-dependent kinase 4 may be useful targets in the therapy of colon cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Cycle Proteins , Colonic Neoplasms/metabolism , Cyclins/metabolism , DNA, Antisense/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Animals , Cell Cycle , Cell Division/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cyclin D1 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/physiology , Down-Regulation , Genetic Vectors , Humans , Mice , Mice, Nude , Microtubule-Associated Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/physiology , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...