Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cancer Res ; 76(8): 2177-85, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26862115

ABSTRACT

VHL-deficient clear cell renal cell carcinomas (ccRCC), the most common form of kidney cancer, express transcripts derived from the novel human endogenous retrovirus HERV-E (named CT-RCC HERV-E). In this study, we define a transcript encoding the entire envelope gene of HERV-E as expressed selectively in ccRCC tumors, as distinct from normal kidney tissues or other tumor types. Sequence analysis of this envelope transcript revealed long open reading frames encoding putative surface and transmembrane envelope proteins. Retroviral envelopes are known to be capable of eliciting immunity in humans. Accordingly, we found that HLA-A*0201-restricted peptides predicted to be products of the CT-RCC HERV-E envelope transcript-stimulated CD8(+) T cells, which could recognize HLA-A*0201-positive HERV-E-expressing kidney tumor cells. Overall, our results offer evidence of unique HERV-E envelope peptides presented on the surface of ccRCC cells, offering potentially useful tumor-restricted targets for T-cell-based immunotherapy of kidney cancer. Cancer Res; 76(8); 2177-85. ©2016 AACR.


Subject(s)
Carcinoma, Renal Cell/virology , Endogenous Retroviruses/isolation & purification , Kidney Neoplasms/virology , Viral Envelope Proteins/metabolism , Amino Acid Sequence , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Renal Cell/immunology , Cell Line, Tumor , Endogenous Retroviruses/genetics , Enzyme-Linked Immunosorbent Assay , Genes, Viral , Humans , Kidney Neoplasms/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
2.
PLoS One ; 10(5): e0123369, 2015.
Article in English | MEDLINE | ID: mdl-25961819

ABSTRACT

The SEMA3B gene is located in the 3p21.3 LUCA region, which is frequently affected in different types of cancer. The objective of our study was to expand our knowledge of the SEMA3B gene as a tumor suppressor and the mechanisms of its inactivation. In this study, several experimental approaches were used: tumor growth analyses and apoptosis assays in vitro and in SCID mice, expression and methylation assays and other. With the use of the small cell lung cancer cell line U2020 we confirmed the function of SEMA3B as a tumor suppressor, and showed that the suppression can be realized through the induction of apoptosis and, possibly, associated with the inhibition of angiogenesis. In addition, for the first time, high methylation frequencies have been observed in both intronic (32-39%) and promoter (44-52%) CpG-islands in 38 non-small cell lung carcinomas, including 16 squamous cell carcinomas (SCC) and 22 adenocarcinomas (ADC), and in 83 clear cell renal cell carcinomas (ccRCC). Correlations between the methylation frequencies of the promoter and the intronic CpG-islands of SEMA3B with tumor stage and grade have been revealed for SCC, ADC and ccRCC. The association between the decrease of the SEMA3B mRNA level and hypermethylation of the promoter and the intronic CpG-islands has been estimated in renal primary tumors (P < 0.01). Using qPCR, we observed on the average 10- and 14-fold decrease of the SEMA3B mRNA level in SCC and ADC, respectively, and a 4-fold decrease in ccRCC. The frequency of this effect was high in both lung (92-95%) and renal (84%) tumor samples. Moreover, we showed a clear difference (P < 0.05) of the SEMA3B relative mRNA levels in ADC with and without lymph node metastases. We conclude that aberrant expression and methylation of SEMA3B could be suggested as markers of lung and renal cancer progression.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , Lung Neoplasms/genetics , Membrane Glycoproteins/genetics , Neoplasms, Squamous Cell/genetics , Semaphorins/genetics , Small Cell Lung Carcinoma/genetics , Animals , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , CpG Islands , DNA Methylation , Humans , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/pathology , Lung/metabolism , Lung/pathology , Lung Neoplasms/pathology , Mice, SCID , Neoplasms, Squamous Cell/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Promoter Regions, Genetic , Small Cell Lung Carcinoma/pathology
3.
Epigenetics ; 8(4): 409-20, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23478628

ABSTRACT

Genetic and epigenetic alterations in cervical carcinomas were investigated using NotI-microarrays containing 180 cloned sequences flanking all NotI-sites associated with genes on chromosome 3. In total, 48 paired normal/tumor DNA samples, specifically enriched in NotI-sites, were hybridized to NotI-microarrays. Thirty genes, including tumor suppressors or candidates (for example, VHL, RBSP3/CTDSPL, ITGA9, LRRC3B, ALDH1L1, EPHB1) and genes previously unknown as cancer-associated (ABHD5, C3orf77, PRL32, LOC285375, FGD5 and others), showed methylation/deletion in 21-44% of tumors. The genes were more frequently altered in squamous cell carcinomas (SCC) than in adenocarcinomas (ADC, p<0.01). A set of seven potential markers (LRRN1, PRICKLE2, VHL, BHLHE40, RBSP3, CGGBP1 and SOX14) is promising for discrimination of ADC and SCC. Alterations of more than 20 genes simultaneously were revealed in 23% of SCC. Bisulfite sequencing analysis confirmed methylation as a frequent event in SCC. High down-regulation frequency was shown for RBSP3, ITGA9, VILL, APRG1/C3orf35 and RASSF1 (isoform A) genes (3p21.3 locus) in SCC. Both frequency and extent of RASSF1A and RBSP3 mRNA level decrease were more pronounced in tumors with lymph node metastases compared with non-metastatic ones (p ≤ 0.05). We confirmed by bisulfite sequencing that RASSF1 promoter methylation was a rare event in SCC and, for the first time, demonstrated RASSF1A down-regulation at both the mRNA and protein levels without promoter methylation in tumors of this histological type. Thus, our data revealed novel tumor suppressor candidates located on chromosome 3 and a frequent loss of epigenetic stability of 3p21.3 locus in combination with down-regulation of genes in cervical cancer.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Squamous Cell/genetics , Chromosomes, Human, Pair 3/genetics , Deoxyribonucleases, Type II Site-Specific/chemistry , Genes, Tumor Suppressor , Uterine Cervical Neoplasms/genetics , Base Sequence , Biomarkers, Tumor/genetics , DNA Methylation , Down-Regulation , Epigenesis, Genetic , Female , Gene Expression Profiling , Humans , Mutation Rate , Oligonucleotide Array Sequence Analysis , Sequence Deletion , Tumor Suppressor Proteins/metabolism
4.
Epigenetics ; 7(5): 502-13, 2012 May.
Article in English | MEDLINE | ID: mdl-22491060

ABSTRACT

This study aimed to clarify genetic and epigenetic alterations that occur during lung carcinogenesis and to design perspective sets of newly identified biomarkers. The original method includes chromosome 3 specific NotI-microarrays containing 180 NotI clones associated with genes for hybridization with 40 paired normal/tumor DNA samples of primary lung tumors: 28 squamous cell carcinomas (SCC) and 12 adenocarcinomas (ADC). The NotI-microarray data were confirmed by qPCR and bisulfite sequencing analyses. Forty-four genes showed methylation and/or deletions in more than 15% of non-small cell lung cancer (NSCLC) samples. In general, SCC samples were more frequently methylated/deleted than ADC. Moreover, the SCC alterations were observed already at stage I of tumor development, whereas in ADC many genes showed tumor progression specific methylation/deletions. Among genes frequently methylated/deleted in NSCLC, only a few were already known tumor suppressor genes: RBSP3 (CTDSPL), VHL and THRB. The RPL32, LOC285205, FGD5 and other genes were previously not shown to be involved in lung carcinogenesis. Ten methylated genes, i.e., IQSEC1, RBSP3, ITGA 9, FOXP1, LRRN1, GNAI2, VHL, FGD5, ALDH1L1 and BCL6 were tested for expression by qPCR and were found downregulated in the majority of cases. Three genes (RBSP3, FBLN2 and ITGA9) demonstrated strong cell growth inhibition activity. A comprehensive statistical analysis suggested the set of 19 gene markers, ANKRD28, BHLHE40, CGGBP1, RBSP3, EPHB1, FGD5, FOXP1, GORASP1/TTC21, IQSEC1, ITGA9, LOC285375, LRRC3B, LRRN1, MITF, NKIRAS1/RPL15, TRH, UBE2E2, VHL, WNT7A, to allow early detection, tumor progression, metastases and to discriminate between SCC and ADC with sensitivity and specificity of 80-100%.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genetic Testing/methods , Oligonucleotide Array Sequence Analysis/methods , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Case-Control Studies , Cell Line, Tumor , Chromosomes, Human, Pair 3/genetics , Chromosomes, Human, Pair 3/metabolism , DNA Methylation , Disease Progression , Female , Gene Deletion , Genes, Neoplasm , Guanine Nucleotide Exchange Factors , Humans , Male , Middle Aged , Sensitivity and Specificity , Transfection , Tumor Suppressor Proteins , Von Hippel-Lindau Tumor Suppressor Protein
5.
Biochimie ; 94(5): 1151-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22321817

ABSTRACT

Chromosome 3 specific NotI microarrays containing 180 NotI linking clones associated with 188 genes were hybridized to NotI representation probes prepared using matched tumor/normal samples from major epithelial cancers: breast (47 pairs), lung (40 pairs) cervical (43 pairs), kidney (34 pairs of clear cell renal cell carcinoma), colon (24 pairs), ovarian (25 pairs) and prostate (18 pairs). In all tested primary tumors (compared to normal controls) methylation and/or deletions was found. For the first time we showed that the gene LRRC3B was frequently methylated and/or deleted in breast carcinoma - 32% of samples, cervical - 35%, lung - 40%, renal - 35%, ovarian - 28%, colon - 33% and prostate cancer - 44%. To check these results bisulfite sequencing using cloned PCR products with representative two breast, one cervical, two renal, two ovarian and two colon cancer samples was performed. In all cases methylation was confirmed. Expression analysis using RT-qPCR showed that LRRC3B is strongly down-regulated at the latest stages of RCC and ovarian cancers. In addition we showed that LRRC3B exhibit strong cell growth inhibiting activity (more than 95%) in colony formation experiments in vitro in KRC/Y renal cell carcinoma line. All these data suggest that LRRC3B gene could be involved in the process of carcinogenesis as a tumor suppressor gene.


Subject(s)
Epigenesis, Genetic/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Base Sequence , Cell Line, Tumor , DNA Methylation/genetics , Female , Genes, Tumor Suppressor/physiology , Humans , In Vitro Techniques , Male , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction
6.
PLoS One ; 6(3): e15612, 2011 Mar 07.
Article in English | MEDLINE | ID: mdl-21408220

ABSTRACT

BACKGROUND: CHL1 gene (also known as CALL) on 3p26.3 encodes a one-pass trans-membrane cell adhesion molecule (CAM). Previously CAMs of this type, including L1, were shown to be involved in cancer growth and metastasis. METHODOLOGY/PRINCIPAL FINDINGS: We used Clontech Cancer Profiling Arrays (19 different types of cancers, 395 samples) to analyze expression of the CHL1 gene. The results were further validated by RT-qPCR for breast, renal and lung cancer. Cancer Profiling Arrays revealed differential expression of the gene: down-regulation/silencing in a majority of primary tumors and up-regulation associated with invasive/metastatic growth. Frequent down-regulation (>40% of cases) was detected in 11 types of cancer (breast, kidney, rectum, colon, thyroid, stomach, skin, small intestine, bladder, vulva and pancreatic cancer) and frequent up-regulation (>40% of cases)--in 5 types (lung, ovary, uterus, liver and trachea) of cancer. Using real-time quantitative PCR (RT-qPCR) we found that CHL1 expression was decreased in 61% of breast, 60% of lung, 87% of clear cell and 89% papillary renal cancer specimens (P<0.03 for all the cases). There was a higher frequency of CHL1 mRNA decrease in lung squamous cell carcinoma compared to adenocarcinoma (81% vs. 38%, P = 0.02) without association with tumor progression. CONCLUSIONS/SIGNIFICANCE: Our results suggested that CHL1 is involved in the development of different human cancers. Initially, during the primary tumor growth CHL1 could act as a putative tumor suppressor and is silenced to facilitate in situ tumor growth for 11 cancer types. We also suggested that re-expression of the gene on the edge of tumor mass might promote local invasive growth and enable further metastatic spread in ovary, colon and breast cancer. Our data also supported the role of CHL1 as a potentially novel specific biomarker in the early pathogenesis of two major histological types of renal cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Neoplasms/genetics , Precancerous Conditions/genetics , Cell Adhesion Molecules , Cell Line, Tumor , Computational Biology , Gene Expression Profiling , Humans , Membrane Proteins/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
BMC Cancer ; 10: 75, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20193080

ABSTRACT

BACKGROUND: The short arm of human chromosome 3 is involved in the development of many cancers including lung cancer. Three bona fide lung cancer tumor suppressor genes namely RBSP3 (AP20 region),NPRL2 and RASSF1A (LUCA region) were identified in the 3p21.3 region. We have shown previously that homozygous deletions in AP20 and LUCA sub-regions often occurred in the same tumor (P < 10-6). METHODS: We estimated the quantity of RBSP3, NPRL2, RASSF1A, GAPDH, RPN1 mRNA and RBSP3 DNA copy number in 59 primary non-small cell lung cancers, including 41 squamous cell and 18 adenocarcinomas by real-time reverse transcription-polymerase chain reaction based on TaqMan technology and relative quantification. RESULTS: We evaluated the relationship between mRNA level and clinicopathologic characteristics in non-small cell lung cancer. A significant expression decrease (> or =2) was found for all three genes early in tumor development: in 85% of cases for RBSP3; 73% for NPRL2 and 67% for RASSF1A (P < 0.001), more strongly pronounced in squamous cell than in adenocarcinomas. Strong suppression of both, NPRL2 and RBSP3 was seen in 100% of cases already at Stage I of squamous cell carcinomas. Deregulation of RASSF1A correlated with tumor progression of squamous cell (P = 0.196) and adenocarcinomas (P < 0.05). Most likely, genetic and epigenetic mechanisms might be responsible for transcriptional inactivation of RBSP3 in non-small cell lung cancers as promoter methylation of RBSP3 according to NotI microarrays data was detected in 80% of squamous cell and in 38% of adenocarcinomas. With NotI microarrays we tested how often LUCA (NPRL2, RASSF1A) and AP20 (RBSP3) regions were deleted or methylated in the same tumor sample and found that this occured in 39% of all studied samples (P < 0.05). CONCLUSION: Our data support the hypothesis that these TSG are involved in tumorigenesis of NSCLC. Both genetic and epigenetic mechanisms contribute to down-regulation of these three genes representing two tumor suppressor clusters in 3p21.3. Most importantly expression of RBSP3, NPRL2 and RASSF1A was simultaneously decreased in the same sample of primary NSCLC: in 39% of cases all these three genes showed reduced expression (P < 0.05).


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Lung Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Adult , Aged , Epigenesis, Genetic , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic
8.
PLoS One ; 4(5): e5231, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19478941

ABSTRACT

BACKGROUND: Many different genetic alterations are observed in cancer cells. Individual cancer genes display point mutations such as base changes, insertions and deletions that initiate and promote cancer growth and spread. Somatic hypermutation is a powerful mechanism for generation of different mutations. It was shown previously that somatic hypermutability of proto-oncogenes can induce development of lymphomas. METHODOLOGY/PRINCIPAL FINDINGS: We found an exceptionally high incidence of single-base mutations in the tumor suppressor genes RASSF1 and RBSP3 (CTDSPL) both located in 3p21.3 regions, LUCA and AP20 respectively. These regions contain clusters of tumor suppressor genes involved in multiple cancer types such as lung, kidney, breast, cervical, head and neck, nasopharyngeal, prostate and other carcinomas. Altogether in 144 sequenced RASSF1A clones (exons 1-2), 129 mutations were detected (mutation frequency, MF = 0.23 per 100 bp) and in 98 clones of exons 3-5 we found 146 mutations (MF = 0.29). In 85 sequenced RBSP3 clones, 89 mutations were found (MF = 0.10). The mutations were not cytidine-specific, as would be expected from alterations generated by AID/APOBEC family enzymes, and appeared de novo during cell proliferation. They diminished the ability of corresponding transgenes to suppress cell and tumor growth implying a loss of function. These high levels of somatic mutations were found both in cancer biopsies and cancer cell lines. CONCLUSIONS/SIGNIFICANCE: This is the first report of high frequencies of somatic mutations in RASSF1 and RBSP3 in different cancers suggesting it may underlay the mutator phenotype of cancer. Somatic hypermutations in tumor suppressor genes involved in major human malignancies offer a novel insight in cancer development, progression and spread.


Subject(s)
Mutation/genetics , Neoplasms/genetics , Tumor Suppressor Proteins/genetics , APOBEC-1 Deaminase , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation , Clone Cells , Computational Biology , Cytidine Deaminase/metabolism , DNA, Bacterial/genetics , DNA, Complementary/genetics , Escherichia coli Proteins/genetics , Expressed Sequence Tags , Founder Effect , Genome/genetics , Hematopoiesis/genetics , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Mice, SCID , Polymerase Chain Reaction
9.
BMC Dev Biol ; 9: 22, 2009 Mar 16.
Article in English | MEDLINE | ID: mdl-19291313

ABSTRACT

BACKGROUND: Transmembrane CAIX and CAXII are members of the alpha carbonic anhydrase (CA) family. They play a crucial role in differentiation, proliferation, and pH regulation. Expression of CAIX and CAXII proteins in tumor tissues is primarily induced by hypoxia and this is particularly true for CAIX, which is regulated by the transcription factor, hypoxia inducible factor-1 (HIF-1). Their distributions in normal adult human tissues are restricted to highly specialized cells that are not always hypoxic. The human fetus exists in a relatively hypoxic environment. We examined expression of CAIX, CAXII and HIF-1alpha in the developing human fetus and postnatal tissues to determine whether expression of CAIX and CAXII is exclusively regulated by HIF-1. RESULTS: The co-localization of CAIX and HIF-1alpha was limited to certain cell types in embryonic and early fetal tissues. Those cells comprised the primitive mesenchyma or involved chondrogenesis and skin development. Transient CAIX expression was limited to immature tissues of mesodermal origin and the skin and ependymal cells. The only tissues that persistently expressed CAIX protein were coelomic epithelium (mesothelium) and its remnants, the epithelium of the stomach and biliary tree, glands and crypt cells of duodenum and small intestine, and the cells located at those sites previously identified as harboring adult stem cells in, for example, the skin and large intestine. In many instances co-localization of CAIX and HIF-1alpha was not evident. CAXII expression is restricted to cells involved in secretion and water absorption such as parietal cells of the stomach, acinar cells of the salivary glands and pancreas, epithelium of the large intestine, and renal tubules. Co-localization of CAXII with CAIX or HIF-1alpha was not observed. CONCLUSION: The study has showed that: 1) HIF-1alpha and CAIX expression co- localized in many, but not all, of the embryonic and early fetal tissues; 2) There is no evidence of co-localization of CAIX and CAXII; 3) CAIX and CAXII expression is closely related to cell origin and secretory activity involving proton transport, respectively. The intriguing finding of rare CAIX-expressing cells in those sites corresponding to stem cell niches requires further investigation.


Subject(s)
Carbonic Anhydrases/genetics , Embryonic Development , Gene Expression Regulation, Developmental , Child , Child, Preschool , Fetus/metabolism , Humans , Hypoxia , Infant , Infant, Newborn
10.
PLoS One ; 3(8): e3031, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18725949

ABSTRACT

BACKGROUND: We identified two 3p21.3 regions (LUCA and AP20) as most frequently affected in lung, breast and other carcinomas and reported their fine physical and gene maps. It is becoming increasingly clear that each of these two regions contains several TSGs. Until now TSGs which were isolated from AP20 and LUCA regions (e.g.G21/NPRL2, RASSF1A, RASSF1C, SEMA3B, SEMA3F, RBSP3) were shown to inhibit tumour cell growth both in vitro and in vivo. METHODOLOGY/PRINCIPAL FINDINGS: The effect of expression HYAL1 and HYAL2 was studied by colony formation inhibition, growth curve and cell proliferation tests in vitro and tumour growth assay in vivo. Very modest growth inhibition was detected in vitro in U2020 lung and KRC/Y renal carcinoma cell lines. In the in vivo experiment stably transfected KRC/Y cells expressing HYAL1 or HYAL2 were inoculated into SCID mice (10 and 12 mice respectively). Tumours grew in eight mice inoculated with HYAL1. Ectopic HYAL1 was deleted in all of them. HYAL2 was inoculated into 12 mice and only four tumours were obtained. In 3 of them the gene was deleted. In one tumour it was present but not expressed. As expected for tumour suppressor genes HYAL1 and HYAL2 were down-expressed in 15 fresh lung squamous cell carcinomas (100%) and clear cell RCC tumours (60-67%). CONCLUSIONS/SIGNIFICANCE: The results suggest that the expression of either gene has led to inhibition of tumour growth in vivo without noticeable effect on growth in vitro. HYAL1 and HYAL2 thus differ in this aspect from other tumour suppressors like P53 or RASSF1A that inhibit growth both in vitro and in vivo. Targeting the microenvironment of cancer cells is one of the most promising venues of cancer therapeutics. As major hyaluronidases in human cells, HYAL1 and HYAL2 may control intercellular interactions and microenvironment of tumour cells providing excellent targets for cancer treatment.


Subject(s)
Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/therapeutic use , Kidney Neoplasms/pathology , Lung Neoplasms/pathology , Animals , Cell Adhesion Molecules/therapeutic use , Cell Division/drug effects , Cell Line, Tumor , Colony-Forming Units Assay/methods , GPI-Linked Proteins , Humans , Hyaluronoglucosaminidase/deficiency , Mice , Mice, Knockout , Mice, SCID , Transfection
11.
Biochem Biophys Res Commun ; 370(4): 536-40, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18359287

ABSTRACT

Mutations in the VHL gene are associated with highly vascular tumors of kidney, brain, retina, and adrenal gland. The inability of the mutant VHL protein to destabilize HIF-1 plays a crucial role in malignant angiogenesis. VHL is also associated with ECM assembly but the molecular mechanisms of this activity remain unclear. We used expression arrays and cell lines with different VHL status to identify ECM-associated genes controlled by VHL. One of them, adhesion-associated TGFBI, was repressed by VHL and overexpressed in renal, gastrointestinal, brain, and other tumors. Analyzing the mechanism of TGFBI up-regulation in clear cell carcinoma, we identified a novel VHL target, a Kruppel-like transcriptional factor 10 (KLF10). The TGFBI promoter, which we isolated and studied in Luc-reporter assay, was induced by KLF10 but not hypoxia. These data provide the molecular basis for the observed VHL effect on TGFBI and stimulate further research into the KLF10 and TGFBI roles in cancer.


Subject(s)
Early Growth Response Transcription Factors/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/genetics , Neoplasms/genetics , Transforming Growth Factor beta/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/genetics , Cell Adhesion/genetics , Humans , Kidney Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Transcription, Genetic , Transforming Growth Factor beta/metabolism , Up-Regulation
12.
J Clin Invest ; 118(3): 1099-109, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18292810

ABSTRACT

Transplanted donor lymphocytes infused during hematopoietic stem cell transplantation (HSCT) have been shown to cure patients with hematological malignancies. However, less is known about the effects of HSCT on metastatic solid tumors. Thus, a better understanding of the immune cells and their target antigens that mediate tumor regression is urgently needed to develop more effective HSCT approaches for solid tumors. Here we report regression of metastatic renal cell carcinoma (RCC) in patients following nonmyeloablative HSCT consistent with a graft-versus-tumor effect. We detected RCC-reactive donor-derived CD8(+) T cells in the blood of patients following nonmyeloablative HSCT. Using cDNA expression cloning, we identified a 10-mer peptide (CT-RCC-1) as a target antigen of RCC-specific CD8(+) T cells. The genes encoding this antigen were found to be derived from human endogenous retrovirus (HERV) type E and were expressed in RCC cell lines and fresh RCC tissue but not in normal kidney or other tissues. We believe this to be the first solid tumor antigen identified using allogeneic T cells from a patient undergoing HSCT. These data suggest that HERV-E is activated in RCC and that it encodes an overexpressed immunogenic antigen, therefore providing a potential target for cellular immunity.


Subject(s)
Antigens, Viral/immunology , Carcinoma, Renal Cell/therapy , Endogenous Retroviruses/immunology , Hematopoietic Stem Cell Transplantation , Kidney Neoplasms/therapy , T-Lymphocytes/immunology , Adult , Amino Acid Sequence , Base Sequence , Carcinoma, Renal Cell/immunology , Cell Line, Tumor , Humans , Kidney Neoplasms/immunology , Male , Middle Aged , Molecular Sequence Data , T-Lymphocytes, Cytotoxic/physiology , Transplantation, Homologous
13.
Cancer Lett ; 257(2): 199-205, 2007 Nov 18.
Article in English | MEDLINE | ID: mdl-17720308

ABSTRACT

Reduced expression or loss of tumor suppressor genes play a key role in many cancers. In this study, we investigated the role of RASSF1A in the pathogenesis of esophageal squamous cell carcinoma (ESCC) and nasopharyngeal carcinoma (NPC). We detected the down-regulated expression of both RASSF1A transcripts and protein in tumor tissues using RT-PCR and tissue microarray immunohistochemical staining analyses. Down-regulated expression of RASSF1A showed a significant association with WHO grade, tumor status, and lymph node metastasis, showing its possible utility as a biomarker for clinical specimens.


Subject(s)
Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Nasopharyngeal Neoplasms/pathology , Tumor Suppressor Proteins/genetics , Adult , Aged , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Neoplasm Staging , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Tumor Suppressor Proteins/metabolism
14.
Int J Cancer ; 119(12): 2821-6, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16929489

ABSTRACT

Chromosome 3p plays an important role in tumorigenesis in many cancers, including nasopharyngeal carcinoma (NPC). We have previously shown chromosome 3p can suppress tumor growth in vivo by using the monochromosome transfer approach, which indicated the chromosome 3p21.3 region was critical for tumor suppression. BLU/ZMYND10 is one of the candidate tumor suppressor genes mapping in the 3p21.3 critical region and is a candidate TSG for NPC. By quantitative RT-PCR, it is frequently downregulated in NPC cell lines (83%) and NPC biopsies (80%). However, no functional studies have yet verified the functional role of BLU/ZMYND10 as a tumor suppressor gene. In the current study, a gene inactivation test (GIT) utilizing a tetracycline regulation system was used to study the functional role of BLU/ZMYND10. When BLU/ZMYND10 is expressed in the absence of doxycycline, the stable transfectants were able to induce tumor suppression in nude mice. In contrast, downregulation of BLU/ZMYND10 in these tumor suppressive clones by doxycycline treatment restored the tumor formation ability. This study provides the first significant evidence to demonstrate BLU/ZMYND10 can functionally suppress tumor formation in vivo and is, therefore, likely to be one of the candidate tumor suppressor genes involved in NPC.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Nasopharyngeal Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Line, Tumor , Cytoskeletal Proteins , Down-Regulation/drug effects , Down-Regulation/genetics , Doxycycline/pharmacology , Doxycycline/therapeutic use , Female , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor , Genetic Predisposition to Disease/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Transfection , Xenograft Model Antitumor Assays/methods
15.
Genes Chromosomes Cancer ; 45(7): 676-91, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16607615

ABSTRACT

Homozygous deletions or loss of heterozygosity (LOH) at human chromosome band 3p12 are consistent features of lung and other malignancies, suggesting the presence of a tumor suppressor gene(s) (TSG) at this location. Only one gene has been cloned thus far from the overlapping region deleted in lung and breast cancer cell lines U2020, NCI H2198, and HCC38. It is DUTT1 (Deleted in U Twenty Twenty), also known as ROBO1, FLJ21882, and SAX3, according to HUGO. DUTT1, the human ortholog of the fly gene ROBO, has homology with NCAM proteins. Extensive analyses of DUTT1 in lung cancer have not revealed any mutations, suggesting that another gene(s) at this location could be of importance in lung cancer initiation and progression. Here, we report the discovery of a new, small, homozygous deletion in the small cell lung cancer (SCLC) cell line GLC20, nested in the overlapping, critical region. The deletion was delineated using several polymorphic markers and three overlapping P1 phage clones. Fiber-FISH experiments revealed the deletion was approximately 130 kb. Comparative genomic sequence analysis uncovered short sequence elements highly conserved among mammalian genomes and the chicken genome. The discovery of two EST clusters within the deleted region led to the isolation of two noncoding RNA (ncRNA) genes. These were subsequently found differentially expressed in various tumors when compared to their normal tissues. The ncRNA and other highly conserved sequence elements in the deleted region may represent miRNA targets of importance in cancer initiation or progression.


Subject(s)
Chromosomes, Human, Pair 3 , Gene Deletion , Genes, Tumor Suppressor , RNA, Neoplasm/genetics , RNA, Untranslated/genetics , Animals , Breast Neoplasms/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Small Cell/genetics , Cell Line, Tumor , Chickens , Chromosome Mapping , Conserved Sequence , Homozygote , Humans , Introns , Kidney Neoplasms/genetics , Lung Neoplasms/genetics , Multigene Family , Nerve Tissue Proteins/genetics , Receptors, Immunologic/genetics , Roundabout Proteins
16.
Cancer Lett ; 234(2): 184-92, 2006 Mar 28.
Article in English | MEDLINE | ID: mdl-15885884

ABSTRACT

The expression of six chromosome 3p21.3 candidate tumor suppressor genes (BLU, FUS2, HYAL2, NPRL2, RASSF1A, and SEMA3B) in esophageal squamous cell carcinoma (ESCC) has been investigated. Reduced expression of BLU was detected in some ESCC cell lines and tumor tissues and the difference was quantitated by real-time quantitative polymerase chain reaction. Methylation specific-PCR revealed the down-regulation of BLU by epigenetic inactivation. However, exogenous expression of BLU did not functionally suppress tumorigenicity in nude mice. These results suggest that over-expression of BLU alone is not sufficient to inhibit tumorigenicity. Further studies on BLU interacting proteins are required to elucidate the possible role of BLU in the development of ESCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Chromosomes, Human, Pair 3 , Esophageal Neoplasms/genetics , Genes, Tumor Suppressor , Animals , Cell Line, Tumor , Cytoskeletal Proteins , DNA Methylation , Down-Regulation , Gene Silencing , Genes, Tumor Suppressor/physiology , Humans , Mice , Mice, Nude , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins
17.
Retrovirology ; 2: 59, 2005 Sep 28.
Article in English | MEDLINE | ID: mdl-16191204

ABSTRACT

BACKGROUND: Infection by jaagsiekte sheep retrovirus (JSRV) and by enzootic nasal tumor virus (ENTV) depends on cell-surface expression of the virus entry receptor, hyaluronidase 2 (Hyal2). Human Hyal2 binds the envelope (Env) proteins of these viruses and is functional as a receptor, but Hyal2 from mice does not bind Env nor does it mediate entry of either virus. Here we have explored the amino acid determinants that account for the difference in receptor function. RESULTS: Analysis of human-mouse Hyal2 chimeric proteins showed that amino acid differences responsible for the difference in Hyal2 receptor activity were localized to the central third of Hyal2. Human Hyal2 mutants containing single or double amino acid replacements with the respective mouse amino acids were generated across this region and were assayed for activity. None of the single or double mutation reduced the receptor activity of human Hyal2 by more than 10-fold, whereas mouse Hyal2 activity is reduced 1,000-fold from that of human Hyal2. While the 3-dimensional structures of mammalian Hyal2 proteins are unknown, bee venom hyaluronidase shows significant amino acid similarity to human and mouse Hyal2 and its structure has been determined. Many mutations having the largest negative effects on human Hyal2 function mapped to a small region of the bee venom hyaluronidase close to but not overlapping the active site of the enzyme, suggesting that this site represents the binding site for Env. Analysis of synonymous and non-synonymous nucleotide substitutions in the coding sequences of multiple mammalian Hyal2 proteins shows that the proteins are undergoing strong selection for amino acid conservation. We found no evidence for positive selection of amino acid changes that might reflect evolution of mammalian hosts to resist JSRV or ENTV infection. CONCLUSION: These results show that the greatly reduced receptor activity of mouse Hyal2 in comparison to that of human Hyal2 is determined by multiple amino acid changes acting in concert. In particular, no one amino acid change blocks infection. However, the most important amino acids map to a small patch on a predicted 3-dimensional Hyal2 structure, which may represent the binding site for Env.


Subject(s)
Cell Adhesion Molecules/chemistry , Hyaluronoglucosaminidase/chemistry , Jaagsiekte sheep retrovirus/physiology , Receptors, Virus/chemistry , Animals , Bee Venoms/enzymology , Cell Adhesion Molecules/physiology , Conserved Sequence , Crystallization , GPI-Linked Proteins , Gene Products, env/chemistry , Humans , Hyaluronoglucosaminidase/physiology , Mice , NIH 3T3 Cells , Rats , Rats, Inbred F344 , Receptors, Virus/physiology , Structure-Activity Relationship
18.
J Mol Biol ; 346(2): 411-22, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15670593

ABSTRACT

Immune responses to invading pathogens are mediated largely through a family of transmembrane Toll-like receptors and modulated by a number of downstream effectors. In particular, a family of four interleukin 1 receptor-associated kinases (IRAK) regulates responsiveness to bacterial endotoxins. Pharmacological targeting of particular IRAK components may be beneficial for treatment of bacterial infections. Here, we studied transcriptional regulation of the human IRAK2 gene. Analysis of the IRAK2 promoter region reveals putative binding sites for several transcriptional factors, including ZIP (EGR1 and SP1), CTCF and AP-2beta. Deletion of the ZIP or AP-2 sites did not significantly affect IRAK2 promoter activity in naive and endotoxin-treated mononuclear cells, in dormant and activated Jurkat T-cells, in lung and kidney cells. In contrast, we found that CTCF plays a major role in IRAK2 transcription. An electrophoretic mobility shift assay of the DNA fragments containing the IRAK2 CpG island, revealed a single high-affinity binding site for the transcriptional regulator and a chromatin insulator protein, CTCF. This assay revealed a CTCF-binding site within the mouse Irak2 promoter. The presence of the CTCF protein in human IRAK2 promoter was confirmed by chromatin immunoprecipitation assay. Specific residues that interacted with the CTCF protein, were identified by methylation interference assay. In all cell lines analyzed, including cells of lung, renal, monocytic and T-cell origin, the IRAK2 luciferase reporter construct, containing an intact CTCF-binding site, showed strong promoter activity. However, IRAK2 promoter activity was decreased dramatically for the constructs with a mutated CTCF-binding site.


Subject(s)
DNA-Binding Proteins/genetics , Promoter Regions, Genetic , Protein Kinases/genetics , Repressor Proteins/genetics , Animals , Base Sequence , Binding Sites , CCCTC-Binding Factor , Cells, Cultured , CpG Islands , DNA Methylation , Gene Expression Regulation , Humans , Interleukin-1 Receptor-Associated Kinases , Mice , Molecular Sequence Data , Transcription Factors , Transcription, Genetic
19.
Cancer Res ; 64(18): 6438-43, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15374952

ABSTRACT

Initial analysis identified the NPRL2/G21 gene located in 3p21.3C, the lung cancer region, as a strong candidate tumor suppressor gene. Here we provide additional evidence of the tumor suppressor function of NPRL2/G21. The gene has highly conserved homologs/orthologs ranging from yeast to humans. The yeast ortholog, NPR2, shows three highly conserved regions with 32 to 36% identity over the whole length. By sequence analysis, the main product of NPRL2/G21 encodes a soluble protein that has a bipartite nuclear localization signal, a protein-binding domain, similarity to the MutS core domain, and a newly identified nitrogen permease regulator 2 domain with unknown function. The gene is highly expressed in many tissues. We report inactivating mutations in a variety of tumors and cancer cell lines, growth suppression of tumor cells with tet-controlled NPRL2/G21 transgenes on plastic Petri dishes, and suppression of tumor formation in SCID mice. Screening of 7 renal, 5 lung, and 7 cervical carcinoma cell lines showed homozygous deletions in the 3' end of NPRL2 in 2 renal, 3 lung, and 1 cervical (HeLa) cell line. Deletions in the 3' part of NPRL2 could result in improper splicing, leading to the loss of the 1.8 kb functional NPRL2 mRNA. We speculate that the NPRL2/G21 nuclear protein may be involved in mismatch repair, cell cycle checkpoint signaling, and activation of apoptotic pathway(s). The yeast NPR2 was shown to be a target of cisplatin, suggesting that the human NPRL2/G21 may play a similar role. At least two homozygous deletions of NPRL2/G21 were detected in 6 tumor biopsies from various locations and with microsatellite instability. This study, together with previously obtained results, indicates that NPRL2 is a multiple tumor suppressor gene.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Genes, Tumor Suppressor , Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Small Cell/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Kidney Neoplasms/genetics , Lung Neoplasms/genetics
20.
Am J Pathol ; 165(3): 1007-18, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331424

ABSTRACT

CACNA2D2 is a putative tumor suppressor gene located in the human chromosome 3p21.3 region that shows frequent allelic imbalances in lung, breast, and other cancers. The alpha2delta-2 protein encoded by the gene is a regulatory subunit of voltage-dependent calcium channels and is expressed in brain, heart, and other tissues. Here we report that mice homozygous for targeted disruption of the Cacna2d2 gene exhibit growth retardation, reduced life span, ataxic gait with apoptosis of cerebellar granule cells followed by Purkinje cell depletion, enhanced susceptibility to seizures, and cardiac abnormalities. The Cacna2d2(tm1NCIF) null phenotype has much in common with that of Cacna1a mutants, such as cerebellar neuro-degeneration associated with ataxia, seizures, and premature death. A tendency to bradycardia and limited response of null mutants to isoflurane implicate alpha2delta-2 in sympathetic regulation of cardiac function. In summary, our findings provide genetic evidence that the alpha2delta-2 subunit serves in vivo as a component of P/Q-type calcium channels, is indispensable for the central nervous system function, and may be involved in hereditary cerebellar ataxias and epileptic disorders in humans.


Subject(s)
Calcium Channels/physiology , Cerebellar Ataxia/pathology , Disease Models, Animal , Growth Disorders/pathology , Heart Diseases/pathology , Seizures/pathology , Animals , Blood Pressure , Calcium Channels/genetics , Cerebellar Ataxia/genetics , Cerebellar Ataxia/mortality , Electrocardiography , Female , Gene Deletion , Gene Targeting , Genes, Tumor Suppressor , Growth Disorders/genetics , Growth Disorders/metabolism , Heart Diseases/genetics , Heart Diseases/metabolism , Homozygote , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Motor Activity , Purkinje Cells/metabolism , Purkinje Cells/pathology , Seizures/genetics , Seizures/mortality
SELECTION OF CITATIONS
SEARCH DETAIL
...