Epigenetic Repression of DNA Mismatch Repair by Inflammation and Hypoxia in Inflammatory Bowel Disease–Associated Colorectal Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Epigenetic Repression of DNA Mismatch Repair by Inflammation and Hypoxia in Inflammatory Bowel Disease–Associated Colorectal Cancer Published OnlineFirst July 28, 2009; DOI: 10.1158/0008-5472.CAN-09-1285 Cell, Tumor, and Stem Cell Biology Epigenetic Repression of DNA Mismatch Repair by Inflammation and Hypoxia in Inflammatory Bowel Disease–Associated Colorectal Cancer Robert A. Edwards,1 Mavee Witherspoon,2 Kehui Wang,1 Kambiz Afrasiabi,2 Trang Pham,2 Lutz Birnbaumer,3 and Steven M. Lipkin2 Departments of 1Pathology and 2Medicine, University of California-Irvine, Irvine, California; and 3Laboratory of Neurobiology/Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina Abstract increased likelihood of right-sided colon cancer location, non- Sporadic human mismatch repair (MMR)–deficient colorectal polyposis pathology, mucinous histology, and presence of a cancers account for f12.5% of all cases of colorectal cancer. Crohn’s-like lymphocytic infiltrate in tumors (2, 3). MMR-deficient colorectal cancers are classically characterized MMR-deficient colorectal cancers arise through three distinct by right-sided location, multifocality, mucinous histology, and genetic mechanisms. First, monoallelic germ-line MMR mutation lymphocytic infiltration. However, tumors in germ-line MMR- and somatic loss of the second MMR allele causes hereditary deficient mouse models lack these histopathologic features. nonpolyposis colon cancer, also known as Lynch syndrome. Lynch Mice lackingthe heterotrimeric G protein A subunit GiA2 syndrome carriers body tissues are MMR-proficient because of the develop chronic colitis and multifocal, right-sided cancers other allele. As predicted by the Knudson hypothesis, the second with mucinous histopathology, similar to human MMR- MMR gene allele is somatically lost in some cells, which then À À deficient colorectal cancer. YoungGiA 2 / colonic epithelium become MMR-deficient and transform into colorectal cancer and has normal MMR expression but selectively loses MLH1 and other cancers. The second mechanism is biallelic germ-line consequently PMS2 expression followinginflammation. constitutive MMR gene mutation, where all body tissues are À À GiA2 / cancers have microsatellite instability. Mlh1 is MMR-deficient. These patients have a different clinical phenotype, epigenetically silenced not by promoter hypermethylation with very early-onset hematologic, brain, and small intestinal but by decreased histone acetylation. Chronically inflamed cancers (4–6). Although colorectal cancers can develop, penetrance À À GiA2 / colonic mucosa contains patchy hypoxia, with is low and arise more commonly in the left colon (4, 5). increased crypt expression of the hypoxia markers DEC-1 The third genetic mechanism, de novo sporadic MMR-deficient f and BNIP3. Chromatin immunoprecipitation identified in- colorectal cancers, accounts for 12.5% of colorectal cancer (6). Both creased bindingof the transcriptional repressor DEC-1 to the MMR alleles are somatically mutated or epigenetically inactivated in proximal Mlh1 promoter in hypoxic YAMC cells and colitic some cells, which transform into cancer. Mlh1 promoter hyper- À À À À GiA2 / crypts. TreatingGi A2 / mice with the histone methylation is the most common cause of sporadic MMR-deficient deacetylase inhibitor suberoylanilide hydroxamic acid signif- colorectal cancer (7–9). In vitro studies have shown that epigenetic icantly decreased colitis activity and rescued MLH1 expression Mlh1 inactivation can also occur via hypoxia-induced transcriptional in crypt epithelial cells, which was associated with increased repression of the proximal Mlh1 promoter by histone deacetylases acetyl histone H3 levels and decreased DEC-1 bindingat the (HDAC; ref. 9). However, whether this mechanism is relevant to proximal Mlh1 promoter, consistent with a histone deacety- in vivo colorectal cancer tumorigenesis is unknown. lase–dependent mechanism. These data link chronic hypoxic Multiple biallelic MMR germ-line knockout mouse models have inflammation, epigenetic MMR protein down-regulation, been created (Supplementary Table S1). These mutations primarily development of MMR-deficient colorectal cancer, and the cause lymphomas and small intestine adenomas (10–16) Only first mouse model of somatically acquired MMR-deficient rarely do these models develop colorectal cancer, and mucinous, colorectal cancer. [Cancer Res 2009;69(16):6423–9] nonpolyposis, or proximal colon colorectal cancers essentially never arise. These models are therefore more similar to human biallelic germ-line constitutive MMR deficiency than somatically Introduction acquired MMR deficiency syndromes. Gia2 is a heterotrimeric G protein a subunit, the deletion of DNA mismatch repair (MMR)–deficient colorectal cancers have which in mice leads to spontaneous colitis and nonpolyposis, distinct features that distinguish them from other colorectal cancer right-sided, multifocal colorectal cancers with mucinous histology types (1). These include increased frameshift mutation rates and a Crohn’s-like inflammatory infiltrate (17–19). The similarities [commonly called microsatellite instability (MSI)], multifocality, À À between Gia2 / mouse colorectal cancer and somatically acquired human MMR-deficient colorectal cancer (Supplementary Note: Supplementary data for this article are available at Cancer Research Online Table S1) led us to investigate whether acquired MMR silencing (http://cancerres.aacrjournals.org/). might contribute as a mechanism of colorectal cancer tumorigen- Requests for reprints: Robert A. Edwards, Department of Pathology, University of a À/À California-Irvine, D449 Med Sci I, Irvine, CA 92697-4800. Phone: 949-824-8576; Fax: 949- esis in Gi 2 mice. We found that MLH1 and PMS2 protein levels 824-2160; E-mail: [email protected] and Steven M. Lipkin, Department of Genetic in crypts are initially normal but are suppressed following the onset Medicine, Weill Cornell Medical College, Room 718 HSS, New York, NY 10021. of hypoxic colitis. Hypoxia induces colonic expression of the bHLH Phone: 212-774-7160; E-mail: [email protected]. I2009 American Association for Cancer Research. transcriptional repressor DEC-1 and enhances its binding to the doi:10.1158/0008-5472.CAN-09-1285 proximal Mlh1 promoter, thereby repressing MLH1 expression and www.aacrjournals.org 6423 Cancer Res 2009; 69: (16). August 15, 2009 Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2009 American Association for Cancer Research. Published OnlineFirst July 28, 2009; DOI: 10.1158/0008-5472.CAN-09-1285 Cancer Research À À leading to MSI. Treatment of Gia2 / mice with HDAC inhibitor forward 5¶-GTTTTACTCCATTCGGAAGCAGT-3¶, murine MLH1 reverse (HDACi) decreases colitis and relieves epigenetic repression of 5¶-TGTGAGCGGAAGGCTTTATAGAT-3¶,murinePMS2forward ¶ ¶ ¶ MLH1 expression. In summary, we describe the first mouse model 5 -ACCTCACGCAGGTTGACTT-3 , murine PMS2 reverse 5 -AGTCGAGTCC- ¶ ¶ of somatically acquired MMR deficiency and show that hypoxia- CAACGCTTG -3 , murine BNIP3 forward 5 -CGAAGCGCACAGCTACTCTC- 3¶, murine BNIP3 reverse 5¶-GCCTTCCAATGTAGATCCCCAAG-3¶, murine induced Mlh1 silencing may be another important epigenetic DEC-1 forward 5¶-TACAAGCTGGTGATTTGTCGG-3¶, and murine DEC-1 mechanism contributing to the development of MMR-deficient reverse 5¶-CTGGGAAGATTTCAGGTCCCG-3¶. Normalization was done colorectal cancer. using murine RPA2 (forward 5¶-ATACCCAGACAACAGAGGG-3¶ and reverse 5¶-GCCAGTCCGCTCAATCACC-3¶). In vitro hypoxia treatment. Subconfluent cultures of YAMC cells Materials and Methods (a kind gift of Dr. Robert Whitehead) were grown in RPMI with 5% fetal À À Mice. Wild-type (WT) and Gia2 / mice on a cross-bred 129Sv/C57BL6 bovine serum, 100 ng/AL IFN-g, and 1Â insulin/transferrin/selenium and j background were genotyped as described previously (19). All experiments incubated in a Biospherix hypoxia chamber at 37 Cin1%O2 for 24 h and were approved by the Institutional Animal Care and Use Committee at total RNA was analyzed by quantitative real-time PCR. À À University of California-Irvine. Suberoylanilide hydroxamic acid treatment of GiA2 / mice. À À In vivo assessment of apoptosis induction. Age-matched WT and Groups of 32- to 36-week-old Gia2 / mice (n = 4) were treated with À À Gia2 / mice were injected i.p. with 20 mg/kg 1,2-dimethylhydrazine for daily gavage feedings with a 5 mg/mL solution of suberoylanilide 24 h. Sections (5 Am) of fixed control and 1,2-dimethylhydrazine–treated hydroxamic acid (SAHA; Cayman Chemical) solubilized in PBS containing colons were stained for apoptotic cells using an Apoptag staining kit a 5 molar excess of h-hydroxypropyl cyclodextrin (Sigma) for 7 days (final (Chemicon). The data are expressed as the mean F SE of the normalized dose 50 mg/kg). Control animals received only cyclodextrin solution. Colons ratio of terminal deoxynucleotidyl transferase–mediated dUTP nick end were harvested and crypts isolated as described above and analyzed for labeling–positive/propidium iodide–positive cells. MLH1 expression by Western blot analysis. Colonic crypt isolation, SDS-PAGE, and Western blotting. Rinsed Chromatin immunoprecipitation. Cells and crypts were cross-linked small intestine and colon segments were reverted on a Labconco vibratory in 1% formaldehyde for 10 min and then lysed in SDS lysis buffer and stirrer and shaken in 30 mmol/L EDTA in HBSS. Crypts liberated between sonicated to a fragment length of 300 to 1,000 bp. The sonicate was 20 and 40 min (>98% pure by cytospin analysis) were used for Western precleared and the supernatant was incubated overnight at 4jC
Recommended publications
  • Paul Modrich Howard Hughes Medical Institute and Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, USA
    Mechanisms in E. coli and Human Mismatch Repair Nobel Lecture, December 8, 2015 by Paul Modrich Howard Hughes Medical Institute and Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, USA. he idea that mismatched base pairs occur in cells and that such lesions trig- T ger their own repair was suggested 50 years ago by Robin Holliday in the context of genetic recombination [1]. Breakage and rejoining of DNA helices was known to occur during this process [2], with precision of rejoining attributed to formation of a heteroduplex joint, a region of helix where the two strands are derived from the diferent recombining partners. Holliday pointed out that if this heteroduplex region should span a genetic diference between the two DNAs, then it will contain one or more mismatched base pairs. He invoked processing of such mismatches to explain the recombination-associated phenomenon of gene conversion [1], noting that “If there are enzymes which can repair points of damage in DNA, it would seem possible that the same enzymes could recognize the abnormality of base pairing, and by exchange reactions rectify this.” Direct evidence that mismatches provoke a repair reaction was provided by bacterial transformation experiments [3–5], and our interest in this efect was prompted by the Escherichia coli (E. coli) work done in Matt Meselson’s lab at Harvard. Using artifcially constructed heteroduplex DNAs containing multiple mismatched base pairs, Wagner and Meselson [6] demonstrated that mismatches elicit a repair reaction upon introduction into the E. coli cell. Tey also showed that closely spaced mismatches, mismatches separated by a 1000 base pairs or so, are usually repaired on the same DNA strand.
    [Show full text]
  • A Dissertation Entitled the Role of Base Excision Repair And
    A Dissertation Entitled The Role of Base Excision Repair and Mismatch Repair Proteins in the Processing of Cisplatin Interstrand Cross-Links. by Akshada Sawant Submitted to the Graduate Faculty as partial fulfillment of the requirements for the Doctor of Philosophy Degree in Biomedical Science Dr. Stephan M. Patrick, Committee Chair Dr. Kandace Williams, Committee Member Dr. William Maltese, Committee Member Dr. Manohar Ratnam, Committee Member Dr. David Giovannucci, Committee Member Dr. Patricia R. Komuniecki, Dean College of Graduate Studies The University of Toledo August 2014 Copyright 2014, Akshada Sawant This document is copyrighted material. Under copyright law, no parts of this document may be reproduced without the expressed permission of the author. An Abstract of The Role of Base Excision Repair and Mismatch Repair Proteins in the Processing of Cisplatin Interstrand Cross-Links By Akshada Sawant Submitted to the Graduate Faculty as partial fulfillment of the requirements for the Doctor of Philosophy Degree in Biomedical Science The University of Toledo August 2014 Cisplatin is a well-known anticancer agent that forms a part of many combination chemotherapeutic treatments used against a variety of human cancers. Despite successful treatment, the development of resistance is the major limitation of the cisplatin based therapy. Base excision repair modulates cisplatin cytotoxicity. Moreover, mismatch repair deficiency gives rise to cisplatin resistance and leads to poor prognosis of the disease. Various models have been proposed to explain this low level of resistance caused due to loss of MMR proteins. In our previous studies, we have shown that BER processing of the cisplatin ICLs is mutagenic. Our studies showed that these mismatches lead to the activation and the recruitment of mismatch repair proteins.
    [Show full text]
  • DNA Repair with Its Consequences (E.G
    Cell Science at a Glance 515 DNA repair with its consequences (e.g. tolerance and pathways each require a number of apoptosis) as well as direct correction of proteins. By contrast, O-alkylated bases, Oliver Fleck* and Olaf Nielsen* the damage by DNA repair mechanisms, such as O6-methylguanine can be Department of Genetics, Institute of Molecular which may require activation of repaired by the action of a single protein, Biology, University of Copenhagen, Øster checkpoint pathways. There are various O6-methylguanine-DNA Farimagsgade 2A, DK-1353 Copenhagen K, Denmark forms of DNA damage, such as base methyltransferase (MGMT). MGMT *Authors for correspondence (e-mail: modifications, strand breaks, crosslinks removes the alkyl group in a suicide fl[email protected]; [email protected]) and mismatches. There are also reaction by transfer to one of its cysteine numerous DNA repair pathways. Each residues. Photolyases are able to split Journal of Cell Science 117, 515-517 repair pathway is directed to specific Published by The Company of Biologists 2004 covalent bonds of pyrimidine dimers doi:10.1242/jcs.00952 types of damage, and a given type of produced by UV radiation. They bind to damage can be targeted by several a UV lesion in a light-independent Organisms are permanently exposed to pathways. Major DNA repair pathways process, but require light (350-450 nm) endogenous and exogenous agents that are mismatch repair (MMR), nucleotide as an energy source for repair. Another damage DNA. If not repaired, such excision repair (NER), base excision NER-independent pathway that can damage can result in mutations, diseases repair (BER), homologous recombi- remove UV-induced damage, UVER, is and cell death.
    [Show full text]
  • Epigenetic Regulation of DNA Repair Genes and Implications for Tumor Therapy ⁎ ⁎ Markus Christmann , Bernd Kaina
    Mutation Research-Reviews in Mutation Research xxx (xxxx) xxx–xxx Contents lists available at ScienceDirect Mutation Research-Reviews in Mutation Research journal homepage: www.elsevier.com/locate/mutrev Review Epigenetic regulation of DNA repair genes and implications for tumor therapy ⁎ ⁎ Markus Christmann , Bernd Kaina Department of Toxicology, University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany ARTICLE INFO ABSTRACT Keywords: DNA repair represents the first barrier against genotoxic stress causing metabolic changes, inflammation and DNA repair cancer. Besides its role in preventing cancer, DNA repair needs also to be considered during cancer treatment Genotoxic stress with radiation and DNA damaging drugs as it impacts therapy outcome. The DNA repair capacity is mainly Epigenetic silencing governed by the expression level of repair genes. Alterations in the expression of repair genes can occur due to tumor formation mutations in their coding or promoter region, changes in the expression of transcription factors activating or Cancer therapy repressing these genes, and/or epigenetic factors changing histone modifications and CpG promoter methylation MGMT Promoter methylation or demethylation levels. In this review we provide an overview on the epigenetic regulation of DNA repair genes. GADD45 We summarize the mechanisms underlying CpG methylation and demethylation, with de novo methyl- TET transferases and DNA repair involved in gain and loss of CpG methylation, respectively. We discuss the role of p53 components of the DNA damage response, p53, PARP-1 and GADD45a on the regulation of the DNA (cytosine-5)- methyltransferase DNMT1, the key enzyme responsible for gene silencing. We stress the relevance of epigenetic silencing of DNA repair genes for tumor formation and tumor therapy.
    [Show full text]
  • The Diagnostic Value of DNA Repair Gene in Breast Cancer Recurrence and Metastasis
    The Diagnostic Value of DNA Repair Gene in Breast Cancer Recurrence and Metastasis Yongxin Yang Southwest Medical University Xiabin Li Southwest Medical University Liyue Hao Southwest Medical University Deyong Jiang Centers for Disease Control and Prevention Bin Wu Southwest Medical University Tao He Southwest Medical University Yan Tang ( [email protected] ) Research Keywords: PARP1, XRCC4, ERCC1, Breast cancer Posted Date: June 25th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-36932/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/19 Abstract Background: DNA repair genes play a vital role in the treatment of many cancers, and DNA repair genes can be used in breast cancer recurrence and metastasis research. We found that the expression of DNA repair genes in breast cancer patients after recurrence and metastasis is abnormal, however, the clinical predictive signicance of DNA repair genes is still elusive. Methods: The nested case-control method was used in patients with breast cancer recurrence and metastasis after surgery (n=109) and patients without recurrence and metastasis after surgery (n=109). The proteins and mRNA of DNA repair genes were detected by immunohistochemistry and Real-time PCR respectively. Results: PARP1(OR=1.485, 95%CI:1.279~1.725, P<0.05), XRCC4(OR= 1.419, 95%CI:1.217~ 1.656, P<0.05) and ERCC1 (OR=1.181, 95%CI: 1.032~1.353, P<0.05) were risk factors for postoperative recurrence and metastasis of breast cancer. Therefore, we used the ROC
    [Show full text]
  • WRN Promoter Methylation Possibly Connects Mucinous Differentiation, Microsatellite Instability and Cpg Island Methylator Phenotype in Colorectal Cancer
    Modern Pathology (2008) 21, 150–158 & 2008 USCAP, Inc All rights reserved 0893-3952/08 $30.00 www.modernpathology.org WRN promoter methylation possibly connects mucinous differentiation, microsatellite instability and CpG island methylator phenotype in colorectal cancer Takako Kawasaki1,2, Mutsuko Ohnishi2, Yuko Suemoto2, Gregory J Kirkner3, Zhiqian Liu2, Hiroyuki Yamamoto4, Massimo Loda1,2, Charles S Fuchs2,3 and Shuji Ogino1,2 1Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA; 2Department of Medical Oncology, Dana–Farber Cancer Institute, Boston, MA, USA; 3Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA and 4First Department of Internal Medicine, Sapporo Medical University, Sapporo, Japan Werner syndrome is a premature aging syndrome characterized by early onset of cancer and abnormal cellular metabolism of glycosaminoglycan. The WRN helicase plays an important role in the maintenance of telomere function. WRN promoter methylation and gene silencing are common in colorectal cancer with the CpG island methylator phenotype (CIMP), which is associated with microsatellite instability (MSI) and mucinous tumors. However, no study has examined the relationship between mucinous differentiation, WRN methylation, CIMP and MSI in colorectal cancer. Utilizing 903 population-based colorectal cancers and real-time PCR (MethyLight), we quantified DNA methylation in WRN and eight other promoters (CACNA1G, CDKN2A, CRABP1, IGF2, MLH1, NEUROG1, RUNX3 and SOCS1) known to be specific for CIMP. Supporting WRN as a good CIMP marker, WRN methylation was correlated well with CIMP-high diagnosis (Z6/8 methylated promoters), demonstrating 89% sensitivity and 81% specificity. WRN methylation was associated with the presence of any mucinous component and Z50% mucinous component (Po0.0001).
    [Show full text]
  • Clinical Significance of ERCC2, XPC, ERCC5 and XRCC3 Gene Polymorphisms in Diffuse Large B Cell Lymphoma
    DOI: 10.14744/ejmi.2020.56831 EJMI 2020;4(3):332–340 Research Article Clinical Significance of ERCC2, XPC, ERCC5 and XRCC3 Gene Polymorphisms in Diffuse Large B Cell Lymphoma Aykut Bahceci,1 Semra Paydas,2 Melek Ergin,3 Gulsah Seydaoglu,4 Gulsum Ucar5 1Department of Medical Oncology, Dr. Ersin Arslan Training and Research Hospital, Gaziantep, Turkey 2Department of Medical Oncology, Cukurova University Faculty of Medicine, Adana, Turkey 3Department of Patology, Cukurova University Faculty of Medicine, Adana, Turkey 4Department of Biostatistics, Cukurova University Faculty of Medicine, Adana, Turkey 5Department of Pediatric Hematology, Cukurova University Faculty of Medicine, Adana, Turkey Abstract Objectives: DNA repair genes protects the genome from DNA damage both of endogenous and exogenous stress fac- tors. Due to DNA repair gene polymorphisms, there are differences in the repair capacity between several cancer types. The aim of this study is to evaluate the association between some of the DNA repair gene polymorphisms and clinical outcome in Diffuse Large B-Cell Lymphoma (DLBCL). Methods: The association between clinical factors including stage at diagnosis, extra-nodal involvement, tumor bur- den, bone marrow involvement, relapse status, disease-free/overall survival times and DNA repair gene polymorphisms including ERCC2 (Lys751Gln), XPC (Gln939Lys), ERCC5 (Asp1104His) and XRCC3 (Thr241Met) in 58 patients with DLBCL. T-Shift Real-Time PCR was used to detect these mutations. Results: The median survival times were 60 months and 109 months in patients with CC genotype and CA/AA geno- type of XPC gene polymorphism, respectively (p=0.017). More interestingly, median survival times were 9 months and 109 months in patients with CC (XPC)/CC (XRCC3) and CA/AA (XPC)/CT/TT (XRCC3) for both XPC and XRCC3 gene polymorphisms, respectively (p=0.004).
    [Show full text]
  • Predisposition to Hematologic Malignancies in Patients With
    LETTERS TO THE EDITOR carcinomas but no internal cancer by the age of 29 years Predisposition to hematologic malignancies in and 9 years, respectively. patients with xeroderma pigmentosum Case XP540BE . This patient had a highly unusual pres - entation of MPAL. She was diagnosed with XP at the age Germline predisposition is a contributing etiology of of 18 months with numerous lentigines on sun-exposed hematologic malignancies, especially in children and skin, when her family emigrated from Morocco to the young adults. Germline predisposition in myeloid neo - USA. The homozygous North African XPC founder muta - plasms was added to the World Health Organization tion was present. 10 She had her first skin cancer at the age 1 2016 classification, and current management recommen - of 8 years, and subsequently developed more than 40 cuta - dations emphasize the importance of screening appropri - neous basal and squamous cell carcinomas, one melanoma 2 ate patients. Rare syndromes of DNA repair defects can in situ , and one ocular surface squamous neoplasm. She 3 lead to myeloid and/or lymphoid neoplasms. Here, we was diagnosed with a multinodular goiter at the age of 9 describe our experience with hematologic neoplasms in years eight months, with several complex nodules leading the defective DNA repair syndrome, xeroderma pigmen - to removal of her thyroid gland. Histopathology showed tosum (XP), including myelodysplastic syndrome (MDS), multinodular adenomatous/papillary hyperplasia. At the secondary acute myeloid leukemia (AML), high-grade age of 19 years, she presented with night sweats, fatigue, lymphoma, and an extremely unusual presentation of and lymphadenopathy. Laboratory studies revealed pancy - mixed phenotype acute leukemia (MPAL) with B, T and topenia with hemoglobin 6.8 g/dL, platelet count myeloid blasts.
    [Show full text]
  • S Na P S H O T: D N a Mism a Tc H R E P a Ir
    SnapShot: Repair DNA Mismatch Scott A. Lujan, and Thomas Kunkel A. Larrea, Andres Park, NC 27709, USA Triangle Health Sciences, NIH, DHHS, Research National Institutes of Environmental 730 Cell 141, May 14, 2010 ©2010 Elsevier Inc. DOI 10.1016/j.cell.2010.05.002 See online version for legend and references. SnapShot: DNA Mismatch Repair Andres A. Larrea, Scott A. Lujan, and Thomas A. Kunkel National Institutes of Environmental Health Sciences, NIH, DHHS, Research Triangle Park, NC 27709, USA Mismatch Repair in Bacteria and Eukaryotes Mismatch repair in the bacterium Escherichia coli is initiated when a homodimer of MutS binds as an asymmetric clamp to DNA containing a variety of base-base and insertion-deletion mismatches. The MutL homodimer then couples MutS recognition to the signal that distinguishes between the template and nascent DNA strands. In E. coli, the lack of adenine methylation, catalyzed by the DNA adenine methyltransferase (Dam) in newly synthesized GATC sequences, allows E. coli MutH to cleave the nascent strand. The resulting nick is used for mismatch removal involving the UvrD helicase, single-strand DNA-binding protein (SSB), and excision by single-stranded DNA exonucleases from either direction, depending upon the polarity of the nick relative to the mismatch. DNA polymerase III correctly resynthesizes DNA and ligase completes repair. In bacteria lacking Dam/MutH, as in eukaryotes, the signal for strand discrimination is uncertain but may be the DNA ends associated with replication forks. In these bacteria, MutL harbors a nick-dependent endonuclease that creates a nick that can be used for mismatch excision. Eukaryotic mismatch repair is similar, although it involves several dif- ferent MutS and MutL homologs: MutSα (MSH2/MSH6) recognizes single base-base mismatches and 1–2 base insertion/deletions; MutSβ (MSH2/MSH3) recognizes insertion/ deletion mismatches containing two or more extra bases.
    [Show full text]
  • Involvement of Nucleotide Excision and Mismatch Repair Mechanisms in Double Strand Break Repair
    250 Current Genomics, 2009, 10, 250-258 Involvement of Nucleotide Excision and Mismatch Repair Mechanisms in Double Strand Break Repair Ye Zhang1,2,*, Larry H. Rohde2 and Honglu Wu1 1NASA Johnson Space Center, Houston, Texas 77058 and 2University of Houston at Clear Lake, Houston, Texas 77058, USA Abstract: Living organisms are constantly threatened by environmental DNA-damaging agents, including UV and ioniz- ing radiation (IR). Repair of various forms of DNA damage caused by IR is normally thought to follow lesion-specific re- pair pathways with distinct enzymatic machinery. DNA double strand break is one of the most serious kinds of damage induced by IR, which is repaired through double strand break (DSB) repair mechanisms, including homologous recombi- nation (HR) and non-homologous end joining (NHEJ). However, recent studies have presented increasing evidence that various DNA repair pathways are not separated, but well interlinked. It has been suggested that non-DSB repair mecha- nisms, such as Nucleotide Excision Repair (NER), Mismatch Repair (MMR) and cell cycle regulation, are highly involved in DSB repairs. These findings revealed previously unrecognized roles of various non-DSB repair genes and indicated that a successful DSB repair requires both DSB repair mechanisms and non-DSB repair systems. One of our recent studies found that suppressed expression of non-DSB repair genes, such as XPA, RPA and MLH1, influenced the yield of IR- induced micronuclei formation and/or chromosome aberrations, suggesting that these genes are highly involved in DSB repair and DSB-related cell cycle arrest, which reveals new roles for these gene products in the DNA repair network.
    [Show full text]
  • Male Factor Infertility and Health Karen Baker, MD Associate Professor Duke University, Division of Urology Fertility And…
    Male Factor Infertility and Health Karen Baker, MD Associate Professor Duke University, Division of Urology Fertility and… • Cancer • Goals: • Heart disease • Review literature linking MFI to poor health • Metabolic syndrome • Diabetes • Discuss possible mechanisms common to MFI and cancer • Early death Cardiovascular risk • 3.5 million AARP members surveyed 1995-1996 • 137,903 men met criteria • 92% +paternity • mean age 62.7 yrs • 96.4% white • Age adjusted cardiovascular mortality risk 2.7/1000 person- years • Childless men 17%↑ cardiovascular mortality compared with men with + paternity Eisenberg, Hum Repro 2011 Cardiovascular risk • 3.5 million AARP members surveyed 1995-1996 • 137,903 men met criteria • 92% +paternity • mean age 62.7 yrs • 96.4% white • Age adjusted cardiovascular mortality risk 2.7/1000 person- years • Childless men 17%↑ cardiovascular mortality compared with men with + paternity Eisenberg, Hum Repro 2011 Chronic disease • Insurance claims 2001 - 2009 • 13,027 infertile men • Semen testing + MFI dx • 23,860 fertile men • Semen testing no MFI dx • 79,099 vasectomy • Age • Smoking by ICD9 • Obesity • Compared rates of common medical conditions • HTN, DM, peripheral vascular disease, cerebrovascular disease, ischemic heart by ICD9 disease, alcohol abuse, bipolar, etc. Chronic disease • ↑ medical disease in MFI vs Risk of developing chronic medical dz after dx MFI “semen testing” and vasectomies • DM HR 1.81 (95%CI 1.57-2.08) • Renal dz HR 1.6 (95%CI 1..14- 2.24) • Peripheral vascular dz HR 1.52 (95% CI 1.12-2.07) •
    [Show full text]
  • MSH2 Gene Muts Homolog 2
    MSH2 gene mutS homolog 2 Normal Function The MSH2 gene provides instructions for making a protein that plays an essential role in repairing DNA. This protein helps fix errors that are made when DNA is copied (DNA replication) in preparation for cell division. The MSH2 protein joins with one of two other proteins, MSH6 or MSH3 (each produced from a different gene), to form a two-protein complex called a dimer. This complex identifies locations on the DNA where errors have been made during DNA replication. Another group of proteins, the MLH1-PMS2 dimer, then binds to the MSH2 dimer and repairs the errors by removing the mismatched DNA and replicating a new segment. The MSH2 gene is one of a set of genes known as the mismatch repair (MMR) genes. Health Conditions Related to Genetic Changes Constitutional mismatch repair deficiency syndrome About 10 mutations in the MSH2 gene have been associated with a condition called constitutional mismatch repair deficiency (CMMRD) syndrome. Individuals with this condition are at increased risk of developing cancers of the colon (large intestine) and rectum (collectively referred to as colorectal cancer), brain, and blood (leukemia or lymphoma). These cancers usually first occur in childhood, with the vast majority of cancers in CMMRD syndrome diagnosed in people under the age of 18. Many people with CMMRD syndrome also develop changes in skin coloring (pigmentation), similar to those that occur in a condition called neurofibromatosis type 1. Individuals with CMMRD syndrome inherit two MSH2 gene mutations, one from each parent, while people with Lynch syndrome (described below) have a mutation in one copy of the MSH2 gene.
    [Show full text]