1
|
Sun C, Zeng B, Zhou J, Li N, Li M, Zhu C, Xie S, Wang Y, Wang S, Wang X. Analysis of SLC genes alternative splicing identifies the SLC7A6 RI isoform as a therapeutic target for colorectal cancer. Cancer Sci 2025; 116:233-247. [PMID: 39403788 PMCID: PMC11711054 DOI: 10.1111/cas.16351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/30/2024] [Accepted: 09/07/2024] [Indexed: 01/11/2025] Open
Abstract
Alternative splicing (AS), a crucial mechanism in post-transcriptional regulation, has been implicated in diverse cancer processes. Several splicing variants of solute carrier (SLC) transporters reportedly play pivotal roles in tumorigenesis and tumor development. However, an in-depth analysis of AS landscapes of SLCs in colon adenocarcinoma (COAD) is lacking. Herein, we analyzed data from The Cancer Genome Atlas and identified 1215 AS events across 243 SLC genes, including 109 differentially expressed AS (DEAS) events involving 62 SLC genes in COAD. Differentially spliced SLCs were enriched in biological processes, including transmembrane transporter activity, transporter activity, ferroptosis, and choline metabolism. In patients with COAD, tumor tissues exhibited higher expression of longer mitochondrial carrier SLC25A16 isoforms than adjacent normal tissues, consistent with bioinformatics analysis. Protein-coding sequences and transmembrane helices of survival-related DEAS were predicted, revealing that shifts in splicing sites altered the number and structure of their transmembrane proteins. We developed a prognostic risk model based on the screened 6-SLC-AS (SLC7A6_RI_37208 (SLC7A6-RI), SLC11A2_AP_21724, SLC2A8_ES_87631, SLC35B1_AA_42317, SLC39A11_AD_43204, and SLC7A8_AP_26712). Knockdown of the intronic region of SLC7A6-RI isoform enhanced colon cancer cell proliferation. In vivo, knockdown of the intronic region of SLC7A6-RI isoform enhanced tumor growth in colon cancer. Mechanistically, si-SLC7A6-RI isoform exerted oncogenic effects by activating the PI3K-Akt-mTOR signaling pathway and promoting cell proliferation, evidenced by increased expression of key regulators Phosphorylated Mammalian Target of Rapamycin (p-mTOR) and a cell proliferation marker Proliferating Cell Nuclear Antigen (PCNA) using western blotting. Our study elucidated SLC-AS in COAD, highlighting its potential as a prognostic and therapeutic target and emphasizing the suppressive influence of SLC7A6-RI in colon cancer progression.
Collapse
Affiliation(s)
- Chao Sun
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
- Integrated Chinese and Western Medicine Postdoctoral Research StationJinan UniversityGuangzhouChina
| | - Boning Zeng
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
- Integrated Chinese and Western Medicine Postdoctoral Research StationJinan UniversityGuangzhouChina
| | - Jilong Zhou
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Nan Li
- School of Pharmaceutical SciencesShenzhen University Health Science CenterShenzhenChina
| | - Mingwei Li
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Chaowei Zhu
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Shouxia Xie
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Yifei Wang
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw MaterialsJinan UniversityGuangzhouChina
| | - Shaoxiang Wang
- School of Pharmaceutical SciencesShenzhen University Health Science CenterShenzhenChina
| | - Xiao Wang
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| |
Collapse
|
2
|
Lai J, Lin X, Cao F, Mok H, Chen B, Liao N. CDKN1C as a prognostic biomarker correlated with immune infiltrates and therapeutic responses in breast cancer patients. J Cell Mol Med 2021; 25:9390-9401. [PMID: 34464504 PMCID: PMC8500970 DOI: 10.1111/jcmm.16880] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/01/2021] [Accepted: 08/09/2021] [Indexed: 12/14/2022] Open
Abstract
Breast cancer (BC) prognosis and therapeutic sensitivity could not be predicted efficiently. Previous evidence have shown the vital roles of CDKN1C in BC. Therefore, we aimed to construct a CDKN1C‐based model to accurately predicting overall survival (OS) and treatment responses in BC patients. In this study, 995 BC patients from The Cancer Genome Atlas database were selected. Kaplan‐Meier curve, Gene set enrichment and immune infiltrates analyses were executed. We developed a novel CDKN1C‐based nomogram to predict the OS, verified by the time‐dependent receiver operating characteristic curve, calibration curve and decision curve. Therapeutic response prediction was followed based on the low‐ and high‐nomogram score groups. Our results indicated that low‐CDKN1C expression was associated with shorter OS and lower proportion of naïve B cells, CD8 T cells, activated NK cells. The predictive accuracy of the nomogram for 5‐year OS was superior to the tumour‐node‐metastasis stage (area under the curve: 0.746 vs. 0.634, p < 0.001). The nomogram exhibited excellent predictive performance, calibration ability and clinical utility. Moreover, low‐risk patients were identified with stronger sensitivity to therapeutic agents. This tool can improve BC prognosis and therapeutic responses prediction, thus guiding individualized treatment decisions.
Collapse
Affiliation(s)
- Jianguo Lai
- Department of Breast Cancer, Guangdong Provincial People's Hospital,Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaoyi Lin
- Department of Breast Cancer, Guangdong Provincial People's Hospital,Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fangrong Cao
- Department of Breast Cancer, Guangdong Provincial People's Hospital,Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hsiaopei Mok
- Department of Breast Cancer, Guangdong Provincial People's Hospital,Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Bo Chen
- Department of Breast Cancer, Guangdong Provincial People's Hospital,Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ning Liao
- Department of Breast Cancer, Guangdong Provincial People's Hospital,Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
3
|
Justice AE, Young K, Gogarten SM, Sofer T, Graff M, Love SAM, Wang Y, Klimentidis YC, Cruz M, Guo X, Hartwig F, Petty L, Yao J, Allison MA, Below JE, Buchanan TA, Chen YDI, Goodarzi MO, Hanis C, Highland HM, Hsueh WA, Ipp E, Parra E, Palmas W, Raffel LJ, Rotter JI, Tan J, Taylor KD, Valladares A, Xiang AH, Sánchez-Johnsen L, Isasi CR, North KE. Genome-wide association study of body fat distribution traits in Hispanics/Latinos from the HCHS/SOL. Hum Mol Genet 2021; 30:2190-2204. [PMID: 34165540 PMCID: PMC8561424 DOI: 10.1093/hmg/ddab166] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 01/02/2023] Open
Abstract
Central obesity is a leading health concern with a great burden carried by ethnic minority populations, especially Hispanics/Latinos. Genetic factors contribute to the obesity burden overall and to inter-population differences. We aimed to identify the loci associated with central adiposity measured as waist-to-hip ratio (WHR), waist circumference (WC) and hip circumference (HIP) adjusted for body mass index (adjBMI) by using the Hispanic Community Health Study/Study of Latinos (HCHS/SOL); determine if differences in associations differ by background group within HCHS/SOL and determine whether previously reported associations generalize to HCHS/SOL. Our analyses included 7472 women and 5200 men of mainland (Mexican, Central and South American) and Caribbean (Puerto Rican, Cuban and Dominican) background residing in the USA. We performed genome-wide association analyses stratified and combined across sexes using linear mixed-model regression. We identified 16 variants for waist-to-hip ratio adjusted for body mass index (WHRadjBMI), 22 for waist circumference adjusted for body mass index (WCadjBMI) and 28 for hip circumference adjusted for body mass index (HIPadjBMI), which reached suggestive significance (P < 1 × 10-6). Many loci exhibited differences in strength of associations by ethnic background and sex. We brought a total of 66 variants forward for validation in cohorts (N = 34 161) with participants of Hispanic/Latino, African and European descent. We confirmed four novel loci (P < 0.05 and consistent direction of effect, and P < 5 × 10-8 after meta-analysis), including two for WHRadjBMI (rs13301996, rs79478137); one for WCadjBMI (rs3168072) and one for HIPadjBMI (rs28692724). Also, we generalized previously reported associations to HCHS/SOL, (8 for WHRadjBMI, 10 for WCadjBMI and 12 for HIPadjBMI). Our study highlights the importance of large-scale genomic studies in ancestrally diverse Hispanic/Latino populations for identifying and characterizing central obesity susceptibility that may be ancestry-specific.
Collapse
Affiliation(s)
- Anne E Justice
- Department of Population Health Sciences, Geisinger Health System, Danville, PA 17822, USA,Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA,To whom correspondence should be addressed at: Department of Population Health Sciences, Geisinger Health System, Danville, PA 17822, USA. Tel: +1 5702141009; Fax: +1 5702143071;
| | - Kristin Young
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | | | - Tamar Sofer
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Misa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Shelly Ann M Love
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Yujie Wang
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Yann C Klimentidis
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ 85724, USA
| | - Miguel Cruz
- Unidad de Investigacion Medica en Bioquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI (CMNSXX1)-IMSS, Mexico City 06720, Mexico
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Fernando Hartwig
- Center for Epidemiological Research, Universidade Federal de Pelotas, Pelotas 96020, Brazil
| | - Lauren Petty
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Matthew A Allison
- Division of Preventive Medicine, Department of Family Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jennifer E Below
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Thomas A Buchanan
- Department of Medicine, Keck School of Medicine and Diabetes and Obesity Research Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Craig Hanis
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Heather M Highland
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Willa A Hsueh
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Eli Ipp
- Department of Medicine, Endocrinology, Diabetes & Metabolism, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Esteban Parra
- Department of Anthropology, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Walter Palmas
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Leslie J Raffel
- Department of PediatrIcs, Division of Genetic and Genomic Medicine, University of California, Irvine, CA 92868, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Jingyi Tan
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Adan Valladares
- Unidad de Investigacion Medica en Bioquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI (CMNSXX1)-IMSS, Mexico City 06720, Mexico
| | - Anny H Xiang
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA 91101, USA
| | - Lisa Sánchez-Johnsen
- Department of Family Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Carmen R Isasi
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY 10467, USA,Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Kari E North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| |
Collapse
|
4
|
Knockdown of Orphan Transporter SLC22A18 Impairs Lipid Metabolism and Increases Invasiveness of HepG2 Cells. Pharm Res 2019; 36:39. [PMID: 30635741 DOI: 10.1007/s11095-018-2565-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE The aim of this work is to investigate the roles of solute carrier family 22 member 18 (SLC22A18) in lipid metabolism and in establishing the tumor phenotype of HepG2 cells. METHODS SLC22A18-knockdown HepG2 cells were established by stable transfection with shRNA. Protein expression levels were measured by quantitative proteomics and Western blot analysis. Cell growth was examined by cell counting kit. Accumulation of triglyceride-rich lipid droplets was measured by Oil-Red O staining. Cell migration and invasion were examined by Transwell assays. RESULTS SLC22A18-knockdown HepG2 cells accumulated triglyceride-rich lipid droplets and showed decreased expression levels of lysosomal/autophagic proteins, suggesting that lipid degradation is suppressed. Growth of HepG2 cells was decreased by SLC22A18 knockdown, but was restored by free fatty acid supplementation. In addition, SLC22A18 knockdown decreased the expression of insulin-like growth factor-binding protein 1 (IGFBP-1) and increased the invasion ability of HepG2 cells. Exogenous IGFBP-1 blocked the increase of invasion activity induced by SLC22A18 knockdown. CONCLUSION Our results suggest that suppression of SLC22A18 decreased the supply of intracellular free fatty acids from triglyceride-rich lipid droplets by impairing the lysosomal/autophagy degradation pathway and reduced the invasive activity of HepG2 cells by decreasing IGFBP-1 expression.
Collapse
|
5
|
Involvement of an Orphan Transporter, SLC22A18, in Cell Growth and Drug Resistance of Human Breast Cancer MCF7 Cells. J Pharm Sci 2018; 107:3163-3170. [DOI: 10.1016/j.xphs.2018.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 01/05/2023]
|
6
|
Yang B, Ma YB, Chu SH. Silencing SATB1 overcomes temozolomide resistance by downregulating MGMT expression and upregulating SLC22A18 expression in human glioblastoma cells. Cancer Gene Ther 2018; 25:309-316. [PMID: 30140041 DOI: 10.1038/s41417-018-0040-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common malignant tumor of the central nervous system and has a very poor prognosis. Currently, patients were treated by resection followed by radiotherapy plus concurrent temozolomide (TMZ) chemotherapy. However, many patients are resistant to TMZ-induced DNA damage because of upregulated expression of the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT). In this study, upregulation of SATB1 and MGMT, and downregulation of SLC22A18 resulted in acquisition of TMZ resistance in GBM U87 cells. Inactivation of special AT-rich sequence-binding protein 1 (SATB1) using short hairpin RNA (shRNA) downregulated MGMT expression and upregulated solute carrier family 22 member 18 (SLC22A18) expression in GBM cells. This suggested SATB1-mediated posttranscriptional regulation of the MGMT and SLC22A18 protein levels. Immunohistochemical analysis of malignant glioma specimens demonstrated a significant positive correlation between the levels of MGMT and SATB1, and a negative correlation between the levels of SLC22A18 and SATB1. Importantly, in recurrent, compared with the primary, lesions in 15 paired identical tumors, the SATB1 and MGMT protein levels were increased and the SLC22A18 levels were decreased. Finally, in TMZ-resistant GBM, SATB1 knockdown enhanced TMZ efficacy. Consequently, SATB1 inhibition might be a promising strategy combined with TMZ chemotherapy to treat TMZ-resistant GBM.
Collapse
Affiliation(s)
- Biao Yang
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China
| | - Yan-Bin Ma
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China
| | - Sheng-Hua Chu
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China.
| |
Collapse
|
7
|
Jung Y, Jun Y, Lee HY, Kim S, Jung Y, Keum J, Lee YS, Cho YB, Lee S, Kim J. Characterization of SLC22A18 as a tumor suppressor and novel biomarker in colorectal cancer. Oncotarget 2016. [PMID: 26196590 PMCID: PMC4694837 DOI: 10.18632/oncotarget.4681] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
SLC22A18, solute carrier family 22, member 18, has been proposed to function as a tumor suppressor based on its chromosomal location at 11p15.5, mutations and aberrant splicing in several types of cancer and down-regulation in glioblastoma. In this study, we sought to demonstrate the significance of SLC22A18 as a tumor suppressor in colorectal cancer (CRC) and provide mechanistic bases for its function. We first showed that the expression of SLC22A18 is significantly down-regulated in tumor tissues using matched normal-tumor samples from CRC patients. This finding was also supported by publically accessible data from The Cancer Genome Atlas (TCGA). Functionally, SLC22A18 inhibits colony formation and induces of G2/M arrest consistent with being a tumor suppressor. Interestingly, suppression of KRAS by RNA interference promotes SLC22A18 expression, and expression of SLC22A18 in turn inhibits KRASG12D-mediated anchorage independent growth of NIH3T3 cells indicating a mutual negative interaction. Finally, we evaluated diagnostic and prognostic values of SLC22A18 using clinical and gene expression data from TCGA which revealed a significantly worse long-term prognosis for patients with low level SLC22A18 expression. In sum, we established SLC22A18 as a tumor suppressor in colon epithelial cells and propose that SLC22A18 is potentially a marker of diagnostic and prognostic values.
Collapse
Affiliation(s)
- Yeonjoo Jung
- Ewha Research Center for Systems Biology, Seoul, Korea.,Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Yukyung Jun
- Ewha Research Center for Systems Biology, Seoul, Korea.,Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Hee-Young Lee
- Ewha Research Center for Systems Biology, Seoul, Korea.,Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Suyeon Kim
- Ewha Research Center for Systems Biology, Seoul, Korea.,Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Yeonhwa Jung
- Ewha Research Center for Systems Biology, Seoul, Korea.,Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Juhee Keum
- Ewha Research Center for Systems Biology, Seoul, Korea.,Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Yeo Song Lee
- Samsung Biomedical Research Institute, Seoul, Korea
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sanghyuk Lee
- Ewha Research Center for Systems Biology, Seoul, Korea.,Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Jaesang Kim
- Ewha Research Center for Systems Biology, Seoul, Korea.,Department of Life Science, Ewha Womans University, Seoul, Korea
| |
Collapse
|
8
|
Nakamura K, Hirayama-Kurogi M, Ito S, Kuno T, Yoneyama T, Obuchi W, Terasaki T, Ohtsuki S. Large-scale multiplex absolute protein quantification of drug-metabolizing enzymes and transporters in human intestine, liver, and kidney microsomes by SWATH-MS: Comparison with MRM/SRM and HR-MRM/PRM. Proteomics 2016; 16:2106-17. [PMID: 27197958 DOI: 10.1002/pmic.201500433] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 05/02/2016] [Accepted: 05/16/2016] [Indexed: 12/21/2022]
Abstract
The purpose of the present study was to examine simultaneously the absolute protein amounts of 152 membrane and membrane-associated proteins, including 30 metabolizing enzymes and 107 transporters, in pooled microsomal fractions of human liver, kidney, and intestine by means of SWATH-MS with stable isotope-labeled internal standard peptides, and to compare the results with those obtained by MRM/SRM and high resolution (HR)-MRM/PRM. The protein expression levels of 27 metabolizing enzymes, 54 transporters, and six other membrane proteins were quantitated by SWATH-MS; other targets were below the lower limits of quantitation. Most of the values determined by SWATH-MS differed by less than 50% from those obtained by MRM/SRM or HR-MRM/PRM. Various metabolizing enzymes were expressed in liver microsomes more abundantly than in other microsomes. Ten, 13, and eight transporters listed as important for drugs by International Transporter Consortium were quantified in liver, kidney, and intestinal microsomes, respectively. Our results indicate that SWATH-MS enables large-scale multiplex absolute protein quantification while retaining similar quantitative capability to MRM/SRM or HR-MRM/PRM. SWATH-MS is expected to be useful methodology in the context of drug development for elucidating the molecular mechanisms of drug absorption, metabolism, and excretion in the human body based on protein profile information.
Collapse
Affiliation(s)
- Kenji Nakamura
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Mio Hirayama-Kurogi
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Shingo Ito
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Takuya Kuno
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Drug Metabolism and Pharmacokinetics, Drug Safety Research Center, Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd, Tokushima, Japan
| | - Toshihiro Yoneyama
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Wataru Obuchi
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Tetsuya Terasaki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
9
|
Hoekstra AS, Addie RD, Ras C, Seifar RM, Ruivenkamp CA, Briaire-de Bruijn IH, Hes FJ, Jansen JC, Corssmit EPM, Corver WE, Morreau H, Bovée JVMG, Bayley JP, Devilee P. Parent-of-origin tumourigenesis is mediated by an essential imprinted modifier in SDHD-linked paragangliomas: SLC22A18 and CDKN1C are candidate tumour modifiers. Hum Mol Genet 2016; 25:3715-3728. [PMID: 27402879 DOI: 10.1093/hmg/ddw218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/28/2016] [Accepted: 06/30/2016] [Indexed: 12/11/2022] Open
Abstract
Mutations in SDHD and SDHAF2 (both located on chromosome 11) give rise to hereditary paraganglioma almost exclusively after paternal transmission of the mutation, and tumours often show loss of the entire maternal copy of chromosome 11. The 'Hensen' model postulates that a tumour modifier gene located on chromosome 11p15, a region known to harbour a cluster of imprinted genes, is essential to tumour formation. We observed decreased protein expression of the 11p15 candidate genes CDKN1C, SLC22A18 and ZNF215 evaluated in 60 SDHD-mutated tumours compared to normal carotid body tissue and non-SDH mutant tumours.We then created stable knockdown in vitro models, reasoning that the simultaneous knockdown of SDHD and a maternally expressed 11p15 modifier gene would enhance paraganglioma-related cellular characteristics compared to SDHD knockdown alone. Knockdown of SDHD in SNB19 and SHSY5Y cells resulted in the accumulation of succinate, the stabilization of HIF1 protein and a reduction in cell proliferation.Compared to single knockdown of SDHD, knockdown of SDHD together with SLC22A18 or with CDKN1C led to small but significant increases in cell proliferation and resistance to apoptosis, and to a gene expression profile closely related to the known transcriptional profile of SDH-deficient tumours. Of the 60 SDHD tumours investigated, four tumours showing retention of chromosome 11 showed SLC22A18 and CDKN1C expression levels comparable to levels in tumours showing loss of chromosome 11, suggesting loss of protein expression despite chromosomal retention.Our data strongly suggest that SLC22A18 and/or CDKN1C are tumour modifier genes involved in the tumourigenesis of SDHD-linked paraganglioma.
Collapse
Affiliation(s)
| | - Ruben D Addie
- Center for Proteomics and Metabolomics
- Department of Pathology
| | - Cor Ras
- Department of Biotechnology, Delft University of Technology, Delft, The Netherlands
| | - Reza M Seifar
- Department of Biotechnology, Delft University of Technology, Delft, The Netherlands
| | | | | | | | | | - Eleonora P M Corssmit
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
10
|
Chu SH, Zhou ZM, Karri S, Li ZQ, Zhao JM. In vitro and in vivo radiosensitization of human glioma U251 cells induced by upregulated expression of SLC22A18. Cancer Gene Ther 2014; 21:103-109. [PMID: 24481489 DOI: 10.1038/cgt.2014.4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 01/01/2014] [Accepted: 01/10/2014] [Indexed: 01/08/2023]
Abstract
Our previous study showed that solute carrier family 22 (organic cation transporter) member 18 (SLC22A18) downregulation via promoter methylation was associated with the development and progression of glioma, and the elevated expression of SLC22A18 was found to increase the sensitivity of glioma U251 cells to the anticancer drug 1,3-bis(2-chloroethyl)-1-nitrosourea. In this study, we investigated the possible upregulated expression of SLC22A18-induced enhancement of radiosensitivity of human glioma U251 cells in order to provide evidence in support of further clinical investigations. Stably overexpressing SLC22A18 human glioma U251 cells were generated to investigate the effect of SLC22A18 on the sensitivity of cells to irradiation in vitro using clonogenic survival assay. The apoptosis of U251 cells was examined with terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay. DNA damage and repair were measured using γH2AX foci. The effect of SLC22A18 on the in vivo tumor radiosensitivity was investigated in the orthotopic mice model. Upregulated expression of SLC22A18 enhanced the radiosensitivity of glioma U251 cells and also enhanced irradiation-induced apoptosis of U251 cells, but irradiation-induced apoptosis did not correlate with radiosensitizing effect of upregulated expression of SLC22A18. The repair of irradiation-induced double-strand-breaks was retarded in stably overexpressing SLC22A18 U251 cells. In the orthotopic mice model, the upregulated expression of SLC22A18 in U251 cells enhanced the effect of irradiation treatment and increased the survival time of mice. These results show that upregulated expression of SLC22A18 radiosensitizes human glioma U251 cells by suppressing DNA repair capacity.
Collapse
Affiliation(s)
- S-H Chu
- Department of Neurosurgery, Shanghai 3rd People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Z-M Zhou
- Department of Neurosurgery, Dujiangyan Medical Center, Chengdu, China
| | - S Karri
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Z-Q Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - J-M Zhao
- Department of Radiology, Shanghai 3rd People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
11
|
A novel link between Slc22a18 and fat accumulation revealed by a mutation in the spontaneously hypertensive rat. Biochem Biophys Res Commun 2013; 440:521-6. [DOI: 10.1016/j.bbrc.2013.09.096] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 09/18/2013] [Indexed: 11/17/2022]
|
12
|
Albacker CE, Storer NY, Langdon EM, DiBiase A, Zhou Y, Langenau DM, Zon LI. The histone methyltransferase SUV39H1 suppresses embryonal rhabdomyosarcoma formation in zebrafish. PLoS One 2013; 8:e64969. [PMID: 23705022 PMCID: PMC3660348 DOI: 10.1371/journal.pone.0064969] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 04/22/2013] [Indexed: 12/22/2022] Open
Abstract
Epigenetics, or the reversible and heritable marks of gene regulation not including DNA sequence, encompasses chromatin modifications on both the DNA and histones and is as important as the DNA sequence itself. Chromatin-modifying factors are playing an increasingly important role in tumorigenesis, particularly among pediatric rhabdomyosarcomas (RMS), revealing potential novel therapeutic targets. We performed an overexpression screen of chromatin-modifying factors in a KRAS(G12D)-driven zebrafish model for RMS. Here, we describe the identification of a histone H3 lysine 9 histone methyltransferase, SUV39H1, as a suppressor of embryonal RMS formation in zebrafish. This suppression is specific to the histone methyltransferase activity of SUV39H1, as point mutations in the SET domain lacked the effect. SUV39H1-overexpressing and control tumors have a similar proliferation rate, muscle differentiation state, and tumor growth rate. Strikingly, SUV39H1-overexpressing fish initiate fewer tumors, which results in the observed suppressive phenotype. We demonstrate that the delayed tumor onset occurs between 5 and 7 days post fertilization. Gene expression profiling at these stages revealed that in the context of KRAS(G12D) overexpression, SUV39H1 may suppress cell cycle progression. Our studies provide evidence for the role of SUV39H1 as a tumor suppressor.
Collapse
Affiliation(s)
- Colleen E. Albacker
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Narie Y. Storer
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Erin M. Langdon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anthony DiBiase
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yi Zhou
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David M. Langenau
- Department of Pathology, Massachusetts General Hospital, Harvard Stem Cell Institute, Charlestown, Massachusetts, United States of America
| | - Leonard I. Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
13
|
Chu SH, Ma YB, Feng DF, Li ZQ, Jiang PC. Predictive value of the SLC22A18 protein expression in glioblastoma patients receiving temozolomide therapy. J Transl Med 2013; 11:69. [PMID: 23514245 PMCID: PMC3610152 DOI: 10.1186/1479-5876-11-69] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 03/13/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Our previous study showed that SLC22A18 downregulation and promoter methylation were associated with the development and progression of glioma and the elevated expression of SLC22A18 was found to increase the sensitivity of glioma U251 cells to the anticancer drug 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). In this study, we investigated the predictive value of SLC22A18 promoter methylation and protein expression in glioblastoma multiforme (GBM) patients receiving temozolomide (TMZ) therapy. PATIENTS AND METHODS SLC22A18 promoter methylation and protein expression were examined by methylation-specific polymerase chain reaction (MSP) and Western blotting respectively, then we compared SLC22A18 promoter methylation and protein expression in tumor cell explants in regard to prediction of TMZ response and survival time of 86 GBM patients. RESULTS SLC22A18 promoter methylation was detected in 61 of 86 (71%) samples, whereas 36 of 86 (42%) cases were scored positive for SLC22A18 protein expression. Overall SLC22A18 promoter methylation was significantly related to SLC22A18 protein expression, but a subgroup of cases did not follow this association. Multivariate Cox regression analysis indicated that SLC22A18 protein expression, but not promoter methylation, was significantly correlated with TMZ therapy. SLC22A18 protein expression predicted a significantly shorter overall survival in 51 patients receiving TMZ therapy, whereas no differences in overall survival were observed in 35 patients without TMZ therapy. CONCLUSIONS These results show that lack of SLC22A18 protein expression is superior to promoter methylation as a predictive tumor biomarker in GBM patients receiving temozolomide therapy.
Collapse
Affiliation(s)
- Sheng-Hua Chu
- Department of Neurosurgery, Shanghai 3rd People's Hospital, School of Medicine, Shanghai Jiao Tong University, 280 Mohe Road, Baoshan District, Shanghai 201900, China
| | - Yan-Bin Ma
- Department of Neurosurgery, Shanghai 3rd People's Hospital, School of Medicine, Shanghai Jiao Tong University, 280 Mohe Road, Baoshan District, Shanghai 201900, China
| | - Dong-Fu Feng
- Department of Neurosurgery, Shanghai 3rd People's Hospital, School of Medicine, Shanghai Jiao Tong University, 280 Mohe Road, Baoshan District, Shanghai 201900, China
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Pu-Cha Jiang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
14
|
Zanola A, Rossi S, Faggi F, Monti E, Fanzani A. Rhabdomyosarcomas: an overview on the experimental animal models. J Cell Mol Med 2012; 16:1377-91. [PMID: 22225829 PMCID: PMC3823208 DOI: 10.1111/j.1582-4934.2011.01518.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Rhabdomyosarcomas (RMS) are aggressive childhood soft-tissue malignancies deriving from mesenchymal progenitors that are committed to muscle-specific lineages. Despite the histopathological signatures associated with three main histological variants, termed embryonal, alveolar and pleomorphic, a plethora of genetic and molecular changes are recognized in RMS. Over the years, exposure to carcinogens or ionizing radiations and gene-targeting approaches in vivo have greatly contributed to disclose some of the mechanisms underlying RMS onset. In this review, we describe the principal distinct features associated with RMS variants and focus on the current available experimental animal models to point out the molecular determinants cooperating with RMS development and progression.
Collapse
Affiliation(s)
- Alessandra Zanola
- Department of Biomedical Sciences and Biotechnologies, Interuniversity Institute of Myology (IIM), University of Brescia, Brescia, Italy
| | | | | | | | | |
Collapse
|
15
|
Abstract
Caveolins are scaffolding proteins that play a pivotal role in numerous processes, including caveolae biogenesis, vesicular transport, cholesterol homeostasis and regulation of signal transduction. There are three different isoforms (Cav-1, -2 and -3) that form homo- and hetero-aggregates at the plasma membrane and modulate the activity of a number of intracellular binding proteins. Cav-1 and Cav-3, in particular, are respectively expressed in the reserve elements (e.g. satellite cells) and in mature myofibres of skeletal muscle and their expression interplay characterizes the switch from muscle precursors to differentiated elements. Recent findings have shown that caveolins are also expressed in rhabdomyosarcoma, a group of heterogeneous childhood soft-tissue sarcomas in which the cancer cells seem to derive from progenitors that resemble myogenic cells. In this review, we will focus on the role of caveolins in rhabdomyosarcomas and on their potential use as markers of the degree of differentiation in these paediatric tumours. Given that the function of Cav-1 as tumour conditional gene in cancer has been well-established, we will also discuss the relationship between Cav-1 and the progression of rhabdomyosarcoma.
Collapse
Affiliation(s)
- Stefania Rossi
- Department of Biomedical Sciences and Biotechnologies, Interuniversity Institute of Myology (IIM), University of Brescia, Brescia, Italy Department of Pathology, University of Brescia, Brescia, Italy
| | | | | | | | | |
Collapse
|
16
|
Chu SH, Ma YB, Feng DF, Zhang H, Zhu ZA, Li ZQ, Jiang PC. Correlation of low SLC22A18 expression with poor prognosis in patients with glioma. J Clin Neurosci 2012; 19:95-98. [PMID: 22153794 DOI: 10.1016/j.jocn.2011.04.032] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 04/11/2011] [Accepted: 04/23/2011] [Indexed: 11/28/2022]
Abstract
We investigated the expression of the putative tumor suppressor SLC22A18 to evaluate it as a prognostic marker in glioma patients. Immunohistochemical and Western blot analyses of clinical tissue samples obtained from 120 patients with glioma were performed. Low expression of SLC22A18 was observed in 71.7% of patients. Loss of SLC22A18 expression in glioma was significantly related to pathological grade (p = 0.003). High pathological grade (World Health Organization III-IV) was correlated with negative (low or absent) expression of SLC22A18, which was correlated with a significantly shorter overall patient survival than in those with positive (high) expression (p = 0.007). Multivariate Cox regression analysis indicated that SLC22A18 expression level is an independent survival prognostic factor for patients with glioma (p = 0.011). Western blotting analysis confirmed decreased expression of SLC22A18 in glioma tissues compared with adjacent brain tissues. This study suggests that SLC22A18 functions as a tumor suppressor in glioma and represents a candidate biomarker for long-term survival in this disease.
Collapse
Affiliation(s)
- Sheng-Hua Chu
- Department of Neurosurgery, No 3 People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 280 Mo He Road, Bao Shan District, Shanghai 201900, China.
| | | | | | | | | | | | | |
Collapse
|
17
|
CHU SHENGHUA, MA YANBIN, FENG DONGFU, ZHANG HONG, QIU JIANHUA, ZHU ZHIAN. Elevated expression of solute carrier family 22 member 18 increases the sensitivity of U251 glioma cells to BCNU. Oncol Lett 2011; 2:1139-1142. [PMID: 22848278 PMCID: PMC3406540 DOI: 10.3892/ol.2011.371] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 07/29/2011] [Indexed: 02/07/2023] Open
Abstract
Previous studies showed that solute carrier family 22 member 18 (SLC22A18) is involved in tumorigenesis. The aim of this study was to examine the role of SLC22A18 in glioma cells. Glioma U251 cells were transfected with the human SLC22A18 gene. Transfection of the empty vector pcDNA3.1 was used as a negative control. Sensitivity to BCNU was measured by Annexin V staining. The expression of caspase-3 and bcl-2 was determined by immunohistochemistry. The transfection was confirmed by PCR, RT-PCR and Western blotting. Augmented apoptotic cell death was observed in the SLC22A18-transfected cells, compared to the non-transfected cells or cells with the empty vector. Caspase-3 expression increased in U251-SLC22A18 cells, whereas the bcl-2 expression decreased. These results indicated that SLC22A18 has a pro-apoptotic function in glioma cells.
Collapse
Affiliation(s)
- SHENG-HUA CHU
- Department of Neurosurgery, No. 3 People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, P.R. China
| | - YAN-BIN MA
- Department of Neurosurgery, No. 3 People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, P.R. China
| | - DONG-FU FENG
- Department of Neurosurgery, No. 3 People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, P.R. China
| | - HONG ZHANG
- Department of Neurosurgery, No. 3 People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, P.R. China
| | - JIAN-HUA QIU
- Department of Neurosurgery, No. 3 People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, P.R. China
| | - ZHI-AN ZHU
- Department of Neurosurgery, No. 3 People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, P.R. China
| |
Collapse
|
18
|
Chu SH, Feng DF, Ma YB, Zhang H, Zhu ZA, Li ZQ, Jiang PC. Promoter methylation and downregulation of SLC22A18 are associated with the development and progression of human glioma. J Transl Med 2011; 9:156. [PMID: 21936894 PMCID: PMC3184631 DOI: 10.1186/1479-5876-9-156] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Accepted: 09/21/2011] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Downregulation of the putative tumor suppressor gene SLC22A18 has been reported in a number of human cancers. The aim of this study was to investigate the relationship between SLC22A18 downregulation, promoter methylation and the development and progression of human glioma. METHOD SLC22A18 expression and promoter methylation was examined in human gliomas and the adjacent normal tissues. U251 glioma cells stably overexpressing SLC22A18 were generated to investigate the effect of SLC22A18 on cell growth and adherence in vitro using the methyl thiazole tetrazolium assay. Apoptosis was quantified using flow cytometry and the growth of SLC22A18 overexpressing U251 cells was measured in an in vivo xenograft model. RESULTS SLC22A18 protein expression is significantly decreased in human gliomas compared to the adjacent normal brain tissues. SLC22A18 protein expression is significantly lower in gliomas which recurred within six months after surgery than gliomas which did not recur within six months. SLC22A18 promoter methylation was detected in 50% of the gliomas, but not in the adjacent normal tissues of any patient. SLC22A18 expression was significantly decreased in gliomas with SLC22A18 promoter methylation, compared to gliomas without methylation. The SLC22A18 promoter is methylated in U251 cells and treatment with the demethylating agent 5-aza-2-deoxycytidine increased SLC22A18 expression and reduced cell proliferation. Stable overexpression of SLC22A18 inhibited growth and adherence, induced apoptosis in vitro and reduced in vivo tumor growth of U251 cells. CONCLUSION SLC22A18 downregulation via promoter methylation is associated with the development and progression of glioma, suggesting that SLC22A18 is an important tumor suppressor in glioma.
Collapse
Affiliation(s)
- Sheng-Hua Chu
- Department of Neurosurgery, NO.3 People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| | - Dong-Fu Feng
- Department of Neurosurgery, NO.3 People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| | - Yan-Bin Ma
- Department of Neurosurgery, NO.3 People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| | - Hong Zhang
- Department of Neurosurgery, NO.3 People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| | - Zhi-An Zhu
- Department of Neurosurgery, NO.3 People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Pu-Cha Jiang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
19
|
Saab R, Spunt SL, Skapek SX. Myogenesis and rhabdomyosarcoma the Jekyll and Hyde of skeletal muscle. Curr Top Dev Biol 2011; 94:197-234. [PMID: 21295688 DOI: 10.1016/b978-0-12-380916-2.00007-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Rhabdomyosarcoma, a neoplasm composed of skeletal myoblast-like cells, represents the most common soft tissue sarcoma in children. The application of intensive chemotherapeutics and refined surgical and radiation therapy approaches have improved survival for children with localized disease over the past 3 decades; however, these approaches have not improved the dismal outcome for children with metastatic and recurrent rhabdomyosarcoma. Elegant studies have defined the molecular mechanisms driving skeletal muscle lineage commitment and differentiation, and the machinery that couples differentiation with irreversible cell proliferation arrest. Further, detailed molecular analyses indicate that rhabdomyosarcoma cells have lost the capacity to fully differentiate when challenged to do so in experimental models. We review the intersection of normal skeletal muscle developmental biology and the molecular genetic defects in rhabdomyosarcoma with the underlying premise that understanding how the differentiation process has gone awry will lead to new treatment strategies aimed at promoting myogenic differentiation and concomitant cell cycle arrest.
Collapse
Affiliation(s)
- Raya Saab
- Children's Cancer Center of Lebanon, Department of Pediatrics, American University of Beirut, Beirut, Lebanon
| | | | | |
Collapse
|
20
|
He H, Xu C, Zhao Z, Qin X, Xu H, Zhang H. Low expression of SLC22A18 predicts poor survival outcome in patients with breast cancer after surgery. Cancer Epidemiol 2010; 35:279-85. [PMID: 21144813 DOI: 10.1016/j.canep.2010.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Revised: 09/09/2010] [Accepted: 09/14/2010] [Indexed: 10/18/2022]
Abstract
PURPOSE The aims of this study were to evaluate the relationship between SLC22A18 expression and clinicopathologic features while investigate the prognostic value of SLC22A18 expression in breast cancer after surgery. SPECIMENS AND METHODS: Immunohistochemistry was used to examine SLC22A18 protein expression in the breast tumors. Prognostic value of SLC22A18 protein and other clinicopathologic factors were evaluated. The Kaplan-Meier method and the Cox proportional hazards model were used to predict factors with a significant independent prognostic value. Digital image analysis was employed to quantify immunostaining. RESULTS SLC22A18 expression was correlated with tumor size, lymph node metastasis, clinical stage, and extensive lyphovascular invasion. The results of Kaplan-Meier analysis indicated that SLC22A18 expression was associated with relapse-free survival (RFS) of breast cancer. The survival of higher expression SLC22A18 group had longer cum survival compared to the group with low expression. The difference was significant (p=0.003, log-rank test). Cox's regression analysis showed that tumor size, lymph nodes metastasis, nuclear stage, extensive lymphovascular invasion, and SLC22A18 expression might be used as prognostic factor for RFS. Nuclear stage and SLC22A18 expression were the most meaningful histopathologic parameter in predicting tumor recurrence. Compared with the group of higher SLC22A18 expression, the lower expression group was more likely to relapse. The HR is 2.624 (p=0.035). CONCLUSION Low expression of SLC22A18 was associated with tumor progression, recurrence and poor survival after breast surgery. Testing expression of SLC22A18 will be helpful for predicting prognosis in breast cancer.
Collapse
Affiliation(s)
- Hongyu He
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
21
|
Bajaj V, Singhmar P, Kumar A. Promoter characterization and regulation of expression of an imprinted gene SLC22A18AS. Gene 2008; 424:40-7. [DOI: 10.1016/j.gene.2008.07.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 07/28/2008] [Accepted: 07/28/2008] [Indexed: 10/21/2022]
|
22
|
Ali AM, Bajaj V, Gopinath KS, Kumar A. Characterization of the human SLC22A18 gene promoter and its regulation by the transcription factor Sp1. Gene 2008; 429:37-43. [PMID: 18996451 DOI: 10.1016/j.gene.2008.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 10/10/2008] [Accepted: 10/11/2008] [Indexed: 11/19/2022]
Abstract
SLC22A18, a poly-specific organic cation transporter, is paternally imprinted in humans and mice. It shows loss-of-heterozygosity in childhood and adult tumors, and gain-of-imprinting in hepatocarcinomas and breast cancers. Despite the importance of this gene, its transcriptional regulation has not been studied, and the promoter has not yet been characterized. We therefore set out to identify the potential cis-regulatory elements including the promoter of this gene. The luciferase reporter assay in human cells indicated that a region from -120 bp to +78 bp is required for the core promoter activity. No consensus TATA or CAAT boxes were found in this region, but two Sp1 binding sites were conserved in human, chimpanzee, mouse and rat. Mutational analysis of the two Sp1 sites suggested their requirement for the promoter activity. Chromatin-immunoprecipitation showed binding of Sp1 to the promoter region in vivo. Overexpression of Sp1 in Drosophila Sp1-null SL2 cells suggested that Sp1 is the transactivator of the promoter. The human core promoter was functional in mouse 3T3 and monkey COS7 cells. We found a CpG island which spanned the core promoter and exon 1. COBRA technique did not reveal promoter methylation in 10 normal oral tissues, 14 oral tumors, and two human cell lines HuH7 and A549. This study provides the first insight into the mechanism that controls expression of this imprinted tumor suppressor gene. A COBRA-based assay has been developed to look for promoter methylation in different cancers. The present data will help to understand the regulation of this gene and its role in tumorigenesis.
Collapse
Affiliation(s)
- Abdullah Mahmood Ali
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | | | | | | |
Collapse
|
23
|
Langenau DM, Keefe MD, Storer NY, Guyon JR, Kutok JL, Le X, Goessling W, Neuberg DS, Kunkel LM, Zon LI. Effects of RAS on the genesis of embryonal rhabdomyosarcoma. Genes Dev 2007; 21:1382-95. [PMID: 17510286 PMCID: PMC1877750 DOI: 10.1101/gad.1545007] [Citation(s) in RCA: 273] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Accepted: 04/03/2007] [Indexed: 02/07/2023]
Abstract
Embryonal rhabdomyosarcoma (ERMS) is a devastating cancer with specific features of muscle differentiation that can result from mutational activation of RAS family members. However, to date, RAS pathway activation has not been reported in a majority of ERMS patients. Here, we have created a zebrafish model of RAS-induced ERMS, in which animals develop externally visible tumors by 10 d of life. Microarray analysis and cross-species comparisons identified two conserved gene signatures found in both zebrafish and human ERMS, one associated with tumor-specific and tissue-restricted gene expression in rhabdomyosarcoma and a second comprising a novel RAS-induced gene signature. Remarkably, our analysis uncovered that RAS pathway activation is exceedingly common in human RMS. We also created a new transgenic coinjection methodology to fluorescently label distinct subpopulations of tumor cells based on muscle differentiation status. In conjunction with fluorescent activated cell sorting, cell transplantation, and limiting dilution analysis, we were able to identify the cancer stem cell in zebrafish ERMS. When coupled with gene expression studies of this cell population, we propose that the zebrafish RMS cancer stem cell shares similar self-renewal programs as those found in activated satellite cells.
Collapse
MESH Headings
- Adenosine Deaminase/genetics
- Animals
- Animals, Genetically Modified
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Differentiation
- Cell Transformation, Neoplastic
- Cells, Cultured
- DNA-Binding Proteins/genetics
- Embryo, Nonmammalian/cytology
- Embryo, Nonmammalian/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Developmental
- Genes, ras/physiology
- Humans
- In Situ Hybridization
- Kidney/cytology
- Kidney/metabolism
- Kidney/pathology
- Microinjections
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Oligonucleotide Array Sequence Analysis
- RNA-Binding Proteins
- Rhabdomyosarcoma, Embryonal/etiology
- Rhabdomyosarcoma, Embryonal/genetics
- Rhabdomyosarcoma, Embryonal/pathology
- Zebrafish/genetics
- Zebrafish/metabolism
Collapse
Affiliation(s)
- David M. Langenau
- Stem Cell Program and Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Matthew D. Keefe
- Stem Cell Program and Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Narie Y. Storer
- Stem Cell Program and Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Jeffrey R. Guyon
- Program in Genomics and Howard Hughes Medical Institute at Children’s Hospital Boston, Boston, Massachusetts 02115, USA
| | - Jeffery L. Kutok
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts 02115, USA
| | - Xiuning Le
- Stem Cell Program and Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Wolfram Goessling
- Stem Cell Program and Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | - Louis M. Kunkel
- Program in Genomics and Howard Hughes Medical Institute at Children’s Hospital Boston, Boston, Massachusetts 02115, USA
| | - Leonard I. Zon
- Stem Cell Program and Division of Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| |
Collapse
|
24
|
Gallagher E, Mc Goldrick A, Chung WY, Mc Cormack O, Harrison M, Kerin M, Dervan PA, Mc Cann A. Gain of imprinting of SLC22A18 sense and antisense transcripts in human breast cancer. Genomics 2006; 88:12-7. [PMID: 16624517 DOI: 10.1016/j.ygeno.2006.02.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 01/27/2006] [Accepted: 02/04/2006] [Indexed: 11/28/2022]
Abstract
The 11p15.5 region harbors three imprinted sense/antisense transcript pairs, SLC22A18/SLC22A18AS, IGF2/IGF2AS (PEG8), and KCNQ1/KCNQ1OT1 (LIT1). SLC22A18 (solute carrier family 22 (organic cation transporter) member 18) and its antisense transcript SLC22A18AS are paternally suppressed in fetal samples. In adult tissue, SLC22A18 displays polymorphic imprinting, but the imprinting status of SLC22A18AS remains elusive. SLC22AI8 DNA-PCR-RFLP analysis using NlaIII restriction digestion identified SLC22A18 heterozygotes within this breast tissue cohort (n = 89). Commercial sequencing identified informative SLC22A18AS samples. Random hexamer-primed cDNA synthesis, SLC22A18/SLC22A18AS-specific PCR, and imprinting evaluation by commercial sequencing demonstrated that SLC22A18AS displays a nonimprinted profile in reduction mastectomies (n = 6). However, SLC22A18 showed a gain of imprinting (GOI) in 1/4 of these normal cases. In the malignant cohort, GOI was also demonstrated in 18% for SLC22A18 and 14% for SLC22A18AS, occurring concomitantly in one case. This study reports the imprinting status of SLC22A18AS in adult tissue, and shows that GOI affects both the sense, and antisense transcripts at this locus in human breast tissue.
Collapse
Affiliation(s)
- E Gallagher
- School of Medicine and Medical Science, UCD Conway Institute of Biomolecular and Biomedical Research, Belfield, Dublin 4, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Yamada HY, Gorbsky GJ. Tumor suppressor candidate TSSC5 is regulated by UbcH6 and a novel ubiquitin ligase RING105. Oncogene 2006; 25:1330-9. [PMID: 16314844 PMCID: PMC2713668 DOI: 10.1038/sj.onc.1209167] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The region of human chromosome 11p15.5 is linked with Beckwith-Wiedemann syndrome that is associated with susceptibility to Wilms' tumor, rhabdomyosarcoma and hepatoblastoma. TSSC5 (tumor-suppressing subchromosomal transferable fragment cDNA; also known as ORCTL2/IMPT1/BWR1A/SLC22A1L) is located in the region. The expression of TSSC5 and other genes in the region is regulated through paternal imprinting. Mutations and/or reduced expression of TSSC5 have been found in certain tumors. TSSC5 encodes an efflux transporter-like protein with 10 transmembrane domains, whose regulation may affect drug sensitivity, cellular metabolism and growth. Here, we present evidences indicating that RING105, a novel conserved RING-finger protein with a PA (protease-associated) domain and a PEST sequence, is a ubiquitin ligase for TSSC5 that can function in concert with the ubiquitin-conjugating enzyme UbcH6. The polyubiquitin target site on TSSC5 was mapped to a region in the 6th hydrophilic loop. Ectopic expression of RING105 in HeLa cells caused an accumulation of cells during G1 that was not observed with the expression of a form of RING105 in which a residue within the RING finger was mutated to inactivate its ligase activity. UbcH6-RING105 may define a novel ubiquitin-proteasome pathway that targets TSSC5 in mammalian cells.
Collapse
Affiliation(s)
- H Y Yamada
- Oklahoma Medical Research Foundation (OMRF), Molecular, Cell and Developmental Biology Research Program, Oklahoma City, OK 73104, USA.
| | | |
Collapse
|
26
|
Bajaj V, Markandaya M, Krishna L, Kumar A. Paternal imprinting of the SLC22A1LS gene located in the human chromosome segment 11p15.5. BMC Genet 2004; 5:13. [PMID: 15175115 PMCID: PMC425576 DOI: 10.1186/1471-2156-5-13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2004] [Accepted: 06/03/2004] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genomic imprinting is an epigenetic chromosomal modification in the gametes or zygotes that results in a non-random monoallelic expression of specific autosomal genes depending upon their parent of origin. Approximately 44 human genes have been reported to be imprinted. A majority of them are clustered, including some on chromosome segment 11p15.5. We report here the imprinting status of the SLC22A1LS gene from the human chromosome segment 11p15.5 RESULTS In order to test for allele specific expression patterns, PCR primer sets from the SLC22A1LS gene were used to look for heterozygosity in DNA samples from 17 spontaneous abortuses using PCR-SSCP and DNA sequence analyses. cDNA samples from different tissues of spontaneous abortuses showing heterozygosity were subjected to PCR-SSCP analysis to determine the allele specific expression pattern. PCR-SSCP analysis revealed heterozygosity in two of the 17 abortuses examined. DNA sequence analysis showed that the heterozygosity is caused by a G>A change at nucleotide position 473 (c.473G>A) in exon 4 of the SLC22A1LS gene. PCR-SSCP analysis suggested that this gene is paternally imprinted in five fetal tissues examined. CONCLUSIONS This study reports the imprinting status of the SLC22A1LS gene for the first time. The results suggest imprinting of the paternal allele of this gene in five fetal tissues: brain, liver, placenta, kidneys and lungs.
Collapse
Affiliation(s)
- Vineeta Bajaj
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Manjunath Markandaya
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Lingegowda Krishna
- Department of Obstetrics and Gynecology, Kempegowda Institute of Medical Sciences, Bangalore, India
| | - Arun Kumar
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| |
Collapse
|
27
|
Albrecht S, Hartmann W, Houshdaran F, Koch A, Gärtner B, Prawitt D, Zabel BU, Russo P, Von Schweinitz D, Pietsch T. Allelic loss but absence of mutations in the polyspecific transporter geneBWR1Aon 11p15.5 in hepatoblastoma. Int J Cancer 2004; 111:627-32. [PMID: 15239143 DOI: 10.1002/ijc.20280] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chromosomal region 11p15.5 shows frequent maternal allelic loss in embryonal tumors, including rhabdomyosarcoma (RMS), Wilms' tumor (WT) and hepatoblastoma (HB), consistent with the presence of at least one tumor suppressor gene in this region, which should be paternally imprinted, i.e., expressed from the maternal allele only. The BWR1A gene encodes a polyspecific transmembrane transporter and is located on 11p15.5. It is highly expressed in liver, paternally imprinted and was found to be mutated in an RMS cell line, making it a plausible tumor suppressor gene for HB. We therefore screened 62 HBs, 3 HB cell lines and 1 pediatric hepatocellular carcinoma for BWR1A mutations using single-strand conformation polymorphism analysis. Allelic loss on 11p15.5 was assessed by PCR-based microsatellite analysis in 56 of the cases for which constitutional DNA was available. BWR1A mRNA expression was determined in 14 HBs by differential RT-PCR of matched cDNA samples from tumor and normal liver. Western blot analysis was performed on 4 tumors and matching normal liver tissue. Except for sequence polymorphisms (in exons 2, 3 and 10 as well as in introns 6 and 7), no mutations were found. Thirteen HBs (23%) had allelic loss on 11p15.5; this included BWR1A in 12 but it was telomeric to BWR1A in 1. Expression of BWR1A mRNA was reduced in 11 out of 14 cases by 19-92%, independent from allelic loss of 11p15.5. By Western blot analysis, all 4 tumors and matching liver samples displayed a 48-51 kd band corresponding to BWR1A. These results make it unlikely that BWR1A is the target of the allelic deletions in HB. However, similar to the putative 11p15.5 tumor suppressor H19, BWR1A appears to be reduced in expression. Reduced expression in the absence of mutations may contribute to HB development; however, to understand the significance of this finding will require further studies on the function of BWR1A, specifically its role in liver development.
Collapse
Affiliation(s)
- Steffen Albrecht
- Department of Pathology, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Tzortzaki EG, Yang M, Glass D, Deng L, Evan AP, Bledsoe SB, Stambrook PJ, Sahota A, Tischfield JA. Impaired expression of an organic cation transporter, IMPT1, in a knockout mouse model for kidney stone disease. UROLOGICAL RESEARCH 2003; 31:257-61. [PMID: 12856169 DOI: 10.1007/s00240-003-0318-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2002] [Accepted: 03/20/2003] [Indexed: 10/26/2022]
Abstract
The imprinted multimembrane-spanning polyspecific transporter-like gene 1 ( IMPT1) encodes a predicted protein with organic cation transport capabilities. As a first step in understanding the function of IMPT1, we identified the renal structures expressing this gene in knockout mice with adenine phosphoribosyltransferase (APRT) deficiency and 2,8-dihydroxyadenine (DHA) nephrolithiasis. IMPT1 mRNA was not detected using a standard in situ hybridization (ISH) protocol, but we observed intense staining in cortico-medullary tubules and glomeruli in wild-type mice using an improved reverse transcription-polymerase chain reaction (RT-PCR) ISH procedure. IMPT1 mRNA expression was significantly decreased in the cortical region in kidney sections from APRT-deficient male mice. APRT-deficient female mice are less severely affected by DHA-induced kidney stone disease, and we observed only a modest reduction in IMPT1 expression in kidneys from these mice. IMPT1 expression in APRT heterozygous mice was comparable to that in wild-type mice, suggesting imprinting of one of the parental alleles. These findings suggest that decreased IMPT1 mRNA expression may contribute to the impaired renal function in APRT-deficient male mice, and that RT-PCR ISH is a valuable tool for localizing the site of expression of transcripts that are not detectable using standard ISH procedures.
Collapse
Affiliation(s)
- Eleni G Tzortzaki
- Department of Genetics, Rutgers University, 604 Allison Road, NJ 08854-8082, Piscataway, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
The diagnostic use of cytogenetic and molecular genetic techniques in the assessment of small round cell tumours. ACTA ACUST UNITED AC 2002. [DOI: 10.1054/cdip.2002.0135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
30
|
Lu R, Moore PA, Pitha PM. Stimulation of IRF-7 gene expression by tumor necrosis factor alpha: requirement for NFkappa B transcription factor and gene accessibility. J Biol Chem 2002; 277:16592-8. [PMID: 11877397 DOI: 10.1074/jbc.m111440200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Interferon regulatory factor 7 (IRF-7) plays an important role in innate immunity, where, together with IRF-3, it controls the expression of interferon A/B genes as well as chemokine RANTES (regulated on activation normal T cell expressed and secreted). Previously, we characterized human IRF-7 promoter and showed that an interferon-stimulated response element site in the first intron binds interferon-stimulated gene factor 3 (ISGF3) and confers the response to interferon. Here we report the stimulation of IRF-7 expression by 12-O-tetradecanoylphorbol-13-acetate (TPA) and tumor necrosis factor alpha (TNFalpha) in human peripheral blood monocytes. Using promoter analysis in combination with electrophoretic mobility shift assays, we have demonstrated that an NFkappaB site located next to the TATA box, binds p50 and p65 heterodimer and is required for the induction of the IRF-7 gene by TPA and TNFalpha. In addition, we report stimulation of IRF-7 gene expression by topoisomerase II (TOPII) inhibitors. We show by chromatin immunoprecipitation assay that treatment with the TOPII inhibitor etoposide induces association of acetylated histone 3 with the promoter of IRF-7 gene, indicating that TOPII-mediated changes in chromatin structure could be responsible for the induction. This suggests that the IRF-7 gene is localized in the condensed area of the chromosome where it is inaccessible to transcription factors that would promote its constitutive expression.
Collapse
Affiliation(s)
- Runqing Lu
- The Sidney Kimmel Comprehensive Cancer Center and the Department of Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | | | | |
Collapse
|
31
|
Scelfo RAM, Schwienbacher C, Veronese A, Gramantieri L, Bolondi L, Querzoli P, Nenci I, Calin GA, Angioni A, Barbanti-Brodano G, Negrini M. Loss of methylation at chromosome 11p15.5 is common in human adult tumors. Oncogene 2002; 21:2564-72. [PMID: 11971191 DOI: 10.1038/sj.onc.1205336] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2001] [Revised: 01/10/2002] [Accepted: 01/11/2002] [Indexed: 11/09/2022]
Abstract
Chromosome 11p15 deletion is frequent in human tumors, suggesting the presence of at least one tumor suppressor gene within this region. While mutation analyses of local genes revealed only rare mutations, we have previously described a mechanism, gain of imprinting, that leads to loss of expression of genes located on the maternal 11p15 chromosome in human hepatocarcinomas. Loss of expression was often associated with loss of maternal-specific methylation at the KvDMR1 locus. Here, we show that loss of the maternal KvDMR1 methylation is common, ranging from 30 to 50%, to a variety of adult neoplasms, including liver, breast, cervical and gastric carcinomas. We found that other 11p15.5 loci were concomitantly hypomethylated, indicating that loss of KvDMR1 methylation occurred in the context of a common mechanism affecting the methylation of a large 11p15 subchromosomal domain. These epigenetic abnormalities were not detected in any normal somatic tissue. Therefore, it seems possible that, contrary to the repression of promoter activity caused by hypermethylation, loss of gene expression at 11p15.5 may result from the activation, by hypomethylation, of one or more negative regulatory elements.
Collapse
Affiliation(s)
- Rosaria A M Scelfo
- Dipartimento di Medicina Sperimentale e Diagnostica, Università di Ferrara, Via Luigi Borsari 46, 44100 Ferrara, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Jonas RE, Kimonis VE. Chest wall hamartoma with Wiedemann-Beckwith syndrome: clinical report and brief review of chromosome 11p15.5-related tumors. AMERICAN JOURNAL OF MEDICAL GENETICS 2001; 101:221-5. [PMID: 11424137 DOI: 10.1002/ajmg.1381] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A girl born with a left chest wall hamartoma, macroglossia, nevus flammeus of the middle forehead, and a small umbilical hernia developed left lower extremity hemihypertrophy by 1 year of age and is assumed to have Wiedemann-Beckwith syndrome. Hamartoma of the bladder and a cardiac fibrous hamartoma have been reported previously in association with Wiedemann-Beckwith syndrome. Infantile hamartomas are exceedingly rare and add to the spectrum of tumor formation in the syndrome.
Collapse
Affiliation(s)
- R E Jonas
- Department of Pediatrics, Southern Illinois University School of Medicine, Springfield, Illinois 62794-9230, USA
| | | |
Collapse
|
33
|
Sakatani T, Wei M, Katoh M, Okita C, Wada D, Mitsuya K, Meguro M, Ikeguchi M, Ito H, Tycko B, Oshimura M. Epigenetic heterogeneity at imprinted loci in normal populations. Biochem Biophys Res Commun 2001; 283:1124-30. [PMID: 11355889 DOI: 10.1006/bbrc.2001.4916] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Genomic imprinting is the phenomenon by which the two alleles of certain genes are differentially expressed according to their parental origin. Extensive analysis of allelic expression at multiple imprinted loci in a normal population has not performed so far. In the present study, we examined the allelic expression pattern of three imprinted genes in a panel of 262 Japanese normal individuals. We observed differences in the extent of maintenance of allele-specific expression of the three genes. The allelic expression of small nuclear ribonucleoprotein N (SNRPN) was stringently regulated while that of multimembrane-spanning polyspecific transporter-like gene 1 (IMPT1) showed a large degree of variation. Significant biallelic expression of insulin-like growth factor II (IGF2) was observed in about 10% of normal individuals. Our findings add to the accumulating evidence for variable allelic expression at multiple loci in a normal human population. This epigenetic heterogeneity can be a stable trait and potentially influence individual phenotypes.
Collapse
Affiliation(s)
- T Sakatani
- CREST Program of the Japan Science and Technology Corporation (JST), Tottori University, Yonago, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Schwienbacher C, Gramantieri L, Scelfo R, Veronese A, Calin GA, Bolondi L, Croce CM, Barbanti-Brodano G, Negrini M. Gain of imprinting at chromosome 11p15: A pathogenetic mechanism identified in human hepatocarcinomas. Proc Natl Acad Sci U S A 2000; 97:5445-9. [PMID: 10779553 PMCID: PMC25848 DOI: 10.1073/pnas.090087497] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Genomic imprinting is a reversible condition that causes parental-specific silencing of maternally or paternally inherited genes. Analysis of DNA and RNA from 52 human hepatocarcinoma samples revealed abnormal imprinting of genes located at chromosome 11p15 in 51% of 37 informative samples. The most frequently detected abnormality was gain of imprinting, which led to loss of expression of genes present on the maternal chromosome. As compared with matched normal liver tissue, hepatocellular carcinomas showed extinction or significant reduction of expression of one of the alleles of the CDKN1C, SLC22A1L, and IGF2 genes. Loss of maternal-specific methylation at the KvDMR1 locus in hepatocarcinoma correlated with abnormal expression of CDKN1C and IGF2, suggesting a function for KvDMR1 as a long-range imprinting center active in adult tissues. These results point to the role of epigenetic mechanisms leading to loss of expression of imprinted genes at chromosome region 11p15 in human tumors.
Collapse
Affiliation(s)
- C Schwienbacher
- Dipartimento di Medicina Sperimentale e Diagnostica, Università di Ferrara, via Luigi Borsari 46, 44100 Ferrara, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Itakura J, Nagayama K, Enomoto N, Sakamoto N, Tazawa J, Izumi N, Marumo F, Sato C. CD81 nucleotide mutation in hepatocellular carcinoma and lack of CD81 polymorphism in patients at stages of hepatitis C virus infection. J Med Virol 2000. [DOI: 10.1002/1096-9071(200101)63:1<22::aid-jmv1003>3.0.co;2-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
36
|
|
37
|
Bl�ker H, Hofmann WJ, Rieker RJ, Penzel R, Graf M, Otto HF. ?-catenin accumulation and mutation of theCTNNB1 gene in hepatoblastoma. Genes Chromosomes Cancer 1999. [DOI: 10.1002/(sici)1098-2264(199908)25:4<399::aid-gcc14>3.0.co;2-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
38
|
Abstract
Genes are recognized as undergoing genomic imprinting when they are capable of being expressed only from the paternal or only from the maternal chromosome. The process can occur coordinately within large physical domains in mammalian chromosomes. One interesting facet of the study of genomic imprinting is that it offers insight into the regulation of large chromosomal regions. Understanding this regulation involves elucidating the cis-acting regulators of gene expression and defining the elements that maintain chromatin insulation, both required for understanding more practically applicable areas of biological research, such as efficient transgene production. This review is focused on the regulation of the imprinted domain of human chromosome 11p15.5, responsible for Beckwith-Wiedemann syndrome (BWS). Recent findings indicate that the maintenance of imprinting within this domain is critically dependent on the stable maintenance of chromatin insulation.
Collapse
Affiliation(s)
- J M Greally
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520-8005, USA.
| |
Collapse
|
39
|
Falls JG, Pulford DJ, Wylie AA, Jirtle RL. Genomic imprinting: implications for human disease. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 154:635-47. [PMID: 10079240 PMCID: PMC1866410 DOI: 10.1016/s0002-9440(10)65309-6] [Citation(s) in RCA: 237] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/06/1999] [Indexed: 01/14/2023]
Abstract
Genomic imprinting refers to an epigenetic marking of genes that results in monoallelic expression. This parent-of-origin dependent phenomenon is a notable exception to the laws of Mendelian genetics. Imprinted genes are intricately involved in fetal and behavioral development. Consequently, abnormal expression of these genes results in numerous human genetic disorders including carcinogenesis. This paper reviews genomic imprinting and its role in human disease. Additional information about imprinted genes can be found on the Genomic Imprinting Website at http://www.geneimprint.com.
Collapse
Affiliation(s)
- J G Falls
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina27710, USA
| | | | | | | |
Collapse
|
40
|
Reece M, Prawitt D, Landers J, Kast C, Gros P, Housman D, Zabel BU, Pelletier J. Functional characterization of ORCTL2--an organic cation transporter expressed in the renal proximal tubules. FEBS Lett 1998; 433:245-50. [PMID: 9744804 DOI: 10.1016/s0014-5793(98)00907-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chromosome 11p15.5 harbors a gene or genes involved in Beckwith-Wiedemann syndrome that confer(s) susceptibility to Wilms' tumor, rhabdomyosarcoma, and hepatoblastoma. We have previously identified a transcript at 11p15.5 which encodes a putative membrane transport protein, designated organic cation transporter-like 2 (ORCTL2), that shares homology with tetracycline resistance proteins and bacterial multidrug resistance proteins. In this report, we have investigated the transport properties of ORCTL2 and show that this protein can confer resistance to chloroquine and quinidine when overexpressed in bacteria. Immunohistochemistry analyses performed with anti-ORCTL2 polyclonal antibodies on human renal sections indicate that ORCTL2 is localized on the apical membrane surface of the proximal tubules. These results suggest that ORCTL2 may play a role in the transport of chloroquine and quinidine related compounds in the kidney.
Collapse
Affiliation(s)
- M Reece
- Department of Biochemistry, McGill University, Montreal, Que., Canada
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Genomic imprinting is the phenomenon by which individual alleles of certain genes are expressed differentially according to their parent of origin. The alleles appear to be differentially marked during gametogenesis or during the early part of development. This mark is heritable but reversible from generation to generation, implying a stable epigenetic modification. Approximately 25 imprinted genes have been identified to date, and dysregulation of a number of these has been implicated in tumour development. The normal physiological role of many imprinted genes is in the control of cell proliferation and fetal growth, indicating potential mechanisms of action in tumour formation. Both dominant and recessive modes of action have been postulated for the role of imprinted genes in neoplasia, as a result of effective gene dosage alterations by epigenetic modification of the normal pattern of allele specific transcription. The aim of this review is to assess the importance of imprinted genes in generating tumours and to discuss the implications for novel mechanisms of transforming mutation.
Collapse
Affiliation(s)
- J A Joyce
- Department of Anatomy, University of Cambridge, UK.
| | | |
Collapse
|