51
|
Chang IW, Lin VCH, Hung CH, Wang HP, Lin YY, Wu WJ, Huang CN, Li CC, Li WM, Wu JY, Li CF. GPX2 underexpression indicates poor prognosis in patients with urothelial carcinomas of the upper urinary tract and urinary bladder. World J Urol 2015; 33:1777-1789. [PMID: 25813210 DOI: 10.1007/s00345-015-1522-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 03/01/2015] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Oxidative stress is believed to be one of the important etiologies in carcinogenesis that has not been systemically investigated in urothelial carcinoma (UC). Through data mining from a published transcriptomic database of UC of urinary bladders (UBUCs) (GSE31684), glutathione peroxidase 2 (GPX2) was identified as the most significant downregulated gene among those response to oxidative stress (GO:0006979). We therefore analyze GPX2 transcript and protein expressions and its clinicopathological significance. METHODS Real-time RT-PCR assay was used to detect GPX2 mRNA level in 20 fresh UBUC specimens. Immunohistochemistry was used to determine GPX2 protein expression in 340 urothelial carcinomas of upper tracts (UTUCs) and 295 UBUCs with mean/median follow-up of 44.7/38.9 and 30.8/23.1 months, respectively. Its expression status was further correlated with clinicopathological features and evaluated for its impact on disease-specific survival and metastasis-free survival (MeFS). RESULTS Decrease in GPX2 transcript level was associated with both higher pT and positive nodal status in 20 UBUCs (all p < 0.05). GPX2 protein underexpression was also significantly associated with advanced pT status, nodal metastasis, high histological grade, vascular invasion, and frequent mitoses in both groups of UCs (all p < 0.05). GPX2 underexpression not only predicted dismal DDS and MeFS at univariate analysis, but also implicated worse DDS (UTUC, p = 0.002; UBUC, p = 0.029) and MeFS (UTUC, p = 0.001; UBUC, p = 0.032) in multivariate analysis. CONCLUSIONS GPX2 underexpression is associated with advanced tumor status and implicated unfavorable clinical outcome of UCs, suggesting its role in tumor progression and may serve as a theranostic biomarker of UCs.
Collapse
Affiliation(s)
- I-Wei Chang
- Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung, Taiwan
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Victor Chia-Hsiang Lin
- Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung, Taiwan
- Department of Urology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chih-Hsin Hung
- Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung, Taiwan
| | - Hua-Pin Wang
- Department of Urology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yung-Yao Lin
- Department of Urology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Wen-Jeng Wu
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan
| | - Chun-Nung Huang
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Chia Li
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Wei-Ming Li
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jui-Yu Wu
- College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Feng Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Pathology, Chi Mei Foundation Medical Center, No. 901, Zhonghua Rd., Yongkang Dist., Tainan, 701, Taiwan.
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| |
Collapse
|
52
|
Hu Y, Chen G. Pathogenic mechanisms of lung adenocarcinoma in smokers and non-smokers determined by gene expression interrogation. Oncol Lett 2015; 10:1350-1370. [PMID: 26622675 DOI: 10.3892/ol.2015.3462] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/19/2015] [Indexed: 12/15/2022] Open
Abstract
Cigarette smoking is the leading risk factor for lung cancer, which accounts for the highest number of cancer-related mortalities worldwide in men and women. Individuals with a history of smoking are 15-30 times more likely to develop lung cancer compared with those who do not smoke. However, our understanding of the underlying molecular mechanisms that contribute to lung tumorigenesis in smokers versus non-smokers remains incomplete. In order to investigate such mechanisms, the present study aimed to systemically interrogate microarray datasets from tumor biopsies and matching normal tissues from stage I and II lung adenocarcinoma patients who had never smoked or were current smokers. The gene expression analysis identified 422 (99 upregulated and 323 downregulated) and 534 (174 upregulated and 360 downregulated) differentially-expressed genes from the never-smokers and current smokers, respectively, and the two groups shared 277 genes that exhibited similar trends of alteration. These genes encode regulators that are involved in a variety of cellular functions, including collagen metabolism and homeostasis of caveolae plasma membranes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes characterization indicated that biological pathways, including extracellular matrix-receptor interaction and cell migration and proliferation, were all affected in the lung cancer patients regardless of the smoking status. However, smoking induced a unique gene expression pattern characterized by upregulation of cell cycle regulators (CDK1, CCNB1 and CDC20), as well as significantly affected biological networks, including p53 signaling pathways. Taken together, these findings suggest novel mechanistic insights, and provide an improved understanding of the smoking-induced molecular alterations that contribute to the pathogenesis of lung adenocarcinoma.
Collapse
Affiliation(s)
- Yunqian Hu
- Department of Respiration, East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Guohan Chen
- Department of Thoracic Surgery, East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| |
Collapse
|
53
|
Samman M, Wood HM, Conway C, Stead L, Daly C, Chalkley R, Berri S, Senguven B, Ross L, Egan P, Chengot P, Ong TK, Pentenero M, Gandolfo S, Cassenti A, Cassoni P, Al Ajlan A, Samkari A, Barrett W, MacLennan K, High A, Rabbitts P. A novel genomic signature reclassifies an oral cancer subtype. Int J Cancer 2015; 137:2364-73. [PMID: 26014678 DOI: 10.1002/ijc.29615] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/15/2015] [Indexed: 12/22/2022]
Abstract
Verrucous carcinoma of the oral cavity (OVC) is considered a subtype of classical oral squamous cell carcinoma (OSCC). Diagnosis is problematic, and additional biomarkers are needed to better stratify patients. To investigate their molecular signature, we performed low-coverage copy number (CN) sequencing on 57 OVC and exome and RNA sequencing on a subset of these and compared the data to the same OSCC parameters. CN results showed that OVC lacked any of the classical OSCC patterns such as gain of 3q and loss of 3p and demonstrated considerably fewer genomic rearrangements compared to the OSCC cohort. OVC and OSCC samples could be clearly differentiated. Exome sequencing showed that OVC samples lacked mutations in genes commonly associated with OSCC (TP53, NOTCH1, NOTCH2, CDKN2A and FAT1). RNA sequencing identified genes that were differentially expressed between the groups. In silico functional analysis showed that the mutated and differentially expressed genes in OVC samples were involved in cell adhesion and keratinocyte proliferation, while those in the OSCC cohort were enriched for cell death and apoptosis pathways. This is the largest and most detailed genomic and transcriptomic analysis yet performed on this tumour type, which, as an example of non-metastatic cancer, may shed light on the nature of metastases. These three independent investigations consistently show substantial differences between the cohorts. Taken together, they lead to the conclusion that OVC is not a subtype of OSCC, but should be classified as a distinct entity.
Collapse
Affiliation(s)
- Manar Samman
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom.,Pathology and Clinical Laboratory Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Henry M Wood
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Caroline Conway
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Lucy Stead
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Catherine Daly
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Rebecca Chalkley
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Stefano Berri
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Burcu Senguven
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Lisa Ross
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Philip Egan
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Preetha Chengot
- St James's Institute of Oncology, St James's University Hospital, Leeds, United Kingdom
| | - Thian K Ong
- Leeds Dental Institute, Leeds General Infirmary, Leeds, United Kingdom
| | - Monica Pentenero
- Oral Medicine and Oral Oncology Unit, Department of Oncology, University of Torino, Turin, Italy
| | - Sergio Gandolfo
- Oral Medicine and Oral Oncology Unit, Department of Oncology, University of Torino, Turin, Italy
| | - Adele Cassenti
- Pathology Unit, Department of Medical Sciences, University of Torino, Turin, Italy
| | - Paola Cassoni
- Pathology Unit, Department of Medical Sciences, University of Torino, Turin, Italy
| | | | - Alaa Samkari
- National Guard Health Affairs, Riyadh, Saudi Arabia
| | | | - Kenneth MacLennan
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom.,St James's Institute of Oncology, St James's University Hospital, Leeds, United Kingdom
| | - Alec High
- St James's Institute of Oncology, St James's University Hospital, Leeds, United Kingdom.,Leeds Dental Institute, Leeds General Infirmary, Leeds, United Kingdom
| | - Pamela Rabbitts
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
54
|
Pastel E, Pointud JC, Loubeau G, Dani C, Slim K, Martin G, Volat F, Sahut-Barnola I, Val P, Martinez A, Lefrançois-Martinez AM. Aldose reductases influence prostaglandin F2α levels and adipocyte differentiation in male mouse and human species. Endocrinology 2015; 156:1671-84. [PMID: 25730106 DOI: 10.1210/en.2014-1750] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aldose reductases (AKR1B) are widely expressed oxidoreductases whose physiological function remains elusive. Some isoforms are genuine prostaglandin F2α (PGF2α) synthases, suggesting they might influence adipose homeostasis because PGF2α inhibits adipogenesis. This was shown by Akr1b7 gene ablation in the mouse, which resulted in increased adiposity related to a lower PGF2α content in fat. Yet humans have no ortholog gene for Akr1b7, so the role of aldose reductases in human adipose homeostasis remains to be explored. We analyzed expression of genes encoding human and mouse aldose reductase isoforms in adipose tissues and differentiating adipocytes to assess conserved mechanisms regulating PGF2α synthesis and adipogenesis. The Akr1b3 gene encoded the most abundant isoform in mouse adipose tissue, whereas Akr1b7 encoded the only isoform enriched in the stromal vascular fraction. Most mouse aldose reductase gene expression peaked in early adipogenesis of 3T3-L1 cells and diminished with differentiation. In contrast with its mouse ortholog Akr1b3, AKR1B1 expression increased throughout differentiation of human multipotent adipose-derived stem cells, paralleling PGF2α release, whereas PGF2α receptor (FP) levels collapsed in early differentiation. Pharmacological inhibition of aldose reductase using Statil altered PGF2α production and enhanced human multipotent adipose-derived stem adipocyte differentiation. As expected, the adipogenic effects of Statil were counteracted by an FP agonist (cloprostenol). Thus, in both species aldose reductase-dependent PGF2α production could be important in early differentiation to restrict adipogenesis. PGF2α antiadipogenic signaling could then be toned down through the FP receptor or aldose reductases down-regulation in human and mouse cells, respectively. Our data suggest that aldose reductase inhibitors could have obesogenic potential.
Collapse
Affiliation(s)
- Emilie Pastel
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 6293 (E.P., J.-C.P., G.L., I.S.-B., P.V., A.M., A.-M.L.-M.), INSERM Unité 1103, Génétique Reproduction et Développement, Clermont Université, 63171 Aubière, France; iBV (C.D.), Institute of Biology Valrose, Université Nice Sophia Antipolis, 06189 Nice, France; Service de Chirurgie Digestive (K.S., G.M.), Centre Hospitalier Universitaire Estaing, 63003 Clermont-Ferrand, France; and INSERM Unité Mixte de Recherche 1048 (F.V.), Institute of Metabolic and Cardiovascular Diseases, Université Paul Sabatier, 31432 Toulouse, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Morikawa Y, Kezuka C, Endo S, Ikari A, Soda M, Yamamura K, Toyooka N, El-Kabbani O, Hara A, Matsunaga T. Acquisition of doxorubicin resistance facilitates migrating and invasive potentials of gastric cancer MKN45 cells through up-regulating aldo-keto reductase 1B10. Chem Biol Interact 2015; 230:30-9. [PMID: 25686905 DOI: 10.1016/j.cbi.2015.02.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/02/2015] [Accepted: 02/06/2015] [Indexed: 12/12/2022]
Abstract
Continuous exposure to doxorubicin (DOX) accelerates hyposensitivity to the drug-elicited lethality of gastric cells, with increased risks of the recurrence and serious cardiovascular side effects. However, the detailed mechanisms underlying the reduction of DOX sensitivity remain unclear. In this study, we generated a DOX-resistant variant upon continuously treating human gastric cancer MKN45 cells with incremental concentrations of the drug, and investigated whether the gain of DOX resistance influences gene expression of four aldo-keto reductases (AKRs: 1B10, 1C1, 1C2 and 1C3). RT-PCR analysis revealed that among the enzymes AKR1B10 is most highly up-regulated during the chemoresistance induction. The up-regulation of AKR1B10 was confirmed by analyses of Western blotting and enzyme activity. The DOX sensitivity of MKN45 cells was reduced and elevated by overexpression and inhibition of AKR1B10, respectively. Compared to the parental MKN45 cells, the DOX-resistant cells had higher migrating and invasive abilities, which were significantly suppressed by addition of AKR1B10 inhibitors. Zymographic and real-time PCR analyses also revealed significant increases in secretion and expression of matrix metalloproteinase (MMP) 2 associated with DOX resistance. Moreover, the overexpression of AKR1B10 in the parental cells remarkably facilitated malignant progression (elevation of migrating and invasive potentials) and MMP2 secretion, which were lowered by the AKR1B10 inhibitors. These results suggest that AKR1B10 is a DOX-resistance gene in the gastric cancer cells, and is responsible for elevating the migrating and invasive potentials of the cells through induction of MMP2.
Collapse
Affiliation(s)
- Yoshifumi Morikawa
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Chihiro Kezuka
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Midori Soda
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, Nagoya 464-8650, Japan
| | - Keiko Yamamura
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, Nagoya 464-8650, Japan
| | - Naoki Toyooka
- Graduate School of Science and Technology for Research, University of Toyama, Toyama 930-8555, Japan
| | - Ossama El-Kabbani
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Akira Hara
- Faculty of Engineering, Gifu University, Gifu 501-1193, Japan
| | - Toshiyuki Matsunaga
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan.
| |
Collapse
|
56
|
Guo X, Song J, Guan T, Wang S, Wang Y, Meng Y, Guo J, Li T, Ma C, Wei J. Characterization of recombinant human gastrointestinal glutathione peroxidase mutant produced inEscherichia coli. Free Radic Res 2015; 49:228-35. [DOI: 10.3109/10715762.2014.995182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
57
|
Vizuete W, Sexton KG, Nguyen H, Smeester L, Aagaard KM, Shope C, Lefer B, Flynn JH, Alvarez S, Erickson MH, Fry RC. From the Field to the Laboratory: Air Pollutant-Induced Genomic Effects in Lung Cells. ENVIRONMENTAL HEALTH INSIGHTS 2015; 9:15-23. [PMID: 26917966 PMCID: PMC4760675 DOI: 10.4137/ehi.s15656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/01/2015] [Accepted: 12/04/2015] [Indexed: 05/18/2023]
Abstract
Current in vitro studies do not typically assess cellular impacts in relation to real-world atmospheric mixtures of gases. In this study, we set out to examine the feasibility of measuring biological responses at the level of gene expression in human lung cells upon direct exposures to air in the field. This study describes the successful deployment of lung cells in the heavily industrialized Houston Ship Channel. By examining messenger RNA (mRNA) levels from exposed lung cells, we identified changes in genes that play a role as inflammatory responders in the cell. The results show anticipated responses from negative and positive controls, confirming the integrity of the experimental protocol and the successful deployment of the in vitro instrument. Furthermore, exposures to ambient conditions displayed robust changes in gene expression. These results demonstrate a methodology that can produce gas-phase toxicity data in the field.
Collapse
Affiliation(s)
- William Vizuete
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- CORRESPONDENCE:
| | - Kenneth G. Sexton
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hang Nguyen
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lisa Smeester
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Cynthia Shope
- Division of Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Barry Lefer
- Department of Earth and Atmospheric Sciences, University of Houston, Houston, TX, USA
| | - James H. Flynn
- Department of Earth and Atmospheric Sciences, University of Houston, Houston, TX, USA
| | - Sergio Alvarez
- Department of Earth and Atmospheric Sciences, University of Houston, Houston, TX, USA
| | - Mathew H. Erickson
- Department of Earth and Atmospheric Sciences, University of Houston, Houston, TX, USA
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
58
|
Zhang W, Li H, Yang Y, Liao J, Yang GY. Knockdown or inhibition of aldo-keto reductase 1B10 inhibits pancreatic carcinoma growth via modulating Kras-E-cadherin pathway. Cancer Lett 2014; 355:273-80. [PMID: 25304374 PMCID: PMC4462172 DOI: 10.1016/j.canlet.2014.09.031] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/12/2014] [Accepted: 09/24/2014] [Indexed: 01/13/2023]
Abstract
Aldo-keto reductase 1B10 (AKR1B10) has relatively specific lipid substrates including carbonyls, retinal and farnesal/geranylgeranial. Metabolizing these lipid substrates appears crucial to carcinogenesis, particularly for farnesal/geranylgeranial that involves protein prenylation. Mutant Kras is a most common active oncogene in pancreatic cancer, and its activation requires protein prenylation. To directly determine the role of AKR1B10 in pancreatic carcinogenesis, we knocked down AKR1B10 in CD18 human pancreatic carcinoma cells using shRNA approach. Silencing AKR1B10 resulted in a significant inhibition of anchor-dependent growth (knockdown cells vs. vector-control cells: 67 ± 9.5 colonies/HPF vs. 170 ± 3.7 colonies/HPF, p < 0.01), invasion index (0.27 vs. 1.00, p < 0.05), and cell migration (at 16 hours 9.2 ± 1.2% vs. 14.0 ± 1.8%, at 24 hours 21.0 ± 1.1% vs. 30.5 ± 3.5%, and at 48 hours 51.9 ± 5.7% vs. 88.9 ± 3.0%, p < 0.01). Inhibition of AKR1B10 by oleanolic acid (OA) showed a dose-dependent inhibition of cell growth with IC50 at 30 µM. Kras pull-down and Western blot analysis revealed a significant down-regulation of active form Kras and phosphorylated C-Raf, and Erk, as well as an up-regulation of E-cadherin. A significant reduction of in vivo tumor growth was observed in nude mice implanted with the CD18 pancreatic carcinoma cells with AKR1B10 knockdown (tumor weight: 0.25 ± 0.06 g vs. 0.52 ± 0.07 g, p = 0.01), and with OA treatment (tumor weight: 0.35 ± 0.05 g vs. 0.52 ± 0.07 g, p = 0.05). Our findings indicate AKR1B10 is a unique enzyme involved in pancreatic carcinogenesis via modulation of the Kras-E-cadherin pathway.
Collapse
Affiliation(s)
- Wanying Zhang
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Haonan Li
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yihe Yang
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jie Liao
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Guang-Yu Yang
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
59
|
Human aldo-keto reductases and the metabolic activation of polycyclic aromatic hydrocarbons. Chem Res Toxicol 2014; 27:1901-17. [PMID: 25279998 PMCID: PMC4237494 DOI: 10.1021/tx500298n] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
Aldo-keto reductases (AKRs) are promiscuous
NAD(P)(H) dependent
oxidoreductases implicated in the metabolic activation of polycyclic
aromatic hydrocarbons (PAH). These enzymes catalyze the oxidation
of non-K-region trans-dihydrodiols to the corresponding o-quinones with the concomitant production of reactive oxygen
species (ROS). The PAH o-quinones are Michael acceptors
and can form adducts but are also redox-active and enter into futile
redox cycles to amplify ROS formation. Evidence exists to support
this metabolic pathway in humans. The human recombinant AKR1A1 and
AKR1C1–AKR1C4 enzymes all catalyze the oxidation of PAH trans-dihydrodiols to PAH o-quinones. Many
human AKRs also catalyze the NADPH-dependent reduction of the o-quinone products to air-sensitive catechols, exacerbating
ROS formation. Moreover, this pathway of PAH activation occurs in
a panel of human lung cell lines, resulting in the production of ROS
and oxidative DNA damage in the form of 8-oxo-2′-deoxyguanosine.
Using stable-isotope dilution liquid chromatography tandem mass spectrometry,
this pathway of benzo[a]pyrene (B[a]P) metabolism was found to contribute equally with the diol-epoxide
pathway to the activation of this human carcinogen in human lung cells.
Evaluation of the mutagenicity of anti-B[a]P-diol epoxide with B[a]P-7,8-dione on
p53 showed that the o-quinone produced by AKRs was
the more potent mutagen, provided that it was permitted to redox cycle,
and that the mutations observed were G to T transversions, reminiscent
of those observed in human lung cancer. It is concluded that there
is sufficient evidence to support the role of human AKRs in the metabolic
activation of PAH in human lung cell lines and that they may contribute
to the causation of human lung cancer.
Collapse
|
60
|
Induction of aldo-keto reductases (AKR1C1 and AKR1C3) abolishes the efficacy of daunorubicin chemotherapy for leukemic U937 cells. Anticancer Drugs 2014; 25:868-77. [DOI: 10.1097/cad.0000000000000112] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
61
|
Karlsson A, Ringnér M, Lauss M, Botling J, Micke P, Planck M, Staaf J. Genomic and transcriptional alterations in lung adenocarcinoma in relation to smoking history. Clin Cancer Res 2014; 20:4912-24. [PMID: 25037737 DOI: 10.1158/1078-0432.ccr-14-0246] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Cigarette smoking is the major pathogenic factor for lung cancer. The precise mechanisms of tobacco-related carcinogenesis and its effect on the genomic and transcriptional landscape in lung cancer are not fully understood. EXPERIMENTAL DESIGN A total of 1,398 (277 never-smokers and 1,121 smokers) genomic and 1,449 (370 never-smokers and 1,079 smokers) transcriptional profiles were assembled from public lung adenocarcinoma cohorts, including matched next-generation DNA-sequencing data (n = 423). Unsupervised and supervised methods were used to identify smoking-related copy-number alterations (CNAs), predictors of smoking status, and molecular subgroups. RESULTS Genomic meta-analyses showed that never-smokers and smokers harbored a similar frequency of total CNAs, although specific regions (5q, 8q, 16p, 19p, and 22q) displayed a 20% to 30% frequency difference between the two groups. Importantly, supervised classification analyses based on CNAs or gene expression could not accurately predict smoking status (balanced accuracies ∼60% to 80%). However, unsupervised multicohort transcriptional profiling stratified adenocarcinomas into distinct molecular subgroups with specific patterns of CNAs, oncogenic mutations, and mutation transversion frequencies that were independent of the smoking status. One subgroup included approximately 55% to 90% of never-smokers and approximately 20% to 40% of smokers (both current and former) with molecular and clinical features of a less aggressive and smoking-unrelated disease. Given the considerable intragroup heterogeneity in smoking-defined subgroups, especially among former smokers, our results emphasize the clinical importance of accurate molecular characterization of lung adenocarcinoma. CONCLUSIONS The landscape of smoking-related CNAs and transcriptional alterations in adenocarcinomas is complex, heterogeneous, and with moderate differences. Our results support a molecularly distinct less aggressive adenocarcinoma entity, arising in never-smokers and a subset of smokers.
Collapse
Affiliation(s)
- Anna Karlsson
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden. CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| | - Markus Ringnér
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden. CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| | - Martin Lauss
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Maria Planck
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden. CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| | - Johan Staaf
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden. CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden.
| |
Collapse
|
62
|
Matsunaga T, Morikawa Y, Haga M, Endo S, Soda M, Yamamura K, El-Kabbani O, Tajima K, Ikari A, Hara A. Exposure to 9,10-phenanthrenequinone accelerates malignant progression of lung cancer cells through up-regulation of aldo-keto reductase 1B10. Toxicol Appl Pharmacol 2014; 278:180-9. [PMID: 24813866 DOI: 10.1016/j.taap.2014.04.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/21/2014] [Accepted: 04/26/2014] [Indexed: 01/13/2023]
Abstract
Inhalation of 9,10-phenanthrenequinone (9,10-PQ), a major quinone in diesel exhaust, exerts fatal damage against a variety of cells involved in respiratory function. Here, we show that treatment with high concentrations of 9,10-PQ evokes apoptosis of lung cancer A549 cells through production of reactive oxygen species (ROS). In contrast, 9,10-PQ at its concentrations of 2 and 5 μM elevated the potentials for proliferation, invasion, metastasis and tumorigenesis, all of which were almost completely inhibited by addition of an antioxidant N-acetyl-l-cysteine, inferring a crucial role of ROS in the overgrowth and malignant progression of lung cancer cells. Comparison of mRNA expression levels of six aldo-keto reductases (AKRs) in the 9,10-PQ-treated cells advocated up-regulation of AKR1B10 as a major cause contributing to the lung cancer malignancy. In support of this, the elevation of invasive, metastatic and tumorigenic activities in the 9,10-PQ-treated cells was significantly abolished by the addition of a selective AKR1B10 inhibitor oleanolic acid. Intriguingly, zymographic and real-time PCR analyses revealed remarkable increases in secretion and expression, respectively, of matrix metalloproteinase 2 during the 9,10-PQ treatment, and suggested that the AKR1B10 up-regulation and resultant activation of mitogen-activated protein kinase cascade are predominant mechanisms underlying the metalloproteinase induction. In addition, HPLC analysis and cytochrome c reduction assay in in vitro 9,10-PQ reduction by AKR1B10 demonstrated that the enzyme catalyzes redox-cycling of this quinone, by which ROS are produced. Collectively, these results suggest that AKR1B10 is a key regulator involved in overgrowth and malignant progression of the lung cancer cells through ROS production due to 9,10-PQ redox-cycling.
Collapse
Affiliation(s)
- Toshiyuki Matsunaga
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan.
| | - Yoshifumi Morikawa
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Mariko Haga
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Midori Soda
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, Nagoya 464-8650, Japan
| | - Keiko Yamamura
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, Nagoya 464-8650, Japan
| | - Ossama El-Kabbani
- Monash Institute of Pharmaceutical Sciences, Monash University, Victoria 3052, Australia
| | - Kazuo Tajima
- Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa 920-1181, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Akira Hara
- Faculty of Engineering, Gifu University, Gifu 501-1193, Japan
| |
Collapse
|
63
|
Cooke SL, Shlien A, Marshall J, Pipinikas CP, Martincorena I, Tubio JM, Li Y, Menzies A, Mudie L, Ramakrishna M, Yates L, Davies H, Bolli N, Bignell GR, Tarpey PS, Behjati S, Nik-Zainal S, Papaemmanuil E, Teixeira VH, Raine K, O’Meara S, Dodoran MS, Teague JW, Butler AP, Iacobuzio-Donahue C, Santarius T, Grundy RG, Malkin D, Greaves M, Munshi N, Flanagan AM, Bowtell D, Martin S, Larsimont D, Reis-Filho JS, Boussioutas A, Taylor JA, Hayes ND, Janes SM, Futreal PA, Stratton MR, McDermott U, Campbell PJ. Processed pseudogenes acquired somatically during cancer development. Nat Commun 2014; 5:3644. [PMID: 24714652 PMCID: PMC3996531 DOI: 10.1038/ncomms4644] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 03/13/2014] [Indexed: 12/14/2022] Open
Abstract
Cancer evolves by mutation, with somatic reactivation of retrotransposons being one such mutational process. Germline retrotransposition can cause processed pseudogenes, but whether this occurs somatically has not been evaluated. Here we screen sequencing data from 660 cancer samples for somatically acquired pseudogenes. We find 42 events in 17 samples, especially non-small cell lung cancer (5/27) and colorectal cancer (2/11). Genomic features mirror those of germline LINE element retrotranspositions, with frequent target-site duplications (67%), consensus TTTTAA sites at insertion points, inverted rearrangements (21%), 5' truncation (74%) and polyA tails (88%). Transcriptional consequences include expression of pseudogenes from UTRs or introns of target genes. In addition, a somatic pseudogene that integrated into the promoter and first exon of the tumour suppressor gene, MGA, abrogated expression from that allele. Thus, formation of processed pseudogenes represents a new class of mutation occurring during cancer development, with potentially diverse functional consequences depending on genomic context.
Collapse
Affiliation(s)
- Susanna L. Cooke
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Adam Shlien
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - John Marshall
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | | | - Inigo Martincorena
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Jose M.C. Tubio
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Yilong Li
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Andrew Menzies
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Laura Mudie
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Manasa Ramakrishna
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Lucy Yates
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Helen Davies
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Niccolo Bolli
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
- University of Cambridge, Cambridge CB2 0XY, UK
| | - Graham R. Bignell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Patrick S. Tarpey
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Sam Behjati
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
- University of Cambridge, Cambridge CB2 0XY, UK
| | - Serena Nik-Zainal
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Elli Papaemmanuil
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Vitor H. Teixeira
- Lungs for Living Research Centre, Rayne Institute, University College London, London WC1E 6JF, UK
| | - Keiran Raine
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Sarah O’Meara
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Maryam S. Dodoran
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Jon W. Teague
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Adam P. Butler
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | | | | | - Richard G. Grundy
- Children’s Brain Tumour Research Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - David Malkin
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada M5G 1X8
| | - Mel Greaves
- Institute for Cancer Research, Sutton, London SM2 5NG, UK
| | - Nikhil Munshi
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Adrienne M. Flanagan
- Lungs for Living Research Centre, Rayne Institute, University College London, London WC1E 6JF, UK
- Royal National Orthopaedic Hospital, Middlesex HA7 4LP, UK
| | - David Bowtell
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3002, Australia
| | - Sancha Martin
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Denis Larsimont
- Department of Pathology, Jules Bordet Institute, 1000 Brussels, Belgium
| | - Jorge S. Reis-Filho
- Department of Pathology, Memorial-Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | - Alex Boussioutas
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3002, Australia
- Department of Gastroenterology, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Jack A. Taylor
- National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27713, USA
| | - Neil D. Hayes
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Sam M. Janes
- Lungs for Living Research Centre, Rayne Institute, University College London, London WC1E 6JF, UK
| | - P. Andrew Futreal
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Michael R. Stratton
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Ultan McDermott
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
- Addenbrooke’s NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Peter J. Campbell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
- University of Cambridge, Cambridge CB2 0XY, UK
- Addenbrooke’s NHS Foundation Trust, Cambridge CB2 0QQ, UK
| |
Collapse
|
64
|
Han SS, Kim WJ, Hong Y, Hong SH, Lee SJ, Ryu DR, Lee W, Cho YH, Lee S, Ryu YJ, Won JY, Rhee H, Park JH, Jang SJ, Lee JS, Choi CM, Lee JC, Lee SD, Oh YM. RNA sequencing identifies novel markers of non-small cell lung cancer. Lung Cancer 2014; 84:229-35. [PMID: 24751108 DOI: 10.1016/j.lungcan.2014.03.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/29/2014] [Accepted: 03/17/2014] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The development of reliable gene expression profiling technology increasingly impacts our understanding of lung cancer biology. Here, we used RNA sequencing (RNA-Seq) to compare the transcriptomes of non-small cell lung cancer (NSCLC) and normal lung tissues and to investigate expression in lung cancer tissues. METHODS We enrolled 88 male patients (mean age, 61.2 years) with NSCLC. RNA-Seq was performed on 88 pairs of NSCLC tumor tissue and non-tumor tissue from 54 patients with adenocarcinoma and 34 patients with squamous cell carcinoma. Immunohistochemistry was performed to validate differential candidate gene expression in a different NSCLC group. RESULTS RNA-Seq produced 25.41 × 10(6) (± 8.90 × 10(6)) reads in NSCLC tissues and 24.70×10(6) (± 4.70 × 10(6)) reads in normal lung tissues [mean (± standard deviation)]. Among the genes expressed in both tissues, 335 were upregulated and 728 were downregulated ≥ 2-fold (p < 0.001). Four upregulated genes - CBX3, GJB2, CRABP2, and DSP - not previously reported in lung cancer were studied further. Their altered expression was verified by immunohistochemistry in a different set of NSCLC tissues (n = 154). CBX3 was positive in 90.3% (139 cases) of the samples; GJB2, in 22.7% (35 cases); CRABP2, in 72.1% (111 cases); and DSP, in 17.5% (27 cases). The positive rate of CRABP2 was higher in adenocarcinoma than squamous cell carcinoma (p < 0.01). CONCLUSIONS CBX3 and CRABP2 expression was markedly increased in lung cancer tissues and especially CRABP2 may be promising candidate genes in lung adenocarcinoma.
Collapse
Affiliation(s)
- Seon-Sook Han
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Woo Jin Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Yoonki Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Seung-Joon Lee
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Dong Ryeol Ryu
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Wonho Lee
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Yo Han Cho
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Seungkoo Lee
- Department of Anatomic Pathology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Young-Joon Ryu
- Department of Pathology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Jun Yeon Won
- Department of Otolaryngology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Kangwon-do 200-701, Republic of Korea
| | - Hwanseok Rhee
- Macrogen Bioinformatics Center, Macrogen, Seoul 153-781, Republic of Korea
| | - Jung Hoon Park
- Macrogen Bioinformatics Center, Macrogen, Seoul 153-781, Republic of Korea
| | - Se Jin Jang
- Department of Pathology and Asan Center for Cancer Genome Discovery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea
| | - Jae Seung Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea; Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea
| | - Chang-Min Choi
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea
| | - Jae Cheol Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea
| | - Sang Do Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea; Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea
| | - Yeon-Mok Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea; Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea.
| |
Collapse
|
65
|
Van Dyck E, Nazarov PV, Muller A, Nicot N, Bosseler M, Pierson S, Van Moer K, Palissot V, Mascaux C, Knolle U, Ninane V, Nati R, Bremnes RM, Vallar L, Berchem G, Schlesser M. Bronchial airway gene expression in smokers with lung or head and neck cancer. Cancer Med 2014; 3:322-36. [PMID: 24497500 PMCID: PMC3987082 DOI: 10.1002/cam4.190] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 09/30/2013] [Accepted: 11/01/2013] [Indexed: 01/10/2023] Open
Abstract
Cigarette smoking is the major cause of cancers of the respiratory tract, including non-small cell lung cancer (NSCLC) and head and neck cancer (HNC). In order to better understand carcinogenesis of the lung and upper airways, we have compared the gene expression profiles of tumor-distant, histologically normal bronchial biopsy specimens obtained from current smokers with NSCLC or HNC (SC, considered as a single group), as well as nonsmokers (NS) and smokers without cancer (SNC). RNA from a total of 97 biopsies was used for gene expression profiling (Affymetrix HG-U133 Plus 2.0 array). Differentially expressed genes were used to compare NS, SNC, and SC, and functional analysis was carried out using Ingenuity Pathway Analysis (IPA). Smoking-related cancer of the respiratory tract was found to affect the expression of genes encoding xenobiotic biotransformation proteins, as well as proteins associated with crucial inflammation/immunity pathways and other processes that protect the airway from the chemicals in cigarette smoke or contribute to carcinogenesis. Finally, we used the prediction analysis for microarray (PAM) method to identify gene signatures of cigarette smoking and cancer, and uncovered a 15-gene signature that distinguished between SNC and SC with an accuracy of 83%. Thus, gene profiling of histologically normal bronchial biopsy specimens provided insight into cigarette-induced carcinogenesis of the respiratory tract and gene signatures of cancer in smokers.
Collapse
Affiliation(s)
- Eric Van Dyck
- Département d'Oncologie, CRP-Santé du Luxembourg, Luxembourg
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Brothers JF, Hijazi K, Mascaux C, El-Zein RA, Spitz MR, Spira A. Bridging the clinical gaps: genetic, epigenetic and transcriptomic biomarkers for the early detection of lung cancer in the post-National Lung Screening Trial era. BMC Med 2013; 11:168. [PMID: 23870182 PMCID: PMC3717087 DOI: 10.1186/1741-7015-11-168] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 06/20/2013] [Indexed: 02/05/2023] Open
Abstract
Lung cancer is the leading cause of cancer death worldwide in part due to our inability to identify which smokers are at highest risk and the lack of effective tools to detect the disease at its earliest and potentially curable stage. Recent results from the National Lung Screening Trial have shown that annual screening of high-risk smokers with low-dose helical computed tomography of the chest can reduce lung cancer mortality. However, molecular biomarkers are needed to identify which current and former smokers would benefit most from annual computed tomography scan screening in order to reduce the costs and morbidity associated with this procedure. Additionally, there is an urgent clinical need to develop biomarkers that can distinguish benign from malignant lesions found on computed tomography of the chest given its very high false positive rate. This review highlights recent genetic, transcriptomic and epigenomic biomarkers that are emerging as tools for the early detection of lung cancer both in the diagnostic and screening setting.
Collapse
|
67
|
Expression of glutathione peroxidase 2 is associated with not only early hepatocarcinogenesis but also late stage metastasis. Toxicology 2013; 311:115-23. [PMID: 23867582 DOI: 10.1016/j.tox.2013.07.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 07/04/2013] [Accepted: 07/05/2013] [Indexed: 01/16/2023]
Abstract
Understanding of mechanisms of cancer progression is very important for reduction of cancer mortality. Of six rat hepatocellular carcinoma (HCC) cell lines, differing in their metastatic potential to the lung after inoculation into the tail vein of nude mice, the most metastatic featured particular overexpression of glutathione peroxidase 2 (GPX2). Therefore, we analyzed the influence of interference in highly metastatic L2 cells by siRNA transfection. Gpx2 siRNA significantly inhibited cell proliferation at 24 and 48h time points with induction of apoptosis but not cell cycle arrest. High expression of mutated p53 was detected in all HCC cell lines, with reduction in Gpx2 siRNA-transfected cells. Migration and invasion in vitro were also suppressed as compared to control siRNA-transfected cells and secretion of matrix metalloproteinase 9 was reduced. In vivo, the numbers and areas of metastatic nodules per area in the lungs were significantly reduced in the mice inoculated with Gpx2 siRNA-transfected cells as compared to control siRNA-transfected cells. In conclusion, expression of GPX2 is associated with cancer metastasis from rat HCCs both in vitro and in vivo. Together with immunohistochemical findings of elevated expression in rat and also human liver lesions, the results point to important roles in hepatocarcinogenesis.
Collapse
|
68
|
Li H, Yang AL, Chung YT, Zhang W, Liao J, Yang GY. Sulindac inhibits pancreatic carcinogenesis in LSL-KrasG12D-LSL-Trp53R172H-Pdx-1-Cre mice via suppressing aldo-keto reductase family 1B10 (AKR1B10). Carcinogenesis 2013; 34:2090-8. [PMID: 23689354 DOI: 10.1093/carcin/bgt170] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sulindac has been identified as a competitive inhibitor of aldo-keto reductase 1B10 (AKR1B10), an enzyme that plays a key role in carcinogenesis. AKR1B10 is overexpressed in pancreatic ductal adenocarcinoma (PDAC) and exhibits lipid substrate specificity, especially for farnesyl and geranylgeranyl. There have been no studies though showing that the inhibition of PDAC by sulindac is via inhibition of AKR1B10, particularly the metabolism of farnesyl/geranylgeranyl and Kras protein prenylation. To determine the chemopreventive effects of sulindac on pancreatic carcinogenesis, 5-week-old LSL-Kras(G12D)-LSL-Trp53(R172H)-Pdx-1-Cre mice (Pan(kras/p53) mice) were fed an AIN93M diet with or without 200 p.p.m. sulindac (n = 20/group). Kaplan-Meier survival analysis showed that average animal survival in Pan(kras/p53) mice was 143.7 ± 8.8 days, and average survival with sulindac was increased to 168.0 ± 8.8 days (P < 0.005). Histopathological analyses revealed that 90% of mice developed PDAC, 10% with metastasis to the liver and lymph nodes. With sulindac, the incidence of PDAC was reduced to 56% (P < 0.01) and only one mouse had lymph node metastasis. Immunochemical analysis showed that sulindac significantly decreased Ki-67-labeled cell proliferation and markedly reduced the expression of phosphorylated extracellular signal-regulated kinases 1 and 2 (ERK1/2), c-Raf and mitogen-activated protein kinase kinase 1 and 2. In in vitro experiments with PDAC cells from Pan(kras/p53) mice, sulindac exhibited dose-dependent inhibition of AKR1B10 activity. By silencing AKR1B10 expression through small interfering RNA or by sulindac treatment, these in vitro models showed a reduction in Kras and human DNA-J homolog 2 protein prenylation, and downregulation of phosphorylated C-raf, ERK1/2 and MEK1/2 expression. Our results demonstrate that sulindac inhibits pancreatic carcinogenesis by the inhibition of Kras protein prenylation by targeting AKR1B10.
Collapse
Affiliation(s)
- Haonan Li
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Ward 6-118, Chicago, IL 60611
| | | | | | | | | | | |
Collapse
|
69
|
Matsunaga T, El-Kabbani O, Hara A. Aldo-Keto Reductases as New Therapeutic Targets for Colon Cancer Chemoresistance. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2013. [DOI: 10.1007/978-1-4614-7070-0_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
70
|
Abstract
Selenium is an essential micronutrient in mammals, but is also recognized as toxic in excess. It is a non-metal with properties that are intermediate between the chalcogen elements sulfur and tellurium. Selenium exerts its biological functions through selenoproteins. Selenoproteins contain selenium in the form of the 21st amino acid, selenocysteine (Sec), which is an analog of cysteine with the sulfur-containing side chain replaced by a Se-containing side chain. Sec is encoded by the codon UGA, which is one of three termination codons for mRNA translation in non-selenoprotein genes. Recognition of the UGA codon as a Sec insertion site instead of stop requires a Sec insertion sequence (SECIS) element in selenoprotein mRNAs and a unique selenocysteyl-tRNA, both of which are recognized by specialized protein factors. Unlike the 20 standard amino acids, Sec is biosynthesized from serine on its tRNA. Twenty-five selenoproteins are encoded in the human genome. Most of the selenoprotein genes were discovered by bioinformatics approaches, searching for SECIS elements downstream of in-frame UGA codons. Sec has been described as having stronger nucleophilic and electrophilic properties than cysteine, and Sec is present in the catalytic site of all selenoenzymes. Most selenoproteins, whose functions are known, are involved in redox systems and signaling pathways. However, several selenoproteins are not well characterized in terms of their function. The selenium field has grown dramatically in the last few decades, and research on selenium biology is providing extensive new information regarding its importance for human health.
Collapse
Affiliation(s)
- Suguru Kurokawa
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA,
| | | |
Collapse
|
71
|
Brody JS. Transcriptome alterations induced by cigarette smoke. Int J Cancer 2012; 131:2754-62. [PMID: 22961494 DOI: 10.1002/ijc.27829] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 08/16/2012] [Indexed: 12/24/2022]
Abstract
Cigarette smoke alters the transcriptome of multiple tissues; those directly exposed to toxic products and those exposed to circulating components and metabolic products of tobacco smoke. In most tissues and organs that have been studied, the smoking transcriptome is characterized by increased expression of antioxidant and xenobiotic genes as well as a wide spectrum of inflammation-related genes, and potential oncogenic genes. Smoking is associated with an increased incidence of cancer in a number of organs both those directly exposed (lungs and airways) and those indirectly exposed (bladder, liver, pancreas). Individual transcriptomic responses vary, based to some degree on as yet to be clarified genetic factors, and likely how and what the individual has smoked. The complexity of individual responses to tobacco exposure and of smoking-related cancers in various organs is beginning to be revealed in transcriptomic and whole genome sequencing studies, of both tumors and cytologically normal appearing cells that have been exposed to cigarette smoke or its products creating a genomic "field of injury." The recent application of next generation sequencing to defining the transcriptome alterations induced by cigarette smoke holds the promise of discovering new approaches to personalized prevention and treatment of smoking-related lung diseases in the future.
Collapse
Affiliation(s)
- Jerome S Brody
- Boston University School of Medicine, Pulmonary Center (R-3), Boston, MA 02118, USA.
| |
Collapse
|
72
|
Abstract
BACKGROUND With increasing evidence that hydroperoxides are not only toxic but rather exert essential physiological functions, also hydroperoxide removing enzymes have to be re-viewed. In mammals, the peroxidases inter alia comprise the 8 glutathione peroxidases (GPx1-GPx8) so far identified. SCOPE OF THE REVIEW Since GPxs have recently been reviewed under various aspects, we here focus on novel findings considering their diverse physiological roles exceeding an antioxidant activity. MAJOR CONCLUSIONS GPxs are involved in balancing the H2O2 homeostasis in signalling cascades, e.g. in the insulin signalling pathway by GPx1; GPx2 plays a dual role in carcinogenesis depending on the mode of initiation and cancer stage; GPx3 is membrane associated possibly explaining a peroxidatic function despite low plasma concentrations of GSH; GPx4 has novel roles in the regulation of apoptosis and, together with GPx5, in male fertility. Functions of GPx6 are still unknown, and the proposed involvement of GPx7 and GPx8 in protein folding awaits elucidation. GENERAL SIGNIFICANCE Collectively, selenium-containing GPxs (GPx1-4 and 6) as well as their non-selenium congeners (GPx5, 7 and 8) became key players in important biological contexts far beyond the detoxification of hydroperoxides. This article is part of a Special Issue entitled Cellular functions of glutathione.
Collapse
Affiliation(s)
- Regina Brigelius-Flohé
- Department of Biochemistry of Micronutrients, German Institute of Human Nutrition, Nuthetal, Germany.
| | | |
Collapse
|
73
|
Zhang L, Jin Y, Huang M, Penning TM. The Role of Human Aldo-Keto Reductases in the Metabolic Activation and Detoxication of Polycyclic Aromatic Hydrocarbons: Interconversion of PAH Catechols and PAH o-Quinones. Front Pharmacol 2012; 3:193. [PMID: 23162467 PMCID: PMC3499756 DOI: 10.3389/fphar.2012.00193] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 10/27/2012] [Indexed: 11/13/2022] Open
Abstract
Polycyclic aromatic hydrocarbons (PAH) are ubiquitous environmental pollutants. They are procarcinogens requiring metabolic activation to elicit their deleterious effects. Aldo-keto reductases (AKR) catalyze the oxidation of proximate carcinogenic PAH trans-dihydrodiols to yield electrophilic and redox-active PAH o-quinones. AKRs are also found to be capable of reducing PAH o-quinones to form PAH catechols. The interconversion of o-quinones and catechols results in the redox-cycling of PAH o-quinones to give rise to the generation of reactive oxygen species and subsequent oxidative DNA damage. On the other hand, PAH catechols can be intercepted through phase II metabolism by which PAH o-quinones could be detoxified and eliminated. The aim of the present review is to summarize the role of human AKRs in the metabolic activation/detoxication of PAH and the relevance of phase II conjugation reactions to human lung carcinogenesis.
Collapse
Affiliation(s)
- Li Zhang
- Center of Excellence in Environmental Toxicology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA
| | | | | | | |
Collapse
|
74
|
Du X, Wang QR, Chan E, Merchant M, Liu J, French D, Ashkenazi A, Qing J. FGFR3 stimulates stearoyl CoA desaturase 1 activity to promote bladder tumor growth. Cancer Res 2012; 72:5843-55. [PMID: 23019225 DOI: 10.1158/0008-5472.can-12-1329] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fibroblast growth factor receptor 3 (FGFR3) belongs to a family of receptor tyrosine kinases that control cell proliferation, differentiation, and survival. Aberrant activation of FGFR3 via overexpression or mutation is a frequent feature of bladder cancer; however, its molecular and cellular consequences and functional relevance to carcinogenesis are not well understood. Through transcriptional profiling of bladder carcinoma cells subjected to short hairpin RNA knockdown of FGFR3, we identified a gene-signature linking FGFR3 signaling with de novo sterol and lipid biosynthesis and metabolism. We found that FGFR3 signaling promotes the cleavage and activation of the master transcriptional regulator of lipogenesis, sterol regulatory element-binding protein 1(SREBP1/SREBF1), in a PI3K-mTORC1-dependent fashion. In turn, SREBP1 regulates the expression of key lipogenic enzymes, including stearoyl CoA desaturase 1 (SCD1/SCD). SCD1 is the rate-limiting enzyme in the biosynthesis of monounsaturated fatty acids and is crucial for lipid homeostasis. In human bladder cancer cell lines expressing constitutively active FGFR3, knockdown of SCD1 by siRNA markedly attenuated cell-cycle progression, reduced proliferation, and induced apoptosis. Furthermore, inducible knockdown of SCD1 in a bladder cancer xenograft model substantially inhibited tumor progression. Pharmacologic inhibition of SCD1 blocked fatty acid desaturation and also exerted antitumor activity in vitro and in vivo. Together, these findings reveal a previously unrecognized role of FGFR3 in regulating lipid metabolism to maintain tumor growth and survival, and also identify SCD1 as a potential therapeutic target for FGFR3-driven bladder cancer.
Collapse
Affiliation(s)
- Xiangnan Du
- Molecular Oncology, Cancer Signaling and Translational Oncology, Bioinformatics, and Pathology, Genentech, Inc, South San Francisco, California 94080, USA
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Abstract
The discovery of multiple selenoproteins has raised tantalizing questions about their role in maintaining normal cellular function. Unfortunately, many of these remain inadequately investigated. While they have a role in maintaining redox balance, other functions are becoming increasingly recognized. As the roles of these selenoproteins are further characterized, a better understanding of the true physiological significance of this trace element will arise. This knowledge will be essential in defining optimum intakes to achieve cellular homeostasis in order to optimize health, including a reduction in cancer, for diverse populations. Human variation in the response to selenium likely reflects significant interactions between the type and amounts of selenium consumed with the genome and a host of environmental factors including the totality of the diet, as discussed in this review.
Collapse
Affiliation(s)
- Cindy D. Davis
- Nutritional Science Research Group, National Cancer Institute, Rockville, Maryland 20892;,
- Current address: Office of Dietary Supplements, National Institutes of Health, Rockville, Maryland 20892
| | - Petra A. Tsuji
- Department of Biological Sciences, Towson University, Towson, Maryland 21252
| | - John A. Milner
- Nutritional Science Research Group, National Cancer Institute, Rockville, Maryland 20892;,
| |
Collapse
|
76
|
Pandiri AR, Sills RC, Ziglioli V, Ton TVT, Hong HHL, Lahousse SA, Gerrish KE, Auerbach SS, Shockley KR, Bushel PR, Peddada SD, Hoenerhoff MJ. Differential transcriptomic analysis of spontaneous lung tumors in B6C3F1 mice: comparison to human non-small cell lung cancer. Toxicol Pathol 2012; 40:1141-59. [PMID: 22688403 DOI: 10.1177/0192623312447543] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lung cancer is the leading cause of cancer-related death in people and is mainly due to environmental factors such as smoking and radon. The National Toxicology Program (NTP) tests various chemicals and mixtures for their carcinogenic hazard potential. In the NTP chronic bioassay using B6C3F1 mice, the incidence of lung tumors in treated and control animals is second only to the liver tumors. In order to study the molecular mechanisms of chemically induced lung tumors, an understanding of the genetic changes that occur in spontaneous lung (SL) tumors from untreated control animals is needed. The authors have evaluated the differential transcriptomic changes within SL tumors compared to normal lungs from untreated age-matched animals. Within SL tumors, several canonical pathways associated with cancer (eukaryotic initiation factor 2 signaling, RhoA signaling, PTEN signaling, and mammalian target of rapamycin signaling), metabolism (Inositol phosphate metabolism, mitochondrial dysfunction, and purine and pyramidine metabolism), and immune responses (FcγR-mediated phagocytosis, clathrin-mediated endocytosis, interleukin 8 signaling, and CXCR4 signaling) were altered. Meta-analysis of murine SL tumors and human non-small cell lung cancer transcriptomic data sets revealed a high concordance. These data provide important information on the differential transcriptomic changes in murine SL tumors that will be critical to our understanding of chemically induced lung tumors and will aid in hazard analysis in the NTP 2-year carcinogenicity bioassays.
Collapse
Affiliation(s)
- Arun R Pandiri
- Cellular and Molecular Pathology Branch, National Toxicology Program-NTP, National Institute of Environmental Health Sciences-NIEHS, Research Triangle Park, North Carolina, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Laffin B, Petrash JM. Expression of the Aldo-Ketoreductases AKR1B1 and AKR1B10 in Human Cancers. Front Pharmacol 2012; 3:104. [PMID: 22685431 PMCID: PMC3368246 DOI: 10.3389/fphar.2012.00104] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/13/2012] [Indexed: 01/27/2023] Open
Abstract
The American Cancer Society estimates that there will be more than 1.5 million new cases of cancer in 2011, underscoring the need for identification of new therapeutic targets and development of novel cancer therapies. Previous studies have implicated the human aldo-ketoreductases AKR1B1 and AKR1B10 in cancer, and therefore we examined AKR1B1 and AKR1B10 expression across all major human cancer types using the Oncomine cancer gene expression database (Compendia Biosciences, www.oncomine.com). Using this database, we found that expression of AKR1B1 and AKR1B10 varies greatly by cancer type and tissue of origin, including agreement with previous reports that AKR1B10 is significantly over-expressed in cancers of the lungs and liver. AKR1B1 is more broadly over-expressed in human cancers than AKR1B10, albeit at a generally lower magnitude. AKR1B1 over-expression was found to be associated with shortened patient survival in acute myelogenous leukemias and multiple myelomas. High AKR1B10 expression tends to predict less aggressive clinical course generally, notably within lung cancers, where it tends to be highly over-expressed compared to normal tissue. These findings suggest that AKR1B1 inhibitors in particular hold great potential as novel cancer therapeutics.
Collapse
Affiliation(s)
- Brian Laffin
- Department of Ophthalmology, The School of Medicine, University of Colorado Aurora, CO, USA
| | | |
Collapse
|
78
|
Bossé Y, Postma DS, Sin DD, Lamontagne M, Couture C, Gaudreault N, Joubert P, Wong V, Elliott M, van den Berge M, Brandsma CA, Tribouley C, Malkov V, Tsou JA, Opiteck GJ, Hogg JC, Sandford AJ, Timens W, Paré PD, Laviolette M. Molecular signature of smoking in human lung tissues. Cancer Res 2012; 72:3753-63. [PMID: 22659451 DOI: 10.1158/0008-5472.can-12-1160] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cigarette smoking is the leading risk factor for lung cancer. To identify genes deregulated by smoking and to distinguish gene expression changes that are reversible and persistent following smoking cessation, we carried out genome-wide gene expression profiling on nontumor lung tissue from 853 patients with lung cancer. Gene expression levels were compared between never and current smokers, and time-dependent changes in gene expression were studied in former smokers. A total of 3,223 transcripts were differentially expressed between smoking groups in the discovery set (n = 344, P < 1.29 × 10(-6)). A substantial number of smoking-induced genes also were validated in two replication sets (n = 285 and 224), and a gene expression signature of 599 transcripts consistently segregated never from current smokers across all three sets. The expression of the majority of these genes reverted to never-smoker levels following smoking cessation, although the time course of normalization differed widely among transcripts. Moreover, some genes showed very slow or no reversibility in expression, including SERPIND1, which was found to be the most consistent gene permanently altered by smoking in the three sets. Our findings therefore indicate that smoking deregulates many genes, many of which reverse to normal following smoking cessation. However, a subset of genes remains altered even decades following smoking cessation and may account, at least in part, for the residual risk of lung cancer among former smokers. Cancer Res; 72(15); 3753-63. ©2012 AACR.
Collapse
Affiliation(s)
- Yohan Bossé
- Department of Molecular Medicine, Laval University, Québec, QC, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
The yin and yang of nrf2-regulated selenoproteins in carcinogenesis. Int J Cell Biol 2012; 2012:486147. [PMID: 22654914 PMCID: PMC3357939 DOI: 10.1155/2012/486147] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 02/20/2012] [Indexed: 02/07/2023] Open
Abstract
The NF-E2-related factor-2 (Nrf2) is a transcription factor which regulates the major cellular defense systems and thereby contributes to the prevention of many diseases including cancer. Selenium deficiency is associated with a higher cancer risk making also this essential trace element a promising candidate for cancer prevention. Two selenoproteins, thioredoxin reductase-1 (TrxR1) and glutathione peroxidase-2 (GPx2), are targets for Nrf2. Selenium deficiency activates Nrf2 as does a TrxR1 knockout making a synergism between both systems plausible. Although this might hold true for healthy cells, the interplay may turn into the opposite in cancer cells. The induction of the detoxifying and antioxidant enzymes by Nrf2 will make cancer cells chemoresistant and will protect them against oxidative damage. The essential role of TrxR1 in maintaining proliferation makes its upregulation in cancer cells detrimental. The anti-inflammatory potential of GPx2 will help to inhibit cancer initiation and inflammation-triggered promotion, but its growth supporting potential will also support tumor growth. This paper considers beneficial and adverse consequences of the activation of Nrf2 and the selenoproteins which appear to depend on the cancer stage.
Collapse
|
80
|
Abstract
INTRODUCTION The recent DNA methylation studies on cancers have revealed the necessity of profiling an entire human genome and not to restrict the profiling to specific regions of the human genome. It has been suggested that genome-wide DNA methylation analysis enables us to identify the genes that are regulated by DNA methylation in carcinogenesis. METHODS So, we performed whole-genome DNA methylation analysis for human lung squamous cell carcinoma (SCC), which is strongly related with smoking. We also performed microarrays using 21 pairs of normal lung tissues and tumors from patients with SCC. By combining these data, 30 hypermethylated and down-regulated genes, and 22 hypomethylated and up-regulated genes were selected. The gene expression level and DNA methylation pattern were confirmed by semiquantitative reverse-transcriptase polymerase chain reaction and pyrosequencing, respectively. RESULTS By these validations, we selected five hypermethylated and down-regulated genes and one hypomethylated and up-regulated gene. Moreover, these six genes were proven to be actually regulated by DNA methylation by confirming the recovery of their DNA methylation pattern and gene expression level using a demethylating agent. The DNA methylation pattern of the CYTL1 promoter region was significantly different between early and advanced stages of SCC. CONCLUSION In conclusion, by combining the whole-genome DNA methylation pattern and the gene expression profile, we identified the six genes (CCDC37, CYTL1, CDO1, SLIT2, LMO3, and SERPINB5) that are regulated by DNA methylation, and we suggest their value as target molecules for further study of SCC.
Collapse
|
81
|
Guo NL, Wan YW. Pathway-based identification of a smoking associated 6-gene signature predictive of lung cancer risk and survival. Artif Intell Med 2012; 55:97-105. [PMID: 22326768 DOI: 10.1016/j.artmed.2012.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 11/07/2011] [Accepted: 01/17/2012] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Smoking is a prominent risk factor for lung cancer. However, it is not an established prognostic factor for lung cancer in clinics. To date, no gene test is available for diagnostic screening of lung cancer risk or prognostication of clinical outcome in smokers. This study sought to identify a smoking associated gene signature in order to provide a more precise diagnosis and prognosis of lung cancer in smokers. METHODS AND MATERIALS An implication network based methodology was used to identify biomarkers by modeling crosstalk with major lung cancer signaling pathways. Specifically, the methodology contains the following steps: (1) identifying genes significantly associated with lung cancer survival; (2) selecting candidate genes which are differentially expressed in smokers versus non-smokers from the survival genes identified in Step 1; (3) from these candidate genes, constructing gene coexpression networks based on prediction logic for the smoker group and the non-smoker group, respectively; (4) identifying smoking-mediated differential components, i.e., the unique gene coexpression patterns specific to each group; and (5) from the differential components, identifying genes directly co-expressed with major lung cancer signaling hallmarks. RESULTS A smoking-associated 6-gene signature was identified for prognosis of lung cancer from a training cohort (n=256). The 6-gene signature could separate lung cancer patients into two risk groups with distinct post-operative survival (log-rank P<0.04, Kaplan-Meier analyses) in three independent cohorts (n=427). The expression-defined prognostic prediction is strongly related to smoking association and smoking cessation (P<0.02; Pearson's Chi-squared tests). The 6-gene signature is an accurate prognostic factor (hazard ratio=1.89, 95% CI: [1.04, 3.43]) compared to common clinical covariates in multivariate Cox analysis. The 6-gene signature also provides an accurate diagnosis of lung cancer with an overall accuracy of 73% in a cohort of smokers (n=164). The coexpression patterns derived from the implication networks were validated with interactions reported in the literature retrieved with STRING8, Ingenuity Pathway Analysis, and Pathway Studio. CONCLUSIONS The pathway-based approach identified a smoking-associated 6-gene signature that predicts lung cancer risk and survival. This gene signature has potential clinical implications in the diagnosis and prognosis of lung cancer in smokers.
Collapse
Affiliation(s)
- Nancy Lan Guo
- Department of Community Medicine, West Virginia University, Morgantown, WV 26506, USA.
| | | |
Collapse
|
82
|
Matsunaga T, Wada Y, Endo S, Soda M, El-Kabbani O, Hara A. Aldo-Keto Reductase 1B10 and Its Role in Proliferation Capacity of Drug-Resistant Cancers. Front Pharmacol 2012; 3:5. [PMID: 22319498 PMCID: PMC3269042 DOI: 10.3389/fphar.2012.00005] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/11/2012] [Indexed: 12/11/2022] Open
Abstract
The human aldo–keto reductase AKR1B10, originally identified as an aldose reductase-like protein and human small intestine aldose reductase, is a cytosolic NADPH-dependent reductase that metabolizes a variety of endogenous compounds, such as aromatic and aliphatic aldehydes and dicarbonyl compounds, and some drug ketones. The enzyme is highly expressed in solid tumors of several tissues including lung and liver, and as such has received considerable interest as a relevant biomarker for the development of those tumors. In addition, AKR1B10 has been recently reported to be significantly up-regulated in some cancer cell lines (medulloblastoma D341 and colon cancer HT29) acquiring resistance toward chemotherapeutic agents (cyclophosphamide and mitomycin c), suggesting the validity of the enzyme as a chemoresistance marker. Although the detailed information on the AKR1B10-mediated mechanisms leading to the drug resistance process is not well understood so far, the enzyme has been proposed to be involved in functional regulations of cell proliferation and metabolism of drugs and endogenous lipids during the development of chemoresistance. This article reviews the current literature focusing mainly on expression profile and roles of AKR1B10 in the drug resistance of cancer cells. Recent developments of AKR1B10 inhibitors and their usefulness in restoring sensitivity to anticancer drugs are also reviewed.
Collapse
|
83
|
Kang MW, Lee ES, Yoon SY, Jo J, Lee J, Kim HK, Choi YS, Kim K, Shim YM, Kim J, Kim H. AKR1B10 is associated with smoking and smoking-related non-small-cell lung cancer. J Int Med Res 2011; 39:78-85. [PMID: 21672310 DOI: 10.1177/147323001103900110] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
This prospective study explored the relationship between expression of AKR1B10 mRNA and various clinical parameters in non-small-cell lung cancer (NSCLC) in terms of its validation as a marker for NSCLC. Tumour tissue samples were collected from 229 patients with NSCLC. Tissue samples from adjacent non-malignant lung tissue (> 5 cm from the tumour) of 89 of these patients and samples from 20 patients with benign lung disease were used as controls. Quantitative reverse transcription- polymerase chain reaction showed significantly higher levels of AKR1B10 mRNA expression in NSCLC tumour tissue than in adjacent non-malignant lung tissue and benign lung tissue. Statistically significant factors for AKR1B10 mRNA over-expression were found to be male gender, smoking, squamous cell carcinoma and moderate or poor cell differentiation. It is concluded that AKR1B10 seems to have potential as a prognostic marker for NSCLC and warrants further investigation.
Collapse
Affiliation(s)
- M-W Kang
- Cancer Research Centre, Centre for Clinical Research, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Lu TP, Lai LC, Tsai MH, Chen PC, Hsu CP, Lee JM, Hsiao CK, Chuang EY. Integrated analyses of copy number variations and gene expression in lung adenocarcinoma. PLoS One 2011; 6:e24829. [PMID: 21935476 PMCID: PMC3173487 DOI: 10.1371/journal.pone.0024829] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Accepted: 08/22/2011] [Indexed: 02/04/2023] Open
Abstract
Numerous efforts have been made to elucidate the etiology and improve the treatment of lung cancer, but the overall five-year survival rate is still only 15%. Identification of prognostic biomarkers for lung cancer using gene expression microarrays poses a major challenge in that very few overlapping genes have been reported among different studies. To address this issue, we have performed concurrent genome-wide analyses of copy number variation and gene expression to identify genes reproducibly associated with tumorigenesis and survival in non-smoking female lung adenocarcinoma. The genomic landscape of frequent copy number variable regions (CNVRs) in at least 30% of samples was revealed, and their aberration patterns were highly similar to several studies reported previously. Further statistical analysis for genes located in the CNVRs identified 475 genes differentially expressed between tumor and normal tissues (p<10(-5)). We demonstrated the reproducibility of these genes in another lung cancer study (p = 0.0034, Fisher's exact test), and showed the concordance between copy number variations and gene expression changes by elevated Pearson correlation coefficients. Pathway analysis revealed two major dysregulated functions in lung tumorigenesis: survival regulation via AKT signaling and cytoskeleton reorganization. Further validation of these enriched pathways using three independent cohorts demonstrated effective prediction of survival. In conclusion, by integrating gene expression profiles and copy number variations, we identified genes/pathways that may serve as prognostic biomarkers for lung tumorigenesis.
Collapse
Affiliation(s)
- Tzu-Pin Lu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Brody JS, Steiling K. Interaction of cigarette exposure and airway epithelial cell gene expression. Annu Rev Physiol 2011; 73:437-56. [PMID: 21090967 DOI: 10.1146/annurev-physiol-012110-142219] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cigarette smoking is responsible for lung cancer and chronic obstructive pulmonary disease (COPD), the leading cause of death from cancer and the second-leading cause of death in the United States. In the United States, 46 million people smoke, with an equal number of former smokers. Moreover, 20-25% of current or former smokers will develop either disease, and smokers with one disease are at increased risk for developing the other. There are no tools for predicting risk of developing either disease; no accepted tools for early diagnosis of potentially curable lung cancer; and no tools for defining molecular pathways or molecular subtypes of these diseases, for predicting rate of progression, or for assessing response to therapy at a biochemical or molecular level. This review discusses current studies and the future potential of measuring global gene expression in epithelial cells that are in the airway field of injury and of using the genomic information derived to begin to answer some of the above questions.
Collapse
Affiliation(s)
- Jerome S Brody
- Pulmonary Center, Boston University School of Medicine, Massachusetts 02218, USA.
| | | |
Collapse
|
86
|
Ahmad I, Singh LB, Foth M, Morris CA, Taketo MM, Wu XR, Leung HY, Sansom OJ, Iwata T. K-Ras and β-catenin mutations cooperate with Fgfr3 mutations in mice to promote tumorigenesis in the skin and lung, but not in the bladder. Dis Model Mech 2011; 4:548-55. [PMID: 21504907 PMCID: PMC3124065 DOI: 10.1242/dmm.006874] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 03/10/2011] [Indexed: 12/31/2022] Open
Abstract
The human fibroblast growth factor receptor 3 (FGFR3) gene is frequently mutated in superficial urothelial cell carcinoma (UCC). To test the functional significance of FGFR3 activating mutations as a 'driver' of UCC, we targeted the expression of mutated Fgfr3 to the murine urothelium using Cre-loxP recombination driven by the uroplakin II promoter. The introduction of the Fgfr3 mutations resulted in no obvious effect on tumorigenesis up to 18 months of age. Furthermore, even when the Fgfr3 mutations were introduced together with K-Ras or β-catenin (Ctnnb1) activating mutations, no urothelial dysplasia or UCC was observed. Interestingly, however, owing to a sporadic ectopic Cre recombinase expression in the skin and lung of these mice, Fgfr3 mutation caused papilloma and promoted lung tumorigenesis in cooperation with K-Ras and β-catenin activation, respectively. These results indicate that activation of FGFR3 can cooperate with other mutations to drive tumorigenesis in a context-dependent manner, and support the hypothesis that activation of FGFR3 signaling contributes to human cancer.
Collapse
Affiliation(s)
- Imran Ahmad
- The Beatson Institute for Cancer Research, Glasgow, G61 1BD, UK
| | | | - Mona Foth
- The Beatson Institute for Cancer Research, Glasgow, G61 1BD, UK
- School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Carol-Ann Morris
- School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Makoto Mark Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Xue-Ru Wu
- Department of Urology and Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Hing Y. Leung
- The Beatson Institute for Cancer Research, Glasgow, G61 1BD, UK
| | - Owen J. Sansom
- The Beatson Institute for Cancer Research, Glasgow, G61 1BD, UK
| | - Tomoko Iwata
- School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
87
|
Gower AC, Steiling K, Brothers JF, Lenburg ME, Spira A. Transcriptomic studies of the airway field of injury associated with smoking-related lung disease. PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY 2011; 8:173-9. [PMID: 21543797 PMCID: PMC3159071 DOI: 10.1513/pats.201011-066ms] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 12/30/2010] [Indexed: 12/12/2022]
Abstract
The "field of injury" hypothesis proposes that exposure to an inhaled insult such as cigarette smoke elicits a common molecular response throughout the respiratory tract. This response can therefore be quantified in any airway tissue, including readily accessible epithelial cells in the bronchus, nose, and mouth. High-throughput technologies, such as whole-genome gene expression microarrays, can be employed to catalog the physiological consequences of such exposures in the airway epithelium. Pulmonary diseases such as chronic obstructive pulmonary disease, lung cancer, and asthma are also thought to be associated with a field of injury, and in patients with these diseases, airway epithelial cells can be a useful surrogate for diseased tissue that is often difficult to obtain. Global measurement of mRNA and microRNA expression in these cells can provide useful information about the molecular pathogenesis of such diseases and may be useful for diagnosis and for predicting prognosis and response to therapy. In this review, our aim is to summarize the history and state of the art of such "transcriptomic" studies in the human airway epithelium, especially in smoking and smoking-related lung diseases, and to highlight future directions for this field.
Collapse
Affiliation(s)
- Adam C. Gower
- Bioinformatics Program, Boston University, Section of Computational Biomedicine, Department of Medicine, and Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Katrina Steiling
- Bioinformatics Program, Boston University, Section of Computational Biomedicine, Department of Medicine, and Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - John F. Brothers
- Bioinformatics Program, Boston University, Section of Computational Biomedicine, Department of Medicine, and Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Marc E. Lenburg
- Bioinformatics Program, Boston University, Section of Computational Biomedicine, Department of Medicine, and Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Avrum Spira
- Bioinformatics Program, Boston University, Section of Computational Biomedicine, Department of Medicine, and Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
88
|
Koelsch B, Winzen-Reichert B, Fischer C, Kutritz A, van den Berg L, Kindler-Röhrborn A. Sex-biased suppression of chemically induced neural carcinogenesis in congenic BDIX.BDIV-Mss4a rats. Physiol Genomics 2011; 43:631-9. [PMID: 21427360 DOI: 10.1152/physiolgenomics.00246.2010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
We previously mapped several gene loci influencing cancer risk of inbred BDIV and BDIX rats, resistant and susceptible, respectively, to N-ethyl-N-nitrosourea (ENU)-induced malignant peripheral nerve sheath tumors (MPNSTs). On the basis of a genomewide association analysis using a (BDIV × BDIX) F(2) generation the Mss4 locus on rat chromosome 6 was predicted to mediate resistance to MPNST development in the trigeminal nerves, preferentially in females. F(2) females homozygous for D6Mit1 proved almost exclusively resistant to peripheral neurooncogenesis, with no effect detectable in males. To functionally verify Mss4, a congenic BDIX rat strain was generated carrying a corresponding BDIV genomic fragment. On treatment with ENU, congenic BDIX.BDIV-Mss4a rats showed a 2.4-fold lower MPNST rate and a 55-day-longer survival time compared with BDIX animals. The sex-specific effect observed in F(2) rats was less pronounced in BDIX.BDIV-Mss4a congenics, with males, too, being protected against MPNST but to a lesser extent than females. Transcription profiling using trigeminal nerve tissue of BDIX, BDIV, and BDIX.BDIV-Mss4a congenics of both sexes revealed 61 genes located in the Mss4a fragment differentially expressed between BDIV and BDIX rats. In congenic rats each gene either displayed trans-regulated BDIX-like expression strength or cis-regulated BDIV-like transcript levels or intermediate expression without marked sex differences. Genomewide a number of genes exhibiting male-biased expression in the BDIX rat strain displayed a reversal of the sexual dimorphism in congenic rats similar to the BDIV expression pattern, which might be the basis of preferential protection of females against MPNST development.
Collapse
Affiliation(s)
- Bernd Koelsch
- Institute of Pathology and Neuropathology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | | | | | | | | | | |
Collapse
|
89
|
Wan YW, Xiao C, Guo NL. Network-Based Identification of Smoking-Associated Gene Signature for Lung Cancer. PROCEEDINGS. IEEE INTERNATIONAL CONFERENCE ON BIOINFORMATICS AND BIOMEDICINE 2010; 2010:479-484. [PMID: 25984394 PMCID: PMC4429297 DOI: 10.1109/bibm.2010.5706613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This study presents a novel computational approach to identifying a smoking-associated gene signature. The methodology contains the following steps: 1) identifying genes significantly associated with lung cancer survival, 2) selecting genes which are differentially expressed in smoker versus non-smoker groups from the survival genes, 3) from these candidate genes, constructing gene co-expression networks based on prediction logic for smokers and non-smokers, 4) identifying smoking-mediated differential components, i.e., the unique gene co-expression patterns specific to each group, and 5) from the differential components, identifying genes directly co-expressed with major lung cancer hallmarks. The identified 7-gene signature could separate lung cancer patients into two risk groups with distinct post-operative survival (log-rank P < 0.05, Kaplan-Meier analysis) in four independent cohorts (n=427). It also has implications in the diagnosis of lung cancer (accuracy = 74%) in a cohort of smokers (n=164). Computationally derived co-expression patterns were validated with Pathway Studio and STRING 8.
Collapse
|
90
|
Wang R, Wang G, Ricard MJ, Ferris B, Strulovici-Barel Y, Salit J, Hackett NR, Gudas LJ, Crystal RG. Smoking-induced upregulation of AKR1B10 expression in the airway epithelium of healthy individuals. Chest 2010; 138:1402-10. [PMID: 20705797 DOI: 10.1378/chest.09-2634] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The aldo-keto reductase (AKR) gene superfamily codes for monomeric, soluble reduced nicotinamide adenine dinucleotide phosphate-dependent oxidoreductases that mediate elimination reactions. AKR1B10, an AKR that eliminates retinals, has been observed as upregulated in squamous metaplasia and non-small cell lung cancer and has been suggested as a diagnostic marker specific to tobacco-related carcinogenesis. We hypothesized that upregulation of AKR1B10 expression may be initiated in healthy smokers prior to the development of evidence of lung cancer. METHODS Expression of AKR1B10 was assessed at the mRNA level using microarrays with TaqMan confirmation in the large airway epithelium (21 healthy nonsmokers, 31 healthy smokers) and small airway epithelium (51 healthy nonsmokers, 58 healthy smokers) obtained by fiberoptic bronchoscopy and brushing. RESULTS Compared with healthy nonsmokers, AKR1B10 mRNA levels were significantly upregulated in both large and small airway epithelia of healthy smokers. Consistent with the mRNA data, AKR1B10 protein was significantly upregulated in the airway epithelium of healthy smokers as assessed by Western blot analysis and immunohistochemistry, with AKR1B10 expressed in both differentiated and basal cells. Finally, cigarette smoke extract mediated upregulation of AKR1B10 in airway epithelial cells in vitro, and transfection of AKR1B10 into airway epithelial cells enhanced the conversion of retinal to retinol. CONCLUSIONS Smoking per se mediates upregulation of AKR1B10 expression in the airway epithelia of healthy smokers with no evidence of lung cancer. In the context of these observations and the link of AKR1B10 to the metabolism of retinals and to lung cancer, the smoking-induced upregulation of AKR1B10 may be an early process in the multiple events leading to lung cancer.
Collapse
Affiliation(s)
- Rui Wang
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Hong WJ, Tibshirani R, Chu G. Local false discovery rate facilitates comparison of different microarray experiments. Nucleic Acids Res 2010; 37:7483-97. [PMID: 19825981 PMCID: PMC2794175 DOI: 10.1093/nar/gkp813] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The local false discovery rate (LFDR) estimates the probability of falsely identifying specific genes with changes in expression. In computer simulations, LFDR <10% successfully identified genes with changes in expression, while LFDR >90% identified genes without changes. We used LFDR to compare different microarray experiments quantitatively: (i) Venn diagrams of genes with and without changes in expression, (ii) scatter plots of the genes, (iii) correlation coefficients in the scatter plots and (iv) distributions of gene function. To illustrate, we compared three methods for pre-processing microarray data. Correlations between methods were high (r = 0.84-0.92). However, responses were often different in magnitude, and sometimes discordant, even though the methods used the same raw data. LFDR complements functional assessments like gene set enrichment analysis. To illustrate, we compared responses to ultraviolet radiation (UV), ionizing radiation (IR) and tobacco smoke. Compared to unresponsive genes, genes responsive to both UV and IR were enriched for cell cycle, mitosis, and DNA repair functions. Genes responsive to UV but not IR were depleted for cell adhesion functions. Genes responsive to tobacco smoke were enriched for detoxification functions. Thus, LFDR reveals differences and similarities among experiments.
Collapse
Affiliation(s)
- Wan-Jen Hong
- Department of Medicine, Department of Biochemistry, Department of Statistics and Health Research and Policy, Stanford University Medical Center, Stanford, CA 94305, USA
| | | | | |
Collapse
|
92
|
Lonergan KM, Chari R, Coe BP, Wilson IM, Tsao MS, Ng RT, MacAulay C, Lam S, Lam WL. Transcriptome profiles of carcinoma-in-situ and invasive non-small cell lung cancer as revealed by SAGE. PLoS One 2010; 5:e9162. [PMID: 20161782 PMCID: PMC2820080 DOI: 10.1371/journal.pone.0009162] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2009] [Accepted: 01/07/2010] [Indexed: 12/29/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) presents as a progressive disease spanning precancerous, preinvasive, locally invasive, and metastatic lesions. Identification of biological pathways reflective of these progressive stages, and aberrantly expressed genes associated with these pathways, would conceivably enhance therapeutic approaches to this devastating disease. Methodology/Principal Findings Through the construction and analysis of SAGE libraries, we have determined transcriptome profiles for preinvasive carcinoma-in-situ (CIS) and invasive squamous cell carcinoma (SCC) of the lung, and compared these with expression profiles generated from both bronchial epithelium, and precancerous metaplastic and dysplastic lesions using Ingenuity Pathway Analysis. Expression of genes associated with epidermal development, and loss of expression of genes associated with mucociliary biology, are predominant features of CIS, largely shared with precancerous lesions. Additionally, expression of genes associated with xenobiotic metabolism/detoxification is a notable feature of CIS, and is largely maintained in invasive cancer. Genes related to tissue fibrosis and acute phase immune response are characteristic of the invasive SCC phenotype. Moreover, the data presented here suggests that tissue remodeling/fibrosis is initiated at the early stages of CIS. Additionally, this study indicates that alteration in copy-number status represents a plausible mechanism for differential gene expression in CIS and invasive SCC. Conclusions/Significance This study is the first report of large-scale expression profiling of CIS of the lung. Unbiased expression profiling of these preinvasive and invasive lesions provides a platform for further investigations into the molecular genetic events relevant to early stages of squamous NSCLC development. Additionally, up-regulated genes detected at extreme differences between CIS and invasive cancer may have potential to serve as biomarkers for early detection.
Collapse
Affiliation(s)
- Kim M. Lonergan
- Genetics Unit, Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- * E-mail:
| | - Raj Chari
- Genetics Unit, Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Bradley P. Coe
- Genetics Unit, Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Ian M. Wilson
- Genetics Unit, Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Ming-Sound Tsao
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Raymond T. Ng
- Genetics Unit, Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Computer Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Calum MacAulay
- Imaging Unit, Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Stephen Lam
- Imaging Unit, Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Wan L. Lam
- Genetics Unit, Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| |
Collapse
|
93
|
Bakkar AA, Allory Y, Iwatsubo Y, de Medina SGD, Maille P, Khreich N, Riou A, Leroy K, Vordos D, Abbou CC, Andujar P, Billebaud T, Chammings S, Conso F, De La Taille A, Fontaine E, Gattegno B, Ravery V, Sibony M, Radvanyi F, Chopin DK, Pairon JC. Occupational exposure to polycyclic aromatic hydrocarbons influenced neither the frequency nor the spectrum of FGFR3 mutations in bladder urothelial carcinoma. Mol Carcinog 2010; 49:25-31. [PMID: 19722178 DOI: 10.1002/mc.20573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Occupational exposure to polycyclic aromatic hydrocarbons (PAH) is associated with an increased risk of urothelial carcinoma (UC). FGFR3 is found mutated in about 70% of Ta tumors, which represent the major group at diagnosis. The influence of PAH on FGFR3 mutations and whether it is related to the emergence or shaping of these mutations is not yet known. We investigated the influence of occupational PAH on the frequency and spectrum of FGFR3 mutations. We included on 170 primary urothelial tumors from five hospitals from France. Patients (median age, 64 yr) were interviewed to gather data on occupational exposure to PAH, revealing 104 non- and possibly PAH exposed patients, 66 probably and definitely exposed patients. Tumors were classified as follows: 75 pTa, 52 pT1, and 43 > or =pT2. Tumor grades were as follows: 6 low malignant potential neoplasms (LMPN) and 41 low-grade and 123 high-grade carcinomas. The SnaPshot method was used to screen for the following FGFR3 mutations: R248C, S249C, G372C, Y375C, A393E, K652E, K652Q, K652M, and K652T. Occupational PAH exposure was not associated with a particular stage or grade of tumors. Thirty-nine percent of the tumors harbored FGFR3 mutations. After adjustment for smoking, occupational exposure to PAH did not influence the frequency [OR, 1.10; 95% CI, 0.78-1.52], or spectrum of FGFR3 mutations. Occupational exposure to PAH influenced neither the frequency nor the spectrum of FGFR3 mutations and there was no direct relationship between these mutations and this occupational hazard.
Collapse
|
94
|
Jackson-Rosario SE, Self WT. Targeting selenium metabolism and selenoproteins: novel avenues for drug discovery. Metallomics 2010; 2:112-6. [PMID: 21069141 PMCID: PMC3039543 DOI: 10.1039/b917141j] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Selenoproteins play a wide range of roles in metabolism and oxidative stress defense and are produced by organisms in all three domains of life. Recent evidence has been presented that metal based cancer drugs target the selenol nucleophile of the active site selenocysteine in thioredoxin reductase isoenzymes. Other metals and metalloids, such as tin, arsenic and gold, have also recently been shown to form stable complexes with hydrogen selenide, a required precursor for the synthesis of selenoproteins in all biological organisms. Moreover these metal based compounds have been shown to inhibit growth of pathogens such as Clostridium difficile and Treponema denticola due to their reactivity with this highly reactive metabolic precursor. This review summarizes the recent finding on these two avenues for drug discovery, and puts this work in context with the larger field of selenium biology.
Collapse
|
95
|
Tu S, Ren Y, Tong W, Zheng S, Xu N, Bhatnagar A, Liu S. A new approach to monitor expression of aldoâketo reductase proteins in mouse tissues. Proteomics 2009; 9:5090-100. [DOI: 10.1002/pmic.200900256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
96
|
Rohrbeck A, Borlak J. Cancer genomics identifies regulatory gene networks associated with the transition from dysplasia to advanced lung adenocarcinomas induced by c-Raf-1. PLoS One 2009; 4:e7315. [PMID: 19812696 PMCID: PMC2754338 DOI: 10.1371/journal.pone.0007315] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 09/13/2009] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Lung cancer is a leading cause of cancer morbidity. To improve an understanding of molecular causes of disease a transgenic mouse model was investigated where targeted expression of the serine threonine kinase c-Raf to respiratory epithelium induced initially dysplasia and subsequently adenocarcinomas. This enables dissection of genetic events associated with precancerous and cancerous lesions. METHODOLOGY/PRINCIPAL FINDINGS By laser microdissection cancer cell populations were harvested and subjected to whole genome expression analyses. Overall 473 and 541 genes were significantly regulated, when cancer versus transgenic and non-transgenic cells were compared, giving rise to three distinct and one common regulatory gene network. At advanced stages of tumor growth predominately repression of gene expression was observed, but genes previously shown to be up-regulated in dysplasia were also up-regulated in solid tumors. Regulation of developmental programs as well as epithelial mesenchymal and mesenchymal endothelial transition was a hall mark of adenocarcinomas. Additionally, genes coding for cell adhesion, i.e. the integrins and the tight and gap junction proteins were repressed, whereas ligands for receptor tyrosine kinase such as epi- and amphiregulin were up-regulated. Notably, Vegfr- 2 and its ligand Vegfd, as well as Notch and Wnt signalling cascades were regulated as were glycosylases that influence cellular recognition. Other regulated signalling molecules included guanine exchange factors that play a role in an activation of the MAP kinases while several tumor suppressors i.e. Mcc, Hey1, Fat3, Armcx1 and Reck were significantly repressed. Finally, probable molecular switches forcing dysplastic cells into malignantly transformed cells could be identified. CONCLUSIONS/SIGNIFICANCE This study provides insight into molecular pertubations allowing dysplasia to progress further to adenocarcinoma induced by exaggerted c-Raf kinase activity.
Collapse
Affiliation(s)
- Astrid Rohrbeck
- Department of Molecular Medicine and Medical Biotechnology, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Jürgen Borlak
- Department of Molecular Medicine and Medical Biotechnology, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
- Center for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
97
|
MacLeod AK, McMahon M, Plummer SM, Higgins LG, Penning TM, Igarashi K, Hayes JD. Characterization of the cancer chemopreventive NRF2-dependent gene battery in human keratinocytes: demonstration that the KEAP1-NRF2 pathway, and not the BACH1-NRF2 pathway, controls cytoprotection against electrophiles as well as redox-cycling compounds. Carcinogenesis 2009; 30:1571-80. [PMID: 19608619 DOI: 10.1093/carcin/bgp176] [Citation(s) in RCA: 253] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To better understand the role of transcription factor NF-E2-related factor (NRF) 2 in the human and its contribution to cancer chemoprevention, we have knocked down its negative regulators, Kelch-like ECH-associated protein 1 (KEAP1) and broad-complex, tramtrack and bric à brac and cap'n'collar homology 1 (BACH1), in HaCaT keratinocytes. Whole-genome microarray revealed that knockdown of KEAP1 resulted in 23 messenger RNAs (mRNAs) being up-regulated > or = 2.0-fold. mRNA for aldo-keto reductase (AKR) 1B10, AKR1C1, AKR1C2 and AKR1C3 were induced to the greatest extent, showing increases of between 12- and 16-fold, whereas mRNA for glutamate-cysteine ligase catalytic and modifier subunits, NAD(P)H:quinone oxidoreductase-1 and haem oxygenase-1 (HMOX1) were induced between 2.0- and 4.8-fold. Knockdown of BACH1 increased HMOX1 135-fold but induced the other genes examined to a maximum of only 2.7-fold. Activation of NRF2, by KEAP1 knockdown, caused a 75% increase in the amount of glutathione in HaCaT cells and a 1.4- to 1.6-fold increase in their resistance to the electrophiles acrolein, chlorambucil and cumene hydroperoxide (CuOOH), as well as the redox-cycling agent menadione. Inhibition of glutathione synthesis during KEAP1 knockdown, by treatment with buthionine sulfoximine, abrogated resistance to acrolein, chlorambucil and CuOOH, but not to menadione. In contrast, knockdown of BACH1 did not increase glutathione levels or resistance to xenobiotics. Knockdown of NRF2 in HaCaT cells decreased glutathione to approximately 80% of normal homeostatic levels and similarly reduced their tolerance of electrophiles. Thus, the KEAP1-NRF2 pathway determines resistance to electrophiles and redox-cycling compounds in human keratinocytes through glutathione-dependent and glutathione-independent mechanisms. This study also shows that AKR1B10, AKR1C1 and AKR1C2 proteins have potential utility as biomarkers for NRF2 activation in the human.
Collapse
Affiliation(s)
- A Kenneth MacLeod
- Biomedical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | | | | | | | | | | | | |
Collapse
|
98
|
Abstract
This review focuses on recent research using genomics to examine lung carcinogenesis, histologic differentiation, and progression.
Collapse
|
99
|
Qing J, Du X, Chen Y, Chan P, Li H, Wu P, Marsters S, Stawicki S, Tien J, Totpal K, Ross S, Stinson S, Dornan D, French D, Wang QR, Stephan JP, Wu Y, Wiesmann C, Ashkenazi A. Antibody-based targeting of FGFR3 in bladder carcinoma and t(4;14)-positive multiple myeloma in mice. J Clin Invest 2009; 119:1216-29. [PMID: 19381019 DOI: 10.1172/jci38017] [Citation(s) in RCA: 188] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Accepted: 03/04/2009] [Indexed: 11/17/2022] Open
Abstract
Overexpression of FGF receptor 3 (FGFR3) is implicated in the development of t(4;14)-positive multiple myeloma. While FGFR3 is frequently overexpressed and/or activated through mutations in bladder cancer, the functional importance of FGFR3 and its potential as a specific therapeutic target in this disease have not been elucidated in vivo. Here we report that inducible knockdown of FGFR3 in human bladder carcinoma cells arrested cell-cycle progression in culture and markedly attenuated tumor progression in xenografted mice. Further, we developed a unique antibody (R3Mab) that inhibited not only WT FGFR3, but also various mutants of the receptor, including disulfide-linked cysteine mutants. Biochemical analysis and 2.1-A resolution crystallography revealed that R3Mab bound to a specific FGFR3 epitope that simultaneously blocked ligand binding, prevented receptor dimerization, and induced substantial conformational changes in the receptor. R3Mab exerted potent antitumor activity against bladder carcinoma and t(4;14)-positive multiple myeloma xenografts in mice by antagonizing FGFR3 signaling and eliciting antibody-dependent cell-mediated cytotoxicity (ADCC). These studies provide in vivo evidence demonstrating an oncogenic role of FGFR3 in bladder cancer and support antibody-based targeting of FGFR3 in hematologic and epithelial cancers driven by WT or mutant FGFR3.
Collapse
Affiliation(s)
- Jing Qing
- Department of Molecular Oncology, Genentech Inc., South San Francisco, California, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Quinn AM, Harvey RG, Penning TM. Oxidation of PAH trans-dihydrodiols by human aldo-keto reductase AKR1B10. Chem Res Toxicol 2009; 21:2207-15. [PMID: 18788756 DOI: 10.1021/tx8002005] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AKR1B10 has been identified as a potential biomarker for human nonsmall cell lung carcinoma and as a tobacco exposure and response gene. AKR1B10 functions as an efficient retinal reductase in vitro and may regulate retinoic acid homeostasis. However, the possibility that this enzyme is able to activate polycyclic aromatic hydrocarbon (PAH) trans-dihydrodiols to form reactive and redox-active o-quinones has not been investigated to date. AKR1B10 was found to oxidize a wide range of PAH trans-dihydrodiol substrates in vitro to yield PAH o-quinones. Reactions of AKR1B10 proceeded with improper stereochemistry, since it was specific for the minor (+)-benzo[a]pyrene-7S,8S-dihydrodiol diastereomer formed in vivo. However, AKR1B10 displayed reasonable activity in the oxidation of both the (-)-R,R and (+)-S,S stereoisomers of benzo[g]chrysene-11,12-dihydrodiol and oxidized the potentially relevant, albeit minor, (+)-benz[a]anthracene-3S,4S-dihydrodiol metabolite. We find that AKR1B10 is therefore likely to play a contributing role in the activation of PAH trans-dihydrodiols in human lung. AKR1B10 retinal reductase activity was confirmed in vitro and found to be 5- to 150-fold greater than the oxidation of PAH trans-dihydrodiols examined. AKR1B10 was highly expressed at the mRNA and protein levels in human lung adenocarcinoma A549 cells, and robust retinal reductase activity was measured in lysates of these cells. The much greater catalytic efficiency of retinal reduction compared to PAH trans-dihydrodiol metabolism suggests AKR1B10 may play a greater role in lung carcinogenesis through dysregulation of retinoic acid homeostasis than through oxidation of PAH trans-dihydrodiols.
Collapse
Affiliation(s)
- Amy M Quinn
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6084, USA
| | | | | |
Collapse
|