Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 612
Filter
1.
J Clin Invest ; 134(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38557488

ABSTRACT

While breast cancer 2 (BRCA2) loss of heterozygosity (LOH) promotes cancer initiation, it can also induce death in nontransformed cells. In contrast, mismatch repair gene mutL homolog 1 (MLH1) is a tumor-suppressor gene that protects cells from cancer development through repairing mismatched base pairs during DNA mismatch repair (MMR). Sengodan et al., in this issue of the JCI, reveal an interplay between the 2 genes: MLH1 promoted the survival of BRCA2-deficient cells independently of its MMR function. MLH1 protected replication forks from degradation, while also resolving R-loops, thereby reducing genomic instability. Moreover, MLH1 expression was regulated directly by estrogen, shedding light into the hormone-responsive nature of many BRCA2 mutant breast cancers. These results provide important insight into the genetics that drive the initiation of BRCA2-mutated breast cancers.


Subject(s)
Breast Neoplasms , MutL Protein Homolog 1 , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Genomic Instability , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism
2.
Plant Sci ; 342: 112050, 2024 May.
Article in English | MEDLINE | ID: mdl-38401766

ABSTRACT

The DNA mismatch repair (MMR) is a postreplicative system that guarantees genomic stability by correcting mispaired and unpaired nucleotides. In eukaryotic nuclei, MMR is initiated by the binding of heterodimeric MutS homologue (MSH) complexes to the DNA error or lesion. Among these proteins, MSH2-MSH6 is the most abundant heterodimer. Even though the MMR mechanism and proteins are highly conserved throughout evolution, physiological differences between species can lead to different regulatory features. Here, we investigated how light, sugar, and/or hormones modulate Arabidopsis thaliana MSH6 expression pattern. We first characterized the promoter region of MSH6. Phylogenetic shadowing revealed three highly conserved regions. These regions were analyzed by the generation of deletion constructs of the MSH6 full-length promoter fused to the ß-glucuronidase (GUS) gene. Combined, our in silico and genetic analyses revealed that a 121-bp promoter fragment was necessary for MSH6 expression and contained potential cis-acting elements involved in light- and hormone-responsive gene expression. Accordingly, light exposure or sugar treatment of four-day old A. thaliana seedlings triggered an upregulation of MSH6 in shoot and root apical meristems. Appropriately, MSH6 was also induced by the stem cell inducer WUSCHEL. Further, the stimulatory effect of light was dependent on the presence of phyA. In addition, treatment of seedlings with auxin or cytokinin also caused an upregulation of MSH6 under darkness. Consistent with auxin signals, MSH6 expression was suppressed in the GATA23 RNAi line compared with the wild type. Our results provide evidence that endogenous factors and environmental signals controlling plant growth and development regulate the MSH6 protein in A. thaliana.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/genetics , Arabidopsis/metabolism , DNA Mismatch Repair/genetics , Phylogeny , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Sugars , Indoleacetic Acids , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism
3.
BMC Gastroenterol ; 24(1): 82, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38395750

ABSTRACT

BACKGROUND: Deficient DNA mismatch repair (MMR) can cause microsatellite instability (MSI) and is more common in colorectal cancer (CRC) patients. Understanding the carcinogenic mechanism of bacteria and their impact on cancer cells is crucial. Bacteroides fragilis (B. fragilis) has been identified as a potential promoter of tumorigenesis through the alteration of signaling pathways. This study aims to assess the expression levels of msh2, msh6, mlh1, and the relative frequency of B. fragilis in biopsy samples from CRC patients. MATERIALS AND METHODS: Based on the sequence of mlh1, msh2, and msh6 genes, B. fragilis specific 16srRNA and bacterial universal 16srRNA specific primers were selected, and the expression levels of the target genes were analyzed using the Real-Time PCR method. RESULTS: Significant increases in the expression levels of mlh1, msh2, and msh6 genes were observed in the cancer group. Additionally, the expression of these MMR genes showed a significant elevation in samples positive for B. fragilis presence. The relative frequency of B. fragilis in the cancer group demonstrated a significant rise compared to the control group. CONCLUSION: The findings suggest a potential correlation between the abundance of B. fragilis and alterations in the expression of MMR genes. Since these genes can play a role in modifying colon cancer, investigating microbial characteristics and gene expression changes in CRC could offer a viable solution for CRC diagnosis.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Colorectal Neoplasms , Humans , DNA Mismatch Repair/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Bacteroides fragilis/genetics , Bacteroides fragilis/metabolism , Iran , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Microsatellite Instability , DNA-Binding Proteins/genetics , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , Biopsy
4.
Neoplasia ; 49: 100970, 2024 03.
Article in English | MEDLINE | ID: mdl-38281411

ABSTRACT

The maintenance of DNA sequence integrity is critical to avoid accumulation of cancer-causing mutations. Inactivation of DNA Mismatch Repair (MMR) genes (e.g., MLH1 and MSH2) is common among many cancers, including colorectal cancer (CRC) and is the driver of classic microsatellite instability (MSI) in tumors. Somatic MSH3 alterations have been linked to a specific form of MSI called elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) that is associated with patient poor prognosis and elevated among African American (AA) rectal cancer patients. Genetic variants of MSH3 and their pathogenicity vary among different populations, such as among AA, which are not well-represented in publicly available databases. Targeted exome sequencing of MSH3 among AA CRC samples followed by computational bioinformatic pipeline and molecular dynamic simulation analysis approach confirmed six identified MSH3 variants (c.G1237A, c.C2759T, c.G1397A, c.G2926A, c.C3028T, c.G3241A) that corresponded to MSH3 amino-acid changes (p.E413K; p.S466N; p.S920F; p.E976K; p.H1010Y; p.E1081K). All identified MSH3 variants were non-synonymous, novel, pathogenic, and show loss or gain of hydrogen bonding, ionic bonding, hydrophobic bonding, and disulfide bonding and have a deleterious effect on the structure of MSH3 protein. Some variants were located within the ATPase site of MSH3, affecting ATP hydrolysis that is critical for MSH3's function. Other variants were in the MSH3-MSH2 interacting domain, important for MSH3's binding to MSH2. Overall, our data suggest that these variants among AA CRC patients affect the function of MSH3 making them pathogenic and likely contributing to the development or advancement of CRC among AA. Further clarifying functional studies will be necessary to fully understand the impact of these variants on MSH3 function and CRC development in AA patients.


Subject(s)
Black or African American , Colorectal Neoplasms , Humans , Black or African American/genetics , Colorectal Neoplasms/ethnology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , High-Throughput Nucleotide Sequencing , Microsatellite Instability , Microsatellite Repeats , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , MutS Homolog 3 Protein/genetics , MutS Homolog 3 Protein/metabolism , Virulence
5.
J Surg Oncol ; 129(5): 876-884, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38173349

ABSTRACT

The aim of the study was to determine DNA mismatch repair (MMR) proteins by immunohistochemically using MLH1, MSH2, MSH6, and PMS2 antibodies in patients diagnosed as pancreatic ductal adenocarcinoma and to assess its relationship with histopathological and clinical prognostic parameters. Fifty cases with a diagnosis of pancreatic ductal adenocarcinoma who underwent surgical resection, were included in the study. Demographic and histopathological features of the patients were collected from the medical records. The relationships between microsatellite status and prognostic parameters were determined. The mean age of the patients was 66.5 ± 9.5 years (range: 47-87) and male/female ratio was 1.63 (31/19). No errors were detected in DNA MMR proteins in any of the cases, and were classified as microsatellite stable. The mean tumor diameter was 4.01 ± 1.77 cm and 74% of the tumors were localized in the pancreatic head. All of the cases had lymphatic invasion, whereas vascular invasion was detected in only 78% and perineural invasion in 98% of the patients. When the relationship between prognostic parameters and survival was evaluated, statistically significant correlation was observed in patient age and histopathological parameters such as tumor diameter, status of surgical margins, and vascular invasion (p < 0.05). Age, tumor size, presence of tumor at surgical margins, vascular invasion, and adjuvant treatment were correlated with survival. Although microsatellite instability was not detected in our cases, it is important to determine the microsatellite status by immunohistochemistry for predicting the chemotherapy response and determining the immunotherapy option in pancreatic adenocarcinomas.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Male , Female , Middle Aged , Aged , Aged, 80 and over , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Adenocarcinoma/metabolism , Prognosis , DNA Mismatch Repair , Margins of Excision , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/therapy , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism
6.
Fam Cancer ; 23(1): 1-7, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37957483

ABSTRACT

Lynch syndrome is an autosomal dominant disorder that usually results from a pathogenic germline variant in one of four genes (MSH2, MSH6, MLH1, PMS2) involved in DNA mismatch repair. Carriers of such variants are at risk of developing numerous cancers during adulthood. Here we report on a family suspected of having Lynch syndrome due to a history of endometrial adenocarcinoma, ovarian clear cell carcinoma, and adenocarcinoma of the duodenum in whom we identified a germline 29 nucleotide in-frame inversion in exon 3 of MSH2. We further show that this variant is almost completely absent at the protein level, and that the associated cancers have complete loss of MSH2 and MSH6 expression by immunohistochemistry. Functional investigation of this inversion in a laboratory setting revealed a resultant abnormal protein function. Thus, we have identified an unusual, small germline inversion in a mismatch repair gene that does not lead to a premature stop codon yet appears likely to be causal for the observed cancers.


Subject(s)
Adenocarcinoma , Colorectal Neoplasms, Hereditary Nonpolyposis , Humans , Adult , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Germ-Line Mutation , Adenocarcinoma/genetics , Exons , DNA Mismatch Repair/genetics , MutL Protein Homolog 1/genetics , Mismatch Repair Endonuclease PMS2/genetics , Mismatch Repair Endonuclease PMS2/metabolism
7.
Nucleic Acids Res ; 51(22): 12185-12206, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-37930834

ABSTRACT

The Msh2-Msh3 mismatch repair (MMR) complex in Saccharomyces cerevisiae recognizes and directs repair of insertion/deletion loops (IDLs) up to ∼17 nucleotides. Msh2-Msh3 also recognizes and binds distinct looped and branched DNA structures with varying affinities, thereby contributing to genome stability outside post-replicative MMR through homologous recombination, double-strand break repair (DSBR) and the DNA damage response. In contrast, Msh2-Msh3 promotes genome instability through trinucleotide repeat (TNR) expansions, presumably by binding structures that form from single-stranded (ss) TNR sequences. We previously demonstrated that Msh2-Msh3 binding to 5' ssDNA flap structures interfered with Rad27 (Fen1 in humans)-mediated Okazaki fragment maturation (OFM) in vitro. Here we demonstrate that elevated Msh2-Msh3 levels interfere with DNA replication and base excision repair in vivo. Elevated Msh2-Msh3 also induced a cell cycle arrest that was dependent on RAD9 and ELG1 and led to PCNA modification. These phenotypes also required Msh2-Msh3 ATPase activity and downstream MMR proteins, indicating an active mechanism that is not simply a result of Msh2-Msh3 DNA-binding activity. This study provides new mechanistic details regarding how excess Msh2-Msh3 can disrupt DNA replication and repair and highlights the role of Msh2-Msh3 protein abundance in Msh2-Msh3-mediated genomic instability.


Subject(s)
Genomic Instability , Saccharomyces cerevisiae Proteins , Humans , DNA/genetics , DNA/metabolism , DNA Mismatch Repair , DNA Repair , DNA-Binding Proteins/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , MutS Homolog 3 Protein/genetics , MutS Homolog 3 Protein/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
8.
Aging (Albany NY) ; 15(22): 13558-13578, 2023 11 27.
Article in English | MEDLINE | ID: mdl-38016120

ABSTRACT

Conserved long non-coding RNAs (lncRNAs) have not thoroughly been studied in many cancers, including gastric cancer (GC). We have identified a novel lncRNA PTCHD4-AS which was highly conserved between humans and mice and naturally downregulated in GC cell lines and tissues. Notably, PTCHD4-AS was found to be transcriptionally induced by DNA damage agents and its upregulation led to cell cycle arrest at the G2/M phase, in parallel, it facilitated the cell apoptosis induced by cisplatin (CDDP) in GC. Mechanistically, PTCHD4-AS directly bound to the DNA mismatch repair protein MSH2-MSH6 dimer, and facilitated the binding of dimer to ATM, thereby promoting the expression of phosphorylated ATM, p53 and p21. Here we conclude that the upregulation of PTCHD4-AS inhibits proliferation and increases CDDP sensitivity of GC cells via binding with MSH2-MSH6 dimer, activating the ATM-p53-p21 pathway.


Subject(s)
Stomach Neoplasms , Tumor Suppressor Protein p53 , Mice , Humans , Animals , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Dimerization , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Cisplatin/pharmacology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism
9.
BMC Cancer ; 23(1): 940, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37798659

ABSTRACT

BACKGROUND: This study aimed to identify patients with upper urinary tract urothelial carcinoma (UTUC) having potential Lynch syndrome (pLS) by immunohistochemistry (IHC) of DNA mismatch repair gene-related proteins (MMRPs) and Amsterdam criteria II and explore their clinical characteristics. METHODS: We retrospectively collected the clinical data of 150 consecutive patients with UTUC who underwent surgical resection at our institution between February 2012 and December 2020, and immunohistochemistry (IHC) of four MMRPs (MLH1, MSH2, MSH6, and PMS2) on all UTUC specimens was performed. Patients who tested positive for Amsterdam criteria (AMS) II and/or IHC screening were classified as having pLS and others as non-pLS, and their characteristics were explored. RESULTS: In this study, 5 (3%) and 6 (4%) patients were positive for AMS II and IHC screening, respectively. Two patient were positive for both AMS II and IHC screening, resulting in 9 (6%) patients with pLS. The pLS group was predominantly female (67% vs. 36%; p = 0.0093) and had more right-sided tumors (100% vs. 43%; p = 0.0009) than the non-pLS group. Of the 6 patients who were positive for IHC screening, 4 showed a combined loss of MSH2/MSH6 (n = 3) and MLH1/PMS2 (n = 1). Other two patients showed single loss of MSH6 and PSM2. CONCLUSIONS: AMS II and IHC screening identified pLS in 6% of patients with UTUC. The IHC screening-positive group tends to have relatively high rate of combined loss, but some patients have single loss. AMS II may overlook patients with LS, and a universal screening may be required for patients with UTUC as well as those with colorectal and endometrial cancer.


Subject(s)
Carcinoma, Transitional Cell , Colorectal Neoplasms, Hereditary Nonpolyposis , Kidney Neoplasms , Ureteral Neoplasms , Urinary Bladder Neoplasms , Urinary Tract , Humans , Female , Male , Carcinoma, Transitional Cell/diagnosis , Carcinoma, Transitional Cell/epidemiology , Carcinoma, Transitional Cell/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/epidemiology , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Mismatch Repair Endonuclease PMS2/genetics , Mismatch Repair Endonuclease PMS2/metabolism , Retrospective Studies , Prevalence , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , Ureteral Neoplasms/diagnosis , Ureteral Neoplasms/epidemiology , Urinary Tract/metabolism , Urinary Tract/pathology , DNA Mismatch Repair
10.
Gene ; 888: 147746, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-37657688

ABSTRACT

Cervical cancer (CACX) is one of the top causes of cancer death in women globally. The involvement of several cellular pathways in carcinogenesis is still poorly understood. Here, we focused to evaluate the contributory role of Mismatch Repair (MMR) pathway genes-MLH1 and MSH2 in CACX and their association with chemo-tolerance of the disease. For this purpose, molecular profiles (expression/promoter methylation/deletion) of the genes were analysed in both normal cervical epithelium and tumour tissue, also validated in in-silico dataset as well. Later on, prognostic importance of the genes was identified through analysis of their methylation/expression status in plasma DNA of circulating tumour cells (CTCs) and cisplatin-tolerant CACX cell lines respectively. It was found that the expression profile of MLH1 and MSH2 genes was considerably reduced from undifferentiated basal-parabasal layers of normal cervical epithelium towards progression of the disease. Further analysis showed that frequent deletion [34-48%] and promoter methylation events [28-46%] of the genes were the plausible reasons for their reduced expression during tumorigenesis. Incidentally, the prevalence of MLH1 [32%] and MSH2 [27%] promoter methylation found in CTCs of plasma of the clinically advanced CACX patients implicated their prognostic importance of the disease. In addition, the patients having high alterations of those genes resulted in poor patient outcomes even after the therapy. In in-depth analysis of this result in cisplatin-tolerant CACX cell lines, we discovered that increased promoter methylation frequency of those genes at higher concentrations of cisplatin and gradual accumulation of the cells in the G2/M phase of the cell cycle were the rational causes for their reduced expression and MMR deficiency in the system. Hence, it is possible to conclude that the gradual down-regulation of MLH1 and MSH2 proteins may be a key event for MMR pathway inactivation in CACX. This might also be associated with chemo-tolerance and overall poor survival among the patients.


Subject(s)
Uterine Cervical Neoplasms , Humans , Female , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Cisplatin/pharmacology , Cisplatin/therapeutic use , Cervix Uteri/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism
11.
Dis Model Mech ; 16(8)2023 08 01.
Article in English | MEDLINE | ID: mdl-37395714

ABSTRACT

ALDH1B1 expressed in the intestinal epithelium metabolises acetaldehyde to acetate, protecting against acetaldehyde-induced DNA damage. MSH2 is a key component of the DNA mismatch repair (MMR) pathway involved in Lynch syndrome (LS)-associated colorectal cancers. Here, we show that defective MMR (dMMR) interacts with acetaldehyde, in a gene/environment interaction, enhancing dMMR-driven colonic tumour formation in a LS murine model of Msh2 conditional inactivation (Lgr5-CreER; Msh2flox/-, or Msh2-LS) combined with Aldh1b1 inactivation. Conditional (Aldh1b1flox/flox) or constitutive (Aldh1b1-/-) Aldh1b1 knockout alleles combined with the conditional Msh2flox/- intestinal knockout mouse model of LS (Msh2-LS) received either ethanol, which is metabolised to acetaldehyde, or water. We demonstrated that 41.7% of ethanol-treated Aldh1b1flox/flox Msh2-LS mice and 66.7% of Aldh1b1-/- Msh2-LS mice developed colonic epithelial hyperproliferation and adenoma formation, in 4.5 and 6 months, respectively, significantly greater than 0% in water-treated control mice. Significantly higher numbers of dMMR colonic crypt foci precursors and increased plasma acetaldehyde levels were observed in ethanol-treated Aldh1b1flox/flox Msh2-LS and Aldh1b1-/- Msh2-LS mice compared with those in water-treated controls. Hence, ALDH1B1 loss increases acetaldehyde levels and DNA damage that interacts with dMMR to accelerate colonic, but not small intestinal, tumour formation.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms, Hereditary Nonpolyposis , Colorectal Neoplasms , Animals , Mice , Acetaldehyde , Colorectal Neoplasms/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Mismatch Repair , Ethanol , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism
12.
Int J Mol Sci ; 24(13)2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37445734

ABSTRACT

Mismatch repair (MMR) is a conserved mechanism that is primarily responsible for the repair of DNA mismatches during DNA replication. Msh2 forms MutS heterodimer complexes that initiate the MMR in eukaryotes. The function of Msh2 is less clear under different chromatin structures. Tetrahymena thermophila contains a transcriptionally active macronucleus (MAC) and a transcriptionally silent micronucleus (MIC) in the same cytoplasm. Msh2 is localized in the MAC and MIC during vegetative growth. Msh2 is localized in the perinuclear region around the MIC and forms a spindle-like structure as the MIC divides. During the early conjugation stage, Msh2 is localized in the MIC and disappears from the parental MAC. Msh2 is localized in the new MAC and new MIC during the late conjugation stage. Msh2 also forms a spindle-like structure with a meiotic MIC and mitotic gametic nucleus. MSH2 knockdown inhibits the division of MAC and MIC during vegetative growth and affects cellular proliferation. MSH2 knockdown mutants are sensitive to cisplatin treatment. MSH2 knockdown also affects micronuclear meiosis and gametogenesis during sexual development. Furthermore, Msh2 interacts with MMR-dependent and MMR-independent factors. Therefore, Msh2 is necessary for macronuclear stability, as well as micronuclear mitosis and meiosis in Tetrahymena.


Subject(s)
Tetrahymena thermophila , Tetrahymena thermophila/genetics , Tetrahymena thermophila/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , DNA Mismatch Repair , Cell Nucleus/metabolism , Macronucleus/genetics , Macronucleus/metabolism
13.
Nucleic Acids Res ; 51(12): 6307-6320, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37224528

ABSTRACT

DNA mismatch repair (MMR) is essential for correction of DNA replication errors. Germline mutations of the human MMR gene MLH1 are the major cause of Lynch syndrome, a heritable cancer predisposition. In the MLH1 protein, a non-conserved, intrinsically disordered region connects two conserved, catalytically active structured domains of MLH1. This region has as yet been regarded as a flexible spacer, and missense alterations in this region have been considered non-pathogenic. However, we have identified and investigated a small motif (ConMot) in this linker which is conserved in eukaryotes. Deletion of the ConMot or scrambling of the motif abolished mismatch repair activity. A mutation from a cancer family within the motif (p.Arg385Pro) also inactivated MMR, suggesting that ConMot alterations can be causative for Lynch syndrome. Intriguingly, the mismatch repair defect of the ConMot variants could be restored by addition of a ConMot peptide containing the deleted sequence. This is the first instance of a DNA mismatch repair defect conferred by a mutation that can be overcome by addition of a small molecule. Based on the experimental data and AlphaFold2 predictions, we suggest that the ConMot may bind close to the C-terminal MLH1-PMS2 endonuclease and modulate its activation during the MMR process.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , DNA Mismatch Repair , MutL Protein Homolog 1 , Humans , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Genetic Predisposition to Disease , Germ-Line Mutation , Mutation , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
14.
Taiwan J Obstet Gynecol ; 62(3): 448-452, 2023 May.
Article in English | MEDLINE | ID: mdl-37188452

ABSTRACT

OBJECTIVE: We aimed to elucidate the pathogenesis of ovarian cancer through the loss of mismatch repair (MMR) proteins in women with Lynch syndrome (LS) in this report. CASE REPORT: Two women with LS underwent surgery for synchronous endometrial cancer and ovarian cancer. In both cases, immunohistochemical examination showed concomitant MMR protein deficiency in endometrial cancer, ovarian cancer, and contiguous ovarian endometriosis. In Case 1, the macroscopically normal ovary included multiple endometrioses with MSH2 and MSH6 expression, and FIGO grade 1 endometrioid carcinoma and contiguous endometriosis without MSH2 and MSH6 expression. In Case 2, all endometriotic cells contiguous with carcinoma in the lumen of the ovarian cyst showed loss of the expression of MSH2 and MSH6. CONCLUSION: Ovarian endometriosis with MMR protein deficiency may progress to endometriosis-associated ovarian cancer in women with LS. Diagnosing endometriosis in women with LS during surveillance is important.


Subject(s)
Carcinoma, Endometrioid , Colorectal Neoplasms, Hereditary Nonpolyposis , Endometrial Neoplasms , Endometriosis , Ovarian Neoplasms , Protein Deficiency , Female , Humans , Endometriosis/complications , Colorectal Neoplasms, Hereditary Nonpolyposis/complications , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Mismatch Repair , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Ovarian Neoplasms/pathology , Carcinoma, Endometrioid/complications , Carcinoma, Endometrioid/pathology , Endometrial Neoplasms/complications , Endometrial Neoplasms/pathology , DNA-Binding Proteins/genetics
15.
Nucleic Acids Res ; 51(11): 5584-5602, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37140056

ABSTRACT

DNA double-strand break (DSB) repair via homologous recombination is initiated by end resection. The extent of DNA end resection determines the choice of the DSB repair pathway. Nucleases for end resection have been extensively studied. However, it is still unclear how the potential DNA structures generated by the initial short resection by MRE11-RAD50-NBS1 are recognized and recruit proteins, such as EXO1, to DSB sites to facilitate long-range resection. We found that the MSH2-MSH3 mismatch repair complex is recruited to DSB sites through interaction with the chromatin remodeling protein SMARCAD1. MSH2-MSH3 facilitates the recruitment of EXO1 for long-range resection and enhances its enzymatic activity. MSH2-MSH3 also inhibits access of POLθ, which promotes polymerase theta-mediated end-joining (TMEJ). Collectively, we present a direct role of MSH2-MSH3 in the initial stages of DSB repair by promoting end resection and influencing the DSB repair pathway by favoring homologous recombination over TMEJ.


Subject(s)
DNA Repair , Exodeoxyribonucleases , MutS Homolog 2 Protein , MutS Homolog 3 Protein , DNA/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Exodeoxyribonucleases/metabolism , Homologous Recombination , MutS Homolog 2 Protein/metabolism , Humans , Cell Line , DNA Helicases/metabolism , MutS Homolog 3 Protein/metabolism
16.
Plant Sci ; 332: 111713, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37068662

ABSTRACT

The MutS homolog 6 (MSH6) is a nuclear DNA mismatch repair (MMR) gene that encodes the MSH6 protein. MSH6 interacts with MSH2 to form the MutSα heterodimer. MutSα corrects DNA mismatches and unpaired nucleotides arising during DNA replication, deamination of 5-methylcytosine, and recombination between non-identical DNA sequences. In addition to correcting DNA biosynthetic errors, MutSα also recognizes chemically damaged DNA bases. Here, we show that inactivation of MSH6 affects the basal susceptibility of Arabidopsis thaliana to Pseudomonas syringae pv tomato DC3000. The msh6 T-DNA insertional mutant exhibited a reduced susceptibility to the bacterial invasion. This heightened basal resistance of msh6 mutants appears to be dependent on an increased stomatal closure, an accumulation of H2O2 and double-strand breaks (DSBs) and a constitutive expression of pathogenesis-related (NPR1 and PR1) and DNA damage response (RAD51D and SOG1) genes. Complementation of this mutant with the MSH6 wild type allele under the control of its own promoter resulted in reversal of the basal bacterial resistance phenotype and the stomatal closure back to wild type levels. Taken together, these results demonstrate that inactivation of MSH6 increases Arabidopsis basal susceptibility to the bacterial pathogen and suggests a link between DNA repair and stress signaling in plants.


Subject(s)
Arabidopsis Proteins , Arabidopsis , DNA-Binding Proteins , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , DNA , DNA Mismatch Repair , DNA-Binding Proteins/genetics , Hydrogen Peroxide , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Pseudomonas syringae/physiology , Repressor Proteins/metabolism , Transcription Factors/genetics
17.
Microb Genom ; 9(4)2023 04.
Article in English | MEDLINE | ID: mdl-37083479

ABSTRACT

Malarial parasites exhibit extensive genomic plasticity, which induces the antigen diversification and the development of antimalarial drug resistance. Only a few studies have examined the genome maintenance mechanisms of parasites. The study aimed at elucidating the impact of a mutation in a DNA mismatch repair gene on genome stability by maintaining the mutant and wild-type parasites through serial in vitro cultures for approximately 400 days and analysing the subsequent spontaneous mutations. A P513T mutant of the DNA mismatch repair protein PfMSH2-1 from Plasmodium falciparum 3D7 was created. The mutation did not influence the base substitution rate but significantly increased the insertion/deletion (indel) mutation rate in short tandem repeats (STRs) and minisatellite loci. STR mutability was affected by allele size, genomic category and certain repeat motifs. In the mutants, significant telomere healing and homologous recombination at chromosomal ends caused extensive gene loss and generation of chimeric genes, resulting in large-scale chromosomal alteration. Additionally, the mutant showed increased tolerance to N-methyl-N'-nitro-N-nitrosoguanidine, suggesting that PfMSH2-1 was involved in recognizing DNA methylation damage. This work provides valuable insights into the role of PfMSH2-1 in genome stability and demonstrates that the genomic destabilization caused by its dysfunction may lead to antigen diversification.


Subject(s)
Genomic Instability , Plasmodium falciparum , Humans , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Mutation , Phenotype
18.
Pathol Res Pract ; 244: 154390, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36905693

ABSTRACT

BACKGROUND: Microsatellite instability (MSI) represents as a molecular hallmark of deficient MMR system at the genomic level. Increasing clinical significance of MSI status highlights the necessity of simple, accurate markers for detection. Although 2B3D NCI panel is the most widely applied, it has been questioned whether the performance of NCI panel is second to none in MSI detection. METHODS: We evaluated the efficacy of the NCI panel versus a 6-mononucleotide site panel (BAT25, BAT26, NR21, NR24, NR27, and MONO-27) in assessing MSI status of 468 Chinese patients with CRC, and compared MSI test results with the results by immunohistochemistry of four MMR proteins (MLH1, PMS2, MSH2, MSH6) in the present study. Clinicopathological variables were also collected, and their associations with MSI or MMR proteins status were analyzed using either the chi-square test or the Fisher's exact test. RESULTS: MSI-H/dMMR was significantly associated with right colon involvement, poor differentiation, early stage, mucinous adenocarcinoma, negative lymph node, less neural invasion, and KRAS/NRAS/BRAF wild-type. As to the efficiency of detecting deficient MMR system, both panels had good concordance with MMR proteins expression by IHC, and 6-mononucleotide site panel outperformed NCI panel in sensitivity, specificity, positive predictive value, and negative predictive value numerically despite the lack of statistical significance. The advantage was more obvious in the sensitivity and specificity analyses of each single microsatellite markers from 6-mononucleotide site panel in comparison with NCI panel. Additionally, the rate of MSI-L detected by 6-mononucleotide site panel was much lower than that detected by the NCI panel (0.64% vs. 2.86%, P = 0.0326). CONCLUSION: 6-mononucleotide site panel had a greater ability to help resolve cases of MSI-L into either MSI-H or MSS. We propose that 6-mononucleotide site panel may be potentially more suitable than NCI panel for Chinese CRC population. Large-scale studies are warranted to validate our findings.


Subject(s)
Adenocarcinoma, Mucinous , Colorectal Neoplasms , Humans , Microsatellite Instability , DNA Mismatch Repair/genetics , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Microsatellite Repeats , Colorectal Neoplasms/pathology , Adenocarcinoma, Mucinous/genetics
19.
J Int Med Res ; 51(3): 3000605231163780, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36994850

ABSTRACT

OBJECTIVE: To explore underlying mechanisms that regulate hMSH2 expression and drug susceptibility in epithelial ovarian cancer (EOC). METHODS: Using data from the Cancer Genome Atlas (TCGA) we used bioinformatical analysis to predict transcription factors (TFs) that potentially regulate hMSH2. RT-qPCR, Western blot, and luciferase assays were undertaken using ovarian cancer cell lines to verify the identified TF. Expressions of the TF were modulated using overexpression or knockdown, and the corresponding cellular responses to cisplatin were examined. RESULTS: The TF, E2F1, was found to regulate the hMSH2 gene. The expression level of E2F1 correlated with cisplatin susceptibility in vitro. Kaplan-Meier analysis of 77 patients with EOC showed that low E2F1 expression was associated with worse survival. CONCLUSIONS: To our knowledge, this is the first report of E2F1 regulated MSH2 expression playing a role in drug resistance of platinum-based treatments for patients with EOC. Further work is need to confirm our results.


Subject(s)
Carcinoma, Ovarian Epithelial , Cisplatin , E2F1 Transcription Factor , MutS Homolog 2 Protein , Ovarian Neoplasms , Female , Humans , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Kaplan-Meier Estimate , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Platinum/pharmacology , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism
20.
Eur J Cancer Prev ; 32(3): 286-297, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36896836

ABSTRACT

BACKGROUND: There is a lack of information on rare germline variants of pancreatic cancer-predisposing genes. Risk genes for multiple primary cancers may overlap with those for pancreatic cancer. METHODS: A retrospective study of autopsy cases with a negative family history in the Japanese single nucleotide polymorphism for geriatric research database examined rare germline variants in the protein-coding regions of 61 genes. Targeted sequencing of these genes was performed and classified for pathogenicity using the American College of Medical Genetics and Genomics guidelines. Polyphen-2, SIFT and LoFtool algorithms were used to predict damage to protein function. RESULTS: Of the 189 subjects used (90 cancer and 99 non-cancer controls), 72 patients had pancreatic cancer (23 had multiple primary cancers) and 18 had no pancreatic cancer in multiple primary cancers. APC, BRCA2, BUB1B, ENG and MSH6 were associated with cancer predisposition, and pathogenic/likely pathogenic (P/LP) variants occurred in 6% [pancreatic cancer (4/72); all-cancer (5/90)] and 54% (49/90) carried only variants of uncertain significance (VUS) among cancer patients. Of these VUS, in pancreatic cancer patients, four DNA mismatch repair (MMR) genes ( MLH1, MSH2, MSH6 and PMS2 ), and POLQ in men were significantly associated (odds ratio = 3.83; P = 0.025; P = 0.027, respectively). The most abundant predictor of functionally damaging variants was POLQ . CONCLUSIONS: The frequency of P/LP variants in patients with sporadic pancreatic cancer suggests the need for genetic evaluation of individuals with no family history. VUS of MMR genes ( MLH1, MSH2, MSH6 and PMS2 ) and POLQ may be useful in predicting genetic trends in the potential risk of pancreatic cancer, especially in individuals lacking P/LP.


Subject(s)
Neoplasms, Multiple Primary , Pancreatic Neoplasms , Male , Humans , Aged , Mismatch Repair Endonuclease PMS2/genetics , Mismatch Repair Endonuclease PMS2/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Autopsy , Retrospective Studies , DNA-Binding Proteins/metabolism , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/genetics , DNA Mismatch Repair , Germ Cells/metabolism , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...