Skip to main content
Top
Published in: Familial Cancer 2/2017

01-04-2017 | Original Article

Truncation of the MSH2 C-terminal 60 amino acids disrupts effective DNA mismatch repair and is causative for Lynch syndrome

Authors: Eva Wielders, Elly Delzenne-Goette, Rob Dekker, Martin van der Valk, Hein te Riele

Published in: Familial Cancer | Issue 2/2017

Login to get access

Abstract

Missense variants of DNA mismatch repair (MMR) genes pose a problem in clinical genetics as long as they cannot unambiguously be assigned as the cause of Lynch syndrome (LS). To study such variants of uncertain clinical significance, we have developed a functional assay based on direct measurement of MMR activity in mouse embryonic stem cells expressing mutant protein from the endogenous alleles. We have applied this protocol to a specific truncation mutant of MSH2 that removes 60 C-terminal amino acids and has been found in suspected LS families. We show that the stability of the MSH2/MSH6 heterodimer is severely perturbed, causing attenuated MMR in in vitro assays and cancer predisposition in mice. This mutation can therefore unambiguously be considered as deleterious and causative for LS.
Appendix
Available only for authorised users
Literature
1.
go back to reference Lynch HT, Lynch J (2000) Lynch syndrome: genetics, natural history, genetic counseling, and prevention. J Clin Oncol 18:19S–31SPubMed Lynch HT, Lynch J (2000) Lynch syndrome: genetics, natural history, genetic counseling, and prevention. J Clin Oncol 18:19S–31SPubMed
2.
go back to reference Ionov Y, Peinado MA, Malkhosyan S, Shibata D, Perucho M (1993) Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature 363:558–561CrossRefPubMed Ionov Y, Peinado MA, Malkhosyan S, Shibata D, Perucho M (1993) Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature 363:558–561CrossRefPubMed
3.
go back to reference Nash GM, Gimbel M, Shia J, Culliford AT, Nathanson DR, Ndubuisi M, Yamaguchi Y, Zeng ZS, Barany F, Paty PB (2003) Automated, multiplex assay for high-frequency microsatellite instability in colorectal cancer. J Clin Oncol 21:3105–3112CrossRefPubMed Nash GM, Gimbel M, Shia J, Culliford AT, Nathanson DR, Ndubuisi M, Yamaguchi Y, Zeng ZS, Barany F, Paty PB (2003) Automated, multiplex assay for high-frequency microsatellite instability in colorectal cancer. J Clin Oncol 21:3105–3112CrossRefPubMed
4.
go back to reference Karran P (2001) Mechanisms of tolerance to DNA damaging therapeutic drugs. Carcinogenesis 22:1931–1937CrossRefPubMed Karran P (2001) Mechanisms of tolerance to DNA damaging therapeutic drugs. Carcinogenesis 22:1931–1937CrossRefPubMed
5.
go back to reference Stojic L, Mojas N, Cejka P, di Pietro M, Ferrari S, Marra G, Jiricny J (2003) Mismatch repair-dependent G2 checkpoint induced by low doses of SN1 type methylating agents requires the ATR kinase. Genes Dev 18:1331–1344CrossRef Stojic L, Mojas N, Cejka P, di Pietro M, Ferrari S, Marra G, Jiricny J (2003) Mismatch repair-dependent G2 checkpoint induced by low doses of SN1 type methylating agents requires the ATR kinase. Genes Dev 18:1331–1344CrossRef
6.
go back to reference Rayssiguier C, Thaler DS, Radman M (1989) The barrier to recombination between Escherichia coli and Salmonella typhimurium is disrupted in mismatch-repair mutants. Nature 342:396–401CrossRefPubMed Rayssiguier C, Thaler DS, Radman M (1989) The barrier to recombination between Escherichia coli and Salmonella typhimurium is disrupted in mismatch-repair mutants. Nature 342:396–401CrossRefPubMed
7.
go back to reference De Wind N, Dekker M, Berns A, Radman M, te Riele H (1995) Inactivation of the mouse Msh2 gene results in mismatch repair deficiency, methylation tolerance, hyperrecombination, and predisposition to cancer. Cell 82:321–330CrossRefPubMed De Wind N, Dekker M, Berns A, Radman M, te Riele H (1995) Inactivation of the mouse Msh2 gene results in mismatch repair deficiency, methylation tolerance, hyperrecombination, and predisposition to cancer. Cell 82:321–330CrossRefPubMed
8.
go back to reference Gradia S, Subramanian D, Wilson T, Acharya S, Makhov A, Griffith J, Fishel R (1999) hMSH2-hMSH6 forms a hydrolysis-independent sliding clamp on mismatched DNA. Mol Cell 3:255–261CrossRefPubMed Gradia S, Subramanian D, Wilson T, Acharya S, Makhov A, Griffith J, Fishel R (1999) hMSH2-hMSH6 forms a hydrolysis-independent sliding clamp on mismatched DNA. Mol Cell 3:255–261CrossRefPubMed
9.
go back to reference Umar A, Risinger JI, Glaab WE, Tindall KR, Barrett JC, Kunkel TA (1998) Functional overlap in mismatch repair by human MSH3 and MSH6. Genetics 148:1637–1646PubMedPubMedCentral Umar A, Risinger JI, Glaab WE, Tindall KR, Barrett JC, Kunkel TA (1998) Functional overlap in mismatch repair by human MSH3 and MSH6. Genetics 148:1637–1646PubMedPubMedCentral
10.
go back to reference Dekker M, de Vries S, Aarts M, Dekker R, Brouwers C, Wiebenga O, de Wind N, Cantelli E, Tonelli R, te Riele H (2011) Transient suppression of MLH1 allows effective single-nucleotide substitution by single-stranded oligonucleotides. Mutat Res 715:52–60CrossRefPubMed Dekker M, de Vries S, Aarts M, Dekker R, Brouwers C, Wiebenga O, de Wind N, Cantelli E, Tonelli R, te Riele H (2011) Transient suppression of MLH1 allows effective single-nucleotide substitution by single-stranded oligonucleotides. Mutat Res 715:52–60CrossRefPubMed
11.
go back to reference Wielders EA, Dekker RJ, Holt I, Morris GE, te Riele H (2011) Characterization of MSH2 variants by endogenous gene modification in mouse embryonic stem cells. Hum Mutat 32:389–396CrossRefPubMed Wielders EA, Dekker RJ, Holt I, Morris GE, te Riele H (2011) Characterization of MSH2 variants by endogenous gene modification in mouse embryonic stem cells. Hum Mutat 32:389–396CrossRefPubMed
12.
go back to reference Mangold E, Pagenstecher C, Friedl W, Mathiak M, Buettner R, Engel C, Loeffler M, Holinski-Feder E, Müller-Koch Y, Keller G, Schackert HK, Krüger S, Goecke T, Moeslein G, Kloor M, Gebert J, Kunstmann E, Schulmann K, Rüschoff J, Propping P (2005) Spectrum and frequencies of mutations in MSH2 and MLH1 identified in 1,721 German families suspected of hereditary nonpolyposis colorectal cancer. Int J Cancer 116:692–702CrossRefPubMed Mangold E, Pagenstecher C, Friedl W, Mathiak M, Buettner R, Engel C, Loeffler M, Holinski-Feder E, Müller-Koch Y, Keller G, Schackert HK, Krüger S, Goecke T, Moeslein G, Kloor M, Gebert J, Kunstmann E, Schulmann K, Rüschoff J, Propping P (2005) Spectrum and frequencies of mutations in MSH2 and MLH1 identified in 1,721 German families suspected of hereditary nonpolyposis colorectal cancer. Int J Cancer 116:692–702CrossRefPubMed
13.
go back to reference Krüger S, Plaschke J, Jeske B, Görgens H, Pistorius SR, Bier A, Kreuz FR, Theissig F, Aust DE, Saeger HD, Schackert HK (2003) Identification of six novel MSH2 and MLH1 germline mutations in HNPCC. Hum Mutat 21:445–446CrossRefPubMed Krüger S, Plaschke J, Jeske B, Görgens H, Pistorius SR, Bier A, Kreuz FR, Theissig F, Aust DE, Saeger HD, Schackert HK (2003) Identification of six novel MSH2 and MLH1 germline mutations in HNPCC. Hum Mutat 21:445–446CrossRefPubMed
14.
go back to reference Wang Q, Lasset C, Desseigne F, Saurin JC, Maugard C, Navarro C, Ruano E, Descos L, Trillet-Lenoir V, Bosset JF, Puisieux A (1999) Prevalence of germline mutations of hMLH1, hMSH2, hPMS1, hPMS2, and hMSH6 genes in 75 French kindreds with nonpolyposis colorectal cancer. Hum Genet 105:79–85PubMed Wang Q, Lasset C, Desseigne F, Saurin JC, Maugard C, Navarro C, Ruano E, Descos L, Trillet-Lenoir V, Bosset JF, Puisieux A (1999) Prevalence of germline mutations of hMLH1, hMSH2, hPMS1, hPMS2, and hMSH6 genes in 75 French kindreds with nonpolyposis colorectal cancer. Hum Genet 105:79–85PubMed
15.
go back to reference Papp J, Kovacs ME, Olah E (2007) Germline MLH1 and MSH2 mutational spectrum including frequent large genomic aberrations in Hungarian hereditary non-polyposis colorectal cancer families: implications for genetic testing. World J Gastroenterol 13:2727–2732CrossRefPubMedPubMedCentral Papp J, Kovacs ME, Olah E (2007) Germline MLH1 and MSH2 mutational spectrum including frequent large genomic aberrations in Hungarian hereditary non-polyposis colorectal cancer families: implications for genetic testing. World J Gastroenterol 13:2727–2732CrossRefPubMedPubMedCentral
16.
go back to reference Miyaki M, Konishi M, Muraoka M, Kikuchi-Yanoshita R, Tanaka K, Iwama T, Mori T, Koike M, Ushio K, Chiba M et al (1995) Germ line mutations of hMSH2 and hMLH1 genes in Japanese families with hereditary nonpolyposis colorectal cancer (HNPCC): usefulness of DNA analysis for screening and diagnosis of HNPCC patients. J Mol Med 73:515–520CrossRefPubMed Miyaki M, Konishi M, Muraoka M, Kikuchi-Yanoshita R, Tanaka K, Iwama T, Mori T, Koike M, Ushio K, Chiba M et al (1995) Germ line mutations of hMSH2 and hMLH1 genes in Japanese families with hereditary nonpolyposis colorectal cancer (HNPCC): usefulness of DNA analysis for screening and diagnosis of HNPCC patients. J Mol Med 73:515–520CrossRefPubMed
17.
go back to reference Yuan Y, Han HJ, Zheng S, Park JG (1998) Germline mutations of hMLH1 and hMSH2 genes in patients with suspected hereditary nonpolyposis colorectal cancer and sporadic early-onset colorectal cancer. Dis Colon Rectum 41:434–440CrossRefPubMed Yuan Y, Han HJ, Zheng S, Park JG (1998) Germline mutations of hMLH1 and hMSH2 genes in patients with suspected hereditary nonpolyposis colorectal cancer and sporadic early-onset colorectal cancer. Dis Colon Rectum 41:434–440CrossRefPubMed
18.
go back to reference Millar AL, Pal T, Madlensky L, Sherman C, Temple L, Mitri A, Cheng H, Marcus V, Gallinger S, Redston M, Bapat B, Narod S (1999) Mismatch repair gene defects contribute to the genetic basis of double primary cancers of the colorectum and endometrium. Hum Mol Genet 8:823–829CrossRefPubMed Millar AL, Pal T, Madlensky L, Sherman C, Temple L, Mitri A, Cheng H, Marcus V, Gallinger S, Redston M, Bapat B, Narod S (1999) Mismatch repair gene defects contribute to the genetic basis of double primary cancers of the colorectum and endometrium. Hum Mol Genet 8:823–829CrossRefPubMed
19.
go back to reference Terdiman JP, Gum JR Jr, Conrad PG, Miller GA, Weinberg V, Crawley SC, Levin TR, Reeves C, Schmitt A, Hepburn M, Sleisenger MH, Kim YS (2001) Efficient detection of hereditary nonpolyposis colorectal cancer gene carriers by screening for tumor microsatellite instability before germline genetic testing. Gastroenterology 120:21–30CrossRefPubMed Terdiman JP, Gum JR Jr, Conrad PG, Miller GA, Weinberg V, Crawley SC, Levin TR, Reeves C, Schmitt A, Hepburn M, Sleisenger MH, Kim YS (2001) Efficient detection of hereditary nonpolyposis colorectal cancer gene carriers by screening for tumor microsatellite instability before germline genetic testing. Gastroenterology 120:21–30CrossRefPubMed
20.
go back to reference Durno C, Aronson M, Bapat B, Cohen Z, Gallinger S (2005) Family history and molecular features of children, adolescents, and young adults with colorectal carcinoma. Gut 54:1146–1150CrossRefPubMedPubMedCentral Durno C, Aronson M, Bapat B, Cohen Z, Gallinger S (2005) Family history and molecular features of children, adolescents, and young adults with colorectal carcinoma. Gut 54:1146–1150CrossRefPubMedPubMedCentral
21.
go back to reference Bianchi F, Galizia E, Bracci R, Belvederesi L, Catalani R, Loretelli C, Giorgetti G, Ferretti C, Bearzi I, Porfiri E, Cellerino R (2007) Effectiveness of the CRCAPRO program in identifying patients suspected for HNPCC. Clin Genet 71:158–164CrossRefPubMed Bianchi F, Galizia E, Bracci R, Belvederesi L, Catalani R, Loretelli C, Giorgetti G, Ferretti C, Bearzi I, Porfiri E, Cellerino R (2007) Effectiveness of the CRCAPRO program in identifying patients suspected for HNPCC. Clin Genet 71:158–164CrossRefPubMed
22.
go back to reference Baudhuin LM, Mai M, French AJ, Kruckeberg KE, Swanson RL, Winters JL, Courteau LK, Thibodeau SN (2005) Analysis of hMLH1 and hMSH2 gene dosage alterations in hereditary nonpolyposis colorectal cancer patients by novel methods. J Mol Diagn 7:226–235CrossRefPubMedPubMedCentral Baudhuin LM, Mai M, French AJ, Kruckeberg KE, Swanson RL, Winters JL, Courteau LK, Thibodeau SN (2005) Analysis of hMLH1 and hMSH2 gene dosage alterations in hereditary nonpolyposis colorectal cancer patients by novel methods. J Mol Diagn 7:226–235CrossRefPubMedPubMedCentral
23.
go back to reference Percesepe A, Borghi F, Menigatti M, Losi L, Foroni M, Di Gregorio C, Rossi G, Pedroni M, Sala E, Vaccina F, Roncucci L, Benatti P, Viel A, Genuardi M, Marra G, Kristo P, Peltomäki P, Ponz de Leon M (2001) Molecular screening for hereditary nonpolyposis colorectal cancer: a prospective, population-based study. J Clin Oncol 19:3944–3950CrossRefPubMed Percesepe A, Borghi F, Menigatti M, Losi L, Foroni M, Di Gregorio C, Rossi G, Pedroni M, Sala E, Vaccina F, Roncucci L, Benatti P, Viel A, Genuardi M, Marra G, Kristo P, Peltomäki P, Ponz de Leon M (2001) Molecular screening for hereditary nonpolyposis colorectal cancer: a prospective, population-based study. J Clin Oncol 19:3944–3950CrossRefPubMed
24.
go back to reference Ponz de Leon M, Benatti P, Borghi F, Pedroni M, Scarselli A, Di Gregorio C, Losi L, Viel A, Genuardi M, Abbati G, Rossi G, Menigatti M, Lamberti I, Ponti G, Roncucci L (2004) Aetiology of colorectal cancer and relevance of monogenic inheritance. Gut 53:115–122CrossRefPubMedPubMedCentral Ponz de Leon M, Benatti P, Borghi F, Pedroni M, Scarselli A, Di Gregorio C, Losi L, Viel A, Genuardi M, Abbati G, Rossi G, Menigatti M, Lamberti I, Ponti G, Roncucci L (2004) Aetiology of colorectal cancer and relevance of monogenic inheritance. Gut 53:115–122CrossRefPubMedPubMedCentral
25.
go back to reference Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, Nakagawa H, Sotamaa K, Prior TW, Westman J, Panescu J, Fix D, Lockman J, Comeras I, de la Chapelle A (2005) Screening for the Lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med 352:1851–1860CrossRefPubMed Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, Nakagawa H, Sotamaa K, Prior TW, Westman J, Panescu J, Fix D, Lockman J, Comeras I, de la Chapelle A (2005) Screening for the Lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med 352:1851–1860CrossRefPubMed
26.
go back to reference Wagner A, Barrows A, Wijnen JT, van der Klift H, Franken PF, Verkuijlen P, Nakagawa H, Geugien M, Jaghmohan-Changur S, Breukel C, Meijers-Heijboer H, Morreau H, van Puijenbroek M, Burn J, Coronel S, Kinarski Y, Okimoto R, Watson P, Lynch JF, de la Chapelle A, Lynch HT, Fodde R (2003) Molecular analysis of hereditary nonpolyposis colorectal cancer in the United States: high mutation detection rate among clinically selected families and characterization of an American founder genomic deletion of the MSH2 gene. Am J Hum Genet 72:1088–1100CrossRefPubMedPubMedCentral Wagner A, Barrows A, Wijnen JT, van der Klift H, Franken PF, Verkuijlen P, Nakagawa H, Geugien M, Jaghmohan-Changur S, Breukel C, Meijers-Heijboer H, Morreau H, van Puijenbroek M, Burn J, Coronel S, Kinarski Y, Okimoto R, Watson P, Lynch JF, de la Chapelle A, Lynch HT, Fodde R (2003) Molecular analysis of hereditary nonpolyposis colorectal cancer in the United States: high mutation detection rate among clinically selected families and characterization of an American founder genomic deletion of the MSH2 gene. Am J Hum Genet 72:1088–1100CrossRefPubMedPubMedCentral
27.
go back to reference Swensen J, Lewis CM, Cannon-Albright LA (1997) Identification of a one-base germline deletion (codon 888 del C) and an intron splice acceptor site polymorphism in hMSH2. Hum Mutat 10:80–81CrossRefPubMed Swensen J, Lewis CM, Cannon-Albright LA (1997) Identification of a one-base germline deletion (codon 888 del C) and an intron splice acceptor site polymorphism in hMSH2. Hum Mutat 10:80–81CrossRefPubMed
28.
go back to reference Warren JJ, Pohlhaus TJ, Changela A, Iyer RR, Modrich PL, Beese LS (2007) Structure of the human MutSalpha DNA lesion recognition complex. Mol Cell 26:579–592CrossRefPubMed Warren JJ, Pohlhaus TJ, Changela A, Iyer RR, Modrich PL, Beese LS (2007) Structure of the human MutSalpha DNA lesion recognition complex. Mol Cell 26:579–592CrossRefPubMed
29.
go back to reference Gupta S, Gellert M, Yang W (2012) Mechanism of mismatch recognition revealed by human MutSβ bound to unpaired DNA loops. Nat Struct Mol Biol 19:72–78CrossRef Gupta S, Gellert M, Yang W (2012) Mechanism of mismatch recognition revealed by human MutSβ bound to unpaired DNA loops. Nat Struct Mol Biol 19:72–78CrossRef
30.
go back to reference Lamers MH, Perrakis A, Enzlin JH, Winterwerp HH, de Wind N, Sixma TK (2000) The crystal structure of DNA mismatch repair protein MutS binding to a G x T mismatch. Nature 407:711–717CrossRefPubMed Lamers MH, Perrakis A, Enzlin JH, Winterwerp HH, de Wind N, Sixma TK (2000) The crystal structure of DNA mismatch repair protein MutS binding to a G x T mismatch. Nature 407:711–717CrossRefPubMed
31.
32.
go back to reference Mendillo ML, Putnam CD, Kolodner RD (2007) Escherichia coli MutS tetramerization domain structure reveals that stable dimers but not tetramers are essential for DNA mismatch repair in vivo. J Biol Chem 282:16345–16354CrossRefPubMed Mendillo ML, Putnam CD, Kolodner RD (2007) Escherichia coli MutS tetramerization domain structure reveals that stable dimers but not tetramers are essential for DNA mismatch repair in vivo. J Biol Chem 282:16345–16354CrossRefPubMed
33.
go back to reference Dekker M, Brouwers C, Aarts M, van der Torre J, de Vries S, van de Vrugt H, te Riele H (2006) Effective oligonucleotide-mediated gene disruption in ES cells lacking the mismatch repair protein MSH3. Gene Ther 13:686–694CrossRefPubMed Dekker M, Brouwers C, Aarts M, van der Torre J, de Vries S, van de Vrugt H, te Riele H (2006) Effective oligonucleotide-mediated gene disruption in ES cells lacking the mismatch repair protein MSH3. Gene Ther 13:686–694CrossRefPubMed
34.
go back to reference De Wind N, Dekker M, Claij N, Jansen L, van Klink Y, Radman M, Riggins G, Van der Valk M, Van ‘t Wout K, Te Riele H (1999) HNPCC-like cancer predisposition in mice through simultaneous loss of Msh3 and Msh6 mismatch-repair protein functions. Nat Genet 23:359–362CrossRefPubMed De Wind N, Dekker M, Claij N, Jansen L, van Klink Y, Radman M, Riggins G, Van der Valk M, Van ‘t Wout K, Te Riele H (1999) HNPCC-like cancer predisposition in mice through simultaneous loss of Msh3 and Msh6 mismatch-repair protein functions. Nat Genet 23:359–362CrossRefPubMed
35.
go back to reference Claij N, Te Riele H (2002) Methylation tolerance in mismatch repair proficient cells with low MSH2 level. Oncogene 21:2873–2879CrossRefPubMed Claij N, Te Riele H (2002) Methylation tolerance in mismatch repair proficient cells with low MSH2 level. Oncogene 21:2873–2879CrossRefPubMed
Metadata
Title
Truncation of the MSH2 C-terminal 60 amino acids disrupts effective DNA mismatch repair and is causative for Lynch syndrome
Authors
Eva Wielders
Elly Delzenne-Goette
Rob Dekker
Martin van der Valk
Hein te Riele
Publication date
01-04-2017
Publisher
Springer Netherlands
Published in
Familial Cancer / Issue 2/2017
Print ISSN: 1389-9600
Electronic ISSN: 1573-7292
DOI
https://doi.org/10.1007/s10689-016-9945-x

Other articles of this Issue 2/2017

Familial Cancer 2/2017 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine