Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Oncogene ; 28(49): 4344-52, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19784068

ABSTRACT

ApoJ/Clusterin (CLU) is a heterodimeric protein localized in the nucleus, cytoplasm or secretory organelles and involved in cell survival and neoplastic transformation. Its function in human cancer is still highly controversial. In this study, we examined the prostate of mice in which CLU has been genetically inactivated. Surprisingly, we observed transformation of the prostate epithelium in the majority of CLU knockout mice. Either PIN (prostate intraepithelial neoplasia) or differentiated carcinoma was observed in 100 and 87% of mice with homozygous or heterozygous deletion of CLU, respectively. Crossing CLU knockout with TRAMP (prostate cancer prone) mice results in a strong enhancement of metastatic spread. Finally, CLU depletion causes tumourigenesis in female TRAMP mice, which are normally cancer free. Mechanistically, deletion of CLU induces activation of nuclear factor-kB, a potentially oncogenic transcription factor important for the proliferation and survival of prostate cells.


Subject(s)
Carcinoma/pathology , Cell Transformation, Neoplastic/drug effects , Clusterin/antagonists & inhibitors , Clusterin/genetics , Prostatic Neoplasms/pathology , RNA, Small Interfering/pharmacology , Animals , Carcinoma/genetics , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/genetics , RNA Interference/physiology
2.
Oncogene ; 28(2): 257-69, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18931701

ABSTRACT

Allelic deletions on human chromosome 12q24 are frequently reported in a variety of malignant neoplasms, indicating the presence of a tumor suppressor gene(s) in this chromosomal region. However, no reasonable candidate has been identified so far. In this study, we report the cloning and functional characterization of a novel mitochondrial protein with tumor suppressor activity, henceforth designated MITOSTATIN. Human MITOSTATIN was found within a 3.2-kb transcript, which encoded a approximately 62 kDa, ubiquitously expressed protein with little homology to any known protein. We found homozygous deletions and mutations of MITOSTATIN gene in approximately 5 and approximately 11% of various cancer-derived cells and solid tumors, respectively. When transiently overexpressed, MITOSTATIN inhibited colony formation, tumor cell growth and was proapoptotic, all features shared by established tumor suppressor genes. We discovered a specific link between MITOSTATIN overexpression and downregulation of Hsp27. Conversely, MITOSTATIN knockdown cells showed an increase in cell growth and cell survival rates. Finally, MITOSTATIN expression was significantly reduced in primary bladder and breast tumors, and its reduction was associated with advanced tumor stages. Our findings support the hypothesis that MITOSTATIN has many hallmarks of a classical tumor suppressor in solid tumors and may play an important role in cancer development and progression.


Subject(s)
Breast Neoplasms/genetics , Chromosomes, Human, Pair 12/genetics , Genes, Tumor Suppressor , Neoplasm Proteins/biosynthesis , Tumor Suppressor Proteins/physiology , Urinary Bladder Neoplasms/genetics , Animals , Apoptosis/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins , Cell Division/genetics , Cell Line, Tumor/metabolism , Cell Line, Tumor/ultrastructure , Cell Transformation, Neoplastic/genetics , Cloning, Molecular , Disease Progression , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , HSP27 Heat-Shock Proteins/biosynthesis , HSP27 Heat-Shock Proteins/genetics , Heat-Shock Proteins , Humans , Male , Mitochondria/metabolism , Mitochondria/ultrastructure , Molecular Chaperones , Molecular Sequence Data , Neoplasm Proteins/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Recombinant Fusion Proteins/physiology , Species Specificity , Tumor Stem Cell Assay , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/isolation & purification , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
3.
J Exp Clin Cancer Res ; 26(1): 25-37, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17550130

ABSTRACT

Prostate cancer (PC) development reflects a complex sequence of biologic and molecular events. Several inheritable and somatic genetic changes have been identified. The knowledge of the molecular basis of PC can improve our understanding of the causes of this common cancer and provide information on prognosis and treatment. To date, however, no molecular studies have yet yielded consistent information that is ready to be incorporated into clinical practice. We reviewed the current literature on the molecular biology of prostate cancer and analyzed different potential tumor markers according to the classical concepts of oncogenes, suppressor genes, and the more modern concepts of genes involved in detoxification or inflammatory pathways of cancer progression. This review aims to identify trends in PC research and suggests potential clinical applications for diagnosis, prognosis, prevention and treatment.


Subject(s)
Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Genetic Predisposition to Disease , Humans , Loss of Heterozygosity , Male , Pedigree , Prognosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy
4.
J Exp Clin Cancer Res ; 25(2): 145-60, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16918124

ABSTRACT

Transitional cell carcinoma of the bladder is a common tumor. While most patients presenting superficial disease can be expected to do well following treatment, still many patients will return to our office with muscle invasive and metastatic disease. Survival in advanced bladder cancer is less than 50%. Tumors of similar histologic grade and stage have variable behavior, suggesting that genetic alterations must be present to explain the diverse behavior of bladder cancer. It is hoped that through the study of the subtle genetic alterations in bladder cancer, important prognostic and therapeutic targets can be exploited. Many new diagnostic tests and gene therapy approaches rely on the identification and targeting of these unique genetic alterations. A review of literature published on the molecular genetics of bladder cancer from 1970 to the present was conducted. A variety of molecular genetic alterations have been identified in bladder cancer. Oncogenes (H-ras, erbB-2, EGFR, MDM2, C-MYC, CCND1), tumor suppressor genes (p53, Rb, p21, p27/KIP1, p16, PTEN, STK15, FHIT, FEZ1/LZTS1, bc10), telomerase, and methylation have all been studied in bladder cancer. Several have proven to be potentially useful clinical targets in the prognosis and therapy of bladder cancer such as staining for p53 and gene therapy strategies such as p53 and fez1. Clinical trials targeting HER2/neu and the EGFR pathways are underway. The UroVysion bladder cancer assay relies on FISH to detect genetic alterations in this disease. Continuing identification of the molecular genetic alterations in bladder cancer will enhance future diagnostic and therapeutic approaches to bladder cancer. Capitalizing on these alterations will allow early detection, providing important prognostic information and unique targets for gene therapy and other therapeutic approaches.


Subject(s)
Biomarkers, Tumor/genetics , Genetic Therapy , Urinary Bladder Neoplasms/genetics , Animals , Humans , Molecular Biology , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/therapy
5.
Cell Death Differ ; 9(9): 995-1006, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12181750

ABSTRACT

Caspase-14 is a recent addition to the caspase family of aspartate proteases involved in apoptotic processes. Human caspase-14 appears to be only weakly processed during apoptosis, and it does not cleave classical caspase substrates. Post partum, caspase-14 is prominently expressed by human keratinocytes and reportedly participates in terminal differentiation of complex epithelia. Here we provide evidence challenging the view that caspase-14 expression or processing is linked exclusively to terminal keratinocyte differentiation. We demonstrate that caspase-14 expression extended to multiple cell lines derived from simple epithelia of the breast, prostate, and stomach. In keratinocytes and breast epithelial cells, caspase-14 expression was upregulated in high-density cultures and during forced suspension culture. These effects were primarily due to transcriptional activation as indicated by reporter gene assays using a 2 kb caspase-14 promoter fragment. Importantly, caspase-14 was not cleaved during forced suspension culture of either cell type although this treatment induced caspase-dependent apoptosis (anoikis). Forced expression of caspase-14 in immortalized human keratinocytes had no effect on cell death in forced suspension nor was the transfected caspase-14 processed in this setting. In contrast to postconfluent and forced suspension culture, terminal differentiation of keratinocytes induced in vitro by Ca2+ treatment was not associated with increased caspase-14 expression or promoter activity. Our results indicate that (1) caspase-14 is expressed not only in complex but also simple epithelia; (2) cells derived from complex and simple epithelia upregulate caspase-14 expression in conditions of high cell density or lack of matrix interaction and; (3) in both cell types this phenomenon is due to transcriptional regulation.


Subject(s)
Caspases/genetics , Cell Differentiation/genetics , Epithelial Cells/enzymology , Epithelium/enzymology , Gene Expression Regulation, Enzymologic/genetics , Genes, Regulator/genetics , Antibody Specificity/immunology , Breast/cytology , Breast/enzymology , Breast/growth & development , Caspase 14 , Cell Adhesion/physiology , Cell Compartmentation/physiology , Cell Cycle/physiology , Cells, Cultured , Epidermal Cells , Epidermis/enzymology , Epidermis/growth & development , Epithelial Cells/cytology , Epithelium/growth & development , Extracellular Matrix/enzymology , Humans , Infant, Newborn , Keratinocytes/cytology , Keratinocytes/enzymology , Male , Promoter Regions, Genetic/genetics , Prostate/cytology , Prostate/enzymology , Prostate/growth & development
6.
JAMA ; 286(19): 2441-9, 2001 Nov 21.
Article in English | MEDLINE | ID: mdl-11712940

ABSTRACT

CONTEXT: The fragile histidine triad gene (FHIT) encompasses a human common fragile site, FRA3B, that is susceptible to environmental carcinogens. Deletion and inactivation of FHIT have been seen in a number of human premalignant and malignant lesions. OBJECTIVE: To review and evaluate preclinical studies of cancer therapy using the FHIT tumor suppressor gene and related studies involving Fhit protein expression. DATA SOURCES: A MEDLINE search of articles published from 1996 to June 2001 was performed; article reference lists were used to retrieve additional relevant articles. STUDY SELECTION: Immunohistochemical studies of primary tumors or relevant lesions were selected to evaluate Fhit expression in premalignant or malignant stages. Preclinical studies on antitumorigenic or therapeutic introduction of FHIT were reviewed for the effects of exogenous Fhit expression. For the immunohistochemical analyses, 26 studies were included that analyzed at least 15 cases of a single type of tumor. For precancerous lesions, 9 studies were included that analyzed at least 4 cases. For studies of FHIT introduction, 9 published studies were included. DATA EXTRACTION: Using primary data from each of the studies, we assessed the rationale and potential contribution of FHIT cancer therapy. Data was independently abstracted by 2 authors and study quality was assessed by 2 other authors. DATA SYNTHESIS: Overall, 60% (1162/1948 cases) of primary tumors showed absent or markedly reduced Fhit protein expression in cancer cells. Studies of preneoplastic lesions or early-stage cancer showed absence or marked reduction of Fhit protein expression in 0% to 93% of samples (overall, 31% [127/408 cases]). Preclinical studies using 26 cancer-derived cell lines from human lung, head and neck, esophageal, gastric, cervical, pancreatic, and kidney cancers, showed that reintroduction of FHIT resulted in inhibition of in vitro tumor cell growth or of in vivo tumorigenicity in 17 (57%) of 30 cell line experiments. Model systems for human preventive cancer therapy suggested that oral introduction of viral vector-mediated FHIT into Fhit-deficient mice may prevent carcinogen-induced tumor development in some cases. CONCLUSION: These findings show that FHIT gene therapy may potentially be clinically useful for treatment of cancer and also prevention of carcinogen-induced tumor development, suggesting a rationale for further research involving FHIT introduction.


Subject(s)
Acid Anhydride Hydrolases , Genes, Tumor Suppressor , Genetic Therapy , Neoplasm Proteins/genetics , Neoplasms/therapy , Precancerous Conditions/therapy , Animals , Evaluation Studies as Topic , Humans , Immunohistochemistry , Mice , Models, Animal , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Transfection
7.
Proc Natl Acad Sci U S A ; 98(18): 10374-9, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11504921

ABSTRACT

The FEZ1/LZTS1 gene maps to chromosome 8p22, a region that is frequently deleted in human tumors. Alterations in FEZ1/LZTS1 expression have been observed in esophageal, breast, and prostate cancers. Here, we show that introduction of FEZ1/LZTS1 into Fez1/Lzts1-negative cancer cells results in suppression of tumorigenicity and reduced cell growth with accumulation of cells at late S-G(2)/M stage of the cell cycle. Fez1/Lzts1 protein is hyperphosphorylated by cAMP-dependent kinase during cell-cycle progression. We found that Fez1/Lzts1 is associated with microtubule components and interacts with p34(cdc2) at late S-G(2)/M stage in vivo. Present data show that FEZ1/LZTS1 inhibits cancer cell growth through regulation of mitosis, and that its alterations result in abnormal cell growth.


Subject(s)
Chromosomes, Human, Pair 8/genetics , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Neoplasms/genetics , Neoplasms/pathology , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing , CDC2 Protein Kinase/metabolism , Cell Cycle/genetics , Cell Division/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Male , Microtubules/metabolism , Mitosis/genetics , Neoplasms/metabolism , Nerve Tissue Proteins , Peptide Elongation Factor 1/metabolism , Phosphorylation , Protein Binding , Transfection , Tumor Cells, Cultured
8.
Clin Cancer Res ; 7(6): 1546-52, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11410489

ABSTRACT

PURPOSE: Loss of heterozygosity (LOH) involving the short arm of chromosome 8 (8p) is a common feature of the malignant progression of human tumors, including gastric cancer. We have cloned and mapped a candidate tumor suppressor gene, FEZ1/LZTS1, to 8p22. Here we have analyzed whether FEZ1/LZTS1 alterations play a role in the development and progression of gastric carcinoma. EXPERIMENTAL DESIGN: We examined Fez1/Lzts1 expression in 8 gastric carcinoma cell lines by Western blot, and in 88 primary gastric carcinomas by immunohistochemistry. Twenty-six of these 88 primary gastric carcinomas were also microdissected and tested for LOH at the FEZ1/LZTS1 locus and for mutation of the FEZ1/LZTS1 gene. Furthermore, we studied the FEZ1/LZTS1 gene regulation and transcriptional control and the methylation status of the 5' region of the gene in all 8 gastric carcinoma cell lines. RESULTS: Fez1/Lzts1 protein was barely detectable in all of the gastric cancer cell lines tested and was absent or significantly reduced in 39 of the 88 (44.3%) gastric carcinomas analyzed by immunohistochemistry, with a significant correlation (P < 0.001) to diffuse histotype. DNA allelotyping analysis showed allelic loss in 3 of 17 (18%) and microsatellite instability in 4 of 17 (23.5%) cases informative for D8S261 at the FEZ1/LZTS1 locus. When we compared the presence of LOH with Fez1/Lzts1 expression, we found loss of protein expression in all three of the tumors with allelic imbalance at D8S261. A missense mutation was detected in one case that did not express Fez1/Lzts1. Hypermethylation of the CpG island flanking the Fez1/Lzts1 promoter was evident in six of the eight cell lines examined as well as in the normal control. CONCLUSIONS: Our findings support FEZ1/LZTS1 as a candidate tumor suppressor gene at 8p in a subtype of gastric cancer and suggest that its inactivation is attributable to several factors including genomic deletion and methylation.


Subject(s)
DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing , Alleles , Base Sequence , Blotting, Southern , Blotting, Western , Brain/metabolism , Cell Nucleus/metabolism , Chromosomes, Human, Pair 8 , CpG Islands , DNA/metabolism , DNA Methylation , DNA Mutational Analysis , DNA, Complementary/metabolism , Gene Deletion , Gene Library , Humans , Immunoblotting , Immunohistochemistry , Loss of Heterozygosity , Molecular Sequence Data , Mutation, Missense , Nerve Tissue Proteins , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Recombinant Proteins/metabolism , Tumor Cells, Cultured
9.
Cancer Res ; 61(12): 4827-36, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11406559

ABSTRACT

The fragile histidine triad (FHIT) gene is a tumor suppressor gene that is altered by deletion in a large fraction of human tumors, including pancreatic cancer. To evaluate the potential of FHIT gene therapy, we developed recombinant adenoviral and adenoassociated viral (AAV) FHIT vectors and tested these vectors in vitro and in vivo for activity against human pancreatic cancer cells. Our data show that viral FHIT gene delivery results in apoptosis by activation of the caspase pathway. Furthermore, Fhit overexpression enhances the susceptibility of pancreatic cancer cells to exogenous inducers of apoptosis. In vivo results show that FHIT gene transfer delays tumor growth and prolongs survival in a murine model mimicking human disease.


Subject(s)
Acid Anhydride Hydrolases , Apoptosis/genetics , Neoplasm Proteins , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proteins/genetics , Adenoviridae/genetics , Animals , Caspases/metabolism , Cell Cycle/physiology , Cell Division/genetics , DNA Fragmentation , Female , Gene Transfer Techniques , Genes, Tumor Suppressor , Genetic Therapy , Genetic Vectors/genetics , Humans , Mice , Mice, Nude , Mitochondria/physiology , Pancreatic Neoplasms/metabolism , Protein Biosynthesis , Signal Transduction/physiology , Transduction, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Cancer Res ; 61(4): 1578-84, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245468

ABSTRACT

Reintroduction of a tumor suppressor gene product in cancer cells is a promising strategy for cancer gene therapy. The fragile histidine triad (FHIT) gene has been identified in a region at chromosome 3p14.2, which is deleted in many tumors, including esophageal cancer. Previous studies have shown frequent biallelic alterations of the FHIT gene in numerous tumors, and have demonstrated a tumor suppressor function of Fhit. We have studied the biological effects of adenoviral-FHIT transduction in esophageal cancer cell lines. Results showed suppression of cell growth in vitro in three of seven esophageal cancer cell lines, all seven of which showed abundant expression of the transgene. Adenoviral-FHIT expression, but not control adenoviral infections, induced caspase-dependent apoptosis in two esophageal cancer cell lines, TE14 and TE4, which express no or very little Fhit, respectively. Treatment of TE14 cells with adenoviral-FHIT vectors resulted in abrogation of tumorigenicity in nude mice. A third esophageal cancer cell line, TE12, without detectable endogenous Fhit, showed accumulation of cells at S to G2-M and a small apoptotic cell fraction after adenoviral-FHIT transduction. Thus, adenoviral-FHIT expression can inhibit the growth of esophageal cancer cells, at least in part through caspase-dependent apoptosis, suggesting that adenoviral-FHIT infection should be explored as a therapeutic strategy.


Subject(s)
Acid Anhydride Hydrolases , Esophageal Neoplasms/genetics , Genetic Therapy/methods , Neoplasm Proteins , Proteins/genetics , Adenoviridae/genetics , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Division/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Genes, Tumor Suppressor , Genetic Vectors/genetics , HeLa Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Biosynthesis , Transduction, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Proc Natl Acad Sci U S A ; 98(6): 3346-51, 2001 Mar 13.
Article in English | MEDLINE | ID: mdl-11248081

ABSTRACT

The tumor suppressor gene FHIT spans a common fragile site and is highly susceptible to environmental carcinogens. FHIT inactivation and loss of expression is found in a large fraction of premaligant and malignant lesions. In this study, we were able to inhibit tumor development by oral gene transfer, using adenoviral or adenoassociated viral vectors expressing the human FHIT gene, in heterozygous Fhit(+/-) knockout mice, that are prone to tumor development after carcinogen exposure. We therefore suggest that FHIT gene therapy could be a novel clinical approach not only in treatment of early stages of cancer, but also in prevention of human cancer.


Subject(s)
Acid Anhydride Hydrolases , Dimethylnitrosamine/analogs & derivatives , Genes, Tumor Suppressor/physiology , Genetic Therapy/methods , Neoplasm Proteins/genetics , Neoplasms, Experimental/prevention & control , Adenoviridae/genetics , Animals , Carcinogens/adverse effects , Dependovirus/genetics , Dimethylnitrosamine/adverse effects , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology
12.
Methods Mol Med ; 53: 81-93, 2001.
Article in English | MEDLINE | ID: mdl-21318789

ABSTRACT

FHIT (fragile histidine triad) is a tumor-suppressor gene located at chromosome band 3p14.2. The genomic locus, which is greater than 1 Mb, contains 10 small exons that make up the 1.1-kb FHIT cDNA. The coding region starts in exon 5 and stops in exon 9, producing a 16.8-kDa cytoplasmic protein. The FHIT locus contains the hereditary renal cell carcinoma (RCC) t(3;8) translocation, and also encompasses the FRA3B common fragile region (for review, 1). Numerous studies have proven that the FHIT gene is inactivated by deletions in both primary tumors and cell lines derived from head and neck, stomach, lung, and kidney cancers (2-6). Since FHIT is inactivated in so many cancers, it is essential to learn its normal function and analyze how the loss of its function contributes to the progression and development of cancer. For example, an early event in the lungs of a smoker is breakage at the FHIT locus, causing a reduced or absent FHIT protein expression in the preneoplastic lesions. Compensation for the functional loss of FHIT via a recombinant, nonfragile FHITgene may prove therapeutically useful (7,8). Our studies have also shown that the FHIT gene is altered or absent in the majority of transitional-cell carcinoma (TCC) cases of the bladder examined (9). Through the utilization of molecular techniques such as those described here, FHIT alterations may be detected in an early stage of cancer, and thus prove to be a useful diagnostic tool to prevent cancer progression.

13.
Tech Urol ; 6(3): 236-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10963500

ABSTRACT

PURPOSE: Insulin-like growth factor 1 (IGF-1) is an important mitogenic and antiapoptotic peptide that affects the proliferation of normal and malignant cells. Contradictory reports on the association between serum IGF-1 level and prostate cancer have been highlighted in the recent literature. The purpose of this study was to investigate the relation between serum levels of IGF-1 and prostate cancer. MATERIALS AND METHODS: We analyzed a population of 57 patients who underwent radical prostatectomy (RP) for adenocarcinoma. Serum samples were collected before RP (T0), 6 months after RP (T6), and from 39 age-matched controls. IGF-1 levels were determined by the active IGF-1 Elisa kit (Diagnostic Systems Laboratories, Inc.). Parallel samples were evaluated for prostate-specific antigen (PSA) levels. Data between groups were analyzed using Welch's t-test and levels before RP and after 6 months were compared by paired t-test. RESULTS: The normal mean serum IGF-1 for case patients at T0 (124.6+/-58.2 ng/mL) was significantly lower than the control subjects (157.5+/-70.8 ng/mL; p = .0192). The normal mean serum IGF-1 for case patients at T0 (124.91+/-58.6 ng/mL) also was significantly lower when it was compared with the T6 group (148.49+/-57.2 ng/mL; p = .0056). No association was found between IGF-1 and PSA blood levels, or IGF-1 and patient weight (p = 0.2434). An inverse relation between IGF-1 levels and age in the normal controls (p = .0041) was observed. CONCLUSION: Findings of this study indicate a significant association between low serum levels of IGF-1 and prostate cancer.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor/analysis , Insulin-Like Growth Factor I/analysis , Prostatic Neoplasms/diagnosis , Adenocarcinoma/blood , Adenocarcinoma/surgery , Aged , Enzyme-Linked Immunosorbent Assay , Humans , Male , Middle Aged , Postoperative Period , Preoperative Care , Prostate-Specific Antigen/analysis , Prostatectomy/methods , Prostatic Neoplasms/blood , Prostatic Neoplasms/surgery , Reference Values , Sensitivity and Specificity
14.
Clin Cancer Res ; 6(4): 1372-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10778965

ABSTRACT

Loss of heterozygosity at several chromosomal loci is a common feature of the malignant progression of human tumors. These regions are thought to harbor one or more putative tumor suppressor gene(s) playing a role in tumor development. Allelic losses on the short arm of chromosome 8 (8p) have been reported as frequent events in several cancers, and three commonly deleted regions have been defined at 8p11.2-12, 8p21-22, and 8p23.1. To evaluate the possible involvement of these regions in gastric cancer, we used eight microsatellite markers to perform an extensive analysis of allele loss at 8p21-22 in 52 cases of primary gastric adenocarcinoma. We found that 44% of tumors showed allelic loss for at least one marker at 8p21-22. The critical region of loss was found to be between markers LPL and D8S258, which displayed loss of heterozygosity in 39% and 33% of cases, respectively. This region is centromeric to the LPL locus and centered on the D8S258 locus. We conclude that 8p22 deletion is a frequent event in gastric cancer and suggest the presence of a putative tumor suppressor gene near the D8S258 locus. Initial steps were taken toward the identification of this gene, which is likely to play an important role in the pathogenesis of gastric cancer and of other tumors as well.


Subject(s)
Chromosomes, Human, Pair 8/genetics , Loss of Heterozygosity , Stomach Neoplasms/genetics , Chromosome Mapping , DNA, Neoplasm/genetics , Genes, Tumor Suppressor , Genetic Markers , Humans , Microsatellite Repeats , Stomach Neoplasms/pathology
15.
Am J Pathol ; 156(2): 419-24, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666370

ABSTRACT

Cytogenetic and loss of heterozygosity (LOH) studies demonstrated chromosome 3p deletions in transitional cell carcinoma (TCC). We recently cloned the tumor suppressor gene FHIT (fragile histidine triad) at 3p14.2, one of the most frequently deleted chromosomal regions in TCC of the bladder, and showed that it is the target of environmental carcinogens. Abnormalities at the FHIT locus have been found in tumors of the lung, breast, cervix, head and neck, stomach, pancreas, and clear cell carcinoma of the kidney. We examined six TCC derived cell lines (SW780, T24, Hs228T, CRL7930, CRL7833, and HTB9) and 30 primary TCC of the bladder for the integrity of the FHIT transcript, using reverse transcriptase-polymerase chain reaction (RT-PCR) to investigate a potential role of the FHIT gene in TCC of the bladder. In addition, we tested expression of the Fhit protein in the six TCC-derived cell lines by Western blot analysis and in 85 specimens of primary TCCs by immunohistochemistry. Three of the six cell lines (50%) did not show the wild-type FHIT transcript, and Fhit protein was not detected in four of the six cell lines (67%) tested. Fhit expression also was correlated with pathological and clinical status. A significant correlation was observed between reduced Fhit expression and advanced stage of the tumors. Overall, 26 of 30 (87%) primary TCCs showed abnormal transcripts. Fhit protein was absent or greatly reduced in 61% of the TCCs analyzed by immunohistochemistry. These results suggested that loss of Fhit expression may be as important in the development of bladder cancer as it is for other neoplasms caused by environmental carcinogens.


Subject(s)
Acid Anhydride Hydrolases , Carcinoma, Transitional Cell/metabolism , Neoplasm Proteins , Proteins/metabolism , Urinary Bladder Neoplasms/metabolism , Blotting, Western , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Female , Gene Deletion , Homozygote , Humans , Immunohistochemistry , Male , Neoplasm Staging , Proteins/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
16.
Cancer ; 88(1): 24-34, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10618602

ABSTRACT

BACKGROUND: The FHIT gene is inactivated by deletion in a large fraction of human tumors, including gastric carcinomas, and the Fhit protein has been proposed to act as a tumor suppressor in multiple tumor types. A large fraction of gastric adenocarcinomas have lost expression of the candidate tumor suppressor protein, Fhit, whereas normal gastric epithelial cells are strongly positive and Fhit loss has been found to correlate with alterations of the FHIT locus. Because the majority of gastric tumors in the current study were found to be entirely negative for Fhit protein, it is possible that alteration of the carcinogen-susceptible fragile region within the FHIT gene is an early event in gastric carcinoma, as it is in lung carcinoma. METHODS: To determine whether the absence of Fhit protein correlates with expression of tumor markers or with clinical parameters, such as grade, stage, and survival time, the authors assessed Fhit expression using immunohistochemistry in a well characterized set of 55 gastric adenocarcinomas resected over several years, with longitudinal follow-up of patients for outcome. RESULTS: In this set of 55 gastric cancers, the absence of Fhit protein correlated with higher tumor stage (P = 0.003) and higher histologic grade (P = 0.007). In addition, patients whose tumors had lost expression of Fhit died of disease significantly earlier than those with Fhit positive tumors (P = 0.017). The absence of Fhit expression did not correlate with the expression of any tumor markers. CONCLUSIONS: Larger studies will be required to elucidate further the relation between tumor stage, grade, and Fhit loss and to determine whether inclusion of Fhit antiserum in immunophenotyping of gastric adenocarcinomas will be a useful indicator of post-diagnosis prognosis.


Subject(s)
Acid Anhydride Hydrolases , Adenocarcinoma/chemistry , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/analysis , Proteins/analysis , Stomach Neoplasms/chemistry , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Biomarkers, Tumor/analysis , Case-Control Studies , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Neoplasm Invasiveness , Neoplasm Staging , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Analysis , Tumor Suppressor Protein p53/analysis
17.
Proc Natl Acad Sci U S A ; 96(7): 3928-33, 1999 Mar 30.
Article in English | MEDLINE | ID: mdl-10097140

ABSTRACT

Alterations of human chromosome 8p occur frequently in many tumors. We identified a 1.5-Mb common region of allelic loss on 8p22 by allelotype analysis. cDNA selection allowed isolation of several genes, including FEZ1. The predicted Fez1 protein contained a leucine-zipper region with similarity to the DNA-binding domain of the cAMP-responsive activating-transcription factor 5. RNA blot analysis revealed that FEZ1 gene expression was undetectable in more than 60% of epithelial tumors. Mutations were found in primary esophageal cancers and in a prostate cancer cell line. Transcript analysis from several FEZ1-expressing tumors revealed truncated mRNAs, including a frameshift. Alteration and inactivation of the FEZ1 gene may play a role in various human tumors.


Subject(s)
Chromosomes, Human, Pair 8 , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Loss of Heterozygosity , Neoplasms/genetics , Transcription, Genetic , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Breast Neoplasms/genetics , Chromosome Mapping , Cloning, Molecular , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/chemistry , Esophageal Neoplasms/genetics , Female , Gene Library , Humans , Leucine Zippers , Male , Molecular Sequence Data , Nerve Tissue Proteins , Ovarian Neoplasms/genetics , Prostatic Neoplasms/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Testis/metabolism , Tumor Cells, Cultured
18.
Cancer Res ; 58(20): 4708-14, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9788626

ABSTRACT

Loss of heterozygosity involving the short arm of chromosome 3 has been reported in gastric and other human tumors. We have cloned and mapped a candidate tumor suppressor gene, FHIT (fragile histidine triad), to this chromosomal region (3p14.2). To investigate the role of FHIT gene alterations in the development of gastric carcinoma, we examined 8 gastric carcinoma-derived cell lines and 32 primary adenocarcinoma samples by Southern blot analysis. We also analyzed the integrity of FHIT transcripts by reverse transcription-PCR. The occurrence of alterations in the FHIT gene and its transcript correlated with the absence of Fhit protein expression by immunoblot analysis in the cancer cell lines. Four of eight cell lines showed deletion or rearrangement within the FHIT gene, together with the absence of the wild-type transcript and the Fhit protein. Among the primary gastric carcinomas, rearrangement of the FHIT gene and/or aberrant reverse transcription-PCR products were detected in 17 of 32 (53%) tumors, and 20 of 30 (67%) samples exhibited an absence of Fhit protein expression. Gastric cancer is thought to develop from carcinogenic exposure, possibly explaining the high frequency of abnormalities in the FHIT gene, a fragile locus exhibiting susceptibility to carcinogen-induced alterations. The consequent absence or reduction of Fhit protein expression is consistent with the proposal that the FHIT gene is a preferential target of environmental carcinogens and that FHIT inactivation plays a role in the development of gastric cancer.


Subject(s)
Acid Anhydride Hydrolases , Genes, Tumor Suppressor , Neoplasm Proteins , Proteins/genetics , Stomach Neoplasms/etiology , Blotting, Southern , Chromosomes, Human, Pair 3 , Humans , Loss of Heterozygosity , Polymerase Chain Reaction , Proteins/analysis , Stomach Neoplasms/genetics , Tumor Cells, Cultured
19.
Mol Pathol ; 51(2): 80-4, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9713590

ABSTRACT

BACKGROUND: Upregulation of the cell cycle associated genes, p16/CDKN2 and the retinoblastoma susceptibility gene (Rb), is commonly seen during the proliferation of normal cells. An inverse relation between the expression of p16/CDKN2 and Rb has been noted in many tumours, but has not yet been determined in oesophageal squamous carcinoma. AIMS: To investigate p16/CDKN2 genetic alterations and both the p16/CDKN2 and the Rb protein (pRb) immunophenotypes in oesophageal squamous carcinoma. METHODS: Twenty primary oesophageal squamous carcinomas were examined for mutations in p16/CDKN2 by the polymerase chain reaction, single stranded conformational polymorphism, and DNA sequencing. Synthesis of p16/CDKN2 and pRb proteins was determined by immunohistochemistry in 19 specimens of formalin fixed, paraffin wax embedded tissues. RESULTS: Mutations of p16/CDKN2 were not detected in exons 1 and 2. In only one case, G to C and C to T base changes were detected in a non-coding region of exon 3. Expression of p16/CDKN2 and Rb was observed in both normal and neoplastic areas of tissue sections, indicating neither consistent homozygous deletion nor consistent hypermethylation of the genes in tumours. Fourteen tumours showed an inverse expression of p16/CDKN2 and Rb. An increased percentage of cells that immunostained positively for p16/CDKN2 but not for pRb was observed in eight tumours, five of which had no detectable pRb, suggesting defective Rb expression in these oesophageal squamous carcinomas. CONCLUSIONS: These results indicate that p16/CDKN2 mutations occur infrequently in oesophageal squamous carcinoma. The alteration of the Rb gene is suggested as an important step in the development of these tumours.


Subject(s)
Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Genes, p16 , Mutation , Retinoblastoma Protein/metabolism , Aged , Carcinoma, Squamous Cell/metabolism , DNA Mutational Analysis , Esophageal Neoplasms/metabolism , Gene Expression , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Proteins/metabolism , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational
20.
Cancer Res ; 57(14): 3010-5, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9230216

ABSTRACT

Spontaneous hydrolytic deamination of 5-methylcytosine leads to T:G mismatches in double-stranded DNA and comprises a major threat for the integrity of both the DNA primary sequence as well as the epigenetic information stored in the DNA methylation pattern. Failure of the cellular DNA repair machinery to recognize and repair such mismatched nucleotides can lead to a mutator phenotype and subsequent carcinogenesis. A thymine-DNA glycosylase (TDG) has been described that initiates T:G mismatch repair by specifically excising the mismatched T. We have studied the TDG genomic locus and the expression of this enzyme to evaluate its role in cancer development. TDG is highly expressed in thymus and is expressed at lower levels in all human tissues analyzed. The TDG gene has 10 exons covering a region of >25 kb and is located on chromosome 12q22-q24.1. Because gastric tumors have been shown to contain a high percentage of C-->T mutations at CpG sites, we used a microsatellite found in intron 8 of the TDG locus to screen gastric tumor samples for loss of heterozygosity. Although our analysis showed loss of heterozygosity in 10 of 24 samples (42%), none of those tumor samples revealed a mutation in the coding sequence of the remaining TDG allele as analyzed by single-strand conformational polymorphism. Expression of the TDG was not determined because of the limited availability of RNA in these primary tumor samples. At present, we have found no evidence that TDG is central to the development of gastric cancer, limiting the importance of TDG in T:G mismatch repair and subsequent carcinogenesis.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 12 , Endodeoxyribonucleases/genetics , Stomach Neoplasms/genetics , Base Sequence , DNA Repair , Deoxyribonuclease (Pyrimidine Dimer) , Humans , Molecular Sequence Data , Polymorphism, Single-Stranded Conformational
SELECTION OF CITATIONS
SEARCH DETAIL