Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 423
Filter
1.
Curr Cancer Drug Targets ; 10(1): 117-26, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20088788

ABSTRACT

Despite recent advancements in treatment modalities, the overall survival and quality of life of patients with head and neck squamous cell carcinoma (HNSCC) have not improved significantly over the past decade. With the increasing emergency of new biological agents, the development of novel treatment schemes based on cancer cell biology may be promising for this group of patients. We previously introduced the "oncogene addiction" concept as a rationale for molecular targeting in cancer therapy and prevention. In this context, an increasing number of preclinical studies have demonstrated that the Signal Transducers and Activators of transcription 3 (Stat3) transcription factor plays critical roles in the development and progression of a variety of tumors including HNSCC, by regulating cell proliferation, cell cycle progression, apoptosis, angiogenesis, immune evasion, Epithelial-Mesenchymal Transition (EMT) and through effects in cancer stem cells. The purpose of this review is to summarize current experimental and clinical evidence that suggest that HNSCC might be addicted to Stat3 and describe the molecular mechanisms that may explain this phenomenon. In addition, we discuss whether this addiction is an exploitable target for developing approaches for the treatment and prevention of HNSCC.


Subject(s)
Head and Neck Neoplasms/metabolism , Neoplasms, Squamous Cell/metabolism , STAT3 Transcription Factor/metabolism , Animals , Apoptosis , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Drug Resistance, Neoplasm , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Humans , Mice , Neoplasms, Squamous Cell/drug therapy , Neoplasms, Squamous Cell/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT3 Transcription Factor/agonists , STAT3 Transcription Factor/antagonists & inhibitors , Tumor Escape
2.
Cancer Epidemiol Biomarkers Prev ; 16(12): 2768-72, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18086786

ABSTRACT

BACKGROUND: Phosphoactivation of a DNA damage response molecule checkpoint kinase 2 (pChk2) may be a marker of oral epithelial cells that have entered the precancerous and squamous cell carcinoma (SCC) stages. We explored whether there was selective expression of pChk2 in precancerous lesions but not in nonneoplastic tissue of the oral mucosa. EXPERIMENTAL DESIGN: In a retrospective cohort design, 96 biopsied clinical leukoplakias and erythroplakias with known subsequent progression to SCC were identified from 48 subjects and assigned as the cases group. Expression status of pChk2 was compared with that of the 97 leukoplakias and erythroplakias that did not progress to SCC (control groups) by immunohistochemical analysis. Included in both groups were lesions with histologically confirmed dysplasia and those that lacked histologic evidence of atypia. RESULTS: Subjects with pChk2-positive but histology-negative (for atypia) lesions had an 8.6 times higher risk of developing SCC compared with those with pChk2-negative and histology-negative lesions. Overall, the presence of detectable pChk2 staining was able to identify lesions at risk of developing SCC within 3 years with a sensitivity of 85.2%, specificity of 74.2%, and predictive accuracy of 78.2% (odds ratio, 19.9; 95% confidence interval, 7.3-55.5). CONCLUSION: This is the first study to include histologically nonatypical cases in the analysis of a putative biomarker for oral precancerous lesions. Our data show that pChk2 merits further investigation as a promising biomarker that can discriminate those lesions at risk for developing SCC, regardless of histologic evidence for atypia.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Precancerous Conditions/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Biomarkers, Tumor/analysis , Checkpoint Kinase 2 , Cohort Studies , Female , Humans , Immunohistochemistry , Male , Precancerous Conditions/pathology , Retrospective Studies
3.
Oncogene ; 25(5): 713-21, 2006 Feb 02.
Article in English | MEDLINE | ID: mdl-16186798

ABSTRACT

The HINT1 protein, a member of the histidine triad (HIT) family, is highly conserved in diverse species and ubiquitously expressed in mammalian tissues. However, its precise function in mammalian cells is not known. As a result of its structural similarity to the tumor-suppressor protein FHIT, we used homozygous-deleted Hint1 mice to study its role in tumorigenesis. We discovered that after 2 to 3 years of age the spontaneous tumor incidence in Hint1 -/- mice was significantly greater than that in wild-type Hint1 +/+ mice (P < 0.05). Using a well-established mouse model of 7,12-dimethylbenz[a]anthracene (DMBA)-induced mammary carcinogenesis we found a marked and significant (P < 0.05) increase in the incidence of mammary and ovarian tumors in both, Hint1 -/- and +/- mice versus +/+ mice. The Hint1 -/- and +/- mice had similar tumor incidence and similar tumor histologies. Therefore, deletion of Hint1 in mice enhances both spontaneous tumor development and susceptibility to tumor induction by DMBA. In addition, since the Hint1 +/- tumors retained expression of the unmutated wild-type allele, Hint1 is haplo-insufficient with respect to tumor suppression in this model system.


Subject(s)
Genes, Tumor Suppressor , Haplotypes , Nerve Tissue Proteins/physiology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Base Sequence , DNA Primers , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/genetics , Mice , Nerve Tissue Proteins/genetics , Ovarian Neoplasms/chemically induced , Ovarian Neoplasms/genetics
4.
Clin Cancer Res ; 7(12): 4220-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751523

ABSTRACT

The antitumor effects of the green tea compound epigallocatechin-3-gallate (EGCG) have not been studied in detail previously in head and neck squamous cell carcinoma (HNSCC) cells. Overexpression of the epidermal growth factor receptor (EGFR) occurs frequently in HNSCC, which is an adverse prognostic factor. Therefore, we examined in detail the molecular effects of EGCG on two human HNSCC cell lines, YCU-N861 and YCU-H891, focusing on the EGFR signaling pathway. The 70% lethal dose (IC(70)) of EGCG for both cell lines was 10 microg/ml. Treatment with EGCG increased the proportion of cells in the G(1) phase of the cell cycle and induced apoptosis. In cells treated with EGCG, there was a decrease in the cyclin D1 protein, an increase in the p21(Cip1) and p27(Kip1) proteins, and a reduction in the hyperphosphorylated form of pRB, changes that may account for the arrest in G(1). EGCG also caused a decrease in the Bcl-2 and Bcl-X(L) proteins, an increase in the Bax protein, and activation of caspase 9, suggesting that EGCG induces apoptosis via a mitochondrial pathway. Treatment with EGCG inhibited phosphorylation of the EGFR, signal transducer and activator of transcription3 (Stat3), and extracellular regulated kinase (ERK) proteins and also inhibited basal and transforming growth factor-alpha-stimulated c-fos and cyclin D1 promoter activity. EGCG at 0.1 microg/ml (a concentration found in serum after oral administration) markedly enhanced the growth-inhibitory effects of 5-fluorouracil. Taken together, these findings provide insights into molecular mechanisms of growth inhibition by EGCG and suggest that this naturally occurring compound may be useful, when used alone or in combination with other agents, in the chemoprevention and/or treatment of HNSCC.


Subject(s)
Antineoplastic Agents/toxicity , Catechin/pharmacology , ErbB Receptors/physiology , Gene Expression Regulation, Neoplastic/drug effects , Signal Transduction/drug effects , Catechin/analogs & derivatives , Cell Division/drug effects , ErbB Receptors/drug effects , Humans
5.
Anticancer Res ; 21(4A): 2297-303, 2001.
Article in English | MEDLINE | ID: mdl-11724286

ABSTRACT

The nonsteroidal anti-inflammatory drug (NSAID) sulindac and its derivatives induce apoptosis in a variety of carcinoma cells in vitro and display antitumor effects in vivo. The effects of these agents have not, however, been studied in detail in leukemia cells. In the present study we compared the effects of sulindac sulfide to those of 12-0-tetradecanoylphorbol ester (TPA) on the human promyelocytic leukemia cell line HL60. The latter compound is known to induce monocytic differentiation in these cells. We found that both sulindac sulfide and TPA caused growth inhibition, cell cycle arrest in G0/G1 and increased levels of the cell cycle inhibitory proteins p21Cip1 and p27KiP1. However, whereas the TPA treated cells underwent subsequent differentiation the sulindac sulfide-treated cells displayed extensive apoptosis and negligible differentiation. Ectopic overexpression of p21Cip1 or p27KiP1 markedly enhanced the apoptosis induced by sulindac sulfide. Therefore, sulindac sulfide and related compounds may be useful in the treatment of leukemia and other neoplasms, especially when used together with agents that increase cellular levels of p21Cip1 or p27KiP1.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/physiology , Cyclins/physiology , Sulindac/analogs & derivatives , Sulindac/pharmacology , Tumor Suppressor Proteins/physiology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/physiology , Cell Cycle/drug effects , Cell Cycle Proteins/biosynthesis , Cell Differentiation/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/biosynthesis , HL-60 Cells/drug effects , HL-60 Cells/pathology , Humans , Tetradecanoylphorbol Acetate/pharmacology , Tumor Suppressor Proteins/biosynthesis
6.
Front Biosci ; 6: E104-18, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11578975

ABSTRACT

Chronic infection of the gastric mucosa by the bacterium H. pylori results in an intense inflammatory response which can last for decades. An associated host response is a chronic hyperproliferative state, in which there is increased cell turnover and also increased apoptosis of the gastric epithelial cells. Recent studies have also demonstrated abnormalities in the expression of cell cycle control proteins. This review describes these events, emphasizing recent studies on the effects of H. pylori infection on cell cycle progression and the expression of cell cycle regulatory proteins. The systems that have been studied include in vivo studies in humans and in experimental animals, and in vitro studies in which gastric epithelial cells were co-cultivated with H. pylori. The earliest event following H. pylori's interaction with epithelial cells appears to be growth inhibition and apoptosis. The hyperproliferative response observed in the gastric mucosa is secondary to this initial insult and is associated with increased expression of cyclin D1, the cyclin dependent kinase inhibitor p16ink4a and of p53 and decreased expression of the cyclin dependent kinase inhibitor p27kip1. Dysregulation of the hyperproliferative response may, ultimately, be responsible for the ability of H. pylori to enhance the development of gastric cancer.


Subject(s)
Cell Cycle/physiology , Gastric Mucosa/physiopathology , Helicobacter Infections/physiopathology , Helicobacter pylori , Animals , Cell Cycle Proteins/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Humans
8.
Anticancer Res ; 21(3B): 1665-71, 2001.
Article in English | MEDLINE | ID: mdl-11497245

ABSTRACT

Propolis has numerous biologic activities including antibiotic, antifungal, antiviral and anti-inflammatory properties. Several components isolated from propolis have been shown to have anticancer activity. This study demonstrates that the compound PM-3 (3-[2-dimethyl-8-(3-methyl-2-butenyl)benzopyran]-6-propenoic acid) isolated from Brazilian propolis markedly inhibits the growth of MCF-7 human breast cancer cells. This effect was associated with inhibition of cell cycle progression and induction of apoptosis. Treatment of MCF-7 cells with PM-3 arrested cells in the G1 phase and resulted in a decrease in the protein levels of cyclin D1 and cyclin E. PM-3 also inhibited the expression of cyclin D1 at the transcriptional level when examined in cyclin D1 promoter luciferase assays. Induction of apoptosis by PM-3 occurred within 48 hours after treatment of MCF-7 cells. The MCF-7 treated cells also displayed a decrease in the level of the estrogen receptor (ER) protein and inhibition of estrogen response element (ERE) promoter activity. Therefore, PM-3 merits further investigation with respect to breast cancer chemoprevention or therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Breast Neoplasms/drug therapy , Methacrylates/pharmacology , Plant Extracts/pharmacology , Propolis/metabolism , Annexin A5/metabolism , Apoptosis , Benzopyrans/chemistry , Blotting, Northern , Blotting, Western , Cell Cycle/drug effects , Cell Division , Coloring Agents/pharmacology , Cyclin D1/biosynthesis , Cyclin E/biosynthesis , Dose-Response Relationship, Drug , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Genes, Reporter , Humans , Methacrylates/chemistry , Models, Chemical , Promoter Regions, Genetic , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/drug effects , Response Elements , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Transcription, Genetic , Transfection , Tumor Cells, Cultured
9.
J Urol ; 166(3): 882-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11490238

ABSTRACT

PURPOSE: We evaluated the safety and efficacy of exisulind for delaying disease progression in men with increasing prostate specific antigen (PSA) after radical prostatectomy. MATERIALS AND METHODS: A total of 96 men with increasing PSA after radical prostatectomy were randomized to receive placebo (49) or 250 mg. exisulind twice daily (47) for 12 months. The primary efficacy parameter was the difference in change from baseline PSA between the placebo and exisulind groups. The PSA doubling time was also evaluated before and during study. A subgroup analysis classified patients based on the risk of developing metastatic disease. RESULTS: Compared with placebo, exisulind significantly suppressed the increase in PSA in all patients (p = 0.017). The results were also statistically significant in men at high risk for metastasis (p = 0.0003) and those who could not be classified according to risk (p = 0.0009). In addition, median PSA doubling time was lengthened in high risk patients on exisulind (2.12 month increase) compared with those on placebo (3.37 month decrease, p = 0.048). Exisulind was well tolerated. CONCLUSIONS: Exisulind inhibited the increase in PSA overall and prolonged PSA doubling time in high risk patients compared with placebo. These results suggest that Exisulind has the potential to extend the time from biochemical recurrence to the need for androgen deprivation therapy. Exisulind was well tolerated in this patient population. Our results support further study of Exisulind in the treatment of patients with prostate cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Prostatectomy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/surgery , Sulindac/therapeutic use , Aged , Aged, 80 and over , Combined Modality Therapy , Double-Blind Method , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/blood , Postoperative Complications/epidemiology , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Sulindac/analogs & derivatives
10.
Cancer Res ; 61(7): 3188-93, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306507

ABSTRACT

CDC25B phosphatase plays a key role in controlling G2-M progression by dephosphorylating two inhibitory residues of CDC2 and also has been suggested to have an oncogenic property. In this study, we investigated the effect of CDC25B overexpression on radiation-induced G2-M arrest and radiation sensitivity in esophageal cancer cells. TE8-CDC25B, in which CDC25B was overexpressed under an inducible system, was more radiosensitive than the vector control (TE8-neo) in a clonogenic survival assay. Without radiation, CDC25B overexpression had little effect on cell cycle fractions or growth rate. After 10-Gy radiation, TE8-CDC25B showed decreased G2-M arrest and increased apoptosis, whereas TE8-neo displayed prolonged G2-M arrest and less apoptosis. During this period, there were no differences in the protein amounts of CDC2 and cyclin B1 between the two cell lines. However, more CDC25B expression, which was reduced immediately by radiation, was sustained in TE8-CDC25B than in TE8-neo. Moreover, induction of tyrosine phosphorylation of CDC2 and reduction of CDC2 kinase activity after irradiation was less significant in TE8-CDC25B than in TE8-neo. These results indicate that cancer cells that overexpress CDC25B override G2-M arrest by retaining CDC2 kinase activity and undergo apoptosis after radiation. This may point to an effective approach toward improving radiotherapy outcomes of various cancers.


Subject(s)
Apoptosis/radiation effects , Cell Cycle Proteins/physiology , Esophageal Neoplasms/pathology , G2 Phase/radiation effects , Mitosis/radiation effects , cdc25 Phosphatases/physiology , Apoptosis/physiology , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Survival/radiation effects , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/radiotherapy , G2 Phase/physiology , Humans , Mitosis/physiology , Radiation Tolerance/physiology , cdc25 Phosphatases/biosynthesis , cdc25 Phosphatases/genetics
11.
J Biol Chem ; 276(19): 16406-10, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11278263

ABSTRACT

We recently obtained evidence that treatment of human colon cancer cells with exisulind (sulindac sulfone) and related compounds induces apoptosis by activation of protein kinase G (PKG) and c-Jun kinase (JNK1). The present study further explores this mechanism. We demonstrate that in NIH3T3 cells a constitutively active mutant of PKG causes a dose-dependent activation of JNK1 and thereby transactivates c-Jun and stimulates transcription from the AP-1 enhancer element. The activation of JNK1 and the transactivation of c-Jun by this mutant of PKG were inhibited by a dominant negative MEKK1. In vitro assays showed that a purified PKG directly phosphorylated the N-terminal domain of MEKK1. PKG also directly phosphorylated a full-length MEKK1, and this was associated with enhanced MEKK1 phosphorylation. Thus, it appears that PKG activates JNK1 through a novel PKG-MEKK1-SEK1-JNK1 pathway, by directly phosphorylating and activating MEKK1.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Genes, jun , MAP Kinase Kinase 4 , MAP Kinase Kinase Kinase 1 , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , 3T3 Cells , Animals , Enzyme Activation , Gene Expression Regulation , Mice , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Transcriptional Activation , Transfection
12.
Cancer Res ; 61(2): 725-31, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11212275

ABSTRACT

Epidemiological studies link increased garlic (Allium sativum) consumption with a reduced incidence of colon cancer in various human populations. Experimental carcinogenesis studies in animal models and in cell culture systems indicate that several allium-derived compounds exhibit inhibitory effects and that the underlying mechanisms may involve both the initiation and promotion phases of carcinogenesis. To provide a better understanding of the effects of allium derivatives on the prevention of colon cancer, we examined two water-soluble derivatives of garlic, S-allylcysteine (SAC) and S-allylmercaptocysteine (SAMC), for their effects on proliferation and cell cycle progression in two human colon cancer cell lines, SW-480 and HT-29. For comparison, we included the compound sulindac sulfide (SS), because sulindac compounds are well-established colon cancer chemopreventive agents. We found that SAMC, but not SAC, inhibited the growth of both cell lines at doses similar to that of SS. SAMC also induced apoptosis, and this was associated with an increase in caspase3-like activity. These affects of SAMC were accompanied by induction of jun kinase activity and a marked increase in endogenous levels of reduced glutathione. Although SS caused inhibition of cell cycle progression from G1 to S, SAMC inhibited progression at G2-M, and a fraction of the SW-480 and HT-29 cells were specifically arrested in mitosis. Coadministration of SS with SAMC enhanced the growth inhibitory and apoptotic effects of SS. These findings suggest that SAMC may be useful in colon cancer prevention when used alone or in combination with SS or other chemopreventive agents.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cysteine/analogs & derivatives , Cysteine/pharmacology , Sulindac/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspase 3 , Caspases/drug effects , Caspases/metabolism , Cell Cycle/drug effects , Cysteine/chemistry , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , G2 Phase/drug effects , Garlic/chemistry , Glutathione/drug effects , Glutathione/metabolism , HT29 Cells , Humans , In Situ Hybridization, Fluorescence , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases , Kinetics , Mitogen-Activated Protein Kinases/metabolism , Mitosis/drug effects , Plants, Medicinal , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulindac/analogs & derivatives , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
13.
Proc Natl Acad Sci U S A ; 98(2): 609-14, 2001 Jan 16.
Article in English | MEDLINE | ID: mdl-11149941

ABSTRACT

Pre-T cell receptor (preTCR)-derived signals mediate the transition of thymocytes from the CD4(-) CD8(-) double-negative (DN) to CD4(+) CD8(+) double-positive stage of T lymphocyte development. This progression, termed beta-selection, is limited to thymocytes that have generated a functional TCR-beta chain able to associate with pTalpha to form the preTCR complex. Formation of the preTCR complex not only induces differentiation, survival, and proliferation of DN thymocytes; it also inhibits further TCR-beta gene rearrangement through an ill-defined process known as allelic exclusion. The signaling pathways controlling this critical developmental checkpoint have not been characterized. Here we demonstrate that formation of the preTCR complex leads to the activation of protein kinase C (PKC), and that activation of PKC is necessary for the differentiation and expansion of DN thymocytes. Importantly, we also show that allelic exclusion at the TCR-beta gene loci is enforced by PKC-mediated signals. These results define PKC as a central mediator of both differentiation and allelic exclusion during thymocyte development.


Subject(s)
Alleles , Clonal Deletion/physiology , Hematopoiesis/physiology , Isoenzymes/physiology , Protein Kinase C/physiology , T-Lymphocyte Subsets/cytology , Thymus Gland/cytology , Adaptor Proteins, Signal Transducing , Animals , Biolistics , Cell Differentiation , DNA-Binding Proteins , Enzyme Activation , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Genes, Dominant , Genes, Reporter , Isoenzymes/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Mice , Mice, Knockout , Organ Culture Techniques , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoproteins/physiology , Protein Kinase C/genetics , Protein Kinase C-alpha , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-raf/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Recombinant Fusion Proteins/physiology , Signal Transduction , Transfection , Type C Phospholipases/physiology , ZAP-70 Protein-Tyrosine Kinase
14.
Anticancer Res ; 21(5): 3535-9, 2001.
Article in English | MEDLINE | ID: mdl-11848520

ABSTRACT

African-American women with breast cancer consistently show a shortened survival when compared with Caucasians with breast cancer, however it is not clear whether this is due to socioeconomic factors or to racial differences in tumor biology. Cyclin D1 overexpression has been demonstrated in 60-80% of female breast cancers, however these studies have not included race or ethnicity data. We examined the level of cyclin D1 protein expression in 139 cases of female breast cancer obtained from different ethnic populations. Using an immunoperoxidase-based technique and a polyclonal anti-cyclin D1 antibody, the rate of overexpression was 68%. Cyclin D1 overexpression tended to be more frequent in cases from non-Caucasian patients when compared with those from Caucasian patients (77% vs. 59%, p=0.051). Our findings suggest that non-Caucasian ethnicity may be important in predicting cyclin D1 overexpression. Cyclin D1 could therefore serve as a possible target in managing breast cancer in the African-American population.


Subject(s)
Black People , Breast Neoplasms/metabolism , Cyclin D1/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Nucleus/metabolism , Cyclin D1/genetics , Female , Humans , Middle Aged , Ploidies , Receptors, Estrogen/physiology
15.
Clin Cancer Res ; 6(10): 4136-41, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11051267

ABSTRACT

Sulindac sulfone (Exisulind) induces apoptosis and exhibits cancer chemopreventive activity, but in contrast to sulindac, it does not inhibit cyclooxygenases 1 or 2. We found that sulindac sulfone and two potent derivatives, CP248 and CP461, inhibited the cyclic GMP (cGMP) phosphodiesterases (PDE) 2 and 5 in human colon cells, and these compounds caused rapid and sustained activation of the c-Jun NH2-terminal kinase 1 (JNK1). Rapid activation of stress-activated protein/ERK kinase 1 (SEK1) and mitogen-activated protein kinase kinase kinase (MEKK1), which are upstream of JNK1, was also observed. Other compounds that increase cellular levels of cGMP also activated JNK1, and an inhibitor of protein kinase G (PKG), Rp-8-pCPT-cGMPS, inhibited JNK1 activation by the sulindac sulfone derivatives. Expression of a dominant-negative JNK1 protein inhibited CP248-induced cleavage of poly(ADP-ribose) polymerase, a marker of apoptosis. Thus, it appears that sulindac sulfone and related compounds induce apoptosis, at least in part, through activation of PKG, which then activates the MEKK1-SEK1-JNK1 cascade. These studies also indicate a role for cGMP and PKG in the JNK pathway.


Subject(s)
Apoptosis , Cyclic GMP/metabolism , MAP Kinase Kinase 4 , MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases/metabolism , Sulindac/pharmacology , Colonic Neoplasms/drug therapy , Dose-Response Relationship, Drug , Enzyme Activation , Genes, Dominant , Humans , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Sulindac/analogs & derivatives , Time Factors , Tumor Cells, Cultured
16.
Infect Immun ; 68(9): 5321-8, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10948161

ABSTRACT

Helicobacter pylori infection is associated with the development of gastric cancer. In short-term coculture with AGS gastric cells, H. pylori inhibits cell cycle progression and induces dose-dependent apoptosis. Based on the concept that an imbalance between proliferation and apoptosis may contribute to the emergence of gastric cancer, we chronically exposed AGS cells to H. pylori as a model of chronic exposure in humans. The AGS derivatives selected by this process were stably resistant not only to H. pylori-induced apoptosis but also to apoptosis induced by other enteric bacteria and by several toxic agents including radiation and cancer chemotherapy. Like the parental AGS cells, the derivatives underwent G(1)/S-phase cell cycle inhibition in response to H. pylori. The AGS derivatives displayed a marked decrease in cellular levels of the cell cycle control protein p27(kip1). We found a similar decrease in epithelial cell p27(kip1) expression in gastric biopsy specimens from H. pylori-infected patients. These findings are consistent with observations that link decreases in the p27(kip1) level to increased susceptibility to cancer in mice with p27(kip1) deleted and to a poor prognosis of gastric cancer in humans. This is the first demonstration that bacterial infection can lead to apoptosis resistance and to cross-resistance to other inducers of apoptosis such as bacteria, chemotherapeutic agents, and radiation. The development of apoptosis resistance and downmodulation of p27(kip1) may contribute to the increased risk for gastric cancer observed in humans chronically exposed to H. pylori.


Subject(s)
Apoptosis , Cell Cycle Proteins , Gastric Mucosa/pathology , Helicobacter Infections/pathology , Helicobacter pylori , Microtubule-Associated Proteins/analysis , Tumor Suppressor Proteins , Bacterial Adhesion , Cell Cycle , Cell Division , Cell Line , Chronic Disease , Cyclin-Dependent Kinase Inhibitor p27 , Gastric Mucosa/chemistry , Genes, p53 , Helicobacter Infections/complications , Helicobacter Infections/metabolism , Humans , Phenotype , Stomach Neoplasms/etiology
17.
Mol Cell Biol ; 20(15): 5392-403, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10891480

ABSTRACT

Although mitogenic and differentiating factors often activate a number of common signaling pathways, the mechanisms leading to their distinct cellular outcomes have not been elucidated. In a previous report, we demonstrated that mitogen-activated protein (MAP) kinase (ERK) activation by the neurogenic agents fibroblast growth factor (FGF) and nerve growth factor is dependent on protein kinase Cdelta (PKCdelta), whereas MAP kinase activation in response to the mitogen epidermal growth factor (EGF) is independent of PKCdelta in rat hippocampal (H19-7) and pheochromocytoma (PC12) cells. We now show that EGF activates MAP kinase through a PKCzeta-dependent pathway involving phosphatidylinositol 3-kinase and PDK1 in H19-7 cells. PKCzeta, like PKCdelta, acts upstream of MEK, and PKCzeta can potentiate Raf-1 activation by EGF. Inhibition of PKCzeta also blocks EGF-induced DNA synthesis as monitored by bromodeoxyuridine incorporation in H19-7 cells. Finally, in embryonic rat brain hippocampal cell cultures, inhibitors of PKCzeta or PKCdelta suppress MAP kinase activation by EGF or FGF, respectively, indicating that these factors activate distinct signaling pathways in primary as well as immortalized neural cells. Taken together, these results implicate different PKC isoforms as determinants of growth factor signaling specificity within the same cell. Furthermore, these data provide a mechanism whereby different growth factors can differentially activate a common signaling intermediate and thereby generate biological diversity.


Subject(s)
Epidermal Growth Factor/metabolism , Isoenzymes/metabolism , Neurons/metabolism , Protein Kinase C/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Cells, Cultured , Epidermal Growth Factor/pharmacology , Hippocampus/cytology , Hippocampus/embryology , Isoenzymes/genetics , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Oligonucleotides, Antisense/pharmacology , PC12 Cells/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/genetics , Protein Kinase C-delta , Protein Kinase C-theta , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Rats , Rats, Sprague-Dawley
18.
Exp Cell Res ; 257(1): 22-32, 2000 May 25.
Article in English | MEDLINE | ID: mdl-10854051

ABSTRACT

Using human cyclin D1 as the "bait" in a yeast two-hybrid system, together with a HL60 cDNA library, we identified a novel human nuclear protein designated DIP1. This protein is expressed in a variety of cell types, and in fibroblasts its level remains constant throughout the cell cycle. However, the level of this protein increases severalfold during the differentiation of HL60 cells. The DIP1 protein can be phosphorylated in vitro by a cellular kinase and this activity reaches its maximum in extracts obtained from cells in the G1 phase of the cell cycle. DIP1 contains a helix-loop-helix motif but lacks an adjacent basic DNA-binding domain, thus resembling the Id family of proteins. The dip1 gene is located on human chromosome 16p11.2-12, a locus that is amplified in several types of human cancer. These results suggest that DIP1 may be involved in the control of gene expression and differentiation, but its precise function remains to be determined.


Subject(s)
Drosophila Proteins , Nuclear Proteins/genetics , Repressor Proteins , Transcription Factors/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Cyclin D1/genetics , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , HL-60 Cells , Helix-Loop-Helix Motifs , Humans , Inhibitor of Differentiation Protein 1 , Molecular Sequence Data , Nuclear Proteins/analysis , Sequence Alignment , Sequence Analysis
19.
Carcinogenesis ; 21(5): 857-64, 2000 May.
Article in English | MEDLINE | ID: mdl-10783304

ABSTRACT

The multistage process of carcinogenesis involves the progressive acquisition of mutations, and epigenetic abnormalities in the expression, of multiple genes that have highly diverse functions. An important group of these genes are involved in cell cycle control. Thus, cyclin D1 is frequently overexpressed in a varety of human cancers. Cylin D1 plays a critical role in carcinogenesis because (i) overexpression enhances cell transformation and tumorigenesis, and enhances the amplification of other genes, and (ii) an antisense cyclin D1 cDNA reverts the malignant phenotype of carcinoma cells. Therefore, cyclin D1 may be a useful biomarker in molecular epidemiology studies, and inhibitors of its function may be useful in both cancer chemoprevention and therapy. We discovered a paradoxical increase in the cell cycle inhibitors protein p27(Kip1) in a subset of human cancers, and obtained evidence for homeostatic feedback loops between cyclins D1 or E and p27(Kip1). Furthermore, derivatives of HT29 colon cancer cells with increased levels of p27(Kip1) showed increased sensitivity to induction of differentiation. This may explain why decreased p27(Kip1) in a subset of human cancers is associated with a high grade (poorly differentiated) histology and poor prognosis. Agents that increase cellular levels of p27(Kip1) may, therefore, also be useful in cancer therapy. Using an antisense Rb oligonucleotide we obtained evidence that the paradoxical increase in pRb often seen in human colon cancers protects these cells from growth inhibition and apopotosis. On the basis of these, and other findings, we hypothesize that homeostatic feedback mechanisms play a critical role in multistage carcinogenesis. Furthermore, because of their bizarre circuitry, cancer cells suffer from 'gene addiction' and 'gene hypersensitivity' disorders that might be exploited in both cancer prevention and chemotherapy.


Subject(s)
Cell Cycle , Cell Transformation, Neoplastic , Homeostasis , Cell Cycle Proteins/physiology , Genes, Tumor Suppressor , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Retinoblastoma Protein/metabolism
20.
Mol Carcinog ; 27(3): 166-76, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10708478

ABSTRACT

The ability of the promyelocytic leukemia HL60 cell line to differentiate in response to various stimuli has provided a widely used model of differentiation. The phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA), acting via its cellular receptor protein kinase C(PKC), induces these cells to acquire a monocytic phenotype. We set out to identify the specific isoform of the multigene PKC family that is involved in this differentiation event. To do so, we utilized a highly specific PKCbeta inhibitor, LY379196. We found that LY379196 could prevent the growth arrest, cellular adherence, and changes in several marker proteins that occur after the addition of TPA to HL60 cells and that these effects were not simply due to nonspecific cytotoxicity. Thus, the present studies provide strong evidence that the beta isoform of PKC plays a critical role in TPA-induced HL60 monocytic differentiation.


Subject(s)
Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Monocytes/drug effects , Protein Kinase C/antagonists & inhibitors , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Blotting, Western , Cell Adhesion/drug effects , Flow Cytometry , HL-60 Cells , Humans , Macrophage-1 Antigen/metabolism , Monocytes/cytology , Protein Kinase C beta , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...