Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Lab Invest ; 96(2): 249-59, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26658453

ABSTRACT

Preclinical studies in ovarian cancer have demonstrated upregulation of the Wnt/ß-catenin pathway promoting tumor proliferation and chemoresistance. Our objective was to evaluate the effect of the Wnt/ß-catenin pathway inhibitor, WNT974, in primary ovarian cancer ascites cells. Ascites cells from patients with papillary serous ovarian cancer were isolated and treated with 1 µM WNT974±100 µM carboplatin. Viability was evaluated with the ATPlite assay. The IC50 was calculated using a dose-response analysis. Immunohistochemistry (IHC) was performed on ascites cells and tumor. Expression of R-spondin 2 (RSPO2), RSPO3, PORCN, WLS, AXIN2, and three previously characterized RSPO fusion transcripts were assessed using Taqman assays. Sixty ascites samples were analyzed for response to WNT974. The ascites samples that showed a decrease in ATP concentration after treatment demonstrated no difference from the untreated cells in percent viability with trypan blue staining. Flow cytometry demonstrated fewer cells in the G2 phase and more in the G1 and S phases after treatment with WNT974. Combination therapy with WNT974 and carboplatin resulted in a higher percentage of samples that showed ≥30% reduction in ATP concentration than either single drug treatment. IHC analysis of Wnt pathway proteins suggests cell cycle arrest rather than cytotoxicity after WNT974 treatment. QPCR indicated that RSPO fusions are not prevalent in ovarian cancer tissues or ascites. However, higher PORCN expression correlated to sensitivity to WNT974 (P=0.0073). In conclusion, WNT974 produces cytostatic effects in patient ascites cells with primary ovarian cancer through inhibition of the Wnt/ß-catenin pathway. The combination of WNT974 and carboplatin induces cytotoxicity plus cell cycle arrest in a higher percentage of ascites samples than with single drug treatment. RSPO fusions do not contribute to WNT974 sensitivity; however, higher PORCN expression indicates increased WNT974 sensitivity.


Subject(s)
Antineoplastic Agents/pharmacology , Ascites/metabolism , Ovarian Neoplasms/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Aged , Antineoplastic Agents/chemistry , Female , Humans , Immunohistochemistry , Middle Aged , Ovarian Neoplasms/chemistry , Ovary/chemistry , Wnt Proteins/metabolism
2.
Cancer Biol Ther ; 15(8): 1053-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25084100

ABSTRACT

TRA-8, a monoclonal antibody targeting death receptor, has demonstrated high therapeutic effect for triple negative breast cancer (TNBC) in preclinical models. Tamoxifen, the standard of care for ERα-positive breast cancer, induces apoptosis via ERß, which commonly presents in TNBC cells. The current study investigates the combination effects of TRA-8 and tamoxifen for TNBC. In vitro assays were implemented with two ERß-positive TNBC cell lines, SUM159 and 2LMP, and in vivo therapy studies were followed using orthotopic breast tumor mouse models. IC50 of tamoxifen for SUM159 and 2LMP were 29 µM and 38 µM, respectively. Synergy between TRA-8 (0-1000 ng/mL) and tamoxifen (20 µM) was observed for both the cell lines. Tamoxifen (400 mg/kg diet) markedly suppressed the growth of SUM159 tumors for 6 weeks after therapy initiation, but it did not induce antitumor effect for 2LMP tumors. TRA-8 (0.1 mg, weekly, i.p.) successfully arrested the growth of both SUM159 and 2LMP tumors during therapy, but an antagonistic effect was observed when tamoxifen was combined. TRA-8 uptake into tumors was not changed by tamoxifen treatment. Histological analysis confirmed that caspase-3 activation induced by TRA-8 was significantly decreased when tamoxifen was used in combination. In conclusion, our findings suggest that the combined use of TRA-8 and tamoxifen may cause antagonistic effects for TNBC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Female , Humans , Inhibitory Concentration 50 , Mice, Nude , Protein Multimerization , Tamoxifen/administration & dosage
3.
J Magn Reson Imaging ; 39(6): 1588-94, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24151215

ABSTRACT

PURPOSE: To assess the early response of triple-negative breast-cancer (TNBC) following TRA-8 and carboplatin therapy using DWI and MRS in 2LMP and SUM159 mouse models. MATERIALS AND METHODS: Four groups (n = 5/group) of each model were untreated or treated with carboplatin, TRA-8, and combination, respectively. DWI and MRS were applied on 0, 3, and 7 days after therapy initiation, and all tumors were collected thereafter for terminal deoxynucleotidyl transferase mediated dUTP nick end labeling (TUNEL) staining. The changes in intratumoral apparent diffusion coefficient (ADC) and fat-water ratios (FWRs) were compared with tumor volume changes and apoptotic cell densities. RESULTS: Mean ADC values of 2LMP and SUM159 tumors significantly increased 4 ± 4% and 37 ± 11% during 7 days of combination therapy, respectively, as compared to control groups (P < 0.05). Similarly, mean FWRs of 2LMP and SUM159 tumors significantly increased 102 ± 30% and 126 ± 52%, respectively, for 7 days of combined treatment (P < 0.05). The changes of the mean ADC values for 3 days (or FWRs for 7 days) were linearly proportional to either the mean volume changes or apoptotic cell densities in both models. CONCLUSION: DWI and MRS assessed the early tumor response to TRA-8 and carboplatin in TNBC mouse models.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carboplatin/therapeutic use , Diffusion Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Mammary Neoplasms, Experimental/drug therapy , Triple Negative Breast Neoplasms/drug therapy , Analysis of Variance , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Female , Heterografts , Humans , Mice , Mice, Nude , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Transplantation, Heterologous , Treatment Outcome
4.
Ann Surg Oncol ; 20(9): 3059-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23525731

ABSTRACT

PURPOSE: Ovarian serous carcinoma is an aggressive cancer that often presents with metastatic disease. Although primary tumor and established metastatic foci in the omentum are generally compared to identify proteins involved in drug resistance, we investigated a potential bridge, the malignant cells from ascites, as facilitator of drug resistance and recurrence. METHODS: We evaluated the expression of drug resistance markers P-glycoprotein (P-gp), canalicular multispecific organic anion transporter (MRP2), and lung resistance-related protein (LRP) in malignant cells from ascites and matched omental metastasis from 25 patients with advanced-stage ovarian serous carcinoma who were chemotherapeutic naïve and undergoing initial cytoreductive surgery. Cell viability in vitro, patient response to chemotherapy, and patient survival were correlated with these biomarkers. RESULTS: Of the 25 patients evaluated for a correlation of LRP to 1-year recurrence, we correctly predicted the 1-year recurrence of 24 patients based solely on the presence of LRP in ascitic tumor cells (p=0.01). P-gp and MRP2 were not expressed in malignant cells of ascites or omental metastases. Malignant cells from ascites had higher expression of LRP and were found to be more resistant to carboplatin treatment than cells from omental metastasis (p=0.00375) by in vitro assay. LRP expression in the malignant cells of ascites correlated with carboplatin resistance (p=0.001) by in vitro assay and recurrence at 1 year (p=0.0125). CONCLUSIONS: LRP expression in malignant cells of ascites is a promising marker to predict response to first-line chemotherapy in patients with advanced ovarian serous carcinoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Ascites/mortality , Cystadenocarcinoma, Serous/mortality , Neoplasm Recurrence, Local/mortality , Ovarian Neoplasms/mortality , Vault Ribonucleoprotein Particles/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Aged , Ascites/metabolism , Ascites/pathology , Biomarkers, Tumor/metabolism , Blotting, Western , Carboplatin/administration & dosage , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Microfilament Proteins/metabolism , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Prognosis , Survival Rate , Tumor Cells, Cultured
5.
J Biomed Opt ; 15(6): 067005, 2010.
Article in English | MEDLINE | ID: mdl-21198209

ABSTRACT

A combination tapered fiber-optic biosensor (CTFOB) dip probe for rapid and cost-effective quantification of proteins in serum samples has been developed. This device relies on diode laser excitation and a charged-coupled device spectrometer and functions on a technique of sandwich immunoassay. As a proof of principle, this technique was applied in a quantitative estimation of interleukin IL-6. The probes detected IL-6 at picomolar levels in serum samples obtained from a patient with lupus, an autoimmune disease, and a patient with lymphoma. The estimated concentration of IL-6 in the lupus sample was 5.9 ± 0.6 pM, and in the lymphoma sample, it was below the detection limit. These concentrations were verified by a procedure involving bead-based xMAP technology. A similar trend in the concentrations was observed. The specificity of the CTFOB dip probes was assessed by analysis with receiver operating characteristics. This analysis suggests that the dip probes can detect 5-pM or higher concentration of IL-6 in these samples with specificities of 100%. The results provide information for guiding further studies in the utilization of these probes to quantify other analytes in body fluids with high specificity and sensitivity.


Subject(s)
Biosensing Techniques/instrumentation , Fiber Optic Technology/instrumentation , Immunoassay/instrumentation , Interleukin-6/blood , Refractometry/instrumentation , Transducers , Equipment Design , Equipment Failure Analysis , Humans
6.
Mol Pharmacol ; 76(1): 81-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19395473

ABSTRACT

Jab1 (Jun activation domain binding protein 1), integrated into COP9 signalosome complex (CSN), induces protein instability of many tumor suppressors and cell cycle regulators and is therefore a novel target in cancer therapy. Curcumin, an inhibitor of Jab1/CSN-associated kinase(s), has been reported to suppress tumor growth; however, curcumin is highly hydrophobic, and this feature prevents its usage as an antitumor drug. To increase the solubility and targeted delivery, we generated a water-soluble polyethylene glycol (PEG)-conjugated curcumin system, in which curcumin is covalently linked to PEG(35kD). PEGylated curcumin showed much greater reduction of cell growth than free curcumin in pancreatic cancer cells. Cells treated with PEGylated curcumin had increased arrest at the mitotic phase with the formation of abnormal multinucleated cells, indicating that this compound affects cell cycle progression, which may contribute to cell growth inhibition. The stabilities of Jab1 target proteins were also examined. PEGylated curcumin increased protein stability of these proteins in pancreatic cancer cells and directly inhibited the activity of Jab1/CSN-associated kinases. Moreover, the inhibitory effect of PEGylated curcumin on cell proliferation was blunted in pancreatic cancer cells with Jab1 knockdown. The results suggest that PEGylated curcumin inhibits cell proliferation through suppression of Jab1/CSN activity. More importantly, the new compound sensitized pancreatic cancer cells to gemcitabine-induced apoptosis and cell proliferation inhibitory effects. Collectively, the PEGylated curcumin conjugate has much more potent effects on pancreatic cancer cell growth inhibition than free curcumin. The current study provides a biologic rationale to treat patients with pancreatic adenocarcinoma with the nontoxic phytochemical conjugated to PEG for systemic delivery.


Subject(s)
Curcumin/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Polyethylene Glycols/pharmacology , Apoptosis/drug effects , COP9 Signalosome Complex , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/physiology , Male , Pancreatic Neoplasms/pathology , Peptide Hydrolases/physiology , Gemcitabine
7.
Cell Cycle ; 6(23): 2944-52, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18000399

ABSTRACT

LIM kinases (LIMK1 and LIMK2) are LIM domain containing serine/threonine kinases that modulate reorganization of actin cytoskeleton through inactivating phosphorylation of cofilin. The Rho family of small GTPases regulates the catalytic activity of LIMK1 and LIMK2 through activating phosphorylation by ROCK or by p21 kinase. Recent studies have suggested that LIMK1 could play a role in modulation of cellular growth by alteration of the cell cycle in breast and prostate tumor cells; however, the direct mitogenic effects of LIMK1 in these tumor cells is yet to be elucidated. Via immunofluorescence, in this study, we show that phosphorylated LIM kinases (pLIMK1/2) are colocalized with gamma-tubulin in the centrosomes during the early mitotic phases of human breast and prostate cancer cells (MDA-MB-231 and DU145); apparent colocalization begins in the centrosomes in prophase. As shown by both bright field (MDA-MB-231) and fluorescent immunohistochemistry (MDA-MB-231 and DU145), pLIMK1/2 does not localize to centrosomes during interphase. By bright field immunohistochemistry, the largest area of the centrosome that is stained with pLIMK1/2 occurs at anaphase. In early telophase, reduced staining of pLIMK1/2 at the spindle poles and concomitant accumulation of pLIMK1/2 at the cleavage furrow begins to occur. In late telophase, loss of staining of pLIMK1/2 and of colocalization with gamma-tubulin occurs at the poles and pLIMK1/2 became further concentrated at the junction between the two daughter cells. Co-immunoprecipitation studies indicated that gamma-tubulin associates with phosphorylated LIMK1 and LIMK2 but not with dephosphorylated LIMK1 or LIMK2. The results suggest that activated LIMK1/2 may associate with gamma-tubulin and play a role in mitotic spindle assembly.


Subject(s)
Centrosome/chemistry , Lim Kinases/metabolism , Mitosis , Tubulin/metabolism , Cell Line, Tumor , Humans , Interphase , Phosphorylation , Protein Transport , Spindle Apparatus
8.
Am J Surg ; 193(2): 274-83, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17236862

ABSTRACT

BACKGROUND: Methionine, an essential amino acid, is important for normal growth and development, as it is required for both protein and polyamine synthesis as well as in methylation reactions. It has been reported that high concentrations of methionine inhibit cellular growth and gene expression in the human breast tumor-derived MCF-7 cells. These effects are thought to be mediated by the modulation of p53. However, the generalizability of this observation and the precise role of p53 in methionine-induced growth suppression needs to be determined. METHODS: To determine if the inhibition of cell growth by methionine applies to other cell lines and to characterize further the role of p53 in methionine-induced growth suppression, we have assessed the effects of methionine on cellular growth and proliferation and p53 expression in cells expressing native p53, eg, breast cancer MCF-7 cells and prostate cancer LNCaP cells, and also in cells expressing a mutated (point) form of p53, eg, prostate cancer DU-145 cells. These cell lines were treated with varying concentrations of L-methionine. The effects of L-methionine on cell growth were assayed by using cell viability assays and immunostaining for Ki-67, a cell proliferation marker. The effects of methionine on p53 expression were assessed by using reverse transcriptase-polymerase chain reaction (RT-PCR), immunohistochemistry, and Western blot analysis. The role of p53 in L-methionine-mediated growth suppression was evaluated using short-interference RNA for p53 (siRNA-p53), immunoprecipitation, and direct DNA sequencing. RESULTS: We demonstrated that methionine at a concentration of 1 to 5 mg/mL inhibited the growth of both MCF-7 and LNCaP cells. In association with the inhibition of growth, methionine also inhibited native p53 expression at the mRNA and protein levels, respectively. Furthermore, transfection with siRNA-p53, to knock down p53 expression, increased cell growth and proliferation of the LNCaP cells even when they were exposed to methionine. In contrast, the same treatment did not diminish growth or proliferation of the DU-145 cells. Also, the expression of mutated p53 at the mRNA or protein levels was not altered. CONCLUSION: Our results extend a prior observation to other cell lines and demonstrate that high concentrations of methionine suppress the expression of native but not mutated p53. These inhibitory effects on cellular growth are, in part, due to inhibition of cellular proliferation probably via a p53-dependent pathway.


Subject(s)
Amino Acids/pharmacology , Cell Proliferation/drug effects , Methionine/pharmacology , Tumor Suppressor Protein p53/biosynthesis , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Male , Prostatic Neoplasms/metabolism
9.
EMBO J ; 26(2): 346-57, 2007 Jan 24.
Article in English | MEDLINE | ID: mdl-17183365

ABSTRACT

Alterations in the signaling pathways of bone morphogenetic proteins (BMPs) and activation of the ERK/MAP kinase (MAPK) pathway by growth factors have been implicated in the development and progression of prostate cancer. Smad1 acts as a substrate for MAPKs and also performs a central role in transmitting signals from BMPs. We found that BMPs/Smad1 signaling inhibits the growth of androgen-sensitive prostate cancer cells. Upon the incorporation of ERK/MAPK signals at its linker region, Smad1 physically interacts with androgen-activated androgen receptor (AR) and suppresses its functions. BMPs induce the function of Smad1 as an AR transcriptional corepressor. We demonstrated in vivo that Smad1 signaling is low in androgen-regulated growth of prostate cancer, is activated after castration, and also is decreased in hormone-independent tumors. The activation status of ERK/MAPK parallels Smad1 in the progression of prostate cancer; thus, our findings indicate a molecular basis for the integration of signals of MAPK and Smad1 in the progression and androgen regulation of prostate cancer.


Subject(s)
Androgens/physiology , Bone Morphogenetic Proteins/physiology , Cell Proliferation , Mitogen-Activated Protein Kinases/physiology , Prostate/cytology , Smad1 Protein/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Androgens/metabolism , Animals , Cells, Cultured , Humans , Male , Mice , Models, Biological , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/physiology , Signal Transduction , Smad1 Protein/metabolism , Xenograft Model Antitumor Assays
10.
Cancer Inform ; 2: 345-9, 2007 Feb 27.
Article in English | MEDLINE | ID: mdl-19458776

ABSTRACT

Early detection of precancerous cells in the cervix and their clinical management is the main purpose of cervical cancer prevention and treatment programs. Cytological findings or testing for high risk (HR)-human papillomavirus (HPV) are inadequately sensitive for use in triage of women at high risk for cervical cancer. The current study is an exploratory study to identify candidate surface-enhanced laser desorption/ionization (SELDI) time of flight (TOF) mass spectrometry (MS) protein profiles in plasma that may distinguish cervical intraepithelial neoplasia (CIN 3) from CIN 1 among women infected with HR-HPV. We evaluated the SELDI-TOF-MS plasma protein profiles of HR-HPV positive 32 women with CIN 3 (cases) and 28 women with CIN1 (controls). Case-control status was kept blinded and triplicates of each sample and quality control plasma samples were randomized and after robotic sample preparations were run on WCX2 chips. After alignment of mass/charge (m-z values), an iterative method was used to develop a classifier on a training data set that had 28 cases and 22 controls. The classifier developed was used to classify the subjects in a test data set that has six cases and six controls. The classifier separated the cases from controls in the test set with 100% sensitivity and 100% specificity suggesting the possibility of using plasma SELDI protein profiles to identify women who are likely to have CIN 3 lesions.

11.
Arch Med Res ; 37(6): 689-95, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16824926

ABSTRACT

BACKGROUND: There is controversy on the safety of inhibitors of cyclooxygenase administered at high doses; however, these drugs have been reported to be effective in the prevention of a variety of human cancers. To determine if celecoxib influences cellular growth, we evaluated several effects in ovarian carcinoma cell lines. METHODS: CAOV3, OVCAR3 and SKOV3 cell lines were exposed to different concentrations of celecoxib (0-100 microM) for 24-96 h. Cellular growth was assessed using a cell viability assay. Immunohistochemistry was performed to evaluate Ki-67 and cleaved caspase-3. Apoptosis was determined by a TUNEL assay, and Western blot was used to determine COX-2 protein expression. RESULTS: We observed a significant decrease in the cellular growth of all cell lines studied exposed to > or = 70 microM of celecoxib for 72 and 96 h (p < 0.02). All cells demonstrated pancytotoxicity at 100 microM of celecoxib. A significant decrease in Ki-67 expression in all cell lines exposed to > or = 30 microM of celecoxib (p < or = 0.05) for 72 h was observed. We observed significant changes in apoptosis and cleaved caspase-3 expression in SKOV3 cells exposed to 50 microM of celecoxib. Downregulation of COX-2 protein expression caused by celecoxib was observed in SKOV3 cells. CONCLUSIONS: We found that celecoxib inhibits cellular growth and proliferation in a dose-dependent manner in all cell lines studied. SKOV3 cells showed an increase in cleaved caspase-3 expression. Additional studies are in progress to evaluate the effects of celecoxib on other aspects of the control of the cell cycle in cancer cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor/drug effects , Ki-67 Antigen/metabolism , Ovarian Neoplasms/metabolism , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Caspase 3/metabolism , Celecoxib , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dose-Response Relationship, Drug , Female , Humans , In Situ Nick-End Labeling
12.
J Biol Chem ; 280(29): 27022-8, 2005 Jul 22.
Article in English | MEDLINE | ID: mdl-15886208

ABSTRACT

Estrogen is a mitogen in most estrogen receptor-alpha (ERalpha)-positive breast cancers. We have found that Smad4, a common signal transducer in the transforming growth factor-beta superfamily, acts as an ERalpha transcriptional corepressor. Here, we show that Smad4 induces apoptosis in ERalpha-positive MCF-7 breast cancer cells, but not in ERalpha-negative MDA-MB-231 cells. Smad4 induced expression of short Bim isoforms (by alternative splicing) and Bax and release of cytochrome c in ERalpha-positive cells only, and expression of these apoptotic marker genes was reduced when ERalpha small interfering RNA was introduced. Notably, Smad4 was able to induce apoptosis in MDA-231 cells with acquired ERalpha expression. Furthermore, Smad4 inhibited ERalpha-positive tumor growth by inducing apoptosis in tumor xenografts in nude mice. The sizes of tumors expressing Smad4 were only one-tenth the size of those expressing green fluorescent protein, whereas in ERalpha-negative cells, Smad4 did not reduce the tumor size. Notably, Smad4 also promoted short Bim isoform and Bax expression and release of cytochrome c only in ERalpha-positive MCF-7 tumor xenografts. Bim was sufficient for induction of apoptosis, and the short form was the most potent inducer. Our results demonstrate that Smad4 induces apoptosis by regulating Bim splicing as an initial intrinsic signal in ERalpha-positive cells. Smad4-induced apoptosis in ERalpha-positive breast cancer cells may explain the invasive nature of ERalpha-negative breast tumors, thereby providing a potential target for breast cancer intervention.


Subject(s)
Apoptosis/genetics , Breast Neoplasms/pathology , DNA-Binding Proteins/physiology , Estrogen Receptor alpha/genetics , Trans-Activators/physiology , Alternative Splicing , Animals , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Breast Neoplasms/chemistry , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Cytochromes c/metabolism , Female , Genetic Therapy , Humans , Membrane Proteins/genetics , Mice , Mice, Nude , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Smad4 Protein , Transplantation, Heterologous , bcl-2-Associated X Protein
13.
Cancer Inform ; 1: 86-97, 2005.
Article in English | MEDLINE | ID: mdl-19305634

ABSTRACT

Multiple studies have reported that surface enhanced laser desorption/ionization time of flight mass spectroscopy (SELDI-TOF-MS) is useful in the early detection of disease based on the analysis of bodily fluids. Use of any multiplex mass spectroscopy based approach as in the analysis of bodily fluids to detect disease must be analyzed with great care due to the susceptibility of multiplex and mass spectroscopy methods to biases introduced via experimental design, patient samples, and/or methodology. Specific biases include those related to experimental design, patients, samples, protein chips, chip reader and spectral analysis. Contributions to biases based on patients include demographics (e.g., age, race, ethnicity, sex), homeostasis (e.g., fasting, medications, stress, time of sampling), and site of analysis (hospital, clinic, other). Biases in samples include conditions of sampling (type of sample container, time of processing, time to storage), conditions of storage, (time and temperature of storage), and prior sample manipulation (freeze thaw cycles). Also, there are many potential biases in methodology which can be avoided by careful experimental design including ensuring that cases and controls are analyzed randomly. All the above forms of biases affect any system based on analyzing multiple analytes and especially all mass spectroscopy based methods, not just SELDI-TOF-MS. Also, all current mass spectroscopy systems have relatively low sensitivity compared with immunoassays (e.g., ELISA). There are several problems which may be unique to the SELDI-TOF-MS system marketed by Ciphergen(®). Of these, the most important is a relatively low resolution (±0.2%) of the bundled mass spectrometer which may cause problems with analysis of data. Foremost, this low resolution results in difficulties in determining what constitutes a "peak" if a peak matching approach is used in analysis. Also, once peaks are selected, the peaks may represent multiple proteins. In addition, because peaks may vary slightly in location due to instrumental drift, long term identification of the same peaks may prove to be a challenge. Finally, the Ciphergen(®) system has some "noise" of the baseline which results from the accumulation of charge in the detector system. Thus, we must be very aware of the factors that may affect the use of proteomics in the early detection of disease, in determining aggressive subsets of cancers, in risk assessment and in monitoring the effectiveness of novel therapies.

14.
Head Neck ; 24(9): 859-67, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12211050

ABSTRACT

BACKGROUND: Despite the importance of reactive oxygen species (ROS) in the development of smoking-related cancers, little is known about the pattern of expression of ROS scavengers in these cancers. METHODS: In this present study, we examined the expression of manganese superoxide dismutase (Mn-SOD) and copper/zinc superoxide dismutase (Cu-Zn-SOD), which are essential enzymes that eliminate ROS, in squamous cell cancers (SCCs) of the lung (n = 12), larynx (n = 13), and oral cavity (n = 20). RESULTS: SCCs of larynx and oral cavity showed significantly enhanced immuhistochemical expression of Mn-SOD compared with the matched uninvolved epithelium. The higher expression of Mn-SOD was shown to be late and early events in the process of SCC development in the larynx and the oral cavity, respectively. The expression of Mn-SOD in SCCs of the lung was significantly lower compared with luminal cells of the uninvolved epithelium but not compared with basal cells or an average expression of SOD in basal and luminal cells. The expression of both Mn-SOD and cytoplasmic or nuclear Cu-Zn-SOD in bronchial epithelium adjacent to invasive cancer was significantly lower compared with its expression in the uninvolved bronchial epithelium away from cancer. This resulted in a significant difference in SOD expression between cancer and uninvolved bronchial epithelium away from cancer but not between cancer and uninvolved epithelium adjacent to cancer. CONCLUSIONS: There are significant differences in the expression of Mn-SOD and Cu-Zn-SOD among SCCs of the lung, larynx, and oral cavity. The results also suggest that variations in distance between cancer and uninvolved tissues evaluated could contribute to conflicting results of SOD expression.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Free Radical Scavengers/metabolism , Laryngeal Neoplasms/enzymology , Lung Neoplasms/enzymology , Mouth Neoplasms/enzymology , Superoxide Dismutase/metabolism , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Case-Control Studies , Cytoplasm/enzymology , Formaldehyde , Free Radical Scavengers/analysis , Humans , Immunoenzyme Techniques , Laryngeal Neoplasms/pathology , Lung Neoplasms/pathology , Middle Aged , Mouth Neoplasms/pathology , Neoplasm Staging , Paraffin Embedding , Reactive Oxygen Species/metabolism , Superoxide Dismutase/analysis
15.
Clin Cancer Res ; 8(1): 202-9, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11801560

ABSTRACT

PURPOSE: Nonsteroidal anti-inflammatory agents may inhibit carcinogenesis in specific tissues including the colon, breast, and pancreas, and, hence, may prove to be effective chemopreventive agents. The purpose of this study was to investigate the cellular effects of acetylsalicylic acid (ASA), acetaminophen, and a COX-2 inhibitor (NS-398) on the growth of cell lines of human ovarian cancer in vitro. EXPERIMENTAL DESIGN: SK-OV-3, Caov-3, and NIH:OVCAR-3 ovarian carcinoma cell lines were treated with ASA (10(-6) M-10(-2) M), acetaminophen (10(-6) M-10(-2) M), and a COX-2 inhibitor (10(-6) M-10(-4) M) for 96 h. The number of viable cells was determined using a tetrazolium conversion assay. Immunohistochemical assessment was performed for alterations in expression of Ki-67, erbB-2, COX enzyme, and apoptosis in primary ovarian cancer cells using terminal deoxynucleotidyl transferase (Tdt)-mediated nick end labeling assay. RESULTS: A decrease in cell number compared with controls was observed for all of the cell lines treated with ASA, acetaminophen, and COX-2 inhibitor by cell count and tetrazolium conversion assay. A significant decrease in Ki-67 compared with controls in the OVCAR-3 (P = 0.005) and SK-OV-3 (P = 0.007) cell lines after treatment with the COX-2 inhibitor was observed. We observed a decrease in mitotic activity compared with controls in each cell line after treatment with the COX-2 inhibitor. Apoptosis was observed in primary ovarian cancer cell culture treated with COX-2 inhibitor. CONCLUSION: Our results suggest additional study for the use of nonsteroidal anti-inflammatory agents, specifically COX-2 inhibitors, as a strategy of chemoprevention for ovarian cancer.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Ovarian Neoplasms/prevention & control , Acetaminophen/pharmacology , Apoptosis/drug effects , Aspirin/pharmacology , Biopsy , Blotting, Western , Cell Division/drug effects , Cyclooxygenase 2 , Drug Evaluation, Preclinical , Female , Humans , Immunoenzyme Techniques , In Situ Nick-End Labeling , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Ki-67 Antigen/metabolism , Membrane Proteins , Nitrobenzenes/pharmacology , Organ Culture Techniques , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prostaglandin-Endoperoxide Synthases/metabolism , Receptor, ErbB-2/metabolism , Sulfonamides/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...