1
|
Uzhachenko R, Shimamoto A, Chirwa SS, Ivanov SV, Ivanova AV, Shanker A. Mitochondrial Fus1/Tusc2 and cellular Ca2 + homeostasis: tumor suppressor, anti-inflammatory and anti-aging implications. Cancer Gene Ther 2022; 29:1307-1320. [PMID: 35181743 PMCID: PMC9576590 DOI: 10.1038/s41417-022-00434-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/22/2021] [Accepted: 01/28/2022] [Indexed: 12/02/2022]
Abstract
FUS1/TUSC2 (FUSion1/TUmor Suppressor Candidate 2) is a tumor suppressor gene (TSG) originally described as a member of the TSG cluster from human 3p21.3 chromosomal region frequently deleted in lung cancer. Its role as a TSG in lung, breast, bone, and other cancers was demonstrated by several groups, but molecular mechanisms of its activities are starting to unveil lately. They suggest that Fus1-dependent mechanisms are relevant in etiologies of diseases beyond cancer, such as chronic inflammation, bacterial and viral infections, premature aging, and geriatric diseases. Here, we revisit the discovery of FUS1 gene in the context of tumor initiation and progression, and review 20 years of research into FUS1 functions and its molecular, structural, and biological aspects that have led to its use in clinical trials and gene therapy. We present a data-driven view on how interactions of Fus1 with the mitochondrial Ca2+ (mitoCa2+) transport machinery maintain cellular Ca2+ homeostasis and control cell apoptosis and senescence. This Fus1-mediated cellular homeostasis is at the crux of tumor suppressor, anti-inflammatory and anti-aging activities.
Collapse
Affiliation(s)
- Roman Uzhachenko
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Akiko Shimamoto
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, TN, USA
| | - Sanika S Chirwa
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Sergey V Ivanov
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Alla V Ivanova
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA.
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA.
- Host-Tumor Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
2
|
Oh SJ, Lee MG, Moon JR, Lee CK, Chi SG, Kim HJ. Ras association domain family 1 isoform A suppresses colonic tumor cell growth through p21 WAF1 activation in a p53-dependent manner. J Gastroenterol Hepatol 2019; 34:890-898. [PMID: 30226276 DOI: 10.1111/jgh.14469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 09/02/2018] [Accepted: 09/06/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIM Despite the frequent loss of Ras association domain family 1 isoform A (RASSF1A) expression in various cancers, the precise mechanism underlying its tumor-suppressive effect is not fully understood. To elucidate the growth-inhibitory role for RASSF1A in colorectal tumorigenesis, this study investigated the RASSF1A regulation of the p53-p21WAF1 pathway. METHODS Ras association domain family 1 isoform A effect on cellular growth was tested in three human colon cancer cell lines by flow cytometry, cell counting, and [3 H]-thymidine incorporation assay. HCT116 p53+/+ and p53-/- isogenic sublines were utilized to determine the p53 dependence of RASSF1A effect on p21WAF1 . Cycloheximide chase experiment and immunoprecipitation assay were carried out to define RASSF1A effect on p53 stability and mouse double minute 2 (MDM2) homolog ubiquitination. RESULTS Ras association domain family 1 isoform A expression inhibits colonic cell proliferation by preventing the G1 to S phase transition of the cell cycle. The RASSF1A-induced G1 cell cycle arrest is accompanied by the increase in the level of p21WAF1 mRNA expression. The p21WAF -inducing activity of RASSF1A was substantially higher in HCT116 p53+/+ cell compared with isogenic p53-/- cells. The cycloheximide chase assay revealed that RASSF1A expression leads to p53 stabilization and MDM2 homolog degradation. Using p53-/- and p21WAF1-/- subline cells, this study finally validated a crucial role of the p53-p21WAF1 axis in RASSF1A-mediated growth inhibition. CONCLUSIONS RASSF1A suppresses colonic tumor growth through the activation of the p53-p21WAF1 pathway. This finding supports that RASSF1A could be a valuable marker for the assessment of colorectal cancer development and progression.
Collapse
Affiliation(s)
- Shin Ju Oh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Korea
| | - Min-Goo Lee
- Department of Life Sciences, Korea University, Seoul, Korea
| | - Jung Rock Moon
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Korea
| | - Chang Kyun Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Korea
| | - Sung-Gil Chi
- Department of Life Sciences, Korea University, Seoul, Korea
| | - Hyo Jong Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Korea
| |
Collapse
|
3
|
Excess of a Rassf1-targeting microRNA, miR-193a-3p, perturbs cell division fidelity. Br J Cancer 2017; 116:1451-1461. [PMID: 28449010 PMCID: PMC5520089 DOI: 10.1038/bjc.2017.110] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/23/2017] [Accepted: 03/29/2017] [Indexed: 01/16/2023] Open
Abstract
Background: Several microRNA (miRNA) molecules have emerged as important post-transcriptional regulators of tumour suppressor and oncogene expression. Ras association domain family member 1 (RASSF1) is a critical tumour suppressor that controls multiple aspects of cell proliferation such as cell cycle, cell division and apoptosis. The expression of RASSF1 is lost in a variety of cancers due to the promoter hypermethylation. Methods: miR-193a-3p was identified as a RASSF1-targeting miRNA by a dual screening approach. In cultured human cancer cells, immunoblotting, qRT–PCR, luciferase reporter assays, time-lapse microscopy and immunofluorescence methods were used to study the effects of excess miR-193a-3p on RASSF1 expression and cell division. Results: Here, we report a new miRNA-mediated mechanism that regulates RASSF1 expression: miR-193a-3p binds directly to RASSF1-3′UTR and represses the mRNA and protein expression. In human cancer cells, excess of miR-193a-3p causes polyploidy through impairment of the Rassf1-Syntaxin 16 signalling pathway that is needed for completion of cytokinesis. In the next cell cycle the miR-193a-3p-overexpressing cells exhibit multipolar mitotic spindles, mitotic delay and elevated frequency of cell death. Conclusions: Our results suggest that besides epigenetic regulation, altered expression of specific miRNAs may contribute to the loss of Rassf1 in cancer cells and cause cell division errors.
Collapse
|
4
|
Augert A, Zhang Q, Bates B, Cui M, Wang X, Wildey G, Dowlati A, MacPherson D. Small Cell Lung Cancer Exhibits Frequent Inactivating Mutations in the Histone Methyltransferase KMT2D/MLL2: CALGB 151111 (Alliance). J Thorac Oncol 2016; 12:704-713. [PMID: 28007623 DOI: 10.1016/j.jtho.2016.12.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/05/2016] [Accepted: 12/09/2016] [Indexed: 12/31/2022]
Abstract
INTRODUCTION SCLC is a lethal neuroendocrine tumor type that is highly prone to metastasis. There is an urgency to understand the mutated genes that promote SCLC, as there are no approved targeted therapies yet available. SCLC is rarely resected, limiting the number of samples available for genomic analyses of somatic mutations. METHODS To identify potential driver mutations in human SCLC we sequenced the whole exomes of 18 primary SCLCs and seven cell lines along with matched normal controls. We extended these data by resequencing a panel of genes across 40 primary SCLCs and 48 cell lines. RESULTS We report frequent mutations in the lysine methyltransferase 2D gene (KMT2D) (also known as MLL2), a key regulator of transcriptional enhancer function. KMT2D exhibited truncating nonsense/frameshift/splice site mutations in 8% of SCLC tumors and 17% of SCLC cell lines. We found that KMT2D mutation in human SCLC cell lines was associated with reduced lysine methyltransferase 2D protein levels and reduced monomethylation of histone H3 lysine 4, a mark associated with transcriptional enhancers. We also found mutations in other genes associated with transcriptional enhancer control, including CREB binding protein gene (CREBBP), E1A binding protein p300 gene (EP300), and chromodomain helicase DNA binding protein 7 gene (CHD7), and we report mutations in additional chromatin remodeling genes such as polybromo 1 gene (PBRM1). CONCLUSIONS These data indicate that KMT2D is one of the major mutated genes in SCLC, and they point to perturbation of transcriptional enhancer control as potentially contributing to SCLC.
Collapse
Affiliation(s)
- Arnaud Augert
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Qing Zhang
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Breanna Bates
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Min Cui
- Carnegie Institution, Department of Embryology, Baltimore, Maryland
| | - Xiaofei Wang
- Duke University School of Medicine and Alliance Data and Statistical Center, Durham, North Carolina
| | - Gary Wildey
- Case Western Reserve Medical School, Division of Hematology and Oncology, Cleveland, Ohio
| | - Afshin Dowlati
- Case Western Reserve Medical School, Division of Hematology and Oncology, Cleveland, Ohio
| | - David MacPherson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington; Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Genome Sciences, University of Washington, Seattle, Washington.
| |
Collapse
|
5
|
Murray N, Noonan KL. Can we expect progress from targeted therapy of SCLC? Lung Cancer 2015. [DOI: 10.1183/2312508x.10010914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
6
|
Grawenda AM, O'Neill E. Clinical utility of RASSF1A methylation in human malignancies. Br J Cancer 2015; 113:372-81. [PMID: 26158424 PMCID: PMC4522630 DOI: 10.1038/bjc.2015.221] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 02/06/2023] Open
Abstract
The high frequency of RASSF1A methylation has been noted in a vast number of patients in a broad spectrum of malignancies, suggesting that RASSF1A inactivation is associated with cancer pathogenesis. However, whether this recurrent incidence of RASSF1A hypermethylation in human malignancies and its association with more aggressive tumour phenotype is a frequent event across different cancer types has not yet been discussed. In this review, we interrogated existing evidence for association of RASSF1A hypermethylation with clinicopathological characteristics that can indicate more invasive lesions.
Collapse
Affiliation(s)
- A M Grawenda
- CRUK/MRC Oxford Institute, Department of Oncology, University of Oxford, Oxford, UK
| | - E O'Neill
- CRUK/MRC Oxford Institute, Department of Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Additive Effects of Retinoic Acid (RA) and Bone Morphogenetic Protein 4 (BMP-4) Apoptosis Signaling in Retinoblastoma Cell Lines. PLoS One 2015; 10:e0131467. [PMID: 26173116 PMCID: PMC4501735 DOI: 10.1371/journal.pone.0131467] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/02/2015] [Indexed: 12/13/2022] Open
Abstract
Retinoids have been shown to serve promising therapeutic agents for human cancers, e.g. the treatment of neuroblastoma. Synthetic retinoids, specific for particular retinoic acid (RA) receptors, are tested as new therapy strategies. In the present study, application of recombinant retinoic acid (RA) lowers retinoblastoma (RB) cell viability and induces apoptosis in RB cell lines. Combined treatment of RA and bone morphogenetic protein 4 (BMP-4) increases the pro-apoptotic effect of RA in the RB cells lines WERI-Rb1, Y-79, RB355, RBL-30 and RBL-15, indicating an additive effect. We could show that in WERI-Rb1 cells RA/BMP-4 mediated cell death is at least partially caspase-dependent, whereby RA and BMP-4 additively increased (i) Apaf-1 mRNA levels, (ii) caspase-9 cleavage activity and (iii) the number of activated, cleaved caspase-3 positive cells. Compared to single application of RA and BMP-4, combined RA/BMP-4 treatment significantly augments mRNA levels of the retinoic acid receptors (RARs) RARα and RARß and the retinoic X receptor (RXR) RXRγ suggesting an interaction in the induction of these RA receptor subtypes in WERI-Rb1 cells. Agonist studies revealed that both, RARs and RXRs are involved in RA/BMP-4 mediated apoptosis in WERI-Rb1 retinoblastoma cells. Employing specific RAR subtype antagonists and a RXRß and RXRγ knockdown, we proved that RA/BMP-4 apoptosis signaling in WERI-Rb1 cells requires the RA receptor subtypes RARα, RARß, RXRß and RXRγ. Deciphering signaling mechanisms underlying apoptosis induction of RA and BMP-4 in WERI-Rb1 cells, our study provides useful starting-points for future retinoid-based therapy strategies in retinoblastoma.
Collapse
|
8
|
Sijmons RH, te Meerman GJ, Hofstra RMW. Charles Buys (1942-2014). Eur J Hum Genet 2014; 22:1343-4. [PMID: 25393683 DOI: 10.1038/ejhg.2014.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Rolf H Sijmons
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerard J te Meerman
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert M W Hofstra
- Department of Clinical Genetics, University of Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
9
|
|
10
|
Tsujiuchi T, Nakae D, Konishi Y. Multi-step lung carcinogenesis model induced by oral administration of N-nitrosobis(2-hydroxypropyl)amine in rats. ACTA ACUST UNITED AC 2013; 66:81-8. [PMID: 24377951 DOI: 10.1016/j.etp.2013.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 11/25/2013] [Accepted: 11/27/2013] [Indexed: 12/12/2022]
Abstract
N-Nitrosobis(2-hydroxypropyl)amine (BHP) was first synthesized by Krüger et al. (1974), and has been shown to primarily induce pancreatic duct adenocarcinomas by a subcutaneous injection in Syrian hamsters. By contrast, the carcinogenic effect of BHP has been indicated at the different target organs in rats, namely the lung. When rats are received by an oral administration of BHP in drinking water for 25 weeks, a high incidence of lung carcinomas are induced, which include adenocarcinomas, squamous cell carcinomas and combined squamous cell and adenocarcinomas. So many similarities are observed in terms of not only histological appearances but also gene alterations between human and BHP-induced rat lung cancers. Moreover, the step by step development of lung lesions, from preneoplastic lesions to cancers in rat lung carcinogenesis by BHP offers a good model to investigate the mechanisms underlying the pathogenesis of lung cancers. Because data for genetic and epigenetic alterations have indeed been accumulated during the BHP-induced rat lung carcinogenesis, we will introduce them in this review and hence demonstrate that this lung carcinogenesis model provides a useful opportunity for the research on the pathogenesis of lung cancers of both humans and rats.
Collapse
Affiliation(s)
- Toshifumi Tsujiuchi
- Division of Cancer Biology and Bioinformatics, Department of Life Science, Faculty of Science and Engineering, Kinki University, 3-4-1, Kowakae, Higashiosaka, Osaka 577-8502, Japan.
| | - Dai Nakae
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunin-cho, Shinjuku-ku, Tokyo 169-0073, Japan
| | - Yoichi Konishi
- Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8501, Japan; Oklahoma Medical Research Foundation, 825 N.E. 13th Street, Oklahoma City, OK 73104, USA
| |
Collapse
|
11
|
William WN, Glisson BS. Novel strategies for the treatment of small-cell lung carcinoma. Nat Rev Clin Oncol 2011; 8:611-9. [PMID: 21691321 DOI: 10.1038/nrclinonc.2011.90] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Small-cell lung cancer (SCLC) is a disease with a poor prognosis and limited treatment options. Over the past 30 years, basic and clinical research have translated to little innovation in the treatment of this disease. The Study of Picoplatin Efficacy After Relapse (SPEAR) evaluated best supportive care with or without picoplatin for second-line SCLC treatment and failed to meet its primary end point of overall survival. As the largest second-line, randomized study in patients with SCLC, SPEAR provides an opportunity to critically examine the drug development model in this disease. In this Review, we discuss the current standard approach for the management of SCLC that progresses after first-line therapy, analyze the preliminary data that supported the evaluation of picoplatin in this setting, and critically evaluate the SPEAR trial design and results. Lastly, we present advances in the understanding of the molecular biology of SCLC that could potentially inform future clinical trials and hopefully lead to the successful development of molecular targeted agents for the treatment of this disease.
Collapse
Affiliation(s)
- William N William
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas M D Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 432, Houston, TX 77030, USA
| | | |
Collapse
|
12
|
Cottage A, Edwards YJ, Elgar G. SAND, a new protein family: from nucleic acid to protein structure and function prediction. Comp Funct Genomics 2010; 2:226-35. [PMID: 18628914 PMCID: PMC2447211 DOI: 10.1002/cfg.93] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2001] [Accepted: 06/21/2001] [Indexed: 11/09/2022] Open
Abstract
As a result of genome, EST and cDNA sequencing projects, there are huge numbers of predicted and/or partially characterised protein sequences compared with a relatively small number of proteins with experimentally determined function and structure. Thus, there is a considerable attention focused on the accurate prediction of gene function and structure from sequence by using bioinformatics. In the course of our analysis of genomic sequence from Fugu rubripes, we identified a novel gene, SAND, with significant sequence identity to hypothetical proteins predicted in Saccharomyces cerevisiae, Schizosaccharomyces pombe, Caenorhabditis elegans, a Drosophila melanogaster gene, and mouse and human cDNAs. Here we identify a further SAND homologue in human and Arabidopsis thaliana by use of standard computational tools. We describe the genomic organisation of SAND in these evolutionarily divergent species and identify sequence homologues from EST database searches confirming the expression of SAND in over 20 different eukaryotes. We confirm the expression of two different SAND paralogues in mammals and determine expression of one SAND in other vertebrates and eukaryotes. Furthermore, we predict structural properties of SAND, and characterise conserved sequence motifs in this protein family.
Collapse
Affiliation(s)
- A Cottage
- UK Human Genome mapping Project Resource Centre, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SB, UK
| | | | | |
Collapse
|
13
|
Duns G, van den Berg E, van Duivenbode I, Osinga J, Hollema H, Hofstra RMW, Kok K. Histone methyltransferase gene SETD2 is a novel tumor suppressor gene in clear cell renal cell carcinoma. Cancer Res 2010; 70:4287-91. [PMID: 20501857 DOI: 10.1158/0008-5472.can-10-0120] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sporadic clear cell renal cell carcinoma (cRCC) is genetically characterized by the recurrent loss of the short arm of chromosome 3, with a hotspot for copy number loss in the 3p21 region. We applied a method called "gene identification by nonsense-mediated mRNA decay inhibition" to a panel of 10 cRCC cell lines with 3p21 copy number loss to identify biallelic inactivated genes located at 3p21. This revealed inactivation of the histone methyltransferase gene SETD2, located on 3p21.31, as a common event in cRCC cells. SETD2 is nonredundantly responsible for trimethylation of the histone mark H3K36. Consistent with this function, we observed loss or a decrease of H3K36me3 in 7 out of the 10 cRCC cell lines. Identification of missense mutations in 2 out of 10 primary cRCC tumor samples added support to the involvement of loss of SETD2 function in the development of cRCC tumors.
Collapse
Affiliation(s)
- Gerben Duns
- Departments of Genetics and Pathology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
14
|
Morrison BH, Haney R, Lamarre E, Drazba J, Prestwich GD, Lindner DJ. Gene deletion of inositol hexakisphosphate kinase 2 predisposes to aerodigestive tract carcinoma. Oncogene 2009; 28:2383-92. [PMID: 19430495 DOI: 10.1038/onc.2009.113] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Inositol hexakisphosphate kinase 2 (IP6K2), a member of the inositol hexakisphosphate kinase family, functions as a growth suppressive and apoptosis-enhancing kinase during cell stress. We created mice with a targeted deletion of IP6K2; these mice display normal embryogenesis, development, growth and fertility. Chronic exposure to the carcinogen 4-nitroquinoline 1-oxide (4-NQO, a UV-mimetic compound) in drinking water resulted in fourfold increased incidence of invasive squamous cell carcinoma (SCC) formation in the oral cavity and esophagus of the knockout (KO) mice compared to the wild-type (WT) littermates. Paradoxically, KO mice displayed relative resistance to ionizing radiation and exhibit enhanced survival following 8-10 Gy total body irradiation. Primary KO fibroblasts displayed resistance to antiproliferative effects of interferon-beta and increased colony forming units following ionizing radiation. Radioresistance of KO fibroblasts was associated with accelerated DNA repair measured by comet assay. Direct microinjection of 5-PP-Ins(1,2,3,4,6)P(5) (the enzymatic product of IP6K2), but not InsP(6) (the substrate of IP6K2) induced cell death in SCC22A squamous carcinoma cells.
Collapse
Affiliation(s)
- B H Morrison
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | | | |
Collapse
|
15
|
Abstract
Small cell lung carcinoma (SCLC) accounts for approximately 15% of all lung cancer cases. Despite a frequently good response to first-line treatment with chemotherapy and/or radiotherapy, early relapse occurs in the majority of patients and 5-year survival is only about 5%. Therefore, there is a need to develop novel treatments to improve the outcome of patients with SCLC. To fulfil this need, it is critical to gain further understanding on the molecular basis of SCLC and specifically to identify novel therapeutic targets. Clinical trials with molecularly targeted agents have been performed with little success in the past, but recently many promising oncogenic pathways have been discovered and novel targeted therapies are under evaluation. In this review, we summarise the most relevant genetic and signalling pathway alterations reported to date in SCLC and discuss the potential therapeutic implications of such events.
Collapse
|
16
|
Nam CW, Shin JW, Park NH. Fragile histidine triad gene alterations are not essential for hepatocellular carcinoma development in South Korea. World J Gastroenterol 2008; 14:3526-33. [PMID: 18567082 PMCID: PMC2716616 DOI: 10.3748/wjg.14.3526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish the role of FHIT in the pathogenesis hepatocellular carcinoma (HCC).
METHODS: We examined genomic alterations, as well as, mRNA and protein expression patterns from the FHIT gene, in 48 surgically resected hepatocellular carcinoma (HCC) tissues. Additionally, p53 mutations were analyzed.
RESULTS: Aberrant FHIT transcripts were detected in 11 of 48 surrounding non-tumor liver tissues and 27 of 48 HCC samples (22.9% vs 56.3%, P = 0.002). No point mutations were identified within the open reading frame region of FHIT. Loss of heterozygosity (LOH) of the FHIT locus was detected in 4 of 42 informative cases for D3S1300, and 3 of 29 informative cases for D3S1313. Reduced expression of FHIT protein (Fhit) was observed in 8 (16.7%) of 48 HCC samples, with complete loss of Fhit in only 1 case. There were no associations with abnormal transcripts, LOH, and Fhit expression. p53 mutations were identified in 9 of the 48 HCC cases. However, none of the cases displayed a G to T transversion at p53 codon 249.
CONCLUSION: Aberrant FHIT transcripts were more common in HCC tissues as compared to non-cancerous liver tissues. However, Fhit expression was lost or reduced in a minor fraction of HCC tissues, while it was strongly expressed in non-cancerous liver tissues. Therefore, our study suggests that FHIT plays a role in relatively few HCC cases in South Korea.
Collapse
|
17
|
Shimizu K, Itsuzaki Y, Fujii H, Honoki K, Tsujiuchi T. Reduced expression of the Rassf1a gene and its aberrant DNA methylation in pancreatic duct adenocarcinomas induced by N-nitrosobis(2-oxopropyl)amine in hamsters. Mol Carcinog 2008; 47:80-7. [PMID: 17849420 DOI: 10.1002/mc.20250] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Alterations of the Rassf1a gene were investigated in pancreatic duct adenocarcinomas (PDAs) induced by N-nitrosobis(2-oxopropyl)amine (BOP) in hamsters. Female Syrian golden hamsters received 70 mg/kg BOP, followed by repeated exposures to an augmentation pressure regimen consisting of a choline-deficient diet combined with a sequential course of DL-ethionine, L-methionine, and 20 mg/kg BOP. A total of 15 PDAs were obtained, and total RNAs were assessed by real-time quantitative reverse transcription (RT)-polymerase chain reaction (PCR). Expression of the Rassf1a was significantly reduced in PDAs (P < 0.005) compared with normal pancreatic tissues. For analysis of methylation status, bisulfite sequencing was performed. Normal tissues were all unmethylated in the 5' upstream region of Rassf1a. In contrast, four PDAs were highly methylated, correlating with reduced expression of the Rassf1a gene. Using reverse transcription (RT)-polymerase chain reaction (PCR)-single strand conformation polymorphism (SSCP) analysis, mutations were detected in 3 out of 15 PDAs (20%). These results suggested that alterations of the Rassf1a gene may be involved in development of PDAs induced by BOP in hamsters.
Collapse
Affiliation(s)
- Kyoko Shimizu
- Department of Life Science, Laboratory of Cancer Biology and Bioinformatics, Faculty of Science and Engineering, Kinki University, Kowakae, Higashiosaka, Osaka, Japan
| | | | | | | | | |
Collapse
|
18
|
Gresner P, Gromadzinska J, Wasowicz W. Polymorphism of selected enzymes involved in detoxification and biotransformation in relation to lung cancer. Lung Cancer 2007; 57:1-25. [PMID: 17337085 DOI: 10.1016/j.lungcan.2007.02.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Revised: 01/30/2007] [Accepted: 02/04/2007] [Indexed: 01/22/2023]
Abstract
Available data indicate that there are significant differences in individual susceptibility to lung cancer within the human population. It is believed to be underlie by inherited genetic predispositions related to the genetic polymorphism of several enzymes involved in the detoxification and xenobiotic metabolism. In this review, we collect and discuss the evidence reported up to date on the association between lung cancer and genetic polymorphism of cytochromes P450, N-acetyltransferase, glutathione S-transferases, microsomal epoxide hydrolase, NAD(P)H:quinone oxidoreductase, myeloperoxidase and glutathione peroxidase. All these genes might appear to be candidates for lung cancer susceptibility genes, nevertheless, the present state of the art still offers only a limited explanation of the link between such polymorphisms and increased risk of lung cancer.
Collapse
Affiliation(s)
- Peter Gresner
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, Lodz, Poland.
| | | | | |
Collapse
|
19
|
Futamura M, Kamino H, Miyamoto Y, Kitamura N, Nakamura Y, Ohnishi S, Masuda Y, Arakawa H. Possible role of semaphorin 3F, a candidate tumor suppressor gene at 3p21.3, in p53-regulated tumor angiogenesis suppression. Cancer Res 2007; 67:1451-60. [PMID: 17308083 DOI: 10.1158/0008-5472.can-06-2485] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the regulation of tumor angiogenesis is believed to be one of the core functions of p53, the mechanism still remains to be elucidated. Here, we report that semaphorin 3F (SEMA3F), an axon guidance molecule, is involved in p53-regulated antiangiogenesis. The expression level of SEMA3F mRNA was increased by both exogenous and endogenous p53. Chromatin immunoprecipitation assay indicated that a potent p53-binding sequence in intron 1 of SEMA3F interacts with p53 and that it has a p53-responsive transcriptional activity. Overexpression of SEMA3F inhibited in vitro cell growth of the lung cancer cell line H1299. In nude mice assay, the size of the H1299 tumors expressing SEMA3F was much smaller, and they showed lesser number of blood vessels as compared with the control tumors. Moreover, tumors derived from the p53-knockdown colorectal cancer cell line LS174T displayed a remarkable enhancement of tumor vessel formation as compared with control tumors containing normal levels of p53. The expression levels of SEMA3F and neuropilin-2 (NRP2), the functional receptor for SEMA3F, in p53-knockdown LS174T tumors were lower than those in the control tumors. Adenovirus-mediated SEMA3F gene transfer induced the remarkable in vitro growth suppression of the stable transformant of H1299 cells, which express high levels of NRP2. These results suggest that p53 negatively regulates tumor vessel formation and cell growth via the SEMA3F-NRP2 pathway.
Collapse
MESH Headings
- Cell Growth Processes/genetics
- Cell Line, Tumor
- Chromosomes, Human, Pair 3
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor
- Genes, p53
- HCT116 Cells
- Humans
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Neoplasms/blood supply
- Neoplasms/genetics
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Nerve Tissue Proteins/biosynthesis
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neuropilin-2/metabolism
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Signal Transduction
- Transcription, Genetic
Collapse
Affiliation(s)
- Manabu Futamura
- Cancer Medicine and Biophysics Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Angeloni D, ter Elst A, Wei MH, van der Veen AY, Braga EA, Klimov EA, Timmer T, Korobeinikova L, Lerman MI, Buys CHCM. Analysis of a new homozygous deletion in the tumor suppressor region at 3p12.3 reveals two novel intronic noncoding RNA genes. Genes Chromosomes Cancer 2006; 45:676-91. [PMID: 16607615 DOI: 10.1002/gcc.20332] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
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.
Collapse
Affiliation(s)
- Debora Angeloni
- Laboratory of Immunobiology, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Shimizu K, Kato A, Shigemura M, Fujii H, Honoki K, Tsujiuchi T. Aberrant methylation patterns of theRassf1a gene in rat lung adenocarcinomas induced by N-nitrosobis(2-hydroxypropyl)amine. Mol Carcinog 2006; 45:112-7. [PMID: 16329149 DOI: 10.1002/mc.20173] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
To clarify the involvement of the Rassf1a gene in lung carcinogenesis, we investigated the methylation status in the 5' upstream region of the RAS-association domain family 1, isoform A (Rassf1a) gene in rat lung adenocarcinomas induced by N-nitrosobis(2-hydroxypropyl)amine (BHP). Six-week-old male Wistar rats were given 2000 ppm BHP in their drinking water for 12 wk and maintained without further treatment until they were sacrificed at 25 wk. A total of 15 lung adenocarcinomas were obtained and total RNAs were extracted from each for assessment of expression of the Rassf1a gene by reverse transcription (RT)-polymerase chain reaction (PCR) analysis. To measure the methylation status of the Rassf1a gene, five adenocarcinomas with a marked reduction of the Rassf1a expression and two normal lung tissues were used for a bisulfite sequencing analysis. While the normal lung tissue was unmethylated, all five adenocarcinomas were highly methylated in the 5' upstream region. Genomic DNAs were also extracted from 15 adenocarcinomas, and mutation analysis of the Rassf1a was performed with PCR-single-strand conformation polymorphism (SSCP) analysis. No mutations were detected throughout exons 1-6. The present results suggest that the aberrant methylation may be involved in the inactivation of the Rassf1a gene in the development of lung adenocarcinomas induced by BHP in rats.
Collapse
Affiliation(s)
- Kyoko Shimizu
- Laboratory of Cancer Biology and Bioinformatics, Department of Life Science, Faculty of Science and Engineering, Kinki University, Kowakae, Higashiosaka, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
22
|
Grote HJ, Schmiemann V, Geddert H, Bocking A, Kappes R, Gabbert HE, Sarbia M. Methylation of RAS association domain family protein 1A as a biomarker of lung cancer. Cancer 2006; 108:129-34. [PMID: 16444703 DOI: 10.1002/cncr.21717] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Promoter hypermethylation is an important mechanism for silencing tumor-suppressor genes in cancer and a promising tool for development of molecular biomarkers. This study aimed to determine the prevalence of RAS association domain family protein 1A (RASSF1A) promoter hypermethylation in bronchial aspirates of patients with suspected lung cancer and to test whether this type of methylation assay could be used as a diagnostic adjunct to conventional cytology. METHODS Two hundred three bronchial aspirates from patients with suspected lung cancer were analyzed for RASSF1A hypermethylation by using a sensitive quantitative methylation-specific polymerase chain reaction (QMSP). RESULTS RASSF1A hypermethylation was found in 88% (35 of 40), 28% (31 of 111), and 100% (6 of 6) of bronchial aspirates collected from patients diagnosed with small cell lung cancer, nonsmall cell lung cancer, and combined small cell lung cancer, respectively. No hypermethylation was detected in patients diagnosed with nonneoplastic lung disease (0 of 46). Depending on histologic subtype, up to 82% of cases presenting with a negative histology showed a positive methylation assay. CONCLUSIONS The QMSP analysis of RASSF1A hypermethylation enabled a highly specific distinction between patients diagnosed with lung cancer and those with nonneoplastic lung disease. These results suggested that a QMSP assay is a promising molecular tool for diagnosis of primary lung cancer.
Collapse
MESH Headings
- Adenocarcinoma/chemistry
- Adenocarcinoma/diagnosis
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biopsy, Needle
- Carcinoma, Non-Small-Cell Lung/chemistry
- Carcinoma, Non-Small-Cell Lung/diagnosis
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Small Cell/chemistry
- Carcinoma, Small Cell/diagnosis
- Carcinoma, Small Cell/genetics
- Carcinoma, Small Cell/pathology
- Case-Control Studies
- Cell Line, Tumor
- DNA Methylation
- DNA, Neoplasm/analysis
- DNA, Neoplasm/genetics
- Diagnosis, Differential
- Female
- Gene Silencing
- Humans
- Lung Neoplasms/chemistry
- Lung Neoplasms/diagnosis
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Male
- Middle Aged
- Molecular Diagnostic Techniques/methods
- Promoter Regions, Genetic
- Retrospective Studies
- Reverse Transcriptase Polymerase Chain Reaction
- Sensitivity and Specificity
- Tumor Suppressor Proteins/analysis
- Tumor Suppressor Proteins/genetics
Collapse
Affiliation(s)
- Hans Juergen Grote
- Institute of Cytopathology, Heinrich Heine University, Duesseldorf, Germany.
| | | | | | | | | | | | | |
Collapse
|
23
|
Meira LB, Burgis NE, Samson LD. Base excision repair. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 570:125-73. [PMID: 18727500 DOI: 10.1007/1-4020-3764-3_5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Lisiane B Meira
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
24
|
Macejova D, Radikova Z, Macho L, Liska J, Brtko J. MNU-induced carcinogenesis of rat mammary gland: effect of thyroid hormone on expression of retinoic acid receptors in tumours of mammary gland. Mol Cell Endocrinol 2005; 244:47-56. [PMID: 16219415 DOI: 10.1016/j.mce.2005.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2004] [Accepted: 01/30/2005] [Indexed: 11/19/2022]
Abstract
The rat model of N-methyl-N-nitrosourea (MNU)-induced mammary carcinomas is well-established animal model for breast cancer. This study was carried out to investigate whether hypothyroid (thyroidectomy or PTU treatment) or hyperthyroid status of female rats would affect MNU-induced mammary carcinogenesis with specific focus on both retinoid and rexinoid receptor expression in mammary tumours. Application of PTU before and during MNU-induced mammary gland carcinogenesis yielded in a marked decrease of the number and volume of tumours per animal, however, there was no effect of hypothyroid state in thyroidectomized rats as well as hyperthyroid state concerning the number and volume of tumours. Mammary tumours of in euthyroid group of MNU animals showed that there was no tumour, in which all of subtypes of retinoid and rexinoid receptors were expressed. A different pattern of expression of retinoid or rexinoid receptors was found either in MNU-induced mammary carcinomas in both hypothyroid and hyperthyroid rats.
Collapse
Affiliation(s)
- Dana Macejova
- Institute of Experimental Endocrinology, Slovak Academy of Sciences, Vlárska 3, 833 06 Bratislava, Slovak Republic
| | | | | | | | | |
Collapse
|
25
|
Philbey AW, Cousens C, Bishop JV, Gill CA, DeMartini JC, Sharp JM. Multiclonal pattern of Jaagsiekte sheep retrovirus integration sites in ovine pulmonary adenocarcinoma. Virus Res 2005; 117:254-63. [PMID: 16310879 DOI: 10.1016/j.virusres.2005.10.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2005] [Revised: 10/25/2005] [Accepted: 10/31/2005] [Indexed: 11/21/2022]
Abstract
Insertional mutagenesis and envelope (Env)-mediated oncogenesis are hypothesized mechanisms by which Jaagsiekte sheep retrovirus (JSRV) causes ovine pulmonary adenocarcinoma (OPA). Twenty-eight JSRV integration sites in lung tumors (LTs) from four sheep with OPA were cloned and sequenced by a multiple step gene walking technique. Using nested PCR, clonal expansion of these integration sites could be detected, if at all, only in the localized regions of LT from which the integration sites were derived. One sheep had a viral integration site in a sequence with 85 and 81% identity, respectively, over 100 bp to exon 2 of the human and mouse receptor protein tyrosine phosphatase gamma genes. Clonal integration of Jaagsiekte sheep retrovirus in this gene was demonstrated by nested PCR and Southern blot hybridization in the DNA sample from which the integration site was cloned, but not in other LT or kidney DNA samples from the same sheep. OPA may develop from multiple independent oncogenic events and a role for insertional mutagenesis cannot be ruled out.
Collapse
|
26
|
Breuer RHJ, Postmus PE, Smit EF. Molecular pathology of non-small-cell lung cancer. Respiration 2005; 72:313-30. [PMID: 15942304 DOI: 10.1159/000085376] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2003] [Accepted: 07/29/2004] [Indexed: 01/22/2023] Open
Abstract
The molecular basis of lung carcinogenesis must be understood more fully and exploited to enhance survival rates of patients suffering from lung cancer. In this review we will discuss the major molecular alterations that occur in lung cancer. Emphasis is placed on alterations that occur early during carcinogenesis since they might be relevant for future screening programs. Finally we will shortly review new approaches that are used to study the molecular pathology of lung cancer and how they can be applied in a clinical setting.
Collapse
Affiliation(s)
- R H J Breuer
- Department of Pulmonology, Free University Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
27
|
Ducruet AP, Vogt A, Wipf P, Lazo JS. DUAL SPECIFICITY PROTEIN PHOSPHATASES: Therapeutic Targets for Cancer and Alzheimer's Disease. Annu Rev Pharmacol Toxicol 2005; 45:725-50. [PMID: 15822194 DOI: 10.1146/annurev.pharmtox.45.120403.100040] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The complete sequencing of the human genome is generating many novel targets for drug discovery. Understanding the pathophysiological roles of these putative targets and assessing their suitability for therapeutic intervention has become the major hurdle for drug discovery efforts. The dual-specificity phosphatases (DSPases), which dephosphorylate serine, threonine, and tyrosine residues in the same protein substrate, have important roles in multiple signaling pathways and appear to be deregulated in cancer and Alzheimer's disease. We examine the potential of DSPases as new molecular therapeutic targets for the treatment of human disease.
Collapse
Affiliation(s)
- Alexander P Ducruet
- Department of Pharmacology, the Combinatorial Chemistry Center and the Fiske Drug Discovery Laboratory, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | |
Collapse
|
28
|
Stern R. Hyaluronan metabolism: a major paradox in cancer biology. ACTA ACUST UNITED AC 2005; 53:372-82. [PMID: 16085113 DOI: 10.1016/j.patbio.2004.12.021] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2004] [Accepted: 12/09/2004] [Indexed: 11/16/2022]
Abstract
Paradoxically, both hyaluronan (HA) and hyaluronidases, the enzymes that eliminate HA, can correlate with cancer progression. Levels of HA on the surface of tumor cells are indicators of poor outcome. Certain hyaluronidases, products of tumor suppressor genes eliminated in the course of tumor spread, are used clinically in anti-cancer chemotherapy regimens. Such information would indicate that cancer progression is inhibited by hyaluronidase. Yet progression of certain cancers correlates with levels of hyaluronidase activity. An attempt is made here to understand such apparent contradictions by examining details of HA metabolism. Anabolic and catabolic pathways are comprised of the HA synthases and hyaluronidases, respectively. There are several enzymes that synthesize HA, each under a different control mechanism, generating products of differing polymer size. The hyaluronidases degrade HA in step-wise fashion, the polymer decreasing in size in quantum steps, each size-specific polymer having a different biological activity. Superimposed on these are the potent hyaluronidase inhibitors, about which very little is known. These components of HA metabolism are reviewed here for possible roles in supporting or suppressing malignant transformation, growth, invasion and metastatic spread of tumors. Such a systematic approach may reveal mechanisms used in the course of cancer progression, resolve some of the apparent disparities, render new prognostic markers, and provide new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Robert Stern
- Department of Pathology, UCSF Comprehensive Cancer Center, University of California San Francisco, CA 94143-0511, USA.
| |
Collapse
|
29
|
Abstract
Over the past 100 years, our understanding of the pathogenesis of lung cancer has advanced impressively. Environmental carcinogens and a gene locus determining susceptibility have been identified. The pathology of lung cancer has been classified into categories with major clinical implications. The cellular and molecular genetic changes underlying lung cancer have become better understood over the past 25 years, but the stepwise progression of respiratory epithelium from normal to neoplastic is not yet well demarcated, limiting abilities to advance early detection and chemoprevention. The translation of improved understanding of dominant signal transduction pathways in lung cancer to rationally designed therapeutic strategies has had recent successes, demonstrating a proof of principle for targeted therapy in lung cancer. Improvement in overall patient outcomes has been stubbornly slow and will require concerted efforts.
Collapse
Affiliation(s)
- York E Miller
- Pulmonary 111A, Denver Veterans Affairs Medical Center, 1055 Clermont Street, Denver, CO 80220-3808, USA.
| |
Collapse
|
30
|
Shimizu K, Kato A, Hinotsume D, Shigemura M, Hanaoka M, Shimoichi Y, Honoki K, Tsujiuchi T. Reduced expressions of Foxp1 and Rassf1a genes in lung adenocarcinomas induced by N-nitrosobis(2-hydroxypropyl)amine in rats. Cancer Lett 2005; 236:186-90. [PMID: 16023287 DOI: 10.1016/j.canlet.2005.05.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2005] [Revised: 04/26/2005] [Accepted: 05/09/2005] [Indexed: 11/28/2022]
Abstract
To clarify the involvement of the Foxp1 and Rassf1a genes in lung carcinogenesis, we investigated their expressions in lung adenocarcinomas induced by N-nitrosobis(2-hydroxypropyl)amine (BHP) in rats. Six week old male Wistar rats were given 2000 ppm BHP in their drinking water for 12 weeks and maintained without further treatment until they were sacrificed at 25 weeks. A total of 10 lung adenocarcinomas were obtained, along with the total RNA from each for assessment of expression by reverse transcription (RT)-polymerase chain reaction (PCR). The reduced expressions of the Foxp1 and Rassf1a genes were observed in some of the lung adenocarcinomas. These analyses were also confirmed by real-time quantitative RT-PCR. These results suggest that reduced expressions of Foxp1 and Rassf1a genes may play a role in the development of lung adenocarcinomas induced by BHP in rats.
Collapse
Affiliation(s)
- Kyoko Shimizu
- Laboratory of Cancer Biology and Bioinformatics, Department of Life Science, Faculty of Science and Technology, Kinki University, 3-4-1, Kowakae, Higashiosaka, Osaka 577-8502, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
In the United States, the 20th century witnessed the emergence of a lung cancer epidemic that peaked and began to decline by the century's end, a decline that continues today. However, lung cancer continues to be an unabating pandemic. In research carried out over the last half of the 20th century, many factors were causally associated with lung cancer and studies were implemented to identify determinants of susceptibility to these factors. Cigarette smoking was identified as the single most predominant cause of the lung cancer epidemic, but other causes were found, including workplace agents (eg, asbestos, arsenic, chromium, nickel, and radon) and other environmental factors (passive smoking, indoor radon, and air pollution). Contemporary epidemiologic research on lung cancer now focuses on a new set of issues, primarily related to susceptibility to the well-identified causal factors, particularly smoking, and on the consequences of changes in tobacco products for risks to smokers. Diet and the possibility of reducing risk through chemoprevention remain a focus of research emphasis through experimental and observational approaches. Questions have also been raised about possible differences in susceptibility to lung cancer by sex and race. Population patterns in smoking prevalence will continue to be the most powerful predictor of the future occurrence of lung cancer. Evaluation of recent US patterns in smoking prevalence indicates that for the next approximately 10 to 15 years, lung cancer rates will decrease, but will then level off starting in approximately 2030. Unless further reductions in the prevalence of cigarette smoking are achieved over the next decade, lung cancer will remain as an all too common, but avoidable, disease.
Collapse
Affiliation(s)
- Anthony J Alberg
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, 21205, USA.
| | | | | |
Collapse
|
32
|
Ito M, Ito G, Kondo M, Uchiyama M, Fukui T, Mori S, Yoshioka H, Ueda Y, Shimokata K, Sekido Y. Frequent inactivation of RASSF1A, BLU, and SEMA3B on 3p21.3 by promoter hypermethylation and allele loss in non-small cell lung cancer. Cancer Lett 2004; 225:131-9. [PMID: 15922865 DOI: 10.1016/j.canlet.2004.10.041] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2004] [Revised: 10/25/2004] [Accepted: 10/28/2004] [Indexed: 02/04/2023]
Abstract
Non-small cell lung cancer frequently shows loss of heterozygosity of the chromosome 3p21.3 region and several genes such as RASSF1A, BLU, and SEMA3B have been identified as candidate tumor suppressor genes at this region since their downregulation and hypermethylation at their promoter regions were frequently detected in lung cancer. To determine whether these three genes are simultaneously inactivated during lung cancer development, we studied 138 primary non-small cell lung cancers for the promoter methylation status of these genes and allelic loss of the chromosome 3p21.3 region. We found promoter hypermethylation at 32% in RASSF1A, 30% in BLU, and 47% in SEMA3B. Allelic loss of 3p21.3 was detected in 54 (58%) of 93 informative tumors. Despite the weak association of methylation status among these three genes, there was no correlation between the methylation status of each gene and loss of heterozygosity. We also studied possible genes downstream of RASSF1A in 16 primary non-small cell lung cancers and found that the expressions of SM22 and SPARC were significantly downregulated in RASSF1A-hypermethylated tumors. Our results showed that, while candidate tumor suppressor genes at this locus can be simultaneously inactivated by epigenetic alterations, loss of heterozygosity without any hypermethylation of the three genes can also occur in some cases, suggesting that just one allelic loss might also be sufficient for the inactivation of any of these genes for lung cancer development.
Collapse
Affiliation(s)
- Masao Ito
- Department of Clinical Preventive Medicine, Nagoya University School of Medicine, Tsurumai 65, Nagoya 466-8560, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Welborn J, Jenks H, Taplett J, Walling P. High-grade neuroendocrine carcinomas display unique cytogenetic aberrations. ACTA ACUST UNITED AC 2004; 155:33-41. [PMID: 15527900 DOI: 10.1016/j.cancergencyto.2004.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2003] [Revised: 03/01/2004] [Accepted: 03/02/2004] [Indexed: 01/12/2023]
Abstract
Neuroendocrine tumors represent a spectrum of tumor types with different biologic and clinical features. The morphologic types include the low-grade typical and atypical carcinoids and the high-grade small cell and large cell neuroendocrine carcinomas (NECs). Cytogenetic descriptions of high-grade NECs are rare. Complete karyotypic descriptions of 34 high-grade NECs are reviewed: 7 extrapulmonary small cell NECs, 3 metastatic NECs of unknown primary, and 24 small cell lung carcinomas (SCLCs). Chromosomal deletions are more frequent than gains and often involve the entire chromosome arm. Typical aberrations are deletions of chromosome 3p, 5q, 10q, and 17p and gains of 1q, 3q, and 5p occurring as isochromosomes. Non-small cell lung cancers (NSCLCs) have different cytogenetic aberrations, but those with a metastatic phenotype display the identical aberrations as SCLC, a tumor known for its metastatic phenotype at onset. A genetic classification of lung cancer that incorporates the pattern of recurrent chromosome aberrations may be a better predictor of clinical outcome than a morphologic classification.
Collapse
Affiliation(s)
- Jeanna Welborn
- Department of Internal Medicine, University of California at Davis Medical Center, UCDMC Cancer Center, Room 3017, 4501 X Street, Sacramento, CA 95817, USA.
| | | | | | | |
Collapse
|
34
|
Tomizawa Y, Iijima H, Nomoto T, Iwasaki Y, Otani Y, Tsuchiya S, Saito R, Dobashi K, Nakajima T, Mori M. Clinicopathological significance of aberrant methylation of RAR?2 at 3p24, RASSF1A at 3p21.3, and FHIT at 3p14.2 in patients with non-small cell lung cancer. Lung Cancer 2004; 46:305-12. [PMID: 15541815 DOI: 10.1016/j.lungcan.2004.05.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2004] [Revised: 04/29/2004] [Accepted: 05/04/2004] [Indexed: 11/18/2022]
Abstract
We investigated the clinicopathological significance of aberrant methylation of the retinoic acid receptor-beta2 (RARbeta2), RAS association domain family 1A (RASSF1A) and fragile histidine triad (FHIT) genes located on choromosome 3p in 120 patients with primary non-small cell lung cancer (NSCLC) by a methylation-specific PCR method. Aberrant methylation of these was detected in 31 (26%), 35 (29%) and 43 (36%) tumors, respectively. There was no correlation with the methylation status of any of the genes. RARbeta2 methylation was more frequently observed in patients with a smoking history (19 of 61, 31%) than in patients without one (3 of 29, 10%, P = 0.0373). RARbeta2 methylation was also preferentially observed in advanced stage NSCLC (12 of 71 (17%) in stage I, 5 of 15 (33%) in stage II, 11 of 24 (46%) in stage III, and 3 of 8 (38%) in stage IV, P = 0.0057 (stage I versus II, III,and IV)). FHIT methylation was predominantly detected in tumors with vascular invasion (21 of 44, 48%, P = 0.0703) or lymphatic permeation (28 of 59, 47%, P = 0.0115). RASSF1A methylation was more frequently observed in adenocarcinomas (28 of 72, 39%) than in squamous cell carcinomas (6 of 45, 13%, P = 0.0033). These results indicate that aberrant methylation of the candidate tumor suppressor genes on 3p plays a respective role in the pathogenesis of NSCLC.
Collapse
Affiliation(s)
- Yoshio Tomizawa
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Markman M, Belinson J, Webster K, Zanotti K, Morrison B, Jacobs B, Borden E, Lindner D. Phase 2 Trial of Interferon-Beta as Second-Line Treatment of Ovarian Cancer, Fallopian Tube Cancer, or Primary Carcinoma of the Peritoneum. Oncology 2004; 66:343-6. [PMID: 15331919 DOI: 10.1159/000079480] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2003] [Accepted: 10/06/2003] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The protocol was designed to examine the biological effects and clinical activity of interferon-beta in patients with platinum/taxane-resistant ovarian cancer. METHODS Patients with resistant ovarian and fallopian tube cancers and primary peritoneal carcinoma were treated with recombinant human interferon-beta (Rebif, Serono International) at doses ranging from 6 to 24 million international units (MIU)/day, based on their tolerance to therapy. Levels of IP-10, an interferon-inducible protein, were measured in the serum to evaluate the biological effects of the drug. Also, the peripheral blood mononuclear cells and serum were examined for the induction of previously described novel regulators of interferon-induced death. RESULTS Eighteen patients were treated, of whom 9 (50%) could be treated at the highest dose level (24 MIU). The major toxicities were fever, chills and fatigue. The median duration of therapy was 6 weeks (range 1-22). No objective responses were observed. IP-10 levels were significantly increased, compared with baseline, at 2, 4, and 6 weeks after initiation of therapy (p < 0.01). CONCLUSIONS Recombinant human interferon-beta produced a definite biological effect in the serum of treated patients, but this outcome was not translated into any clinically observable or meaningful impact on the disease process.
Collapse
Affiliation(s)
- Maurie Markman
- Cleveland Clinic Taussig Cancer Center, and the Departments of Hematology/Medical Oncology, The Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Sarli L, Bottarelli L, Azzoni C, Campanini N, Di Cola G, Bader G, Iusco D, Salvemini C, Caruso G, Donadei E, Pizzi S, D'Adda T, Renato C, Roncoroni L, Bordi C. Abnormal Fhit protein expression and high frequency of microsatellite instability in sporadic colorectal cancer. Eur J Cancer 2004; 40:1581-8. [PMID: 15196543 DOI: 10.1016/j.ejca.2004.02.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2003] [Revised: 02/13/2004] [Accepted: 02/26/2004] [Indexed: 10/26/2022]
Abstract
The role of Fhit protein in the oncogenesis of colorectal cancer is still in debate. Recent studies have revealed that reduced Fhit protein expression is associated with a deficiency of the mismatch repair protein. One hundred and twenty unselected patients who underwent curative resection for sporadic colorectal cancer in a three-year period were evaluated for microsatellite instability (MSI) using six microsatellite markers, and for the presence of Fhit and mismatch repair (MMR) proteins (Mlh1 and Msh2) by means of immunostaining. The relations between these markers were analysed. Reduced or absent Fhit expression was noted in 18 out of 118 patients. This altered expression was significantly higher in right-sided cancer (P = 0.005), mucinous tumours (P = 0.005) and in poorly differentiated histological types (P = 0.0001). MSI was found in 22 out of 109 patients, more so in right-sided cancer (P = 0.0001), poorly differentiated histology (P = 0.0001), and mucinous tumours (P = 0.0001). No association was found with TNM stage. MSI was present in 66.7% of tumours with altered Fhit expression and in only 10% of tumours with preserved or intermediate Fhit expression (P = 0.0001). Of the tumours with reduced or absent Fhit expression, 72.2% had loss of nuclear Mlh1 or Msh2 expression compared with only 14% of the preserved or intermediate Fhit expression tumours (P = 0.0001). These results support the hypothesis that deficiency in a MMR gene could be a cause of the high frequency of alterations in Fhit expression, and they permit the suggestion that FHIT gene alteration may be part of the genetic pathway involving MSI through which some colorectal cancers arise.
Collapse
Affiliation(s)
- Leopoldo Sarli
- Department of Surgical Sciences, Section of General Surgical Clinics and Surgical Therapy, Parma University, Medical School, Via Gramsci 14, 43100 Parma, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Tsujiuchi T, Sasaki Y, Oka Y, Kuniyasu H, Tsutsumi M. Alterations of the Dutt1/Robo1 gene in lung adenocarcinomas induced by N-nitrosobis(2-hydroxypropyl)amine in rats. Mol Carcinog 2004; 40:241-6. [PMID: 15264215 DOI: 10.1002/mc.20037] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abnormalities of tumor suppressor genes (TSGs) on chromosome 3p are known to be important for the development of human lung cancers. In the present study, we investigated alterations of the Dutt1/Robo1 gene, as a possible tumor suppressor in this region, in rat lung adenocarcinomas induced by N-nitrosobis(2-hydroxypropyl)amine (BHP). Male Wistar rats, 6-wk-old, were given 2000 ppm BHP in their drinking water for 12 wk and maintained without further treatment until killed at wk 25. A total of 12 lung adenocarcinomas were obtained and total RNAs were extracted from each for assessment of aberrant transcripts of the Dutt1/Robo1 gene by reverse transcription (RT)-polymerase chain reaction (PCR) analysis. Aberrant transcripts bearing deletions of nucleotides (nt) 55-4318, 89-4346, 605-4221, and 929-4318 were detected in four of 12 adenocarcinomas (33.3%). Loss or reduced expression of the Dutt1/Robo1 gene was not found in any of the adenocarcinomas. Genomic DNAs extracted from six adenocarcinomas for Southern blot analysis did not show any evidence of deletion or gross rearrangement of the Dutt1/Robo1 gene. These results suggest that alterations of the Dutt1/Robo1 gene may be involved in the development of some lung adenocarcinomas induced by BHP in rats.
Collapse
Affiliation(s)
- Toshifumi Tsujiuchi
- Department of Oncological Pathology, Cancer Center, Nara Medical University, Kashihara, Nara, Japan
| | | | | | | | | |
Collapse
|
39
|
Amare Kadam PS, Ghule P, Jose J, Bamne M, Kurkure P, Banavali S, Sarin R, Advani S. Constitutional genomic instability, chromosome aberrations in tumor cells and retinoblastoma. ACTA ACUST UNITED AC 2004; 150:33-43. [PMID: 15041221 DOI: 10.1016/j.cancergencyto.2003.08.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2003] [Revised: 08/21/2003] [Accepted: 08/22/2003] [Indexed: 11/21/2022]
Abstract
Although retinoblastoma (Rb) is initiated as a result of biallelic inactivation of the RB1 gene, additional genetic events (M3) in tumor cells are indicative of their role in the full transformation of retinal cells. We investigated the constitutional genetic instability by fragile site (FS) expression studies and checked its relationship with loci of tumor cytogenetics in a series of 36 retinoblastoma patients (34 nonfamilial and 2 familial cases). Tumor cytogenetics revealed -13/+13, del/t(13)(q14) (50%), +1/del/t(1p/q) (65%), +6/i(6p) (60%), and del(16)(q13)/(q22 approximately q23) (60%). Conventional cytogenetics in leukocytes revealed constitutional del(13q14) in five unilateral Rb (URB) and one trilateral Rb (TRB). Constitutional del(16)(q22) and t(6;12) were also identified in two cases. Constitutional FS analysis showed a significant increase in the cellular fragility, with high prevalence at 13q14, 3p14, 6p23, 16q22 approximately q23, and 13q22 loci in retinoblastoma patients (P<0.05). Patients with constitutional del(13)(q14) demonstrated higher fragility than those with normal constitution. A strong correlation between loci of constitutional FSs and loci of recurrent chromosomal abnormalities in tumors strengthen and support the proposal that FS loci present as inherent genomic instability in retinoblastoma. The chromosomal changes and resultant genetic mutations, along with RB1 mutation events, probably contribute synergistically to the development and progression of Rb malignancy. Implementation of fluorescence in situ hybridization to nonfamilial Rb on a large scale (113 cases) could detect constitutional RB1 deletion in 12.3% of cases, with equally higher incidence in URB (14.7%) and bilateral Rb (13.6%), demonstrating that the true prevalence of patients with predisposition to RB1 mutation in sporadic URB is definitely higher in our populations. Also, higher incidence of constitutional RB1 deletion mosaicism in unilateral than in bilateral Rb indicates that the constitutional genetic mosaicism in URB should be given serious consideration during genetic counseling.
Collapse
Affiliation(s)
- P S Amare Kadam
- Cancer Cytogenetics Laboratory, Tata Memorial Hospital, Dr Ernest Borges Marg, Parel, Mumbai 400012, India.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Pisick E, Jagadeesh S, Salgia R. Small cell lung cancer: from molecular biology to novel therapeutics. JOURNAL OF EXPERIMENTAL THERAPEUTICS AND ONCOLOGY 2004; 3:305-18. [PMID: 14678519 DOI: 10.1111/j.1533-869x.2003.01103.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Small cell lung cancer (SCLC) is an aggressive tumor which metastasizes early. Patients with this disease have a poor prognosis even with immediate treatment. Because of the aggressive nature of this disease, all aspects of this tumor are studied extensively. This review will provide an update of the biology of SCLC at both the molecular and cellular levels. Cellular pathways and their relationship to cellular function will also be discussed. Treatment of both primary limited- and extensive-stage diseases as well as recurrent disease will be discussed including chemotherapy, thoracic radiotherapy, and surgery. The role of novel therapeutics being investigated will also be addressed.
Collapse
Affiliation(s)
- Evan Pisick
- Tufts- New England Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | | | | |
Collapse
|
41
|
Xue Jun H, Gemma A, Hosoya Y, Matsuda K, Nara M, Hosomi Y, Okano T, Kurimoto F, Seike M, Takenaka K, Yoshimura A, Toyota M, Kudoh S. Reduced transcription of the RB2/p130 gene in human lung cancer. Mol Carcinog 2003; 38:124-9. [PMID: 14587097 DOI: 10.1002/mc.10152] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Reduced expression of the retinoblastoma gene (RB)2/p130 protein, as well as mutation of exons 19, 20, 21, and 22 of the same gene, has been reported in primary lung cancer. However, it has been suggested by other investigators that mutational inactivation and loss of the RB2/p130 gene and protein, respectively, are rare events in lung cancer. In order to determine the contribution and mechanisms of RB2/p130 gene inactivation to lung cancer development and progression, we quantified RB2/p130 mRNA expression levels in a range of human lung cancer cell lines (n = 13) by real-time reverse transcription (RT)-polymerase chain reaction (PCR) analysis. In comparison to normal lung tissue, reduced transcription of the RB2/p130 gene was found in all small cell lung cancer cell lines examined, along with six out of the eight nonsmall cell lung cancers tested, most of which had inactivation of RB/p16 pathway. On the basis of Western blot analysis, the expression of RB2/p130 protein was consistent with RNA expression levels in all lung cancer cell lines examined. In addition, the mutational status of the RB2/p130 gene (specifically, exons 19, 20, 21, and 22) was determined in 30 primary lung cancers (from patients with distant metastasis) and 30 lung cancer cell lines by PCR-single strand conformation polymorphism (SSCP) analysis and direct DNA sequencing. There was no evidence of somatic mutations within the RB2/p130 gene in the 60 lung cancer samples (both cell lines and tumors) assessed, including the 11 lung cancer cell lines that displayed reduced expression of the gene. Furthermore, hypermethylation of the RB2/p130 promoter was not found in any of the above-mentioned 11 cell lines, as determined by a DNA methylation assay, combined bisulfite restriction analysis (COBRA). The results of the present study suggest that the reduced RB2/p130 expression seen in lung cancer may be in part transcriptionally mediated, albeit not likely via a mechanism involving hypermethylation of the RB2/p130 promoter. The observed reduction in RB2/p130 gene expression may be due to histone deacetylation, altered mRNA stability, and/or other forms of transcriptional regulation.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Small Cell/genetics
- Carcinoma, Small Cell/metabolism
- Carcinoma, Small Cell/pathology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Case-Control Studies
- DNA Methylation
- DNA Mutational Analysis
- DNA, Neoplasm/metabolism
- Exons
- Gene Expression Regulation, Neoplastic
- Humans
- Lung/metabolism
- Lung/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Mutation
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- Polymerase Chain Reaction
- Polymorphism, Single-Stranded Conformational
- Proteins
- Retinoblastoma Protein/genetics
- Retinoblastoma Protein/metabolism
- Retinoblastoma-Like Protein p130
- Transcription, Genetic
Collapse
Affiliation(s)
- Hu Xue Jun
- Fourth Department of Medicine, Nippon Medical School, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Fukami T, Fukuhara H, Kuramochi M, Maruyama T, Isogai K, Sakamoto M, Takamoto S, Murakami Y. Promoter methylation of the TSLC1 gene in advanced lung tumors and various cancer cell lines. Int J Cancer 2003; 107:53-9. [PMID: 12925956 DOI: 10.1002/ijc.11348] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We previously identified TSLC1, a tumor suppressor gene in human nonsmall cell lung cancer (NSCLC). TSLC1 belongs to immunoglobulin superfamily molecules and is involved in cell adhesion. Loss of TSLC1 expression was strongly correlated with the promoter hypermethylation in several NSCLC cell lines. Here, we examined the methylation status of the TSLC1 gene promoter in 48 primary NSCLC tumors by bisulfite SSCP in combination with bisulfite sequencing. Six CpG sites around the promoter regions were significantly methylated in 21 of 48 primary NSCLC tumors (44%). Promoter methylation was more likely to be observed in relatively advanced tumors with TNM classification of pT2, pT3 or pT4 (19 of 33, 58%) than in those with pT1 (2 of 15, 13%), suggesting that alteration of TSLC1 would be involved in the progression of human NSCLC. Loss of TSLC1 expression was also observed in 20 of 46 (43%) human cancer cell lines, including those from esophageal (3 of 3), gastric (8 of 9), ovarian (2 of 5), endometrial (2 of 2), breast (1 of 3), colorectal (2 of 8) and small cell lung cancers (2 of 10). Combined analysis of promoter methylation and the allelic state in these cell lines indicated that the TSLC1 gene was often silenced not only by mono-allelic methylation associated with loss of the other allele but also through bi-allelic methylation. These results suggest that alteration of TSLC1 would be involved in advanced NSCLC as well as in many other human cancers.
Collapse
Affiliation(s)
- Takeshi Fukami
- Tumor Suppression & Functional Genomics Project, University of Tokyo, School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Uchino K, Ito A, Wakayama T, Koma YI, Okada T, Ohbayashi C, Iseki S, Kitamura Y, Tsubota N, Okita Y, Okada M. Clinical implication and prognostic significance of the tumor suppressor TSLC1 gene detected in adenocarcinoma of the lung. Cancer 2003; 98:1002-7. [PMID: 12942568 DOI: 10.1002/cncr.11599] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Recently, the TSLC1 (tumor suppressor in lung cancer 1) gene has been identified as a novel tumor suppressor in human nonsmall cell lung carcinoma. To the authors' knowledge, the clinical relevance of TSLC1 gene expression has not been studied using patient data and surgical samples. The current study was designed to evaluate whether the TSLC1 gene can serve as a target for the prognostic determination of patients with pulmonary adenocarcinoma. METHODS A total of 38 patients who were surgically treated for proven primary lung adenocarcinoma were enrolled in the current study. Surgical specimens were examined for TSLC1 protein expression immunohistochemically and by Western blot analysis. The correlation between levels of TSLC1 expression and pathologic characteristics, as well as prognosis, was investigated. RESULTS All patients underwent a potentially curative resection of their tumor. TSLC1 antigen expression as evaluated by immunohistochemistry was confirmed by immunoblotting. The expression of TSLC1 protein was found to be inversely correlated with advanced disease stage, lymph node involvement, lymphatic permeation, and vascular invasion. The 4-year overall survival rates of patients with a tumor demonstrating high (> 70% positive cells [n = 14 patients]), intermediate (20-70% positive cells [n = 10 patients]), and low (< 20% positive cells [n = 14 patients]) expression of the TSLC1 antigen were 84%, 28%, and 7%, respectively. In addition, the disease-free survival of patients with a tumor that demonstrated a high percentage of TSLC1 protein-positive cells was reported to be significantly better than that of patients with a tumor that showed a low percentage of TSLC1 protein-positive cells. CONCLUSIONS The loss or reduction of TSLC1 expression in resected lung adenocarcinoma cases was associated with a poor prognosis, indicating that TSLC1 represents a central effector gene for controlling the biologic aggressiveness of the tumor and that it is an essential biomarker for predicting patient prognosis. These data may help to detect those patients at high risk for recurrence who might benefit from additional therapeutic strategies such as adjuvant therapy.
Collapse
Affiliation(s)
- Kazuya Uchino
- Department of Cardiothoracic Surgery, Kobe University Medical School, Kobe, Hyogo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Tsujiuchi T, Sasaki Y, Kubozoe T, Konishi Y, Tsutsumi M. Alterations in the Fhit gene in pancreatic duct adenocarcinomas induced by N-nitrosobis(2-oxopropyl)amine in hamsters. Mol Carcinog 2003; 36:60-6. [PMID: 12557261 DOI: 10.1002/mc.10099] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Alteration of the Fhit gene was investigated in pancreatic duct adenocarcinomas induced by N-nitrosobis(2-oxopropyl)amine (BOP) in Syrian golden hamsters. The animals received 70 mg/kg BOP, followed by repeated exposure to an augmentation pressure regimen consisting of a choline-deficient diet combined with DL-ethionine and then L-methionine and administration of 20 mg/kg BOP. A total of 15 pancreatic duct adenocarcinomas were obtained 10 wk after the beginning of the experiment, and total RNAs were extracted from each for assessment of aberrant transcription of the Fhit gene by reverse transcription-polymerase chain reaction analysis. Aberrant transcripts lacking nucleotides in the regions of nt -75 to 348, nt -15 to 348, or nt -75 to 178 were detected in 11 adenocarcinomas (73.3%). Southern blot analysis of eight tumors did not show any evidence of gross rearrangement or deletion. These results indicated that changes in the Fhit gene occurred frequently and thus may have played a role in the development of pancreatic duct adenocarcinomas induced by BOP in hamsters.
Collapse
Affiliation(s)
- Toshifumi Tsujiuchi
- Department of Oncological Pathology, Cancer Center, Nara Medical University, Kashihara, Nara, Japan
| | | | | | | | | |
Collapse
|
45
|
Abstract
For any tumor to become cancerous, various genetic mutations and biologic alterations must occur in the cell that in combination render it a malignant neoplasm. Small cell lung cancer (SCLC) is a neoplasm associated with several molecular and cellular abnormalities. SCLC is associated with early and frequent metastasis as well as a poor ultimate response to chemotherapy. New and novel therapies based on understanding the mechanisms of transformation are needed. SCLC is associated with multiple chromosomal abnormalities, the most common of which is chromosome 3p deletion, as well as with abnormal oncogenes and tumor-suppressor genes. Along with the genetic alterations, SCLC has been shown to overexpress various cell surface receptors, including receptor tyrosine kinases (RTKs), G-protein-coupled receptors, integrins, and others. Some downstream molecules are also activated, such as phosphatidylinositol 3'-kinase, and would serve as good candidates for therapeutic strategies.
Collapse
Affiliation(s)
- Martin Sattler
- Department of Medical Oncology, Division of Thoracic Oncology Program, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
46
|
Abstract
A wealth of cytogenetic data has demonstrated that numerous somatic genetic changes are involved in the pathogenesis of human lung cancer. Despite the complexity of the genomic changes observed in these neoplasms, recurrent chromosomal patterns have emerged. In this review, we summarize chromosomal alterations identified in small cell and non-small cell lung cancer, using classical and molecular cytogenetic techniques. These analyses have uncovered a set of chromosome regions implicated in lung cancer development and progression. However, many of the target genes remain unknown. Newer technology, such as array-CGH, when combined with cDNA microarrays and tissue microarrays, will facilitate the integration of genomic and gene expression data and pave the way toward a molecular classification of lung carcinomas. The molecular implications of consistent chromosome imbalances found in lung cancer to date are also discussed.
Collapse
Affiliation(s)
- Binaifer R Balsara
- Human Genetics Program, Fox Chase Cancer Center, 7701 Burholme Avenue, Philadelphia, Pennsylvania 19111, USA
| | | |
Collapse
|
47
|
Sekido Y, Sato M, Usami N, Shigemitsu K, Mori S, Maeda O, Yokoi T, Hasegawa Y, Yoshioka H, Shimokata K. Establishment of a large cell lung cancer cell line (Y-ML-1B) producing granulocyte colony-stimulating factor. CANCER GENETICS AND CYTOGENETICS 2002; 137:33-42. [PMID: 12377411 DOI: 10.1016/s0165-4608(02)00536-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We established a new lung cancer cell line, designated Y-ML-1B, from a lung cancer of a 70-year-old Japanese man with leukocytosis and thrombocytosis. Before surgical resection, the white blood cell and platelet counts were elevated to 34,400/mm3 and 668,000/mm3, respectively, and the granulocyte colony-stimulating factor (G-CSF) level in the serum was increased at 141 pg/mL. The primary tumor showed an undifferentiated morphology with large cells and induced extensive thickening of the pleura in the right hemithorax. The Y-ML-1B cells grow as a monolayer, with a doubling time of 19 hours, and are tumorigenic in nude mice, which showed a morphology similar to the primary tumor in xenografts. Analysis of the supernatant of cell culture medium of Y-ML-1B showed elevated levels of G-CSF and other cytokines such as interleukin (IL)-6, IL-8, and granulocyte-macrophage colony-stimulating factor (GM-CSF), consistent with the high levels detected in the patient's serum. Cytogenetic analysis revealed aneuploidy of greater than 56 in metaphases with many structural abnormalities. Mutation analysis of the tumor suppressor genes showed that Y-ML-1B is inactivated in TP53 and RASSF1A, but not in p14(ARF), p16(INK4A), or RB. Neither activating mutations of KRAS or NRAS nor amplification of MYC or MDM2 were detected. Y-ML-1B expressed N-cadherin but not E-cadherin. This newly established cell line might serve as a useful model for studying the molecular pathogenesis for large cell cancers of the lung which express high levels of cytokines.
Collapse
Affiliation(s)
- Yoshitaka Sekido
- Department of Clinical Preventive Medicine, Nagoya University School of Medicine, 466-8560, Nagoya, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bourdon JC, Renzing J, Robertson PL, Fernandes KN, Lane DP. Scotin, a novel p53-inducible proapoptotic protein located in the ER and the nuclear membrane. J Cell Biol 2002; 158:235-46. [PMID: 12135983 PMCID: PMC2173124 DOI: 10.1083/jcb.200203006] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
p53 is a transcription factor that induces growth arrest or apoptosis in response to cellular stress. To identify new p53-inducible proapoptotic genes, we compared, by differential display, the expression of genes in spleen or thymus of normal and p53 nullizygote mice after gamma-irradiation of whole animals. We report the identification and characterization of human and mouse Scotin homologues, a novel gene directly transactivated by p53. The Scotin protein is localized to the ER and the nuclear membrane. Scotin can induce apoptosis in a caspase-dependent manner. Inhibition of endogenous Scotin expression increases resistance to p53-dependent apoptosis induced by DNA damage, suggesting that Scotin plays a role in p53-dependent apoptosis. The discovery of Scotin brings to light a role of the ER in p53-dependent apoptosis.
Collapse
Affiliation(s)
- J-C Bourdon
- Department of Surgery and Molecular Oncology, Ninewells Hospital and Medical School, Cancer Research Campaign (CRC) Cell Transformation Research Group, University of Dundee, Dundee DD1 9SY, Scotland, UK.
| | | | | | | | | |
Collapse
|
49
|
Tsujiuchi T, Sasaki Y, Kubozoe T, Tsutsumi M, Konishi Y, Nakae D. Alterations of the Fhit gene in hepatocellular carcinomas induced by N-nitrosodiethylamine in rats. Mol Carcinog 2002; 34:19-24. [PMID: 12112319 DOI: 10.1002/mc.10044] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The present study was conducted to assess whether Fhit gene alterations are a feature of hepatocellular carcinomas (HCCs) induced by N-nitrosodiethylamine (DEN) in male Fischer 344 rats. Animals, 6 wk old, received a single intraperitoneal injection of DEN at a dose of 10 mg/kg body weight, followed by combined treatment with partial hepatectomy and colchicine to induce cell-cycle disturbance and a selection procedure, consisting of 2-acetylaminofluorene and carbon tetrachloride. Fourteen HCCs were obtained 42 wk after the beginning of the experiment; total RNA was extracted for the assessment of aberrant transcription of the Fhit gene by reverse transcriptase-polymerase chain reaction analysis. Aberrant transcripts were detected in nine of the 14 HCCs (64.3%). Sequence analysis showed that these resulted from the absence of nt -9 to 279, nt -9 to 348, nt -98 to 279, nt -26 to 365, or nt -98 to 348. Western blot analysis demonstrated reduced expression of Fhit protein in six of 10 HCCs (60.0%), with a perfect correlation with Fhit gene alterations. These results indicated that changes in the Fhit gene occur frequently and may thus play some role in the development of HCCs induced by DEN in rats.
Collapse
Affiliation(s)
- Toshifumi Tsujiuchi
- Department of Oncological Pathology, Cancer Center, Nara Medical University, Kashihara, Nara, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Morrison BH, Bauer JA, Hu J, Grane RW, Ozdemir AM, Chawla-Sarkar M, Gong B, Almasan A, Kalvakolanu DV, Lindner DJ. Inositol hexakisphosphate kinase 2 sensitizes ovarian carcinoma cells to multiple cancer therapeutics. Oncogene 2002; 21:1882-9. [PMID: 11896621 PMCID: PMC2043497 DOI: 10.1038/sj.onc.1205265] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2001] [Revised: 12/11/2001] [Accepted: 12/18/2001] [Indexed: 12/29/2022]
Abstract
We recently identified inositol hexakisphosphate kinase 2 (IP6K2) as a positive regulator of apoptosis. Overexpression of IP6K2 enhances apoptosis induced by interferon-beta (IFN-beta) and cytotoxic agents in NIH-OVCAR-3 ovarian carcinoma cells. In this study, we contrast and compare IFN-beta and radiation-induced death, and show that IP6K2 expression sensitizes tumor cells. Unirradiated NIH-OVCAR-3 cells transfected with IP6K2 formed fewer colonies compared to unirradiated vector-expressing cells. IP6K2 overexpression caused increased radiosensitivity, evidenced by decreased colony forming units (CFU). Both IFN-beta and radiation induced caspase 8. IFN-beta, but not gamma-irradiation, induced TRAIL in NIH-OVCAR-3 cells. Gamma irradiation, but not IFN-beta, induced DR4 mRNA. Apoptotic effects of IFN-beta or gamma-irradiation were blocked by expression of a dominant negative mutant death receptor 5 (DR5Delta) or by Bcl-2. Caspase-8 mRNA induction was more pronounced in IP6K2-expressing cells compared to vector-expressing cells. These data suggest that overexpression of IP6K2 enhances sensitivity of some ovarian carcinomas to radiation and IFN-beta. IP6K2 may function to enhance the expression and/or function of caspase 8 and DR4 following cell injury. Both IFN-beta and gamma-irradiation induce apoptosis through the extrinsic, receptor-mediated pathway, IFN-beta through TRAIL, radiation through DR4, and both through caspase 8. The function of both death inducers is positively regulated by IP6K2.
Collapse
Affiliation(s)
- Bei H Morrison
- Center for Cancer Drug Discovery and Development, Taussig Cancer Center, The Cleveland Clinic Foundation, Cleveland, Ohio, OH 44195, USA
| | - Joseph A Bauer
- Center for Cancer Drug Discovery and Development, Taussig Cancer Center, The Cleveland Clinic Foundation, Cleveland, Ohio, OH 44195, USA
| | - Jiadi Hu
- Department of Microbiology and Immunology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201, USA
| | - Ronald W Grane
- Center for Cancer Drug Discovery and Development, Taussig Cancer Center, The Cleveland Clinic Foundation, Cleveland, Ohio, OH 44195, USA
| | - Aylin M Ozdemir
- Center for Cancer Drug Discovery and Development, Taussig Cancer Center, The Cleveland Clinic Foundation, Cleveland, Ohio, OH 44195, USA
| | - Mamta Chawla-Sarkar
- Center for Cancer Drug Discovery and Development, Taussig Cancer Center, The Cleveland Clinic Foundation, Cleveland, Ohio, OH 44195, USA
| | - Bendi Gong
- Department of Cancer Biology, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, Ohio, OH 44195, USA
| | - Alex Almasan
- Department of Cancer Biology, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, Ohio, OH 44195, USA
- Department of Radiation Oncology, The Cleveland Clinic Foundation, Cleveland, Ohio, OH 44195, USA
| | - Dhananjaya V Kalvakolanu
- Department of Microbiology and Immunology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201, USA
| | - Daniel J Lindner
- Center for Cancer Drug Discovery and Development, Taussig Cancer Center, The Cleveland Clinic Foundation, Cleveland, Ohio, OH 44195, USA
- Department of Cancer Biology, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, Ohio, OH 44195, USA
- Correspondence: DJ Lindner, 9500 Euclid Avenue, R40, Cleveland, OH 44195, USA; E-mail:
| |
Collapse
|