Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Genetics and Genomics

Lynch syndrome, molecular mechanisms and variant classification

Abstract

Patients with the heritable cancer disease, Lynch syndrome, carry germline variants in the MLH1, MSH2, MSH6 and PMS2 genes, encoding the central components of the DNA mismatch repair system. Loss-of-function variants disrupt the DNA mismatch repair system and give rise to a detrimental increase in the cellular mutational burden and cancer development. The treatment prospects for Lynch syndrome rely heavily on early diagnosis; however, accurate diagnosis is inextricably linked to correct clinical interpretation of individual variants. Protein variant classification traditionally relies on cumulative information from occurrence in patients, as well as experimental testing of the individual variants. The complexity of variant classification is due to (1) that variants of unknown significance are rare in the population and phenotypic information on the specific variants is missing, and (2) that individual variant testing is challenging, costly and slow. Here, we summarise recent developments in high-throughput technologies and computational prediction tools for the assessment of variants of unknown significance in Lynch syndrome. These approaches may vastly increase the number of interpretable variants and could also provide important mechanistic insights into the disease. These insights may in turn pave the road towards developing personalised treatment approaches for Lynch syndrome.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The human DNA mismatch repair (MMR) system.
Fig. 2: Proteasomal degradation of misfolded proteins.
Fig. 3: Overview of tools used for testing individual variant effects.

Similar content being viewed by others

References

  1. Pearlman R, Frankel WL, Swanson B, Zhao W, Yilmaz A, Miller K, et al. Prevalence and spectrum of germline cancer susceptibility gene mutations among patients with early-onset colorectal cancer. JAMA Oncol. 2017;3:464–71.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Yurgelun MB, Kulke MH, Fuchs CS, Allen BA, Uno H, Hornick JL, et al. Cancer susceptibility gene mutations in individuals with colorectal cancer. J Clin Oncol. 2017;35:1086–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Dominguez-Valentin M, Sampson JR, Seppälä TT, ten Broeke SW, Plazzer J-P, Nakken S, et al. Cancer risks by gene, age, and gender in 6350 carriers of pathogenic mismatch repair variants: findings from the Prospective Lynch Syndrome Database. Genet Med. 2020;22:15–25.

    Article  CAS  PubMed  Google Scholar 

  4. Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, et al. Screening for the Lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med. 2005;352:1851–60.

    Article  CAS  PubMed  Google Scholar 

  5. Møller P, Seppälä T, Bernstein I, Holinski-Feder E, Sala P, Evans DG, et al. Cancer incidence and survival in Lynch syndrome patients receiving colonoscopic and gynaecological surveillance: first report from the prospective Lynch syndrome database. Gut. 2017;66:464–72.

    Article  PubMed  Google Scholar 

  6. Boland PM, Yurgelun MB, Boland CR. Recent progress in lynch syndrome and other familial colorectal cancer syndromes. CA Cancer J Clin. 2018;68:217–31.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Seppälä TT, Latchford A, Negoi I, Sampaio Soares A, Jimenez-Rodriguez R, Sánchez-Guillén L, et al. European guidelines from the EHTG and ESCP for Lynch syndrome: an updated third edition of the Mallorca guidelines based on gene and gender. Br J Surg. 2021;108:484–98.

    Article  PubMed  Google Scholar 

  8. Crosbie EJ, Ryan NAJ, Arends MJ, Bosse T, Burn J, Cornes JM, et al. The Manchester International Consensus Group recommendations for the management of gynecological cancers in Lynch syndrome. Genet Med J Am Coll Med Genet. 2019;21:2390–400.

    Google Scholar 

  9. Grindedal EM, Renkonen-Sinisalo L, Vasen H, Evans G, Sala P, Blanco I, et al. Survival in women with MMR mutations and ovarian cancer: a multicentre study in Lynch syndrome kindreds. J Med Genet. 2010;47:99–102.

    Article  CAS  PubMed  Google Scholar 

  10. Schmeler KM, Lu KH. Gynecologic cancers associated with Lynch syndrome/HNPCC. Clin Transl Oncol. 2008;10:313–7.

    Article  CAS  PubMed  Google Scholar 

  11. Therkildsen C, Jensen LH, Rasmussen M, Bernstein I. An update on immune checkpoint therapy for the treatment of Lynch syndrome. Clin Exp Gastroenterol. 2021;14:181–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. StCharles JA, Liberti SE, Williams JS, Lujan SA, Kunkel TA. Quantifying the contributions of base selectivity, proofreading and mismatch repair to nuclear DNA replication in Saccharomyces cerevisiae. DNA Repair. 2015;31:41–51.

    Article  CAS  Google Scholar 

  13. Huang Y, Li G-M. DNA mismatch repair preferentially safeguards actively transcribed genes. DNA Repair. 2018;71:82–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Preston BD, Albertson TM, Herr AJ. DNA replication fidelity and cancer. Semin Cancer Biol 2010;20:281–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hitchins MP. Inheritance of epigenetic aberrations (constitutional epimutations) in cancer susceptibility. Adv Genet. 2010;70:201–43.

    Article  CAS  PubMed  Google Scholar 

  16. Hitchins MP, Ward RL. Constitutional (germline) MLH1 epimutation as an aetiological mechanism for hereditary non-polyposis colorectal cancer. J Med Genet. 2009;46:793–802.

    Article  CAS  PubMed  Google Scholar 

  17. Ward RL, Dobbins T, Lindor NM, Rapkins RW, Hitchins MP. Identification of constitutional MLH1 epimutations and promoter variants in colorectal cancer patients from the Colon Cancer Family Registry. Genet Med. 2013;15:25–35.

    Article  CAS  PubMed  Google Scholar 

  18. Ligtenberg MJL, Kuiper RP, Chan TL, Goossens M, Hebeda KM, Voorendt M, et al. Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3′ exons of TACSTD1. Nat Genet. 2009;41:112–7.

    Article  CAS  PubMed  Google Scholar 

  19. Duraturo F, Liccardo R, Cavallo A, De Rosa M, Grosso M, Izzo P. Association of low-risk MSH3 and MSH2 variant alleles with Lynch syndrome: probability of synergistic effects. Int J Cancer. 2011;129:1643–50.

    Article  CAS  PubMed  Google Scholar 

  20. Jiricny J. The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol. 2006;7:335–46.

    Article  CAS  PubMed  Google Scholar 

  21. Dufner P, Marra G, Räschle M, Jiricny J. Mismatch recognition and DNA-dependent stimulation of the ATPase activity of hMutSα is abolished by a single mutation in the hMSH6 subunit *. J Biol Chem. 2000;275:36550–5.

    Article  CAS  PubMed  Google Scholar 

  22. Gupta S, Gellert M, Yang W. Mechanism of mismatch recognition revealed by human MutSβ bound to unpaired DNA loops. Nat Struct Mol Biol. 2012;19:72–8.

    Article  CAS  Google Scholar 

  23. Reyes GX, Schmidt TT, Kolodner RD, Hombauer H. New insights into the mechanism of DNA mismatch repair. Chromosoma. 2015;124:443–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Groothuizen FS, Winkler I, Cristóvão M, Fish A, Winterwerp HH, Reumer A, et al. MutS/MutL crystal structure reveals that the MutS sliding clamp loads MutL onto DNA. eLife. 2015;4:e06744.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Bowen N, Smith CE, Srivatsan A, Willcox S, Griffith JD, Kolodner RD. Reconstitution of long and short patch mismatch repair reactions using Saccharomyces cerevisiae proteins. Proc Natl Acad Sci USA. 2013;110:18472–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jiricny J. Postreplicative mismatch repair. Cold Spring Harb Perspect Biol. 2013;5:a012633.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Groothuizen FS, Sixma TK. The conserved molecular machinery in DNA mismatch repair enzyme structures. DNA Repair. 2016;38:14–23.

    Article  CAS  PubMed  Google Scholar 

  28. Gueneau E, Dherin C, Legrand P, Tellier-Lebegue C, Gilquin B, Bonnesoeur P, et al. Structure of the MutLα C-terminal domain reveals how Mlh1 contributes to Pms1 endonuclease site. Nat Struct Mol Biol 2013;20:461–8.

    Article  CAS  PubMed  Google Scholar 

  29. Goellner EM, Putnama CD, Kolodnera RD. Exonuclease 1-dependent and independent mismatch repair. DNA Repair. 2015;32:24–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cannavo E, Sanchez A, Anand R, Ranjha L, Hugener J, Adam C, et al. Regulation of the MLH1–MLH3 endonuclease in meiosis. Nature. 2020;586:618–22.

    Article  CAS  PubMed  Google Scholar 

  31. Kadyrova LY, Gujar V, Burdett V, Modrich PL, Kadyrov FA. Human MutLγ, the MLH1–MLH3 heterodimer, is an endonuclease that promotes DNA expansion. Proc Natl Acad Sci USA. 2020;117:3535–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Campbell CS, Hombauer H, Srivatsan A, Bowen N, Gries K, Desai A, et al. Mlh2 is an accessory factor for DNA mismatch repair in Saccharomyces cerevisiae. PLOS Genet. 2014;10:e1004327.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Gupta D, Heinen CD. The mismatch repair-dependent DNA damage response: mechanisms and implications. DNA Repair. 2019;78:60–9.

    Article  CAS  PubMed  Google Scholar 

  34. Lynch HT, Snyder CL, Shaw TG, Heinen CD, Hitchins MP. Milestones of Lynch syndrome: 1895–2015. Nat Rev Cancer. 2015;15:181–94.

    Article  CAS  PubMed  Google Scholar 

  35. De’ Angelis GL, Bottarelli L, Azzoni C, De’ Angelis N, Leandro G, Di Mario F, et al. Microsatellite instability in colorectal cancer. Acta Bio-Med Atenei Parm. 2018;89:97–101.

    Google Scholar 

  36. Imai K, Yamamoto H. Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis. 2008;29:673–80.

    Article  CAS  PubMed  Google Scholar 

  37. Yamamoto H, Imai K. Microsatellite instability: an update. Arch Toxicol. 2015;89:899–921.

    Article  CAS  PubMed  Google Scholar 

  38. Herman JG, Umar A, Polyak K, Graff JR, Ahuja N, Issa JP, et al. Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma. Proc Natl Acad Sci USA. 1998;95:6870–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Vasen HF, Watson P, Mecklin JP, Lynch HT. New clinical criteria for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed by the International Collaborative group on HNPCC. Gastroenterology. 1999;116:1453–6.

    Article  CAS  PubMed  Google Scholar 

  40. Umar A, Boland CR, Terdiman JP, Syngal S, Chapelle A, de la, Rüschoff J, et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. JNCI J Natl Cancer Inst. 2004;96:261–8.

    Article  CAS  PubMed  Google Scholar 

  41. Kastrinos F, Allen JI, Stockwell DH, Stoffel EM, Cook EF, Mutinga ML, et al. Development and validation of a colon cancer risk assessment tool for patients undergoing colonoscopy. Am J Gastroenterol. 2009;104:1508–18.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Giardiello FM, Allen JI, Axilbund JE, Boland RC, Burke CA, Burt RW, et al. Guidelines on genetic evaluation and management of Lynch syndrome: a consensus statement by the US Multi-Society Task Force on colorectal cancer. J Am Coll Gastroenterol ACG. 2014;109:1159–79.

    Article  Google Scholar 

  43. Win AK, Jenkins MA, Dowty JG, Antoniou AC, Lee A, Giles GG, et al. Prevalence and penetrance of major genes and polygenes for colorectal cancer. Cancer Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol. 2017;26:404–12.

    Article  CAS  Google Scholar 

  44. Moreira L, Balaguer F, Lindor N, de la Chapelle A, Hampel H, Aaltonen LA, et al. Identification of Lynch syndrome among patients with colorectal cancer. J Am Med Assoc. 2012;308:1555–65.

    Article  CAS  Google Scholar 

  45. Peltomäki P. Update on Lynch syndrome genomics. Fam Cancer. 2016;15:385–93.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Dominguez-Valentin M, Plazzer J-P, Sampson JR, Engel C, Aretz S, Jenkins MA, et al. No difference in penetrance between truncating and missense/aberrant splicing pathogenic variants in MLH1 and MSH2: a prospective Lynch Syndrome Database Study. J Clin Med. 2021;10:2856.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Knudson AG. Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci USA 1971;68:820–3.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Hemminki A, Peltomäki P, Mecklin J-P, Järvinen H, Salovaara R, Nyström-Lahti M, et al. Loss of the wild type MLH1 gene is a feature of hereditary nonpolyposis colorectal cancer. Nat Genet. 1994;8:405–10.

    Article  CAS  PubMed  Google Scholar 

  49. Zhang M, Xiang S, Joo H-Y, Wang L, Williams KA, Liu W, et al. HDAC6 deacetylates and ubiquitinates MSH2 to maintain proper levels of MutSα. Mol Cell. 2014;55:31–46.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Abildgaard AB, Stein A, Nielsen SV, Schultz-Knudsen K, Papaleo E, Shrikhande A, et al. Computational and cellular studies reveal structural destabilization and degradation of MLH1 variants in Lynch syndrome. eLife. 2019;8:e49138.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hinrichsen I, Weßbecher IM, Huhn M, Passmann S, Zeuzem S, Plotz G, et al. Phosphorylation-dependent signaling controls degradation of DNA mismatch repair protein PMS2. Mol Carcinog. 2017;56:2663–8.

    Article  CAS  PubMed  Google Scholar 

  52. Mohd AB, Palama B, Nelson SE, Tomer G, Nguyen M, Huo X, et al. Truncation of the C-terminus of human MLH1 blocks intracellular stabilization of PMS2 and disrupts DNA mismatch repair. DNA Repair. 2006;5:347–61.

    Article  CAS  PubMed  Google Scholar 

  53. Rosty C, Clendenning M, Walsh MD, Eriksen SV, Southey MC, Winship IM, et al. Germline mutations in PMS2 and MLH1 in individuals with solitary loss of PMS2 expression in colorectal carcinomas from the Colon Cancer Family Registry Cohort. BMJ Open. 2016;6:e010293.

    Article  PubMed  PubMed Central  Google Scholar 

  54. ten Broeke SW, van der Klift HM, Tops CMJ, Aretz S, Bernstein I, Buchanan DD, et al. Cancer risks for PMS2-associated Lynch syndrome. J Clin Oncol. 2018;36:2961–8.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Wang C, Wang Y, Hughes KS, Parmigiani G, Braun D. Penetrance of colorectal cancer among mismatch repair gene mutation carriers: a meta-analysis. JNCI Cancer Spectr. 2020;4:pkaa027.

  56. Thompson BA, Spurdle AB, Plazzer J-P, Greenblatt MS, Akagi K, Al-Mulla F, et al. Application of a five-tiered scheme for standardized classification of 2,360 unique mismatch repair gene variants lodged on the InSiGHT locus-specific database. Nat Genet. 2014;46:107–15.

    Article  CAS  PubMed  Google Scholar 

  57. Williams PD, Pollock DD, Goldstein RA. Functionality and the evolution of marginal stability in proteins: inferences from lattice simulations. Evol Bioinforma Online. 2007;2:91–101.

    Google Scholar 

  58. Abildgaard AB, Gersing SK, Larsen-Ledet S, Nielsen SV, Stein A, Lindorff-Larsen K, et al. Co-chaperones in targeting and delivery of misfolded proteins to the 26S proteasome. Biomolecules. 2020;10:1141.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Clausen L, Abildgaard AB, Gersing SK, Stein A, Lindorff-Larsen K, Hartmann-Petersen R. Chapter two - protein stability and degradation in health and disease. In: Advances in protein chemistry and structural biology. Donev R, editor. Vol. 114. London, United Kingdom: Academic Press; 2019. p. 61–83.

  60. Kohler V, Andréasson C. Hsp70-mediated quality control: should I stay or should I go? Biol Chem. 2020;401:1233–48.

    Article  CAS  PubMed  Google Scholar 

  61. Reinle K, Mogk A, Bukau B. The diverse functions of small heat shock proteins in the proteostasis network. J Mol Biol. 2022;434:167157.

    Article  CAS  PubMed  Google Scholar 

  62. Rosenzweig R, Nillegoda NB, Mayer MP, Bukau B. The Hsp70 chaperone network. Nat Rev Mol Cell Biol. 2019;20:665–80.

    Article  CAS  PubMed  Google Scholar 

  63. Hernandez-Pigeon H, Laurent G, Humbert O, Salles B, Lautier D. Degadration of mismatch repair hMutSα heterodimer by the ubiquitin-proteasome pathway. FEBS Lett. 2004;562:40–4.

    Article  CAS  PubMed  Google Scholar 

  64. Wu Q, Huang Y, Gu L, Chang Z, Li G-M. OTUB1 stabilizes mismatch repair protein MSH2 by blocking ubiquitination. J Biol Chem. 2021;296:100466.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhang M, Hu C, Tong D, Xiang S, Williams K, Bai W, et al. Ubiquitin-specific peptidase 10 (USP10) deubiquitinates and stabilizes MutS homolog 2 (MSH2) to regulate cellular sensitivity to DNA damage*. J Biol Chem. 2016;291:10783–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Jia X, Burugula BB, Chen V, Lemons RM, Jayakody S, Maksutova M, et al. Massively parallel functional testing of MSH2 missense variants conferring Lynch syndrome risk. Am J Hum Genet. 2021;108:163–75.

    Article  CAS  PubMed  Google Scholar 

  67. Nielsen SV, Hartmann-Petersen R, Stein A, Lindorff-Larsen K. Multiplexed assays reveal effects of missense variants in MSH2 and cancer predisposition. PLOS Genet. 2021;17:e1009496.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Nielsen SV, Stein A, Dinitzen AB, Papaleo E, Tatham MH, Poulsen EG, et al. Predicting the impact of Lynch syndrome-causing missense mutations from structural calculations. PLoS Genet. 2017;13:e1006739.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Ollodart AR, Yeh C-LC, Miller AW, Shirts BH, Gordon AS, Dunham MJ. Multiplexing mutation rate assessment: determining pathogenicity of Msh2 variants in Saccharomyces cerevisiae. Genetics. 2021;218:iyab058.

  70. Arlow T, Scott K, Wagenseller A, Gammie A. Proteasome inhibition rescues clinically significant unstable variants of the mismatch repair protein Msh2. Proc Natl Acad Sci USA. 2013;110:246–51.

    Article  CAS  PubMed  Google Scholar 

  71. Kampmeyer C, Nielsen SV, Clausen L, Stein A, Gerdes A-M, Lindorff-Larsen K, et al. Blocking protein quality control to counter hereditary cancers. Genes Chromosomes Cancer. 2017;56:823–31.

    Article  CAS  PubMed  Google Scholar 

  72. Stein A, Fowler DM, Hartmann-Petersen R, Lindorff-Larsen K. Biophysical and mechanistic models for disease-causing protein variants. Trends Biochem Sci. 2019;44:575–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Livesey BJ, Marsh JA. Using deep mutational scanning to benchmark variant effect predictors and identify disease mutations. Mol Syst Biol. 2020;16:e9380.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Starita LM, Ahituv N, Dunham MJ, Kitzman JO, Roth FP, Seelig G, et al. Variant interpretation: functional assays to the rescue. Am J Hum Genet. 2017;101:315–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Ponti G, Castellsagué E, Ruini C, Percesepe A, Tomasi A. Mismatch repair genes founder mutations and cancer susceptibility in Lynch syndrome: mismatch repair genes founder mutations in Lynch syndrome. Clin Genet. 2015;87:507–16.

    Article  CAS  PubMed  Google Scholar 

  76. Foulkes WD, Thiffault I, Gruber SB, Horwitz M, Hamel N, Lee C, et al. The founder mutation MSH2*1906G->C is an important cause of hereditary nonpolyposis colorectal cancer in the Ashkenazi Jewish population. Am J Hum Genet. 2002;71:1395–412.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Brnich SE, Rivera‐Muñoz EA, Berg JS. Quantifying the potential of functional evidence to reclassify variants of uncertain significance in the categorical and Bayesian interpretation frameworks. Hum Mutat. 2018;39:1531–41.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Houlleberghs H, Goverde A, Lusseveld J, Dekker M, Bruno MJ, Menko FH, et al. Suspected Lynch syndrome associated MSH6 variants: a functional assay to determine their pathogenicity. PLOS Genet. 2017;13:e1006765.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Houlleberghs H, Dekker M, Lantermans H, Kleinendorst R, Dubbink HJ, Hofstra RMW, et al. Oligonucleotide-directed mutagenesis screen to identify pathogenic Lynch syndrome-associated MSH2 DNA mismatch repair gene variants. Proc Natl Acad Sci USA. 2016;113:4128–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Alim I, Loke J, Yam S, Templeton AS, Newcomb P, Lindor NM, et al. Cancer risk C (CR-C), a functional genomics test is a sensitive and rapid test for germline mismatch repair deficiency. Genet. Med. 2022;S1098360022007584. https://doi.org/10.1016/j.gim.2022.05.003.

  81. Cagiada M, Johansson KE, Valanciute A, Nielsen SV, Hartmann-Petersen R, Yang JJ, et al. Understanding the origins of loss of protein function by analyzing the effects of thousands of variants on activity and abundance. Mol Biol Evol. 2021;38:3235–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Arora S, Huwe PJ, Sikder R, Shah M, Browne AJ, Lesh R, et al. Functional analysis of rare variants in mismatch repair proteins augments results from computation-based predictive methods. Cancer Biol Ther. 2017;18:519–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Niroula A, Vihinen M. Classification of amino acid substitutions in mismatch repair proteins using PON-MMR2. Hum Mutat. 2015;36:1128–34.

    Article  CAS  PubMed  Google Scholar 

  84. Adzhubei I, Jordan DM, Sunyaev SR. Predicting functional effect of human missense mutations using PolyPhen-2. Curr Protoc Hum Genet. 2013;Chapter 7:Unit7.20.

  85. Choi Y, Chan AP. PROVEAN web server: a tool to predict the functional effect of amino acid substitutions and indels. Bioinformatics. 2015;31:2745–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Kumar P, Henikoff S, Ng PC. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4:1073–81.

    Article  CAS  PubMed  Google Scholar 

  87. Hopf TA, Ingraham JB, Poelwijk FJ, Schärfe CPI, Springer M, Sander C, et al. Mutation effects predicted from sequence co-variation. Nat Biotechnol. 2017;35:128–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Laine E, Karami Y, Carbone A. GEMME: a simple and fast global epistatic model predicting mutational effects. Mol Biol Evol. 2019;36:2604–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Riesselman AJ, Ingraham JB, Marks DS. Deep generative models of genetic variation capture the effects of mutations. Nat Methods. 2018;15:816–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Frazer J, Notin P, Dias M, Gomez A, Min JK, Brock K, et al. Disease variant prediction with deep generative models of evolutionary data. Nature. 2021;599:91–5.

    Article  CAS  PubMed  Google Scholar 

  91. Park H, DiMaio F, Baker D. CASP11 refinement experiments with ROSETTA. Proteins Struct Funct Bioinforma. 2016;84:314–22.

    Article  Google Scholar 

  92. Guerois R, Nielsen JE, Serrano L. Predicting changes in the stability of proteins and protein complexes: a study of more than 1000 mutations. J Mol Biol. 2002;320:369–87.

    Article  CAS  PubMed  Google Scholar 

  93. Schymkowitz J, Borg J, Stricher F, Nys R, Rousseau F, Serrano L. The FoldX web server: an online force field. Nucleic Acids Res. 2005;33:W382–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Raevaara TE, Korhonen MK, Lohi H, Hampel H, Lynch E, Lönnqvist KE, et al. Functional significance and clinical phenotype of nontruncating mismatch repair variants of MLH1. Gastroenterology. 2005;129:537–49.

    CAS  PubMed  Google Scholar 

  95. Takahashi M, Shimodaira H, Andreutti-Zaugg C, Iggo R, Kolodner RD, Ishioka C. Functional analysis of human MLH1 variants using yeast and in vitro mismatch repair assays. Cancer Res. 2007;67:4595–604.

    Article  CAS  PubMed  Google Scholar 

  96. De Baets G, Van Durme J, Reumers J, Maurer-Stroh S, Vanhee P, Dopazo J, et al. SNPeffect 4.0: on-line prediction of molecular and structural effects of protein-coding variants. Nucleic Acids Res. 2012;40:D935–9.

    Article  PubMed  Google Scholar 

  97. Gerasimavicius L, Liu X, Marsh JA. Identification of pathogenic missense mutations using protein stability predictors. Sci Rep. 2020;10:15387.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Pey AL, Stricher F, Serrano L, Martinez A. Predicted effects of missense mutations on native-state stability account for phenotypic outcome in phenylketonuria, a paradigm of misfolding diseases. Am J Hum Genet. 2007;81:1006–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Fowler DM, Fields S. Deep mutational scanning: a new style of protein science. Nat Methods. 2014;11:801–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Raraigh KS, Han ST, Davis E, Evans TA, Pellicore MJ, McCague AF, et al. Functional assays are essential for interpretation of missense variants associated with variable expressivity. Am J Hum Genet. 2018;102:1062–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Sun S, Yang F, Tan G, Costanzo M, Oughtred R, Hirschman J, et al. An extended set of yeast-based functional assays accurately identifies human disease mutations. Genome Res. 2016;26:670–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Jepsen MM, Fowler DM, Hartmann-Petersen R, Stein A, Lindorff-Larsen K. Chapter 5 - Classifying disease-associated variants using measures of protein activity and stability. In: Protein homeostasis diseases. Pey AL, editor. London, United Kingdom: Academic Press; 2020, p. 91–107.

  103. Matreyek KA, Starita LM, Stephany JJ, Martin B, Chiasson MA, Gray VE, et al. Multiplex assessment of protein variant abundance by massively parallel sequencing. Nat Genet. 2018;50:874–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Brnich SE, On behalf of the Clinical Genome Resource Sequence Variant Interpretation Working Group, Abou Tayoun AN, Couch FJ, Cutting GR, Greenblatt MS, et al. Recommendations for application of the functional evidence PS3/BS3 criterion using the ACMG/AMP sequence variant interpretation framework. Genome Med. 2020;12:3.

    Article  Google Scholar 

  105. Ernst C, Hahnen E, Engel C, Nothnagel M, Weber J, Schmutzler RK, et al. Performance of in silico prediction tools for the classification of rare BRCA1/2 missense variants in clinical diagnostics. BMC Med Genomics. 2018;11:35.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Miosge LA, Field MA, Sontani Y, Cho V, Johnson S, Palkova A, et al. Comparison of predicted and actual consequences of missense mutations. Proc Natl Acad Sci USA. 2015;112:E5189–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Grimm DG, Azencott C-A, Aicheler F, Gieraths U, MacArthur DG, Samocha KE, et al. The evaluation of tools used to predict the impact of missense variants is hindered by two types of circularity. Hum Mutat. 2015;36:513–23.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Wu Y, Li R, Sun S, Weile J, Roth FP. Improved pathogenicity prediction for rare human missense variants. Am J Hum Genet. 2021. https://doi.org/10.1016/j.ajhg.2021.08.012.

  109. Høie MH, Cagiada M, Beck Frederiksen AH, Stein A, Lindorff-Larsen K. Predicting and interpreting large-scale mutagenesis data using analyses of protein stability and conservation. Cell Rep. 2022;38:110207.

    Article  PubMed  Google Scholar 

  110. Thompson S, Zhang Y, Ingle C, Reynolds KA, Kortemme T. Altered expression of a quality control protease in E. coli reshapes the in vivo mutational landscape of a model enzyme. eLife. 2020;9:e53476.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Brusa I, Sondo E, Falchi F, Pedemonte N, Roberti M, Cavalli A. Proteostasis regulators in cystic fibrosis: current development and future perspectives. J Med Chem. 2022. https://doi.org/10.1021/acs.jmedchem.1c01897.

  112. Hutt DM, Herman D, Rodrigues APC, Noel S, Pilewski JM, Matteson J, et al. Reduced histone deacetylase 7 activity restores function to misfolded CFTR in cystic fibrosis. Nat Chem Biol. 2010;6:25–33.

    Article  CAS  PubMed  Google Scholar 

  113. Okiyoneda T, Veit G, Dekkers JF, Bagdany M, Soya N, Xu H, et al. Mechanism-based corrector combination restores ΔF508-CFTR folding and function. Nat Chem Biol. 2013;9:444–54.

    Article  CAS  PubMed  Google Scholar 

  114. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405–24.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Fayer S, Horton C, Dines JN, Rubin AF, Richardson ME, McGoldrick K, et al. Closing the gap: systematic integration of multiplexed functional data resolves variants of uncertain significance in BRCA1, TP53, and PTEN. Am J Hum Genet. 2021;S0002-9297(21)00411–0. https://doi.org/10.1016/j.ajhg.2021.11.001.

  116. Kuang D, Truty R, Weile J, Johnson B, Nykamp K, Araya C, et al. Prioritizing genes for systematic variant effect mapping. Bioinformatics. 2020;36:5448–55.

    Article  CAS  PubMed Central  Google Scholar 

  117. Manolio TA, Fowler DM, Starita LM, Haendel MA, MacArthur DG, Biesecker LG, et al. Bedside back to bench: building bridges between basic and clinical genomic research. Cell. 2017;169:6–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

Our research in this area is funded by the Novo Nordisk Foundation Challenge programme PRISM (NNFOC180033950; to AS, KLL and RHP), the Lundbeck Foundation (R272-2017-4528 to AS), and the Danish Council for Independent Research, Natural Sciences (7014-00039B to RHP).

Author information

Authors and Affiliations

Authors

Contributions

ABA and SVN prepared the figures. ABA, SVN, IB, AS, KLL and RHP wrote the manuscript.

Corresponding authors

Correspondence to Sofie V. Nielsen, Kresten Lindorff-Larsen or Rasmus Hartmann-Petersen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

Not applicable.

Consent to publish

Not applicable.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Abildgaard, A.B., Nielsen, S.V., Bernstein, I. et al. Lynch syndrome, molecular mechanisms and variant classification. Br J Cancer 128, 726–734 (2023). https://doi.org/10.1038/s41416-022-02059-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-022-02059-z

This article is cited by

Search

Quick links