1
|
Tao S, Pu Y, Yang EJ, Ren G, Shi C, Chen LJ, Chen L, Shim JS. Inhibition of GSK3β is synthetic lethal with FHIT loss in lung cancer by blocking homologous recombination repair. Exp Mol Med 2025; 57:167-183. [PMID: 39762409 PMCID: PMC11799392 DOI: 10.1038/s12276-024-01374-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/06/2024] [Accepted: 10/06/2024] [Indexed: 02/07/2025] Open
Abstract
FHIT is a fragile site tumor suppressor that is primarily inactivated upon tobacco smoking. FHIT loss is frequently observed in lung cancer, making it an important biomarker for the development of targeted therapy for lung cancer. Here, we report that inhibitors of glycogen synthase kinase 3 beta (GSK3β) and the homologous recombination DNA repair (HRR) pathway are synthetic lethal with FHIT loss in lung cancer. Pharmacological inhibition or siRNA depletion of GSK3β selectively suppressed the growth of FHIT-deficient lung cancer tumors in vitro and in animal models. We further showed that FHIT inactivation leads to the activation of DNA damage repair pathways, including the HRR and NHEJ pathways, in lung cancer cells. Conversely, FHIT-deficient cells are highly dependent on HRR for survival under DNA damage stress. The inhibition of GSK3β in FHIT-deficient cells suppressed the ATR/BRCA1/RAD51 axis in HRR signaling via two distinct pathways and suppressed DNA double-strand break repair, leading to the accumulation of DNA damage and apoptosis. Small molecule inhibitors of HRR, but not NHEJ or PARP, induced synthetic lethality in FHIT-deficient lung cancer cells. The findings of this study suggest that the GSK3β and HRR pathways are potential drug targets in lung cancer patients with FHIT loss.
Collapse
Affiliation(s)
- Shishi Tao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Yue Pu
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Eun Ju Yang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Guowen Ren
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518055, Guangdong, China
| | - Changxiang Shi
- Nanjing Key Laboratory of Female Fertility Preservation and Restoration, Nanjing Women and Children's Healthcare Institute, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing, 210004, China
| | - Li-Jie Chen
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Liang Chen
- Shenzhen Laboratory of Tumor Cell Biology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Joong Sup Shim
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
- MOE Frontiers Science Centre for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
2
|
Zhang X, Felter SP, Api AM, Joshi K, Selechnik D. A Cautionary tale for using read-across for cancer hazard classification: Case study of isoeugenol and methyl eugenol. Regul Toxicol Pharmacol 2022; 136:105280. [DOI: 10.1016/j.yrtph.2022.105280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/16/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
|
3
|
Bellon M, Bialuk I, Galli V, Bai XT, Farre L, Bittencourt A, Marçais A, Petrus MN, Ratner L, Waldmann TA, Asnafi V, Gessain A, Matsuoka M, Franchini G, Hermine O, Watanabe T, Nicot C. Germinal epimutation of Fragile Histidine Triad (FHIT) gene is associated with progression to acute and chronic adult T-cell leukemia diseases. Mol Cancer 2021; 20:86. [PMID: 34092254 PMCID: PMC8183032 DOI: 10.1186/s12943-021-01370-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human T cell Leukemia virus type 1 (HTLV-I) is etiologically linked to adult T cell leukemia/lymphoma (ATL) and an inflammatory neurodegenerative disease called HTLV-I-associated myelopathy or tropical spastic paraparesis (HAM/TSP). The exact genetic or epigenetic events and/or environmental factors that influence the development of ATL, or HAM/TSP diseases are largely unknown. The tumor suppressor gene, Fragile Histidine Triad Diadenosine Triphosphatase (FHIT), is frequently lost in cancer through epigenetic modifications and/or deletion. FHIT is a tumor suppressor acting as genome caretaker by regulating cellular DNA repair. Indeed, FHIT loss leads to replicative stress and accumulation of double DNA strand breaks. Therefore, loss of FHIT expression plays a key role in cellular transformation. METHODS Here, we studied over 400 samples from HTLV-I-infected individuals with ATL, TSP/HAM, or asymptomatic carriers (AC) for FHIT loss and expression. We examined the epigenetic status of FHIT through methylation specific PCR and bisulfite sequencing; and correlated these results to FHIT expression in patient samples. RESULTS We found that epigenetic alteration of FHIT is specifically found in chronic and acute ATL but is absent in asymptomatic HTLV-I carriers and TSP/HAM patients' samples. Furthermore, the extent of FHIT methylation in ATL patients was quantitatively comparable in virus-infected and virus non-infected cells. We also found that longitudinal HTLV-I carriers that progressed to smoldering ATL and descendants of ATL patients harbor FHIT methylation. CONCLUSIONS These results suggest that germinal epigenetic mutation of FHIT represents a preexisting mark predisposing to the development of ATL diseases. These findings have important clinical implications as patients with acute ATL are rarely cured. Our study suggests an alternative strategy to the current "wait and see approach" in that early screening of HTLV-I-infected individuals for germinal epimutation of FHIT and early treatment may offer significant clinical benefits.
Collapse
Affiliation(s)
- Marcia Bellon
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Izabela Bialuk
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Veronica Galli
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xue-Tao Bai
- Comprehensive Cancer Center, Department of Health Sciences, Ohio State University, Columbus, OH, USA
| | - Lourdes Farre
- Program in Molecular Mechanisms and Experimental Therapy in Oncology, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Achilea Bittencourt
- Department of Pathology, Prof. Edgard Santos Teaching Hospital, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Ambroise Marçais
- Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151, Laboratoire Onco-Hématologie, Paris, France
| | - Michael N Petrus
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lee Ratner
- Division of Oncology, Department of Medicine, Washington University, St Louis, MO, USA
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vahid Asnafi
- Université de Paris (Descartes), Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151 Laboratoire Onco-Hematology, Paris, France
| | - Antoine Gessain
- Unité d'épidémiologie et de Physiopathologie des virus Oncogene, Institut Pasteur, 75015, Paris, France.,Centre National de la Recherche Scientifique (CNRS) UMR 3569, 75015, Paris, France
| | - Masao Matsuoka
- Laboratory of Virus Control, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Hematology, Rheumatology, and Infectious Disease, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Genoveffa Franchini
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Olivier Hermine
- Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151, Laboratoire Onco-Hématologie, Paris, France
| | - Toshiki Watanabe
- Department of Hematology/Oncology, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Christophe Nicot
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
4
|
Chae HJ, Seo JB, Kim SH, Jeon YJ, Suh SS. Fhit induces the reciprocal suppressions between Lin28/Let-7 and miR-17/92miR. Int J Med Sci 2021; 18:706-714. [PMID: 33437205 PMCID: PMC7797533 DOI: 10.7150/ijms.51429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Objective: Fhit gene is known as a genome "caretaker" and frequently inactivated by deletion or hypermethylation on the promoter in several cancers. In spite of several lines of evidence, the exact mechanism underlying Fhit-induced biology is relatively less studied. This study will focus the role of Fhit in regulating Lin28 and microRNAs (miRNAs) loop. Material and Methods: To this end, we employed Fhit overexpressing isogenic cell lines to conduct miRNA nanostring array, and differentially expressed miRNAs were identified. Using real-time PCR and Western blot analysis, expression levels of Lin28b or miRNAs were investigated in response to the overexpression of Fhit gene in H1299 lung cancer cells. Results: A series of in vitro including gene nanostring analyses revealed that Lin28B protein was induced by Fhit gene overexpression, which consequently suppressed Let-7 miRNAs. Also, we found that miRNAs in miR-17/92 clusters are redundantly increased and there is an inverse correlation between Let-7 and miR-17/92 clusters in Fhit-expressing cells. Also, a series of in vitro experiments suggests that ELF-1- and/or STAT1-dependent Lin28b regulation is responsible for Let-7 induction in Fhit-expressing cancer cells. Conclusions: Based on the same experimental system proving that Fhit gene has a robust role in suppressing tumor progression and epithelial-mesenchymal transition, our data show that Fhit mediates the negative feedback between Lin28/Let-7 axis and miR-17/-92 miRNA although the physiological relevance of current interesting observation should be further investigated.
Collapse
Affiliation(s)
- Hae-Jung Chae
- Department of Biosciences, Mokpo National University, Joennam 58554, South Korea
| | - Jong Bae Seo
- Department of Biosciences, Mokpo National University, Joennam 58554, South Korea.,Department of Biomedicine, Health & Life Convergence Science, BK21 Four, Mokpo National University, Joennam 58554, South Korea
| | - Sung-Hak Kim
- Lab of Animal Molecular Biochemistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, South Korea
| | - Sung-Suk Suh
- Department of Biosciences, Mokpo National University, Joennam 58554, South Korea.,Department of Biomedicine, Health & Life Convergence Science, BK21 Four, Mokpo National University, Joennam 58554, South Korea
| |
Collapse
|
5
|
Role of HSP60/HSP10 in Lung Cancer: Simple Biomarkers or Leading Actors? JOURNAL OF ONCOLOGY 2020; 2020:4701868. [PMID: 32318107 PMCID: PMC7149434 DOI: 10.1155/2020/4701868] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 02/22/2020] [Accepted: 03/02/2020] [Indexed: 02/08/2023]
Abstract
Cancers are one of the major challenges faced by modern medicine both because of their impact in terms of the amount of cases and of the ineffectiveness of therapies used today. A concrete support to the fight against them can be found in the analysis and understanding of the molecular mechanisms involving molecular chaperones. In particular, HSP60 and HSP10 seem to play an important role in carcinogenesis, supporting tumours in their proliferation, survival, and metastasis. Efforts must be directed toward finding ways to eliminate or block this "mistaken" chaperone. Therefore, the scientific community must develop therapeutic strategies that consider HSP60 and HSP10 as the possible target of an anti-tumoural treatment and not only as diagnostic biomarkers, since they contribute to the evolution of pre-cancerous respiratory pathologies in lung tumours. HSP60 acts at the mitochondrial, cytoplasmic, and extracellular levels in the development of cancer pathologies. The molecular mechanisms in which these chaperones are involved concern cell survival, the restoration of a condition of absence of replicative senescence, the promotion of pro-inflammatory environments, and an increase in the ability to form metastases. In this review, we will also present examples of interactions between HSP60 and HSP10 and different molecules and ways to exploit this knowledge in anticancer therapies for lung tumours. In order to improve not only chances for an earlier diagnosis but also treatments for patients suffering from this type of disease, chaperones must be considered as key agents in carcinogenesis and primary targets in therapeutics.
Collapse
|
6
|
Kiss DL, Baez W, Huebner K, Bundschuh R, Schoenberg DR. Impact of FHIT loss on the translation of cancer-associated mRNAs. Mol Cancer 2017; 16:179. [PMID: 29282095 PMCID: PMC5745650 DOI: 10.1186/s12943-017-0749-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/10/2017] [Indexed: 11/30/2022] Open
Abstract
Background FHIT is a genome caretaker/tumor suppressor that is silenced in >50% of cancers. Although it was identified more than 20 years ago, questions remain as to how FHIT loss contributes to cancer, and conversely, how FHIT acts to maintain genome integrity and suppress malignancy. Fhit belongs to the histidine triad family of enzymes that catalyze the degradation of nucleoside 5′,5′-triphosphates, including the m7GpppN ‘caps’ that are generated when mRNAs undergo 3′-5′ decay. This raised the possibility that Fhit loss might affect changes in the translation of cancer-associated mRNAs, possibly as a consequence of increased intracellular concentrations of these molecules. Results Ribosome profiling identified several hundred mRNAs for which coding region ribosome occupancy changed as a function of Fhit expression. While many of these changes could be explained by changes in mRNA steady-state, a subset of these showed changes in translation efficiency as a function of Fhit expression. The onset of malignancy has been linked to changes in 5’-UTR ribosome occupancy and this analysis also identified ribosome binding to 5′-untranslated regions (UTRs) of a number of cancer-associated mRNAs. 5’-UTR ribosome occupancy of these mRNAs differed between Fhit-negative and Fhit-positive cells, and in some cases these differences correlated with differences in coding region ribosome occupancy. Conclusions In summary, these findings show Fhit expression impacts the translation of a number of cancer associated genes, and they support the hypothesis that Fhit’s genome protective/tumor suppressor function is associated with post-transcriptional changes in expression of genes whose dysregulation contributes to malignancy. Electronic supplementary material The online version of this article (10.1186/s12943-017-0749-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel L Kiss
- Center for RNA Biology, The Ohio State University, Columbus, OH, 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.,Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA.,Biomarker Research Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - William Baez
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA
| | - Kay Huebner
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.,Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA
| | - Ralf Bundschuh
- Center for RNA Biology, The Ohio State University, Columbus, OH, 43210, USA.,Department of Physics, The Ohio State University, Columbus, OH, 43210, USA.,Department of Chemistry & Biochemistry, The Ohio State University, Columbus, OH, 43210, USA.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Daniel R Schoenberg
- Center for RNA Biology, The Ohio State University, Columbus, OH, 43210, USA. .,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA. .,Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
7
|
Fassan M, Rusev B, Corbo V, Gasparini P, Luchini C, Vicentini C, Mafficini A, Paiella S, Salvia R, Cataldo I, Scarpa A, Huebner K. Fhit down-regulation is an early event in pancreatic carcinogenesis. Virchows Arch 2017; 470:647-653. [PMID: 28289900 PMCID: PMC5568551 DOI: 10.1007/s00428-017-2105-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 02/22/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
Aberrant Fhit expression characterizes a large proportion of primary pancreatic ductal adenocarcinomas (PDACs), but fragmentary information is available on Fhit expression during the phenotypic changes of pancreatic ductal epithelium during multistep transformation. We assessed Fhit expression by immunohistochemistry in two different multistep pancreatic carcinogenic processes: pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasia (IPMN). We considered 105 surgically treated PDACs/IPMNs and selected 30 samples of non-neoplastic pancreatic parenchyma, 50 PanIN lesions, 30 IPMNs, 15 IPMNs with associated invasive carcinoma, and 60 adenocarcinomas. Normal pancreatic ducts and surrounding acinar cells consistently showed moderate to strong Fhit immunoreactivity. Significant down-regulation of Fhit expression was observed in association with increasing severity of dysplastia/neoplastia in both carcinogenic processes. This was further confirmed by studying multiple lesions obtained from the same surgical specimen. Of 60 PDACs, only 14 showed Fhit expression comparable to normal pancreatic ductal epithelium, while the remainder (77%) showed clearly negative or reduced Fhit expression. This study demonstrates that Fhit down-regulation is an early event in both multistep carcinogenic processes leading to PDAC.
Collapse
Affiliation(s)
- Matteo Fassan
- ARC-NET Research Centre, Department of Diagnostics and Public Health, University of Verona, Verona, Italy.
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Via Gabelli 61, 35121, Padua, Italy.
| | - Borislav Rusev
- ARC-NET Research Centre, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Vincenzo Corbo
- ARC-NET Research Centre, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Pierluigi Gasparini
- Comprehensive Cancer Center, Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Claudio Luchini
- ARC-NET Research Centre, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- Department of Diagnostics and Public Health, Surgical Pathology Unit, University and Hospital Trust of Verona, Verona, Italy
- Department of Pathology, Santa Chiara Hospital, Trento, Italy
| | - Caterina Vicentini
- ARC-NET Research Centre, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Andrea Mafficini
- ARC-NET Research Centre, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Salvatore Paiella
- Department of Surgery, Unit of General Surgery B, University and Hospital Trust of Verona, Verona, Italy
| | - Roberto Salvia
- Department of Surgery, Unit of General Surgery B, University and Hospital Trust of Verona, Verona, Italy
| | - Ivana Cataldo
- ARC-NET Research Centre, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Aldo Scarpa
- ARC-NET Research Centre, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- Department of Diagnostics and Public Health, Surgical Pathology Unit, University and Hospital Trust of Verona, Verona, Italy
| | - Kay Huebner
- Comprehensive Cancer Center, Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
8
|
Kiss DL, Waters CE, Ouda IM, Saldivar JC, Karras JR, Amin ZA, Mahrous S, Druck T, Bundschuh RA, Schoenberg DR, Huebner K. Identification of Fhit as a post-transcriptional effector of Thymidine Kinase 1 expression. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:374-382. [PMID: 28093273 DOI: 10.1016/j.bbagrm.2017.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 11/30/2022]
Abstract
FHIT is a genome caretaker gene that is silenced in >50% of cancers. Loss of Fhit protein expression promotes accumulation of DNA damage, affects apoptosis and epithelial-mesenchymal transition, though molecular mechanisms underlying these alterations have not been fully elucidated. Initiation of genome instability directly follows Fhit loss and the associated reduced Thymidine Kinase 1 (TK1) protein expression. The effects on TK1 of Fhit knockdown and Fhit induction in the current study confirmed the role of Fhit in regulating TK1 expression. Changes in Fhit expression did not impact TK1 protein turnover or transcription from the TK1 promoter, nor steady-state levels of TK1 mRNA or turnover. Polysome profile analysis showed that up-regulated Fhit expression resulted in decreased TK1 RNA in non-translating messenger ribonucleoproteins and increased ribosome density on TK1 mRNA. Fhit does not bind RNA but its expression increased luciferase expression from a transgene bearing the TK1 5'-UTR. Fhit has been reported to act as a scavenger decapping enzyme, and a similar result with a mutant (H96) that binds but does not cleave nucleoside 5',5'-triphosphates suggests the impact on TK1 translation is due to its ability to modulate the intracellular level of cap-like molecules. Consistent with this, cells expressing Fhit mutants with reduced activity toward cap-like dinucleotides exhibit DNA damage resulting from TK1 deficiency, whereas cells expressing wild-type Fhit or the H96N mutant do not. The results have implications for the mechanism by which Fhit regulates TK1 mRNA, and more broadly, for its modulation of multiple functions as tumor suppressor/genome caretaker.
Collapse
Affiliation(s)
- Daniel L Kiss
- Center for RNA Biology, 484 West 12th Ave., Columbus, OH 43210 USA
- Department of Biological Chemistry and Pharmacology, The Ohio State University, 1060 Carmack Rd., Columbus, OH 43210 USA
| | - Catherine E Waters
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12 Ave., Columbus, OH 43210 USA
| | - Iman M Ouda
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12 Ave., Columbus, OH 43210 USA
- Department of Clinical Pathology, Faculty of Medicine, 2nd floor, Surgery Hospital, Zagazig University, Zagazig 44519, Egypt
| | - Joshua C Saldivar
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12 Ave., Columbus, OH 43210 USA
| | - Jenna R Karras
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12 Ave., Columbus, OH 43210 USA
| | - Zaynab A Amin
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12 Ave., Columbus, OH 43210 USA
| | - Seham Mahrous
- Department of Clinical Pathology, Faculty of Medicine, 2nd floor, Surgery Hospital, Zagazig University, Zagazig 44519, Egypt
| | - Teresa Druck
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12 Ave., Columbus, OH 43210 USA
| | - Ralf A Bundschuh
- Center for RNA Biology, 484 West 12th Ave., Columbus, OH 43210 USA
- Department of Physics, Department of Chemistry and Biochemistry, and Division of Hematology, Department of Internal Medicine, The Ohio State University, 191 West Woodruff Ave., Columbus, OH 43210 USA
| | - Daniel R Schoenberg
- Center for RNA Biology, 484 West 12th Ave., Columbus, OH 43210 USA
- Department of Biological Chemistry and Pharmacology, The Ohio State University, 1060 Carmack Rd., Columbus, OH 43210 USA
| | - Kay Huebner
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12 Ave., Columbus, OH 43210 USA
| |
Collapse
|
9
|
Gaudio E, Paduano F, Croce CM, Trapasso F. The Fhit protein: an opportunity to overcome chemoresistance. Aging (Albany NY) 2016; 8:3147-3150. [PMID: 27852977 PMCID: PMC5191891 DOI: 10.18632/aging.101123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/10/2016] [Indexed: 11/25/2022]
Affiliation(s)
- Eugenio Gaudio
- Department of Molecular Immunology, Virology and Medical Genetics, The Ohio State University, Columbus, OH, USA
- Lymphoma & Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
- Dipartimento di Medicina Sperimentale e Clinica, University , Catanzaro, Italy
| | - Francesco Paduano
- Dipartimento di Medicina Sperimentale e Clinica, University , Catanzaro, Italy
- Tecnologica Research Institute, Biomedical Section, Crotone, Italy
| | - Carlo M Croce
- Department of Molecular Immunology, Virology and Medical Genetics, The Ohio State University, Columbus, OH, USA
| | - Francesco Trapasso
- Dipartimento di Medicina Sperimentale e Clinica, University , Catanzaro, Italy
| |
Collapse
|
10
|
Karras JR, Schrock MS, Batar B, Zhang J, La Perle K, Druck T, Huebner K. Fhit loss-associated initiation and progression of neoplasia in vitro. Cancer Sci 2016; 107:1590-1598. [PMID: 27513973 PMCID: PMC5132276 DOI: 10.1111/cas.13032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/05/2016] [Accepted: 08/09/2016] [Indexed: 12/28/2022] Open
Abstract
The FHIT gene, encompassing an active common fragile site, FRA3B, is frequently silenced in preneoplasia and cancer, through gene rearrangement or methylation of regulatory sequences. Silencing of Fhit protein expression causes thymidine kinase 1 downregulation, resulting in dNTP imbalance, and spontaneous replication stress that leads to chromosomal aberrations, allele copy number variations, insertions/deletions, and single-base substitutions. Thus, Fhit, which is reduced in expression in the majority of human cancers, is a genome "caretaker" whose loss initiates genome instability in preneoplastic lesions. To follow the early genetic alterations and functional changes induced by Fhit loss that may recapitulate the neoplastic process in vitro, we established epithelial cell lines from kidney tissues of Fhit-/- and +/+ mouse pups early after weaning, and subjected cell cultures to nutritional and carcinogen stress, which +/+ cells did not survive. Through transcriptome profiling and protein expression analysis, we observed changes in the Trp53/p21 and survivin apoptotic pathways in -/- cells, and in expression of proteins involved in epithelial-mesenchymal transition. Some Fhit-deficient cell lines showed anchorage-independent colony formation and increased invasive capacity in vitro. Furthermore, cells of stressed Fhit-/- cell lines formed s.c. and metastatic tumors in nude mice. Collectively, we show that Fhit loss and subsequent thymidine kinase 1 inactivation, combined with selective pressures, leads to neoplasia-associated alterations in genes and gene expression patterns in vitro and in vivo.
Collapse
Affiliation(s)
- Jenna R. Karras
- Department of Cancer Biology and GeneticsOhio State University Wexner Medical CenterColumbusOhioUSA
| | - Morgan S. Schrock
- Department of Cancer Biology and GeneticsOhio State University Wexner Medical CenterColumbusOhioUSA
| | - Bahadir Batar
- Department of Cancer Biology and GeneticsOhio State University Wexner Medical CenterColumbusOhioUSA
| | - Jie Zhang
- Department of Biomedical InformaticsOhio State University Wexner Medical CenterColumbusOhioUSA
| | - Krista La Perle
- Department of Veterinary BiosciencesCollege of Veterinary MedicineOhio State UniversityColumbusOhioUSA
| | - Teresa Druck
- Department of Cancer Biology and GeneticsOhio State University Wexner Medical CenterColumbusOhioUSA
| | - Kay Huebner
- Department of Cancer Biology and GeneticsOhio State University Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|
11
|
Sun J, Shen R, Schrock MS, Liu J, Pan X, Quimby D, Zanesi N, Druck T, Fong LY, Huebner K. Reduction in squamous cell carcinomas in mouse skin by dietary zinc supplementation. Cancer Med 2016; 5:2032-42. [PMID: 27185213 PMCID: PMC4873604 DOI: 10.1002/cam4.768] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/12/2016] [Accepted: 04/21/2016] [Indexed: 11/10/2022] Open
Abstract
Inadequate dietary Zn consumption increases susceptibility to esophageal and other cancers in humans and model organisms. Since Zn supplementation can prevent cancers in rodent squamous cell carcinoma (SCC) models, we were interested in determining if it could have a preventive effect in a rodent skin cancer model, as a preclinical basis for considering a role for Zn in prevention of human nonmelanoma skin cancers, the most frequent cancers in humans. We used the 7,12-dimethyl benzanthracene carcinogen/phorbol myristate acetate tumor promoter treatment method to induce skin tumors in Zn-sufficient wild-type and Fhit (human or mouse protein) knockout mice. Fhit protein expression is lost in >50% of human cancers, including skin SCCs, and Fhit-deficient mice show increased sensitivity to carcinogen induction of tumors. We hypothesized that: (1) the skin cancer burdens would be reduced by Zn supplementation; (2) Fhit(-/-) (Fhit, murine fragile histidine triad gene) mice would show increased susceptibility to skin tumor induction versus wild-type mice. 30 weeks after initiating treatment, the tumor burden was increased ~2-fold in Fhit(-/-) versus wild-type mice (16.2 versus 7.6 tumors, P < 0.001); Zn supplementation significantly reduced tumor burdens in Fhit(-/-) mice (males and females combined, 16.2 unsupplemented versus 10.3 supplemented, P = 0.001). Most importantly, the SCC burden was reduced after Zn supplementation in both strains and genders of mice, most significantly in the wild-type males (P = 0.035). Although the mechanism(s) of action of Zn supplementation in skin tumor prevention is not known in detail, the Zn-supplemented tumors showed evidence of reduced DNA damage and some cohorts showed reduced inflammation scores. The results suggest that mild Zn supplementation should be tested for prevention of skin cancer in high-risk human cohorts.
Collapse
Affiliation(s)
- Jin Sun
- Department of Molecular Virology, Immunology and Medical GeneticsThe Ohio State University Comprehensive Cancer CenterColumbusOhio
- Present address: Department of PathologyMolecular PathologyThe Ohio State University Wexner Medical CenterColumbusOhio
| | - Rulong Shen
- Department of PathologyCollege of MedicineColumbusOhio
| | - Morgan S. Schrock
- Department of Molecular Virology, Immunology and Medical GeneticsThe Ohio State University Comprehensive Cancer CenterColumbusOhio
- Biomedical Sciences Graduate ProgramColumbusOhio
| | - James Liu
- Department of Molecular Virology, Immunology and Medical GeneticsThe Ohio State University Comprehensive Cancer CenterColumbusOhio
- Present address: Beaumont‐Oakland University Medical SchoolGrosse PointeMichigan
| | - Xueliang Pan
- Department of Biomedical InformaticsCenter for BiostatisticsThe Ohio State UniversityColumbusOhio
| | - Donald Quimby
- Department of Molecular Virology, Immunology and Medical GeneticsThe Ohio State University Comprehensive Cancer CenterColumbusOhio
- Present address: University of CincinnatiDepartment of Internal MedicineCincinnatiOhio
| | - Nicola Zanesi
- Department of Molecular Virology, Immunology and Medical GeneticsThe Ohio State University Comprehensive Cancer CenterColumbusOhio
| | - Teresa Druck
- Department of Molecular Virology, Immunology and Medical GeneticsThe Ohio State University Comprehensive Cancer CenterColumbusOhio
| | - Louise Y. Fong
- Department of Pathology, Anatomy & Cell BiologySidney Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvania
| | - Kay Huebner
- Department of Molecular Virology, Immunology and Medical GeneticsThe Ohio State University Comprehensive Cancer CenterColumbusOhio
| |
Collapse
|
12
|
Gaudio E, Paduano F, Ngankeu A, Ortuso F, Lovat F, Pinton S, D'Agostino S, Zanesi N, Aqeilan RI, Campiglia P, Novellino E, Alcaro S, Croce CM, Trapasso F. A Fhit-mimetic peptide suppresses annexin A4-mediated chemoresistance to paclitaxel in lung cancer cells. Oncotarget 2016; 7:29927-29936. [PMID: 27166255 PMCID: PMC5058653 DOI: 10.18632/oncotarget.9179] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/11/2016] [Indexed: 01/04/2023] Open
Abstract
We recently reported that Fhit is in a molecular complex with annexin A4 (ANXA4); following to their binding, Fhit delocalizes ANXA4 from plasma membrane to cytosol in paclitaxel-resistant lung cancer cells, thus restoring their chemosensitivity to the drug. Here, we demonstrate that Fhit physically interacts with A4 through its N-terminus; molecular dynamics simulations were performed on a 3D Fhit model to rationalize its mechanism of action. This approach allowed for the identification of the QHLIKPS heptapeptide (position 7 to 13 of the wild-type Fhit protein) as the smallest Fhit sequence still able to preserve its ability to bind ANXA4. Interestingly, Fhit peptide also recapitulates the property of the native protein in inhibiting Annexin A4 translocation from cytosol to plasma membrane in A549 and Calu-2 lung cancer cells treated with paclitaxel. Finally, the combination of Tat-Fhit peptide and paclitaxel synergistically increases the apoptotic rate of cultured lung cancer cells and blocks in vivo tumor formation.Our findings address to the identification of chemically simplified Fhit derivatives that mimic Fhit tumor suppressor functions; intriguingly, this approach might lead to the generation of novel anticancer drugs to be used in combination with conventional therapies in Fhit-negative tumors to prevent or delay chemoresistance.
Collapse
Affiliation(s)
- Eugenio Gaudio
- Department of Molecular Immunology, Virology and Medical Genetics, The Ohio State University, Columbus, Ohio, USA
- Lymphoma & Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
- Dipartimento di Medicina Sperimentale e Clinica, University Magna Græcia, Campus “S. Venuta”, Catanzaro, Italy
| | - Francesco Paduano
- Dipartimento di Medicina Sperimentale e Clinica, University Magna Græcia, Campus “S. Venuta”, Catanzaro, Italy
| | - Apollinaire Ngankeu
- Department of Molecular Immunology, Virology and Medical Genetics, The Ohio State University, Columbus, Ohio, USA
| | - Francesco Ortuso
- Dipartimento di Scienze della Salute, University Magna Græcia, Campus “S. Venuta”, Catanzaro, Italy
| | - Francesca Lovat
- Department of Molecular Immunology, Virology and Medical Genetics, The Ohio State University, Columbus, Ohio, USA
| | - Sandra Pinton
- Lymphoma & Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
| | - Sabrina D'Agostino
- Dipartimento di Medicina Sperimentale e Clinica, University Magna Græcia, Campus “S. Venuta”, Catanzaro, Italy
| | - Nicola Zanesi
- Department of Molecular Immunology, Virology and Medical Genetics, The Ohio State University, Columbus, Ohio, USA
| | - Rami I. Aqeilan
- Department of Molecular Immunology, Virology and Medical Genetics, The Ohio State University, Columbus, Ohio, USA
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research, The Hebrew University, Jerusalem, Israel
| | - Pietro Campiglia
- Dipartimento di Farmacia, Università di Salerno, Fisciano, Italy
| | - Ettore Novellino
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, University Magna Græcia, Campus “S. Venuta”, Catanzaro, Italy
| | - Carlo M. Croce
- Department of Molecular Immunology, Virology and Medical Genetics, The Ohio State University, Columbus, Ohio, USA
| | - Francesco Trapasso
- Dipartimento di Medicina Sperimentale e Clinica, University Magna Græcia, Campus “S. Venuta”, Catanzaro, Italy
| |
Collapse
|
13
|
Paisie CA, Schrock MS, Karras JR, Zhang J, Miuma S, Ouda IM, Waters CE, Saldivar JC, Druck T, Huebner K. Exome-wide single-base substitutions in tissues and derived cell lines of the constitutive Fhit knockout mouse. Cancer Sci 2016; 107:528-35. [PMID: 26782170 PMCID: PMC4832848 DOI: 10.1111/cas.12887] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 02/03/2023] Open
Abstract
Loss of expression of Fhit, a tumor suppressor and genome caretaker, occurs in preneoplastic lesions during development of many human cancers. Furthermore, Fhit-deficient mouse models are exquisitely susceptible to carcinogen induction of cancers of the lung and forestomach. Due to absence of Fhit genome caretaker function, cultured cells and tissues of the constitutive Fhit knockout strain develop chromosome aneuploidy and allele copy number gains and losses and we hypothesized that Fhit-deficient cells would also develop point mutations. On analysis of whole exome sequences of Fhit-deficient tissues and cultured cells, we found 300 to >1000 single-base substitutions associated with Fhit loss in the 2% of the genome included in exomes, relative to the C57Bl6 reference genome. The mutation signature is characterized by increased C>T and T>C mutations, similar to the "age at diagnosis" signature identified in human cancers. The Fhit-deficiency mutation signature also resembles a C>T and T>C mutation signature reported for human papillary kidney cancers and a similar signature recently reported for esophageal and bladder cancers, cancers that are frequently Fhit deficient. The increase in T>C mutations in -/- exomes may be due to dNTP imbalance, particularly in thymidine triphosphate, resulting from decreased expression of thymidine kinase 1 in Fhit-deficient cells. Fhit-deficient kidney cells that survived in vitro dimethylbenz(a)anthracene treatment additionally showed increased T>A mutations, a signature generated by treatment with this carcinogen, suggesting that these T>A transversions may be evidence of carcinogen-induced preneoplastic changes.
Collapse
Affiliation(s)
- Carolyn A. Paisie
- Department of Molecular Virology, Immunology and Medical GeneticsColumbusOhioUSA
- Present address: The Center for Infectious Disease ResearchSeattleWashingtonUSA
| | - Morgan S. Schrock
- Department of Molecular Virology, Immunology and Medical GeneticsColumbusOhioUSA
| | - Jenna R. Karras
- Department of Molecular Virology, Immunology and Medical GeneticsColumbusOhioUSA
| | - Jie Zhang
- Department of Biomedical InformaticsThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Satoshi Miuma
- Department of Gastroenterology and HepatologyGraduate School of Biomedical SciencesNagasaki UniversityNagasakiJapan
| | - Iman M. Ouda
- Department of Molecular Virology, Immunology and Medical GeneticsColumbusOhioUSA
| | - Catherine E. Waters
- Department of Molecular Virology, Immunology and Medical GeneticsColumbusOhioUSA
- Present address: Department of BiochemistryMolecular Biology and Biophysics Institute for Molecular VirologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Joshua C. Saldivar
- Department of Chemical and Systems BiologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Teresa Druck
- Department of Molecular Virology, Immunology and Medical GeneticsColumbusOhioUSA
| | - Kay Huebner
- Department of Molecular Virology, Immunology and Medical GeneticsColumbusOhioUSA
| |
Collapse
|
14
|
Wu X, Wu G, Yao X, Hou G, Jiang F. The clinicopathological significance and ethnic difference of FHIT hypermethylation in non-small-cell lung carcinoma: a meta-analysis and literature review. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:699-709. [PMID: 26929601 PMCID: PMC4760666 DOI: 10.2147/dddt.s85253] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Emerging evidence indicates that FHIT is a candidate tumor suppressor in many types of tumors including non-small-cell lung carcinoma (NSCLC). However, the prognostic value and correlation between FHIT hypermethylation and clinicopathological characteristics of NSCLC remains unclear. In this report, we performed a meta-analysis to evaluate the effects of FHIT hypermethylation on the incidence of NSCLC and clinicopathological characteristics of human NSCLC patients. Final analysis of 1,801 NSCLC patients from 18 eligible studies was performed. FHIT hypermethylation was found to be significantly higher in NSCLC than in normal lung tissue. The pooled odds ratio (OR) from ten studies included 819 NSCLC and 792 normal lung tissues (OR =7.51, 95% confidence interval [CI] =2.98-18.91, P<0.0001). Subgroup analysis based on ethnicity implied that FHIT hypermethylation level was higher in NSCLC tissues than in normal tissues in both Caucasians (P=0.02) and Asians (P<0.0001), indicating that the difference in Asians was much more significant. FHIT hypermethylation was also correlated with sex status, smoking status, as well as pathological types. In addition, patients with FHIT hypermethylation had a lower survival rate than those without (hazard ratio =1.73, 95% CI =1.10-2.71, P=0.02). The results of this meta-analysis suggest that FHIT hypermethylation is associated with an increased risk and poor survival in NSCLC patients. FHIT hypermethylation, which induces the inactivation of FHIT gene, plays an important role in the carcinogenesis and clinical outcome and may serve as a potential diagnostic marker and drug target of NSCLC.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Department of Surgical Oncology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Guannan Wu
- Department of Surgical Oncology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Xuequan Yao
- Department of Surgical Oncology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Gang Hou
- Department of Respiratory Medicine, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Feng Jiang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
15
|
The clinicopathological significance of FHIT hypermethylation in non-small cell lung cancer, a meta-analysis and literature review. Sci Rep 2016; 6:19303. [PMID: 26796853 PMCID: PMC4726317 DOI: 10.1038/srep19303] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 11/18/2015] [Indexed: 12/17/2022] Open
Abstract
Emerging evidence indicates that FHIT is a candidate tumor suppressor in non-small cell lung cancer (NSCLC). However, the correlation between FHIT hypermethylation and clinicopathological characteristics of NSCLC remains unclear. Thus, we conducted a meta-analysis to quantitatively evaluate the effects of FHIT hypermethylation on the incidence of NSCLC and clinicopathological characteristics. Final analysis of 1717 NSCLC patients from 16 eligible studies was performed. FHIT hypermethylation was found to be significantly higher in NSCLC than in normal lung tissue, the pooled OR from 8 studies including 735 NSCLC and 708 normal lung tissue, OR = 5.45, 95% CI = 2.15-13.79, p = 0.0003. FHIT hypermethylation was also correlated with sex status, smoking status, as well as pathological types. We did not find that FHIT hypermethylation was correlated with the differentiated types or clinical stages in NSCLC patients. However, patients with FHIT hypermethylation had a lower survival rate than those without, HR = 1.73, 95% CI = 1.10-2.71, p = 0.02. The results of this meta-analysis suggest that FHIT hypermethylation is associated with an increased risk and worsen survival in NSCLC patients. FHIT hypermethylation, which induces the inactivation of FHIT gene, plays an important role in the carcinogenesis and clinical outcome and may serve as a potential drug target of NSCLC.
Collapse
|
16
|
Molitor M, Junker K, Eltze E, Toma M, Denzinger S, Siegert S, Knuechel R, Gaisa NT. Comparison of structural genetics of non-schistosoma-associated squamous cell carcinoma of the urinary bladder. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:8143-58. [PMID: 26339383 PMCID: PMC4555711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 05/31/2015] [Indexed: 06/05/2023]
Abstract
Little is known about genetic changes in squamous differentiation of non-schistosomiasis-associated bladder cancer. Therefore, we investigated pure squamous cell carcinomas (SqCC), squamous parts of mixed urothelial carcinomas with squamous differentiation (MIX) and mere urothelial cancers (UC) for structural genetic differences. Tissue microarray slides (n = 29 SqCC, n = 35 MIX and n = 23 UC) were analyzed by ZytoLight SPEC p16/CEN3/7/17 Quadruple Color Probe fluorescence-in-situ-hybridization (FISH) and DNA was investigated by comparative genomic hybridization (CGH) (n = 35 SqCCs, n = 40 MIX and n = 36 UC). By FISH the mean number of polysomic cells was lowest in SqCC (CEN3 P = 0.0498, CEN17 P = 0.0009). A slight tendency of lower copy numbers of chromosomes 3, 7 and 17 and higher numbers of the p16-locus in SqCC (P = 0.45) indicated less aneuploid tumor cells in SqCC compared to MIX and UC. In CGH SqCC showed the lowest mean number of aberrations per tumor (SqCC 5.37 changes, MIX 6.75 and UC 7.64; P = 0.1754). Significant differences between the three groups were found for loss of chromosome 3p (P = 0.004), 6q (P = 0.028), 11p (P = 0.024) and gains of 5p (P = 0.020). Loss of 3p was more frequent in SqCC (51.4%) than in MIX (37.5%) or UC (13.9%). To conclude, SqCCs show less polysomy and genetic alterations than MIX and UC. Loss of 3p is more frequent in SqCC but there are no absolute specific alterations for each tumor group. Squamous parts of mixed tumors show similar alterations than UC and should be considered as further development of UC, while pure SqCC seem to be a separate tumor group.
Collapse
Affiliation(s)
- Marina Molitor
- Institute of Pathology, RWTH Aachen UniversityAachen, Germany
| | - Kerstin Junker
- Department of Urology, Friedrich-Schiller-UniversityJena, Germany
- Department of Urology, Saarland UniversityHomburg, Germany
| | - Elke Eltze
- Institute of Pathology Saarbruecken-RastpfuhlSaarbruecken, Germany
| | - Marieta Toma
- Institute of Pathology, University DresdenDresden, Germany
| | - Stefan Denzinger
- Department of Urology, University Hospital RegensburgRegensburg, Germany
| | - Sabine Siegert
- Institute of Pathology LMU MunichMunich, Germany
- Institute of Pathology Munich-NordMunich, Germany
| | - Ruth Knuechel
- Institute of Pathology, RWTH Aachen UniversityAachen, Germany
| | - Nadine T Gaisa
- Institute of Pathology, RWTH Aachen UniversityAachen, Germany
| |
Collapse
|
17
|
Affiliation(s)
- Ramin Nazarian
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | |
Collapse
|
18
|
Stewart DJ, Nunez MI, Jelinek J, Hong D, Gupta S, Aldaz M, Issa JP, Kurzrock R, Wistuba II. Impact of decitabine on immunohistochemistry expression of the putative tumor suppressor genes FHIT, WWOX, FUS1 and PTEN in clinical tumor samples. Clin Epigenetics 2014; 6:13. [PMID: 25024751 PMCID: PMC4094901 DOI: 10.1186/1868-7083-6-13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 06/02/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Since tumor suppressor gene function may be lost through hypermethylation, we assessed whether the demethylating agent decitabine could increase tumor suppressor gene expression clinically. For fragile histidine triad (FHIT), WW domain-containing oxidoreductase (WWOX), fused in sarcoma-1 (FUS1) and phosphatase and tensin homolog (PTEN), immunohistochemistry scores from pre- and post-decitabine tumor biopsies (25 patients) were correlated with methylation of the long interspersed nuclear element-1 (LINE-1) repetitive DNA element (as a surrogate for global DNA methylation) and with tumor regression. RESULTS With negative staining pre-decitabine (score = 0), the number of patients converting to positive staining post-decitabine was 1 of 1 for FHIT, 3 of 6 for WWOX, 2 of 3 for FUS1 and 1 of 10 for PTEN. In tumors with low pre-decitabine tumor suppressor gene scores (≤150), expression was higher post-treatment in 8 of 8 cases for FHIT (P = 0.014), 7 of 17 for WWOX (P = 0.0547), 7 of 12 for FUS1 (P = 0.0726), and 1 of 16 for PTEN (P = 0.2034). If FHIT, WWOX and FUS1 were considered together, median pre- versus post-decitabine scores were 60 versus 100 (P = 0.0002). Overall, tumor suppressor gene expression change did not correlate with LINE-1 demethylation, although tumors converting from negative to positive had a median decrease in LINE-1 methylation of 24%, compared to 6% in those not converting (P = 0.069). Five of 15 fully evaluable patients had reductions in tumor diameter (range 0.2% to 33.4%). Of these, three had simultaneous increases in three tumor suppressor genes (including the two patients with the greatest tumor regression) compared to 2 of 10 with tumor growth (P = 0.25). CONCLUSIONS In tumors with low tumor suppressor gene expression, decitabine may be associated with increased expression of the tumor suppressor genes FHIT, FUS1, and WWOX, but not PTEN.
Collapse
Affiliation(s)
- David J Stewart
- University of Ottawa, 501 Smyth Rd, Ottawa, ON K1H 8 L6, Canada
| | - Maria I Nunez
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jaroslav Jelinek
- Fels Institute for Cancer Research and Molecular Biology, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - David Hong
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Sanjay Gupta
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Marcelo Aldaz
- UT MD Anderson Cancer Center, 1808 Park Road 1C, Smithville, TX 78957, USA
| | - Jean-Pierre Issa
- Fels Institute for Cancer Research and Molecular Biology, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Razelle Kurzrock
- University of California San Diego, 3855 Health Sciences Dr, La Jolla, CA 92093, USA
| | - Ignacio I Wistuba
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
19
|
Replicative Stress and the FHIT Gene: Roles in Tumor Suppression, Genome Stability and Prevention of Carcinogenesis. Cancers (Basel) 2014; 6:1208-19. [PMID: 24901304 PMCID: PMC4074825 DOI: 10.3390/cancers6021208] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/21/2014] [Accepted: 05/26/2014] [Indexed: 12/25/2022] Open
Abstract
The fragile FHIT gene, encompassing the chromosomal fragile site FRA3B, is an early target of DNA damage in precancerous cells. While vulnerable to DNA damage itself, FHIT protein expression is essential to protect from DNA damage-induced cancer initiation and progression by modulating genome stability, oxidative stress and levels of accumulating DNA damage. Thus, FHIT, whose expression is lost or reduced in many human cancers, is a tumor suppressor and genome caretaker whose loss initiates genome instability in preneoplastic lesions. Ongoing studies are seeking more detailed understanding of the role of FHIT in the cellular response to oxidative damage. This review discusses the relationship between FHIT, reactive oxygen species production, and DNA damage in the context of cancer initiation and progression.
Collapse
|
20
|
Kondratov AG, Nekrasov KA, Lototska LV, Panasenko GV, Stoliar LA, Lapska YV, Kolesnyk OO, Shchepotin IB, Rynditch AV, Kashuba VI. Comparative analysis of epigenetic markers in plasma and tissue of patients with colorectal cancer. ACTA ACUST UNITED AC 2014. [DOI: 10.7124/bc.00088b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
| | - K. A. Nekrasov
- Institute of Molecular Biology and Genetics, NAS of Ukraine
| | - L. V. Lototska
- Institute of Molecular Biology and Genetics, NAS of Ukraine
| | | | - L. A. Stoliar
- Institute of Molecular Biology and Genetics, NAS of Ukraine
| | - Y. V. Lapska
- Institute of Molecular Biology and Genetics, NAS of Ukraine
| | | | | | - A. V. Rynditch
- Institute of Molecular Biology and Genetics, NAS of Ukraine
| | - V. I. Kashuba
- Institute of Molecular Biology and Genetics, NAS of Ukraine
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute
| |
Collapse
|
21
|
Sun Z, Huang C, He J, Lamb KL, Kang X, Gu T, Shen WH, Yin Y. PTEN C-terminal deletion causes genomic instability and tumor development. Cell Rep 2014; 6:844-54. [PMID: 24561254 DOI: 10.1016/j.celrep.2014.01.030] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 12/22/2013] [Accepted: 01/23/2014] [Indexed: 12/18/2022] Open
Abstract
Tumor suppressor PTEN controls genomic stability and inhibits tumorigenesis. The N-terminal phosphatase domain of PTEN antagonizes the PI3K/AKT pathway, but its C-terminal function is less defined. Here, we describe a knockin mouse model of a nonsense mutation that results in the deletion of the entire Pten C-terminal region, referred to as Pten(ΔC). Mice heterozygous for Pten(ΔC) develop multiple spontaneous tumors, including cancers and B cell lymphoma. Heterozygous deletion of the Pten C-terminal domain also causes genomic instability and common fragile site rearrangement. We found that Pten C-terminal disruption induces p53 and its downstream targets. Simultaneous depletion of p53 promotes metastasis without influencing the initiation of tumors, suggesting that p53 mainly suppresses tumor progression. Our data highlight the essential role of the PTEN C terminus in the maintenance of genomic stability and suppression of tumorigenesis.
Collapse
Affiliation(s)
- Zhuo Sun
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PRC; Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Chuanxin Huang
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Jinxue He
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Kristy L Lamb
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Xi Kang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PRC; Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Tingting Gu
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Wen Hong Shen
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA.
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PRC; Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
22
|
Gao G, Kasperbauer JL, Tombers NM, Wang V, Mayer K, Smith DI. A selected group of large common fragile site genes have decreased expression in oropharyngeal squamous cell carcinomas. Genes Chromosomes Cancer 2014; 53:392-401. [DOI: 10.1002/gcc.22150] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 01/14/2013] [Accepted: 01/15/2013] [Indexed: 01/11/2023] Open
Affiliation(s)
- Ge Gao
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester MN
| | | | | | - Vivian Wang
- Mayo Medical Genome Facilities; Mayo Clinic; Rochester MN
| | - Kevin Mayer
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester MN
| | - David I. Smith
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester MN
| |
Collapse
|
23
|
Pyne NJ, Ohotski J, Bittman R, Pyne S. The role of sphingosine 1-phosphate in inflammation and cancer. Adv Biol Regul 2014; 54:121-129. [PMID: 24070975 DOI: 10.1016/j.jbior.2013.08.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 08/31/2013] [Accepted: 08/31/2013] [Indexed: 06/02/2023]
Abstract
The enzymes that catalyze formation of the bioactive sphingolipid, sphingosine 1-phosphate, sphingosine kinase 1 and 2, are predictive markers in inflammatory diseases and cancer as evidenced by data from patients, knockout mice and the use of available molecular and chemical inhibitors. Thus, there is a compelling case for therapeutic targeting of sphingosine kinase. In addition, there are several examples of functional interaction between sphingosine 1-phosphate receptors and sphingosine kinase 1 that can drive malicious amplification loops that promote cancer cell growth. These novel aspects of sphingosine 1-phosphate pathobiology are reviewed herein.
Collapse
Affiliation(s)
- Nigel J Pyne
- Cell Biology Research Group, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Jan Ohotski
- Cell Biology Research Group, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, New York 11367-1597, USA
| | - Susan Pyne
- Cell Biology Research Group, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
24
|
Miuma S, Saldivar JC, Karras JR, Waters CE, Paisie CA, Wang Y, Jin V, Sun J, Druck T, Zhang J, Huebner K. Fhit deficiency-induced global genome instability promotes mutation and clonal expansion. PLoS One 2013; 8:e80730. [PMID: 24244712 PMCID: PMC3828255 DOI: 10.1371/journal.pone.0080730] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/07/2013] [Indexed: 01/25/2023] Open
Abstract
Loss of Fhit expression, encoded at chromosome fragile site FRA3B, leads to increased replication stress, genome instability and accumulation of genetic alterations. We have proposed that Fhit is a genome ‘caretaker’ whose loss initiates genome instability in preneoplastic lesions. We have characterized allele copy number alterations and expression changes observed in Fhit-deficient cells in conjunction with alterations in cellular proliferation and exome mutations, using cells from mouse embryo fibroblasts (MEFs), mouse kidney, early and late after establishment in culture, and in response to carcinogen treatment. Fhit-/- MEFs escape senescence to become immortal more rapidly than Fhit+/+ MEFs; -/- MEFs and kidney cultures show allele losses and gains, while +/+ derived cells show few genomic alterations. Striking alterations in expression of p53, p21, Mcl1 and active caspase 3 occurred in mouse kidney -/- cells during progressive tissue culture passage. To define genomic changes associated with preneoplastic changes in vivo, exome DNAs were sequenced for +/+ and -/- liver tissue after treatment of mice with the carcinogen, 7,12-dimethylbenz[a]anthracene, and for +/+ and -/- kidney cells treated in vitro with this carcinogen. The -/- exome DNAs, in comparison with +/+ DNA, showed small insertions, deletions and point mutations in more genes, some likely related to preneoplastic changes. Thus, Fhit loss provides a ‘mutator’ phenotype, a cellular environment in which mild genome instability permits clonal expansion, through proliferative advantage and escape from apoptosis, in response to pressures to survive.
Collapse
Affiliation(s)
- Satoshi Miuma
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Joshua C. Saldivar
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Jenna R. Karras
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Catherine E. Waters
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Carolyn A. Paisie
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Yao Wang
- Department of Biomedical Informatics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Victor Jin
- Department of Biomedical Informatics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Jin Sun
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Teresa Druck
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Jie Zhang
- Department of Biomedical Informatics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Kay Huebner
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
25
|
Al-Temaimi RA, Jacob S, Al-Ali W, Thomas DA, Al-Mulla F. Reduced FHIT expression is associated with mismatch repair deficient and high CpG island methylator phenotype colorectal cancer. J Histochem Cytochem 2013; 61:627-38. [PMID: 23797051 DOI: 10.1369/0022155413497367] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease and a major contributor to world cancer mortality rates. Molecular subtypes of CRC have become standards for CRC classification and have established prognostic potential. Here, we attempt to corroborate and provide further insight pertinent to the fragile histidine triad (FHIT) gene in microsatellite instable (MSI), microsatellite stable (MSS), and CpG island methylator phenotype (CIMP) CRC subtypes. We employed array comparative genomic hybridization and multiplex ligation-dependent probe amplification (MLPA) techniques to survey genomic aberrations in FHIT gene and their effects on FHIT protein expression using immunohistochemistry (IHC) in a CRC cohort. We further studied FHIT protein expression by IHC in a larger CRC cohort defined for its mismatch repair (MMR) protein expression and genomic methylation profiles. Our results show FHIT genomic deletions centered in exons 4 and 5 in most of MSI-CRC samples. Moreover, we confirmed the significant association of FHIT protein expression diminution (p=0.035) with MSI-CRC. In the larger cohort, reduced FHIT protein expression was significantly associated with CIMP-high subtype of CRC (p=0.009) and loss of PMS2 protein expression (p=0.017). We conclude that FHIT expression may be a valuable marker for CRC subtyping, and its diagnostic, prognostic, and therapeutic potential should be perused.
Collapse
Affiliation(s)
- Rabeah Abbas Al-Temaimi
- Human Genetics Unit, Pathology Department, Faculty of Medicine, Health Sciences Center, Kuwait University, State of Kuwait
| | | | | | | | | |
Collapse
|
26
|
The roles of sphingosine kinase 1 and 2 in regulating the metabolome and survival of prostate cancer cells. Biomolecules 2013; 3:316-33. [PMID: 24970170 PMCID: PMC4030851 DOI: 10.3390/biom3020316] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 06/03/2013] [Accepted: 06/04/2013] [Indexed: 12/15/2022] Open
Abstract
We have previously shown that treatment of androgen-sensitive LNCaP cells with the sphingosine kinase (SK) inhibitor SKi (2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole) induces the proteasomal degradation of two N-terminal variants of SK1 (SK1a and SK1b), increases C22:0-ceramide and diadenosine 5′,5′′′-P1,P3-triphosphate (Ap3A) and reduces S1P levels, and promotes apoptosis. We have now investigated the effects of three SK inhibitors (SKi, (S)-FTY720 vinylphosphonate, and (R)-FTY720 methyl ether) on metabolite and sphingolipid levels in androgen-sensitive LNCaP and androgen-independent LNCaP-AI prostate cancer cells. The 51 kDa N-terminal variant of SK1 (SK1b) evades the proteasome in LNCaP-AI cells, and these cells do not exhibit an increase in C22:0-ceramide or Ap3A levels and do not undergo apoptosis in response to SKi. In contrast, the SK inhibitor (S)-FTY720 vinylphosphonate induces degradation of SK1b in LNCaP-AI, but not in LNCaP cells. In LNCaP-AI cells, (S)-FTY720 vinylphosphonate induces a small increase in C16:0-ceramide levels and cleavage of polyADPribose polymerase (indicative of apoptosis). Surprisingly, the level of S1P is increased by 7.8- and 12.8-fold in LNCaP and LNCaP-AI cells, respectively, on treatment with (S)-FTY720 vinylphosphonate. Finally, treatment of androgen-sensitive LNCaP cells with the SK2-selective inhibitor (R)-FTY720 methyl ether increases lysophosphatidylinositol levels, suggesting that SK2 may regulate lyso-PI metabolism in prostate cancer cells.
Collapse
|
27
|
Watson DG, Tonelli F, Osaimi MA, Williamson L, Chan E, Gorshkova I, Berdyshev E, Bittman R, Pyne NJ, Pyne S. The roles of sphingosine kinases 1 and 2 in regulating the Warburg effect in prostate cancer cells. Cell Signal 2013; 25:1011-7. [PMID: 23314175 PMCID: PMC3595369 DOI: 10.1016/j.cellsig.2013.01.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/06/2013] [Indexed: 12/30/2022]
Abstract
Two isoforms of sphingosine kinase, SK1 and SK2, catalyze the formation of the bioactive lipid sphingosine 1-phosphate (S1P) in mammalian cells. We have previously shown that treatment of androgen-sensitive LNCaP prostate cancer cells with a non-selective SK isoform inhibitor, 2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole (SKi), induces the proteasomal degradation of SK1. This is concomitant with a significant increase in C22:0-ceramide and sphingosine levels and a reduction in S1P levels, resulting in the apoptosis of LNCaP cells. In contrast, we show here that a SK2-selective inhibitor, (R)-FTY720 methyl ether (ROME), increases sphingosine and decreases S1P levels but has no effect on ceramide levels and does not induce apoptosis in LNCaP cells. We also show that several glycolytic metabolites and (R)-S-lactoylglutathione are increased upon treatment of LNCaP cells with SKi, which induces the proteasomal degradation of c-Myc. These changes reflect an indirect antagonism of the Warburg effect. LNCaP cells also respond to SKi by diverting glucose 6-phosphate into the pentose phosphate pathway to provide NADPH, which serves as an antioxidant to counter an oxidative stress response. SKi also promotes the formation of a novel pro-apoptotic molecule called diadenosine 5',5'''-P(1),P(3)-triphosphate (Ap3A), which binds to the tumor suppressor fragile histidine triad protein (FHIT). In contrast, the SK2-selective inhibitor, ROME, induces a reduction in some glycolytic metabolites and does not affect oxidative stress. We conclude that SK1 functions to increase the stability of c-Myc and suppresses Ap3A formation, which might maintain the Warburg effect and cell survival, while SK2 exhibits a non-overlapping function.
Collapse
Affiliation(s)
| | | | | | | | | | - Irina Gorshkova
- The University of Illinois at Chicago Section of Pulmonary, Critical Care, Sleep, and Allergy, 909 S. Wolcott Ave., Chicago, IL 60612, USA
| | - Evgeny Berdyshev
- The University of Illinois at Chicago Section of Pulmonary, Critical Care, Sleep, and Allergy, 909 S. Wolcott Ave., Chicago, IL 60612, USA
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, New York 11367-1597, USA
| | | | - Susan Pyne
- To whom correspondence should be addressed (Tel: 441415482012; Fax: 441415522562; )
| |
Collapse
|
28
|
Amino-acyl tRNA synthetases generate dinucleotide polyphosphates as second messengers: functional implications. Top Curr Chem (Cham) 2013; 344:189-206. [PMID: 23536246 DOI: 10.1007/128_2013_426] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this chapter we describe aminoacyl-tRNA synthetase (aaRS) production of dinucleotide polyphosphate in response to stimuli, their interaction with various signaling pathways, and the role of diadenosine tetraphosphate and diadenosine triphosphate as second messengers. The primary role of aaRS is to mediate aminoacylation of cognate tRNAs, thereby providing a central role for the decoding of genetic code during protein translation. However, recent studies suggest that during evolution, "moonlighting" or non-canonical roles were acquired through incorporation of additional domains, leading to regulation by aaRSs of a spectrum of important biological processes, including cell cycle control, tissue differentiation, cellular chemotaxis, and inflammation. In addition to aminoacylation of tRNA, most aaRSs can also produce dinucleotide polyphosphates in a variety of physiological conditions. The dinucleotide polyphosphates produced by aaRS are biologically active both extra- and intra-cellularly, and seem to function as important signaling molecules. Recent findings established the role of dinucleotide polyphosphates as second messengers.
Collapse
|
29
|
Recurrent R-spondin fusions in colon cancer. Nature 2012; 488:660-4. [PMID: 22895193 DOI: 10.1038/nature11282] [Citation(s) in RCA: 777] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 06/06/2012] [Indexed: 12/15/2022]
Abstract
Identifying and understanding changes in cancer genomes is essential for the development of targeted therapeutics. Here we analyse systematically more than 70 pairs of primary human colon tumours by applying next-generation sequencing to characterize their exomes, transcriptomes and copy-number alterations. We have identified 36,303 protein-altering somatic changes that include several new recurrent mutations in the Wnt pathway gene TCF7L2, chromatin-remodelling genes such as TET2 and TET3 and receptor tyrosine kinases including ERBB3. Our analysis for significantly mutated cancer genes identified 23 candidates, including the cell cycle checkpoint kinase ATM. Copy-number and RNA-seq data analysis identified amplifications and corresponding overexpression of IGF2 in a subset of colon tumours. Furthermore, using RNA-seq data we identified multiple fusion transcripts including recurrent gene fusions involving R-spondin family members RSPO2 and RSPO3 that together occur in 10% of colon tumours. The RSPO fusions were mutually exclusive with APC mutations, indicating that they probably have a role in the activation of Wnt signalling and tumorigenesis. Consistent with this we show that the RSPO fusion proteins were capable of potentiating Wnt signalling. The R-spondin gene fusions and several other gene mutations identified in this study provide new potential opportunities for therapeutic intervention in colon cancer.
Collapse
|
30
|
Saldivar JC, Bene J, Hosseini SA, Miuma S, Horton S, Heerema NA, Huebner K. Characterization of the role of Fhit in suppression of DNA damage. Adv Biol Regul 2012; 53:77-85. [PMID: 23102829 DOI: 10.1016/j.jbior.2012.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 10/04/2012] [Indexed: 01/08/2023]
Abstract
The fragile histidine triad protein, Fhit, has a number of reported tumor suppressive functions which include signaling of apoptosis in cancer cells in vitro and in vivo, modulation of the DNA damage response, down-regulation of target oncogene expression, suppression of tumor growth in vivo, and suppression of cancer cell invasion and metastasis. Most of these functions of Fhit have been observed on exogenous re-expression of Fhit in Fhit-negative cancer cells. However, little is known about the tumorigenic changes that occur in normal or precancerous cells following loss of Fhit expression. Recently, we have shown that shortly after loss of Fhit expression, cells exhibit signs of DNA replication stress-induced DNA damage and develop genomic instability. Here, we extend these findings through investigation of different factors that affect Fhit function to prevent DNA damage. We found that Fhit activity is dependent upon a functional HIT domain and the tyrosine-114 residue, previously shown to be required for tumor suppression by Fhit. Furthermore, Fhit function was shown to be independent of exogenous and endogenous sources of oxidative stress. Finally, Fhit function was shown to be dependent upon Chk1 kinase activity, but independent of Atr or Atm kinases. Evidence suggests that Fhit and Chk1 kinase cooperate to prevent replication stress-induced DNA damage. These findings provide important and unexpected insights into the mechanism whereby loss of Fhit expression contributes to cell transformation.
Collapse
Affiliation(s)
- Joshua C Saldivar
- Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Li H, Zhao P, Lu Y, Lu Y. Correlation of aberrant expression of CD133 with FHIT and malignant phenotype of colorectal adenocarcinoma. Int J Colorectal Dis 2012; 27:1015-20. [PMID: 22315171 DOI: 10.1007/s00384-012-1421-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2012] [Indexed: 02/04/2023]
Abstract
BACKGROUND The relationship between the expression of CD133 and fragile histidine triad (FHIT) or prognosis in Chinese colorectal adenocarcinoma is unknown and needs to be explored. MATERIAL AND METHODS The samples of colorectal adenocarcinoma from 200 Chinese patients with follow-up were analyzed for the expression of CD133 and FHIT proteins by immunohistochemical method. RESULTS CD133 was highly expressed in up to 42.0%(84/200) of this group of colorectal adenocarcinomas. The expression of CD133 was significantly higher in carcinoma than in normal (P=0.0001) and in adenomatous mucosas (P=0.004). CD133 positively corresponds to histological grade, clinical stage, regional lymphatic metastasis, and distant metastasis (all P<0.05). The mean overall survival time was shorter in patients with CD133 high expression than in those with CD133 low expression (P00.0001). The expression of CD133 was inversely correlative with that of FHIT (r=-0.464, P=0.0001) in colorectal adenocarcinoma. CD133 was an independent prognostic factor (P=0.0001). CONCLUSIONS The expression of CD133 may be inversely correlated with the expression of FHIT. It is suggested that CD133 may play an important role in the evolution of colorectal adenocarcinoma and be considered as a potential marker for the prognosis.
Collapse
Affiliation(s)
- Hong Li
- Health Medical Center, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | | | | | | |
Collapse
|
32
|
Tian Y, Zhang J, Yan S, Qiu L, Li Z. FATS expression is associated with cisplatin sensitivity in non small cell lung cancer. Lung Cancer 2012; 76:416-22. [DOI: 10.1016/j.lungcan.2011.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 09/26/2011] [Accepted: 11/07/2011] [Indexed: 01/21/2023]
|
33
|
Josse R, Dumont J, Fautrel A, Robin MA, Guillouzo A. Identification of early target genes of aflatoxin B1 in human hepatocytes, inter-individual variability and comparison with other genotoxic compounds. Toxicol Appl Pharmacol 2012; 258:176-87. [DOI: 10.1016/j.taap.2011.10.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 10/17/2011] [Accepted: 10/26/2011] [Indexed: 12/26/2022]
|
34
|
Barresi V, Romano A, Musso N, Capizzi C, Consoli C, Martelli MP, Palumbo G, Di Raimondo F, Condorelli DF. Broad copy neutral-loss of heterozygosity regions and rare recurring copy number abnormalities in normal karyotype-acute myeloid leukemia genomes. Genes Chromosomes Cancer 2011; 49:1014-23. [PMID: 20725993 DOI: 10.1002/gcc.20810] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
We analyzed, by the latest high-resolution SNP arrays, 19 Normal Karyotype (NK)-AML patients at diagnosis (Dx) and remission (R) phases, to determine the number of tumor-associated copy number abnormalities (CNAs) and copy neutral-loss of heterozygosity (CN-LOH) regions per patient and to identify possible recurring genomic abnormalities. The number of tumor-associated CNAs was determined after comparison of matched Dx/R samples using stringent conditions able to reduce the number of false positive CNAs. With the exception of a single outlier case, a low number of CNAs per patient was detected (median value of 1 somatic loss or gain per patient). However, a high prevalence of CNAs (60-70% of the patients showed at least one tumor-associated gain or loss) and few recurring CNAs were observed, thus providing new hints towards identification of cooperating mutations. An extensive search of all tumor-associated CN-LOH regions >1 Mb revealed only three broad regions (terminal 12Mb of 22q, terminal 27Mb of 1p and the whole chromosome 21) in three patients out of 19 (16%). CN-LOH of the whole chromosome 21 was responsible for homozygosity of a missense mutation (R80C) of RUNX1/AML1. Our study suggests that a relative submicroscopic copy number stability NK-AML genomes is associated with low recurrence of specific CNAs and CN-LOH in NK-AML patient population. Sequencing of candidate genes in the identified CNAs and CN-LOH regions should be considered a priority in the search of novel driver mutations of AML.
Collapse
Affiliation(s)
- Vincenza Barresi
- Laboratorio sui Sistemi Complessi, Scuola Superiore di Catania, Università di Catania, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kelley K, Berberich SJ. FHIT gene expression is repressed by mitogenic signaling through the PI3K/AKT/FOXO pathway. Am J Cancer Res 2011; 1:62-70. [PMID: 21743838 PMCID: PMC3130617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2010] [Accepted: 10/10/2010] [Indexed: 05/31/2023] Open
Abstract
The Fragile Histidine Triad gene or FHIT functions as tumor suppressor in many epithelial cell types. Although its tumor suppressive mechanism is the subject of intense study, less is known about how FHIT gene expression itself is regulated. Here we show that PI3 kinase and its downstream target AKT suppress FHIT gene expression in response to growth factor stimulation in actively cycling cells. Upon removal of mitogens from the culture environment, FHIT mRNA and protein levels are observed to increase as a result of derepression from these protooncogenic kinases. AKT signaling through the FOXO transcription factors appears to be the basis for FHIT gene regulation. Increases in FHIT gene expression are directly dependent on endogenous FOXO3a in MCF7 breast carcinoma cells as evidenced by experiments with RNAi targeting FOXO transcription factor family members. Thus, this is the first report demonstrating that FHIT gene expression is normally repressed in actively cycling cells through the PI3K/AKT/FOXO3a axis.
Collapse
Affiliation(s)
- Kevin Kelley
- Wright State University, Boonshoft School of Medicine, Biochemistry & Molecular Biology Department, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA
| | | |
Collapse
|
36
|
Sun J, Liu J, Pan X, Quimby D, Zanesi N, Druck T, Pfeifer GP, Croce CM, Fong LY, Huebner K. Effect of zinc supplementation on N-nitrosomethylbenzylamine-induced forestomach tumor development and progression in tumor suppressor-deficient mouse strains. Carcinogenesis 2010; 32:351-8. [PMID: 21097531 DOI: 10.1093/carcin/bgq251] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Zinc deficiency is associated with high incidences of esophageal and other cancers in humans and leads to a highly proliferative hyperplastic condition in the upper gastrointestinal tract in laboratory rodents. Zn replenishment reduces the incidence of lingual, esophageal and forestomach tumors in Zn-deficient rats and mice. While previous animal studies focused on Zn deficiency, we have investigated the effect of Zn supplementation on carcinogenesis in Zn-sufficient mice of wild-type and tumor suppressor-deficient mouse strains. All mice received N-nitrosomethylbenzylamine and half the mice of each strain then received Zn supplementation. At killing, mice without Zn supplementation had developed more tumors than Zn-supplemented mice: wild-type C57BL/6 mice developed an average of 7.0 versus 5.0 tumors for Zn supplemented (P < 0.05); Zn-supplemented Fhit-/- mice averaged 5.7 versus 8.0 for control mice (P < 0.01); Zn-supplemented Fhit-/-Nit1-/- mice averaged 5.4 versus 9.2 for control mice (P < 0.01) and Zn-supplemented Fhit-/-Rassf1a-/- (the murine gene) mice averaged 5.9 versus 9.1 for control mice (P < 0.01). Zn supplementation reduced tumor burdens by 28% (wild-type) to 42% (Fhit-/-Nit1-/-). Histological analysis of forestomach tissues also showed significant decreases in severity of preneoplastic and neoplastic lesions in Zn-supplemented cohorts of each mouse strain. Thus, Zn supplementation significantly reduced tumor burdens in mice with multiple tumor suppressor deficiencies. When Zn supplementation was begun at 7 weeks after the final carcinogen dose, the reduction in tumor burden was the same as observed when supplementation began immediately after carcinogen dosing, suggesting that Zn supplementation may affect tumor progression rather than tumor initiation.
Collapse
Affiliation(s)
- Jin Sun
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Guler G, Himmetoglu C, Jimenez RE, Geyer SM, Wang WP, Costinean S, Pilarski RT, Morrison C, Suren D, Liu J, Chen J, Kamal J, Shapiro CL, Huebner K. Aberrant expression of DNA damage response proteins is associated with breast cancer subtype and clinical features. Breast Cancer Res Treat 2010; 129:421-32. [PMID: 21069451 DOI: 10.1007/s10549-010-1248-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 10/26/2010] [Indexed: 12/18/2022]
Abstract
Landmark studies of the status of DNA damage checkpoints and associated repair functions in preneoplastic and neoplastic cells has focused attention on importance of these pathways in cancer development, and inhibitors of repair pathways are in clinical trials for treatment of triple negative breast cancer. Cancer heterogeneity suggests that specific cancer subtypes will have distinct mechanisms of DNA damage survival, dependent on biological context. In this study, status of DNA damage response (DDR)-associated proteins was examined in breast cancer subtypes in association with clinical features; 479 breast cancers were examined for expression of DDR proteins γH2AX, BRCA1, pChk2, and p53, DNA damage-sensitive tumor suppressors Fhit and Wwox, and Wwox-interacting proteins Ap2α, Ap2γ, ErbB4, and correlations among proteins, tumor subtypes, and clinical features were assessed. In a multivariable model, triple negative cancers showed significantly reduced Fhit and Wwox, increased p53 and Ap2γ protein expression, and were significantly more likely than other subtype tumors to exhibit aberrant expression of two or more DDR-associated proteins. Disease-free survival was associated with subtype, Fhit and membrane ErbB4 expression level and aberrant expression of multiple DDR-associated proteins. These results suggest that definition of specific DNA repair and checkpoint defects in subgroups of triple negative cancer might identify new treatment targets. Expression of Wwox and its interactor, ErbB4, was highly significantly reduced in metastatic tissues vs. matched primary tissues, suggesting that Wwox signal pathway loss contributes to lymph node metastasis, perhaps by allowing survival of tumor cells that have detached from basement membranes, as proposed for the role of Wwox in ovarian cancer spread.
Collapse
Affiliation(s)
- Gulnur Guler
- Department of Pathology, Hacettepe University, Ankara, Turkey
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhao P, Lu Y, Han W. Clinicopathological significance and prognostic value of leukemia-related protein 16 expression in invasive ductal breast carcinoma. Cancer Sci 2010; 101:2262-8. [PMID: 20649898 PMCID: PMC11159915 DOI: 10.1111/j.1349-7006.2010.01658.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
To explore the expression of leukemia-related protein 16 (LRP16) in invasive ductal breast carcinoma and analyze its correlation with clinicopathological feature and prognosis, immunohistochemistry was performed on 100 cases of invasive ductal breast carcinoma. Medical records were reviewed and clinicopathological analysis was performed. Leukemia-related protein 16 expression was detected in 33 of 100 cases (33%) of the invasive ductal breast carcinoma. Expression of LRP16 in carcinoma was obviously higher than that in normal breast tissue. LRP16 protein expression was found in 27.6% (21/76) of carcinoma at stage I and II, and 50.0% (12/24) of carcinoma at stage III and IV. LRP16 expression was found correlative with metastasis in the axillary lymph node (P = 0.001), stage (P = 0.042), estrogen receptor (ER) expression (P = 0.001), fragile histidine triad (FHIT) expression (P = 0.015) and CD133 expression (P = 0.038), but not with grade (P = 0.543), tumor size (P = 0.263), age (P = 0.840), menopause (P = 0.701) and HER-2 gene amplification (P = 0.463). The difference of the mean disease free survival (DFS) time between cancer patients with LRP16 expression (43.7 months) and those without (77.7 months) was statistically significant (Log rank = 9.989, P = 0.002). The difference of the mean overall survival (OS) time between cancer patients with LRP16 expression (50.0 months) and those without (120.0 months) was statistically significant (Log rank = 9.977, P = 0.002). Our finding suggests that expression of LRP16 protein is correlated with the stage, metastasis, prognosis and expression of ER, progesterone receptor, Ki-67, CD133 and FHIT in invasive ductal breast carcinoma.
Collapse
Affiliation(s)
- Po Zhao
- Department of Pathology, Chinese People's Liberation Army General Hospital, Beijing, China.
| | | | | |
Collapse
|
39
|
Saldivar JC, Shibata H, Huebner K. Pathology and biology associated with the fragile FHIT gene and gene product. J Cell Biochem 2010; 109:858-65. [PMID: 20082323 DOI: 10.1002/jcb.22481] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
More than 12 years and >800 scientific publications after the discovery of the first gene at a chromosome fragile site, the FHIT gene at FRA3B, there are still questions to pursue concerning the selective advantage conferred to cells by loss of expression of FHIT, the most frequent target of allele deletion in precancerous lesions and cancers. These questions are considered in light of recent investigations of genetic and epigenetic alterations to the locus and in a retrospective consideration of biological roles of the Fhit protein discovered through functional studies.
Collapse
Affiliation(s)
- Joshua C Saldivar
- Integrated Biomedical Science Graduate Program, Ohio State University Medical Center, Columbus, Ohio, USA
| | | | | |
Collapse
|
40
|
Salah Z, Aqeilan R, Huebner K. WWOX gene and gene product: tumor suppression through specific protein interactions. Future Oncol 2010; 6:249-59. [PMID: 20146584 DOI: 10.2217/fon.09.152] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The WWOX gene, an archetypal fragile gene, encompasses a chromosomal fragile site at 16q23.2, and encodes the approximately 46-kDa Wwox protein, with WW domains that interact with a growing list of interesting proteins. If the function of a protein is defined by the company it keeps, then Wwox is involved in numerous important signal pathways for bone and germ-cell development, cellular and animal growth and death, transcriptional control and suppression of cancer development. Because alterations to genes at fragile sites are exquisitely sensitive to replication stress-induced DNA damage, there has been an ongoing scientific discussion questioning whether such gene expression alterations provide a selective advantage for clonal expansion of neoplastic cells, and a parallel discussion on why important genes would be present at sites that are susceptible to inactivation. We offer some answers through a description of known WWOX functions.
Collapse
Affiliation(s)
- Zaidoun Salah
- The Lautenberg Center for Immunology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Pharmacy Building, Jerusalem 91120, Israel.
| | | | | |
Collapse
|
41
|
Liu GY, Liu KH, Zhang Y, Wang YZ, Wu XH, Lu YZ, Pan C, Yin P, Liao HF, Su JQ, Ge Q, Luo Q, Xiong B. Alterations of tumor-related genes do not exactly match the histopathological grade in gastric adenocarcinomas. World J Gastroenterol 2010; 16:1129-37. [PMID: 20205286 PMCID: PMC2835792 DOI: 10.3748/wjg.v16.i9.1129] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [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 investigate the diverse characteristics of different pathological gradings of gastric adenocarcinoma (GA) using tumor-related genes.
METHODS: GA tissues in different pathological gradings and normal tissues were subjected to tissue arrays. Expressions of 15 major tumor-related genes were detected by RNA in situ hybridization along with 3’ terminal digoxin-labeled anti-sense single stranded oligonucleotide and locked nucleic acid modifying probe within the tissue array. The data obtained were processed by support vector machines by four different feature selection methods to discover the respective critical gene/gene subsets contributing to the GA activities of different pathological gradings.
RESULTS: In comparison of poorly differentiated GA with normal tissues, tumor-related gene TP53 plays a key role, although other six tumor-related genes could also achieve the Area Under Curve (AUC) of the receiver operating characteristic independently by more than 80%. Comparing the well differentiated GA with normal tissues, we found that 11 tumor-related genes could independently obtain the AUC by more than 80%, but only the gene subsets, TP53, RB and PTEN, play a key role. Only the gene subsets, Bcl10, UVRAG, APC, Beclin1, NM23, PTEN and RB could distinguish between the poorly differentiated and well differentiated GA. None of a single gene could obtain a valid distinction.
CONCLUSION: Different from the traditional point of view, the well differentiated cancer tissues have more alterations of important tumor-related genes than the poorly differentiated cancer tissues.
Collapse
|
42
|
Enlightened protein: Fhit tumor suppressor protein structure and function and its role in the toxicity of protoporphyrin IX-mediated photodynamic reaction. Toxicol Appl Pharmacol 2009; 241:246-52. [PMID: 19716840 DOI: 10.1016/j.taap.2009.08.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 08/18/2009] [Accepted: 08/20/2009] [Indexed: 12/12/2022]
Abstract
The Fhit tumor suppressor protein possesses Ap(3)A (diadenosine triphosphate - ApppA) hydrolytic activity in vitro and its gene is found inactive in many pre-malignant states due to gene inactivation. For several years Fhit has been a widely investigated protein as its cellular function still remains largely unsolved. Fhit was shown to act as a molecular 'switch' of cell death via cascade operating on the influence of ATR-Chk1 pathway but also through the mitochondrial apoptotic pathway. Notably, Fhit was reported by our group to enhance the overall eradication effect of porphyrin-mediated photodynamic treatment (PDT). In this review the up-to-date findings on Fhit protein as a tumor suppressor and its role in PDT are presented.
Collapse
|