Epigenetic MLH1 silencing concurs with mismatch repair deficiency in sporadic, naturally occurring colorectal cancer in rhesus macaques

J Transl Med. 2024 Mar 19;22(1):292. doi: 10.1186/s12967-024-04869-6.

Abstract

Background: Naturally occurring colorectal cancers (CRC) in rhesus macaques share many features with their human counterparts and are useful models for cancer immunotherapy; but mechanistic data are lacking regarding the comparative molecular pathogenesis of these cancers.

Methods: We conducted state-of-the-art imaging including CT and PET, clinical assessments, and pathological review of 24 rhesus macaques with naturally occurring CRC. Additionally, we molecularly characterized these tumors utilizing immunohistochemistry (IHC), microsatellite instability assays, DNAseq, transcriptomics, and developed a DNA methylation-specific qPCR assay for MLH1, CACNA1G, CDKN2A, CRABP1, and NEUROG1, human markers for CpG island methylator phenotype (CIMP). We furthermore employed Monte-Carlo simulations to in-silico model alterations in DNA topology in transcription-factor binding site-rich promoter regions upon experimentally demonstrated DNA methylation.

Results: Similar cancer histology, progression patterns, and co-morbidities could be observed in rhesus as reported for human CRC patients. IHC identified loss of MLH1 and PMS2 in all cases, with functional microsatellite instability. DNA sequencing revealed the close genetic relatedness to human CRCs, including a similar mutational signature, chromosomal instability, and functionally-relevant mutations affecting KRAS (G12D), TP53 (R175H, R273*), APC, AMER1, ALK, and ARID1A. Interestingly, MLH1 mutations were rarely identified on a somatic or germline level. Transcriptomics not only corroborated the similarities of rhesus and human CRCs, but also demonstrated the significant downregulation of MLH1 but not MSH2, MSH6, or PMS2 in rhesus CRCs. Methylation-specific qPCR suggested CIMP-positivity in 9/16 rhesus CRCs, but all 16/16 exhibited significant MLH1 promoter hypermethylation. DNA hypermethylation was modelled to affect DNA topology, particularly propeller twist and roll profiles. Modelling the DNA topology of a transcription factor binding motif (TFAP2A) in the MLH1 promoter that overlapped with a methylation-specific probe, we observed significant differences in DNA topology upon experimentally shown DNA methylation. This suggests a role of transcription factor binding interference in epigenetic silencing of MLH1 in rhesus CRCs.

Conclusions: These data indicate that epigenetic silencing suppresses MLH1 transcription, induces the loss of MLH1 protein, abrogates mismatch repair, and drives genomic instability in naturally occurring CRC in rhesus macaques. We consider this spontaneous, uninduced CRC in immunocompetent, treatment-naïve rhesus macaques to be a uniquely informative model for human CRC.

Keywords: Colorectal cancer; Epigenetic silencing; Mismatch repair deficiency; Nonhuman primates; Translational oncology.

MeSH terms

  • Animals
  • Brain Neoplasms*
  • Colorectal Neoplasms* / pathology
  • DNA / metabolism
  • DNA Methylation / genetics
  • DNA Mismatch Repair / genetics
  • Epigenesis, Genetic
  • Humans
  • Macaca mulatta / genetics
  • Macaca mulatta / metabolism
  • Microsatellite Instability*
  • Mismatch Repair Endonuclease PMS2 / genetics
  • Mismatch Repair Endonuclease PMS2 / metabolism
  • MutL Protein Homolog 1 / genetics
  • Neoplastic Syndromes, Hereditary*
  • Transcription Factors / genetics
  • Transcription Factors / metabolism

Substances

  • MutL Protein Homolog 1
  • Mismatch Repair Endonuclease PMS2
  • Transcription Factors
  • DNA
  • MLH1 protein, human

Supplementary concepts

  • Turcot syndrome