Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 759
Filtrer
1.
Int J Gynecol Cancer ; 34(7): 993-1000, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38950928

RÉSUMÉ

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.


Sujet(s)
Réparation de mésappariement de l'ADN , Protéines de liaison à l'ADN , Protéine-2 homologue de MutS , Tumeurs du col de l'utérus , Humains , Femelle , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/mortalité , Pronostic , Adulte d'âge moyen , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Protéine-2 homologue de MutS/métabolisme , Protéine-2 homologue de MutS/biosynthèse , Protéine-2 homologue de MutS/génétique , Adulte , Sujet âgé , Mismatch repair endonuclease PMS2/métabolisme , Mismatch repair endonuclease PMS2/génétique , Protéine-1 homologue de MutL/métabolisme , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/biosynthèse
2.
Zhonghua Bing Li Xue Za Zhi ; 53(7): 702-708, 2024 Jul 08.
Article de Chinois | MEDLINE | ID: mdl-38955702

RÉSUMÉ

Objective: To investigate the clinicopathological characteristics and prognostic factors of sporadic mismatch repair deficient (dMMR) colorectal cancer. Methods: A total of 120 cases of sporadic dMMR colorectal cancer from July 2015 to April 2021 were retrospectively collected in Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College. Patients with Lynch syndrome; synchronous multiple colorectal cancers; preoperative anti-tumor treatments such as chemotherapy and radiotherapy; and those with incomplete follow-up information were excluded based on family history and next-generation sequencing (NGS) test results. Immunohistochemical stains were used to detect the expression of mismatch repair proteins, methylation-specific PCR for methylation testing, and fluorescent PCR for BRAF V600E gene mutation detection. The clinical and pathological data, and gene mutation status were analyzed. Follow-up was done to assess survival and prognosis including progression-free survival and overall survival rate. Results: Sporadic dMMR colorectal cancer occurred more frequently in the right side of the colon, in females, and in the elderly. Morphologically, it was mostly moderately-differentiated, and most patients had low-grade tumor budding. In terms of immunohistochemical expression, MLH1 and PMS2 loss were dominant, and there were age and location-specificities in protein expression. MLH1 methylation was commonly detected in elderly female patients and rare in young male patients; while MLH1 and PMS2 deficiency, and BRAF V600E mutation occurred more often on the right side (P<0.05). The 3-year and 5-year progression-free survival rates were 90.7% and 88.7% respectively, and the 3-year and 5-year overall survival rates were 92.8% and 90.7% respectively. Tumor budding status was an independent risk factor affecting patient recurrence (hazard ratio=3.375, 95% confidence interval: 1.060-10.741, P=0.039), patients with low-grade tumor budding had better prognosis, and those with medium or high-grade tumor budding had poor prognosis. Conclusion: For dMMR colorectal cancer patients, tumor budding status is an independent risk factor for recurrence.


Sujet(s)
Tumeurs colorectales , Réparation de mésappariement de l'ADN , Protéines proto-oncogènes B-raf , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Mâle , Femelle , Pronostic , Études rétrospectives , Protéines proto-oncogènes B-raf/génétique , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/métabolisme , Mutation , Taux de survie , Adulte d'âge moyen , Sujet âgé , Méthylation de l'ADN , Adulte , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Tumeurs colorectales héréditaires sans polypose/métabolisme
3.
Mol Biol Rep ; 51(1): 764, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38874740

RÉSUMÉ

BACKGROUND AND AIM: Colorectal cancer (CRC) originates from pre-existing polyps in the colon. The development of different subtypes of CRC is influenced by various genetic and epigenetic characteristics. CpG island methylator phenotype (CIMP) is found in about 15-20% of sporadic CRCs and is associated with hypermethylation of certain gene promoters. This study aims to find prognostic genes and compare their expression and methylation status as potential biomarkers in patients with serrated sessile adenomas/polyps (SSAP) and CRC, in order to evaluate which, one is a better predictor of disease. METHOD: This study employed a multi-phase approach to investigate genes associated with CRC and SSAP. Initially, two gene expression datasets were analyzed using R and Limma package to identify differentially expressed genes (DEGs). Venn diagram analysis further refined the selection, revealing four genes from the Weissenberg panel with significant changes. These genes, underwent thorough in silico evaluations. Once confirmed, they proceeded to wet lab experimentation, focusing on expression and methylation status. This comprehensive methodology ensured a robust examination of the genes involved in CRC and SSAP. RESULT: This study identified cancer-specific genes, with 8,351 and 1,769 genes specifically down-regulated in SSAP and CRC tissues, respectively. The down-regulated genes were associated with cell adhesion, negative regulation of cell proliferation, and drug response. Four highly downregulated genes in the Weissenberg panel, including CACNA1G, IGF2, MLH1, and SOCS1. In vitro analysis showed that they are hypermethylated in both SSAP and CRC samples while their expressions decreased only in CRC samples. CONCLUSION: This suggests that the decrease in gene expression could help determine whether a polyp will become cancerous. Using both methylation status and gene expression status of genes in the Weissenberg panel in prognostic tests may lead to better prognoses for patients.


Sujet(s)
Tumeurs colorectales , Ilots CpG , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux , Facteur de croissance IGF-II , Protéine-1 homologue de MutL , Protéine-1 suppressive de la signalisation des cytokines , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Protéine-1 suppressive de la signalisation des cytokines/génétique , Protéine-1 suppressive de la signalisation des cytokines/métabolisme , Méthylation de l'ADN/génétique , Facteur de croissance IGF-II/génétique , Facteur de croissance IGF-II/métabolisme , Régulation de l'expression des gènes tumoraux/génétique , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/métabolisme , Ilots CpG/génétique , Femelle , Polypes coliques/génétique , Polypes coliques/métabolisme , Polypes coliques/anatomopathologie , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Mâle , Régulation négative/génétique , Simulation numérique , Adulte d'âge moyen , Adénomes/génétique , Adénomes/métabolisme , Adénomes/anatomopathologie , Régions promotrices (génétique)/génétique , Canaux calciques de type T/génétique , Canaux calciques de type T/métabolisme , Analyse de profil d'expression de gènes/méthodes , Sujet âgé , Pronostic
4.
Cancer Epidemiol ; 91: 102601, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38905781

RÉSUMÉ

BACKGROUND: Serrated lesions and polyps (SP) are precursors of up to 30 % of colorectal cancers (CRC) through the serrated pathway. This often entails early BRAF mutations and MLH1 hypermethylation leading to mismatch repair deficient (dMMR) CRC. We investigated predictors of dMMR CRC among patients with co-occurrence of CRC and SP to increase our knowledge on the serrated pathway. METHODS: We used data from The Danish Pathology Registry and Danish Colorectal Cancer Groups Database from the period 2010-2021 to investigate risk factors for development of dMMR CRC. We used logistic regression models to identify difference in risk factors of developing dMMR CRC in comparison to CRC with proficient MMR (pMMR). RESULTS: We included 3273 patients with a median age of 70.7 years [64.3,76.4] of which 1850 (56.5 %) were male. dMMR CRC was present in 592 patients (18.1 %), with loss of MLH1/PMS2 being most common. The risk of dMMR CRC was significantly higher in females OR 3.47 [2.87;4.20]. When adjusting for age, SP subtype, conventional adenomas (CA), anatomical location and lifestyle factors, female sex remained the strongest predictor OR 2.84 [2.27;3.56]. The presence of sessile serrated lesions with or without dysplasia was related to higher risk OR 1.60 [1.11;2.31] and OR 1.42 [1.11;1.82] respectively, while conventional adenomas constituted a lower risk OR 0.68 [0.55;0.84]. CONCLUSION: In conclusion we found several predictors of whom female sex had the strongest correlation with dMMR CRC in patients with SP.


Sujet(s)
Tumeurs colorectales , Enregistrements , Humains , Mâle , Femelle , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/épidémiologie , Tumeurs colorectales/génétique , Sujet âgé , Adulte d'âge moyen , Danemark/épidémiologie , Polypes coliques/anatomopathologie , Polypes coliques/épidémiologie , Facteurs de risque , Réparation de mésappariement de l'ADN , Études de cohortes , Protéine-1 homologue de MutL/génétique
5.
Int J Mol Sci ; 25(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891846

RÉSUMÉ

Tumor recurrence and drug resistance are responsible for poor prognosis in colorectal cancer (CRC). DNA mismatch repair (MMR) deficiency or elevated interleukin-8 (IL-8) levels are characteristics of CRCs, which have been independently correlated with treatment resistance to common therapies. We recently demonstrated significantly impaired therapeutical response and increased IL-8 release of CRC cell lines with reduced expression of MMR protein MLH1 as well as cytoskeletal non-erythrocytic spectrin alpha II (SPTAN1). In the present study, decreased intratumoral MLH1 and SPTAN1 expression in CRCs could be significantly correlated with enhanced serum IL-8. Furthermore, using stably reduced SPTAN1-expressing SW480, SW620 or HT-29 cell lines, the RAS-mediated RAF/MEK/ERK pathway was analyzed. Here, a close connection between low SPTAN1 expression, increased IL-8 secretion, enhanced extracellular-signal-regulated kinase (ERK) phosphorylation and a mesenchymal phenotype were detected. The inhibition of ERK by U0126 led to a significant reduction in IL-8 secretion, and the combination therapy of U0126 with FOLFOX optimizes the response of corresponding cancer cell lines. Therefore, we hypothesize that the combination therapy of FOLFOX and U0126 may have great potential to improve drug efficacy on this subgroup of CRCs, showing decreased MLH1 and SPTAN1 accompanied with high serum IL-8 in affected patients.


Sujet(s)
Butadiènes , Tumeurs colorectales , Fluorouracil , Interleukine-8 , Nitriles , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Interleukine-8/métabolisme , Interleukine-8/génétique , Fluorouracil/pharmacologie , Fluorouracil/usage thérapeutique , Butadiènes/pharmacologie , Nitriles/pharmacologie , Lignée cellulaire tumorale , Composés organiques du platine/pharmacologie , Composés organiques du platine/usage thérapeutique , Leucovorine/usage thérapeutique , Leucovorine/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Femelle , Mâle , Extracellular Signal-Regulated MAP Kinases/métabolisme , Cellules HT29 , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Protéine-1 homologue de MutL/métabolisme , Protéine-1 homologue de MutL/génétique , Adulte d'âge moyen , Sujet âgé , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques
6.
PLoS One ; 19(6): e0304141, 2024.
Article de Anglais | MEDLINE | ID: mdl-38843250

RÉSUMÉ

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.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Réparation de mésappariement de l'ADN , Protéine-1 homologue de MutL , Tumeurs colorectales héréditaires sans polypose/génétique , Humains , Mâle , Protéine-1 homologue de MutL/génétique , Femelle , Réparation de mésappariement de l'ADN/génétique , Adulte d'âge moyen , Protéine-2 homologue de MutS/génétique , Adulte , Tunisie , Pedigree , Turquie , Sujet âgé , Mutation faux-sens
7.
Zhonghua Bing Li Xue Za Zhi ; 53(6): 605-609, 2024 Jun 08.
Article de Chinois | MEDLINE | ID: mdl-38825907

RÉSUMÉ

Objective: To investigate the clinicopathological features of children with metachronous or synchronous primary tumors and to identify related genetic tumor syndromes. Methods: The clinicopathological data of 4 children with multiple primary tumors diagnosed in the Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China from 2011 to 2023 were collected. The histological, immunophenotypic and molecular characteristics were examined using H&E staining, immunohistochemical staining, PCR, Sanger sequencing and next-generation sequencing (NGS). The patients were followed up. Results: Case 1 was an 8-year-old boy with the adrenal cortical carcinoma, and 5 years later a poorly differentiated gastric adenocarcinoma was detected. Case 2 was a 2-year-old boy, presented with a left ventricular choroid plexus carcinoma, and a hepatoblastoma was detected 8 months later. Case 3 was a 9-month-old girl, diagnosed with renal rhabdoid tumor first and intracranial atypical teratoid/rhabdoid tumor (AT/RT) 3 months later. Case 4 was a 7-year-old boy and had a sigmoid colon adenocarcinoma 3 years after the diagnosis of a glioblastoma. The morphology and immunohistochemical features of the metachronous or synchronous primary tumors in the 4 cases were similar to the corresponding symptom-presenting/first-diagnosed tumors. No characteristic germ line mutations were detected in cases 1 and 2 by relevant molecular detection, and the rhabdoid tumor predisposition syndrome was confirmed in case 3 using NGS. Case 4 was clearly related to constitutional mismatch repair deficiency as shown by the molecular testing and clinical features. Conclusions: Childhood multiple primary tumors are a rare disease with histological morphology and immunophenotype similar to the symptom-presenting tumors. They are either sporadic or associated with a genetic (tumor) syndrome. The development of both tumors can occur simultaneously (synchronously) or at different times (metachronously). Early identification of the children associated with genetic tumor syndromes can facilitate routine tumor screening and early treatment.


Sujet(s)
Hépatoblastome , Tumeurs du rein , Tumeurs du foie , Tumeurs primitives multiples , Tumeur rhabdoïde , Tumeurs de l'estomac , Humains , Mâle , Enfant , Femelle , Enfant d'âge préscolaire , Tumeurs primitives multiples/génétique , Tumeurs primitives multiples/anatomopathologie , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du rein/anatomopathologie , Tumeurs du rein/génétique , Nourrisson , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/génétique , Tumeur rhabdoïde/génétique , Tumeur rhabdoïde/anatomopathologie , Hépatoblastome/génétique , Hépatoblastome/anatomopathologie , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Adénocarcinome/diagnostic , Tumeurs du plexus choroïde/génétique , Tumeurs du plexus choroïde/anatomopathologie , Tumeurs du plexus choroïde/diagnostic , Carcinome corticosurrénalien/génétique , Carcinome corticosurrénalien/anatomopathologie , Tumeurs corticosurrénaliennes/anatomopathologie , Tumeurs corticosurrénaliennes/génétique , Tératome/anatomopathologie , Tératome/génétique , Tératome/chirurgie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Protéine SMARCB1/génétique , Protéine-1 homologue de MutL/génétique , Seconde tumeur primitive/anatomopathologie , Seconde tumeur primitive/génétique , Séquençage nucléotidique à haut débit , Syndromes néoplasiques héréditaires/génétique , Syndromes néoplasiques héréditaires/anatomopathologie
8.
J Int Med Res ; 52(6): 3000605241259747, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38902203

RÉSUMÉ

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.


Sujet(s)
Réparation de mésappariement de l'ADN , Protéines de liaison à l'ADN , Mismatch repair endonuclease PMS2 , Tumeurs du sein triple-négatives , Humains , Femelle , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Adulte d'âge moyen , Adulte , Études rétrospectives , Mismatch repair endonuclease PMS2/métabolisme , Mismatch repair endonuclease PMS2/génétique , Sujet âgé , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Protéine-1 homologue de MutL/métabolisme , Protéine-1 homologue de MutL/génétique , Protéine-2 homologue de MutS/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Taux de survie , Immunohistochimie , Sujet âgé de 80 ans ou plus , Pronostic
9.
Cells ; 13(10)2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38786078

RÉSUMÉ

Prime editing (PE), a recent progression in CRISPR-based technologies, holds promise for precise genome editing without the risks associated with double-strand breaks. It can introduce a wide range of changes, including single-nucleotide variants, insertions, and small deletions. Despite these advancements, there is a need for further optimization to overcome certain limitations to increase efficiency. One such approach to enhance PE efficiency involves the inhibition of the DNA mismatch repair (MMR) system, specifically MLH1. The rationale behind this approach lies in the MMR system's role in correcting mismatched nucleotides during DNA replication. Inhibiting this repair pathway creates a window of opportunity for the PE machinery to incorporate the desired edits before permanent DNA repair actions. However, as the MMR system plays a crucial role in various cellular processes, it is important to consider the potential risks associated with manipulating this system. The new versions of PE with enhanced efficiency while blocking MLH1 are called PE4 and PE5. Here, we explore the potential risks associated with manipulating the MMR system. We pay special attention to the possible implications for human health, particularly the development of cancer.


Sujet(s)
Systèmes CRISPR-Cas , Réparation de mésappariement de l'ADN , Édition de gène , Humains , Édition de gène/méthodes , Systèmes CRISPR-Cas/génétique , Réparation de l'ADN , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/métabolisme , Animaux
10.
Gastric Cancer ; 27(4): 760-771, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38744779

RÉSUMÉ

BACKGROUND: Microsatellite instability-high (MSI-H) tumors are distinct molecular subtypes in gastric cancer. However, a few studies have comprehensively reported the molecular features of MSI-H tumors and their prognostic factors in locally advanced gastric cancer. This study aimed to clarify the molecular features and prognostic factors of locally advanced MSI-H gastric cancer. METHODS: This study included 499 patients with locally advanced gastric cancer who underwent radical gastrectomy. We evaluated the MSI status and compared with previously published whole-exome sequencing, panel sequencing, and gene expression profiling data. Clinicopathological characteristics and molecular profiles were compared between patients with MSI-H and microsatellite stable (MSS) gastric cancer. A subgroup analysis of survival was performed in patients with MSI-H gastric cancer. RESULTS: MSI-H tumors were detected in 79 of 499 patients (15.8%). MSI-H tumors were associated with an increased tumor mutational burden, MLH1 downregulation, CD274 (PD-L1) upregulation, and enrichment of cell cycle pathways. Among patients with MSI-H gastric cancer, the disease-specific survival (DSS) tended to be better in the surgery plus tegafur, gimeracil, and oteracil potassium (S-1) adjuvant chemotherapy group than in the surgery alone group, especially for stage III patients. Furthermore, DSS was better in the T cell-inflamed gene expression signature-high group, and it tended to be worse in the non-solid type poorly differentiated adenocarcinoma group. CONCLUSIONS: The molecular features and prognostic factors of locally advanced MSI-H gastric cancer were clarified. S-1 adjuvant chemotherapy appears to be beneficial, and the T cell-inflamed gene expression signature and histopathological type are prognostic factors in MSI-H tumors.


Sujet(s)
Gastrectomie , Instabilité des microsatellites , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/mortalité , Femelle , Mâle , Pronostic , Sujet âgé , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Tégafur/usage thérapeutique , Adulte , Association médicamenteuse , Acide oxonique/usage thérapeutique , Sujet âgé de 80 ans ou plus , Antigène CD274/génétique , Antigène CD274/métabolisme , Taux de survie , Mutation , Analyse de profil d'expression de gènes , , Protéine-1 homologue de MutL/génétique , Régulation de l'expression des gènes tumoraux
11.
Am J Hum Genet ; 111(6): 1165-1183, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38749429

RÉSUMÉ

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.


Sujet(s)
Réparation de mésappariement de l'ADN , Protéine huntingtine , Maladie de Huntington , Cellules souches pluripotentes induites , Expansion de trinucléotide répété , Humains , Maladie de Huntington/génétique , Maladie de Huntington/métabolisme , Réparation de mésappariement de l'ADN/génétique , Cellules souches pluripotentes induites/métabolisme , Expansion de trinucléotide répété/génétique , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/métabolisme , Protéine-2 homologue de MutS/génétique , Protéine-2 homologue de MutS/métabolisme , Gènes modificateurs , Protéine-3 homologue de MutS/génétique , Protéine-3 homologue de MutS/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines MutL/génétique , Protéines MutL/métabolisme , Systèmes CRISPR-Cas , Étude d'association pangénomique
12.
Br J Cancer ; 131(2): 334-346, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38796599

RÉSUMÉ

BACKGROUND: Identifying the target of natural killer (NK) cells in colorectal cancer (CRC) is critical for optimising the clinical use of NK cell-mediated immunotherapy. Mismatch repair deficiency (dMMR) is associated with high immune cell infiltration and MHC Class I defects. Whether dMMR CRC responses to NK cell therapy remains unclear. METHODS: MLH1, DR4, and DR5 knockout cell lines were established using CRISPR-Cas9 system. NK92-MI or NK cell isolated from BABL/C mice were used as effector cells against tumour cells. Inflammatory cytokines secretion by CRC cells was assessed via cytokine analysis. NK-cell-deficient/proficient animal models were used to validate the NK cell sensitivity. RESULTS: We observed that dMMR CRC cells were more sensitive to NK cell-mediated cytotoxicity than were mismatch-repair-proficient (pMMR) CRC cells. In dMMR CRC, Death receptor (DR)4/5 was upregulated and mediated sensitivity to NK cell-mediated cytotoxicity. DR4/5-mediated secretion of interleukin -12 sustained NK cell viability in dMMR CRC. NK cell depletion induced dMMR CRC tumour growth, and NK cell transfer inhibited lung metastasis of dMMR CRC with DR4/5 expression in vivo. TP53 upregulated DR4/DR5 expression in dMMR CRC. CONCLUSIONS: dMMR associated with increased sensitivity to NK cell-mediated cytotoxicity in CRC. DR4/DR5 sensitise dMMR CRC to NK cell-mediated cytotoxicity.


Sujet(s)
Tumeurs colorectales , Cellules tueuses naturelles , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Animaux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/immunologie , Souris , Humains , Lignée cellulaire tumorale , Réparation de mésappariement de l'ADN , Cytotoxicité immunologique , Récepteurs de TRAIL/génétique , Récepteurs de TRAIL/métabolisme , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/déficit , Protéine-1 homologue de MutL/métabolisme , Souris de lignée BALB C , Syndromes néoplasiques héréditaires/génétique , Syndromes néoplasiques héréditaires/immunologie , Syndromes néoplasiques héréditaires/anatomopathologie , Tumeurs du cerveau
13.
Genome Biol ; 25(1): 131, 2024 05 21.
Article de Anglais | MEDLINE | ID: mdl-38773623

RÉSUMÉ

BACKGROUND: High-efficiency prime editing (PE) is desirable for precise genome manipulation. The activity of mammalian PE systems can be largely improved by inhibiting DNA mismatch repair by coexpressing a dominant-negative variant of MLH1. However, this strategy has not been widely used for PE optimization in plants, possibly because of its less conspicuous effects and inconsistent performance at different sites. RESULTS: We show that direct RNAi knockdown of OsMLH1 in an ePE5c system increases the efficiency of our most recently updated PE tool by 1.30- to 2.11-fold in stably transformed rice cells, resulting in as many as 85.42% homozygous mutants in the T0 generation. The high specificity of ePE5c is revealed by whole-genome sequencing. To overcome the partial sterility induced by OsMLH1 knockdown of ePE5c, a conditional excision system is introduced to remove the RNAi module by Cre-mediated site-specific recombination. Using a simple approach of enriching excision events, we generate 100% RNAi module-free plants in the T0 generation. The increase in efficiency due to OsMLH1 knockdown is maintained in the excised plants, whose fertility is not impaired. CONCLUSIONS: This study provides a safe and reliable plant PE optimization strategy for improving editing efficiency without disturbing plant development via transient MMR inhibition with an excisable RNAi module of MLH1.


Sujet(s)
Édition de gène , Oryza , Protéines végétales , Oryza/génétique , Protéines végétales/génétique , Protéines végétales/métabolisme , Fécondité/génétique , Techniques de knock-down de gènes , Protéine-1 homologue de MutL/génétique , Interférence par ARN , Systèmes CRISPR-Cas , Végétaux génétiquement modifiés
14.
Gulf J Oncolog ; 1(45): 35-41, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38774931

RÉSUMÉ

BACKGROUND: Microsatellite instability (MSI) is a pattern of hyper mutation that occurs at microsatellite level in the genome and result due to error in the mismatch repair system. MSI is caused by defective mismatch repair (MMR) genes associated with either hyper methylation of MMR genes or BRAF mutations. Anti-MLH-1, anti-MSH-2, anti-MSH-6 and anti-PMS2 monoclonal antibodies are used for Immunohistochemical analysis. METHODS: The immunohistochemical expression of MSI proteins were assessed in 72 cases of colorectal carcinoma. These were classified based on the expression of MLH1, MSH2, MSH6 and PMS2 proteins. RESULTS: There were 57 percent of cases showing loss of at least one antibodies, and 43 percent cases showing intact expression of all antibodies (MLH1, MSH2, MSH6 and PMS2). CONCLUSION: In conclusion, our study provides valuable insights into the expression of mismatch repair in colorectal adenocarcinoma through immunohistochemistry analysis conducted at our tertiary care centre. These findings hold significant clinical implications, suggesting further testing for BRAF and MLH1 Promoter Hypermethylation to confirm possibility of Lynch syndrome. KEY WORDS: IHC, MMR, CRC.


Sujet(s)
Adénocarcinome , Tumeurs colorectales , Réparation de mésappariement de l'ADN , Immunohistochimie , Centres de soins tertiaires , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Mâle , Adénocarcinome/génétique , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Immunohistochimie/méthodes , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte , Protéine-1 homologue de MutL/génétique
15.
Dermatol Online J ; 30(1)2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38762859

RÉSUMÉ

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.


Sujet(s)
Réparation de mésappariement de l'ADN , Mutation germinale , Immunosuppresseurs , Syndrome de Muir-Torre , Tumeurs des glandes sébacées , Humains , Syndrome de Muir-Torre/génétique , Mâle , Réparation de mésappariement de l'ADN/génétique , Immunosuppresseurs/usage thérapeutique , Immunosuppresseurs/effets indésirables , Tumeurs des glandes sébacées/génétique , Adulte d'âge moyen , Protéine-2 homologue de MutS/génétique , Sujet immunodéprimé , Protéine-1 homologue de MutL/génétique , Tumeurs cutanées/génétique , Analyse de mutations d'ADN
16.
Nat Commun ; 15(1): 4002, 2024 May 11.
Article de Anglais | MEDLINE | ID: mdl-38734692

RÉSUMÉ

Precise genome editing is crucial for establishing isogenic human disease models and ex vivo stem cell therapy from the patient-derived hPSCs. Unlike Cas9-mediated knock-in, cytosine base editor and prime editor achieve the desirable gene correction without inducing DNA double strand breaks. However, hPSCs possess highly active DNA repair pathways and are particularly susceptible to p53-dependent cell death. These unique characteristics impede the efficiency of gene editing in hPSCs. Here, we demonstrate that dual inhibition of p53-mediated cell death and distinct activation of the DNA damage repair system upon DNA damage by cytosine base editor or prime editor additively enhanced editing efficiency in hPSCs. The BE4stem system comprised of p53DD, a dominant negative p53, and three UNG inhibitor, engineered to specifically diminish base excision repair, improves cytosine base editor efficiency in hPSCs. Addition of dominant negative MLH1 to inhibit mismatch repair activity and p53DD in the conventional prime editor system also significantly enhances prime editor efficiency in hPSCs. Thus, combined inhibition of the distinct cellular cascades engaged in hPSCs upon gene editing could significantly enhance precise genome editing in these cells.


Sujet(s)
Systèmes CRISPR-Cas , Altération de l'ADN , Réparation de l'ADN , Édition de gène , Protéine p53 suppresseur de tumeur , Édition de gène/méthodes , Humains , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Lignée cellulaire , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/métabolisme , Cytosine/métabolisme
17.
Genetics ; 227(3)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38657110

RÉSUMÉ

The accurate segregation of homologous chromosomes during the Meiosis I reductional division in most sexually reproducing eukaryotes requires crossing over between homologs. In baker's yeast approximately 80% of meiotic crossovers result from Mlh1-Mlh3 and Exo1 acting to resolve double-Holliday junction intermediates in a biased manner. Little is known about how Mlh1-Mlh3 is recruited to recombination intermediates to perform its role in crossover resolution. We performed a gene dosage screen in baker's yeast to identify novel genetic interactors with Mlh1-Mlh3. Specifically, we looked for genes whose lowered dosage reduced meiotic crossing over using sensitized mlh3 alleles that disrupt the stability of the Mlh1-Mlh3 complex and confer defects in mismatch repair but do not disrupt meiotic crossing over. To our surprise we identified genetic interactions between MLH3 and DMC1, the recombinase responsible for recombination between homologous chromosomes during meiosis. We then showed that Mlh3 physically interacts with Dmc1 in vitro and in vivo. Partial complementation of Mlh3 crossover functions was observed when MLH3 was expressed under the control of the CLB1 promoter (NDT80 regulon), suggesting that Mlh3 function can be provided late in meiotic prophase at some functional cost. A model for how Dmc1 could facilitate Mlh1-Mlh3's role in crossover resolution is presented.


Sujet(s)
Protéines du cycle cellulaire , Crossing-over , Méiose , Protéine-1 homologue de MutL , Protéines MutL , Protéines de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/métabolisme , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Protéines MutL/métabolisme , Protéines MutL/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Liaison aux protéines
18.
Int J Clin Oncol ; 29(7): 944-952, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38642190

RÉSUMÉ

BACKGROUND: Lynch-like syndrome (LLS) has recently been proposed as a third type of microsatellite instability (MSI) tumor after Lynch syndrome (LS) and sporadic MSI colorectal cancer (CRC) without either a germline variant of mismatch repair (MMR) genes or hypermethylation of the MLH1 gene. The present study aimed to clarify and compare the clinicopathological characteristics of LLS with those of the other MSI CRC subtypes. METHODS: In total, 2634 consecutive patients with CRC who underwent surgical resection and subsequently received universal tumor screening (UTS), including MSI analysis were enrolled between January 2008 and November 2019. Genetic testing was performed in patients suspected of having Lynch syndrome. RESULTS: UTS of the cohort found 146 patients with MSI CRC (5.5%). Of these, excluding sporadic MSI CRC, 30 (1.1%) had a diagnosis of LS, and 19 (0.7%) had no germline pathogenic variants of the MMR gene. The CRC type in the latter group was identified as LLS. LLS occurred significantly more often in young patients, was left-sided, involved a KRAS variant and BRAF wild-type, and had a higher concordance rate with the Revised Bethesda Guidelines than sporadic MSI CRC. No significant differences were observed in terms of the clinicopathological factors between LLS and LS-associated MSI CRC; however, LLS had a lower frequency of LS-related neoplasms compared with LS. CONCLUSIONS: Distinguishing clinically between LS and LLS was challenging, but the incidence of neoplasms was higher in LS than in LLS, suggesting the need for different screening and surveillance methods for the two subtypes.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Instabilité des microsatellites , Humains , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , Adulte , Protéine-1 homologue de MutL/génétique , Réparation de mésappariement de l'ADN/génétique , Protéines proto-oncogènes B-raf/génétique , Sujet âgé de 80 ans ou plus , Protéines proto-oncogènes p21(ras)/génétique , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Dépistage génétique
19.
Int J Clin Oncol ; 29(7): 953-963, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38615286

RÉSUMÉ

BACKGROUND: Based on molecular characteristics, deficient DNA mismatch repair (dMMR) solid tumors are largely divided into three categories: somatically MLH1-hypermethylated tumors, Lynch syndrome (LS)-associated tumors, and Lynch-like syndrome (LLS)-associated tumors. The incidence of each of these conditions and the corresponding pathogenic genes related to LLS remain elusive. METHODS: We identified dMMR tumors in 3609 tumors from 9 different solid organs, including colorectal cancer, gastric cancer, small-bowel cancer, endometrial cancer, ovarian cancer, upper urinary tract cancer, urinary bladder cancer, prostate cancer, and sebaceous tumor, and comprehensively summarized the characterization of dMMR tumors. Characterization of dMMR tumors were performed as loss of at least one of MMR proteins (MLH1, MSH2, MSH6, and PMS2), by immunohistochemistry, followed by MLH1 promotor methylation analysis and genetic testing for MMR genes where appropriate. Somatic variant analysis of MMR genes and whole exome sequencing (WES) were performed in patients with LLS. RESULTS: In total, the incidence of dMMR tumors was 5.9% (24/3609). The incidence of dMMR tumors and the proportion of the three categorized dMMR tumors varied considerably with different tumor types. One to three likely pathogenic/pathogenic somatic MMR gene variants were detected in 15 out of the 16 available LLS tumors. One patient each from 12 patients who gave consent to WES demonstrated non-MMR germline variants affect function (POLQ or BRCA1). CONCLUSIONS: Our data regarding the LS to LLS ratio would be useful for genetic counseling in patients who are suspected to have LS, though the genetic backgrounds for the pathogenesis of LLS need further investigation.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Réparation de mésappariement de l'ADN , Mutation germinale , Humains , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Réparation de mésappariement de l'ADN/génétique , Femelle , Mâle , Incidence , Adulte d'âge moyen , Sujet âgé , Adulte , Protéine-1 homologue de MutL/génétique , Méthylation de l'ADN ,
20.
EBioMedicine ; 103: 105111, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38583260

RÉSUMÉ

BACKGROUND: Lynch syndrome (LS) is one of the most common hereditary cancer syndromes worldwide. Dominantly inherited mutation in one of four DNA mismatch repair genes combined with somatic events leads to mismatch repair deficiency and microsatellite instability (MSI) in tumours. Due to a high lifetime risk of cancer, regular surveillance plays a key role in cancer prevention; yet the observation of frequent interval cancers points to insufficient cancer prevention by colonoscopy-based methods alone. This study aimed to identify precancerous functional changes in colonic mucosa that could facilitate the monitoring and prevention of cancer development in LS. METHODS: The study material comprised colon biopsy specimens (n = 71) collected during colonoscopy examinations from LS carriers (tumour-free, or diagnosed with adenoma, or diagnosed with carcinoma) and a control group, which included sporadic cases without LS or neoplasia. The majority (80%) of LS carriers had an inherited genetic MLH1 mutation. The remaining 20% included MSH2 mutation carriers (13%) and MSH6 mutation carriers (7%). The transcriptomes were first analysed with RNA-sequencing and followed up with Gorilla Ontology analysis and Reactome Knowledgebase and Ingenuity Pathway Analyses to detect functional changes that might be associated with the initiation of the neoplastic process in LS individuals. FINDINGS: With pathway and gene ontology analyses combined with measurement of mitotic perimeters from colonic mucosa and tumours, we found an increased tendency to chromosomal instability (CIN), already present in macroscopically normal LS mucosa. Our results suggest that CIN is an earlier aberration than MSI and may be the initial cancer driving aberration, whereas MSI accelerates tumour formation. Furthermore, our results suggest that MLH1 deficiency plays a significant role in the development of CIN. INTERPRETATION: The results validate our previous findings from mice and highlight early mitotic abnormalities as an important contributor and precancerous marker of colorectal tumourigenesis in LS. FUNDING: This work was supported by grants from the Jane and Aatos Erkko Foundation, the Academy of Finland (330606 and 331284), Cancer Foundation Finland sr, and the Sigrid Jusélius Foundation. Open access is funded by Helsinki University Library.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Instabilité des microsatellites , Mitose , Humains , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Tumeurs colorectales héréditaires sans polypose/complications , Femelle , Mâle , Mitose/génétique , Adulte d'âge moyen , Mutation , Adulte , Sujet âgé , Protéine-1 homologue de MutL/génétique , Analyse de profil d'expression de gènes , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/étiologie , Carcinogenèse/génétique , Réparation de mésappariement de l'ADN/génétique , Transcriptome
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE