Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.191
Filter
1.
Front Immunol ; 15: 1455907, 2024.
Article in English | MEDLINE | ID: mdl-39247185

ABSTRACT

Pembrolizumab and other immunotherapies have become central in treating metastatic colon cancer, particularly effective in patients with mismatch repair deficiencies. We report a case involving a man who initially underwent radical surgery for sigmoid colon cancer on April 27, 2011, followed by hepatic tumor resection on September 21, 2017. Post-surgery, he received eight cycles of adjuvant chemotherapy with the CAPEOX regimen and was regularly monitored through CT and MRI scans. On August 24, 2022, liver metastases were detected, and he was diagnosed with Lynch syndrome (LS) due to germline mutation in the MSH2 and EPCAM genes. He commenced treatment with 200mg of pembrolizumab intravenously every three weeks on September 2, 2022, and demonstrated a sustained response. However, after 17 cycles, he developed a treatment related adverse event (TRAE) of pancreatic endocrine dysfunction, leading to type 1 diabetes, managed with subcutaneous insulin injections. After 30 cycles of treatment, no evidence of disease was observed. This case underscores the significant clinical benefits of first-line pembrolizumab in managing hepatic metastasis in colonic carcinoma associated with LS, despite the occurrence of TRAEs. It raises critical questions regarding the optimal duration of immunotherapy following a complete or partial response and whether treatment should be discontinued upon the emergency of TRAEs. Continued research and forthcoming clinical trials with checkpoint inhibitors are expected to refine treatment protocols for LS-associated carcinoma.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological , Colorectal Neoplasms, Hereditary Nonpolyposis , Liver Neoplasms , Humans , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Male , Colorectal Neoplasms, Hereditary Nonpolyposis/drug therapy , Liver Neoplasms/secondary , Liver Neoplasms/drug therapy , Antineoplastic Agents, Immunological/therapeutic use , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Middle Aged , Treatment Outcome , MutS Homolog 2 Protein/genetics , Epithelial Cell Adhesion Molecule/genetics
2.
Mol Cell ; 84(16): 3044-3060.e11, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39142279

ABSTRACT

G-quadruplexes (G4s) form throughout the genome and influence important cellular processes. Their deregulation can challenge DNA replication fork progression and threaten genome stability. Here, we demonstrate an unexpected role for the double-stranded DNA (dsDNA) translocase helicase-like transcription factor (HLTF) in responding to G4s. We show that HLTF, which is enriched at G4s in the human genome, can directly unfold G4s in vitro and uses this ATP-dependent translocase function to suppress G4 accumulation throughout the cell cycle. Additionally, MSH2 (a component of MutS heterodimers that bind G4s) and HLTF act synergistically to suppress G4 accumulation, restrict alternative lengthening of telomeres, and promote resistance to G4-stabilizing drugs. In a discrete but complementary role, HLTF restrains DNA synthesis when G4s are stabilized by suppressing primase-polymerase (PrimPol)-dependent repriming. Together, the distinct roles of HLTF in the G4 response prevent DNA damage and potentially mutagenic replication to safeguard genome stability.


Subject(s)
DNA Primase , DNA Replication , DNA-Binding Proteins , G-Quadruplexes , Genomic Instability , MutS Homolog 2 Protein , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , MutS Homolog 2 Protein/metabolism , MutS Homolog 2 Protein/genetics , DNA Primase/metabolism , DNA Primase/genetics , Telomere Homeostasis , DNA Damage , HEK293 Cells , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , DNA-Directed DNA Polymerase
3.
J Med Genet ; 61(9): 861-869, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39004446

ABSTRACT

BackgroundLynch syndrome (LS) is an inherited cancer predisposition syndrome caused by genetic variants affecting DNA mismatch repair (MMR) genes MLH1, MSH2, MSH6 and PMS2 Cancer risk in LS is estimated from cohorts of individuals ascertained by individual or family history of cancer, which may upwardly bias estimates. METHODS: 830 carriers of pathogenic or likely pathogenic (path_MMR) MMR gene variants classified by InSiGHT were identified in 454 756 UK Biobank (UKB) participants using whole-exome sequence. Nelson-Aalen survival analysis was used to estimate cumulative incidence of colorectal, endometrial and breast cancer (BC). RESULTS: Cumulative incidence of colorectal and endometrial cancer (EC) by age 70 years was elevated in path_MMR carriers compared with non-carriers (colorectal: 11.8% (95% confidence interval (CI): 9.5% to 14.6%) vs 1.7% (95% CI: 1.6% to 1.7%), endometrial: 13.4% (95% CI: 10.2% to 17.6%) vs 1.0% (95% CI: 0.9% to 1.0%)), but the magnitude of this increase differed between genes. Cumulative BC incidence by age 70 years was not elevated in path_MMR carriers compared with non-carriers (8.9% (95% CI: 6.3% to 12.4%) vs 7.5% (95% CI: 7.4% to 7.6%)). Cumulative cancer incidence estimates in UKB were similar to estimates from the Prospective Lynch Syndrome Database for all genes and cancers, except there was no evidence for elevated EC risk in carriers of pathogenic PMS2 variants in UKB. CONCLUSION: These results support offering incidentally identified carriers of any path_MMR surveillance to manage colorectal cancer risk. Incidentally identified carriers of pathogenic variants in MLH1, MSH2 and MSH6 would also benefit from interventions to reduce EC risk. The results suggest that BC is not an LS-related cancer.


Subject(s)
Biological Specimen Banks , Colorectal Neoplasms, Hereditary Nonpolyposis , DNA Mismatch Repair , Genetic Predisposition to Disease , Humans , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/epidemiology , Female , United Kingdom/epidemiology , Male , Middle Aged , DNA Mismatch Repair/genetics , Aged , Heterozygote , Exome Sequencing , Incidence , Adult , Breast Neoplasms/genetics , Breast Neoplasms/epidemiology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/epidemiology , DNA-Binding Proteins/genetics , MutL Protein Homolog 1/genetics , UK Biobank , MutS Homolog 2 Protein
4.
Mol Syst Biol ; 20(9): 1006-1024, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39026103

ABSTRACT

Somatic hypermutation in cancer has gained momentum with the increased use of tumour mutation burden as a biomarker for immune checkpoint inhibitors. Spontaneous deamination of 5-methylcytosine to thymine at CpG dinucleotides is one of the most ubiquitous endogenous mutational processes in normal and cancer cells. Here, we performed a systematic investigation of somatic CpG hypermutation at a pan-cancer level. We studied 30,191 cancer patients and 103 cancer types and developed an algorithm to identify somatic CpG hypermutation. Across cancer types, we observed the highest prevalence in paediatric leukaemia (3.5%), paediatric high-grade glioma (1.7%), and colorectal cancer (1%). We discovered germline variants and somatic mutations in the mismatch repair complex MutSα (MSH2-MSH6) as genetic drivers of somatic CpG hypermutation in cancer, which frequently converged on CpG sites and TP53 driver mutations. We further observe an association between somatic CpG hypermutation and response to immune checkpoint inhibitors. Overall, our study identified novel cancer types that display somatic CpG hypermutation, strong association with MutSα-deficiency, and potential utility in cancer immunotherapy.


Subject(s)
CpG Islands , DNA Mismatch Repair , Mutation , Neoplasms , Humans , DNA Mismatch Repair/genetics , Neoplasms/genetics , Neoplasms/immunology , CpG Islands/genetics , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , DNA-Binding Proteins/genetics , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Tumor Suppressor Protein p53/genetics
5.
Nucleic Acids Res ; 52(16): 9574-9585, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39016170

ABSTRACT

The endonuclease activity of Pms1 directs mismatch repair by generating a nick in the newly replicated DNA strand. Inactivating Pms2, the human homologue of yeast Pms1, increases the chances of colorectal and uterine cancers. Here we use whole genome sequencing to show that loss of this endonuclease activity, via the pms1-DE variant, results in strong mutator effects throughout the Saccharomyces cerevisiae genome. Mutation rates are strongly increased for mutations resulting from all types of single-base substitutions and for a wide variety of single- and multi-base indel mutations. Rates for these events are further increased in strains combining pms1-DE with mutator variants of each of the three major leading and lagging strand replicases. In all cases, mutation rates, spectra, biases, and context preferences are statistically indistinguishable from strains with equivalent polymerases but lacking initial mismatch recognition due to deletion of MSH2. This implies that, across the nuclear genome, strand discrimination via the Pms1 endonuclease is as important for MMR as is initial mismatch recognition by Msh2 heterodimers.


Subject(s)
DNA Mismatch Repair , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , DNA Mismatch Repair/genetics , Genome, Fungal , Genomic Instability/genetics , Mutation Rate , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Mutation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Whole Genome Sequencing , MutL Proteins
6.
Int J Gynecol Cancer ; 34(7): 993-1000, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38950928

ABSTRACT

OBJECTIVE: Although early-detected cervical cancer is associated with good survival, the prognosis for late-stage disease is poor and treatment options are sparse. Mismatch repair deficiency (MMR-D) has surfaced as a predictor of prognosis and response to immune checkpoint inhibitor(s) in several cancer types, but its value in cervical cancer remains unclear. This study aimed to define the prevalence of MMR-D in cervical cancer and assess the prognostic value of MMR protein expression. METHODS: Expression of the MMR proteins MLH-1, PMS-2, MSH-2, and MSH-6 was investigated by immunohistochemical staining in a prospectively collected cervical cancer cohort (n=508) with corresponding clinicopathological and follow-up data. Sections were scored as either loss or intact expression to define MMR-D, and by a staining index, based on staining intensity and area, evaluating the prognostic potential. RNA and whole exome sequencing data were available for 72 and 75 of the patients and were used for gene set enrichment and mutational analyses, respectively. RESULTS: Five (1%) tumors were MMR-deficient, three of which were of neuroendocrine histology. MMR status did not predict survival (HR 1.93, p=0.17). MSH-2 low (n=48) was associated with poor survival (HR 1.94, p=0.02), also when adjusting for tumor stage, tumor type, and patient age (HR 2.06, p=0.013). MSH-2 low tumors had higher tumor mutational burden (p=0.003) and higher frequency of (frameshift) mutations in the double-strand break repair gene RAD50 (p<0.01). CONCLUSION: MMR-D is rare in cervical cancer, yet low MSH-2 expression is an independent predictor of poor survival.


Subject(s)
DNA Mismatch Repair , DNA-Binding Proteins , MutS Homolog 2 Protein , Uterine Cervical Neoplasms , Humans , Female , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/mortality , Prognosis , Middle Aged , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , MutS Homolog 2 Protein/metabolism , MutS Homolog 2 Protein/biosynthesis , MutS Homolog 2 Protein/genetics , Adult , Aged , Mismatch Repair Endonuclease PMS2/metabolism , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/metabolism , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/biosynthesis
7.
Genes (Basel) ; 15(7)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39062638

ABSTRACT

BACKGROUND: Lynch syndrome (LS) is a genetic disease with increased risk of colorectal cancer and other malignancies. There are few reported cases of thyroid cancer in LS patients. The aim of this study is to investigate the presence of thyroid nodules in LS patients and to explore their association with the genetic features of the disease. METHODS: A retrospective and descriptive analysis was conducted to include all LS patients followed at the CEMAD (Centro Malattie Apparato Digerente) of Fondazione Policlinico Universitario A. Gemelli IRCCS. The characteristics of LS disease, gene mutations, and previous history of thyroid disease were evaluated. Majority of patients underwent thyroid ultrasound (US), and nodule cytology was performed when needed. RESULTS: Of a total of 139 patients with LS, 110 patients were included in the study. A total of 103 patients (74%) underwent thyroid ultrasound examinations, and 7 patients (5%) had a previous history of thyroid disease (cancer or multinodular goiter). The mean age was 51.9 years. Thyroid nodules were found in 62 patients (60%) who underwent US, and 9 of them (14%) had suspicious features of malignancy, inducing a fine-needle aspiration biopsy. A cytologic analysis classified 7 of 9 cases (78%) as TIR2 and 2 (22%) as TIR3a. Between patients with nodular thyroid disease (single nodule, multinodular goiter, and cancer), most of them (25 patients, 36% of total) were carriers of the MSH6 mutation, while 22 (32%), 17 (24%), and 5 (7%) had MSH2, MLH1, and PMS2 mutations, respectively. CONCLUSIONS: A high prevalence of thyroid nodules was found in patients with LS, especially in MSH6-carrying patients. Performing at least one thyroid ultrasound examination is suggested for the detection of nodular thyroid disease in LS patients. Systematic investigations are needed to estimate their prevalence, features, and risk of malignant transformation.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Thyroid Nodule , Humans , Thyroid Nodule/genetics , Thyroid Nodule/pathology , Thyroid Nodule/diagnostic imaging , Middle Aged , Female , Male , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnostic imaging , Adult , Retrospective Studies , Aged , MutL Protein Homolog 1/genetics , Ultrasonography , MutS Homolog 2 Protein/genetics , Mutation
8.
J Mol Diagn ; 26(9): 805-814, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38925454

ABSTRACT

Replication-coupled gene editing using locked nucleic acid-modified single-stranded DNA oligonucleotides (LMOs) can genetically engineer mammalian cells with high precision at single nucleotide resolution. Based on this method, oligonucleotide-directed mutation screening (ODMS) was developed to determine whether variants of uncertain clinical significance of DNA mismatch repair (MMR) genes can cause Lynch syndrome. In ODMS, the appearance of 6-thioguanine-resistant colonies upon introduction of the variant is indicative for defective MMR and hence pathogenicity. Whereas mouse embryonic stem cells (mESCs) hemizygous for MMR genes were used previously, we now show that ODMS can also be applied in wild-type mESCs carrying two functional alleles of each MMR gene. 6-Thioguanine resistance can result from two possible events: first, the mutation is present in only one allele, which is indicative for dominant-negative activity of the variant; and second, both alleles contain the planned modification, which is indicative for a regular loss-of-function variant. Thus, ODMS in wild-type mESCs can discriminate fully disruptive and dominant-negative MMR variants. The feasibility of biallelic targeting suggests that the efficiency of LMO-mediated gene targeting at a nonselectable locus may be enriched in cells that had undergone a simultaneous selectable LMO targeting event. This turned out to be the case and provided a protocol to improve recovery of LMO-mediated gene modification events.


Subject(s)
Alleles , DNA Mismatch Repair , Gene Editing , DNA Mismatch Repair/genetics , Gene Editing/methods , Mice , Animals , Mouse Embryonic Stem Cells/metabolism , Humans , MutS Homolog 2 Protein/genetics , Loss of Function Mutation , DNA Replication/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , MutS Homolog 3 Protein/genetics , Mutation , Genes, Dominant , DNA-Binding Proteins
9.
J Gastroenterol ; 59(8): 699-708, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38902413

ABSTRACT

BACKGROUND: The optimal interval of colonoscopy (CS) surveillance in cases with Lynch syndrome (LS), and stratification according to the causative mismatch repair gene mutation, has received much attention. To verify a feasible and effective CS surveillance strategy, we investigated the colorectal cancer (CRC) incidence at different intervals and the characteristics of precancerous colorectal lesions of LS cases. METHODS: This retrospective multicenter study was conducted in Japan. CRCs and advanced adenomas (AAs) in 316 LS cases with germline pathogenic variants (path_) were analyzed according to the data of 1,756 registered CS. RESULTS: The mean time interval for advanced CRCs (ACs) detected via CS surveillance was 28.7 months (95% confidence interval: 13.8-43.5). The rate of AC detection within (2.1%) and beyond 2 years (8.7%) differed significantly (p = 0.0003). AAs accounted for 43%, 46%, and 41% of lesions < 10 mm in size in the MLH1-, MSH2-, and MSH6-groups, respectively. The lifetime incidence of metachronous CRCs requiring intestinal resection for path_MLH1, path_MSH2, and path_MSH6 cases was 34%, 23%, and 14% in these cases, respectively. The cumulative CRC incidence showed a trend towards a 10-year delay for path_MSH6 cases as compared with that for path_MLH1 and path_MSH2 cases. CONCLUSIONS: In cases with path_MLH1, path_MSH2, and path_MSH6, maintaining an appropriate CS surveillance interval of within 2 years is advisable to detect of the colorectal lesion amenable to endoscopic treatment. path_MSH6 cases could be stratified with path_MLH1 and MSH2 cases in terms of risk of metachronous CRC and age of onset.


Subject(s)
Adenoma , Colonoscopy , Colorectal Neoplasms, Hereditary Nonpolyposis , Colorectal Neoplasms , DNA Mismatch Repair , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Humans , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Male , Female , Middle Aged , Retrospective Studies , Japan/epidemiology , Adenoma/genetics , Adenoma/pathology , Adenoma/epidemiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/epidemiology , Aged , MutL Protein Homolog 1/genetics , Adult , DNA Mismatch Repair/genetics , MutS Homolog 2 Protein/genetics , Incidence , DNA-Binding Proteins/genetics , Germ-Line Mutation , Time Factors , Early Detection of Cancer/methods , East Asian People
10.
Cancer Treat Res Commun ; 40: 100826, 2024.
Article in English | MEDLINE | ID: mdl-38870667

ABSTRACT

BACKGROUND: Recently, some evidence emphasized the value of MSH2 and MSH6 inactivation and their hypermutation in predicting different cancers. The present consideration is to evaluate the value of MSH2 and MSH6 protein deficient studied by the immunohistochemistry (IHC) method and the tumor behaviors and aggressiveness in prostatic carcinoma. METHODS: This cross-sectional study was performed on 80 examples extricated from patients who endured prostate cancer and were planned for radical prostatectomy surgery. The expression levels of the genes were studied by IHC staining. RESULTS: The deficiency in MSH2 and MSH6 expression was revealed in 10.0 % and 11.3 % of patients respectively, while the reduction of simultaneous expression in two genes was found in 6.2 % of patients. In the two subgroups with and without MSH2 and/or MSH6 staining, there was no difference in patients' mean age and history of prostate cancer. There was also no difference in tumor-related behaviors including combined Gleason grade group, tumor stage, vascular invasion, perineural invasion, and prostatic capsular invasion between the groups with and without gene loss. CONCLUSION: The evaluation of the deficient rate of two genes among patients with prostate cancer to predict the tumor grade and its aggressive behavior needs further study in every population.


Subject(s)
DNA-Binding Proteins , Disease Progression , MutS Homolog 2 Protein , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Cross-Sectional Studies , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Aged , Middle Aged , Immunohistochemistry , Neoplasm Grading , Prostatectomy , Gene Expression Regulation, Neoplastic
11.
J Int Med Res ; 52(6): 3000605241259747, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38902203

ABSTRACT

BACKGROUND: Breast cancer, particularly triple-negative breast cancer (TNBC), poses a significant global health burden. Chemotherapy was the mainstay treatment for TNBC patients until immunotherapy was introduced. Studies indicate a noteworthy prevalence (0.2% to 18.6%) of mismatch repair protein (MMRP) deficiency in TNBC, with recent research highlighting the potential of immunotherapy for MMRP-deficient metastatic breast cancer. This study aims to identify MMRP deficiency in TNBC patients using immunohistochemistry. METHODS: A retrospective cohort study design was used and included TNBC patients treated between 2015 and 2021 at King Hussein Cancer Center. Immunohistochemistry was conducted to assess MMRP expression. RESULTS: Among 152 patients, 14 (9.2%) exhibited deficient MMR (dMMR). Loss of PMS2 expression was observed in 13 patients, 5 of whom showed loss of MLH1 expression. Loss of MSH6 and MSH2 expression was observed in one patient. The median follow-up duration was 44 (3-102) months. Despite the higher survival rate (80.8%, 5 years) of dMMR patients than of proficient MMR patients (62.3%), overall survival did not significantly differ between the two groups. CONCLUSION: Approximately 9% of TNBC patients exhibit dMMR. dMMR could be used to predict outcomes and identify patients with TNBC who may benefit from immunotherapy.


Subject(s)
DNA Mismatch Repair , DNA-Binding Proteins , Mismatch Repair Endonuclease PMS2 , Triple Negative Breast Neoplasms , Humans , Female , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Middle Aged , Adult , Retrospective Studies , Mismatch Repair Endonuclease PMS2/metabolism , Mismatch Repair Endonuclease PMS2/genetics , Aged , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , MutL Protein Homolog 1/metabolism , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Survival Rate , Immunohistochemistry , Aged, 80 and over , Prognosis
12.
PLoS One ; 19(6): e0304141, 2024.
Article in English | MEDLINE | ID: mdl-38843250

ABSTRACT

Lynch syndrome is caused by inactivating variants in DNA mismatch repair genes, namely MLH1, MSH2, MSH6 and PMS2. We have investigated five MLH1 and one MSH2 variants that we have identified in Turkish and Tunisian colorectal cancer patients. These variants comprised two small deletions causing frameshifts resulting in premature stops which could be classified pathogenic (MLH1 p.(His727Profs*57) and MSH2 p.(Thr788Asnfs*11)), but also two missense variants (MLH1 p.(Asn338Ser) and p.(Gly181Ser)) and two small, in-frame deletion variants (p.(Val647-Leu650del) and p.(Lys678_Cys680del)). For such small coding genetic variants, it is unclear if they are inactivating or not. We here provide clinical description of the variant carriers and their families, and we performed biochemical laboratory testing on the variant proteins to test if their stability or their MMR activity are compromised. Subsequently, we compared the results to in-silico predictions on structure and conservation. We demonstrate that neither missense alteration affected function, while both deletion variants caused a dramatic instability of the MLH1 protein, resulting in MMR deficiency. These results were consistent with the structural analyses that were performed. The study shows that knowledge of protein function may provide molecular explanations of results obtained with functional biochemical testing and can thereby, in conjunction with clinical information, elevate the evidential value and facilitate clinical management in affected families.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , DNA Mismatch Repair , MutL Protein Homolog 1 , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Humans , Male , MutL Protein Homolog 1/genetics , Female , DNA Mismatch Repair/genetics , Middle Aged , MutS Homolog 2 Protein/genetics , Adult , Tunisia , Pedigree , Turkey , Aged , Mutation, Missense
13.
Nat Commun ; 15(1): 5044, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890315

ABSTRACT

Homology-dependent targeted DNA integration, generally referred to as gene targeting, provides a powerful tool for precise genome modification; however, its fundamental mechanisms remain poorly understood in human cells. Here we reveal a noncanonical gene targeting mechanism that does not rely on the homologous recombination (HR) protein Rad51. This mechanism is suppressed by Rad52 inhibition, suggesting the involvement of single-strand annealing (SSA). The SSA-mediated gene targeting becomes prominent when DSB repair by HR or end-joining pathways is defective and does not require isogenic DNA, permitting 5% sequence divergence. Intriguingly, loss of Msh2, loss of BLM, and induction of a target-site DNA break all significantly and synergistically enhance SSA-mediated targeted integration. Most notably, SSA-mediated integration is cell cycle-independent, occurring in the G1 phase as well. Our findings provide unequivocal evidence for Rad51-independent targeted integration and unveil multiple mechanisms to regulate SSA-mediated targeted as well as random integration.


Subject(s)
Cell Cycle , Gene Targeting , MutS Homolog 2 Protein , Rad51 Recombinase , Rad52 DNA Repair and Recombination Protein , Humans , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Cell Cycle/genetics , MutS Homolog 2 Protein/metabolism , MutS Homolog 2 Protein/genetics , RecQ Helicases/metabolism , RecQ Helicases/genetics , Homologous Recombination , DNA Breaks, Double-Stranded , DNA Repair , DNA End-Joining Repair , G1 Phase/genetics
14.
Dermatol Online J ; 30(1)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38762859

ABSTRACT

Patients with Muir-Torre syndrome may have a systemic malignancy and a sebaceous neoplasm such as an adenoma, epithelioma, and/or carcinoma. The syndrome usually results from a germline mutation in one or more mismatch repair genes. Iatrogenic or acquired immunosuppression can promote the appearance of sebaceous tumors, either as an isolated event or as a feature of Muir-Torre syndrome and may unmask individuals genetically predisposed to the syndrome. Two iatrogenically immunosuppressed men with Muir-Torre syndrome features are described. Similar to these immunocompromised men, Muir-Torre syndrome-associated sebaceous neoplasms have occurred in solid organ transplant recipients, human immunodeficiency virus-infected individuals, and patients with chronic diseases who are treated with immunosuppressive agents. Muir-Torre syndrome-associated sebaceous neoplasms occur more frequently and earlier in kidney recipients, who are receiving more post-transplant immunosuppressive agents, than in liver recipients. The development of sebaceous neoplasms is decreased by replacing cyclosporine or tacrolimus with sirolimus or everolimus. Specific anti-cancer vaccines or checkpoint blockade immunotherapy may merit exploration for immune-interception of Muir-Torre syndrome-associated sebaceous neoplasms and syndrome-related visceral cancers. We suggest germline testing for genomic aberrations of mismatch repair genes should routinely be performed in all patients-both immunocompetent and immunosuppressed-who develop a Muir-Torre syndrome-associated sebaceous neoplasm.


Subject(s)
DNA Mismatch Repair , Germ-Line Mutation , Immunosuppressive Agents , Muir-Torre Syndrome , Sebaceous Gland Neoplasms , Humans , Muir-Torre Syndrome/genetics , Male , DNA Mismatch Repair/genetics , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/adverse effects , Sebaceous Gland Neoplasms/genetics , Middle Aged , MutS Homolog 2 Protein/genetics , Immunocompromised Host , MutL Protein Homolog 1/genetics , Skin Neoplasms/genetics , DNA Mutational Analysis
15.
Am J Hum Genet ; 111(6): 1165-1183, 2024 06 06.
Article in English | MEDLINE | ID: mdl-38749429

ABSTRACT

The pathological huntingtin (HTT) trinucleotide repeat underlying Huntington disease (HD) continues to expand throughout life. Repeat length correlates both with earlier age at onset (AaO) and faster progression, making slowing its expansion an attractive therapeutic approach. Genome-wide association studies have identified candidate variants associated with altered AaO and progression, with many found in DNA mismatch repair (MMR)-associated genes. We examine whether lowering expression of these genes affects the rate of repeat expansion in human ex vivo models using HD iPSCs and HD iPSC-derived striatal medium spiny neuron-enriched cultures. We have generated a stable CRISPR interference HD iPSC line in which we can specifically and efficiently lower gene expression from a donor carrying over 125 CAG repeats. Lowering expression of each member of the MMR complexes MutS (MSH2, MSH3, and MSH6), MutL (MLH1, PMS1, PMS2, and MLH3), and LIG1 resulted in characteristic MMR deficiencies. Reduced MSH2, MSH3, and MLH1 slowed repeat expansion to the largest degree, while lowering either PMS1, PMS2, or MLH3 slowed it to a lesser degree. These effects were recapitulated in iPSC-derived striatal cultures where MutL factor expression was lowered. CRISPRi-mediated lowering of key MMR factor expression to levels feasibly achievable by current therapeutic approaches was able to effectively slow the expansion of the HTT CAG tract. We highlight members of the MutL family as potential targets to slow pathogenic repeat expansion with the aim to delay onset and progression of HD and potentially other repeat expansion disorders exhibiting somatic instability.


Subject(s)
DNA Mismatch Repair , Huntingtin Protein , Huntington Disease , Induced Pluripotent Stem Cells , Trinucleotide Repeat Expansion , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , DNA Mismatch Repair/genetics , Induced Pluripotent Stem Cells/metabolism , Trinucleotide Repeat Expansion/genetics , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Genes, Modifier , MutS Homolog 3 Protein/genetics , MutS Homolog 3 Protein/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , MutL Proteins/genetics , MutL Proteins/metabolism , CRISPR-Cas Systems , Genome-Wide Association Study
16.
Genetics ; 227(3)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38691577

ABSTRACT

Although gene conversion (GC) in Saccharomyces cerevisiae is the most error-free way to repair double-strand breaks (DSBs), the mutation rate during homologous recombination is 1,000 times greater than during replication. Many mutations involve dissociating a partially copied strand from its repair template and re-aligning with the same or another template, leading to -1 frameshifts in homonucleotide runs, quasipalindrome (QP)-associated mutations and microhomology-mediated interchromosomal template switches. We studied GC induced by HO endonuclease cleavage at MATα, repaired by an HMR::KI-URA3 donor. We inserted into HMR::KI-URA3 an 18-bp inverted repeat where one arm had a 4-bp insertion. Most GCs yield MAT::KI-ura3::QP + 4 (Ura-) outcomes, but template-switching produces Ura+ colonies, losing the 4-bp insertion. If the QP arm without the insertion is first encountered by repair DNA polymerase and is then (mis)used as a template, the palindrome is perfected. When the QP + 4 arm is encountered first, Ura+ derivatives only occur after second-end capture and second-strand synthesis. QP + 4 mutations are suppressed by mismatch repair (MMR) proteins Msh2, Msh3, and Mlh1, but not Msh6. Deleting Rdh54 significantly reduces QP mutations only when events creating Ura+ occur in the context of a D-loop but not during second-strand synthesis. A similar bias is found with a proofreading-defective DNA polymerase mutation (poI3-01). DSB-induced mutations differed in several genetic requirements from spontaneous events. We also created a + 1 frameshift in the donor, expanding a run of 4 Cs to 5 Cs. Again, Ura+ recombinants markedly increased by disabling MMR, suggesting that MMR acts during GC but favors the unbroken, template strand.


Subject(s)
DNA Breaks, Double-Stranded , DNA Mismatch Repair , Frameshift Mutation , Mutagenesis , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Gene Conversion , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , MutS Homolog 3 Protein/genetics , MutS Homolog 3 Protein/metabolism , MutL Protein Homolog 1
17.
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
18.
Prostate ; 84(10): 945-953, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38629217

ABSTRACT

BACKGROUND: Pathogenic germline variants in the mismatch repair (MMR) genes are associated with an increased risk of prostate cancer (PCa). Since 2010 we have recommended MMR carriers annual PSA testing from the age of 40. Prospective studies of the outcome of long-term PSA screening are lacking. This study aimed to investigate the incidence and characteristics of PCa in Norwegian MMR carriers attending annual PSA screening (PSA threshold >3.0 ng/mL) to evaluate whether our recommendations should be continued. METHODS: This is a prospective observational study of 225 male MMR carriers who were recommended annual PSA screening by the Section of Inherited Cancer, Oslo University Hospital from 2010 and onwards. Incidence and tumor characteristics (age, PSA at diagnosis, Gleason score, TNM score) were described. IHC and MSI-analyses were done on available tumors. Standardized incidence ratio (SIR) was calculated based on data from the Cancer Registry of Norway. RESULTS: Twenty-two of 225 (9.8%) had been diagnosed with PCa, including 10/69 (14.5%) MSH2 carriers and 8/61 (13.1%) MSH6 carriers. Ten of 20 (50%) tumors had Gleason score ≥4 + 3 on biopsy and 6/11 (54.5%) had a pathological T3a/b stage. Eight of 17 (47.1%) tumors showed abnormal staining on IHC and 3/13 (23.1%) were MSI-high. SIR was 9.54 (95% CI 5.98-14.45) for all MMR genes, 13.0 (95% CI 6.23-23.9) for MSH2 and 13.74 for MSH6 (95% CI 5.93-27.08). CONCLUSIONS: Our results indicate that the MMR genes, and especially MSH2 and MSH6, are associated with a significant risk of PCa, and a high number of tumors show aggressive characteristics. While the impact of screening on patient outcomes remains to be more firmly established, the high SIR values we observe provide support for continued PSA screening of MSH2 and MSH6 carriers. Studies are needed to provide optimal recommendations for PSA-threshold and to evaluate whether MLH1 and PMS2 carriers should not be recommended screening.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Early Detection of Cancer , Prostate-Specific Antigen , Prostatic Neoplasms , Humans , Male , Norway/epidemiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/blood , Prostate-Specific Antigen/blood , Middle Aged , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/epidemiology , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Prospective Studies , Early Detection of Cancer/methods , Aged , Adult , MutS Homolog 2 Protein/genetics , Incidence , DNA Mismatch Repair/genetics , Neoplasm Grading , DNA-Binding Proteins/genetics
19.
Cancer Prev Res (Phila) ; 17(7): 311-324, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38643981

ABSTRACT

Lynch syndrome (LS) is the most prevalent heritable form of colorectal cancer. Its early onset and high lifetime risk for colorectal cancer emphasize the necessity for effective chemoprevention. NFE2L2 (NRF2) is often considered a potential druggable target, and many chemopreventive compounds induce NRF2. However, although NRF2 counteracts oxidative stress, it is also overexpressed in colorectal cancer and may promote tumorigenesis. In this study, we evaluated the role of NRF2 in the prevention of LS-associated neoplasia. We found increased levels of NRF2 in intestinal epithelia of mice with intestinal epithelium-specific Msh2 deletion (MSH2ΔIEC) compared with C57BL/6 (wild-type) mice, as well as an increase in downstream NRF2 targets NAD(P)H dehydrogenase (quinone 1) and glutamate-cysteine ligase catalytic subunit. Likewise, NRF2 levels were increased in human MSH2-deficient LS tumors compared with healthy human controls. In silico analysis of a publicly accessible RNA sequencing LS dataset also found an increase in downstream NRF2 targets. Upon crossing MSH2ΔIEC with Nrf2null (MSH2ΔIECNrf2null) mice, we unexpectedly found reduced tumorigenesis in MSH2ΔIECNrf2null mice compared with MSH2ΔIEC mice after 40 weeks, which occurred despite an increase in oxidative damage in MSH2ΔIECNrf2null mice. The loss of NRF2 impaired proliferation as seen by Ki67 intestinal staining and in organoid cultures. This was accompanied by diminished WNT/ß-catenin signaling, but apoptosis was unaffected. Microbial α-diversity increased over time with the loss of NRF2 based upon 16S rRNA gene amplicon sequencing of murine fecal samples. Altogether, we show that NRF2 protein levels are increased in MSH2 deficiency and associated neoplasia, but the loss of NRF2 attenuates tumorigenesis. Activation of NRF2 may not be a feasible strategy for chemoprevention in LS. Prevention Relevance: Patients with LS have an early onset and high lifetime risk for colorectal cancer. In this study, we show that NRF2 protein levels are increased in MSH2 deficiency and associated neoplasia, but the loss of NRF2 attenuates tumorigenesis. This suggests that NRF2 may not be a tumor suppressor in this specific context.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Disease Models, Animal , Mice, Inbred C57BL , MutS Homolog 2 Protein , NF-E2-Related Factor 2 , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Mice , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Humans , Carcinogenesis/genetics , Carcinogenesis/pathology , Mice, Knockout , Female , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male
20.
Nucleic Acids Res ; 52(10): 5732-5755, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38597682

ABSTRACT

Expansion of a G4C2 repeat in the C9orf72 gene is associated with familial Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD). To investigate the underlying mechanisms of repeat instability, which occurs both somatically and intergenerationally, we created a novel mouse model of familial ALS/FTD that harbors 96 copies of G4C2 repeats at a humanized C9orf72 locus. In mouse embryonic stem cells, we observed two modes of repeat expansion. First, we noted minor increases in repeat length per expansion event, which was dependent on a mismatch repair pathway protein Msh2. Second, we found major increases in repeat length per event when a DNA double- or single-strand break (DSB/SSB) was artificially introduced proximal to the repeats, and which was dependent on the homology-directed repair (HDR) pathway. In mice, the first mode primarily drove somatic repeat expansion. Major changes in repeat length, including expansion, were observed when SSB was introduced in one-cell embryos, or intergenerationally without DSB/SSB introduction if G4C2 repeats exceeded 400 copies, although spontaneous HDR-mediated expansion has yet to be identified. These findings provide a novel strategy to model repeat expansion in a non-human genome and offer insights into the mechanism behind C9orf72 G4C2 repeat instability.


Subject(s)
C9orf72 Protein , DNA Repeat Expansion , Genomic Instability , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , Disease Models, Animal , DNA Breaks, Double-Stranded , DNA Repeat Expansion/genetics , Frontotemporal Dementia/genetics , Gene Knock-In Techniques , Genomic Instability/genetics , MutS Homolog 2 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL