1
|
Wang X, Yu J, Wang J. Neural Tube Defects and Folate Deficiency: Is DNA Repair Defective? Int J Mol Sci 2023; 24:ijms24032220. [PMID: 36768542 PMCID: PMC9916799 DOI: 10.3390/ijms24032220] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Neural tube defects (NTDs) are complex congenital malformations resulting from failure of neural tube closure during embryogenesis, which is affected by the interaction of genetic and environmental factors. It is well known that folate deficiency increases the incidence of NTDs; however, the underlying mechanism remains unclear. Folate deficiency not only causes DNA hypomethylation, but also blocks the synthesis of 2'-deoxythymidine-5'-monophosphate (dTMP) and increases uracil misincorporation, resulting in genomic instabilities such as base mismatch, DNA breakage, and even chromosome aberration. DNA repair pathways are essential for ensuring normal DNA synthesis, genomic stability and integrity during embryonic neural development. Genomic instability or lack of DNA repair has been implicated in risk of development of NTDs. Here, we reviewed the relationship between folate deficiency, DNA repair pathways and NTDs so as to reveal the role and significance of DNA repair system in the pathogenesis of NTDs and better understand the pathogenesis of NTDs.
Collapse
|
2
|
Li H, Wang X, Zhao H, Wang F, Bao Y, Guo J, Chang S, Wu L, Cheng H, Chen S, Zou J, Cui X, Niswander L, Finnell RH, Wang H, Zhang T. Low folate concentration impacts mismatch repair deficiency in neural tube defects. Epigenomics 2019; 12:5-18. [PMID: 31769301 DOI: 10.2217/epi-2019-0279] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aim: To know the cause of sequence variants in neural tube defect (NTD). Materials & methods: We sequenced genes implicated in neural tube closure (NTC) in a Chinese cohort and elucidated the molecular mechanism-driving mutations. Results: In NTD cases, an increase in specific variants was identified, potentially deleterious rare variants harbored in H3K36me3 occupancy regions that recruits mismatch repair (MMR) machinery. Lower folate concentrations in local brain tissues were also observed. In neuroectoderm cells, folic acid insufficiency attenuated association of Msh6 to H3K36me3, and reduced bindings to NTC genes. Rare variants in human NTDs were featured by MMR deficiency and more severe microsatellite instability. Conclusion: Our work suggests a mechanistic link between folate insufficiency and MMR deficiency that correlates with an increase of rare variants in NTC genes.
Collapse
Affiliation(s)
- Huili Li
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China.,Department of Molecular, Cellular & Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Xiaolei Wang
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Huizhi Zhao
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Fang Wang
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Yihua Bao
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Jin Guo
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Shaoyan Chang
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Lihua Wu
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Haiqin Cheng
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Shuyuan Chen
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Jizhen Zou
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Xiaodai Cui
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Lee Niswander
- Department of Molecular, Cellular & Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Richard H Finnell
- Obstetrics & Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction & Development, Fudan University, Shanghai 200011, China.,Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hongyan Wang
- Obstetrics & Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction & Development, Fudan University, Shanghai 200011, China.,Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center of Genetics & Development, Fudan University, Shanghai 200032, China.,Children's Hospital, Fudan University, Shanghai 201102, China
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development & Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| |
Collapse
|
3
|
Kok DE, Steegenga WT, Smid EJ, Zoetendal EG, Ulrich CM, Kampman E. Bacterial folate biosynthesis and colorectal cancer risk: more than just a gut feeling. Crit Rev Food Sci Nutr 2018; 60:244-256. [DOI: 10.1080/10408398.2018.1522499] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Dieuwertje E. Kok
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Wilma T. Steegenga
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Eddy J. Smid
- Laboratory of Food Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Erwin G. Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Cornelia M. Ulrich
- Department of Population Health Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Ellen Kampman
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
4
|
Khalili H, Gong J, Brenner H, Austin TR, Hutter CM, Baba Y, Baron JA, Berndt SI, Bézieau S, Caan B, Campbell PT, Chang-Claude J, Chanock SJ, Chen C, Hsu L, Jiao S, Conti DV, Duggan D, Fuchs CS, Gala M, Gallinger S, Haile RW, Harrison TA, Hayes R, Hazra A, Henderson B, Haiman C, Hoffmeister M, Hopper JL, Jenkins MA, Kolonel LN, Küry S, LaCroix A, Marchand LL, Lemire M, Lindor NM, Ma J, Manson JE, Morikawa T, Nan H, Ng K, Newcomb PA, Nishihara R, Potter JD, Qu C, Schoen RE, Schumacher FR, Seminara D, Taverna D, Thibodeau S, Wactawski-Wende J, White E, Wu K, Zanke BW, Casey G, Hudson TJ, Kraft P, Peters U, Slattery ML, Ogino S, Chan AT. Identification of a common variant with potential pleiotropic effect on risk of inflammatory bowel disease and colorectal cancer. Carcinogenesis 2015; 36:999-1007. [PMID: 26071399 DOI: 10.1093/carcin/bgv086] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 06/06/2015] [Indexed: 12/12/2022] Open
Abstract
Although genome-wide association studies (GWAS) have separately identified many genetic susceptibility loci for ulcerative colitis (UC), Crohn's disease (CD) and colorectal cancer (CRC), there has been no large-scale examination for pleiotropy, or shared genetic susceptibility, for these conditions. We used logistic regression modeling to examine the associations of 181 UC and CD susceptibility variants previously identified by GWAS with risk of CRC using data from the Genetics and Epidemiology of Colorectal Cancer Consortium and the Colon Cancer Family Registry. We also examined associations of significant variants with clinical and molecular characteristics in a subset of the studies. Among 11794 CRC cases and 14190 controls, rs11676348, the susceptibility single nucleotide polymorphism (SNP) for UC, was significantly associated with reduced risk of CRC (P = 7E-05). The multivariate-adjusted odds ratio of CRC with each copy of the T allele was 0.93 (95% CI 0.89-0.96). The association of the SNP with risk of CRC differed according to mucinous histological features (P heterogeneity = 0.008). In addition, the (T) allele was associated with lower risk of tumors with Crohn's-like reaction but not tumors without such immune infiltrate (P heterogeneity = 0.02) and microsatellite instability-high (MSI-high) but not microsatellite stable or MSI-low tumors (P heterogeneity = 0.03). The minor allele (T) in SNP rs11676348, located downstream from CXCR2 that has been implicated in CRC progression, is associated with a lower risk of CRC, particularly tumors with a mucinous component, Crohn's-like reaction and MSI-high. Our findings offer the promise of risk stratification of inflammatory bowel disease patients for complications such as CRC.
Collapse
Affiliation(s)
- Hamed Khalili
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Jian Gong
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany, German Cancer Cosortium (DKTK), Heidelberg, Germany
| | | | - Carolyn M Hutter
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Kumamoto University, Kumamoto, Japan
| | - John A Baron
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | - Bette Caan
- Division of Hematology, Faculty of Medicine, The University of Ottawa, Ottawa, ON, Canada
| | - Peter T Campbell
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Constance Chen
- Program in Molecular and Genetic Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shuo Jiao
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - David V Conti
- Keck School of Medicine, University of Southern California, Los Angles, CA, USA
| | - David Duggan
- Systems Imagination, Computational Biology, Pheonix, AZ, USA
| | - Charles S Fuchs
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA, Department of Epidemiology, Indiana University School of Public Health, Indianapolis, IN, USA
| | - Manish Gala
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Steven Gallinger
- Department of Surgery, University Health Network Toronto General Hospital, Toronto, ON, Canada
| | | | - Tabitha A Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Richard Hayes
- Division of Epidemiology, New York University School of Medicine, New York, NY, USA
| | - Aditi Hazra
- Department of Epidemiology, Indiana University School of Public Health, Indianapolis, IN, USA
| | - Brian Henderson
- Keck School of Medicine, University of Southern California, Los Angles, CA, USA
| | - Chris Haiman
- Keck School of Medicine, University of Southern California, Los Angles, CA, USA
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - John L Hopper
- Melbourne School of Population Health, The University of Melbourne, Melbourne, Australia
| | - Mark A Jenkins
- Division of Epidemiology, New York University School of Medicine, New York, NY, USA
| | - Laurence N Kolonel
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Sébastien Küry
- CHU Nantes, Service de Génétique Médicale, Nantes, France
| | - Andrea LaCroix
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Mathieu Lemire
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Noralane M Lindor
- Department of Health Science Services, Mayo Clinic, Scottsdale, AZ, USA
| | - Jing Ma
- Department of Epidemiology, Indiana University School of Public Health, Indianapolis, IN, USA
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Teppei Morikawa
- Department of Pathology, The University of Tokyo Hospital, Tokyo, Japan
| | - Hongmei Nan
- Department of Epidemiology, Indiana University School of Public Health, Indianapolis, IN, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Polly A Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Reiko Nishihara
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA, Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, Department of Epidemiology, University of Washington, Seattle, WA, USA, Center for Public Health Research, Massey University, Wellington, New Zealand
| | - Conghui Qu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Robert E Schoen
- Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Daniela Seminara
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Darin Taverna
- Systems Imagination, Computational Biology, Pheonix, AZ, USA
| | - Stephen Thibodeau
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Jean Wactawski-Wende
- Department of Social and Preventive Medicine, University of Buffalo, Buffalo, NY, USA
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kana Wu
- Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - Brent W Zanke
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, USA
| | - Graham Casey
- Keck School of Medicine, University of Southern California, Los Angles, CA, USA
| | - Thomas J Hudson
- Ontario Institute for Cancer Research, Toronto, ON, Canada, Department of Medical Biophysics, University of Toronto, Toronto, ON, USA
| | - Peter Kraft
- Program in Molecular and Genetic Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA, Department of Epidemiology, Indiana University School of Public Health, Indianapolis, IN, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Martha L Slattery
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, UT, USA
| | - Shuji Ogino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrew T Chan
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA, Department of Epidemiology, Indiana University School of Public Health, Indianapolis, IN, USA,
| | | |
Collapse
|
5
|
The risk of colorectal cancer in patients with ulcerative colitis. Dig Dis Sci 2015; 60:492-501. [PMID: 25280558 DOI: 10.1007/s10620-014-3373-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 09/24/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIM Ulcerative colitis increases the risk of developing dysplasia and colitis-associated cancer (CAC). The purpose of this study was to determine the risk factors as well as protective measures for disease burden, need for colectomy and the development of CAC in ulcerative colitis (UC) patients. METHODS A cohort of n = 434 UC patients was evaluated. Data analysis was performed by univariate and multivariate logistic regression. Odds ratios (OR) and 95 % confidence intervals (CI) were calculated, and significance was assessed by the likelihood ratio test. RESULTS Mean patient age at UC diagnosis was 45.7 ± 15.1 years which manifested mainly as pancolitis (47 %) or left-sided colitis (45.2 %). CAC was detected in ten patients (2.3 %). UC disease duration was strongly associated with the risk of CAC (P < 0.0014); disease duration between 9 and 15 years: OR of 2.5 (95 % CI 0.2-41.1), more than 15 years: OR of 21.4 (95 % CI 2.6-173.6). The risk of developing dysplasia (low-grade intraepithelial neoplasia, LGIEN and high-grade intraepithelial neoplasia, HGIEN) or the need to undergo colectomy was also significantly related to disease duration (P = 0.006, P = 0.002, respectively). Established anti-inflammatory medication (e.g., 5-ASA, anti-TNF-α) significantly reduced the risk of both dysplasia and CAC (P = 0.02). CONCLUSIONS Despite the use of modern therapies for UC, CAC rates remain high. In our study, risk factors included disease duration while anti-inflammatory therapies reduced the risk. Effective control of the intestinal inflammation also reduced the disease burden as indicated by decreased risk of requiring colectomy, underscoring the need for sufficient surveillance and anti-inflammatory therapies.
Collapse
|
6
|
Li IC, Chiu CY, Wu CL, Chi JY, Jian SR, Wang SW, Chang CL. A dual-fluorescent reporter facilitates identification of thiol compounds that suppress microsatellite instability induced by oxidative stress. Free Radic Biol Med 2014; 69:86-95. [PMID: 24412704 DOI: 10.1016/j.freeradbiomed.2013.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 12/13/2013] [Accepted: 12/19/2013] [Indexed: 01/22/2023]
Abstract
The DNA mismatch-repair (MMR) system corrects replicative errors and minimizes mutations that occur at a high rate in microsatellites. Patients with chronic inflammation or inflammation-associated cancer display microsatellite instability (MSI), indicating a possible MMR inactivation. In fact, H2O2-generated oxidative stress inactivates the MMR function and increases mutation accumulation in a reporter microsatellite. However, it remains unclear whether MSI induced by oxidative stress is preventable because of the lack of a sufficiently sensitive detection assay. Here, we developed and characterized a dual-fluorescent system, utilizing DsRed harboring the (CA)13 microsatellite as a reporter and GFP for normalization, in near-isogenic human colorectal cancer cell lines. Via flow cytometry, this reporter sensitively detected H2O2-generated oxidative microsatellite mutations in a dose-dependent manner. The reporter further revealed that glutathione or N-acetylcysteine was better than aspirin and ascorbic acid for suppressing oxidative microsatellite mutations. These two thiol compounds also partially suppressed oxidative frameshift mutations in the coding microsatellites of the hMSH6 and CHK1 genes based on a fluoresceinated PCR-based assay. MSI suppression by N-acetylcysteine appears to be mediated through reduction of oxidative frameshift mutations in the coding microsatellite of hMSH6 and protection of hMSH6 and other MMR protein levels from being decreased by H2O2. Our findings suggest a linkage between oxidative damage, MMR deficiency, and MSI. The two thiol compounds are potentially valuable for preventing inflammation-associated MSI. The dual-fluorescent reporter with improved features will facilitate identification of additional compounds that modulate MSI, which is relevant to cancer initiation and progression.
Collapse
Affiliation(s)
- I-Chen Li
- Institute of Molecular Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chien-Yuan Chiu
- Institute of Oral Medicine, and National Cheng Kung University, Tainan 70101, Taiwan
| | - Chang-Lin Wu
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan
| | - Jhih-Ying Chi
- Institute of Molecular Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Siao-Ru Jian
- Institute of Oral Medicine, and National Cheng Kung University, Tainan 70101, Taiwan
| | - Shainn-Wei Wang
- Institute of Molecular Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan
| | - Christina L Chang
- Institute of Molecular Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Institute of Oral Medicine, and National Cheng Kung University, Tainan 70101, Taiwan; Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
7
|
Bacalini MG, Friso S, Olivieri F, Pirazzini C, Giuliani C, Capri M, Santoro A, Franceschi C, Garagnani P. Present and future of anti-ageing epigenetic diets. Mech Ageing Dev 2014; 136-137:101-15. [DOI: 10.1016/j.mad.2013.12.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 12/06/2013] [Accepted: 12/20/2013] [Indexed: 12/13/2022]
|
8
|
Abstract
Inflammatory bowel diseases (IBD) are accompanied by an increased risk of developing colitis-associated carcinoma (CAC). These tumors are one of the most important causes of morbidity and mortality in patients with IBD and distinctly differ from sporadic colorectal cancer in their biology and underlying mechanisms. First, this review discusses risk factors for the development of CAC and summarizes some of the most important genetic alterations and molecular pathways involved in inflammatory carcinogenesis. Then, new endoscopic techniques, such as chromoendoscopy and confocal laser endomicroscopy, and their contribution to surveillance and early detection of CAC are presented. Last, we briefly address different types of concepts for prevention (i.e. anti-inflammatory agents) and treatment (i.e. surgical resection) of CAC and give an outlook on this important aspect of IBD.
Collapse
Affiliation(s)
- Sebastian Foersch
- Department of Medicine I, Friedrich Alexander University, Erlangen, Germany
| | | | | |
Collapse
|
9
|
Abstract
SIGNIFICANCE The progressive, dose-dependent, and potentially reversible epigenetic changes observed in cancer present new opportunities in cancer risk modification and prevention using dietary and lifestyle factors. Folate, a water-soluble B vitamin, has been of intense interest because of an inverse association between folate status and the risk of several malignancies (particularly colorectal cancer) and its potential to modulate DNA methylation. Aberrant patterns and dysregulation of DNA methylation are mechanistically related to carcinogenesis. RECENT ADVANCES The effects of folate on DNA methylation patterns have recently been investigated in two important life stages: pre- and early postnatal life and aging. Recent studies have demonstrated that folate exposure in the intrauterine environment and early life and during the aging process may have profound effects on DNA methylation with significant functional ramifications, including the risk of cancer. CRITICAL ISSUES Evidence from animal, human, and in vitro studies suggest that the epigenetic effects of folate on DNA methylation are highly complex. The effects are gene and site specific and appear to depend on cell type, target organ, stage of transformation, the degree and duration of folate manipulations, interactions with other methyl group donors and dietary factors, and genetic variants in the folate metabolic pathways. FUTURE DIRECTIONS The potential for folate to modulate DNA methylation and, thus, modify the risk of cancer in humans is worthy of further investigation. Due to the complex relationship between folate exposure and DNA methylation, more elaborate epidemiological, clinical, and mechanistic studies that determine the clinical, biological, and molecular effects of folate are warranted.
Collapse
Affiliation(s)
- Anna Ly
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
10
|
Duthie SJ. Folate and cancer: how DNA damage, repair and methylation impact on colon carcinogenesis. J Inherit Metab Dis 2011; 34:101-9. [PMID: 20544289 DOI: 10.1007/s10545-010-9128-0] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 02/24/2010] [Accepted: 05/05/2010] [Indexed: 12/18/2022]
Abstract
Inappropriate diet may contribute to one third of cancer deaths. Folates, a group of water-soluble B vitamins present in high concentrations in green, leafy vegetables, maintain DNA stability through their ability to donate one-carbon units for cellular metabolism. Folate deficiency has been implicated in the development of several cancers, including cancer of the colorectum, breast, ovary, pancreas, brain, lung and cervix. Generally, data from the majority of human studies suggest that people who habitually consume the highest level of folate, or with the highest blood folate concentrations, have a significantly reduced risk of developing colon polyps or cancer. However, an entirely protective role for folate against carcinogenesis has been questioned, and recent data indicate that an excessive intake of synthetic folic acid (from high-dose supplements or fortified foods) may increase human cancers by accelerating growth of precancerous lesions. Nonetheless, on balance, evidence from the majority of human studies indicates that dietary folate is genoprotective against colon cancer. Suboptimal folate status in humans is widespread. Folate maintains genomic stability by regulating DNA biosynthesis, repair and methylation. Folate deficiency induces and accelerates carcinogenesis by perturbing each of these processes. This review presents recent evidence describing how these mechanisms act, and interact, to modify colon cancer risk.
Collapse
Affiliation(s)
- Susan J Duthie
- Nutrition and Epigenetics Group, Division of Vascular Health, Rowett Institute of Nutrition and Health, University of Aberdeen, Bucksburn, Aberdeen, UK.
| |
Collapse
|
11
|
Harpaz N, Polydorides AD. Colorectal dysplasia in chronic inflammatory bowel disease: pathology, clinical implications, and pathogenesis. Arch Pathol Lab Med 2010; 134:876-95. [PMID: 20524866 DOI: 10.5858/134.6.876] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
CONTEXT Colorectal cancer, the most lethal long-term complication of chronic inflammatory bowel disease (IBD), is the culmination of a complex sequence of molecular and histologic derangements of the intestinal epithelium that are initiated and at least partially sustained by chronic inflammation. Dysplasia, the earliest histologic manifestation of this process, plays an important role in cancer prevention by providing the first clinical alert that this sequence is underway and serving as an endpoint in colonoscopic surveillance of patients at high risk for colorectal cancer. OBJECTIVE To review the histology, nomenclature, clinical implications, and molecular pathogenesis of dysplasia in IBD. DATA SOURCE Literature review and illustrations from case material. CONCLUSIONS The diagnosis and grading of dysplasia in endoscopic surveillance biopsies play a decisive role in the management of patients with IBD. Although interpathologist variation, endoscopic sampling problems, and incomplete information regarding the natural history of dysplastic lesions are important limiting factors, indirect evidence that surveillance may be an effective means of reducing cancer-related mortality in the population with IBD has helped validate the histologic criteria, nomenclature, and clinical recommendations that are the basis of current practice among pathologists and clinicians. Emerging technologic advances in endoscopy may permit more effective surveillance, but ultimately the greatest promise for cancer prevention in IBD lies in expanding our thus far limited understanding of the molecular pathogenetic relationships between neoplasia and chronic inflammation.
Collapse
Affiliation(s)
- Noam Harpaz
- Department of Pathology, The Mount Sinai School of Medicine, New York, New York 10092, USA.
| | | |
Collapse
|
12
|
Duthie SJ, Grant G, Pirie LP, Watson AJ, Margison GP. Folate deficiency alters hepatic and colon MGMT and OGG-1 DNA repair protein expression in rats but has no effect on genome-wide DNA methylation. Cancer Prev Res (Phila) 2010; 3:92-100. [PMID: 20051376 DOI: 10.1158/1940-6207.capr-09-0231] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Folate deficiency is implicated in human colon cancer. The effects of feeding rats a folate-deficient diet for 24 weeks on DNA damage (8-oxo-7,8-dihydroguanine), DNA repair [O(6)-methylguanine-DNA methyltransferase (MGMT) and 8-oxoguanine-DNA glycosylase (OGG-1) activity], and epigenetic parameters (genome-wide cytosine methylation and indices of cellular methylation status) were investigated. Relative to control diet, the folate-deficient diet resulted in significantly reduced levels of serum ( approximately 80%; P < 0.0001), whole blood ( approximately 40%; P < 0.0001), and tissue folate (between 25% and 60% depending on the tissue sampled; P < 0.05); increased plasma total homocysteine ( approximately 35%; P < 0.05); and decreased S-adenosylmethionine to S-adenosylhomocysteine concentrations ( approximately 11%; P < 0.05). There was no significant change in the levels of 5-methyldeoxycytidine in liver or colon DNA, nor in the activity of liver DNA cytosine methyltransferase. However, there were significant increases in 8-oxo-7,8-dihydroguanine (P < 0.001) in lymphocyte DNA and in levels of the DNA repair proteins OGG-1 ( approximately 27%; P < 0.03) and MGMT ( approximately 25%; P < 0.003) in the liver, but not in the colon. This may reflect the ability of the liver, but not the colon, to upregulate DNA repair enzymes in response to either elevated DNA damage or an imbalance in the nucleotide precursor pool. These results show that folate deficiency can significantly modulate DNA damage and DNA repair, providing mechanisms by which it plays a role in the etiology of human cancer. We speculate that the inability of colon tissue to respond to folate deficiency occurs in humans and may increase the potential for malignant transformation.
Collapse
Affiliation(s)
- Susan J Duthie
- Rowett Institute of Nutrition and Health, University of Aberdeen, United Kingdom.
| | | | | | | | | |
Collapse
|
13
|
Du W, Li WY, Lu R, Fang JY. Folate and fiber in the prevention of colorectal cancer: Between shadows and the light. World J Gastroenterol 2010; 16:921-6. [PMID: 20180229 PMCID: PMC2828595 DOI: 10.3748/wjg.v16.i8.921] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies and causes of cancer deaths throughout the world. Endoscopy has its functional and financial limitations; therefore, chemoprevention might be crucial in reducing the incidence of CRC. Although a number of studies have demonstrated the potential chemopreventive effects of folate (or folic acid), many challenges still remain. The relationship between folate intake and CRC risk is a complex association that might depend on many factors including gender, age, alcohol consumption, and smoking, all of which interfere with folate metabolism. The supplementary dose of fiber, the length of time required to observe the effects, and confounding factors exposed during the trial might also influence these findings. Therefore, more evidence from clinical studies is needed regarding the mechanisms that underlie the actions of bioactive food components in minimizing the risk of CRC.
Collapse
|
14
|
Diosdado B, Buffart TE, Watkins R, Carvalho B, Ylstra B, Tijssen M, Bolijn AS, Lewis F, Maude K, Verbeke C, Nagtegaal ID, Grabsch H, Mulder CJJ, Quirke P, Howdle P, Meijer GA. High-resolution array comparative genomic hybridization in sporadic and celiac disease-related small bowel adenocarcinomas. Clin Cancer Res 2010; 16:1391-401. [PMID: 20179237 DOI: 10.1158/1078-0432.ccr-09-1773] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE The molecular pathogenesis of small intestinal adenocarcinomas is not well understood. Understanding the molecular characteristics of small bowel adenocarcinoma may lead to more effective patient treatment. EXPERIMENTAL DESIGN Forty-eight small bowel adenocarcinomas (33 non-celiac disease related and 15 celiac disease related) were characterized for chromosomal aberrations by high-resolution array comparative hybridization, microsatellite instability, and APC promoter methylation and mutation status. Findings were compared with clinicopathologic and survival data. Furthermore, molecular alterations were compared between celiac disease-related and non-celiac disease-related small bowel adenocarcinomas. RESULTS DNA copy number changes were observed in 77% small bowel adenocarcinomas. The most frequent DNA copy number changes found were gains on 5p15.33-5p12, 7p22.3-7q11.21, 7q21.2-7q21.3, 7q22.1-7q34, 7q36.1, 7q36.3, 8q11.21-8q24.3, 9q34.11-9q34.3, 13q11-13q34, 16p13.3, 16p11.2, 19q13.2, and 20p13-20q13.33, and losses on 4p13-4q35.2, 5q15-5q21.1, and 21p11.2-21q22.11. Seven highly amplified regions were identified on 6p21.1, 7q21.1, 8p23.1, 11p13, 16p11.2, 17q12-q21.1, and 19q13.2. Celiac disease-related and non-celiac disease-related small bowel adenocarcinomas displayed similar chromosomal aberrations. Promoter hypermethylation of the APC gene was found in 48% non-celiac disease-related and 73% celiac disease-related small bowel adenocarcinomas. No nonsense mutations were found. Thirty-three percent of non-celiac disease-related small bowel adenocarcinomas showed microsatellite instability, whereas 67% of celiac disease-related small bowel adenocarcinomas were microsatellite unstable. CONCLUSIONS Our study characterized chromosomal aberrations and amplifications involved in small bowel adenocarcinoma. At the chromosomal level, celiac disease-related and non-celiac disease-related small bowel adenocarcinomas did not differ. A defect in the mismatch repair pathways seems to be more common in celiac disease-related than in non-celiac disease-related small bowel adenocarcinomas. In contrast to colon and gastric cancers, no APC nonsense mutations were found in small bowel adenocarcinoma. However, APC promoter methylation seems to be a common event in celiac disease-related small bowel adenocarcinoma. Clin Cancer Res; 16(5); 1391-401.
Collapse
Affiliation(s)
- Begoña Diosdado
- Departments of Pathology and Gastroenterology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
GOEL A, MITTAL A, EVSTATIEV R, NEMETH M, KRUIS W, STOLTE M, BOLAND CR, GASCHE C. In vivo effects of mesalazine or E. coli Nissle 1917 on microsatellite instability in ulcerative colitis. Aliment Pharmacol Ther 2009; 30:634-42. [PMID: 19558562 PMCID: PMC2853877 DOI: 10.1111/j.1365-2036.2009.04076.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Microsatellite instability (MSI) occurs in chronically inflamed colorectal tissue and may evolve to colitis-associated cancer. In vitro data suggest that mesalazine (5-ASA) improves MSI. AIM To analyse the changes in MSI in 156 distal colonic biopsies of 39 patients with ulcerative colitis that had been treated within a randomized, double-blind trial comparing 5-ASA with E. coli Nissle (EcN). METHODS Two biopsies had been collected before and after 1 year of treatment. MSI testing was performed using a panel of eight markers, including 3 dinucleotide and 5 mononucleotide repeats. RESULTS No MSI was observed with any of the mono-repeats, and among dinucleotide repeats, only D5S346 (maps to APC) and D17S250 (p53) were consistently informative. Overall, 31/156 (20%) biopsies displayed MSI. After 1 year, 3/11 patients displayed MSI improvement [change to microsatellite stability (MSS); 1 on 5-ASA, 2 on EcN] at D5S346 and 4/11 showed MSI worsening (change from MSS to MSI; all 5-ASA). For D17S250, the corresponding data were for 3/9 patients (2 on 5-ASA, 1 on EcN) and 2/9 (both on 5-ASA), respectively. CONCLUSIONS In the set of biopsies taken from patients treated with 1.5 g 5-ASA for 1 year, there was no improvement in the prevalence of MSI in the distal colon.
Collapse
Affiliation(s)
- A. GOEL
- Division of Gastroenterology, Department of Internal Medicine and Charles A Sammons Cancer Center, Baylor University Medical Center, Dallas, TX 75246, USA
| | - A. MITTAL
- Division of Gastroenterology, Department of Internal Medicine and Charles A Sammons Cancer Center, Baylor University Medical Center, Dallas, TX 75246, USA
| | - R. EVSTATIEV
- Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - M. NEMETH
- Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - W. KRUIS
- Evangelisches Krankenhaus Kalk, Cologne, Germany
| | - M. STOLTE
- Department of Pathology, Klinikum Bayreuth, Germany
| | - C. R. BOLAND
- Division of Gastroenterology, Department of Internal Medicine and Charles A Sammons Cancer Center, Baylor University Medical Center, Dallas, TX 75246, USA
| | - C. GASCHE
- Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
,Christian Doppler Laboratory on Molecular Cancer Chemoprevention, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Odin E, Wettergren Y, Nilsson S, Carlsson G, Gustavsson B. Colorectal carcinomas with microsatellite instability display increased thymidylate synthase gene expression levels. Clin Colorectal Cancer 2008; 6:720-7. [PMID: 18039426 DOI: 10.3816/ccc.2007.n.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND This study investigated whether patients with colorectal cancer (CRC) who have tumors with high microsatellite instability (MSI; MSI-H) had an altered expression of the folate and methyl-group metabolism. The gene expression levels of thymidylate synthase (TS), reduced folate carrier (RFC-1), folylpolyglutamate synthase (FPGS), and methylenetetrahydrofolate reductase (MTHFR) in mucosa and tumor were compared with patients with MSS. Furthermore, the influence of TS polymorphisms on TS gene expression levels and MSI-H was studied. PATIENTS AND METHODS The microsatellite status (MSI-H, low instability [MSI-L], or stable [MSS]) and TS polymorphisms were analyzed in genomic DNA from 181 patients with CRC. Gene expression levels of TS, RFC-1, FPGS, and MTHFR in mucosa and tumors were quantified and the difference in TS expression between tumor and mucosa was designated DeltaTS. RESULTS Significantly higher gene expression levels of TS (P < .0001) were detected in patients with CRC with MSI-H compared with MSS/MSI-L tumors. Gene expression of TS and FPGS were significantly higher in right-sided MSI-H tumors compared with right-sided MSS/MSI-L tumors (P < .0001, P = .041, respectively). A significant correlation between DeltaTS and the number of unstable markers was found (P < .0001). An inverse association between age and TS expression was found in MSI tumors (r = -0.57; P = .0004) and also in right-sided tumors (r = -0.25, P = .011) regardless of MSI status. No relation was detected between MSI status and the TS polymorphisms or between the TS polymorphisms and TS expression. CONCLUSION This study has revealed, for the first time, that age and the frequency of unstable MSI markers were factors that were linked to the variability in TS gene expression in tumors.
Collapse
Affiliation(s)
- Elisabeth Odin
- Department of General Surgery, Göteborg University, Sahlgrenska University Hospital/Ostra, Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
17
|
Systemic folate status and risk of colorectal cancer. Cancer Causes Control 2008; 19:1005-7; author reply 1003. [DOI: 10.1007/s10552-008-9161-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2007] [Accepted: 04/08/2008] [Indexed: 11/24/2022]
|
18
|
Abstract
Currently available evidence from epidemiologic, animal, and intervention studies does not unequivocally support the role of folate, a water-soluble B vitamin and important cofactor in one-carbon transfer, in the development and progression of colorectal cancer (CRC). However, when the portfolio of evidence from these studies is analyzed critically, the overall conclusion supports the inverse association between folate status and CRC risk. It is becoming increasingly evident that folate possesses dual modulatory effects on colorectal carcinogenesis depending on the timing and dose of folate intervention. Folate deficiency has an inhibitory effect whereas folate supplementation has a promoting effect on the progression of established colorectal neoplasms. In contrast, folate deficiency in normal colorectal mucosa appears to predispose it to neoplastic transformation, and modest levels of folic acid supplementation suppress, whereas supraphysiologic supplemental doses enhance, the development of cancer in normal colorectal mucosa. Several potential mechanisms relating to the disruption of one-carbon transfer reactions exist to support the dual modulatory role of folate in colorectal carcinogenesis. Based on the lack of compelling supportive evidence and on the potential tumor-promoting effect, routine folic acid supplementation should not be recommended as a chemopreventive measure against CRC at present.
Collapse
Affiliation(s)
- Young-In Kim
- Department of Medicine and Nutritional Sciences, Medical Sciences Building, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada.
| |
Collapse
|
19
|
Brockton NT. Localized depletion: the key to colorectal cancer risk mediated by MTHFR genotype and folate? Cancer Causes Control 2007; 17:1005-16. [PMID: 16933051 DOI: 10.1007/s10552-006-0051-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Accepted: 06/15/2006] [Indexed: 10/24/2022]
Abstract
Dietary folate has been consistently associated with reduced risk of colorectal cancer (CRC). One of the known biochemical roles of folate is donation of methyl moieties. DNA hypomethylation is an early and almost ubiquitous occurrence in tumor tissue. Therefore, it was originally suggested that adequate folate intake contributed to reduced risk of CRC by facilitating methyl-mediated silencing of oncogenes. Methylene tetrahydrofolate reductase (MTHFR) metabolizes 5,10-MTHF (important in DNA synthesis) to 5-MTHF (contributes to downstream methylation reactions by regeneration of methionine from homocysteine). A common polymorphism in the MTHFR gene (C677T) results in a thermolabile phenotype associated with increased homocysteine levels and DNA hypomethylation. Consistent with the folate/methylation hypothesis, it was originally proposed that C677T may increase risk of CRC due to hypomethylation of oncogenes. However, most subsequent studies have reported a reduced risk associated with this polymorphism. This is inconsistent with methylation as the mechanism by which folate and MTHFR genotype mediate CRC risk. The hypothesis presented here proposes that localized folate depletion combined with the effect of the C677T polymorphism on enzyme stability, impacts on the DNA synthesis pathway and accounts for the observed variation in risk associated with genotype and folate status.
Collapse
Affiliation(s)
- N T Brockton
- University of Dundee, Maternal and Child Health Sciences, Ninewells Hospital, Dundee, UK.
| |
Collapse
|
20
|
Ozaki K, Nagasaka T, Notohara K, Kambara T, Takeda M, Sasamoto H, Jass JR, Tanaka N, Matsubara N. Heterogeneous microsatellite instability observed within epithelium of ulcerative colitis. Int J Cancer 2006; 119:2513-9. [PMID: 16929496 DOI: 10.1002/ijc.22095] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Microsatellite instability (MSI) has been associated with colitic cancer. However, reported frequency of MSI was varied and the association of MSI with mismatch repair (MMR) deficiency was unclear. In addition, the occurrence of genetic alterations in stromal cells within ulcerative colitis (UC) is still controversial. We therefore sampled 164 microareas in various pathological lesions of UC with or without colitic cancer and studied the MSI status in relation to the DNA repair protein expressions. A total of 129 microfoci from colorectal tissue of 5 colitic cancer patients and 35 microfoci of 7 UC patients (without neoplasm) were carefully sampled by laser-capture microdissection. MSI was analyzed in each microsamples. The protein expression of MMR genes (MLH1, MSH2, MSH6), O(6)-methylguanine-DNA methyltransferase and p53 were assessed by immunohistochemical analysis. Variety of di-nulcleotide microsatellite markers was altered in individual microfoci from different morphological epithelial lesions, in full range of nonneoplastic epithelium to colitic cancer. Interestingly, MSI was not observed in stromal cells at any sites, including those within colitic cancer/dysplasia lesions. Expression of the MMR proteins was not lost in any of the lesions examined. Microsatellite alterations rather seem to be related to the initiation than to the progression of colitic cancer.
Collapse
Affiliation(s)
- Kazuhide Ozaki
- Department of Gastroenterological Surgery and Surgical Oncology, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer--a survey. Biochim Biophys Acta Rev Cancer 2006; 1775:181-232. [PMID: 17137717 DOI: 10.1016/j.bbcan.2006.10.001] [Citation(s) in RCA: 418] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 10/04/2006] [Accepted: 10/04/2006] [Indexed: 12/23/2022]
Abstract
It has been known for decades that it is possible to detect small amounts of extracellular nucleic acids in plasma and serum of healthy and diseased human beings. The unequivocal proof that part of these circulating nucleic acids (CNAs) is of tumor origin, initiated a surge of studies which confirmed and extended the original observations. In the past few years many experiments showed that tumor-associated alterations can be detected at the DNA and RNA level. At the DNA level the detection of point mutations, microsatellite alterations, chromosomal alterations, i.e. inversion and deletion, and hypermethylation of promoter sequences were demonstrated. At the RNA level the overexpression of tumor-associated genes was shown. These observations laid the foundation for the development of assays for an early detection of cancer as well as for other clinical means.
Collapse
Affiliation(s)
- M Fleischhacker
- Charité, Universitätsmedizin Berlin, Medizinische Klinik mS Onkologie u Hämatologie, CCM, Charitéplatz 1, 10117 Berlin, Germany.
| | | |
Collapse
|
22
|
Itzkowitz SH. Molecular biology of dysplasia and cancer in inflammatory bowel disease. Gastroenterol Clin North Am 2006; 35:553-71. [PMID: 16952740 DOI: 10.1016/j.gtc.2006.07.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) develops from a dysplastic precursor lesion, regardless of whether it arises sporadically, in the setting of high-risk hereditary conditions, or in the context of chronic inflammation like inflammatory bowel disease (IBD). This review focuses on the molecular alterations associated with CRC pathogenesis in IBD. Although none of the molecular alterations to be discussed have yet been integrated into clinical practice, there is potential for molecular diagnostics to enhance the management of patients with long-standing IBD.
Collapse
Affiliation(s)
- Steven H Itzkowitz
- The Dr. Henry D. Janowitz Division of Gastroenterology, Mount Sinai School of Medicine, One Gustave Levy Place, New York, NY 10029, USA.
| |
Collapse
|
23
|
Risques RA, Rabinovitch PS, Brentnall TA. Cancer surveillance in inflammatory bowel disease: new molecular approaches. Curr Opin Gastroenterol 2006; 22:382-90. [PMID: 16760754 DOI: 10.1097/01.mog.0000231812.95525.a7] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Patients with chronic inflammatory bowel disease, such as ulcerative colitis and Crohn's disease, have an increased risk of colorectal cancer. Life-long colonoscopy surveillance is performed to detect the presence of dysplasia, but this approach is expensive and time-consuming. Thus, there is intensive research to identify molecular factors with prognostic value. This review summarizes recent research, with a special emphasis on the mechanisms underlying these molecular alterations. RECENT FINDINGS The role of chromosomal instability in the progression to inflammatory bowel disease-associated colorectal cancer is clear and likely relates to chronic cycles of injury, inflammation, repair and telomere shortening. The role of microsatellite instability has been a subject of discussion, and data suggest that microsatellite instability in inflammatory bowel disease might be different from microsatellite instability in sporadic colorectal cancer. Methylation, as a mechanism of gene silencing, also plays a role in ulcerative colitis tumorigenesis. Chronic inflammation has been linked to p53 activation and oxidative stress, contributing to the extensive genomic DNA damage observed in ulcerative colitis. SUMMARY Improved understanding of the molecular biology of cancer progression in inflammatory bowel disease will hopefully lead to the identification of useful prognostic biomarkers. Efforts are needed to prove the clinical utility of the most promising markers now identified.
Collapse
|
24
|
Michael-Robinson JM, Pandeya N, Walsh MD, Biemer-Huttmann AE, Eri RD, Buttenshaw RL, Lincoln D, Clouston AD, Jass JR, Radford-Smith GL. Characterization of tumour-infiltrating lymphocytes and apoptosis in colitis-associated neoplasia: comparison with sporadic colorectal cancer. J Pathol 2006; 208:381-7. [PMID: 16315333 DOI: 10.1002/path.1895] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The development of colorectal cancer is a major complication for patients with chronic idiopathic colitis. Colitis-associated tumours tend to occur at a younger age and be more aggressive than sporadic colorectal cancers. While we have previously associated the presence of tumour-infiltrating lymphocytes (TILs) and increased apoptosis in sporadic colorectal cancer with high-level microsatellite instability and improved prognosis, little is known of the relationship between these variables in colitis-associated colorectal cancer. The aim of this study was to correlate TILs and tumour cell apoptosis in colitis-associated neoplasms stratified according to microsatellite instability. Twenty tumour and 11 dysplastic samples resected from 21 patients with long-standing colitis were analysed for microsatellite instability at 10 microsatellite markers. TIL distribution (CD3, CD8) and function (granzyme B) were quantified by immunohistochemistry. Neoplastic cell apoptosis was assessed using the M30 CytoDEATH antibody. These findings were compared with 40 microsatellite stable (MSS) sporadic colorectal cancers previously evaluated for TILs and neoplastic apoptosis. Low-level microsatellite instability was found in 1/20 colitis-associated tumours. All other colitis-associated lesions were designated MSS. CD3(+) and CD8(+) TIL counts were significantly higher in colitis-associated lesions compared with MSS sporadic colorectal cancer (p < 0.0001, p = 0.001 respectively). Despite their higher TIL density, colitis-associated tumours were more likely to present late (Dukes' stage C or D) (p = 0.02). Functionally, colitis-associated TILs demonstrated significantly less granzyme B expression compared to sporadic cancers (p = 0.002). The level of tumour cell apoptosis was similar between the two groups (sporadic, 1.53%; colitis cancers, 1.45%). In conclusion, MSS colitis-associated tumours have a higher prevalence of CD3(+)/CD8(+) TILs but no associated increase in tumour cell killing by apoptosis. Unlike cytotoxic T cells in sporadic colorectal cancer, TILs do not appear to enhance the prognosis of colitis-associated colorectal cancer. This may be related to an impairment of granzyme B expression within these lesions.
Collapse
Affiliation(s)
- J M Michael-Robinson
- Crohn's and Colitis Laboratory, Clinical Research Centre, Royal Brisbane and Women's Hospital Research Foundation, Queensland Institute of Medical Research, Brisbane, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Patients with ulcerative colitis are at greater risk of developing colorectal cancer than the general population. Prophylactic colectomy is the only certain method of avoiding this risk. To avoid indiscriminate surgery, efforts have focused on colonoscopic surveillance in order to select for prophylactic surgery only patients at ultra-high risk. However this policy has not always been proven to reduce mortality for colorectal cancer in these patients. Recently, there has been growing interest in chemoprevention using folate. Folate maintains the normal DNA methylation process and steady-state levels of DNA precursors. These properties indicate that folate has potential as chemopreventive agent.
Collapse
Affiliation(s)
- G Biasco
- Universita' di Bologna, Istituto di Ematologia e Oncologia Medica L e A Seràgnoli, Policlinico S. Orsola-Malpighi, Via Massarenti, 9, 40138 Bologna, Italy.
| | | |
Collapse
|
26
|
Wettergren Y, Odin E, Nilsson S, Willen R, Carlsson G, Gustavsson B. Low expression of reduced folate carrier-1 and folylpolyglutamate synthase correlates with lack of a deleted in colorectal carcinoma mRNA splice variant in normal-appearing mucosa of colorectal carcinoma patients. ACTA ACUST UNITED AC 2005; 29:348-55. [PMID: 16122883 DOI: 10.1016/j.cdp.2005.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2005] [Indexed: 11/19/2022]
Abstract
BACKGROUND Cellular folate deficiency leads to DNA strand breaks, mutations, and aberrant methylation and might be a risk factor for colorectal cancer (CRC). The putative tumor suppressor gene deleted in colorectal carcinoma (DCC) is one of several genes the expression of which seems to be affected by the folate concentration at the tissue level. Decreased expression of DCC may be caused by LOH or hypermethylation, i.e. by events that might be linked to folate deficiency. The purpose of this study was to analyze if the folate level and the gene expression levels of reduced folate carrier (RFC-1) and folylpolyglutamate synthase (FPGS) had impact on the expression of DCC splice variants. METHODS Quantification of RFC-1 and FPGS expression in mucosa of 53 CRC patients was performed using real-time PCR whereas DCC splicing variants were detected by automated capillary gel electrophoresis. Total reduced folate concentration was measured with the FdUMP-binding assay (n = 22). RESULTS Significantly higher expression levels of RFC-1 (p = 0.026) and FPGS (p = 0.05) were found in mucosa expressing the splice variant DCC342 compared to mucosa that did not. Furthermore, multivariate analysis showed that RFC-1 and FPGS (r = 0.49, p = 0.01) as well as folate and RFC-1 (r = 0.56, p = 0.023) were correlated only in mucosa expressing DCC342. CONCLUSIONS In conclusion, the present study points to a potential influence of folates in regulating DCC expression at multiple levels involving post-transcriptional pathways. The results may provide a basis for a detailed investigation of molecular mechanisms involved in folate regulation of DCC expression.
Collapse
Affiliation(s)
- Yvonne Wettergren
- Department of General Surgery, Sahlgrenska University Hospital/Ostra, Goteborg University, S-416 85 Goteborg, Sweden.
| | | | | | | | | | | |
Collapse
|
27
|
Potter DD, Murray JA, Donohue JH, Burgart LJ, Nagorney DM, van Heerden JA, Plevak MF, Zinsmeister AR, Thibodeau SN. The role of defective mismatch repair in small bowel adenocarcinoma in celiac disease. Cancer Res 2004; 64:7073-7. [PMID: 15466202 DOI: 10.1158/0008-5472.can-04-1096] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Celiac disease is associated with an increased risk of small bowel adenocarcinoma. The aims of this study were to investigate the molecular basis, assess outcomes, and identify clinicopathologic characteristics of small bowel adenocarcinoma in celiac disease. Retrospective case control cohort study of all celiac disease patients treated at our institution for small bowel adenocarcinoma and matched control patients with sporadic small bowel adenocarcinoma from July 1960 to November 2002. Mismatch repair (MMR) status was accessed by testing tissue for microsatellite instability (MSI) and for hMLH1 and hMSH2 protein expression. Over a 40-year time period, 18 patients with small bowel adenocarcinoma and celiac disease were treated at the Mayo Clinic. One celiac disease patient was excluded. High-frequency MSI (MSI-H) was identified in 8 of 11 (73%) and 2 of 22 (9%) available small bowel adenocarcinoma specimens in the celiac disease and control groups, respectively. In the celiac disease group, MSI-H was associated with loss of hMLH1 and hMSH2 in 6 and 1 specimens, respectively. Loss of hMLH1 occurred in both control tumors. Stage was associated with celiac disease status (P = 0.018), and 78% of controls were stage III or IV compared with 47% of celiac disease patients. Overall, survival was better (P = 0.025) in the celiac disease group compared with stage-matched controls. Celiac disease patients with small bowel adenocarcinoma had a high incidence defective MMR (73%) compared with controls and had better survival compared with stage-matched controls. In addition, celiac disease patients presented more frequently with early-stage small bowel adenocarcinoma. The better survival and earlier presentation of small bowel adenocarcinoma in celiac disease appears to be biologically associated with defective MMR.
Collapse
Affiliation(s)
- D Dean Potter
- Division of Gastroenterologic and General Surgery, Mayo Clinic and Mayo Foundation, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Li GM, Presnell SR, Gu L. Folate deficiency, mismatch repair-dependent apoptosis, and human disease. J Nutr Biochem 2004; 14:568-75. [PMID: 14559107 DOI: 10.1016/s0955-2863(03)00115-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The vitamin that is most commonly deficient in the American diet is folate. Severe folate deficiency in humans is known to cause megaloblastic anemia and developmental defects, and is associated with an increased incidence of several forms of human cancer. Although the exact mechanisms by which this vitamin deficiency may cause these diseases are not known at the present time, recent work has shown that folate deficiency also causes genomic instability and programmed cell death (or apoptosis). Additionally, it is known that the DNA mismatch repair pathway mediates folate deficiency-induced apoptosis. This review will first describe work suggesting that folate deficiency causes genomic instability and apoptosis, then discuss possible mechanisms by which the mismatch repair pathway could trigger folate deficiency-induced apoptosis, which has either protective or destructive effects on tissue.
Collapse
Affiliation(s)
- Guo Min Li
- Department of Pathology and Laboratory Medicine, Markey Cancer Center, University of Kentucky Medical Center, Lexington, KY 40536, USA.
| | | | | |
Collapse
|
29
|
Abstract
The development and dissemination of sophisticated detection technologies have recently exposed the high prevalence of preinvasive colorectal neoplasia in the adult U.S. population. Although cancer screening and surveillance provide opportunities for risk stratification, they achieve risk reduction only when coupled with effective interventions. This review surveys the lead compounds for colorectal cancer prevention and the measures by which they may be prioritized for clinical testing. Clinical trials remain the rate-limiting step in agent development, and novel trial designs are needed to hasten agent identification and testing for cancer prevention. Innovative research models include the nesting of prevention end points within cancer treatment trials and within trials testing promising preventive compounds intended for nononcologic indications.
Collapse
Affiliation(s)
- Ernest T Hawk
- Gastrointentinal and Other Cancers Research Group, Division of Cancer Prevention, National Cancer Institute/NIH, EPN Suite 2141, 6130 Executive Boulevard, Bethesda, MD 20892-7317, USA.
| | | | | |
Collapse
|
30
|
Kim YI. Folate, colorectal carcinogenesis, and DNA methylation: lessons from animal studies. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2004; 44:10-25. [PMID: 15199543 DOI: 10.1002/em.20025] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Folate, a water-soluble B vitamin and cofactor in one-carbon transfer, is an important nutritional factor that may modulate the development of colorectal cancer (CRC). Epidemiologic and clinical studies indicate that dietary folate intake and blood folate levels are inversely associated with CRC risk. Collectively, these studies suggest an approximately 40% reduction in the risk of CRC in individuals with the highest dietary folate intake compared with those with the lowest intake. Animal studies using chemical and genetically predisposed rodent models have provided considerable support for a causal relationship between folate depletion and colorectal carcinogenesis as well as a dose-dependent protective effect of folate supplementation. However, animal studies also have shown that the dose and timing of folate intervention are critical in providing safe and effective chemoprevention; exceptionally high supplemental folate levels and folate intervention after microscopic neoplastic foci are established in the colorectal mucosa promote, rather than suppress, colorectal carcinogenesis. These animal studies, in conjunction with clinical observations, suggest that folate possesses dual modulatory effects on carcinogenesis depending on the timing and dose of folate intervention. Folate deficiency has an inhibitory effect, whereas folate supplementation has a promoting effect on the progression of established neoplasms. In contrast, folate deficiency in normal epithelial tissues appears to predispose them to neoplastic transformation, and modest levels of folate supplementation suppress the development of tumors in normal tissues. Notwithstanding the limitations associated with animal models, these studies suggest that the optimal timing and dose of folate intervention must be established for safe and effective chemoprevention in humans. Folate is an important factor in DNA synthesis, stability, and integrity, the repair aberrations of which have been implicated in colorectal carcinogenesis. Folate may also modulate DNA methylation, which is an important epigenetic determinant in gene expression (an inverse relationship), in the maintenance of DNA integrity and stability, in chromosomal modifications, and in the development of mutations. A mechanistic understanding of how folate status modulates colorectal carcinogenesis further strengthens the case for a causal relationship and provides insight into a possible chemopreventive role of folate.
Collapse
Affiliation(s)
- Young-In Kim
- Departments of Medicine and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
31
|
Courtemanche C, Huang AC, Elson-Schwab I, Kerry N, Ng BY, Ames BN. Folate deficiency and ionizing radiation cause DNA breaks in primary human lymphocytes: a comparison. FASEB J 2003; 18:209-11. [PMID: 14597554 DOI: 10.1096/fj.03-0382fje] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
DNA double-strand breaks, the most serious DNA lesion caused by ionizing radiation, are also caused by several vitamin or mineral deficiencies, such as for folate. Primary human lymphocytes were either irradiated or cultured at different levels of folate deficiency to assess cell proliferation, apoptosis, cell cycle, DNA breaks, and changes in gene expression. Both radiation and folate deficiency decreased cell proliferation and induced DNA breaks, apoptosis, and cell cycle arrest. Levels of folate deficiency commonly found resulted in effects similar to those caused by 1 Gy of radiation, a relatively high dose. Though both radiation and folate deficiency caused DNA breaks, they affected the expression of different genes. Radiation activated excision and DNA double-strand break repair genes and repressed mitochondrially encoded genes. Folate deficiency activated base and nucleotide excision repair genes and repressed folate-related genes. No DNA double-strand break repair gene was activated by folate deficiency. These findings suggest that a diet poor in folate may pose a risk of DNA damage comparable to that of a relatively high dose of radiation. Our results also suggest that research on biological effects of low-dose radiation should take into account the nutritional status of the subjects, because folate deficiency could confound the effects of low-dose radiation.
Collapse
Affiliation(s)
- Chantal Courtemanche
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California, USA
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
Folate, a water-soluble B vitamin and important cofactor in 1-carbon transfer, is an important nutritional factor that may modulate the development of colorectal cancer. Epidemiologic and clinical studies indicate that dietary folate intake and blood folate levels are inversely associated with colorectal cancer risk. Collectively, these studies suggest an approximately 40% reduction in the risk of colorectal cancer in individuals with the highest dietary folate intake compared with those with the lowest intake. Animal studies using chemical and genetically predisposed rodent models have provided considerable support for a causal relationship between folate depletion and colorectal carcinogenesis as well as a dose-dependent protective effect of folate supplementation. However, animal studies have also shown that the dose and timing of folate intervention are critical in providing safe and effective chemoprevention; exceptionally high supplemental folate levels and folate intervention after microscopic neoplastic foci are established in the colorectal mucosa promote rather than suppress colorectal carcinogenesis. These animal studies in conjunction with clinical observations suggest that folate possesses the dual modulatory effects on carcinogenesis depending on the timing and dose of folate intervention. Folate deficiency has an inhibitory effect whereas folate supplementation has a promoting effect on progression of established neoplasms. In contrast, folate deficiency in normal epithelial tissues appears to predispose them to neoplastic transformation, and modest levels of folate supplementation suppress the development of tumors in normal tissues. Notwithstanding the limitations associated with animal models, these animal studies suggest that the optimal timing and dose of folate intervention need to be established for safe and effective chemoprevention in humans.
Collapse
Affiliation(s)
- Young-In Kim
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
33
|
Lamprecht SA, Lipkin M. Chemoprevention of colon cancer by calcium, vitamin D and folate: molecular mechanisms. Nat Rev Cancer 2003; 3:601-14. [PMID: 12894248 DOI: 10.1038/nrc1144] [Citation(s) in RCA: 374] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recent findings have indicated that dietary calcium, vitamin D and folate can modulate and inhibit colon carcinogenesis. Supporting evidence has been obtained from a wide variety of preclinical experimental studies, epidemiological findings and a few human clinical trials. Important molecular events and cellular actions of these micronutrients that contribute to their tumour-modulating effects are discussed. They include a complex series of signalling events that affect the structural and functional organization of colon cells.
Collapse
Affiliation(s)
- Sergio A Lamprecht
- Strang Cancer Prevention Center and Strang Cancer Research Laboratory at The Rockefeller University, 1230 York Avenue, New York, New York 10021, USA.
| | | |
Collapse
|
34
|
Abstract
Major advances in the understanding of the aetio-pathogenesis and genetics of inflammatory bowel disease have been accompanied by an escalation in the sophistication of immunomodulatory inflammatory bowel disease therapeutics. However, the basic 'triple' therapy (5-aminosalicylates, corticosteroids, azathioprine) and nutrition have maintained their central role in the management of patients with inflammatory bowel disease over recent decades. This review provides an overview of the supportive and therapeutic perspectives of nutrition in adult inflammatory bowel disease. The objective of supportive nutrition is to correct malnutrition in terms of calorie intake or specific macro- or micronutrients. Of particular clinical relevance is deficiency in calcium, vitamin D, folate, vitamin B12 and zinc. There is justifiably a growing sense of unease amongst clinicians and patients with regard to the long-term use of corticosteroids in inflammatory bowel disease. This, rather than arguments about efficacy, should be the catalyst for revisiting the use of enteral nutrition as primary treatment in Crohn's disease. Treatment failure is usually related to a failure to comply with enteral nutrition. Potential factors that militate against successful completion of enteral nutrition are feed palatability, inability to stay on a solid-free diet for weeks, social inconvenience and transient feed-related adverse reactions. Actions that can be taken to improve treatment outcome include the provision of good support from dietitians and clinicians for the duration of treatment and the subsequent 'weaning' period. There is evidence to support a gradual return to a normal diet through exclusion-re-introduction or other dietary regimen following the completion of enteral nutrition to increase remission rates. We also review the evidence for emerging therapies, such as glutamine, growth factors and short-chain fatty acids. The future may see the evolution of enteral nutrition into an important therapeutic strategy, and the design of a 'Crohn's disease-specific formulation' that is individually tailored, acceptable to patients, cost-effective, free from adverse side-effects and combines enteral nutrition with novel pre- and pro-biotics and other factors.
Collapse
Affiliation(s)
- J Goh
- Gastrointestinal Unit, University Hospital Birmingham NHS Trust, Queen Elizabeth and Selly Oak Hospitals, UK.
| | | |
Collapse
|
35
|
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease of the colon that can lead to cancer. Patients with UC have approximately 20% lifetime risk of colon cancer, with the risk increasing with the duration of disease. Why some patients develop colorectal cancer and others do not is not clearly understood. Evidence reveals that the UC patients who develop cancer have an underlying process of instability in the colonic mucosa as measured by DNA fingerprinting and fluorescent in situ hybridization techniques. The cause of instability may be shortened telomeres, the protective ends of chromosomes. Patients who progress to dysplasia and cancer have shorter telomeres, which become sticky and cause telomeric bridges to form, with subsequent chromosomal breakage. Thus, UC patients who have dysplasia and cancer appear to have a mutator phenotype present in the colonic mucosa that underlies the process of tumorigenesis.
Collapse
Affiliation(s)
- Teresa A Brentnall
- University of Washington Medical Center, Seattle, Washington 98195, USA.
| |
Collapse
|
36
|
Gu L, Wu J, Qiu L, Jennings CD, Li GM. Involvement of DNA mismatch repair in folate deficiency-induced apoptosis small star, filled. J Nutr Biochem 2002; 13:355-363. [PMID: 12088801 DOI: 10.1016/s0955-2863(02)00178-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Folate is a critical factor for DNA metabolism and its deficiency is associated with a number of human diseases and cancers. Although it has been shown that folate deficiency induces genomic instability and apoptotic cell death, the underlying mechanism is largely unknown. Given the role of mismatch repair in maintaining genomic integrity, mismatch repair was tested for its involvement in folate deficiency-induced genomic instability and cell death. Cells proficient in mismatch repair were highly sensitive to folate deficiency compared with cells defective in either hMutSalpha or hMutLalpha. Since wild-type cells but not mutant cells underwent apoptosis upon extensive folate depletion, the apoptotic response is dependent on a functional mismatch repair system. Our data also indicate that p53 is required for the folate depletion-induced apoptosis. In vitro biochemical studies demonstrated that hMutSalpha specifically recognized DNA damage induced by folate deficiency, suggesting a direct participation of mismatch repair proteins in mediating the apoptotic response. We conclude that while the mismatch repair-dependent apoptosis is necessary to protect damaged cells from tumorigenesis, it may damage a whole tissue or organ, as seen in patients with megaloblastic anemia, during extensive folate deficiency.
Collapse
Affiliation(s)
- Liya Gu
- Department of Pathology and Laboratory Medicine, Markey Cancer Center, University of Kentucky Medical Center, University of Kentucky Medical Center, 40536, Lexington, Kentucky, USA
| | | | | | | | | |
Collapse
|
37
|
Aust DE, Terdiman JP, Willenbucher RF, Chang CG, Molinaro-Clark A, Baretton GB, Loehrs U, Waldman FM. The APC/beta-catenin pathway in ulcerative colitis-related colorectal carcinomas: a mutational analysis. Cancer 2002; 94:1421-7. [PMID: 11920497 DOI: 10.1002/cncr.10334] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Although the APC/beta-catenin pathway is known to play a crucial role in sporadic colorectal carcinogenesis, its influence on ulcerative colitis (UC)-related neoplastic progression is unknown. To elucidate the role of the APC-/beta-catenin pathway in UC-related carcinogenesis, the authors identified APC and beta-catenin mutations in a set of UC-related and sporadic colorectal carcinomas. METHODS The mutational cluster region of APC (codon 1267 to 1529) and exon 3 of the beta-catenin were directly sequenced. RESULTS Only 1 of 30 UC-related tumors (3%) showed an APC mutation whereas 11 of the 42 sporadic carcinomas (26%) had mutations within the mutational cluster region. Within the sporadic carcinoma group, only 8% of the right-sided carcinomas showed APC mutations whereas 50% of the left-sided carcinomas had mutations within the mutational cluster region. None of the tumors in either group showed a beta-catenin mutation. CONCLUSIONS Mutations of the APC and beta-catenin are rare in UC-related tumors. These genes may be altered because of mutations outside the regions studied, or by epigenetic silencing. Alternatively, other proteins involved in the APC/beta-catenin signaling cascade may be altered, or this pathway may be involved infrequently in UC-related carcinogenesis. The significant difference in frequency of APC mutations between right- and left-sided sporadic tumors suggests different molecular pathways in these two tumor sites.
Collapse
Affiliation(s)
- Daniela E Aust
- Pathologisches Institut der Ludwig-Maximilians-Universität, München, Germany
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Fuchs CS. Dietary and Lifestyle Influences on Colorectal Carcinogenesis. COLORECTAL CANCER 2002. [DOI: 10.1007/978-1-59259-160-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
39
|
Ishitsuka T, Kashiwagi H, Konishi F. Microsatellite instability in inflamed and neoplastic epithelium in ulcerative colitis. J Clin Pathol 2001; 54:526-32. [PMID: 11429424 PMCID: PMC1731482 DOI: 10.1136/jcp.54.7.526] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Several genetic alterations have been documented in dysplasia and cancer developing in ulcerative colitis (UC). However, the microsatellite instability (MSI) status has rarely been described, especially in the inflamed epithelium of UC. AIMS To study MSI status during neoplastic and inflammatory changes in UC. METHODS Seventy five surgically resected samples of colorectal mucosa, taken from 16 colectomy specimens of patients with UC were examined: five patients had a long duration with dysplasia or cancer (UC-LD with neoplasm), seven patients had a long duration without neoplastic changes (UC-LD without neoplasm), and four patients had a short duration without neoplastic changes (UC-SD). In addition to MSI status examined by six microsatellite markers, p53 expression was compared among the three groups. RESULTS With regard to non-neoplastic inflamed epithelium, MSI in two or more loci (MSI> or =2) was seen more frequently in the UC-LD without neoplasm group than in the UC-SD group (six of 14 v one of 12; p = 0.060), and significantly more often than in the UC-LD with neoplasm group (six of 14 v two of 23; p = 0.016). In the UC-LD without neoplasm group, MSI> or =2 was detected significantly more frequently in patients with severe inflammation than in those with mild inflammation (six of nine v none of five; p = 0.028). With regard to neoplastic epithelium in the UC-LD with neoplasm group, MSI in two or more loci was found in three of 17, and p53 overexpression was seen in 11 of 17 of the neoplastic lesions. CONCLUSIONS A high incidence of MSI in long standing UC with severe inflammation probably reflects genomic instability caused by repeated inflammatory stress. Thus, the influence of inflammation should be considered when estimating MSI in UC. It is possible that changes in p53 expression are important in the development of cancer in UC.
Collapse
Affiliation(s)
- T Ishitsuka
- Department of Surgery, Jichi Medical School, 3311-1, Yakushiji, Minami-Kawachi, Kawachi, Tochigi, Japan.
| | | | | |
Collapse
|
40
|
Mason JB, Choi SW. Folate and carcinogenesis: developing a unifying hypothesis. ADVANCES IN ENZYME REGULATION 2000; 40:127-41. [PMID: 10828349 DOI: 10.1016/s0065-2571(99)00037-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- J B Mason
- Vitamin and Carcinogenesis Program, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| | | |
Collapse
|
41
|
Abstract
Collectively, the evidence from epidemiologic, animal and human studies strongly suggests that folate status modulates the risk of developing cancers in selected tissues, the most notable of which is the colorectum. Folate depletion appears to enhance carcinogenesis whereas folate supplementation above what is presently considered to be the basal requirement appears to convey a protective effect. The means by which this modulation of cancer risk is mediated is not known with certainty, but there are several plausible mechanisms which have been described. Folate plays a major role in the formation of S-adenosylmethionine, the universal methyl donor, as well as in the formation of purine and thymidine synthesis for DNA and RNA. Therefore, most mechanistic studies performed to date have focused on alterations in DNA methylation, disruption of DNA integrity and disruption of DNA repair, all of which have been observed with folate depletion. These aberrations in DNA are believed to enhance carcinogenesis by altering the expression of critical tumor suppressor genes and proto-oncogenes. Recently, the role of a common polymorphism of the methylenetetrahydrofolate reductase gene has been highlighted as well. This review presents those mechanisms which are the most likely candidates to explain folate's effects and it proposes an integrated scheme to explain how these mechanisms might interact.
Collapse
Affiliation(s)
- S W Choi
- Vitamin and Carcinogenesis Program, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA
| | | |
Collapse
|
42
|
Umetani N, Sasaki S, Watanabe T, Shinozaki M, Matsuda K, Ishigami H, Ueda E, Muto T. Genetic alterations in ulcerative colitis-associated neoplasia focusing on APC, K-ras gene and microsatellite instability. Jpn J Cancer Res 1999; 90:1081-7. [PMID: 10595736 PMCID: PMC5925993 DOI: 10.1111/j.1349-7006.1999.tb00681.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The status of genetic alterations in ulcerative colitis (UC)-associated neoplasia (UCAN) was investigated focusing on microsatellite instability (MSI) which is seen in a certain fraction of colorectal carcinomas, and adenomatous polyposis coli (APC) gene and K-ras gene, in which mutations occur in the early stage of sporadic colorectal tumorigenesis. Thirty-one UCAN from 15 UC patients who had undergone colorectal resection at our institution were investigated. There were 8 lesions of invasive carcinoma, 15 high-grade dysplasia (HGD) and 8 low-grade dysplasia (LGD). DNA was extracted from each neoplastic lesion and corresponding non-neoplastic tissue by a microdissection method. MSI status at 9 microsatellite loci, loss of heterozygosity (LOH) at the APC locus, and K-ras codon 12 point mutation were examined. As for MSI, 4/31 (13%) UCAN (carcinoma: 1/8 (13%), HGD: 2/15 (13%), LGD: 1/8 (13%)) were MSI-high (3 or more unstable loci) and 12/31 (39%) UCAN (carcinoma: 3/8 (38%), HGD: 6/15 (40%), LGD: 3/8 (38%)) were MSI-low (1 or 2 unstable loci). LOH at the APC locus was not found in 9 UCAN from 6 informative (heterozygous) cases. The K-ras mutation rate of UCAN was 3/31 (9.7%) (carcinoma: 2/8 (25%), HGD: 1/15 (7%) and LGD: 0/8). MSI is relatively common in UCAN and is present at the early stage of tumorigenesis of UCAN, while the involvement of genetic alterations of the APC gene and K-ras gene is small. MSI may be one of the mechanisms of the increased neoplastic risk in UC, and UCAN may develop through a different carcinogenic pathway from sporadic carcinomas.
Collapse
Affiliation(s)
- N Umetani
- Department of Surgical Oncology, School of Medicine, The University of Tokyo.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Loeb KR, Loeb LA. Genetic instability and the mutator phenotype. Studies in ulcerative colitis. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 154:1621-6. [PMID: 10362784 PMCID: PMC1866616 DOI: 10.1016/s0002-9440(10)65415-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Keith R. Loeb
- University of Washington School of Medicine and the Fred Hutchinson Cancer Research Center,†
| | - Lawrence A. Loeb
- University of Washington School of Medicine and the Fred Hutchinson Cancer Research Center,†
| |
Collapse
|